[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
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
remove_circle_outline
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
remove_circle_outline
remove_circle_outline

Search Results (2,888)

Search Parameters:
Keywords = cartilage

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 5375 KiB  
Article
Tunable Alginate-Polyvinyl Alcohol Bioinks for 3D Printing in Cartilage Tissue Engineering
by Alexandra Hunter Aitchison, Nicholas B. Allen, Kishen Mitra, Bijan Abar, Conor N. O’Neill, Kian Bagheri, Albert T. Anastasio and Samuel B. Adams
Gels 2024, 10(12), 829; https://doi.org/10.3390/gels10120829 (registering DOI) - 14 Dec 2024
Viewed by 400
Abstract
This study investigates 3D extrusion bioinks for cartilage tissue engineering by characterizing the physical properties of 3D-printed scaffolds containing varying alginate and polyvinyl alcohol (PVA) concentrations. We systematically investigated the effects of increasing PVA and alginate concentrations on swelling, degradation, and the elastic [...] Read more.
This study investigates 3D extrusion bioinks for cartilage tissue engineering by characterizing the physical properties of 3D-printed scaffolds containing varying alginate and polyvinyl alcohol (PVA) concentrations. We systematically investigated the effects of increasing PVA and alginate concentrations on swelling, degradation, and the elastic modulus of printed hydrogels. Swelling decreased significantly with increased PVA concentrations, while degradation rates rose with higher PVA concentrations, underscoring the role of PVA in modulating hydrogel matrix stability. The highest elastic modulus value was achieved with a composite of 5% PVA and 20% alginate, reaching 0.22 MPa, which approaches that of native cartilage. These findings demonstrate that adjusting PVA and alginate concentrations enables the development of bioinks with tailored physical and mechanical properties, supporting their potential use in cartilage tissue engineering and other biomedical applications. Full article
(This article belongs to the Special Issue Hydrogel for Tissue Regeneration (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>Schematic overview of hydrogel fabrication and functional characterization. The top panel illustrates the process of hydrogel synthesis, combining bioprinting technology and crosslinking to produce stable constructs. The bottom panel summarizes the key methods used to evaluate hydrogel performance, including swelling behavior, mechanical properties, and degradation over time.</p>
Full article ">Figure 2
<p>Percent swelling behavior of hydrogel samples in response to varying concentrations of alginate and PVA. (<b>A</b>) Box-and-whisker plots showing percent swelling across different alginate concentrations. The boxes represent the interquartile range (IQR), the lines indicate the median, whiskers extend to the data range, and individual data points displayed as circles are outliers. (<b>B</b>) Box-and-whisker plots showing percent swelling across different PVA concentrations, with statistical significance indicated between specific pairs of PVA concentrations. (<b>C</b>) Three-dimensional scatter plot illustrating the combined effect of alginate and PVA concentrations on percent swelling, providing an integrated view of their joint impact on swelling behavior. Asterisks denote statistical significance: * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Percent swelling of hydrogel samples at varying concentrations of PVA with fixed alginate concentrations. Bar graphs represent mean percent swelling at different PVA concentrations for each specified alginate concentration. Error bars indicate the standard error of the mean (SEM). Individual data points are overlaid as navy circles, showing variability within each concentration group. Statistical significance between selected PVA concentration pairs is indicated by asterisks above the bars, with the following notation: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, ANOVA F-statistic and <span class="html-italic">p</span>-value for each alginate concentration level are included in the title of each panel, highlighting significant differences in percent swelling across PVA concentrations within each fixed alginate concentration level.</p>
Full article ">Figure 4
<p>Heatmap displaying the 48-h percent swelling ratio of hydrogel samples across varying concentrations of high-molecular-weight PVA and medium-viscosity alginate. The color intensity corresponds to the swelling ratio, with darker blue shades representing higher swelling percentages. Each cell is annotated with the mean swelling ratio (%) for the corresponding combination of PVA and alginate concentrations. This visualization highlights the inverse relationship between PVA concentration and swelling, as higher PVA levels generally correspond to lower swelling ratios, particularly at lower alginate concentrations.</p>
Full article ">Figure 5
<p>Percent degradation of constructs after 28 days in culture in response to varying concentrations of alginate and PVA. Box-and-whisker plots show percent degradation across alginate concentrations (<b>A</b>) and PVA concentrations (<b>B</b>), with boxes representing the interquartile range (IQR), and the lines indicating the median. Whiskers extend to the data range, and individual data points displayed as circles are outliers. A 3D scatter plot (<b>C</b>) illustrates the combined effects of alginate and PVA concentrations on percent degradation, offering an integrated view of their joint impact on material degradation. Asterisks denote statistical significance: * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Percent degradation of hydrogel samples after 28 days, shown across varying PVA concentrations with fixed alginate concentrations. Each bar represents the mean degradation percentage for a given PVA concentration at a fixed alginate level. Error bars represent the standard error of the mean (SEM), and individual data points are displayed as circles, providing insights into data spread and individual variation. Statistical significance between selected PVA concentration pairs is indicated by asterisks above the bars, with * <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. ANOVA F-statistic and <span class="html-italic">p</span>-value are included in each panel title, highlighting significant differences in degradation across PVA concentrations within each fixed alginate concentration level.</p>
Full article ">Figure 7
<p>Heatmap displaying the percent degradation of hydrogel samples after 28 days across varying concentrations of high-molecular-weight PVA and medium-viscosity alginate. The color intensity corresponds to the degradation percentage, with darker red shades representing higher degradation levels. Each cell is annotated with the mean degradation percentage (%) for the respective combination of PVA and alginate concentrations.</p>
Full article ">Figure 8
<p>Elastic modulus behavior of hydrogel samples in response to varying alginate and PVA concentrations. (<b>A</b>) Box-and-whisker plots of modulus values across alginate concentrations, with boxes representing the interquartile range (IQR), the lines indicating the median, and whiskers extending to the data range, with outliers represented as individual data points. (<b>B</b>) Box-and-whisker plots of modulus values across different PVA concentrations, with significant differences between specific PVA concentrations. (<b>C</b>) Three-dimensional scatter plot illustrating the combined effect of alginate and PVA concentrations on modulus, offering an integrated view of their joint impact on mechanical properties. Asterisks denote statistical significance: * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 9
<p>Heatmap of elastic modulus by PVA and alginate concentrations. This heatmap illustrates the elastic modulus (MPa) of constructs at varying concentrations of PVA and alginate, as determined by compression testing. Each cell represents the mean elastic modulus for a specific combination of PVA and alginate concentrations, with values shown to three decimal places. Darker shades of purple indicate higher elastic modulus values, signifying stronger constructs.</p>
Full article ">Figure 10
<p>Schematic representation of the bioprinting process for alginate–PVA hydrogel constructs. From left to right: (1) Three-dimensional model of the cylindrical construct designed for bioprinting; (2) slicing of the 3D model to create a 50% gyroid infill pattern; (3) extrusion of the hydrogel bioink in the specified design; (4) final printed hydrogel construct after crosslinking. Scale bar represents 1 mm.</p>
Full article ">Figure 11
<p>Uniaxial unconfined compression testing setup. A cylindrical 3D bioprinted hydrogel sample is positioned between stainless-steel compression platens in the mechanical testing machine. The close-up inset shows the hydrogel sample’s gyroid infill structure with a scale for size reference. True measurements of the hydrogels were performed using digital calipers prior to compression testing.</p>
Full article ">
20 pages, 3659 KiB  
Article
Exploring the Osteoinductive Potential of Bacterial Pyomelanin Derived from Pseudomonas aeruginosa in a Human Osteoblast Model
by Mateusz M. Urbaniak, Karolina Rudnicka, Przemysław Płociński and Magdalena Chmiela
Int. J. Mol. Sci. 2024, 25(24), 13406; https://doi.org/10.3390/ijms252413406 - 14 Dec 2024
Viewed by 343
Abstract
Alkaptonuria (AKU) is a genetically determined disease associated with disorders of tyrosine metabolism. In AKU, the deposition of homogentisic acid polymers contributes to the pathological ossification of cartilage tissue. The controlled use of biomimetics similar to deposits observed in cartilage during AKU potentially [...] Read more.
Alkaptonuria (AKU) is a genetically determined disease associated with disorders of tyrosine metabolism. In AKU, the deposition of homogentisic acid polymers contributes to the pathological ossification of cartilage tissue. The controlled use of biomimetics similar to deposits observed in cartilage during AKU potentially may serve the development of new bone regeneration therapy based on the activation of osteoblasts. The proposed biomimetic is pyomelanin (PyoM), a polymeric biomacromolecule synthesized by Pseudomonas aeruginosa. This work presents comprehensive data on the osteoinductive, pro-regenerative, and antibacterial properties, as well as the cytocompatibility, of water-soluble (PyoMsol) or water-insoluble (PyoMinsol) PyoM. Both variants of PyoM support osteoinductive processes as well as the maturation of osteoblasts in cell cultures in vitro due to the upregulation of bone-formation markers, osteocalcin (OC), and alkaline phosphatase (ALP). Furthermore, the cytokines involved in these processes were elevated in cell cultures of osteoblasts exposed to PyoM: tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-10. The PyoM variants are cytocompatible in a wide concentration range and limit the doxorubicin-induced apoptosis of osteoblasts. This cytoprotective PyoM activity is correlated with an increased migration of osteoblasts. Moreover, PyoMsol and PyoMinsol exhibit antibacterial activity against staphylococci isolated from infected bones. The osteoinductive, pro-regenerative, and antiapoptotic effects achieved through PyoM stimulation prompt the development of new biocomposites modified with this bacterial biopolymer for medical use. Full article
(This article belongs to the Section Macromolecules)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) The percentage of viable hFOB 1.19 osteoblasts after 24 h exposure to different concentrations of the water-soluble (PyoM<sub>sol</sub>) or water-insoluble pyomelanin (PyoM<sub>insol</sub>). Osteoblasts incubated only in medium (NC = 100% cell viability). (<b>B</b>) Diminishing cell apoptosis induced by doxorubicin (DOX) in the milieu of PyoM<sub>sol</sub> or PyoM<sub>insol</sub> at the concentration of 1 µg/mL. The Apoptotic Index was determined from the relative fluorescence units (RFUs) of cells exposed to PyoM vs. RFUs of non-stimulated osteoblasts (NS). The mean ± standard deviation results of four separate experiments are shown. Statistical significance for ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>(<b>A</b>) The migration effectiveness of hFOB 1.19 osteoblasts determined in a scratch assay. Osteoblasts were cultivated for 24 h, 48 h, or 72 h in the presence of water-soluble (PyoM<sub>sol</sub>) or water-insoluble pyomelanin (PyoM<sub>insol</sub>) at a concentration of 1 µg/mL Mean ± standard deviation of five separate experiments are shown. * <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, statistically significant differences. Cells not stimulated with PyoM (NS). The reference wound closure of non-stimulated cells is marked on the graph with dashed green line. (<b>B</b>) Representative images of hFOB 1.19 osteoblast migration after 24, 48, and 72 h of water-soluble (PyoM<sub>sol</sub>) or water-insoluble pyomelanin (PyoM<sub>insol</sub>) stimulation. In the microscopic images, the red dashed lines indicate the width of the overgrown crack.</p>
Full article ">Figure 3
<p>Transcriptomic changes in hFOB 1.19 osteoblasts upon treatment with soluble pyomelanin (PyoM<sub>sol</sub>). (<b>A</b>) Ontology analysis of differentially expressed genes overexpressed in hFOB1.19 osteoblasts during differentiation in osteoinductive media utilizing the ShinyGo 0.77 online platform [<a href="#B30-ijms-25-13406" class="html-bibr">30</a>]. (<b>B</b>) List of transcripts relevant to osteoblastic differentiation of hFOB cells in comparison between proliferative and differentiation conditions for PyoM<sub>sol</sub>-treated and non-treated cells. (<b>C</b>) A summary of transcriptomic changes observed between hFOB 1.19 cells differentiating under standard conditions versus cells differentiating in media supplemented with PyoM<sub>sol</sub>. RNA was isolated from cells incubated in proliferative or differentiation conditions for 14 days. The change in BMP-2 expression between non-treated and treated cells (marked in red) exceeded the Log 2 FC of 2, preset as the threshold for our analysis. ALPL, liver-/bone-/kidney-specific or tissue-nonspecific (TNSALP) ALP form, ALPP, placental ALP form; BMP, bone morphogenic protein; CGMP-PKG, cyclic guanosine monophosphate protein kinase G; COL, collagen; OCN, osteocalcin; OPG, osteoprotegrin, RUNX, runt-related transcription factors; TGF-Beta, transforming growth factor beta; PI3-Akt, PI3-Akt; phosphatidylinositol 3-kinase, serine/threonine kinase (protein kinase B); Rap1, Ras-proximate-1 or Ras-related protein-1.</p>
Full article ">Figure 4
<p>The influence of pyomelanin (PyoM) on cell growth, alkaline phosphatase production, and bone cell calcification. (<b>A</b>) Number of hFOB 1.19 osteoblasts after 1, 7, 14, 21, or 28 days of incubation with water-soluble pyomelanin (PyoM<sub>sol</sub>), water-insoluble pyomelanin (PyoM<sub>insol</sub>), or culture medium alone, i.e., unstimulated cells (NS). (<b>B</b>) The activity of alkaline phosphatase (ALP) produced by hFOB 1.19 osteoblasts in cell cultures exposed to PyoM<sub>sol</sub>, PyoM<sub>insol</sub>, or culture medium alone (NS) after 7, 14, 21, or 28 days are shown in international units (IU). Results are shown as mean ± standard deviation. The experiment was performed four times. Statistical significance is indicated by * at <span class="html-italic">p</span> &lt; 0.05. (<b>C</b>) Representative images of calcification process in cell culture of osteoblasts exposed for 24 days to PyoM<sub>sol</sub> or PyoM<sub>insol</sub> or not stimulated (NS). The cells were stained with 4% alizarin The stained mineralized extracellular matrix of osteoblasts was observed under an inverted-phase contrast microscope. Calcium deposits were assessed quantitatively on the basis of absorbance values at 405 nm using a standard curve developed with hydroxyapatite.</p>
Full article ">Figure 5
<p>Secretion of osteocalcin and cytokines by osteoblasts exposed to PyoM. (<b>A</b>) The levels of osteocalcin (OC), (<b>B</b>) the levels of interleukin (IL)-6, (<b>C</b>) the levels of IL-10, and (<b>D</b>) the levels of tumor necrosis factor (TNF)-α in cell cultures of hFOB 1.19 exposed to water-soluble pyomelanin (PyoM<sub>sol</sub>), water-insoluble pyomelanin (PyoM<sub>insol</sub>), or culture medium alone, i.e., non-stimulated cells (NS), after 1, 4, 7, 11, 14, 18, 21, 25, and 28 days. Results are shown as mean ± standard deviation. The experiment was performed four times. * <span class="html-italic">p</span> &lt; 0.05 indicates statistically significant differences.</p>
Full article ">Figure 6
<p>Antibacterial activity of studied PyoM variants. The figure shows dose–response curves supplemented with the minimum inhibitory concentration (MIC)<sub>50</sub> determined for (<b>1</b>) water-soluble piomelanin (PyoM<sub>sol</sub>) or (<b>2</b>) water-insoluble pyomelanin (PyoM<sub>insol</sub>). <span class="html-italic">Staphylococcus</span> strains: (<b>A</b>) reference <span class="html-italic">S. aureus</span> ATTC 29213, (<b>B</b>) clinical <span class="html-italic">S. aureus</span> strain resistant to methicillin (MRSA), and (<b>C</b>) <span class="html-italic">S. felis</span>. Results are shown as mean ± standard deviation. The experiment was performed five times. The green dashed line shows the 50% bacterial viability.</p>
Full article ">
16 pages, 7709 KiB  
Article
Blocking the Sphingosine-1-Phosphate Receptor 2 (S1P2) Reduces the Severity of Collagen-Induced Arthritis in DBA-1J Mice
by Ju-Hyun Lee, Jung-Eun Lee and Dong-Soon Im
Int. J. Mol. Sci. 2024, 25(24), 13393; https://doi.org/10.3390/ijms252413393 - 13 Dec 2024
Viewed by 250
Abstract
The amount of sphingosine 1-phosphate (S1P) found in the synovial tissue of individuals with rheumatoid arthritis is five times greater than that in those with osteoarthritis. Our study aims to determine whether inhibiting S1P2 can mitigate collagen-induced rheumatoid arthritis (CIA) by using [...] Read more.
The amount of sphingosine 1-phosphate (S1P) found in the synovial tissue of individuals with rheumatoid arthritis is five times greater than that in those with osteoarthritis. Our study aims to determine whether inhibiting S1P2 can mitigate collagen-induced rheumatoid arthritis (CIA) by using an S1P2 antagonist, JTE-013, alongside DBA-1J S1pr2 wild-type (WT) and knock-out (KO) mice. CIA causes increases in arthritis scores, foot swelling, synovial hyperplasia, pannus formation, proteoglycan depletion, cartilage damage, and bone erosion, but these effects are markedly reduced when JTE-013 is administered to S1pr2 WT mice. CIA also elevates mRNA expression levels of pro-inflammatory Th1/Th17 cytokines in the foot and spleen, which are significantly decreased by JTE-013 in S1pr2 WT mice. Additionally, CIA raises Th1/Th17 and Treg cell counts, while JTE-013 reduces these elevations in the spleens of S1pr2 WT mice. Treatment with JTE-013 or the absence of S1pr2 curtails the differentiation of naïve T cells into Th1 and Th17 cells in a dose-dependent manner. In SW982 human synovial cells, JTE-013 lowers LPS-induced increases in pro-inflammatory cytokine levels. Overall, these findings propose that blocking S1P2 in immune and synovial cells may alleviate rheumatoid arthritis symptoms and offer a potential therapeutic approach. Full article
(This article belongs to the Special Issue Osteoarthritis: From Pathogenesis to Treatment)
Show Figures

Figure 1

Figure 1
<p>JTE-013 reduced the CIA-induced increase in arthritis score and paw thickness in <span class="html-italic">S1pr2</span> DBA-1J WT mice. Male <span class="html-italic">S1pr2</span> WT and KO DBA-1J mice, each aged 7 to 9 weeks, were injected via the tails with 100 μL of complete Freund’s adjuvant (CFA) emulsion on day 0. A booster injection of incomplete Freund’s adjuvant (IFA) was given on day 21. JTE-013, at a dose of 3 mg/kg body weight, was administered via intraperitoneal injection 30 min prior to the emulsion injection. The treatment with JTE-013 began on day 21 and continued through day 41. (<b>A</b>) Arthritis scores from day 21 to day 41 in <span class="html-italic">S1pr2</span> WT and KO mice. The mice were assessed for arthritis severity every other day using a specific scoring system. The criteria for scoring were: 0 for no arthritis symptoms, 1 for swelling and/or redness of one paw or digit, 2 for involvement of two joints, 3 for more than two joints being affected, and 4 for severe arthritis impacting the entire paw and digits. Each mouse’s arthritis index score was calculated by summing the scores from all individual paws. (<b>B</b>) Final paw thickness on day 42 in <span class="html-italic">S1pr2</span> WT and KO mice. (<b>C</b>) Representative foot images from <span class="html-italic">S1pr2</span> WT and KO mice. Data are presented as mean± SEM (n = 8). *** <span class="html-italic">p</span> &lt; 0.001, * <span class="html-italic">p</span> &lt; 0.05 compared to the control group, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared to the CIA group, <sup><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.001 compared to the CIA group in <span class="html-italic">S1pr2</span> WT mice.</p>
Full article ">Figure 2
<p>JTE-013 diminished CIA-induced arthritic changes in <span class="html-italic">S1pr2</span> WT mice. (<b>A</b>) H&amp;E staining (100×) of the ankle joint in <span class="html-italic">S1pr2</span> WT and KO mice. (<b>B</b>) Safranin O (100×) of the ankle joint in <span class="html-italic">S1pr2</span> WT and KO mice. Histologic scores based on (<b>C</b>) inflammation, (<b>D</b>) bone erosion, (<b>E</b>) cartilage damage, and (<b>F</b>) proteoglycan loss in <span class="html-italic">S1pr2</span> WT and KO mice. Data are shown as mean ± SEM (<span class="html-italic">n</span> = 8). *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01 compared to the control group, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared to the CIA group, <sup><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.001 compared to the CIA group in <span class="html-italic">S1pr2</span> WT mice.</p>
Full article ">Figure 3
<p>JTE-013 reduced the CIA-induced elevation of inflammatory cytokine levels in the foot of <span class="html-italic">S1pr2</span> WT mice. Cytokine mRNA levels were normalized to <span class="html-italic">Gapdh</span> mRNA level. (<b>A</b>) <span class="html-italic">Il-1b</span>, (<b>B</b>) <span class="html-italic">Tnf-a</span>, (<b>C</b>) <span class="html-italic">Il-6</span>, (<b>D</b>) <span class="html-italic">Il-8</span>, (<b>E</b>) <span class="html-italic">Ifn-g</span>, (<b>F</b>) <span class="html-italic">Il-17a</span>, (<b>G</b>) <span class="html-italic">Rankl</span>, and (<b>H</b>) <span class="html-italic">Mmp-3</span> in <span class="html-italic">S1pr2</span> WT and KO mice. Significance levels: *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01 compared to the control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the CIA group; <sup><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.001, <sup><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01 compared to the CIA group in <span class="html-italic">S1pr2</span> WT mice.</p>
Full article ">Figure 4
<p>JTE-013 effectively lowered the CIA-induced elevations in serum IgG1 and IgG2a levels in <span class="html-italic">S1pr2</span> WT mice. Blood samples were obtained on day 42, and the serum levels of IgG1 (<b>A</b>) and IgG2a (<b>B</b>) were measured using ELISA in both S1pr2 WT and KO mice. The data are presented as mean ± SEM (n = 8). Significance levels are indicated as *** <span class="html-italic">p</span> &lt; 0.001 compared to the control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the CIA group; <sup><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.001 compared to the CIA group in <span class="html-italic">S1pr2</span> WT mice.</p>
Full article ">Figure 5
<p>JTE-013 reduced CIA-induced spleen enlargement in <span class="html-italic">S1pr2</span> WT mice. (<b>A</b>) Images of spleens. (<b>B</b>) Spleen weights in <span class="html-italic">S1pr2</span> WT and KO mice. Results are shown as the mean ± SEM (<span class="html-italic">n</span> = 8). *** <span class="html-italic">p</span> &lt; 0.001 compared to the control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared to the CIA group; <sup><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.001 compared to the CIA group in <span class="html-italic">S1pr2</span> WT mice.</p>
Full article ">Figure 6
<p>JTE-013 reduced CIA-induced increases in inflammatory cytokine levels in the spleens of <span class="html-italic">S1pr2</span> WT mice. Cytokine mRNA levels were quantified as ratios to <span class="html-italic">Gapdh</span> mRNA. (<b>A</b>) <span class="html-italic">Il-1b</span>, (<b>B</b>) <span class="html-italic">Tnf-a</span>, (<b>C</b>) <span class="html-italic">Il-6</span>, (<b>D</b>) <span class="html-italic">Il-8</span>, (<b>E</b>) <span class="html-italic">Ifn-g</span>, and (<b>F</b>) <span class="html-italic">Il-17a</span> in <span class="html-italic">S1pr2</span> WT and KO mice. Significance: *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 compared to the control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the CIA group; <sup><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.001, <sup><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01, <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05 compared to the CIA group in <span class="html-italic">S1pr2</span> WT mice.</p>
Full article ">Figure 7
<p>JTE-013 reduced CIA-induced increases in the populations of Th1, Th17, and Treg cells in <span class="html-italic">S1pr2</span> WT mice. Representative flow cytometry results of CD4<sup>+</sup>T-bet<sup>+</sup> Th1 cells (<b>A</b>), CD4<sup>+</sup>T RORγt<sup>+</sup> Th17 cells (<b>B</b>), and CD4<sup>+</sup>T FoxP3<sup>+</sup> Treg cells (<b>C</b>) in <span class="html-italic">S1pr2</span> WT and KO mice. (<b>D</b>) Percentage of CD4<sup>+</sup>T-bet<sup>+</sup> Th1 cells in <span class="html-italic">S1pr2</span> WT and KO mice. (<b>E</b>) Percentage of CD4<sup>+</sup>T RORγt<sup>+</sup> Th17 cells in <span class="html-italic">S1pr2</span> WT and KO mice. (<b>F</b>) Percentage of CD4<sup>+</sup>T FoxP3<sup>+</sup> Treg cells in <span class="html-italic">S1pr2</span> WT and KO mice. Significance: *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 compared to the control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the CIA group; <sup><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.001, <sup><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01 compared to the CIA group in <span class="html-italic">S1pr2</span> WT mice.</p>
Full article ">Figure 8
<p>Suppressive effect of JTE-013 on T cell differentiation into Th1 and Th17 cells. CD4<sup>+</sup> T cells, isolated from splenocytes, were cultured in media for Th1 or Th17 cell differentiation for 5 days in plates pre-coated with an antibody to mouse CD3. Representative flow cytometry results for CD4<sup>+</sup>IFN-γ<sup>+</sup> Th1 cell differentiation (<b>A</b>) and CD4<sup>+</sup>IL-17A<sup>+</sup> Th17 cell differentiation (<b>B</b>). Histograms show the percentage of CD4<sup>+</sup>IFN-γ<sup>+</sup> cells (<b>C</b>) and CD4<sup>+</sup>IL-17A<sup>+</sup> cells (<b>D</b>) (<span class="html-italic">n</span> = 5). Significance: *** <span class="html-italic">p</span> &lt; 0.001 compared to the control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the CIA group; <sup><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.001 compared to the CIA group in <span class="html-italic">S1pr2</span> WT mice.</p>
Full article ">Figure 9
<p>JTE-013 inhibited LPS-induced increases in pro-inflammatory cytokine mRNA expression in SW982 cells. SW982 cells were seeded at 1 × 10<sup>5</sup>/mL. After 24 h, JTE-013 (10, 15, 30 mM) was added, followed by LPS (100 ng/mL) incubation for 30 min. qPCR confirmed mRNA expression levels of inflammatory cytokines in SW982 cells. (<b>A</b>) IL-1β, (<b>B</b>) TNF-α, (<b>C</b>) IL-6, (<b>D</b>) IL-8, (<b>E</b>) IFN-g, (<b>F</b>) IL-17A, (<b>G</b>) RANKL, and (<b>H</b>) MMP-3. Results are shown as mean ± SEM (<span class="html-italic">n</span> = 5). *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01 vs. the control group. <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 vs. the LPS-induced group.</p>
Full article ">
12 pages, 3011 KiB  
Article
Chondroprotective Effect of Campylaephora hypnaeoides Extract in Primary Chondrocytes and Rat OA Model
by Ji Yun Jang, Seul Ah Lee, Do Kyung Kim, Sook-Young Lee and Chun Sung Kim
Int. J. Mol. Sci. 2024, 25(24), 13391; https://doi.org/10.3390/ijms252413391 - 13 Dec 2024
Viewed by 281
Abstract
Campylaephora hypnaeoides (C. hypnaeoides) was extracted using fermented ethanol. The effect of fermented ethanol extract of C. hypnaeoides (FeCH) on chondrocyte viability was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-iphenyltetrazolium bromide assay, which showed no cytotoxicity at 2 mg/mL. FeCH pretreatment in IL-1β-stimulated chondrocytes significantly [...] Read more.
Campylaephora hypnaeoides (C. hypnaeoides) was extracted using fermented ethanol. The effect of fermented ethanol extract of C. hypnaeoides (FeCH) on chondrocyte viability was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-iphenyltetrazolium bromide assay, which showed no cytotoxicity at 2 mg/mL. FeCH pretreatment in IL-1β-stimulated chondrocytes significantly inhibited the accumulation of nitric oxide and prostaglandin E2, which was analyzed using the ELISA assay. In addition, protein expression levels of inflammatory-related factors, such as inducible nitric oxide synthase, cyclooxygenase-2, interleukin-6, tumor necrosis factor-alpha, and cartilage-degrading-related enzymes, such as matrix metalloproteinases-1, -3, and -13, and a disintegrin and metalloproteinase with thrombospondin motifs-4 and -5 were significantly decreased in IL-1β-stimulated chondrocytes pretreated with FeCH, which were analyzed using western blot analysis. In addition, as a result of analyzing the content of collagen type II (Col II) and proteoglycan through western blot analysis and alcian blue staining, FeCH pretreatment prevented the degradation of Col II and proteoglycan. It was analyzed through western blot analysis that the chondroprotective effect of FeCH may be mediated through MAPKs and NF-κB-signaling mechanisms. In an in vivo study, an osteoarthritis experimental animal model with damaged medial meniscus (DMM) was utilized and orally administered daily for 8 weeks after surgery. At the study end point, knee joints were harvested and subjected to histological analysis with safranin O staining. As a result, articular cartilage was significantly protected in the FeCH group compared to the DMM group. These results suggest FeCH as a candidate material for the development of pharmaceutical materials for the treatment or prevention of degenerative arthritis. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
Show Figures

Figure 1

Figure 1
<p>Effects of fermentation ethanol of <span class="html-italic">Campylaephora hypnaeoides</span> (FeCH) on primary rat chondrocyte viability. (<b>A</b>) primary rat chondrocytes were treated with FeCH (0.25, 0.5, 1, and 2 mg/mL) for 24 h, and viability was determined by MTT assay. (<b>B</b>) ICC staining of chondrocytes using collagen type II antibody after 3 days of culture (×40). Data are represented as mean ± SD of three independent experiments.</p>
Full article ">Figure 2
<p>Inhibitory effects of FeCH on IL-1β-induced nitrite, PGE<sub>2</sub>, TNF-α, IL-6, and COX-2 in primary rat chondrocytes. Primary rat chondrocytes were pretreated with FeCH (0.125, 0.25, and 0.5 mg/mL) for 1 h, followed by IL-1β (5 ng/mL) for 24 h. Nitrite production (<b>A</b>), PGE₂ production (<b>B</b>), and TNF-α production (<b>C</b>) in the cell culture medium were determined by ELISA kit. (<b>D</b>) Expression of iNOS, COX-2, TNF-α, IL-6, and COX-1 expression was determined by western blot. (<b>E</b>) Quantitative data of (<b>D</b>) were analyzed using Image J software (version 8). Data are represented as mean ± SD of three independent experiments. α-tubulin served as an internal control. ## <span class="html-italic">p</span> &lt; 0.01 vs. control; * <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 vs. IL-1β-treated group.</p>
Full article ">Figure 3
<p>Inhibitory effects of FeCH on IL-1β-induced ADAMTS-5 and -4 and MMP-1, -3, and -13 in primary rat chondrocytes. Primary rat chondrocytes were pretreated with FeCH (0.125, 0.25, and 0.5 mg/mL) for 1 h, followed by IL-1β (5 ng/mL) for 24 h. (<b>A</b>) Protein levels of matrix-degrading enzymes (ADAMTS-5 and -4 and MMP-1, -3, and -13) was determined by western blot. (<b>B</b>) Quantitative data of (<b>A</b>) were analyzed using Image J software. Data are represented as mean ± SD of three independent experiments. α-tubulin served as an internal control. ## <span class="html-italic">p</span> &lt; 0.01 vs. control; ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. IL-1β-treated group.</p>
Full article ">Figure 4
<p>Effect of FeCH on collagen type II and proteoglycan in IL-1β-stimulated primary rat chondrocytes. Primary rat chondrocytes were pretreated with FeCH (0.125, 0.25, and 0.5 mg/mL) for 1 h, followed by IL-1β (5 ng/mL) for 24 h. (<b>A</b>) Protein levels of collagen type II degradation were determined by western blot. (<b>B</b>) Quantitative data of (<b>A</b>) were analyzed using Image J software. (<b>C</b>) Proteoglycan degradation was determined by alcian blue stain. (<b>D</b>) After dissolving alcian blue dye in 6M guanidine-HCL, the absorbance at 620 nm was measured. Data are represented as mean ± SD of three independent experiments. α-tubulin served as an internal control. <span class="html-italic">## p</span> &lt; 0.01 vs. control; <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 vs. IL-1β-treated group.</p>
Full article ">Figure 5
<p>Effects of FeCH on phosphorylation of MAPKs and activation of NF-κB in IL-1β-stimulated primary rat chondrocytes. Primary rat chondrocytes were pretreated with FeCH (0.125, 0.25, and 0.5 mg/mL) for 1 h, followed by IL-1β (5 ng/mL) for 24 h. (<b>A</b>) Protein levels of phosphorylation of MAPKs were determined by western blot. (<b>B</b>) Quantitative data of (<b>A</b>) were analyzed using Image J software. (<b>C</b>) Phosphorylation levels of NF-κB p65 were determined by western blot. (<b>D</b>) Quantitative data of (<b>C</b>) were analyzed using Image J software. Data are represented as mean ± SD of three independent experiments. α-tubulin and Lamin B1 served as internal controls. <span class="html-italic">## p</span> &lt; 0.01 vs. control; <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 vs. IL-1β-treated group.</p>
Full article ">Figure 6
<p>Histological evaluation of cartilage-protective effect of FeCH against cartilage degradation in a DMM model. (<b>A</b>) In vivo schemic (×100). White box indicates the enlarged area. (<b>B</b>) Rats underwent surgical destabilization of the medial meniscus (DMM). The day after the DMM surgery, rats were orally administered with FeCH (25 and 50 mg/kg) or celecoxib (2 mg/kg) daily for 8 weeks. Histological analysis of cartilage destruction was evaluated by safranin O/fast green staining.</p>
Full article ">
20 pages, 1333 KiB  
Review
Innovative Biotherapies and Nanotechnology in Osteoarthritis: Advancements in Inflammation Control and Cartilage Regeneration
by Binhan Liu, Tao Liu, Yanhong Li and Chunyu Tan
Int. J. Mol. Sci. 2024, 25(24), 13384; https://doi.org/10.3390/ijms252413384 - 13 Dec 2024
Viewed by 309
Abstract
Osteoarthritis (OA) is among the most prevalent degenerative joint disorders worldwide, particularly affecting the aging population and imposing significant disability and economic burdens. The disease is characterized by progressive degradation of articular cartilage and chronic inflammation, with no effective long-term treatments currently available [...] Read more.
Osteoarthritis (OA) is among the most prevalent degenerative joint disorders worldwide, particularly affecting the aging population and imposing significant disability and economic burdens. The disease is characterized by progressive degradation of articular cartilage and chronic inflammation, with no effective long-term treatments currently available to address the underlying causes of its progression. Conventional therapies primarily manage symptoms such as pain and inflammation but fail to repair damaged tissues. Emerging biotherapies and regenerative medicine approaches offer promising alternatives by addressing cartilage repair and inflammation control at the molecular level. This review explores the recent advancements in biotherapeutic strategies, including mesenchymal stem cell (MSC) therapy, growth factors, and tissue engineering, which hold the potential for promoting cartilage regeneration and modulating the inflammatory microenvironment. Additionally, the integration of nanotechnology has opened new avenues for targeted drug delivery systems and the development of innovative nanomaterials that can further enhance the efficacy of biotherapies by precisely targeting inflammation and cartilage damage. This article concludes by discussing the current clinical applications, the ongoing clinical trials, and the future research directions necessary to confirm the safety and efficacy of these advanced therapies for OA management. With these advancements, biotherapies combined with nanotechnology may revolutionize the future of OA treatment by offering precise and effective solutions for long-term disease management and improved patient outcomes. Full article
(This article belongs to the Section Molecular Nanoscience)
Show Figures

Figure 1

Figure 1
<p>Clinical manifestations of OA.</p>
Full article ">Figure 2
<p>Biological treatment of OA. Biological therapy for OA refers to the use of biologics or bioengineering methods to treat OA. These therapies can include biological products such as biological agents, growth factors, stem cell therapy, gene therapy, and others.</p>
Full article ">Figure 3
<p>Application of nanotechnology in OA.</p>
Full article ">
7 pages, 1646 KiB  
Case Report
Osteochondral Allograft Transplant in a Young Patient with a Traumatic Hip Fracture Dislocation: A Case Report
by Morgan Turnow, Trent Davis, Thomas Seebacher, Grant Chudik, Taylor Manes, Hunter Pharis, Daniel Degenova and Sanjay Mehta
Surg. Tech. Dev. 2024, 13(4), 402-408; https://doi.org/10.3390/std13040032 - 13 Dec 2024
Viewed by 272
Abstract
Background: Femoral head fractures with osteochondral defects are rare injuries often resulting from traumatic hip dislocations. These injuries create a significant risk for post-traumatic osteoarthritis. Various surgical methods for repair have been utilized to restore these osteochondral defects, including mosaicplasty, autologous cartilage implantation, [...] Read more.
Background: Femoral head fractures with osteochondral defects are rare injuries often resulting from traumatic hip dislocations. These injuries create a significant risk for post-traumatic osteoarthritis. Various surgical methods for repair have been utilized to restore these osteochondral defects, including mosaicplasty, autologous cartilage implantation, osteochondral allograft transplant (OAT), and demineralized bone matrix (DBM). Methods: We present a case of a 21-year-old male who sustained a fracture-dislocation of the left femoral head with impaction of the weight-bearing surface due to a motor vehicle collision. Due to the patient’s relatively young age, OAT plugs from a fresh-frozen proximal humerus with DBM supplementation during fracture fragment fixation were chosen to reduce the likelihood of post-traumatic arthritis. Results: The patient regained subjective function and full strength on exam with no pain at 2 years postoperatively. Conclusions: We propose that a proximal humerus allograft is a suitable alternative in an urgent setting when a femoral head allograft is not available. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Anterior-to-posterior radiograph of the pelvis demonstrating left posterior hip dislocation with impaction of the femoral head into the posterior wall. (<b>B</b>) Anterior-to-posterior radiograph of the pelvis post-reduction film demonstrating the suspected left femoral head and/or a posterior wall of the acetabulum fracture.</p>
Full article ">Figure 2
<p>Intraoperative images of the large osteochondral defect and the definitive osteochondral allograft transplant. (<b>A</b>) The 5 × 4 cm impacted osteochondral lesion of the femoral head with a small surface of the weightbearing surface intact superior to the fovea. (<b>B</b>) Reduction and fixation of the inferior portion of the weight–bearing surface with two 2.5 mm headless compression screws. (<b>C</b>) Surface area demonstration of the proximal humerus osteochondral allograft (OAT) plug. (<b>D</b>) Placement of the 22 × 8 and 15 × 9 mm OAT plugs.</p>
Full article ">Figure 3
<p>(<b>A</b>) Anterior-to-posterior radiograph of the left hip after open reduction internal fixation of the left femoral head osteochondral lesion. (<b>B</b>) Anterior-to-posterior radiograph of the left hip at 2 weeks following open reduction internal fixation of the left femoral head osteochondral lesion. (<b>C</b>) Anterior-to-posterior radiograph of the left hip at 8 weeks following open reduction internal fixation of the left femoral head osteochondral lesion. (<b>D</b>) Anterior-to-posterior radiograph of the left hip at 2 years following open reduction internal fixation of the left femoral head osteochondral lesion.</p>
Full article ">
9 pages, 2780 KiB  
Article
Morphology of the Calcaneofibular Ligament Reflects Degeneration of the Talonavicular Articular Surface: A Cadaver Study
by Ryuta Tanaka, Daisuke Kiyoshima, Kaori Suyama, Ning Qu, Miyu Inagawa and Shogo Hayashi
J. Clin. Med. 2024, 13(24), 7565; https://doi.org/10.3390/jcm13247565 - 12 Dec 2024
Viewed by 402
Abstract
Background: Osteoarthritis is caused by damage to the articular cartilage due to bone-on-bone collisions and friction. The length, width, and thickness of the ligaments are expected to change in order to regulate excessive bone-to-bone movement. We aimed to clarify the relationship between ligament [...] Read more.
Background: Osteoarthritis is caused by damage to the articular cartilage due to bone-on-bone collisions and friction. The length, width, and thickness of the ligaments are expected to change in order to regulate excessive bone-to-bone movement. We aimed to clarify the relationship between ligament morphology and joint surface degeneration in the ankle joints using macroscopic observations and measurements. Methods: The participants were 50 feet of 45 Japanese cadavers. The lengths, widths, and thicknesses of the tibionavicular, tibiospring, tibiocalcaneal, posterior tibiotalar, anterior tibiotalar, and calcaneofibular ligaments (CFLs) were measured. The degeneration of the talonavicular joint surface was investigated macroscopically and classified into two groups: the Degeneration (+) group and Degeneration (−) group. Unpaired t-tests were performed for each measurement. Logistic regression analysis was performed on the significantly different items to obtain cutoff values, sensitivity, and specificity. Results: Only the width of the CFL differed significantly between the Degeneration (+) (20 feet) and Degeneration (−) groups (p < 0.001). In the logistic regression analysis, the width of the CFL had an R2 of 0.262, sensitivity of 75.0%, and specificity of 83.3%, with a cutoff value of 8.7 mm. Conclusions: A wide CFL indicates a high likelihood of talonavicular articular surface degeneration. Full article
(This article belongs to the Special Issue Clinical Advancements in Foot and Ankle Surgery)
Show Figures

Figure 1

Figure 1
<p>Measurements of each ankle ligament: (<b>a</b>) Deltoid ligaments measured in this study: 1. TNL, tibionavicular ligament, 2. TSL, tibiospring ligament, 3. TCL, tibiocalcaneal ligament, and 4. PTTL, superficial posterior tibiotalar ligament. (<b>b</b>) Lateral ligaments measured in this study were as follows: 5. ATFL, anterior talofibular ligament, and 6. CFL, calcaneofibular ligament.</p>
Full article ">Figure 2
<p>Cases of degeneration of the talocrural articular surface: (<b>a</b>) diagram showing the degeneration of the medial grade 4 articular surface; and (<b>b</b>) diagram of an ankle joint with grade 3 and 2 joint degeneration on the medial and lateral sides, respectively.</p>
Full article ">Figure 3
<p>Cases with wide and narrow CFL observed in this study: (<b>a</b>) CFL width with talar articular surface degeneration; and (<b>b</b>) CFL width in the absence of talar surface degeneration. CFL: calcaneofibular ligament.</p>
Full article ">Figure 4
<p>ROC curves for determining CFL width cutoff value, sensitivity, and specificity. The sensitivity and specificity were 75% and 83.3%, respectively, and the cutoff value for CFL width calculated from the ROC curve was 8.7 mm. CFL, calcaneofibular ligament; ROC, receiver operating characteristic.</p>
Full article ">Figure 5
<p>Method used to measure the width of the CFL using ultrasound imaging equipment: (<b>a</b>) after palpation, the CFL is extracted from the fibula to the talus in the long axial direction; (<b>b</b>) CFL width was measured by placing it in the short-axis direction at the midpoint; and (<b>c</b>) width measured in the short-axis image (6.7 mm). CFL: calcaneofibular ligament.</p>
Full article ">
18 pages, 9960 KiB  
Article
Comparative Analysis of Extracorporeal Shockwave Therapy, Bisphosphonate, and Wharton Jelly-Derived Mesenchymal Stem Cells in Preserving Bone and Cartilage Integrity and Modulating IL31, IL33, and BMP2 in the Cartilage of Ovariectomized Rat Model
by Jai-Hong Cheng, Cheng-Wei Chen, Wen-Yi Chou, Po-Cheng Chen, Kuan-Ting Wu, Shun-Wun Jhan, Shan-Ling Hsu, Yi-No Wu and Hou-Tsung Chen
Biomedicines 2024, 12(12), 2823; https://doi.org/10.3390/biomedicines12122823 - 12 Dec 2024
Viewed by 276
Abstract
Background: Osteoporosis (OP) is a chronic inflammatory bone disease characterized by reduced bone structure and strength, leading to increased fracture risk. Effective therapies targeting both bone and cartilage are limited. This study compared the therapeutic effects of extracorporeal shockwave therapy (ESWT), bisphosphonate (Aclasta), [...] Read more.
Background: Osteoporosis (OP) is a chronic inflammatory bone disease characterized by reduced bone structure and strength, leading to increased fracture risk. Effective therapies targeting both bone and cartilage are limited. This study compared the therapeutic effects of extracorporeal shockwave therapy (ESWT), bisphosphonate (Aclasta), and human Wharton jelly-derived mesenchymal stem cells (WJMSCs) in a rat model of OP. Methods: Female rats were assigned to four groups: Sham (no surgery or treatment), OP (bilateral ovariectomy, OVX), ESWT (OVX + ESWT on both tibias at 0.25 mJ/mm2, 1500 impulses per tibia), Aclasta (OVX + zoledronic acid 0.1 mg/kg via tail vein injection), and WJMSC (OVX + 2 × 10⁶ WJMSCs). Pathological changes, bone microarchitecture (by micro-CT), serum cytokines (by ELISA), and tissue-specific molecular markers (by immunohistochemistry) were evaluated. Results: All treatments improved bone density, preserved cartilage, and modulated cytokines (IL31, IL33, VEGF, and BMP2), with Aclasta showing the greatest improvements in bone parameters and cartilage preservation. ESWT and WJMSC also demonstrated significant effects, with ESWT highlighting non-invasive chondroprotective potential. Conclusions: Aclasta provided the best overall therapeutic response, particularly in bone regeneration. However, ESWT and WJMSC also showed comparable chondroprotective effects. ESWT emerges as a promising non-invasive alternative for OP management when pharmacological or cell-based therapies are not feasible. Full article
(This article belongs to the Special Issue Diseases and Regeneration for Muscle, Joint and Bone)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The study design and application of ESWT. (<b>A</b>) Flowchart displaying the experiments and timing for knee surgery, shockwave application, Aclasta treatment, WJMSC treatments, and sacrifice of the animals. Eight rats were used for each group. (<b>B</b>) Focused ESWT application (0.25 mJ/mm<sup>2</sup>, 4 Hz, 1500 impulses for each location) on right and left tibia at 0.5 cm below the skin of rats at each location, indicated as black circles. OP group = osteoporosis, OVX = ovariectomy, ESWT group = extracorporeal shockwave therapy, WJMSC group = Wharton Jelly-derived mesenchymal stem cell treatment, micro-CT = micro computed tomography, IHC = immunohistochemistry, ELISA = enzyme-linked immunosorbent assay.</p>
Full article ">Figure 2
<p>Micro-CT scan of proximal femur, tibia, and spine in different groups. (<b>A</b>) The results displayed photomicrographs of the knee in transverse (tibia, up) and sagittal (femur and tibia, below) views from micro-CT. (<b>B</b>) The photomicrographs of the spine (T6 to T8) in transverse and sagittal views. The region of the red line was the region of interest in vertebral bone (T7). F indicated the femur and T indicated the tibia. R was the right lower limb and L was the left lower limb. Eight rats were used for each group. OP group was osteoporosis, OVX was ovariectomy, ESWT group was extracorporeal shockwave therapy, and WJMSC group was Wharton jelly-derived mesenchymal stem cell treatment.</p>
Full article ">Figure 3
<p>The microphotographs of the left knees and spine. (<b>A</b>) The hematoxylin and eosin staining showed the cartilage change in the articular cartilage and epiphyseal plate of the left tibia (×100 magnification) in different groups. (<b>B</b>) Safranin-O staining showing the articular cartilage of the left knee (1000 μm) and epiphyseal plate of the left tibia (×100 magnification). (<b>C</b>) Safranin-O staining showing the spine (×50 magnification), vertebral bone (×100 magnification), and vertebral cartilage (×100 magnification). Eight rats were used for each group. The red arrowhead indicates cartilage. The black arrowhead indicated epiphyseal plate. The black triangle indicated cancellous bone. The red triangle indicated primary spongiosa. The tibia indicated by T and the femur indicated by F. Osteoporosis indicated by OP group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group.</p>
Full article ">Figure 4
<p>Immunohistochemical analysis for (<b>A</b>) IL31, (<b>B</b>) IL33, and (<b>C</b>) ST2 in the articular cartilage (×100 magnification) of the left knee and epiphyseal plate of the left tibia in Sham, OP, ESWT, Aclasta, and WJMSC groups (right). The expression levels are assessed following the treatments (left). *** <span class="html-italic">p</span> &lt; 0.001 as compared with the OP group. Eight rats were used for each group. Osteoporosis indicated OP group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group. T represented tibia. Red arrowhead indicated the expression of proteins.</p>
Full article ">Figure 4 Cont.
<p>Immunohistochemical analysis for (<b>A</b>) IL31, (<b>B</b>) IL33, and (<b>C</b>) ST2 in the articular cartilage (×100 magnification) of the left knee and epiphyseal plate of the left tibia in Sham, OP, ESWT, Aclasta, and WJMSC groups (right). The expression levels are assessed following the treatments (left). *** <span class="html-italic">p</span> &lt; 0.001 as compared with the OP group. Eight rats were used for each group. Osteoporosis indicated OP group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group. T represented tibia. Red arrowhead indicated the expression of proteins.</p>
Full article ">Figure 5
<p>Immunohistochemical analysis for BMP2 in the articular cartilage of the (<b>A</b>) tibia, (<b>B</b>) femur, and (<b>C</b>) epiphyseal plate of the left tibia in Sham, OP, SW, Aclasta, and WJMSC groups (×100 magnification) (right). The expression level was assessed following the treatments (left). *** <span class="html-italic">p</span> &lt; 0.001 as compared with the OP group. T represented tibia and Eight rats were used for each group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group. Red arrowhead indicated the expression of proteins.</p>
Full article ">Figure 6
<p>The immunohistochemical analysis of the vertebral cartilage in OVX rats. The immunohistochemical staining (×100 magnification, left panel) and the expression levels (right) of IL31, IL33, ST2, and BMP2 in the vertebral cartilage of the spines for each group. *** <span class="html-italic">p</span> &lt; 0.001 as compared with the OP group. Eight rats were used for each group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group. Red arrowhead indicated the expression of proteins.</p>
Full article ">
13 pages, 1822 KiB  
Article
Enhanced Pain Reduction at Different Stages of Knee Osteoarthritis via Repeated Injections of Hyaluronic Acid with Niacinamide: A Comparative Study
by Sophie Pennekamp, Stephan Hegelmaier, Wolfgang Hitzl, Markus Geßlein, Hermann Josef Bail, Kim Loose, Andreas Kopf, Niklas Engel, Johannes Rüther, Maximilian Willauschus and Michael Millrose
J. Clin. Med. 2024, 13(24), 7553; https://doi.org/10.3390/jcm13247553 - 12 Dec 2024
Viewed by 456
Abstract
Background: Osteoarthritis (OA) of the knee is the most common joint disease, characterized by the degeneration of joint cartilage. Intra-articular hyaluronic acid (IAHA) injections are a well-established non-surgical treatment. Methods: This retrospective study analyzed knee OA patients receiving IAHA combined with niacinamide injections, [...] Read more.
Background: Osteoarthritis (OA) of the knee is the most common joint disease, characterized by the degeneration of joint cartilage. Intra-articular hyaluronic acid (IAHA) injections are a well-established non-surgical treatment. Methods: This retrospective study analyzed knee OA patients receiving IAHA combined with niacinamide injections, assessing pain reduction in relation to patient data, the number of injections, and radiological findings. Results: IAHA injections led to significant pain reduction on the numeric rating scale (NRS) (0–10), with a mean decrease of 3.34 ± 1.65. Pain relief was greater with multiple injections. A comparison of subgroups by injection frequency (1, 2, or >2) showed significant pain reduction between 1 and 2 injections (p = 0.027) and between 1 and >2 injections (p = 0.032). The OA grade measured using the Kellgren–Lawrence (p = 0.95) and Vallotton MRI classifications (p = 0.50) did not correlate with pain reduction. However, patients with meniscal damage (p = 0.02) showed a greater benefit. A strong positive correlation was found between baseline pain intensity and pain reduction (p < 0.001; r = 0.61). Conclusions: IAHA with niacinamide significantly reduces knee OA pain, with more injections enhancing pain relief. Greater benefits were observed in patients with higher baseline pain and meniscal damage. The favorable safety profile and potential for repeated treatments make IAHA a valuable option in knee OA management. Full article
(This article belongs to the Section Orthopedics)
Show Figures

Figure 1

Figure 1
<p>Study flowchart. <sup>1</sup> Intra-articular hyaluronic acid; <sup>2</sup> osteoarthritis; <sup>3</sup> hyaluronic acid; <sup>4</sup> magnetic resonance imaging.</p>
Full article ">Figure 2
<p>Pain on the numeric rating scale (NRS) before and after intra-articular hyaluronic acid (IAHA) injection.</p>
Full article ">Figure 3
<p>Correlation between number of infiltrations and pain reduction. The asterisk (*) represents the <span class="html-italic">p</span>-value of the statistical test. One asterisk = <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>The pain reduction after 1, 2, or more than 2 infiltrations on the numeric rating scale.</p>
Full article ">
15 pages, 2991 KiB  
Article
Elevated IL-6 Expression in Autologous Adipose-Derived Stem Cells Regulates RANKL Mediated Inflammation in Osteoarthritis
by Hyun-Joo Lee, Dae-Yong Kim, Hyeon jeong Noh, Song Yi Lee, Ji Ae Yoo, Samuel Jaeyoon Won, Yoon Sang Jeon, Ji Hoon Baek and Dong Jin Ryu
Cells 2024, 13(24), 2046; https://doi.org/10.3390/cells13242046 - 11 Dec 2024
Viewed by 317
Abstract
Interleukin-6 (IL-6) expression in mesenchymal stem cells (MSCs) has been shown to play a pivotal role in modulating cartilage regeneration and immune responses, particularly in the context of diseases that involve both degenerative processes and inflammation, such as osteoarthritis (OA). However, the precise [...] Read more.
Interleukin-6 (IL-6) expression in mesenchymal stem cells (MSCs) has been shown to play a pivotal role in modulating cartilage regeneration and immune responses, particularly in the context of diseases that involve both degenerative processes and inflammation, such as osteoarthritis (OA). However, the precise mechanism through which IL-6 and other immune-regulatory factors influence the therapeutic efficacy of autologous adipose-derived stem cells (ASCs) transplantation in OA treatment remains to be fully elucidated. This study aims to investigate the relationship between IL-6 expression in autologous ASCs isolated from OA patients and their impact on immune modulation, particularly focusing on the regulation of Receptor Activator of Nuclear factor Kappa-Β Ligand (RANKL), a key mediator of immune-driven cartilage degradation in OA. Autologous ASCs were isolated from the stromal vascular fraction (SVF) of adipose tissue obtained from 22 OA patients. The isolated ASCs were cultured and characterized using reverse transcription polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and flow cytometry to the phenotype and immune regulatory factors of MSCs. Based on IL-6 expression levels, ASCs were divided into high and low IL-6 expression groups. These groups were then co-cultured with activated peripheral blood mononuclear cells (PBMCs) to evaluate their immune-modulatory capacity, including the induction of regulatory T cells, inhibition of immune cell proliferation, and regulation of key cytokines, such as interferon-gamma (IFN-γ). Additionally, RANKL expression, a critical factor in osteoclastogenesis and cartilage degradation, was assessed in both ASC groups. High IL-6-expressing ASCs demonstrated a significantly greater capacity to inhibit immune cell proliferation and IFN-γ production compared to their low IL-6-expressing counterparts under co-culture conditions. Moreover, the group of ASCs with high IL-6 expression showed a marked reduction in RANKL expression, suggesting enhanced potential to control osteoclast activity and subsequent cartilage defect in OA. Conclusion: Autologous ASCs with elevated IL-6 expression exhibit enhanced immunomodulatory properties, particularly in regulating over-activated immune response and reducing osteoclastogenesis through RANKL suppression. These findings indicate that selecting ASCs based on IL-6 expression could enhance the therapeutic efficacy of ASC-based treatments for OA by mitigating immune-driven joint inflammation and cartilage degradation, potentially slowing disease progression. Full article
Show Figures

Figure 1

Figure 1
<p>The characterization of ASC-derived OA patients. (<b>A</b>) FACS analysis showing the expression of MSC characterization markers in representative ASCs used in this study. Green histograms represent staining with isotype-matched control antibodies, while red histograms depict the specific expression of each indicated marker. (<b>B</b>) Immunofluorescence staining for FABP4, Osteocalcin, and Aggrecan in ASCs induced to undergo adipogenic, osteogenic, and chondrogenic differentiation, respectively. DAPI was used for nuclear staining. Scale bars: FABP4, 400 μm; Osteocalcin, 200 μm; Aggrecan, 400 μm).</p>
Full article ">Figure 2
<p>Gene and protein expression analysis of ASCs related to immunoregulatory factor, IL-6, TGF-b, and CCL2. The analysis was conducted on ASCs isolated from 22 OA patients. Gene and protein expression levels are presented relative to the levels observed in ASCs isolated from patients S01 to S22. Expression of (<b>A</b>) IL-6, (<b>B</b>) TGF-β, and (<b>C</b>) CCL2 in ASCs. The left panel shows mRNA expression levels, and the right panel presents protein expression both relative to gene and protein levels in ASCs isolated from patients S01 to S22.</p>
Full article ">Figure 3
<p>Comparative analysis of IL-6 expression in ASCs from two groups of osteoarthritis (OA) patients based on IL-6 levels. Based on the IL-6 protein expression, the ASCs of OA patients were divided into two groups: ASCs of 5 patients with high IL-6 expression (ASC<sup>IL-6H</sup>) and five patients with low IL-6 expression (ASC<sup>IL-6L</sup>). (<b>A</b>) Individual IL-6 protein expression values in each patient, divided into the ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup> groups. (<b>B</b>) The average levels of protein expression for IL-6, TGF-β, and CCL2 in the ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup> groups. (<b>C</b>) Table summarizing MSC characterization marker expression for the ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup> groups. The experiments were conducted in triplicates. The results are shown as the mean ± SD. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Comparison of Regulatory T Cell Induction between ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. (<b>A</b>) Flow cytometry analysis of CD4<sup>+</sup>CD25<sup>+</sup>FoxP3<sup>+</sup> regulatory T cells (Tregs) under allogeneic mixed lymphocyte reaction (MLR) cultures with the addition of ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. Five ASCs from each group were cultured under MLR conditions to determine Treg inductivity. (<b>B</b>) In the presence of ASC<sup>IL-6 H</sup> and ASC<sup>IL-6L</sup>, the bars represent the median and range of results obtained from the five ASCs in each group co-cultured under MLR conditions. (<b>C</b>) Cytokine levels in the supernatant of MLR cultures, as measured by CBA assay. The levels of IFN-γ, TNF-α, IL-10, and IL-17 were quantified. The experiments were conducted in triplicates. The results are shown as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Comparison of Immunomodulatory effect between ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. PBMCs stimulated with phytohaemagglutinin (PHA) were cultured alone or in the presence of the five ASCs from each group, ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. (<b>A</b>) After 72 h of co-culture, the proliferating cells and CD25 expression were analyzed by flow cytometer. (<b>B</b>) In the presence of ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>, the bars represent the median and range of results obtained from the five ASCs in each group co-cultured with PHA-stimulated PBMCs. (<b>C</b>) The production of IFN-g, TNF-a, IL-10, and IL-17 in the supernatant of PHA stimulation was analyzed by CBA assay. The experiments were conducted in triplicates. The results are shown as the 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 6
<p>Regulatory Effect on RANKL of immune cells by IL-6 Expression in ASCs. PBMCs stimulated with cultured alone PHA or in the presence of the five ASCs from each group: ASC<sup>IL-6H</sup> and ASC<sup>IL-6L</sup>. After 72 h of co-culture, The RANKL expression of T cells was analyzed by flow cytometer. (<b>A</b>) The gating strategy for RANKL expression of CD3<sup>+</sup> T cells under PHA stimulation. (<b>B</b>) Flow cytometry analysis for RANKL expression of CD3<sup>+</sup> T cells. (<b>C</b>) In the presence of ASC_IL-6H and ASC_IL-6L, the bars represent the median and range of results obtained from the five ASCs in each group co-cultured with PHA-stimulated PBMCs. The experiments were conducted in triplicates. The results are shown as the mean ± SD. ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
13 pages, 1627 KiB  
Review
Innovative Approaches in Knee Osteoarthritis Treatment: A Comprehensive Review of Bone Marrow-Derived Products
by José Fábio Lana, Joseph Purita, Madhan Jeyaraman, Bianca Freitas de Souza, Bruno Lima Rodrigues, Stephany Cares Huber, Carolina Caliari, Gabriel Silva Santos, Lucas Furtado da Fonseca, Ignacio Dallo, Annu Navani, Marco Antônio Percope De Andrade and Peter Albert Everts
Biomedicines 2024, 12(12), 2812; https://doi.org/10.3390/biomedicines12122812 - 11 Dec 2024
Viewed by 369
Abstract
Knee osteoarthritis (OA) is a chronic articular disease characterized by the progressive degeneration of cartilage and bone tissue, leading to the appearance of subchondral cysts, osteophyte formation, and synovial inflammation. Conventional treatments consist of non-steroidal anti-inflammatory drugs (NSAIDs), analgesics, and glucocorticoids. However, the [...] Read more.
Knee osteoarthritis (OA) is a chronic articular disease characterized by the progressive degeneration of cartilage and bone tissue, leading to the appearance of subchondral cysts, osteophyte formation, and synovial inflammation. Conventional treatments consist of non-steroidal anti-inflammatory drugs (NSAIDs), analgesics, and glucocorticoids. However, the prolonged use of these drugs causes adverse effects. NSAIDs, for instance, are known to be nephrotoxic, increasing the damage to articular cartilage. New therapies capable of accelerating the process of tissue regeneration and repair are being discussed, such as the use of orthobiologics that are naturally found in the body and obtained through minimally invasive collection and/or laboratory manipulations. Bone marrow aspirate (BMA) and bone marrow aspirate concentrate (BMAC) are both rich in hematopoietic stem cells, mesenchymal stem cells (MSCs), and growth factors (GFs) that can be used in the healing process due to their anabolic and anti-inflammatory effects. The aim of this literature review is to assess the efficacy of BMA and BMAC in the treatment of knee OA based on the favorable results that researchers have obtained with the use of both orthobiologics envisaging an accelerated healing process and the prevention of OA progression. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

Figure 1
<p>Progression and changes in cartilage and bone due to KOA and the high levels of pro-inflammatory cytokines. Created with BioRender.com, accessed on 4 March 2024.</p>
Full article ">Figure 2
<p>Properties of mesenchymal stem cells and hematopoietic stem cells. Created with BioRender.com, accessed on 4 March 2024.</p>
Full article ">Figure 3
<p>Collection procedure and BMA and BMAC components. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 27 February 2024.</p>
Full article ">Figure 4
<p>BMAC procedure in the laboratory. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 27 February 2024.</p>
Full article ">
14 pages, 4450 KiB  
Article
Integrated Transcriptomic and Proteomic Analyses of Antler Growth and Ossification Mechanisms
by Ruijia Liu, Pan Zhang, Jiade Bai, Zhenyu Zhong, Yunfang Shan, Zhibin Cheng, Qingxun Zhang, Qingyun Guo, Hao Zhang and Bo Zhang
Int. J. Mol. Sci. 2024, 25(23), 13215; https://doi.org/10.3390/ijms252313215 - 9 Dec 2024
Viewed by 478
Abstract
Antlers are the sole mammalian organs capable of continuous regeneration. This distinctive feature has evolved into various biomedical models. Research on mechanisms of antler growth, development, and ossification provides valuable insights for limb regeneration, cartilage-related diseases, and cancer mechanisms. Here, ribonucleic acid sequencing [...] Read more.
Antlers are the sole mammalian organs capable of continuous regeneration. This distinctive feature has evolved into various biomedical models. Research on mechanisms of antler growth, development, and ossification provides valuable insights for limb regeneration, cartilage-related diseases, and cancer mechanisms. Here, ribonucleic acid sequencing (RNA-seq) and four-dimensional data-independent acquisition (4D DIA) technologies were employed to examine gene and protein expression differences among four tissue layers of the Chinese milu deer antler: reserve mesenchyme (RM), precartilage (PC), transition zone (TZ), cartilage (CA). Overall, 4611 differentially expressed genes (DEGs) and 2388 differentially expressed proteins (DEPs) were identified in the transcriptome and proteome, respectively. Among the 828 DEGs common to both omics approaches, genes from the collagen, integrin, and solute carrier families, and signaling molecules were emphasized for their roles in the regulation of antler growth, development, and ossification. Bioinformatics analysis revealed that in addition to being regulated by vascular and nerve regeneration pathways, antler growth and development are significantly influenced by numerous cancer-related signaling pathways. This indicates that antler growth mechanisms may be similar to those of cancer cell proliferation and development. This study lays a foundation for future research on the mechanisms underlying the rapid growth and ossification of antlers. Full article
(This article belongs to the Section Molecular Informatics)
Show Figures

Figure 1

Figure 1
<p>Number of differentially expressed genes (DEGs) and functional enrichment analysis and statistics of DEGs. (<b>a</b>) DEGs statistics of 6 groups. (<b>b</b>) Overlapping statistics of DEGs obtained by comparing six groups. (<b>c</b>) The top 15 most significant pathways enriched in DEGs across the six groups. In all pairwise comparisons, upregulated/downregulated genes refer to those that are upregulated/downregulated in the first group of the comparison. For example, in RM_ vs. _PC, upregulated genes are those that are upregulated in RM and downregulated in PC.</p>
Full article ">Figure 2
<p>Short Time-series Expression Miner (STEM) analysis was used to explore gene regulatory networks in four tissue layers of velvet. (<b>a</b>) The trend analysis of DEGs at different tissue layers. Each profile corresponds to a rectangle, with the number in the top-left corner representing the profile’s ID. The line chart inside shows the trend of expression changes across tissue layers (RM, PC, TZ, CA), and the value in the bottom-left corner is the corresponding significance level (<span class="html-italic">p</span>-value). Color-coded trend graphs: These indicate that the time-series pattern of the profile exhibits a significant change trend. Profiles with the same color represent those belonging to the same cluster. Non-color-coded trend graphs: These indicate that the time-series pattern of the profile shows a statistically insignificant change trend. (<b>b</b>–<b>f</b>) Kyoto Encyclopedia of Genes and Genomics (KEGG) enrichment analysis of DEGs in Clusters 1–5. (<b>g</b>) Heatmap of functional DEGs in the 5 clusters. C1: Cluster 1 (Profiles 3, 0, 9, 1, 10); C2: Cluster 2 (Profiles 24, 23, 15); C3: Cluster 3 (Profile 21); C4: Cluster 4 (Profile 2); C5: Cluster 5 (Profile 14).</p>
Full article ">Figure 3
<p>Protein expression characteristics and functional enrichment analysis of differential proteins (DEPs) in the four tissue layers of antlers. (<b>a</b>) Venn diagram for protein identification. (<b>b</b>) Bar chart of DEPs statistics for the six comparisons. (<b>c</b>) KEGG enrichment analysis of DEPs in the six groups. (<b>d</b>) Gene ontology (GO) enrichment chord diagram of DEPs in PC vs. CA. In all pairwise comparisons, upregulated/downregulated proteins refer to those that are upregulated/downregulated in the first group of the comparison. For example, in RM_ vs. _PC, upregulated proteins are those that are upregulated in RM and downregulated in PC.</p>
Full article ">Figure 4
<p>Correlation analysis between transcriptomics and proteomics. (<b>a</b>) Overlapping genes between transcriptomics and proteomics. (<b>b</b>) Histogram showing gene-wise mRNA–protein Spearman’s correlations. (<b>c</b>) Overlapping statistics of DEGs and DEPs.</p>
Full article ">Figure 5
<p>KEGG functional enrichment analysis of overlapping genes between six groups of DEGs and DEPs.</p>
Full article ">Figure 6
<p>Validation of the DEGs in the four tissue layers of antlers using quantitative real-time polymerase chain reaction (qRT-PCR). * <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.</p>
Full article ">
17 pages, 2293 KiB  
Article
Pararamosis, a Neglected Tropical Disease Induced by Premolis semirufa Caterpillar Toxins: Investigating Their Effects on Synovial Cell Inflammation
by Paula C. Pohl, Isadora M. Villas-Boas, Giselle Pidde and Denise V. Tambourgi
Int. J. Mol. Sci. 2024, 25(23), 13149; https://doi.org/10.3390/ijms252313149 - 6 Dec 2024
Viewed by 365
Abstract
Pararamosis, also known as Pararama-associated phalangeal periarthritis, is a neglected tropical disease primarily affecting rubber tappers in the Amazon region. It is caused by contact with the urticating hairs of the Premolis semirufa moth caterpillar, which resides in rubber plantations. The condition is [...] Read more.
Pararamosis, also known as Pararama-associated phalangeal periarthritis, is a neglected tropical disease primarily affecting rubber tappers in the Amazon region. It is caused by contact with the urticating hairs of the Premolis semirufa moth caterpillar, which resides in rubber plantations. The condition is marked by the thickening of the articular synovial membrane and cartilage impairment, features associated with chronic synovitis. Given the significance of synovial inflammation in osteoarticular diseases, in this study, the role of synoviocytes and their interactions with macrophages and chondrocytes are examined when stimulated by Pararama toxins. Synoviocytes and macrophages treated with Pararama hair extract showed an increased production of cytokines IL-6, IL-1β, and TNF-α, indicating a direct effect on these cells. In cocultures, there was a significant rise in inflammation, with levels of IL-1β, IL-6, and chemokines CCL2, CCL5, and CXCL8 increasing up to seven times compared to monocultures. Additionally, matrix-degrading enzymes MMP-1 and MMP-3 were significantly elevated in cocultures. Chondrocytes exposed to the extract also produced IL-6, CCL2, and CCL5, and in cocultures with synoviocytes, there was a notable increase in IL-6, CCL5, and CXCL8, as well as a doubling of MMP-1 and MMP-3 levels. These findings underscore the critical role of cell crosstalk in the inflammatory and catabolic processes associated with pararamosis and demonstrate how Pararama hair extract can influence factors affecting cartilage health, providing valuable insights into this condition. Full article
(This article belongs to the Special Issue Molecular Toxicity Research of Biological Venoms)
Show Figures

Figure 1

Figure 1
<p>Cytokine and chemokine levels in synoviocyte cultures after Pararama hair extract stimulation. Synoviocytes (1 × 10<sup>4</sup> cells/well) were treated with Pararama hair extract (EXT 15, 30, or 60 μg/mL) or PBS for 24, 48, or 72 h. After each treatment period, the supernatants were collected to assess the concentrations of cytokines and chemokines via a cytometric bead array (CBA) (<b>A</b>–<b>D</b>). The results represent two separate experiments performed in triplicate and are expressed as the mean of the concentrations of the molecules ± SEM. Statistical analyses within the same incubation time were performed using one-way ANOVA and Tukey’s post hoc test, while statistical analyses between different incubation times were conducted using two-way ANOVA and Tukey’s post hoc test. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤ 0.001; and **** <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 2
<p>Metalloproteinase levels in synoviocyte culture after Pararama hair extract stimulation. Synoviocytes (1 × 10<sup>4</sup> cells/well) were treated with Pararama hair extract (EXT 15, 30, or 60 μg/mL) or PBS for 24, 48, or 72 h. After stimulation, the production of metalloproteinases in the supernatants of the cultures was determined by enzyme-linked immunosorbent assay (ELISA) (<b>A</b>–<b>E</b>). The results represent two separate experiments performed in triplicate and are expressed as the mean of the concentrations of the molecules ± SEM. Statistical analyses within the same incubation time were performed using one-way ANOVA and Tukey’s post hoc test, while statistical analyses between different incubation times were conducted using two-way ANOVA and Tukey’s post hoc test. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤ 0.001; and **** <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 3
<p>Cytokine and chemokine levels in cocultured synoviocytes and macrophages induced by Pararama hair extract. THP1-derived macrophages (MΦs), synoviocytes, and cocultured cells in the transwell system were treated with Pararama hair extract (EXT- 48 µg/mL) or PBS (as a negative control) for 48 h. Supernatants of the cultures were collected to assess the concentrations of cytokines and chemokines via a cytometric bead array (CBA) (<b>A</b>–<b>H</b>). The results summarize two independent experiments, each performed in triplicate, and are expressed as the means of the concentrations of the molecules ± SEM. The data were analyzed using two-way ANOVA and Tukey’s post hoc test. ns = not significant; * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001, and **** <span class="html-italic">p</span> ≤ 0.0001. The dotted line represents the limit of detection for each analyte.</p>
Full article ">Figure 4
<p>Metalloproteinase levels in cocultured synoviocytes and macrophages induced by Pararama hair extract. THP1-derived macrophages (MΦ), synoviocytes, and cocultured cells in the transwell system were treated with Pararama hair extract (EXT- 48 µg/mL) or PBS (as a negative control) for 48 h. Supernatants of the cultures were collected to assess the concentration of metalloproteinases by enzyme-linked immunosorbent assay (ELISA) (<b>A</b>–<b>C</b>). The results summarize two independent experiments, each performed in triplicate, and are expressed as the mean of the concentrations of the molecules ± SEM. The data were analyzed using two-way ANOVA and Tukey’s post hoc test. ns = not significant; * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001, and **** <span class="html-italic">p</span> ≤ 0.0001. The dotted line represents the limit of detection for each analyte.</p>
Full article ">Figure 5
<p>Cytokine and chemokine levels in cocultured synoviocytes and chondrocytes induced by Pararama hair extract. Synoviocytes, chondrocytes, and cocultured cells in the transwell system were treated with Pararama hair extract (EXT; 48 µg/mL) or PBS (as a negative control) for 48 h. Supernatants of the cultures were collected to assess the concentrations of cytokines and chemokines via a cytometric bead array (CBA) (<b>A</b>–<b>D</b>). The results summarize two independent experiments, each performed in triplicate, and are expressed as the means of the concentrations of the molecules ± SEM. The data were analyzed using two-way ANOVA and Tukey’s post hoc test. ns = not significant; *** <span class="html-italic">p</span> ≤ 0.001, and **** <span class="html-italic">p</span> ≤ 0.0001. The dotted line represents the limit of detection for each analyte.</p>
Full article ">Figure 6
<p>Metalloproteinase levels in synoviocyte and chondrocyte cocultures induced by Pararama hair extract. Synoviocytes, chondrocytes, and cocultured cells in the transwell system were treated with Pararama hair extract (EXT- 48 µg/mL) or PBS (as a negative control) for 48 h. Supernatants of the cultures were collected to assess the concentration of metalloproteinases by enzyme-linked immunosorbent assay (ELISA) (<b>A</b>–<b>C</b>). The results summarize two independent experiments, each performed in triplicate, and are expressed as the means of the concentrations of the molecules ± SEM. The data were analyzed using two-way ANOVA and Tukey’s post hoc test. ns-not significant, ** <span class="html-italic">p</span> ≤ 0.01; and **** <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">
24 pages, 3604 KiB  
Article
Enhancement of Chondrogenic Differentiation in Bone Marrow-Derived Stem Cell Spheroids by Cuminum cyminum Methanolic Extract: Insights into Concentration-Dependent mRNA Expression and Gene Clustering Analysis
by Kyung-Hwan Na, Hyun-Jin Lee, Ju-Hwan Kim, Md. Salah Uddin, Yoon-Hee Park, Young-Min Song, Chul-Sung Park and Jun-Beom Park
J. Pers. Med. 2024, 14(12), 1142; https://doi.org/10.3390/jpm14121142 - 5 Dec 2024
Viewed by 598
Abstract
Background/Objectives: Cuminum cyminum L. has been utilized as a medicinal plant for centuries. This research sought to examine the effects of cumin methanolic extract (CMT) on the chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells. Methods: Spheroids were generated using [...] Read more.
Background/Objectives: Cuminum cyminum L. has been utilized as a medicinal plant for centuries. This research sought to examine the effects of cumin methanolic extract (CMT) on the chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells. Methods: Spheroids were generated using human stem cells and cultured with CMT at concentrations between 0 and 1 µg/mL. Morphological assessments and cell viability tests were conducted on days 1 and 3. Chondrogenic differentiation expression was evaluated through quantitative polymerase chain reaction, Western blot, and RNA sequencing. SOX9, FAM20B, COL2A1, and COL1A1 mRNA expression levels were determined using real-time polymerase chain reaction. Protein expression was analyzed via Western blot. Results: Throughout this study, the spheroids maintained their integrity and shape. No significant variations in spheroid diameter were observed among the groups. CMT treatment enhanced the expression of SOX9 and FAM20B. Conclusions: The methanolic extract of Cuminum cyminum facilitated chondrogenic differentiation in human bone marrow-derived mesenchymal stem cells by modulating SOX9 and FAM20B expression. This indicates its potential application in cartilage tissue engineering. Full article
(This article belongs to the Section Regenerative Medicine and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Evaluation of morphology and cellular viability of stem cell spheroids. (<b>A</b>) Assessment of cell morphology on days 1, 3, 7, and 14 for various concentrations of cumin methanolic extract, as seen under 100× original magnification. The scale bar in the image represents 200 μm. (<b>B</b>) Diameters of the cell spheroids on days 1, 3, 7, and 14 for different concentrations of the cumin methanolic extract. *: There was statistical difference when compared with the control on day 1. **: Statistical differences were noted when compared with the control on day 1. ***: There was statistical difference when compared with the control on day 3. ****: Statistical differences were noted when compared with the control on day 7. *****: There was statistical difference when compared with the control on day 14. (<b>C</b>) Cellular viability of the spheroids using the Cell Counting Kit-8 on days 1, 3, 7, and 14 for various concentrations of the extract. *: There was a significantly higher value for the 1 μg/mL group when compared with the control group on day 14.</p>
Full article ">Figure 1 Cont.
<p>Evaluation of morphology and cellular viability of stem cell spheroids. (<b>A</b>) Assessment of cell morphology on days 1, 3, 7, and 14 for various concentrations of cumin methanolic extract, as seen under 100× original magnification. The scale bar in the image represents 200 μm. (<b>B</b>) Diameters of the cell spheroids on days 1, 3, 7, and 14 for different concentrations of the cumin methanolic extract. *: There was statistical difference when compared with the control on day 1. **: Statistical differences were noted when compared with the control on day 1. ***: There was statistical difference when compared with the control on day 3. ****: Statistical differences were noted when compared with the control on day 7. *****: There was statistical difference when compared with the control on day 14. (<b>C</b>) Cellular viability of the spheroids using the Cell Counting Kit-8 on days 1, 3, 7, and 14 for various concentrations of the extract. *: There was a significantly higher value for the 1 μg/mL group when compared with the control group on day 14.</p>
Full article ">Figure 2
<p>Quantification of gene expression was performed using real-time polymerase chain reaction on day 14 for various concentrations of cumin methanolic extracts. Significant differences compared to the control group are indicated (*). (<b>A</b>) The expression of SOX9 was quantified, showing a significant increase in mRNA levels at a concentration of 0.001 μg/mL. (<b>B</b>) The expression of FAM20B mRNA was quantified, revealing significant differences in comparison to the control group. (<b>C</b>) The expression of COL2A1 mRNA was quantified. (<b>D</b>) The expression of COL1A1 was quantified, showing significant differences in mRNA levels compared to the control group.</p>
Full article ">Figure 2 Cont.
<p>Quantification of gene expression was performed using real-time polymerase chain reaction on day 14 for various concentrations of cumin methanolic extracts. Significant differences compared to the control group are indicated (*). (<b>A</b>) The expression of SOX9 was quantified, showing a significant increase in mRNA levels at a concentration of 0.001 μg/mL. (<b>B</b>) The expression of FAM20B mRNA was quantified, revealing significant differences in comparison to the control group. (<b>C</b>) The expression of COL2A1 mRNA was quantified. (<b>D</b>) The expression of COL1A1 was quantified, showing significant differences in mRNA levels compared to the control group.</p>
Full article ">Figure 3
<p>Western blot analysis. (<b>A</b>) Western blot analysis to evaluate the expression levels of SOX9, collagen II, collagen I, and β-catenin at different concentrations of cumin methanolic extracts. (<b>B</b>) Quantification of expression of SOX9 protein by Western blot analysis on day 14 for different concentrations of cumin methanolic extracts. Significant increases in SOX9 mRNA expression were observed at a concentration of 0.001 and 0.01 μg/mL when compared to the control group (*). (<b>C</b>) Quantification of expression of collagen I protein by Western blot analysis on day 14 for different concentrations of cumin methanolic extracts.</p>
Full article ">Figure 3 Cont.
<p>Western blot analysis. (<b>A</b>) Western blot analysis to evaluate the expression levels of SOX9, collagen II, collagen I, and β-catenin at different concentrations of cumin methanolic extracts. (<b>B</b>) Quantification of expression of SOX9 protein by Western blot analysis on day 14 for different concentrations of cumin methanolic extracts. Significant increases in SOX9 mRNA expression were observed at a concentration of 0.001 and 0.01 μg/mL when compared to the control group (*). (<b>C</b>) Quantification of expression of collagen I protein by Western blot analysis on day 14 for different concentrations of cumin methanolic extracts.</p>
Full article ">Figure 4
<p>Differentially expressed mRNAs in four different groups. The mRNAs were clustered based on their expression levels, and only those with a fold change greater than 2.0 and a log2 normalized data greater than 3 were included in the analysis. (<b>A</b>) The clustering analysis of differentially expressed mRNAs in the 0.001 μg/mL group compared to the control group. (<b>B</b>) The analysis of differentially expressed mRNAs in the 0.01 μg/mL group compared to the control group. (<b>C</b>) The clustering analysis of differentially expressed mRNAs in the 0.1 μg/mL group vs. the control group. (<b>D</b>) The analysis of mRNAs with differential expression in the 1 μg/mL group compared to the control group.</p>
Full article ">Figure 4 Cont.
<p>Differentially expressed mRNAs in four different groups. The mRNAs were clustered based on their expression levels, and only those with a fold change greater than 2.0 and a log2 normalized data greater than 3 were included in the analysis. (<b>A</b>) The clustering analysis of differentially expressed mRNAs in the 0.001 μg/mL group compared to the control group. (<b>B</b>) The analysis of differentially expressed mRNAs in the 0.01 μg/mL group compared to the control group. (<b>C</b>) The clustering analysis of differentially expressed mRNAs in the 0.1 μg/mL group vs. the control group. (<b>D</b>) The analysis of mRNAs with differential expression in the 1 μg/mL group compared to the control group.</p>
Full article ">Figure 4 Cont.
<p>Differentially expressed mRNAs in four different groups. The mRNAs were clustered based on their expression levels, and only those with a fold change greater than 2.0 and a log2 normalized data greater than 3 were included in the analysis. (<b>A</b>) The clustering analysis of differentially expressed mRNAs in the 0.001 μg/mL group compared to the control group. (<b>B</b>) The analysis of differentially expressed mRNAs in the 0.01 μg/mL group compared to the control group. (<b>C</b>) The clustering analysis of differentially expressed mRNAs in the 0.1 μg/mL group vs. the control group. (<b>D</b>) The analysis of mRNAs with differential expression in the 1 μg/mL group compared to the control group.</p>
Full article ">Figure 4 Cont.
<p>Differentially expressed mRNAs in four different groups. The mRNAs were clustered based on their expression levels, and only those with a fold change greater than 2.0 and a log2 normalized data greater than 3 were included in the analysis. (<b>A</b>) The clustering analysis of differentially expressed mRNAs in the 0.001 μg/mL group compared to the control group. (<b>B</b>) The analysis of differentially expressed mRNAs in the 0.01 μg/mL group compared to the control group. (<b>C</b>) The clustering analysis of differentially expressed mRNAs in the 0.1 μg/mL group vs. the control group. (<b>D</b>) The analysis of mRNAs with differential expression in the 1 μg/mL group compared to the control group.</p>
Full article ">Figure 5
<p>Clustering heatmap of differentially expressed mRNAs in response to cumin methanolic extract (CMT) treatment. The clustering heatmap shows the expression patterns of 76 differentially expressed mRNAs (fold change &gt; 2, <span class="html-italic">p</span>-value &lt; 0.05) in bone marrow-derived mesenchymal stem cells (BMSCs) treated with cumin methanolic extract (CMT) at various concentrations (0.001–1 µg/mL) compared to the control group. Upregulated genes are shown in red, downregulated genes in blue, and the intensity of the color represents the magnitude of fold change.</p>
Full article ">Figure 6
<p>The expression of genes related to chondrogenesis. The extracellular matrix (ECM)receptor interaction pathway map generated using the Kyoto Encyclopedia of Genes and Genomes database. Differentially expressed genes involved in this pathway are highlighted, demonstrating the impact of cumin methanolic extracts on ECM interactions during chondrogenesis.</p>
Full article ">
15 pages, 6353 KiB  
Article
Tissue Engineering Construct for Articular Cartilage Restoration with Stromal Cells from Synovium vs. Dental Pulp—A Pre-Clinical Study
by Tiago Lazzaretti Fernandes, João Paulo Cortez Santanna, Rafaella Rogatto de Faria, Enzo Radaic Pastore, Daniela Franco Bueno and Arnaldo José Hernandez
Pharmaceutics 2024, 16(12), 1558; https://doi.org/10.3390/pharmaceutics16121558 - 5 Dec 2024
Viewed by 391
Abstract
Background/Objectives: Cartilage injuries and osteoarthritis are prevalent public health problems, due to their disabling nature and economic impact. Mesenchymal stromal cells (MSCs) isolated from different tissues have the immunomodulatory capacity to regulate local joint environment. This translational study aims to compare cartilage restoration [...] Read more.
Background/Objectives: Cartilage injuries and osteoarthritis are prevalent public health problems, due to their disabling nature and economic impact. Mesenchymal stromal cells (MSCs) isolated from different tissues have the immunomodulatory capacity to regulate local joint environment. This translational study aims to compare cartilage restoration from MSCs from the synovial membrane (SM) and dental pulp (DP) by a tissue-engineered construct with Good Manufacturing Practices. Methods: A controlled experimental study was conducted on fourteen miniature pigs, using scaffold-free Tissue Engineering Constructs (TECs) from DP and SM MSCs, with a 6-month follow-up. Total thickness cartilage defects were created in both hind knees; one side was left untreated and the other received a TEC from either DP (n = 7) or SM (n = 7). An MRI assessed the morphology using the MOCART scoring system, T2 mapping evaluated water, and collagen fiber composition, and histological analysis was performed using the ICRS-2 score. Results: The untreated group had a mean MOCART value of 46.2 ± 13.4, while the SM-treated group was 65.7 ± 15.5 (p < 0.05) and the DP-treated group was 59.0 ± 7.9 (n.s.). The T2 mapping indicated a mean value of T2 of 54.9 ± 1.9 for native cartilage, with the untreated group at 50.9 ± 2.4 (p < 0.05). No difference was found between the T2 value of native cartilage and the treated groups. The ICRS-2 mean values were 42.1 ± 14.8 for the untreated group, 64.3 ± 19.0 for SM (p < 0.05), and 54.3 ± 12.2 for DP (n.s.). Conclusion: MRI and histological analysis indicated that TEC treatment led to superior cartilage coverage and quality compared to the defect group. TECs from SM demonstrated better results than the defect group in the histological assessment. Full article
(This article belongs to the Special Issue Osteoarthritis and Cartilage Biologics)
Show Figures

Figure 1

Figure 1
<p>Cell differentiation: (<b>A</b>) Osteogenic differentiation visualized with Alizarin Red S staining (21 days), (<b>B</b>) adipogenic differentiation visualized with Oil Red O staining (18 days), and (<b>C</b>) chondrogenic differentiation visualized with Alcian Blue 8G staining (21 days), with respective controls shown below. Images acquired via optical microscopy (EVOS XL Cell Imaging System).</p>
Full article ">Figure 2
<p>Three confluent tissue engineering construct samples after 15 days of culture forming a three-dimensional structure.</p>
Full article ">Figure 3
<p>Full-thickness cartilage defect measuring 6 mm in the medial femoral condyle of the right knee’s hind limb.</p>
Full article ">Figure 4
<p>Flow cytometry analysis showing positive reactions to mesenchymal markers (CD29, CD73, CD105, CD44, CD90, and CD166) and negative reactions to hematopoietic (CD34 and CD45). In purple, the cell population that presents the respective marker.</p>
Full article ">Figure 5
<p>Cartilage defect 6 months after surgery, arrows indicate the region where the defect was made. (<b>A</b>) Defect-only (solid arrow); (<b>B</b>) defect treated with a tissue engineering construct (TEC) (dashed arrow).</p>
Full article ">Figure 6
<p>Overall assessment score values in MOCART 3D score for the defect-only (untreated), dental pulp treatment, and synovial treatment groups, showing a significant difference * (<span class="html-italic">p</span> &lt; 0.05) between the defect-only group and the group treated with a TEC from the synovial.</p>
Full article ">Figure 7
<p>Mean T2 values of the groups from native (adjacent cartilage), defect-only (untreated), dental pulp treatment, and synovial treatment groups. There was a significant difference * (<span class="html-italic">p</span> &lt; 0.05) between native cartilage and defect-only groups.</p>
Full article ">Figure 8
<p>Mean value of overall assessment of cartilage repair using the ICRS-2 histological score for the defect-only (untreated), dental pulp treatment, and synovial treatment groups. A significant difference * (<span class="html-italic">p</span> &lt; 0.05) was found between the defect-only group and the group treated with a TEC from synovial membrane.</p>
Full article ">Figure 9
<p>Histological evaluation of (<b>A</b>) defect-only in posterior knee; (<b>B</b>) defect treated with a TEC from synovial membrane in posterior knee.</p>
Full article ">Figure 10
<p>Histological evaluation of (<b>A</b>) defect-only in posterior knee; (<b>B</b>) defect treated with a TEC from dental pulp in posterior knee.</p>
Full article ">
Back to TopTop