[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

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
remove_circle_outline

Search Results (9,689)

Search Parameters:
Keywords = TNF-α

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
10 pages, 2020 KiB  
Article
Influence of Apis mellifera syriaca Bee Venom on Nociception and Inflammatory Cytokine Profiles in Experimental Hyperalgesia
by Mohamad Ayoub, Salma Fayjaloun, Rabih Roufayel, Dany El Obeid, Ziad Fajloun, Mohamad Rima and Marc Karam
Toxins 2025, 17(1), 18; https://doi.org/10.3390/toxins17010018 (registering DOI) - 1 Jan 2025
Abstract
Hyperalgesia is a condition marked by an abnormal increase in pain sensitivity, often occurring in response to tissue injury, inflammation, or prolonged exposure to certain medications. Inflammatory mediators, such as cytokines IL-1β, IL-6, and TNF-α, play a central role in this process, amplifying [...] Read more.
Hyperalgesia is a condition marked by an abnormal increase in pain sensitivity, often occurring in response to tissue injury, inflammation, or prolonged exposure to certain medications. Inflammatory mediators, such as cytokines IL-1β, IL-6, and TNF-α, play a central role in this process, amplifying pain perception. Developing effective treatments that address the underlying mechanisms of hyperalgesia is an active field of research. Apis mellifera syriaca venom demonstrated potential immunomodulatory activity associated with cytokine release in vivo. Therefore, the aim of this study is to evaluate the effect of Apis mellifera syriaca bee venom (AmsBV) on pain sensitivity in a formalin-induced hyperalgesia mice model and to evaluate the potential role of cytokines associated with the nociception of pain. The hotplate test, used to measure pain latency, showed that hypersensitivity to pain was induced in formalin-injected male mice only, with no changes in females, suggesting a sex-based response to formalin. When applied, AmsBV reduced pain sensitivity in males, suggesting pain relief potential. At the molecular level, AmsBV was able to reduce pro-inflammatory interleukin IL-4 and cytokine IFN-γ, emphasizing its immunomodulatory potential. Interestingly, the venom restored anti-inflammatory IL-10 levels that were significantly decreased in hyperalgesia males. Together, these findings highlight the therapeutic potential for AmsBV in managing inflammation and reducing pain, particularly hyperalgesia. Full article
(This article belongs to the Special Issue Animals Venom in Drug Discovery: A Valuable Therapeutic Tool)
Show Figures

Figure 1

Figure 1
<p>Validation of formalin-induced hyperalgesia in mice models. Changes in pain sensitivity were reported using the hotplate test. Male and female mice were injected with formalin or an equivalent volume of PBS (at T = 0) after recording baseline sensitivity daily over 4 days. Pain sensitivity was tracked up to 2 days post-formalin injection. <span class="html-italic">* p</span> ≤ 0.05; <span class="html-italic">** p</span> ≤ 0.01.</p>
Full article ">Figure 2
<p><span class="html-italic">Ams</span>BV does not affect pain threshold in mice. Alteration in pain sensitivity in male and female mice injected with <span class="html-italic">Ams</span>BV or an equivalent volume of PBS (at T = 0), after recording baseline sensitivity daily over 4 days, was investigated up to 2 days post-formalin injection.</p>
Full article ">Figure 3
<p><span class="html-italic">Ams</span>BV decreases pain sensitivity in formalin-induced hyperalgesia in male mice models. Male and female mice were injected with formalin or an equivalent volume of PBS (at T = 0) after recording baseline sensitivity daily over 4 days. BV was injected 10 min post-formalin injection and pain sensitivity was tracked up to 2 days using the hotplate test. <span class="html-italic">* p</span> ≤ 0.05; <span class="html-italic">** p</span> ≤ 0.01.</p>
Full article ">Figure 4
<p>Evaluation of interleukin-level changes upon <span class="html-italic">Ams</span>BV in formalin-induced hyperalgesia mice model. (<b>A</b>) IL-4 levels were investigated in male and (<b>B</b>) female mice. (<b>C</b>) IL-6 levels were quantified in male and (<b>D</b>) female mice following venom injection with/without formalin preinjection. Values are means ± SEM for <span class="html-italic">n</span> = 5 per group. <span class="html-italic">*** p</span> ≤ 0.001; <span class="html-italic">**** p</span> ≤ 0.0001.</p>
Full article ">Figure 5
<p>Evaluation of interleukin and cytokine-level changes upon <span class="html-italic">Ams</span>BV in formalin-induced hyperalgesia mice model. (<b>A</b>) IL-10 levels were investigated in male and (<b>B</b>) female mice. (<b>C</b>) IFN-γ levels were quantified in male and (<b>D</b>) female mice following venom injection with/without formalin preinjection. Values are means ± SEM for <span class="html-italic">n</span> = 5 per group. <span class="html-italic">** p ≤ 0.01;</span> <span class="html-italic">*** p</span> ≤ 0.001.</p>
Full article ">
16 pages, 2656 KiB  
Article
Innovative Lipid Nanoparticles Co-Delivering Hydroxychloroquine and siRNA for Enhanced Rheumatoid Arthritis Therapy
by Yanru Feng, Xintong Pan, Ziqian Li, Yue Li, Ya’nan Sun, Shaokun Yang, Chaoxing He, Yunjie Dang, Lu Huang and Bai Xiang
Pharmaceutics 2025, 17(1), 45; https://doi.org/10.3390/pharmaceutics17010045 (registering DOI) - 1 Jan 2025
Viewed by 101
Abstract
Background: Rheumatoid arthritis (RA) is a debilitating autoimmune disorder characterized by chronic inflammation and joint damage. Despite advancements in treatment, complete remission remains elusive. Methods: In this study, we introduce a novel lipid nanoparticle formulation co-delivering hydroxychloroquine (HCQ) and siRNA targeting TNF-α (si [...] Read more.
Background: Rheumatoid arthritis (RA) is a debilitating autoimmune disorder characterized by chronic inflammation and joint damage. Despite advancements in treatment, complete remission remains elusive. Methods: In this study, we introduce a novel lipid nanoparticle formulation co-delivering hydroxychloroquine (HCQ) and siRNA targeting TNF-α (siTNF-α) using microfluidic technology, marking the first use of such a combination for RA therapy. Results: In LPS-stimulated RAW 264.7 cells, the nanoparticles effectively reduced inflammatory markers. When administered via an intra-articular injection in a rat model, they significantly decreased joint inflammation and demonstrated good biological safety. Conclusions: This pioneering approach highlights the potential of lipid nanoparticles as a dual-delivery platform for enhanced RA treatment through targeted intra-articular administration. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Characterization of LNP-si<span class="html-italic">TNF-α</span> and LNP-si<span class="html-italic">TNF-α</span>-HCQ. (<b>A</b>) Preparation process of LNP-si<span class="html-italic">TNF-α</span>-HCQ. Morphology of LNP-si<span class="html-italic">TNF-α</span> (<b>B</b>) and LNP-si<span class="html-italic">TNF-α</span>-HCQ (<b>C</b>) characterized by cryo-electron microscopy. (<b>D</b>) The high-performance liquid chromatogram of HCQ. (<b>E</b>) The serum stability of free si<span class="html-italic">N.C.</span>, LNP-si<span class="html-italic">N.C.</span>, and LNP-si<span class="html-italic">N.C.</span>-HCQ. (<b>F</b>) The particle size and PDI stability of LNP-si<span class="html-italic">TNF-α</span> and LNP-si<span class="html-italic">TNF-α</span>-HCQ.</p>
Full article ">Figure 2
<p>Cytotoxicity in RAW 264.7 cells of LNP-si<span class="html-italic">N.C.</span> (<b>A</b>), LNP-si<span class="html-italic">TNF-α</span> (<b>B</b>), LNP-si<span class="html-italic">N.C.</span>-HCQ (<b>C</b>), and LNP-si<span class="html-italic">TNF-α</span>-HCQ (<b>D</b>) prepared with different concentrations of siRNA; ns: no significance.</p>
Full article ">Figure 3
<p>The cellular uptake of free si<span class="html-italic">N.C.</span>-Cy5, LNP-si<span class="html-italic">N.C.</span>-Cy5, and LNP-si<span class="html-italic">N.C.</span>-Cy5-HCQ by RAW 264.7 cells was studied using confocal laser microscopy (<b>A</b>) and flow cytometry (<b>B</b>). (<b>C</b>) The in vitro anti-inflammatory effect of free si<span class="html-italic">TNF-α</span>, Free HCQ, LNP-si<span class="html-italic">TNF-α</span>, LNP-si<span class="html-italic">N.C.</span>-HCQ, and LNP-si<span class="html-italic">TNF-α</span>-HCQ on RAW 264.7 cells. Scale bar: 10 μm (<b>A</b>); ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, ns: no significance.</p>
Full article ">Figure 4
<p>Therapeutic efficacy of LNP-si<span class="html-italic">TNF-α</span>-HCQ on adjuvant-induced rheumatoid arthritis rat model. (<b>A</b>) Timeline of in vivo study. (<b>B</b>) Biodistribution of LNP-si<span class="html-italic">TNF-α</span>-HCQ in rheumatoid arthritis rat articular cavity. Arthritis score (<b>C</b>), paw volume (<b>D</b>), and TNF-α content (<b>F</b>) in plasma of AIA rats were assessed in different groups. (<b>E</b>) The picture of the rat’s hind paw in different groups before sacrifice. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Safety evaluation of AIA rats treated with different administration groups. (<b>A</b>) H&amp;E staining of various organs. (<b>B</b>) The body weight changes in rats. (<b>C</b>–<b>F</b>) The number of WBCs, RBCs, PLTs, and Lymphs in the blood after rats were sacrificed. (<b>G</b>–<b>J</b>) The concentration of AST, ALT, ALP, and CREA in the blood. Scale bar: 100 μm; ns: no significance.</p>
Full article ">
19 pages, 4268 KiB  
Article
Effects of Cordyceps cicadae Polysaccharide on Gut Microbiota, the Intestinal Mucosal Barrier, and Inflammation in Diabetic Mice
by Lijia Sun, Huaibo Yuan, Huiqing Ma and Yani Wang
Metabolites 2025, 15(1), 8; https://doi.org/10.3390/metabo15010008 (registering DOI) - 1 Jan 2025
Viewed by 118
Abstract
Background: Polysaccharides produced by the edible fungus Cordyceps cicadae can regulate blood sugar levels and may represent a suitable candidate for the treatment of diabetes and its complications. However, there is limited information available about the mechanism of how C. cicadae polysaccharide (CCP) [...] Read more.
Background: Polysaccharides produced by the edible fungus Cordyceps cicadae can regulate blood sugar levels and may represent a suitable candidate for the treatment of diabetes and its complications. However, there is limited information available about the mechanism of how C. cicadae polysaccharide (CCP) might improve diabetic conditions. Methods: This study investigated its effects on the intestinal microbiota, intestinal mucosal barrier, and inflammation in mice with type 2 diabetes mellitus (T2DM) induced by streptozotocin, and its potential mechanisms. Results: Compared with the DC (diabetes model control group), CCPH oral treatment significantly increased the number of beneficial bifidobacteria, bifidobacteria, and lactobacilli (p < 0.01), restored the diversity of intestinal microorganisms in diabetic mice, and the proportions of Firmicutes and Bacteroidetes (34.36%/54.65%) were significantly lower than those of the DC (52.15%/32.09%). Moreover, CCPH significantly reduced the content of endotoxin (lipopolysaccharide, LPS) and D-lactic acid(D-LA) (p < 0.05), the activities of antioxidant enzymes and total antioxidant capacity were significantly increased (p < 0.01), and the content of proinflammatory cytokines TNF-α, IL-6, and IL-1β were reduced by 42.05%, 51.28%, and 52.79%, respectively, compared with the DC. The TLR4/NF-κB signaling pathway, as a therapeutic target for diabetic intestinal diseases, plays a role in regulating the inflammatory response and protecting the intestinal barrier function. Molecular mechanism studies showed that oral treatment with CCPH down-regulated the expression of NF-κB, TLR-4, and TNF-α genes by 18.66%, 21.58%, and 34.87%, respectively, while up-regulating the expression of ZO-1 and occludin genes by 32.70% and 25.11%, respectively. CCPH regulates the expression of short-chain fatty acid levels, increases microbial diversity, and ameliorates mouse colon lesions by inhibiting the TLR4/NF-κB signaling pathway. Conclusions: In conclusion, it is demonstrated that in this murine model, the treatment of diabetes with C. cicadae polysaccharide can effectively regulate intestinal microbiota imbalance, protect intestinal mucosal barrier function, and reduce inflammation in vivo, suggesting this natural product can provide a suitable strategy for the treatment of T2D-induced gut dysbiosis and intestinal health. Full article
(This article belongs to the Section Endocrinology and Clinical Metabolic Research)
Show Figures

Figure 1

Figure 1
<p>Quantification by cultivation of beneficial bacteria in the intestines of diabetic mice under various treatments (n = 10 per group), with Bifidobacterium (<b>A</b>), Lactobacillus (<b>B</b>), and Bacteroides (<b>C</b>). Data are presented as mean ± standard deviation (SD) values. Statistical significance is indicated as A (<span class="html-italic">p</span> &lt; 0.01) and a (<span class="html-italic">p</span> &lt; 0.05) vs. the normal control and as B (<span class="html-italic">p</span> &lt; 0.01) and b (<span class="html-italic">p</span> &lt; 0.05) vs. the diabetic control.</p>
Full article ">Figure 2
<p>Intestinal mucosal barrier indicators of the diabetic mice (n = 10 per group). (<b>A</b>) LPS serum levels and (<b>B</b>) lactic acid content. The compared statistical significance is given as a: <span class="html-italic">p</span> &lt; 0.05, A: <span class="html-italic">p</span> &lt; 0.01; compared with the DC group as B: <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Levels of intestinal antioxidant capacity (<span class="html-italic">n</span> = 10 per group). (<b>A</b>) CAT level, (<b>B</b>) SOD level, (<b>C</b>) T-AOC level. Compared with the NC group, a: <span class="html-italic">p</span> &lt; 0.05, A: <span class="html-italic">p</span> &lt; 0.01; compared with the DC group, B: <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Content of proinflammatory cytokines (<b>A</b>–<b>C</b>) in diabetic mice (n = 10 per group). Compared with the NC group, A: <span class="html-italic">p</span> &lt; 0.01; compared with the DC group, B: <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Venn diagram of OTU distribution.</p>
Full article ">Figure 6
<p>Distribution of microbial communities at the phylum level. (<b>A</b>–<b>D</b>) microbial species composition from the NC group, DC group, PC group and CCPH group.</p>
Full article ">Figure 7
<p>PCA analysis.</p>
Full article ">Figure 8
<p>HE (<b>left</b>) and AB-PAS (<b>right</b>) stained mouse colon sections. (<b>A</b>–<b>D</b>) Colon from the NC group, DC group, PC group, and CCPH group, respectively.</p>
Full article ">Figure 9
<p>mRNA expression levels of NF-kB, TLR-4, TNF-α, ZO-1, and occludin in the colon.</p>
Full article ">
16 pages, 1687 KiB  
Article
Preconditioning with Wound Fluid Enhances Immunosuppressive Properties of Mesenchymal Stromal Cells In Vitro
by Helena Moratin, Isabel Mache, Miguel Goncalves, Totta Ehret Kasemo, Manuel Stöth, Till Jasper Meyer, Stephan Hackenberg, Agmal Scherzad and Marietta Herrmann
Int. J. Mol. Sci. 2025, 26(1), 293; https://doi.org/10.3390/ijms26010293 - 31 Dec 2024
Viewed by 154
Abstract
Immunosuppression is one key feature of mesenchymal stromal cells (MSCs) that has high expectations for therapeutic use. The influence of pro-inflammatory stimuli can modify the characteristics of MSCs and enhance immunosuppressive properties. The local postoperative environment contains cytokines, MSCs, and immune cells in [...] Read more.
Immunosuppression is one key feature of mesenchymal stromal cells (MSCs) that has high expectations for therapeutic use. The influence of pro-inflammatory stimuli can modify the characteristics of MSCs and enhance immunosuppressive properties. The local postoperative environment contains cytokines, MSCs, and immune cells in high quantities, and their mutual influence is still unclear. Knowledge of in vivo processes is pivotal for potential therapeutic applications, and therefore, the aim of this study was to investigate the influence of wound fluid (WF) on the immunomodulatory potential of MSCs. CD4+ cells were co-cultured with native or WF-preconditioned MSCs for 5 days. CFSE staining revealed significant suppression of T cell proliferation after co-culture that was even more distinct in co-culture with WF-MSCs. The concentration of IDO-1, TGF-β1 and IFN-γ was higher while TNF-α was reduced in co-culture supernatants, indicating a transition to an anti-inflammatory milieu. In summary, the results provide evidence that the influence of WF alters the immunomodulatory potential of MSCs. These findings should serve as the basis for further investigations with a focus on T cell subpopulations. Full article
(This article belongs to the Special Issue Mesenchymal Stem Cells: Cross-Talk with the Microenvironment)
27 pages, 3932 KiB  
Article
Evaluation of the Anti-Amyloid and Anti-Inflammatory Properties of a Novel Vanadium(IV)–Curcumin Complex in Lipopolysaccharides-Stimulated Primary Rat Neuron-Microglia Mixed Cultures
by Georgios Katsipis, Sophia N. Lavrentiadou, George D. Geromichalos, Maria P. Tsantarliotou, Eleftherios Halevas, George Litsardakis and Anastasia A. Pantazaki
Int. J. Mol. Sci. 2025, 26(1), 282; https://doi.org/10.3390/ijms26010282 - 31 Dec 2024
Viewed by 193
Abstract
Lipopolysaccharides (LPS) are bacterial mediators of neuroinflammation that have been detected in close association with pathological protein aggregations of Alzheimer’s disease. LPS induce the release of cytokines by microglia and mediate the upregulation of inducible nitric oxide synthase (iNOS)—a mechanism also associated with [...] Read more.
Lipopolysaccharides (LPS) are bacterial mediators of neuroinflammation that have been detected in close association with pathological protein aggregations of Alzheimer’s disease. LPS induce the release of cytokines by microglia and mediate the upregulation of inducible nitric oxide synthase (iNOS)—a mechanism also associated with amyloidosis. Curcumin is a recognized natural medicine but has extremely low bioavailability. V-Cur, a novel hemocompatible Vanadium(IV)-curcumin complex with higher solubility and bioactivity than curcumin, is studied here. Co-cultures consisting of rat primary neurons and microglia were treated with LPS and/or curcumin or V-Cur. V-Cur disrupted LPS-induced overexpression of amyloid precursor protein (APP) and the in vitro aggregation of human insulin (HI), more effectively than curcumin. Cell stimulation with LPS also increased full-length, inactive, and total iNOS levels, and the inflammation markers IL-1β and TNF-α. Both curcumin and V-Cur alleviated these effects, with V-Cur reducing iNOS levels more than curcumin. Complementary insights into possible bioactivity mechanisms of both curcumin and V-Cur were provided by In silico molecular docking calculations on Aβ1-42, APP, Aβ fibrils, HI, and iNOS. This study renders curcumin-based compounds a promising anti-inflammatory intervention that may be proven a strong tool in the effort to mitigate neurodegenerative disease pathology and neuroinflammatory conditions. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Levels of amyloid precursor protein (APP) in mixed cultures of primary neuron-microglia, in the absence or presence of 0.1, 1, or 10 μg/mL of LPS (<b>a</b>,<b>b</b>). Effect of LPS (1 μg/mL) in the presence or absence of 2 μΜ curcumin or V-Cur complex on APP levels (<b>c</b>,<b>d</b>). Analysis performed with Western blotting. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used to verify equal loading. The density of the blots was semi-quantified with ImageJ 1.54. Results are presented as fractional changes in comparison with the control sample and are the mean (±SD) of three independent biological experiments. One-way ANOVA was employed to compare untreated or LPS-treated samples. Statistical significance when compared with: * untreated sample (control); # LPS-treated sample; <span>$</span> curcumin-treated: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001; #### <span class="html-italic">p</span> &lt; 0.0001; <span>$</span><span>$</span><span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Docking poses orientation of curcumin and V-Cur in the crystal structure of the Kunitz protease inhibitor domain (APPI) of APP (PDB accession number 1AAP). The target protein is illustrated as a semi-transparent cartoon and surface colored in yellow orange and chocolate (chains A and B, respectively), while curcumin and V-Cur molecules are rendered in ball-and-stick mode and colored according to atom type in light pink and violet purple C atoms, respectively. The ligand binding site of both molecules depicting the architecture of the binding interactions is also illustrated (in the upper part) with an additional depiction of selected contacting amino acid residues of the binding pocket rendered in line and colored according to the cartoon. Binding interactions are illustrated in light pink (for curcumin) and violet (for V-Cur). Heteroatom color code: V: grey, N: blue, and O: red. Molecular docking simulations of both ligands were performed individually. Hydrogen atoms are omitted for clarity. The final structure was ray-traced and illustrated with the aid of PyMol Molecular Graphics.</p>
Full article ">Figure 3
<p>In vitro fibrillation assay with insulin in the presence of several concentrations (0–100 μΜ) of either curcumin (■) or V-Cur (●). The insulin amyloid fibers formed in the absence or presence of either curcumin or V-Cur were semi-quantified by employing Thioflavin T fluorescence, with excitation at 450 nm and recording the emission spectrum at 490 nm. The results from three independent experiments are provided as mean normalized fibrillization rates (±SEM), setting the value of the control sample as 100%. Some error bars are not visible due to very small values (&lt;1%).</p>
Full article ">Figure 4
<p>Docking pose orientation of curcumin and V-Cur in the crystal structure of dimer (PDB ID 1GUJ) and hexamer (PDB ID 6GNQ) HΙ target proteins. In the hexameric structure of HI are also illustrated the six chain-stabilizing Zn<sup>2+</sup> ions, the co-crystallized meta-cresol (CRS, depicted in gold sticks), and some critical to self-assembly and aggregation resides of HI (represented in stick mode colored in orange). Both HI proteins’ structures are depicted as cartoon colored in wheat and firebrick for A and B chains, respectively. Curcumin and V-Cur are rendered in sphere representation colored according to atom type in light pink and violet purple, respectively. The two Zn ions co-crystallized in the hexameric structure are depicted in sphere representation in lemon color and are shown to be connected with polar contact with Nε2 of His10 in the three double chains (A and B). Heteroatom color code: V: grey, N: blue, and O: red. Molecular docking simulations of both ligands were performed individually. Hydrogen atoms are omitted for clarity. The final structure was ray-traced and illustrated with the aid of PyMol Molecular Graphics.</p>
Full article ">Figure 5
<p>Levels of (<b>a</b>) active inducible NO synthase (iNOS) (100 kDa), (<b>b</b>) inactive iNOS (50 and 75 kDa), and (<b>c</b>) total iNOS levels, after 24 h of treatment with LPS 1 μg/mL, in the presence or absence of 2 μΜ of curcumin or V-Cur complex, in mixed cultures of primary neurons-microglia. iNOS levels were determined by Western blotting (<b>d</b>). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was employed to verify equal loading. The density of the blots was semi-quantified with ImageJ 1.54. Results are presented as fractional changes in comparison with the control sample and are the mean (±SD) of three independent biological experiments. One-way ANOVA was employed to compare untreated or LPS-treated samples. Statistical significance when compared with: * untreated sample (control); # LPS-treated sample; <span>$</span> curcumin-treated: * <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; # <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; <span>$</span> <span class="html-italic">p</span> &lt; 0.05; <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Docking pose orientation of curcumin and V-Cur in the crystal structure of iNOS monomer enzyme (PDB accession number 4NOS). The target protein is illustrated as cartoon colored in the sand along with a semi-transparent surface colored in the dark sand. Curcumin and V-Cur molecules, as well as the co-crystallized iNOS inhibitor ethylisothiourea (ITU) are rendered in sphere mode and colored according to atom type in light pink, violet purple, and hot pink C atoms, respectively. The co-crystallized molecules heme (HEM) (iron protoporphyrin IX) and H2B superimposed with the docked molecules are rendered in stick representation and colored according to atom type in orange and yellow-orange C atoms, respectively. H4B, essential for the dimerization of the protein, is not shown since it is located farther down the binding cavity, near the dimerization interface. The target protein structure model in the lower panel, depicting in a close-up view of the binding cavity of the target enzyme the architecture of the binding interactions, is illustrated as a semi-transparent surface colored in dark sand with an additional depiction of selected contacting amino acid residues of the binding pocket highlighted in the molecular surface in smudge green (for V-Cur) and white (for curcumin). Binding interaction residues are labeled in white (for curcumin) and smudge green (for V-Cur). Heteroatom color code: V: grey, N: blue, and O: red. Molecular docking simulations of both ligands were performed individually. Hydrogen atoms are omitted for clarity. The final structure was ray-traced and illustrated with the aid of PyMol Molecular Graphics.</p>
Full article ">Figure 7
<p>Levels of (<b>a</b>) tumor necrosis factor-α (TNF-α), and (<b>b</b>) interleukin-1β (IL-1β), in mixed cultures of primary neurons-microglia after treatment with LPS (1 μg/mL) in the presence or absence of 2 μΜ curcumin or V-Cur. Cytokine levels were determined with Western blotting (<b>c</b>,<b>d</b>). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was employed to verify equal loading. The density of the blots was semi-quantified with ImageJ 1.54. Results are presented as fractional changes in comparison with the control sample and are the mean (± SD) of three independent biological experiments. One-way ANOVA was employed to compare untreated or LPS-treated samples. Statistical significance when compared with: * untreated sample (control); # LPS-treated sample; <span>$</span> curcumin-treated: ** <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; # <span class="html-italic">p</span> &lt; 0.05; #### <span class="html-italic">p</span> &lt; 0.0001; <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
14 pages, 3714 KiB  
Article
Experimental Validation of Antiobesogenic and Osteoprotective Efficacy of Ginsenoside CK via Targeting Lipid and Atherosclerosis Pathways
by Md. Niaj Morshed, Reshmi Akter, Imran Mahmud, Ah-Yeong Gwon, Jin Woo Jeang, Yeong-Geun Lee, Dae Won Park, Deok Chun Yang, Yeon Ju Kim and Se-Chan Kang
Life 2025, 15(1), 41; https://doi.org/10.3390/life15010041 - 31 Dec 2024
Viewed by 209
Abstract
The present study explored the possible antiobesogenic and osteoprotective properties of the gut metabolite ginsenoside CK to clarify its influence on lipid and atherosclerosis pathways, thereby validating previously published hypotheses. These hypotheses were validated by harvesting and cultivating 3T3-L1 and MC3T3-E1 in adipogenic [...] Read more.
The present study explored the possible antiobesogenic and osteoprotective properties of the gut metabolite ginsenoside CK to clarify its influence on lipid and atherosclerosis pathways, thereby validating previously published hypotheses. These hypotheses were validated by harvesting and cultivating 3T3-L1 and MC3T3-E1 in adipogenic and osteogenic media with varying concentrations of CK. We assessed the differentiation of adipocytes and osteoblasts in these cell lines by applying the most effective doses of CK that we initially selected. Using 3T3-L1 adipocytes in vitro assessments, CK could effectively decrease intracellular lipid accumulation, inhibit α-glucosidase enzyme, increase 2-NBDG glucose uptake, reduce inflammation-associated cytokines (TNFα, and IL-6), adipogenic regulatory genes (PPARγ, FAS, C/EBPα), lipogenic gene LPL, and increase the expression of thermogenic gene UCP1. Additionally, CK treatment induced osteoblast development in MC3T3-E1 cells as shown by increased mineralization and calcium distribution, collagen content, alkaline phosphatase activity, and decreased inflammatory cytokines TNFα, and IL-6 and increased the regulated expressions of osteogenic genes including Runx2, ALP, BGLAP, OCN, and Col1a1. Significantly, as a major inhibitory regulator, the TP53 gene was down-regulated in both 3T3-L1 and MC3T3E1 cells after the treatment of CK. These encouraging results demonstrate the possible use of CK as an innovative treatment for controlling obesity and osteoporosis, targeting the underlying mechanisms of obesogenic and bone loss. Further studies are necessary to explore the clinical implications of these results and the potential of CK in future treatment strategies. This research highlights the promise of CK in addressing significant health issues. Full article
(This article belongs to the Section Pharmaceutical Science)
Show Figures

Figure 1

Figure 1
<p>The MTT assay was used to measure cell viability. CK (3.125–50 μM) was administered to (<b>A</b>) 3T3-L1 pre-adipocyte (Orange-colored columns) and (<b>B</b>) MC3T3-E1 pre-osteoblast cells (1 × 10<sup>4</sup> cells/well) (Tan-colored columns) for 24 h. A two-tailed Student’s <span class="html-italic">t</span>-test was used to determine whether there was a substantial distinction between the groups; ns denotes a non-significant change, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 when compared to a control group (Black-colored column) that was not treated.</p>
Full article ">Figure 2
<p>The inhibitory effect of CK on lipid accumulation in MDI-induced 3T3-L1 adipocytes was assessed as follows: (<b>A</b>) Oil Red O staining was performed to visualize fat droplets, which were then observed under a light microscope at 20× magnification. (<b>B</b>) Lipid accumulation was quantified by measuring the absorbance of Oil Red O dissolved in isopropyl alcohol at 520 nm. Data represent the mean ± SEM from three independent experiments. Statistical significance is indicated as ### <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, compared to the MDI-treated group.</p>
Full article ">Figure 3
<p>(<b>A</b>) The proportion of a-glucosidase inhibition at various CK concentrations. Acarbose, or ACR, was employed as a control. We also analyzed the half-maximum inhibitory concentration (IC<sub>50</sub>) of CK and ACR (<b>B</b>) 3T3L1 cells using the 2-NBDG uptake test. Using a fluorescent derivative of glucose 2-NBDG, the impact of CK on 3T3L1 cells’ glucose absorption was examined for 24 h with and without CK. Insulin was employed as a positive control at 100 nM. Data are presented as a control percentage. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>The relative expression of inflammatory genes (<span class="html-italic">TNFα</span>, <span class="html-italic">IL-6</span>), adipogenic genes (<span class="html-italic">PPARγ</span>, <span class="html-italic">C/EBPα</span>, <span class="html-italic">LPL</span>), the lipogenic gene <span class="html-italic">FAS</span>, and the thermogenic gene <span class="html-italic">UCP1</span>, along with <span class="html-italic">TP</span><sup>53</sup>, was measured in differentiated 3T3-L1 cells treated with CK at concentrations of 2.5 μM and 10 μM, or RSG at 20 μM. Results are presented as the mean ± standard deviation from three independent experiments. Statistical significance was assessed using a two-tailed Student’s <span class="html-italic">t</span>-test. Significant differences in gene expression between untreated and treated groups are indicated as ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>(<b>A</b>) ALP activity in CK and E<sub>2</sub>-treated differentiated MC3T3-E1 cells. (<b>B</b>) The impact of CK on MC3T3-E1 cell mineralization. A 96-well plate was used to measure the quantity of Alizarin Red S at 562 nm. (<b>C</b>) The calcium-binding Alizarin Red S dye was used to evaluate the calcium deposits in the extracellular matrix for matrix mineralization. Up to Day 28, CK treatment accelerated the mineralization of the extracellular matrix. The images are representative of over three different concentrations of CK and E<sub>2.</sub> The results are presented as the mean ± standard deviation from three independent experiments. Statistical significance was determined using a two-tailed Student’s <span class="html-italic">t</span>-test. Significant differences in lipid production compared to the untreated (control) group are denoted by ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Collagen content in MC3T3-E1 cells is increased by CK extracts. For 12 days, cells were exposed to extracts at doses ranging from 2.5 to 10 μM, either with or without a differentiation medium. E<sub>2</sub> was used at a concentration of 100 nm. (<b>A</b>) Picro-Sirius red staining was carried out and seen using a microscope (magnification of ×100). (<b>B</b>) Absorbance was measured at 550 nm to determine the amount of collagen. The presented data are the mean ± standard deviation (SD) of three studies. Statistical analysis revealed significant differences, denoted as * <span class="html-italic">p</span> &lt; 0.1, *** <span class="html-italic">p</span> &lt; 0.001 when compared with the indicated ascorbic acid and β-glycerophosphate treated group.</p>
Full article ">Figure 7
<p>Relative expression of proinflammatory cytokines TNFα, and IL-6, and osteogenic genes <span class="html-italic">RunX2</span>, <span class="html-italic">ALP</span>, <span class="html-italic">BGLAP</span>, <span class="html-italic">OCN</span>, and <span class="html-italic">COL1a1</span> with <span class="html-italic">TP</span><sup>53</sup> in differentiated MC3T3-E1 cells on treatment with CK at 2.5 μM and E<sub>2</sub> at 100 nm concentrations, respectively. A two-tailed Student’s <span class="html-italic">t</span>-test was used to determine whether the difference was statistically significant. The non-treated and treated groups’ significant differences in gene expression are indicated by * <span class="html-italic">p</span> &lt; 0.1, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. control.</p>
Full article ">
16 pages, 2882 KiB  
Article
Blocking TNF-α Reduces Leishmania major-Induced Hyperalgesia and Changes the Cytokine Profile in the Paw Skin of BALB/c Mice with a Potential Positive Effect on Parasite Clearance
by Muriel Tahtouh Zaatar, Sara Salman, Reem Hoblos, Rabih Roufayel, Ziad Fajloun, Jean-Marc Sabatier and Marc Karam
Microbiol. Res. 2025, 16(1), 8; https://doi.org/10.3390/microbiolres16010008 (registering DOI) - 31 Dec 2024
Viewed by 158
Abstract
The course and outcome of infection with the parasitic protozoa Leishmania major depends on the host immune response which, itself, depends mainly on the cytokine milieu, especially early in the infection. It is widely accepted that INF-γ, TNF-α, and IL-12 usually favor a [...] Read more.
The course and outcome of infection with the parasitic protozoa Leishmania major depends on the host immune response which, itself, depends mainly on the cytokine milieu, especially early in the infection. It is widely accepted that INF-γ, TNF-α, and IL-12 usually favor a protective response, while IL-4, IL-5, IL-10, and IL-13 favor a pathogenic one. These and other cytokines also play a major role in Leishmania-induced hyperalgesia via two possible pathways, one involving prostaglandins and the other sympathetic amines as final mediators, preceded by a cascade of cytokines, among which TNF-α seems to play a pivotal role via a still unclear mechanism of action. This study investigates the effects of anti-TNF-α antibody (Infliximab) on L. major-induced hyperalgesia in susceptible BALB/c mice using the hot plate and tail flicks tests, as well as the levels of many cytokines in the infected paws of mice using the ELISA technique. In addition, the parasite burden was assessed using the serial dilution method. Our results show that Infliximab can reduce the induced hyperalgesia, up-regulate TNF-α, IL-1β, and keratinocyte-derived chemokine (KC), and down-regulate IL-10 and IL-17 in the paws of infected mice. Infliximab may also have beneficial effects on the prognosis of cutaneous leishmaniasis by reducing the parasite burden. Full article
Show Figures

Figure 1

Figure 1
<p>Timeline of <span class="html-italic">L. major</span> Infection, Infliximab injections, and tissue sampling for ELISA analysis; n = 5 mice in each group.</p>
Full article ">Figure 2
<p>The effect of Infliximab on pain thresholds in non-infected mice as assessed by the hot plate (<b>A</b>) and tail flick (<b>B</b>) tests. Mice were injected intraperitoneally with different doses of Infliximab. Hot plate and tail flick tests were performed on a daily basis. Results are displayed as mean of 5 mice ± SEM and the degree of significance was calculated with reference to the control (naive).</p>
Full article ">Figure 3
<p>The time course of the effect of Infliximab (5 mg/kg) on <span class="html-italic">L. major</span> (high dose)-induced hyperalgesia as assessed by the hot plate (<b>A</b>) and tail flick (<b>B</b>) tests. The infected mice were treated (i.p.) with Infliximab on days 0, 4, 7, and 10 post-infection. Each result is the mean of 5 mice ± SEM and the degree of significance was calculated with reference to the uninfected mice (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>The time course of the effect of Infliximab (10 mg/kg) on <span class="html-italic">L. major</span> (high dose)-induced hyperalgesia as assessed by the hot plate (<b>A</b>) and tail flick (<b>B</b>) tests. The infected mice were treated (i.p.) with Infliximab on days 0, 4, 7, and 10 post-infection. Each result is the mean of 5 mice ± SEM and the degree of significance was calculated with reference to the uninfected mice (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 5
<p>The time course of the effect of Infliximab (20 mg/kg) on <span class="html-italic">L. major</span> (high dose)-induced hyperalgesia as assessed by the hot plate (<b>A</b>) and tail flick (<b>B</b>) tests. The infected mice were treated (i.p.) with Infliximab on days 0, 4, 7, and 10 post-infection. Each result is the mean of 5 mice ± SEM and the degree of significance was calculated with reference to the uninfected mice (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 6
<p>The effect of Infliximab (10 mg/kg) on TNF-α (<b>A</b>), IL-10 (<b>B</b>), IL-17 (<b>C</b>), IL-1β (<b>D</b>), IFN-γ (<b>E</b>), and KC (<b>F</b>) levels in the paws of mice infected with a high dose of <span class="html-italic">L. major</span>. Mice were injected (i.p.) with Infliximab on days 0, 4, 7, and 10 post-infection. Each result is the mean of 5 mice ± SEM and the degree of significance was calculated with reference to infected but non-Infliximab-treated mice (# <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01) and with reference to the uninfected control mice (* <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). Asterisks (*) denote comparisons between uninfected and infected groups, while hashes (#) indicate comparisons between infected and Infliximab-treated groups. The ## in the figure indicates a significance level of <span class="html-italic">p</span> &lt; 0.01 in comparisons between infected and Infliximab-treated groups.</p>
Full article ">
20 pages, 4034 KiB  
Article
Nicotinamide Mononucleotide Restores NAD+ Levels to Alleviate LPS-Induced Inflammation via the TLR4/NF-κB/MAPK Signaling Pathway in Mice Granulosa Cells
by Mehboob Ahmed, Umair Riaz, Haimiao Lv, Muhammad Amjad, Sohail Ahmed, Shaokat Ali, Muhammad Usman Ghani, Guohua Hua and Liguo Yang
Antioxidants 2025, 14(1), 39; https://doi.org/10.3390/antiox14010039 - 31 Dec 2024
Viewed by 249
Abstract
Inflammation disrupts the normal function of granulosa cells (GCs), which leads to ovarian dysfunction and fertility decline. Inflammatory conditions such as polycystic ovary syndrome (PCOS), primary ovarian insufficiency (POI), endometriosis, and age-related ovarian decline are often associated with chronic low-grade inflammation. Nicotinamide mononucleotide [...] Read more.
Inflammation disrupts the normal function of granulosa cells (GCs), which leads to ovarian dysfunction and fertility decline. Inflammatory conditions such as polycystic ovary syndrome (PCOS), primary ovarian insufficiency (POI), endometriosis, and age-related ovarian decline are often associated with chronic low-grade inflammation. Nicotinamide mononucleotide (NMN) is an important precursor of NAD+ and has gained attention for its potential to modulate cellular metabolism, redox homeostasis, and mitigate inflammation. This study investigated the protective roles of NMN against lipopolysaccharide LPS-mediated inflammation in GCs. The results of this experiment demonstrated that LPS had negative effects on GCs in term of reduced viability and proliferation rates and upregulated the production of pro-inflammatory cytokines, including interleukin-1 beta (IL-1β), interleukin-6 (IL-6), cyclooxygenase-2 (Cox-2), and tumor necrosis factor-alpha (TNF-α). Notably, the levels of NAD+ and NAD+/NADH ratio in GCs were reduced in response to inflammation. On the other hand, NMN supplementation restored the NAD+ levels and the NAD+/NADH ratio in GCs and significantly reduced the expression of pro-inflammatory markers at both mRNA and protein levels. It also enhanced cell viability and proliferation rates of GCs. Furthermore, NMN also reduced apoptosis rates in GCs by downregulating pro-apoptotic markers, including Caspase-3, Caspase-9, and Bax while upregulating anti-apoptotic marker Bcl-2. NMN supplementation significantly reduced reactive oxygen species ROS and improved steroidogenesis activity by restoring the estradiol (E2) and progesterone (P4) levels in LPS-treated GCs. Mechanistically, this study found that NMN suppressed the activation of the TLR4/NF-κB/MAPK signaling pathways in GCs, which regulates inflammatory processes. In conclusion, the findings of this study revealed that NMN has the potential to reduce LPS-mediated inflammatory changes in GCs by modulating NAD+ metabolism and inflammatory signaling pathways. NMN supplementation can be used as a potential therapeutic agent for ovarian inflammation and related fertility disorders. Full article
(This article belongs to the Section Antioxidant Enzyme Systems)
Show Figures

Figure 1

Figure 1
<p>Effects of LPS on granulosa cells viability, proliferation, mRNA expression, and protein levels of pro-inflammatory markers. (<b>A</b>) Percentage of granulosa cell (GC) viability following treatment with varying concentrations of LPS (0.1, 1, 5, 10, 20, and 100 µg/mL) in culture media, showing a significant reduction at concentrations ≥ 10 µg/mL. (<b>B</b>) GC proliferation presented as the number of cells, with a significant decrease observed at 10 µg/mL LPS and higher. (<b>C</b>) mRNA expression of TLR4, IL-1β, IL-6, Cox-2, and TNF-α analyzed by qPCR, demonstrating significant upregulation in LPS-treated GCs compared to the control. (<b>D</b>) Western blot representative images and analysis showing increased protein expression of TLR4, phosphorylated NF-κB (p-NF-κB), COX-2, and TNF-α in response to LPS treatment, while GAPDH serves as the loading control. Values are expressed as mean ± SEM. Compared to the control group, * <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.</p>
Full article ">Figure 2
<p>Effects of LPS on the NAD<sup>+</sup> levels, NADH levels, and NAD<sup>+</sup>/NADH ratio in granulosa cells. (<b>A</b>) The NAD<sup>+</sup> and NADH levels were measured in GCs (~1 × 10<sup>6</sup> cells). (<b>A</b>) NAD<sup>+</sup> levels in GCs after LPS treatment, showing a significant decrease compared to the control. (<b>B</b>) The NADH levels remained unchanged between the control and LPS-treated groups, indicating no significant effect of LPS on the NADH level. (<b>C</b>) The NAD<sup>+</sup>/NADH ratio in GCs, demonstrating a significant reduction in LPS-treated cells compared to the control. These data illustrate the metabolic shift induced by LPS in GCs, primarily driven by a reduction in the NAD<sup>+</sup> levels and the NAD<sup>+</sup>/NADH ratio. Values are expressed as mean ± SEM. Compared to the control group, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Effects of NMN supplementation on LPS-treated granulosa cells viability, proliferation, mRNA expression, and protein levels of pro-inflammatory markers. (<b>A</b>) Percentage of cell viability following treatment with varying concentrations of NMN (1, 10, 100, 500, 1000, and 2000 µM) in LPS-treated GCs, showing a significant increase at 100 µM, with the highest viability observed at 500 and 1000 µM. (<b>B</b>) GC proliferation presented as the number of cells, with a significant improvement observed at 100 µM and the highest rates observed at 500 and 1000 µM of NMN in LPS-treated GCs. (<b>C</b>) mRNA expression of IL-1β, IL-6, Cox-2, and TNF-α analyzed by qPCR, showing significant upregulation in LPS-treated GCs compared to the control, and a marked reduction in the LPS + NMN group as compared to LPS-treated GCs (<b>D</b>) Western blot representative images and analysis showing a reduced protein expression of Cox-2 and TNF-α in LPS + NMN-treated GCs compared to the LPS group. GAPDH was used as the loading control. Values are expressed as mean ± SEM. ns = non-significant; <span class="html-italic">p</span> ≥ 0.05, * <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.</p>
Full article ">Figure 4
<p>Effects of NMN supplementation on the NAD<sup>+</sup> levels, NADH levels, and NAD<sup>+</sup>/NADH ratio in granulosa cells. The NAD<sup>+</sup> and NADH levels were measured in GCs (~1 × 10<sup>6</sup> cells) across four groups: Control, LPS, NMN, and LPS + NMN. (<b>A</b>) The NAD<sup>+</sup> levels significantly decreased in LPS-treated GCs compared to the controls, while NMN supplementation restored the NAD<sup>+</sup> levels to baseline. (<b>B</b>) The NADH levels showed no significant differences across the experimental groups. (<b>C</b>) The NAD<sup>+</sup>/NADH ratio, reduced by LPS treatment, was significantly restored with NMN supplementation. (<b>D</b>) The mRNA expression levels of apoptosis-related markers were assessed by qPCR. LPS treatment significantly upregulated the pro-apoptotic markers, including Caspase-3 (Cas-3), Caspase-9 (Cas-9), and Bax, while reducing the expression of the anti-apoptotic marker Bcl-2 compared to the control group. NMN supplementation (LPS + NMN) significantly reduced the expression of Cas-3, Cas-9, and Bax while restoring Bcl-2 expression in LPS-treated GCs. (<b>E</b>) Western blot analysis confirmed the protein-level changes in the apoptosis markers. LPS treatment increased the protein expression of Caspase-3 and Bax and reduced Bcl-2 expression compared to the control group. NMN supplementation significantly decreased the Caspase-3 and Bax levels and increased Bcl-2 protein expression in LPS-treated GCs. Data are presented as mean ± SEM with significance levels indicated: ns = non-significant; <span class="html-italic">p</span> ≥ 0.05, * <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.</p>
Full article ">Figure 5
<p>Effects of NMN supplementation on the ROS levels and steroidogenesis activity in granulosa cells. (<b>A</b>) Intracellular ROS levels were measured in GCs to assess oxidative stress. LPS-treated GCs showed a significant increase in ROS compared to the control group, while NMN supplementation reduced ROS in the LPS + NMN group, indicating protective effects against oxidative stress. (<b>B</b>,<b>C</b>) The levels of estradiol (E2) and progesterone (P4) in GCs were also evaluated. LPS treatment significantly reduced E2 and P4 production, whereas NMN supplementation restored their levels to the control values, supporting steroidogenic function. Values are presented as mean ± SEM, with significance levels indicated: ns = non-significant; <span class="html-italic">p</span> ≥ 0.05, * <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.</p>
Full article ">Figure 6
<p>Effects of NMN on TLR4/NF-κB p65/MAPK pathway activation in granulosa cells. Western blot analysis shows that LPS treatment increased the expression of TLR4 (relative to GAPDH), p-NF-κB p65 (relative to NF-κB p65), p-ERK1/2 (relative to ERK1/2), p-JNK (relative to JNK), and p-P38 (relative to GAPDH) compared to the control group, while NMN supplementation in LPS-treated GCs reduced the expression of these proteins, indicating that NMN alleviates LPS-induced inflammatory signaling through the TLR4/NF-κB p65/MAPK pathway. Values are expressed as mean ± SEM; with significance levels indicated: ns = non-significant; <span class="html-italic">p</span> ≥ 0.05, * <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.</p>
Full article ">Figure 7
<p>Graphical illustration of TLR4/NF-κB p65/MAPK pathway activation and inhibition by NMN. This illustration depicts the activation of the TLR4/NF-κB p65 and MAPK pathways in granulosa cells in response to LPS. LPS binding to TLR4 triggers downstream signaling, leading to NF-κB p65 phosphorylation and MAPK pathway activation, as indicated by the increased p-ERK1/2, p-JNK, and p-P38. The ROS levels and apoptosis are also increased in response to LPS-mediated TLR4 activation. NMN supplementation counteracts this activation by increasing the NAD+ levels and reducing TLR4 signaling and subsequently downregulating NF-κB p65 and MAPK phosphorylation, effectively mitigating the inflammatory responses, ROS production, and apoptosis markers. This graphical overview highlights the anti-inflammatory and anti-apoptotic roles of NMN and NAD+ in cellular signaling.</p>
Full article ">
19 pages, 6005 KiB  
Article
Dapsone Alters Phenotypical and Functional Properties of Human Neutrophils In Vitro
by Sara Rakočević, Vanja Mališ, Ljiljana Kozić, Anđela Dubovina, Marija Drakul, Dejan Bokonjić, Miodrag Čolić and Dušan Mihajlović
Molecules 2025, 30(1), 113; https://doi.org/10.3390/molecules30010113 - 30 Dec 2024
Viewed by 250
Abstract
Dapsone is a sulfone used in treating inflammatory skin conditions. Despite its widespread dermatological use, the pharmacological actions of dapsone remain poorly understood. Here, we examined how different aspects of neutrophil functions are affected by dapsone. Peripheral blood neutrophils from healthy donors were [...] Read more.
Dapsone is a sulfone used in treating inflammatory skin conditions. Despite its widespread dermatological use, the pharmacological actions of dapsone remain poorly understood. Here, we examined how different aspects of neutrophil functions are affected by dapsone. Peripheral blood neutrophils from healthy donors were stimulated with phorbol-12-myristate-13-acetate (PMA), N-formyl-L-methionyl-L-leucyl-L-phenylalanine (fMLP), or calcium ionophore (CaI) or primed with cytokines prior to stimulation, in the presence of different concentrations of dapsone (from 10 to 50 µg/mL), followed by analyses of their survival, phenotype, and functional properties. We found that dapsone at the concentration of 50 μg/mL induced a significant neutrophil apoptotic rate during 6 h and 18 h, while other concentrations were well tolerated compared to control non-treated cells. However, dapsone significantly decreased the induced oxidative burst of neutrophils at all non-cytotoxic concentrations. Additionally, dapsone showed a dose-dependent suppression of NETosis in activated neutrophils. The production of IL-8 by dapsone-treated neutrophils was decreased under both stimulated (fMLP) and primed (TNF-α/fMLP) conditions. Moreover, dapsone inhibited the expression of CD11b/CD18, CD66, and CD89 and reversed or significantly mitigated the downregulation of CD16, CD32, CD181, CD88, and CD62L on neutrophils after priming and fMLP stimulation. In conclusion, our results indicate the complexity of dapsone actions on neutrophil functions, extending previous knowledge on the suppression of oxidative burst and IL-8 production upon neutrophils’ activation. Suppressed NETosis and modulation of marker expression associated with different neutrophil functions under inflammatory conditions are new findings, not recognized previously. Full article
(This article belongs to the Special Issue New Strategies for Drug Development)
Show Figures

Figure 1

Figure 1
<p>Cytotoxic effect of dapsone on neutrophils. Neutrophils were cultivated alone or with different concentrations of dapsone as indicated for 6 h. Cytotoxicity was evaluated using an Annexin V-APC/Propidium Iodide apoptosis/necrosis assay. (<b>A</b>) The summarized results showing % of apoptosis. Values are given as mean ± SD from 3 independent experiments carried out with different donors. ** <span class="html-italic">p</span> &lt; 0.01 compared to the corresponding control (dapsone-untreated) group. (<b>B</b>) Representative plots of early apoptosis (AnnexineV+), late apoptosis/secondary necrosis (AnnexinV+/Pi+), and primary necrosis (PI+) of neutrophils from one representative experiment. The doublets and the death (FCS low) cells were gated out (see <a href="#app1-molecules-30-00113" class="html-app">Supplementary Figure S1</a>), and the quadrants were set according to the single-labeled samples.</p>
Full article ">Figure 2
<p>Cytotoxic effect of dapsone on neutrophils. Neutrophils were cultivated alone or with different concentrations of dapsone for 18 h. Cell viability was evaluated by apoptosis/necrosis assay by flow cytometry. Values are given as mean ± SD from 3 independent experiments carried out with different donors. * <span class="html-italic">p</span> &lt; 0.05 compared to corresponding control groups (non-treated neutrophils).</p>
Full article ">Figure 3
<p>Effect of dapsone on ROS production (<b>A</b>,<b>B</b>). Neutrophils were cultured alone or with dapsone as described in the Materials and Methods section. ROS production was evaluated by measuring luminescence intensity in real time. The values are shown as the area under the curve (AUC). (<b>A</b>) A representative histogram showing real-time ROS production. (<b>B</b>) Summarized results are presented as mean ± SD from 4 independent experiments carried out with different donors. * <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 compared with corresponding control (dapsone-untreated) groups.</p>
Full article ">Figure 4
<p>Effect of dapsone on NETosis. Neutrophils were treated with two concentrations of dapsone and stimulated for 4 h. Afterward, the cells were stained with the fluorescent dye Syto Green to measure NETosis by fluorescence intensity. The results were presented as a percentage of NETosis. Values are given as mean ± SD from 3 independent experiments carried out with different donors. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to the corresponding control groups, as indicated.</p>
Full article ">Figure 5
<p>Effect of dapsone on the secretion of IL8. After incubation with two concentrations of dapsone (10 μg/mL and 40 μg/mL), neutrophils were either left resting or stimulated with fMLP or primed with TNF-α before stimulation. The levels of IL-8 in culture supernatants were measured by a commercial ELISA kit. The summarized results are presented as mean ± SD from 5 independent experiments carried out with different donors. * <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 between the indicated samples.</p>
Full article ">Figure 6
<p>Effect of dapsone on the neutrophil phenotype. After incubation with dapsone (10 μg/mL and 40 μg/mL) neutrophils were activated with fMLP or primed with cytokines and then subsequently stimulated with fMLP. (<b>A</b>) The expressions of CD11b and CD18 are represented as mean fluorescence intensity (MFI) (mean ± SD from 3 independent experiments). (<b>B</b>) The representative histograms from one experiment. * <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 compared with corresponding controls as indicated. C = control, unstimulated neutrophils.</p>
Full article ">Figure 7
<p>Effect of dapsone on the phenotype of primed neutrophils. After incubation with dapsone (10 μg/mL and 40 μg/mL), neutrophils were activated with fMLP or primed with cytokines and then subsequently stimulated with fMLP. (<b>A</b>) The expressions of CD89 and CD66 are represented as mean fluorescence intensity (MFI) (mean ± SD from 3 independent experiments). (<b>B</b>) The representative histograms from one experiment. * <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 compared with corresponding controls as indicated. C = control, unstimulated neutrophils.</p>
Full article ">Figure 8
<p>Effect of dapsone on the neutrophil phenotype. After incubation with dapsone (10 μg/mL and 40 μg/mL), neutrophils were activated with fMLP or primed with cytokines and then subsequently stimulated with fMLP. (<b>A</b>) The expressions of CD16 and CD32 are represented as mean fluorescence intensity (MFI) (mean ± SD from 3 independent experiments). (<b>B</b>) The representative histograms from one experiment. * <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 compared with corresponding controls as indicated. C = control, unstimulated neutrophils.</p>
Full article ">Figure 9
<p>Effect of dapsone on the neutrophil phenotype. After incubation with dapsone (10 μg/mL and 40 μg/mL), neutrophils were activated with fMLP or primed with cytokines and then subsequently stimulated with fMLP. (<b>A</b>) The expressions of CD181 and CD88 are represented as mean fluorescence intensity (MFI) (mean ± SD from 3 independent experiments). (<b>B</b>) The representative histograms from one experiment. * <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 compared with corresponding controls as indicated. C = control, unstimulated neutrophils.</p>
Full article ">Figure 10
<p>Effect of dapsone on the neutrophil phenotype. After incubation with dapsone (10 μg/mL and 40 μg/mL), neutrophils were activated with fMLP or primed with cytokines and then subsequently stimulated with fMLP. (<b>A</b>) The expression of CD62L is represented as mean fluorescence intensity (MFI) (mean ± SD from 3 independent experiments). (<b>B</b>) The representative histograms from one experiment. * <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 compared with corresponding controls as indicated. C = control, unstimulated neutrophils.</p>
Full article ">
10 pages, 653 KiB  
Article
Cytokine and Chemokine Responses of Peripheral Blood Mononuclear Cells from Dogs Infected with Mycobacterium bovis
by Tyler Morrison, Danielle A. Gunn-Moore, Jayne C. Hope and Conor O’Halloran
Pathogens 2025, 14(1), 17; https://doi.org/10.3390/pathogens14010017 - 30 Dec 2024
Viewed by 195
Abstract
Mycobacterial infections are an important emerging zoonosis in companion animals for which diagnostic options remain imperfect, and the canine immunological response to these infections has been poorly investigated. We sought to further define the cellular response of peripheral blood mononuclear cells (PBMCs) from [...] Read more.
Mycobacterial infections are an important emerging zoonosis in companion animals for which diagnostic options remain imperfect, and the canine immunological response to these infections has been poorly investigated. We sought to further define the cellular response of peripheral blood mononuclear cells (PBMCs) from dogs infected with Mycobacterium bovis, as determined using a commercial interferon-gamma response assay (IGRA). To this end, PBMCs from healthy or infected dogs were collected. Serum samples were tested to further classify dogs as seropositive or seronegative for circulating antibodies against M. bovis using the DPP® VetTB Assay, Idexx M. bovis antibody ELISA, and a novel purified protein derivative ELISA. Isolated PBMCs were stimulated with mycobacterial proteins (PPDB or ESAT-6/CFP-10), and 13 cytokines/chemokines were measured in the supernatant. These concentrations were determined using the CYTOMAG-90K MILLIPLEX MAP Canine Cytokine/Chemokine system. PBMCs from infected dogs released IFN-γ in response to stimulation, but this response was reduced in those that had seroconverted. Similarly, cells stimulated with PPDB secreted increased amounts of TNF-α when dogs were seronegative, but cells taken from seropositive dogs did not. Finally, the IL-18 response of seropositive dogs was reduced compared to those that were seronegative in response to PPDB, potentially suggesting that these dogs have a reduced macrophage functionality. This work demonstrates that the inflammatory cytokine response may wane following seroconversion with deleterious consequences for the host response. Overall, combining IFN-γ and TNF-α assessment during diagnosis may increase IGRA sensitivity, whilst further work is needed to better understand the prognostic and diagnostic implications of seroconversion in dogs. Full article
(This article belongs to the Section Immunological Responses and Immune Defense Mechanisms)
Show Figures

Figure 1

Figure 1
<p>Cytokine production by canine peripheral blood mononuclear cells (PBMCs) following stimulation by purified protein derived from <span class="html-italic">Mycobacterium bovis</span> (PPDB). Data represent the individual values of the antigen-specific cytokine concentration for each dog, along with the median ± interquartile range of the group. Dogs are grouped as IGRA+/seronegative (pink), IGRA+/seropositive (turquoise), or as uninfected control (black). ** <span class="html-italic">p</span> &lt; 0.01; * <span class="html-italic">p</span> &lt; 0.05; AU—arbitrary units.</p>
Full article ">Figure 2
<p>Cytokine production by canine peripheral blood mononuclear cells (PBMCs) following stimulation with a cocktail of ESAT-6/CFP-10. Data represent the individual values of the antigen-specific cytokine concentration for each dog along with the median ± interquartile range of the group. Dogs are grouped as IGRA+/seronegative (pink), IGRA+/seropositive (turquoise), or as uninfected control (black). * <span class="html-italic">p</span> &lt; 0.05; AU—arbitrary units.</p>
Full article ">
13 pages, 10000 KiB  
Article
Impacts of Copper Deficiency on Oxidative Stress and Immune Function in Mouse Spleen
by Xiaocong Li, Xin Zeng, Wanqin Yang, Peng Ren, Hengxiao Zhai and Heng Yin
Nutrients 2025, 17(1), 117; https://doi.org/10.3390/nu17010117 - 30 Dec 2024
Viewed by 247
Abstract
Introduction: Copper is an essential trace element crucial for enzyme synthesis and metabolism. Adequate copper levels are beneficial for maintaining the normal immune function of the spleen. Copper deficiency disrupts the metabolic processes within the spleen and impairs its immune function. This research [...] Read more.
Introduction: Copper is an essential trace element crucial for enzyme synthesis and metabolism. Adequate copper levels are beneficial for maintaining the normal immune function of the spleen. Copper deficiency disrupts the metabolic processes within the spleen and impairs its immune function. This research examines the impact of copper deficiency on the spleen and the potential recovery following copper supplementation. Methods: Weaned mice underwent a 4-week copper-deficient diet, succeeded by 1-week of copper repletion via intraperitoneal copper sulfate injection. Histological examination was used to assess pathological changes in the spleen. Biochemical assays were performed to measure oxidative stress levels in the spleen. ELISA, qPCR, and Western blot were employed to examine alterations in inflammatory markers, immune indicators, and oxidative regulatory factors across various levels. Results: Copper deficiency caused histological damage to the spleen, altered the expression of oxidative stress regulatory pathways (Nrf2, Keap1, and HO-1), and affected the expression of key inflammatory enzymes (iNOS, COX2) and transcription factor NF-κB, leading to oxidative damage. This was reflected by decreased levels of SOD, GSH, and T-AOC, along with increased levels of CAT and MDA. The levels of inflammatory cytokines IL-1β, TNF-α, IL-6, and IL-8 were notably increased. Copper supplementation significantly improved these changes. Conclusions: Copper deficiency leads to spleen tissue damage in mice, affecting the Nrf2 regulatory pathway and inducing oxidative damage. Subsequent copper supplementation with copper sulfate effectively ameliorates the damage caused by copper deficiency. Full article
(This article belongs to the Section Nutritional Immunology)
Show Figures

Figure 1

Figure 1
<p>Effects of Cu deficiency and supplementation on spleen index and histological morphology in mice. (<b>A</b>) Spleen microscopic structure of mice, H&amp;E, 100×. (<b>B</b>) Appearance and size of spleen. (<b>C</b>) Spleen index of the groups of mice. * <span class="html-italic">p</span> &lt; 0.05, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 2
<p>Effects of Cu deficiency and supplementation on oxidative damage of spleen in mice. (<b>A</b>–<b>E</b>) represent the CAT activity, SOD activity, GSH concentration, total antioxidant capacity, and MDA content in the spleens of mice from each group, respectively. * <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, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 3
<p>Effects of Cu deficiency and supplementation on cytokine of serum in mice. (<b>A</b>–<b>D</b>) represent concentrations of IL-1β, IL-6, IL-8, and TNF-α in mouse serum, respectively. * <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, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 4
<p>Effects of Cu deficiency and supplementation on mRNA expression of spleen inflammation-related genes. (<b>A</b>–<b>G</b>) represent the mRNA relative expression levels of IL-1β, IL-6, IL-8, TNF-α, NF-κB, iNOS, and COX2 in the spleens of mice, respectively. * <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, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 5
<p>Effects of Cu deficiency and supplementation on mRNA expression of spleen nrf2 signaling pathway. (<b>A</b>–<b>C</b>) represent the mRNA relative expression levels of Nrf2, Keap1, and HO-1 in the spleens of mice, respectively. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 6
<p>Impact of Cu deficiency and supplementation on spleen inflammation-related protein expression. (<b>A</b>) Protein levels of p-NF-κB, NF-κB, TNF-α, IL-1β, and IL-6 in the spleen, using β-actin as the loading control. (<b>B</b>) The ratio of p-NF-κB to NF-κB. (<b>C</b>–<b>E</b>) The relative expression levels of TNF-α, IL-1β, and IL-6 were normalized relative to β-actin. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>Effects of Cu deficiency and supplementation on protein expression of nrf2 signaling pathway of spleen. (<b>A</b>) Protein levels of Nrf2, Keap1, and HO-1 in spleen, using β-actin as loading control. (<b>B</b>–<b>D</b>) The relative expression levels of Nrf2, Keap1, and HO-1 were normalized to β-actin expression. *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">
24 pages, 9508 KiB  
Article
From High Protection to Lethal Effect: Diverse Outcomes of Immunization Against Invasive Candidiasis with Different Candida albicans Extracellular Vesicles
by Raquel Martínez-López, Gloria Molero, Claudia Marcela Parra-Giraldo, Matías Sebastián Cabeza, Guillermo Castejón, Carmen García-Durán, Luis Felipe Clemente, María Luisa Hernáez, Concha Gil and Lucía Monteoliva
Int. J. Mol. Sci. 2025, 26(1), 244; https://doi.org/10.3390/ijms26010244 - 30 Dec 2024
Viewed by 270
Abstract
Extracellular vesicles (EVs) from Candida albicans can elicit immune responses, positioning them as promising acellular vaccine candidates. We characterized EVs from an avirulent C. albicans cell wall mutant (ecm33Δ) and evaluated their protective potential against invasive candidiasis. EVs from the yeast [...] Read more.
Extracellular vesicles (EVs) from Candida albicans can elicit immune responses, positioning them as promising acellular vaccine candidates. We characterized EVs from an avirulent C. albicans cell wall mutant (ecm33Δ) and evaluated their protective potential against invasive candidiasis. EVs from the yeast (YEVs) and hyphal (HEVs) forms of the SC5314 wild-type strain were also tested, yielding high survival rates with SC5314 YEV (91%) and ecm33 YEV immunization (64%). Surprisingly, HEV immunization showed a dual effect, resulting in 36% protection but also causing premature death in some mice. Proteomic analyses revealed distinct profiles among the top 100 proteins in the different EVs, which may explain these effects: a shared core of 50 immunogenic proteins such as Pgk1, Cdc19, and Fba1; unique, relevant immunogenic proteins in SC5314 YEVs; and proteins linked to pathogenesis, like Ece1 in SC5314 HEVs. Sera from SC5314 YEV-immunized mice showed the highest IgG2a titers and moderate IL-17, IFN-γ, and TNF-α levels, indicating the importance of both humoral and cellular responses for protection. These findings highlight the distinct immunogenic properties of C. albicans EVs, suggesting their potential in acellular vaccine development while emphasizing the need to carefully evaluate pathogenic risks associated with certain EVs. Full article
(This article belongs to the Special Issue Fungal Pathogen-Host Interactions)
Show Figures

Figure 1

Figure 1
<p>NTA analysis of SC5314 YEVs and <span class="html-italic">ecm33</span> YEVs. TEM images showing spherical electron-dense bilayered structures, typical of extracellular vesicles, are provided for visual reference. The X10, X50, and X90 indicate a 10%, 50%, or 90% of EVs, respectively, with the specified size or smaller.</p>
Full article ">Figure 2
<p>(<b>a</b>) Functional enrichment analysis for proteins identified in SC5314 YEVs (1219 proteins) and <span class="html-italic">ecm33</span> YEVs (1333 proteins). The word clouds represent enriched terms based on the Fungal Database (CGD GO) analysis. These terms correspond to cellular compartments and are derived from Gene Ontology (GO). The size and color intensity of the terms indicate the significance of the enrichment, with larger and darker terms representing lower <span class="html-italic">p</span>-values. (<b>b</b>) Quantitative comparison of proteins present in EVs from both strains. The volcano plot illustrates proteins with significant differences in abundance between EV types (log2(fold change) vs. −log10(q-value)). Proteins more abundant in SC5314 YEVs or ecm33 YEVs are highlighted in blue and pink, respectively. Additionally, functional enrichment analyses were performed on the differentially abundant protein sets, focusing on “cellular component” (<b>top</b>) and “biological process” (<b>bottom</b>). The red asterisks indicate cellular components or biological processes related to the cell wall or cell surface.</p>
Full article ">Figure 3
<p>Vaccination schedule (<b>a</b>) and survival curves (<b>b</b>) of a mouse model of IC, showing the protective effect of immunization with different <span class="html-italic">C. albicans</span> EVs. Mice were immunized with different doses of EVs with adjuvant (Ad) or without adjuvant and subsequently challenged with an intravenous lethal dose of SC5314 (1 × 10<sup>6</sup> cells). Statistical differences between immunization and control groups are marked with an asterisk. * Mantel–Cox test, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Vaccination schedule (<b>a</b>) and survival curves (<b>b</b>) of a murine model of IC, showing the protective effect achieved with immunization with SC5314 YEVs and <span class="html-italic">ecm33</span> YEVs. Mice were immunized with different doses of EVs with adjuvant (Ad) and subsequently challenged with an intravenous lethal dose of SC5314 (1 × 10<sup>6</sup> cells). Vaccination with live cells (2.5 × 10<sup>6</sup> cells, one dose) of the completely avirulent <span class="html-italic">ecm33</span>Δ mutant was included as a positive vaccination control (<b>a</b>,<b>b</b>). * Mantel–Cox test, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Vaccination schedule (<b>a</b>) and survival curves (<b>b</b>) of a mouse model of invasive candidiasis, showing the protective effect of immunization with different <span class="html-italic">C. albicans</span> EVs. Mice were immunized with different doses of EVs with adjuvant (Ad) and subsequently challenged with an intravenous lethal dose of SC5314 (5 <span class="html-italic">×</span> 10<sup>5</sup> cells). Mantel–Cox test, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.0005 (black asterisks represent statistically significant differences between the control group and the different EV-immunized groups. Red asterisks represent statistically significant differences in the protection acquired between the different EV-immunized groups).</p>
Full article ">Figure 6
<p>(<b>a</b>) Hierarchical heatmap depicting the relative abundance of each protein across the three different types of EVs, with darker shades of red indicating higher relative abundance (measured by NSAF) (protein names shown represent 1 out of every 22 proteins for clarity). (<b>b</b>) Zoomed-in view on the region of the heatmap with a higher abundance in cell surface proteins in SC5314 YEVs. Proteins described as immunogenic in the Candida Genome Database (CGD) are marked with an asterisk.</p>
Full article ">Figure 7
<p>Venn diagram illustrating the top 100 most abundant proteins in each type of extracellular vesicle (EV). Proteins previously reported as immunogenic in other studies are highlighted in bold. Proteins associated with virulence are shaded.</p>
Full article ">Figure 8
<p>IgG and IgG2a antibody titers. ELISAs with pooled sera obtained prior to infection from immunized mice with SC5314 YEVs (blue), <span class="html-italic">ecm33</span> YEVs (pink), and SC5314 HEVs (green). Protein samples from SC5314 YEVs, SC5314 HEVs, and total cytoplasmic extracts from the wild-type strain SC5314 were used for detection purposes. Statistically significant differences in antibody titers were observed across all groups compared to the control group. Two-way ANOVA test, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 9
<p>Cytokine profile for each of the pooled sera obtained after complete immunization with the different EVs (SC5314 YEVs (blue), <span class="html-italic">ecm33</span> YEVs (pink), and SC5314 HEVs (green)) but prior to infection (immunized), and 30 days after infection (surviving). Adjuvant group (control group) received only the adjuvant. Statistically significant differences in cytokine levels were observed across all cases compared to the control group. Two-way ANOVA test, *** <span class="html-italic">p</span> &lt;0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
15 pages, 5792 KiB  
Article
Structural Characterization and Immune Activation Capacity of Peptidoglycan from Corynebacterium glutamicum in RAW264.7 Cells
by Xiaoying Wang, Shuzhen Li, Aijuan Zheng, Zhimin Chen, Jiang Chen, Zhiheng Zou and Guohua Liu
Int. J. Mol. Sci. 2025, 26(1), 237; https://doi.org/10.3390/ijms26010237 - 30 Dec 2024
Viewed by 252
Abstract
Peptidoglycan (PGN) is a unique component of prokaryotic cell walls with immune-enhancing capacities. Here, we extracted PGN from Corynebacterium glutamicum, a by-product of amino acid fermentation, using the trichloroacetic acid (TCA) method. SDS-PAGE analysis confirmed the presence of PGN, with a band [...] Read more.
Peptidoglycan (PGN) is a unique component of prokaryotic cell walls with immune-enhancing capacities. Here, we extracted PGN from Corynebacterium glutamicum, a by-product of amino acid fermentation, using the trichloroacetic acid (TCA) method. SDS-PAGE analysis confirmed the presence of PGN, with a band of approximately 28 kDa. Further analysis was conducted through amino acid analysis, FTIR, and MALDI-TOF/TOF MS, and the results showed that the chemical structural monomer of PGN is NAG-(β-1,4-)-NAM-l-Ala-d-Glu-l-Lis-d-Ala. The immune activation effects of PGN were evaluated in a RAW264.7 cell model. Our results showed that PGN could increase the secretion level of NO, ROS, and immune regulatory substances, including TNF-α and IL-1β, and up-regulated the mRNA expression of TNF-α and iNOS. In addition, PGN stimulated the expression of ERK2, MyD88, RIP2, and the related receptor NOD1 in the NF-κB and MAPK pathways. Comparative RNA sequencing was conducted to analyze the gene expression profiles in RAW264.7 cells. KEGG analysis indicated that most of the genes were enriched in the NF-κB, MAPK, and TNF signaling pathways. Taken together, these findings suggest that PGN may have immune-activating potential for the development and application of immune adjuvants. Importantly, the application of PGN also provides a new way to utilize amino acid fermentation by-products. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structure of PGN from <span class="html-italic">Corynebacterium glutamicum</span>. (<b>a</b>) Examination via SEM at original magnification (1000×) and 5000×. (<b>b</b>) SDS-PAGE analysis of PGN proteins. (<b>c</b>) MALDI-TOF/TOF MS analysis of PGN. (<b>d</b>) FTIR analysis of PGN. (<b>e</b>) Chemical structure of PGN.</p>
Full article ">Figure 2
<p>Effects of different PGN concentrations on the immune-active substances in RAW264.7 cells. (<b>a</b>) Cell viability. (<b>b</b>) NO production. (<b>c</b>) ROS release. (<b>d</b>) TNF-α and IL-1β secretion. PGN refers to peptidoglycan extract. PGN concentrations of 0, 12.5, 25, and 50 μg/mL were used as control (CK), low, medium, and high PGN groups, respectively. Data are presented as mean ± SD from three independent experiments. Values indicated with different letters differ significantly (<span class="html-italic">p</span> &lt; 0.05) compared to the control (CK) group, as determined by one-way ANOVA.</p>
Full article ">Figure 3
<p>Effects of different concentrations of PGN on the gene expression of immune-active substances in RAW264.7 cells. (<b>a</b>) <span class="html-italic">TNF-α</span>, <span class="html-italic">IL-1β</span>, and <span class="html-italic">IL-6</span>. (<b>b</b>) <span class="html-italic">iNOS</span>. (<b>c</b>) <span class="html-italic">COX-2</span>. PGN refers to peptidoglycan extract. PGN concentrations of 0, 12.5, 25, and 50 μg/mL were used as control (CK), low, medium, and high PGN groups, respectively. Data are presented as mean ± SD from three independent experiments. Values indicated with different letters differ significantly (<span class="html-italic">p</span> &lt; 0.05) from the control (CK) group, as determined by one-way ANOVA.</p>
Full article ">Figure 4
<p>Effects of PGN on the expression of related signaling pathway genes in RAW264.7 cells. (<b>a</b>) <span class="html-italic">NOD1</span>. (<b>b</b>) <span class="html-italic">NOD2</span>. (<b>c</b>) <span class="html-italic">TRL2</span>. (<b>d</b>) <span class="html-italic">RIP2</span>. (<b>e</b>) <span class="html-italic">ERK2</span>. (<b>f</b>) <span class="html-italic">MyD88</span>. (<b>g</b>) <span class="html-italic">IKKα</span>. (<b>h</b>) <span class="html-italic">IKKβ</span>. PGN refers to peptidoglycan extract. PGN concentrations of 0, 12.5, 25, and 50 μg/mL were used as control (CK), low, medium, high PGN groups, respectively. Data are presented as mean ± SD from three independent experiments. Values indicated with different letters differ significantly (<span class="html-italic">p</span> &lt; 0.05) compared to the control (CK) group, as determined by one-way ANOVA.</p>
Full article ">Figure 5
<p>Transcriptomic changes in gene expression in RAW264.7 cells by PGN. (<b>a</b>) Volcano plot of mRNA expression in CK and low-, medium-, and high-PGN-dose groups. (<b>b</b>) Identification of DEGs (differentially expressed genes) by RNA sequencing. (<b>c</b>) Venn diagram analysis of differentially expressed mRNAs among the three groups. (<b>d</b>) Heat map of differentially expressed genes showing stratified clustering of the four groups. (<b>e</b>) KEGG top 20 enrichment of differentially expressed mRNAs between the CK and the three PGN groups.</p>
Full article ">
15 pages, 3793 KiB  
Article
Panduratin A Inhibits TNF Alpha-Stimulated Endothelial Cell Activation Through Suppressing the NF-κB Pathway
by Kriangkrai Kiatsoonthon, Nitchakarn Phimthong, Saranyapin Potikanond, Nitwara Wikan and Wutigri Nimlamool
Biomolecules 2025, 15(1), 34; https://doi.org/10.3390/biom15010034 - 30 Dec 2024
Viewed by 287
Abstract
Upon exposure to inflammatory stimuli including TNF-α, endothelial cells are activated leading to the adhesion of monocytes to their surface. These events are involved in the pathophysiology of atherosclerosis. Since TNF-α activates the NF-κB pathway, which contributes to atherosclerosis, targeting this signaling pathway [...] Read more.
Upon exposure to inflammatory stimuli including TNF-α, endothelial cells are activated leading to the adhesion of monocytes to their surface. These events are involved in the pathophysiology of atherosclerosis. Since TNF-α activates the NF-κB pathway, which contributes to atherosclerosis, targeting this signaling pathway may help prevent the risk of developing the disease. The current study elucidated the inhibitory effect of panduratin A (PA) on TNF-α-induced endothelial activation and monocyte adhesion. We discovered that PA reduced the level of pro-inflammatory cytokine IL-6 and chemokine MCP-1 in the media collected from endothelial cells stimulated with TNF-α. In addition, PA inhibited the expression of ICAM-1 and VCAM-1 on the surface of TNF-α-induced endothelial cells resulting in a decrease in the number of monocytes attached to endothelial cell surface. Mechanistically, PA prevented IκB degradation and specifically suppressed NF-κB phosphorylation and nuclear translocation in endothelial cells. However, PA had no inhibitory effect on the phosphorylation of AKT, ERK1/2, p38, and JNK. Taken together, PA blocked the production of cytokine and chemokine, adhesion molecules, and monocyte adhesion in response to TNF-α stimulation, in part, through NF-κB inhibition. Our study suggests that PA may possibly be effective in blocking the pathophysiology of atherosclerosis. Full article
(This article belongs to the Special Issue The Value of Natural Compounds as Therapeutic Agents: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Effect of PA on the viability or proliferation of endothelial cells. Cells were treated with PA (at concentrations ranging from 0 to 10 μM) or DMSO (vehicle control) for 48 h prior to the cell viability test. Data are obtained from at least 3 individual experiments and presented as mean ± SD.</p>
Full article ">Figure 2
<p>Effect of PA on secretion of IL-6 and MCP-1 in the TNF-α-induced endothelial cell culture media. The level of IL-6 (<b>A</b>) and MCP-1 (<b>B</b>) in the media of the PA-treated cells stimulated with 10 TNF-α for 24 h evaluated by ELISA. Data are presented as mean ± SD * <span class="html-italic">p</span> &lt; 0.05 (compared with the DMSO-treated group).</p>
Full article ">Figure 3
<p>PA decreased TNF-α- induced expression of vascular adhesion molecules on endothelial cell surface. Endothelial cells (untreated, TNF-α-simulated, or TNF-α-simulated with the presence of PA at 4 μM for 24 h) were subject to immunofluorescence for detecting VCAM-1 (<b>A</b>) and ICAM-1 (<b>B</b>). Nuclei (blue) were visualized by DAPI staining. The micrographs were captured at 100× magnification.</p>
Full article ">Figure 4
<p>Effects of TNF-α and PA on the morphology and cytoskeletal arrangement of endothelial cells: (<b>A</b>) Phase-contrast microscopy visualizing endothelial cells without any treatment (Untreated), induced with TNF-α, or induced with TNF-α with the presence of 4 μM of PA at 0, 12, and 24 h (Scale bar = 300 μm). Immunofluorescence study detecting α-tubulin (<b>B</b>), β-catenin (<b>C</b>), and vimentin (<b>D</b>) in endothelial cells after treatment for 24 h. Cells were also stained for F-actin (red) and nuclei (blue). The micrographs (immunofluorescence study) were captured at 100× magnification.</p>
Full article ">Figure 5
<p>The effect of PA on THP-1 cell binding to TNF-α-activated endothelial cells. Adhesion of the labeled THP-1 monocytes to endothelial cells (untreated, induced with TNF-α, induced with TNF-α with the presence of PA at 4 μM, or induced with TNF-α with the presence of DMSO (vehicle control) for 24 h) was visualized and quantified by using a live-cell imager: (<b>A</b>) Phase-contrast microscopy showing THP-1 monocytes bound to endothelial monolayer, and fluorescent microscopy showing the nuclei (blue) of THP-1 monocytes. Scale bar = 300 μm. (<b>B</b>) Quantitative analysis of THP-1 cells bound to endothelial cells. Data are expressed as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>The effect of PA on TNF-α-activated phosphorylation of NF-κB (pNF-κB) and the degradation of IκB. (<b>A</b>) Western blot analysis showing the level of pNF-κB and IκB in cells without any treatment, TNF-α induction, and TNF-α induction with the presence of PA, or TNF-α induction with the presence of DMSO (vehicle control). (<b>B</b>) Densitometry of pNF-κB immunoreactive bands from cells without any treatment, TNF-α induction, and TNF-α induction with the presence of PA, or TNF-α induction with the presence of DMSO. (<b>C</b>) Densitometry of IκB immunoreactive bands from cells without any treatment, TNF-α induction, and TNF-α induction with the presence of PA, or TNF-α induction with the presence of DMSO. * <span class="html-italic">p</span> &lt; 0.05. Original images of (<b>A</b>) can be found in <a href="#app1-biomolecules-15-00034" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 7
<p>The effect of PA on inhibiting TNF-α-activated nuclear relocalization of NF-κB and IκB degradation: (<b>A</b>) The presence of NF-κB in the nuclear extract of TNF-α-activated endothelial cells with or without the presence of PA. (<b>B</b>) Densitometric analysis of NF-κB immunoreactive bands from the Western blot of the nuclear extracts. (<b>C</b>) Immunofluorescence study visualizing relocalization of NF-κB into the nucleus of TNF-α-activated endothelial cells with or without the presence of PA. (<b>D</b>) Western blot analysis detecting IκB in the cytoplasmic extracts of TNF-α-activated endothelial cells with or without the presence of PA. (<b>E</b>) Densitometric analysis of IκB immunoreactive bands from the Western blot of the cytoplasmic extracts. (<b>F</b>) Immunofluorescence study visualizing the existence of IκB in the cytosol of TNF-α-activated endothelial cells with or without the presence of PA. * <span class="html-italic">p</span> &lt; 0.05. The micrographs were captured at 100× magnification. Original images of (<b>A</b>,<b>D</b>) can be found in <a href="#app1-biomolecules-15-00034" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 8
<p>The effect of PA on the phosphorylation status of important molecular markers for growth and survival: (<b>A</b>) Western blot determining the level of pAKT. (<b>B</b>) Quantitative analysis of pAKT immunoreactive bands. (<b>C</b>) Western blot determining the level of pERK1/2. (<b>D</b>) Quantitative analysis of pERK1/2 immunoreactive bands. (<b>E</b>) Western blot analysis detecting pp38. (<b>F</b>) Quantitative analysis of pp38 immunoreactive bands. (<b>G</b>) Western blot analysis detecting pJNK. (<b>H</b>) Quantitative analysis of pJNK immunoreactive bands. * <span class="html-italic">p</span> &lt; 0.05. Original images of (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) can be found in <a href="#app1-biomolecules-15-00034" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 9
<p>The proposed model for the mechanism of PA in reducing TNF-α-stimulated endothelial activation and monocyte adhesion. This figure was created with an approval from <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 28 November 2024).</p>
Full article ">
14 pages, 2853 KiB  
Article
Dysregulation of Podocyte BK Channels and Nephrosis: Effects of Circulating Factors and Auxiliary β4 Subunits
by Eun Young Kim, Patrycja Rachubik and Stuart E. Dryer
Cells 2025, 14(1), 22; https://doi.org/10.3390/cells14010022 - 30 Dec 2024
Viewed by 214
Abstract
Podocytes express large-conductance Ca2+-activated K+ channels (BK channels) and at least two different pore-forming KCa1.1 subunit C-terminal splice variants, known as VEDEC and EMVYR, along with auxiliary β and γ subunits. Podocyte KCa1.1 subunits interact directly with TRPC6 channels and [...] Read more.
Podocytes express large-conductance Ca2+-activated K+ channels (BK channels) and at least two different pore-forming KCa1.1 subunit C-terminal splice variants, known as VEDEC and EMVYR, along with auxiliary β and γ subunits. Podocyte KCa1.1 subunits interact directly with TRPC6 channels and BK channels become active in response to Ca2+ influx through TRPC6. Here, we confirmed that Ca2+ influx through TRPC channels is reduced following the blockade of BK channels by paxilline. The overall abundance of KCa1.1 subunits, as well as that of β4 and γ3 subunits, were increased in glomeruli isolated from Sprague Dawley rats during chronic puromycin aminonucleoside (PAN) nephrosis. Exposing cultured mouse podocytes for 24 h to recombinant TNFα, a circulating factor implicated in pediatric nephrotic syndromes, did not affect the total abundance of KCa1.1, but did evoke significant increases in both β4 and γ3. However, TNFα evoked a marked increase in the surface abundance of KCa1.1 subunits, similar to that of its previously reported effects on TRPC6 channels. The effect of TNFα on the surface expression of KCa1.1 was eliminated following siRNA knockdown of the β4 subunits, suggesting a role for this subunit in KCa1.1 trafficking to the cell surface. By contrast, treating podocytes with suPAR did not affect the total or surface expression of KCa1.1. The coordinated activation of KCa1.1 channels may promote Ca2+ influx through TRPC channels during normal and abnormal podocyte function by maintaining a membrane potential that allows for the efficient permeation of divalent cations through TRPC pores. Full article
Show Figures

Figure 1

Figure 1
<p>Quantitative fluorescence measurement of cytosolic free Ca<sup>2+</sup> in immortalized podocytes in response to a hypoosmotic stretch stimulus known to cause robust and sustained activation of TRPC6. The Ca<sup>2+</sup> signal was measured using the indicator dye Calybryte-520™: (<b>a</b>) a hypoosmotic stretch stimulus caused a marked increase in cytosolic free Ca<sup>2+</sup> that was maintained for as long as the stretch stimulus was maintained, as we have reported previously for TRPC6 channels [<a href="#B13-cells-14-00022" class="html-bibr">13</a>]; (<b>b</b>) pre-exposure for 10 min to the TRPC6 blocker SAR-7334 (100 nM) caused a marked reduction in the amplitude of this signal; and (<b>c</b>) a similar reduction was observed in podocytes pre-treated for 10 min with the BK channel inhibitor paxilline (20 μM). Points represent means amplitudes relative to baseline fluorescence, error bars represent SEM, and the number of cells examined is indicated. The inhibition by paxilline suggests that BK channels allow for enhanced Ca<sup>2+</sup> influx through TRPC6.</p>
Full article ">Figure 2
<p>Evidence of kidney disease in Sprague Dawley rats 30-days after treatment with PAN or saline, as indicated. This experimental design is the chronic PAN nephrosis model: (<b>a</b>) increase in spot urine albumin:creatinine ratios (ACR) following PAN treatment; (<b>b</b>) increase in serum creatinine in PAN-treated rats compared to the control, indicating a decrease in the glomerular filtration rate; (<b>c</b>) glomerulosclerosis in PAN-treated rats compared to the saline-treated controls shown in representative PAS-stained section; and (<b>d</b>) mean glomerular score in saline- and PAN-treated rats based on the analyses of PAS-stained sections by an observer blind to the treatment group being scored. Points are results from individual animals. Error bars represent SD. Data were analyzed by Student’s unpaired <span class="html-italic">t</span>-test. In this experiment, there were <span class="html-italic">N</span> = 4 animals in each group.</p>
Full article ">Figure 3
<p>Increase in the expression of pore-forming and auxiliary subunits of BK channels in whole glomeruli isolated from saline- and PAN-treated rats during chronic PAN nephrosis. Representative immunoblots measuring pore-forming KCa1.1 subunits and auxiliary β4 and γ3 subunits are shown on the left (all from the same animal); quantitative densitometric analyses from the two groups of animals are shown on the right. Graphs show individual values and SD for <span class="html-italic">N</span> = 4 animals in each group.</p>
Full article ">Figure 4
<p>Effects of TNFα on BK channel subunits. Exposing cultured podocytes to 10 ng/mL TNFα for 24 h does not increase overall abundance of KCa1.1 (<b>a</b>) but increases overall abundance of auxiliary β4 and γ3 subunits (<b>b</b>). Representative blots are shown to the left. Points in graphs on the right are the results from each repetition and error bars represent SD. Data were analyzed by Student’s unpaired <span class="html-italic">t</span>-test.</p>
Full article ">Figure 5
<p>Exposing cultured podocytes to 20 ng/mL suPAR for 24 h had no effect on the overall abundance of pore-forming KCa1.1 subunits and caused a decrease in auxiliary β4 and γ3. Data were analyzed by Student’s unpaired <span class="html-italic">t</span>-test. n.s. denotes statistically non-significant difference.</p>
Full article ">Figure 6
<p>Exposing cultured podocytes to TNFα for 24 h increased cell surface expression of KCa1.1 (<b>a</b>); whereas suPAR had no effect (<b>b</b>). Graphs show individual values of the relative expression of surface and total KCa1.1 based on cell-surface biotinylation assays. Error bars represent SD. Data were analyzed by Student’s unpaired <span class="html-italic">t</span>-test. n.s. denotes statistically non-significant difference.</p>
Full article ">Figure 7
<p>The stimulatory effect of TNFα on the surface expression of KCa1.1 subunits in podocytes was inhibited by siRNA knockdown of β4 subunits: (<b>a</b>) immunoblot analysis showing reduced expression of the β4 subunit by the siRNA targeting that subunit compared to the control siRNA; and (<b>b</b>) representative cell-surface biotinylation assays are shown above the quantitative analyses of these types of experiments. There was a significant difference in the groups by one-way ANOVA (<span class="html-italic">F</span> = 11.05, <span class="html-italic">p</span> &lt; 0.01); the siRNA targeting the β4 subunit significantly reduced the effects of TNFα (<span class="html-italic">p</span> &lt; 0.01, Tukey’s honestly significant difference post hoc test).</p>
Full article ">
Back to TopTop