Nothing Special   »   [go: up one dir, main page]

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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (441)

Search Parameters:
Keywords = KEAP1/NRF2 pathway

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 4663 KiB  
Article
Microplastics Exposure Aggravates Synovitis and Pyroptosis in SLE by Activating NF-κB and NRF2/KEAP1 Signaling
by Wenxiang Zeng, Shiqiao He, Ying Zhao, Minjian Jiang, Wenla Wang, Limeng Yang, Weibin Du and Wei Zhuang
Toxics 2024, 12(12), 840; https://doi.org/10.3390/toxics12120840 - 22 Nov 2024
Viewed by 346
Abstract
Microplastics (MPs) represent an emerging pollutant capable of entering the human body through the respiratory and digestive systems, thereby posing significant health risks. Systemic lupus erythematosus (SLE) is a complex autoimmune disease that affects multiple organ systems, often presenting with polyarticular joint manifestations. [...] Read more.
Microplastics (MPs) represent an emerging pollutant capable of entering the human body through the respiratory and digestive systems, thereby posing significant health risks. Systemic lupus erythematosus (SLE) is a complex autoimmune disease that affects multiple organ systems, often presenting with polyarticular joint manifestations. Despite its relevance, there is currently limited research on the impact of MPs on lupus arthritis. This study aims to investigate the effects of MPs on joint inflammation in SLE. MRL/lpr mice exhibit SLE similar to that of humans. We administered either 0.5 mg/kg or 5 mg/kg of MPs to 8-week-old female MRL/lpr mice via oral ingestion. Our findings indicate that exposure to MPs can lead to synovial damage, adversely affecting the morphology and function of the knee joint, along with increased oxidative stress, apoptosis, synovial fibrosis, and the secretion of inflammatory cytokines. Notably, MPs significantly enhanced synovial cell pyroptosis by upregulating the expression of NLRP3, CASPASE-1, GSDMD, IL-1β, and IL-18. Mechanistic analyses further demonstrated that MPs exposure activates the NF-κB and NRF2/KEAP1 signaling pathways. Overall, our in vivo findings suggest that MPs exposure promotes synovial cell pyroptosis through increased oxidative stress and NF-κB signaling, thereby disrupting the structure and function of synovial tissue. This research provides new insights into the synovial damage associated with MPs exposure. Full article
(This article belongs to the Section Reproductive and Developmental Toxicity)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The MP structure damages the synovial structure and aggravate synovitis in SLE mice: (<b>A</b>) flow chart of the experimental design; (<b>B</b>) HE staining of mouse knee synovium, demonstrating thickening of synovial layers (Black arrow) and increased vasculature (Blue arrow); (<b>C</b>) histological scoring of the synovium based on HE staining; (<b>D</b>) MASSON’s trichrome staining of mouse knee synovium, indicating increased fiber content (Black arrow); (<b>E</b>–<b>H</b>) immunohistochemical staining illustrating the expression of MMP-13 and MMP-19 in the synovium of the knee joint of mice, along with quantification of their expression levels. Red arrowheads denote positively stained cells. Data are presented as mean ± standard deviation (SD). ns indicates no statistical significance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BL/6J group), with <span class="html-italic">n</span> = 6 per group.</p>
Full article ">Figure 2
<p>MPs promote synovial cell apoptosis in SLE mice: (<b>A</b>,<b>C</b>) immunofluorescence staining demonstrating the expression of CASPASE-3 and BCL-2 in the synovium of the knee joints of mice, along with (<b>B</b>,<b>D</b>) quantification of expression; (<b>E</b>) TUNEL staining and (<b>F</b>) quantification of the rate of TUNEL-positive cells. DAPI stains the nuclei blue, and white arrowheads indicate positively stained cells. Data are presented as mean ± SD. ns indicates no statistical significance. ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BL/6J group), <span class="html-italic">n</span> = 6 per group.</p>
Full article ">Figure 3
<p>MPs compound synovial inflammation in SLE mice. (<b>A</b>–<b>H</b>) Immunofluorescence staining illustrating the expression of IL-1β, IL-18, IL-6, and TNF-α in the synovium of the knee joint of mice, along with quantification of expression. DAPI stains the nuclei blue, and white arrowheads indicate positively stained cells. Data are presented as mean ± SD. ns indicates no statistical significance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BJ/6 group), with <span class="html-italic">n</span> = 6 per group.</p>
Full article ">Figure 4
<p>MPs aggravate synovial pyroptosis in SLE mice. (<b>A</b>–<b>C</b>) Immunofluorescence staining showing the expression of NLRP3, CASPASE-1, and GSDMD in the synovium of the knee joint of mice, along with (<b>D</b>–<b>F</b>) quantification of expression. DAPI stains nuclei blue, and white arrowheads indicate positively stained cells. Data are expressed as mean ± SD. ns indicates no statistical significance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BL/6J group), with <span class="html-italic">n</span> = 6 per group.</p>
Full article ">Figure 5
<p>Effects of MPs exposure on oxidative stress and the NF-κB signaling pathway in SLE mice. (<b>A</b>–<b>C</b>) Immunofluorescence staining was conducted to assess the expression of NRF2, KEAP1, and HO-1 in the synovium of the knee joints of mice, while (<b>H</b>–<b>J</b>) illustrates the quantification of these expressions. (<b>D</b>–<b>G</b>) Immunofluorescence staining for P65, P-P65, IκBα, and p-IκBα in the knee joint synovium of mice is presented, with (<b>K</b>–<b>N</b>) showing the corresponding quantification. DAPI was used to stain nuclei in blue, and white arrowheads indicate positively stained cells. Data are expressed as mean ± SD. ns indicates no statistical significance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BL/6J group), with <span class="html-italic">n</span> = 6 per group.</p>
Full article ">
25 pages, 6608 KiB  
Article
Microbiome–Metabolomics Analysis Insight into the Effects of Starvation and Refeeding on Intestinal Integrity in the Juvenile Largemouth Bass (Micropterus salmoides)
by Zhenxin Zhao, Xianbo Zhang, Fei Zhao and Tianxun Luo
Int. J. Mol. Sci. 2024, 25(23), 12500; https://doi.org/10.3390/ijms252312500 - 21 Nov 2024
Viewed by 408
Abstract
The effects of starvation and refeeding on the gut condition of juvenile largemouth bass (Micropterus salmoides) remain unclear. Therefore, our research aimed to explore these effects. Amylase and lipase activities were remarkably decreased in the starvation (ST) group, yet prominently increased [...] Read more.
The effects of starvation and refeeding on the gut condition of juvenile largemouth bass (Micropterus salmoides) remain unclear. Therefore, our research aimed to explore these effects. Amylase and lipase activities were remarkably decreased in the starvation (ST) group, yet prominently increased in the refeeding (RE) group (p < 0.05). In addition to the malondialdehyde (MDA) level, catalase (CAT) and superoxide dismutase (SOD) activities were significantly upregulated in the ST group (p < 0.05) in marked contrast to those in the controls; however, the RE group showed no substantial variations in CAT and SOD activities or the MDA level (p > 0.05). During starvation, the expression of Nrf2-Keap1 pathway-associated genes was significantly upregulated (p < 0.05). The comparative levels of TNF-α, IL-1β, and IL-15 were highly increased, with the levels of TGF-β1 and IL-10 apparently downregulated in the ST group; in contrast, these levels were restored to their original values in the RE group (p < 0.05). In contrast to the controls, the ST group showed significantly lower height and width of the villi, muscle thickness, and crypt depth and a higher goblet cell number; however, these values were recovered to some extent in the RE group (p < 0.05). The dominant bacterial phyla in the intestines of both groups were Proteobacteria, Firmicutes, Bacteroidetes, Acidobacteria, and Actinobacteria, with marked inter-group differences in the genera Serratia and Lactobacillus. Metabolomics analysis showed that amino acid metabolism is disrupted during starvation and is restored after refeeding. In summary, this study expands our comprehension of the interaction between oxidative stress and antioxidant defenses among juvenile largemouth bass subjected to starvation and refeeding. Full article
(This article belongs to the Special Issue Fish Immunology, 5th Edition)
Show Figures

Figure 1

Figure 1
<p>Digestive and antioxidant enzyme activities of juvenile largemouth bass subjected to starvation and refeeding at the end of the experiment. Note: Values are means ± SEM. Different letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05). SOD: superoxide dismutase, CAT: catalase, MDA: malondialdehyde.</p>
Full article ">Figure 2
<p>qRT-PCR analysis of the expression of Nrf2-Keap1 pathway-related genes in the anterior intestine of juvenile largemouth bass after the onset of starvation and refeeding. Note: Values are means ± SEM. Different letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05). <span class="html-italic">Nrf2</span>: nuclear factor erythroid 2-related factor 2, <span class="html-italic">Keap1</span>: Kelch-like ECH-associated protein 1, <span class="html-italic">GPx</span>: glutathione peroxidase, <span class="html-italic">GCLC</span>: glutamate-cysteine ligase catalytic subunit, <span class="html-italic">HO-1</span>: heme oxygenase-1.</p>
Full article ">Figure 3
<p>qRT-PCR analysis of the expression of inflammation-related genes in the anterior intestine of juvenile largemouth bass after the onset of starvation and refeeding. Note: Values are means ± SEM. Different letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05). <span class="html-italic">IL-1β</span>: interleukin-1β, <span class="html-italic">IL-8</span>: interleukin-8, <span class="html-italic">IL-10</span>: interleukin-10, <span class="html-italic">IL-15</span>: interleukin-15, <span class="html-italic">TNF-α</span>: tumor necrosis factor, <span class="html-italic">TGF-β1</span>: transforming growth factor-β1.</p>
Full article ">Figure 4
<p>Histological examination the mid-gut morphology after the onset of starvation and refeeding using H&amp;E and AB-PAS staining in juvenile largemouth bass. VH: villus height (µm), VW: villus width (µm), MT: muscle thickness (µm), CD: crypt depth (µm), GC: goblet cells. Note: Values are means ± SEM. Different letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>The effects of starvation and refeeding on 16S rRNA gene sequencing of the intestinal microbiota in juvenile largemouth bass (<span class="html-italic">n</span> = 6 per group). (<b>A</b>) Venn diagram; (<b>B</b>) PCA diversity; (<b>C</b>) Alterations in microbiota at the phylum level; (<b>D</b>) The abundance of the Firmicutes/Bacteroidetes (F/B) ratio in different groups, diverse lowercase letters above the bars show significant differences (<span class="html-italic">p</span> &lt; 0.05) in different groups; (<b>E</b>) Alterations in microbiota at the genus level.</p>
Full article ">Figure 5 Cont.
<p>The effects of starvation and refeeding on 16S rRNA gene sequencing of the intestinal microbiota in juvenile largemouth bass (<span class="html-italic">n</span> = 6 per group). (<b>A</b>) Venn diagram; (<b>B</b>) PCA diversity; (<b>C</b>) Alterations in microbiota at the phylum level; (<b>D</b>) The abundance of the Firmicutes/Bacteroidetes (F/B) ratio in different groups, diverse lowercase letters above the bars show significant differences (<span class="html-italic">p</span> &lt; 0.05) in different groups; (<b>E</b>) Alterations in microbiota at the genus level.</p>
Full article ">Figure 5 Cont.
<p>The effects of starvation and refeeding on 16S rRNA gene sequencing of the intestinal microbiota in juvenile largemouth bass (<span class="html-italic">n</span> = 6 per group). (<b>A</b>) Venn diagram; (<b>B</b>) PCA diversity; (<b>C</b>) Alterations in microbiota at the phylum level; (<b>D</b>) The abundance of the Firmicutes/Bacteroidetes (F/B) ratio in different groups, diverse lowercase letters above the bars show significant differences (<span class="html-italic">p</span> &lt; 0.05) in different groups; (<b>E</b>) Alterations in microbiota at the genus level.</p>
Full article ">Figure 6
<p>Spearman’s correlation-based network analysis of bacterial communities for interactions among genera. Nodes (colored dots) represent genera, whose size is proportional to their relative abundance. A line between two nodes indicates a significant positive (orange, Spearman’s correlation, <span class="html-italic">rs</span> &gt; 0.1 and <span class="html-italic">p</span> &lt; 0.05) or negative (green, Spearman’s correlation, <span class="html-italic">rs</span> &gt; −0.1 and <span class="html-italic">p</span> &lt; 0.05) correlation, with its thickness reflecting the relative strength.</p>
Full article ">Figure 7
<p>The functional pathways showing the highest differences between the (<b>A</b>) CON and ST, and (<b>B</b>) CON and RE using PICRUSt2 analysis, generated by the STAMP program. Bars on the left indicate the percentage of each category at different levels. Only categories with Bonferroni-corrected <span class="html-italic">p</span>-values exhibiting statistical differences at <span class="html-italic">p</span> &lt; 0.05 are shown.</p>
Full article ">Figure 7 Cont.
<p>The functional pathways showing the highest differences between the (<b>A</b>) CON and ST, and (<b>B</b>) CON and RE using PICRUSt2 analysis, generated by the STAMP program. Bars on the left indicate the percentage of each category at different levels. Only categories with Bonferroni-corrected <span class="html-italic">p</span>-values exhibiting statistical differences at <span class="html-italic">p</span> &lt; 0.05 are shown.</p>
Full article ">Figure 8
<p>The effects of starvation and refeeding on the intestinal metabolic profile in juvenile largemouth bass (<span class="html-italic">n</span> = 6 per group). (<b>A</b>) PCA scores; (<b>B</b>) Volcano plot; (<b>C</b>) Venn analysis.</p>
Full article ">Figure 8 Cont.
<p>The effects of starvation and refeeding on the intestinal metabolic profile in juvenile largemouth bass (<span class="html-italic">n</span> = 6 per group). (<b>A</b>) PCA scores; (<b>B</b>) Volcano plot; (<b>C</b>) Venn analysis.</p>
Full article ">Figure 9
<p>The KEGG functional annotation of significantly differential metabolites between the groups CON and ST. (<b>A</b>) Classification diagram. The horizontal axis is the number of different metabolites annotated to the pathway, and the vertical axis is the pathway name; (<b>B</b>) Network diagram. Note: The light-yellow nodes in the figure are the pathways, and the small nodes connected to each are the specific metabolites annotated to that pathway. The depth of the color indicates the difference multiple based on the log2 value.</p>
Full article ">Figure 10
<p>Intestinal microbial and immune responses of juvenile largemouth bass after the onset of starvation and refeeding. Green and red arrows indicate decreased and increased responses, respectively. (For the interpretation of the references to color in this figure legend, the reader is referred to the web version of this article).</p>
Full article ">
22 pages, 3391 KiB  
Article
Bioactivity of Grape Pomace Extract and Sodium Selenite, Key Components of the OenoGrape Advanced Complex, on Target Human Cells: Intracellular ROS Scavenging and Nrf2/ARE Induction Following In Vitro Intestinal Absorption
by Cécile Dufour, Camille Gironde, Mylène Rigal, Christophe Furger and Erwan Le Roux
Antioxidants 2024, 13(11), 1392; https://doi.org/10.3390/antiox13111392 - 14 Nov 2024
Viewed by 412
Abstract
Oenobiol Sun Expert, a food formulation designed to enhance skin health prior to sun exposure, has been optimized by incorporating the OenoGrape Advanced Complex, which includes grape pomace extract, increased selenium content and 10% lycopene-rich tomato extract, with these constituents exhibiting high antioxidant [...] Read more.
Oenobiol Sun Expert, a food formulation designed to enhance skin health prior to sun exposure, has been optimized by incorporating the OenoGrape Advanced Complex, which includes grape pomace extract, increased selenium content and 10% lycopene-rich tomato extract, with these constituents exhibiting high antioxidant potential. To evaluate the effects of these individual ingredients and the overall formulation at the cellular level, the AOP1 cell antioxidant efficacy assay was employed to measure the intracellular free radical scavenging activity, while the Cell Antioxidant Assay (CAA or DCFH-DA) assay was used to assess peroxidation scavenging at the plasma membrane level. The indirect antioxidant activity was examined using stably transfected cell lines containing a luciferase reporter gene controlled by the Antioxidant Response Element (ARE), which activates the endogenous antioxidant system via the Nrf2/Keap1-ARE pathway. Our results indicate that among the individual components, grape pomace extract and sodium selenite possess high and complementary antioxidant properties. Grape pomace extract was particularly effective in inhibiting free radicals (AOP1 EC50 = 6.80 μg/mL) and activating the ARE pathway (ARE EC50 = 231.1 μg/mL), whereas sodium selenite exerted its effects through potent ARE activation at sub-microgram levels (EC50 = 0.367 μg/mL). In contrast, the lycopene-rich tomato extract did not show a notable contribution to the antioxidant effects. The antiradical activity of the OenoGrape Advanced Complex, comprising these three ingredients, was very efficient and consistent with the results obtained for the individual components (AOP1 EC50 = 15.78 µg/mL and ARE EC50 of 707.7 μg/mL). Similarly, the free radical scavenging activity still persisted in the Oenobiol Sun Expert formulation (AOP1 EC50 = 36.63 µg/mL). Next, in vitro intestinal transepithelial transfer experiments were performed. The basolateral compartments of cells exposed to the ingredients were collected and assessed using the same antioxidant cell assays. The direct and indirect antioxidant activities were measured on both hepatocytes and keratinocytes, demonstrating the bioavailability and bioactivity of grape pomace extract and sodium selenite. These finding suggest that the ingredients of this food supplement contribute to enhanced cytoprotection following ingestion. Full article
(This article belongs to the Special Issue Antioxidant and Protective Effects of Plant Extracts—2nd Edition)
Show Figures

Figure 1

Figure 1
<p>The intracellular ROS scavenging activity of three individual ingredients was assessed on HepG2 cells using the AOP1 bioassay. HepG2 cells were incubated for 1 h with increasing concentrations of a 10% lycopene-rich tomato extract (<b>A</b>), sodium selenite (<b>B</b>) and grape pomace extract (<b>C</b>). Left panel: kinetic fluorescence profiles, where the <span class="html-italic">x</span>-axis represents the light flash number, and the <span class="html-italic">y</span>-axis displays the Relative Fluorescence Unit (RFU) values for each sample concentration. <b>Middle panel</b>: Antioxidant Index (AI) values calculated for each concentration. <b>Right panel</b>: dose–response curves with the log-transformed concentration on the <span class="html-italic">x</span>-axis and the AI on the <span class="html-italic">y</span>-axis. Data points: mean RFU value from triplicate wells; error bars: standard deviation (SD); EC<sub>50</sub>: efficacy concentration required to achieve 50% of the maximum effect; R<sup>2</sup>: coefficient of determination for the dose–response fit.</p>
Full article ">Figure 2
<p>Intracellular ROS scavenging activity on HepG2 cells was assessed for OenoGrape Advanced Complex, Oenobiol Sun Expert and Oenobiol Solaire Intensif formulations. HepG2 cells were incubated for 1 h with increasing concentrations of OenoGrape Advanced Complex (<b>A</b>), Oenobiol Sun Expert (<b>B</b>) and Oenobiol Solaire Intensif formulations (<b>C</b>). <b>Left panel:</b> kinetic profile of AOP1 biosensor fluorescence, where the <span class="html-italic">x</span>-axis represents light flashes, and the <span class="html-italic">y</span>-axis shows relative fluorescence unit (RFU) values recorded for each concentration. <b>Middle panel:</b> Antioxidant Index (AI) calculated for each concentration. <b>Right panel:</b> dose–response curves, with the <span class="html-italic">x</span>-axis representing the log-transformed concentration and the <span class="html-italic">y</span>-axis the Antioxidant Index (AI) values. Data points: mean RFU values from triplicate measurements; error bars: SD; EC<sub>50</sub>: efficacy concentration required for 50% efficacity; R<sup>2</sup>: coefficient of determination for the dose–response fit.</p>
Full article ">Figure 3
<p>The cell membrane radical scavenging activity of a 10% lycopene-rich extract, the OenoGrape Advanced Complex and Oenobiol Sun Expert formulation was evaluated in HepG2 cells using the CAA or AAPH/DCFH-DA assay. HepG2 cells were incubated for 4 h with varying concentrations of the 10% lycopene-rich tomato extract (<b>A</b>), OenoGrape Advanced Complex (<b>B</b>) and Oenobiol Sun Expert formulation (<b>C</b>). <b>Left panel:</b> fluorescence emission kinetics of the DCFH probe. <b>Middle panel:</b> Antioxidant Index (AI) calculated for each concentration. <b>Right panel:</b> dose–response curves. Data points: mean RFUs of triplicate wells; error bars: SD; EC<sub>50</sub>: efficacy concentrations at which 50% efficacity is observed; R<sup>2</sup>: coefficient of determination for the dose–response fit.</p>
Full article ">Figure 4
<p>The ARE transcriptional activity of the three individual ingredients was assessed on ARE-luc-HepG2 cells. ARE–luciferase–HepG2 cells were treated for 17 h with a range of concentrations of 10% lycopene-rich tomato extract (<b>A</b>), sodium selenite (<b>B</b>) and grape pomace extract (<b>C</b>), and the luciferase luminescence was measured as relative luminescence units. <b>Left panel:</b> the graphs display the luciferase gene expression as the fold increase (FI) relative to the vehicle control. <b>Right panel</b>: the dose–response curves are represented, where the log-transformed concentrations are plotted on the <span class="html-italic">x</span>-axis against the fold increase in the gene expression (FI). Data points: mean FI of duplicate measurements; error bars: SD; EC<sub>50</sub>: efficacy concentration required for 50% of the maximum effect; R<sup>2</sup>: coefficient of determination for the dose–response curve.</p>
Full article ">Figure 5
<p>The ARE transcriptional activity of the OenoGrape Advanced Complex, Oenobiol Sun Expert and Solaire Intensif formulations was evaluated on ARE-luc-HepG2 cells. ARE–luciferase–HepG2 cells were treated for 17 h with a range of concentrations of the OenoGrape Advanced Complex (<b>A</b>), Oenobiol Sun Expert (<b>B</b>) and Solaire Intensif (<b>C</b>) formulations, and the luciferase luminescence was measured. The left panel displays the fold increase in gene expression (FI) relative to the vehicle control, while the right panel presents the dose–response curves with the log-transformed concentrations plotted on the <span class="html-italic">x</span>-axis and the fold increase in gene expression (FI) on the <span class="html-italic">y</span>-axis. Data points: the mean fold increase in gene expression (FI) of duplicate measurements; error bars: SD; EC<sub>50</sub>: efficacy concentration required to achieve 50% of the maximum effect; R<sup>2</sup>: coefficient of determination for the dose–response fit.</p>
Full article ">Figure 6
<p>A comparative analysis of the ROS scavenging activity (AOP1 assay) in HepG2 cells was performed for the Oenobiol Sun Expert formulation, before and after intestinal transepithelial transfer. HepG2 cells were treated with increasing concentrations of raw Oenobiol Sun Expert (panel (<b>A</b>)) and with serial dilutions of the basolateral fractions obtained from Caco2 cells following a 1 h incubation with Oenobiol Sun Expert (panel (<b>B</b>)). Top panel: kinetic fluorescence profiles, with the <span class="html-italic">x</span>-axis representing the light flash number and the <span class="html-italic">y</span>-axis showing the normalized Relative Fluorescence Unit (RFU) values. Bottom panel: dose–response curves, where log concentrations or log dilutions are plotted on the <span class="html-italic">x</span>-axis and the Antioxidant Index on the <span class="html-italic">y</span>-axis. Data points: mean RFUs of triplicate measurements; error bars: SD; EC<sub>50</sub>: efficacy concentrations at 50% effect; R<sup>2</sup>: coefficient of determination.</p>
Full article ">Figure 7
<p>A comparison of the ROS scavenging activity (AOP1 assay) in HepG2 cells was conducted for grape pomace extract before and after intestinal transepithelial transfer. HepG2 cells were treated with increasing concentrations of raw grape pomace extract (panel (<b>A</b>)) and increasing dilutions of basolateral fractions collected from Caco2 cells following a 1 h incubation with either 25 mg/mL (panel (<b>B</b>)) or 6 mg/mL (panel (<b>C</b>)) grape pomace extract. Top panel: kinetic fluorescence profiles, with the light flash number on the <span class="html-italic">x</span>-axis and the normalized Relative Fluorescence Unit (RFU) values on the <span class="html-italic">y</span>-axis. Bottom panel: dose–response curves, with log concentrations or log dilutions plotted on the <span class="html-italic">x</span>-axis and the Antioxidant Index on the <span class="html-italic">y</span>-axis. Data points: mean RFUs from triplicate measurements; bars: SD; EC<sub>50</sub>: efficacy concentrations at 50% effect; R<sup>2</sup>: coefficient of determination.</p>
Full article ">Figure 8
<p>A comparison of the ROS scavenging activity (AOP1 assay) in HaCaT cells was performed for the Oenobiol Sun Expert formulation before and after intestinal transepithelial transfer. HaCaT cells were treated with increasing concentrations of raw Oenobiol Sun Expert (panel (<b>A</b>)) and increasing dilutions of basolateral fractions obtained from Caco2 cells after a 1 h incubation with Oenobiol Sun Expert (panel (<b>B</b>)). Top panel: kinetic fluorescence profiles, with the light flash number on the <span class="html-italic">x</span>-axis and the normalized Relative Fluorescence Unit (RFU) values on the <span class="html-italic">y</span>-axis. Bottom panel: dose–response curves, with log concentrations or log dilutions plotted on the <span class="html-italic">x</span>-axis and the Antioxidant Index on the <span class="html-italic">y</span>-axis. Data points: mean RFUs of triplicate measurements; bars: SD; EC<sub>50</sub>: efficacy concentrations at 50% effect; R<sup>2</sup>: coefficient of determination.</p>
Full article ">Figure 9
<p>A comparison of the ROS scavenging activity (AOP1 test) was conducted on HacaT cells for grape pomace extract before and after intestinal transepithelial transfer. HaCaT cells were treated with increasing concentrations of raw grape pomace extract (panel (<b>A</b>)) and increasing dilutions of basolateral fractions from Caco2 cells after a 1 h incubation with 25 mg/mL (panel (<b>B</b>)) or 6 mg/mL (panel (<b>C</b>)) of grape pomace extract. Top panel: kinetic fluorescence profiles, with the light flash number on the <span class="html-italic">x</span>-axis and the normalized Relative Fluorescence Unit (RFU) values on the <span class="html-italic">y</span>-axis. Bottom panel: dose–response curves with log concentrations or log dilutions plotted on the <span class="html-italic">x</span>-axis and the Antioxidant Index on the <span class="html-italic">y</span>-axis. Data points: mean RFUs of triplicate measurements; bars: SD; EC<sub>50</sub>: efficacy concentrations at 50% effect; R<sup>2</sup>: coefficient of determination.</p>
Full article ">Figure 10
<p>A comparison of ARE transcriptional activity comparison in ARE–luciferase–HepG2 cells was conducted for sodium selenite before and after intestinal transfer. ARE–luciferase–HepG2 cells were treated for 17 h with a range of sodium selenite concentrations (panel (<b>A</b>)) or dilutions of basolateral compartments collected from Caco2 cells after a 1 h incubation with 10 or 20 μg/mL sodium selenite (panel (<b>B</b>)). Luciferase luminescence was measured as an indicator of ARE pathway activation. The graphs depict the gene expression fold increase (FI) relative to the vehicle control for either decreasing concentrations of sodium selenite or varying dilutions of the basolateral fractions. Data points: mean fold increase (FI) of duplicate measurements; bars: SD.</p>
Full article ">Figure 11
<p>A comparison of ARE transcriptional activity in ARE–luciferase–HacaT cells was conducted for sodium selenite before and after intestinal transfer. ARE–luciferase–HacaT cells were treated for 17 h with a range of concentrations of sodium selenite (panel (<b>A</b>)) or dilutions of basolateral fractions collected from Caco2 cells following a 1 h incubation with 10 or 20 μg/mL sodium selenite (panel (<b>B</b>)). Luciferase luminescence was measured to assess ARE pathway activation. The graphs display the fold increase (FI) in gene expression compared to the vehicle control for either decreasing concentrations of sodium selenite or different dilutions of the basolateral fractions. Data points: mean fold increase (FI) of duplicate measurements; bars: SD.</p>
Full article ">
19 pages, 2688 KiB  
Review
Neuroprotective Potential of Glycyrrhizic Acid in Ischemic Stroke: Mechanisms and Therapeutic Prospects
by Yanwen Li, Juan Wu, Fang Du, Tao Tang, Jonathan Chee Woei Lim, Thilakavathy Karuppiah, Jiaxin Liu and Zhong Sun
Pharmaceuticals 2024, 17(11), 1493; https://doi.org/10.3390/ph17111493 - 7 Nov 2024
Viewed by 464
Abstract
Background/Objectives: Ischemic stroke is a leading cause of disability and mortality worldwide, with current therapies limited in addressing its complex pathophysiological mechanisms, such as inflammation, oxidative stress, apoptosis, and impaired autophagy. Glycyrrhizic acid (GA), a bioactive compound from licorice (Glycyrrhiza glabra L.), [...] Read more.
Background/Objectives: Ischemic stroke is a leading cause of disability and mortality worldwide, with current therapies limited in addressing its complex pathophysiological mechanisms, such as inflammation, oxidative stress, apoptosis, and impaired autophagy. Glycyrrhizic acid (GA), a bioactive compound from licorice (Glycyrrhiza glabra L.), has demonstrated neuroprotective properties in preclinical studies. This review consolidates current evidence on GA’s pharmacological mechanisms and assesses its potential as a therapeutic agent for ischemic stroke. Methods: This review examines findings from recent preclinical studies and reviews on GA’s neuroprotective effects, focusing on its modulation of inflammation, oxidative stress, apoptosis, and autophagy. Studies were identified from major scientific databases, including PubMed, Web of Science, and Embase, covering research from January 2000 to August 2024. Results: GA has demonstrated significant neuroprotective effects through the modulation of key pathways, including HMGB1/TLR4/NF-κB and Keap1/Nrf2, thereby reducing neuroinflammation, oxidative stress, and apoptosis. Additionally, GA promotes autophagy and modulates immune responses, suggesting it could serve as an adjunct therapy to enhance the efficacy and safety of existing treatments, such as thrombolysis. Conclusions: Current findings underscore GA’s potential as a multi-targeted neuroprotective agent in ischemic stroke, highlighting its anti-inflammatory, antioxidant, and anti-apoptotic properties. However, while preclinical data are promising, further clinical trials are necessary to validate GA’s therapeutic potential in humans. This review provides a comprehensive overview of GA’s mechanisms of action, proposing directions for future research to explore its role in ischemic stroke management. Full article
Show Figures

Figure 1

Figure 1
<p>Structural configurations of glycyrrhizic acid stereoisomers. (<b>a</b>) 18α-Glycyrrhizic Acid: The α stereoisomer of glycyrrhizic acid, featuring a specific spatial arrangement around the 18th carbon atom. (<b>b</b>) 18β-Glycyrrhizic Acid: The β stereoisomer, which is the predominant and biologically active form found in licorice root. Glycyrrhizic acid generally refers to this 18β form, known for its anti-inflammatory, antiviral, and hepatoprotective properties. Structural diagrams were downloaded from PubChem.</p>
Full article ">Figure 2
<p>PRISMA flow diagram illustrating the study selection process.</p>
Full article ">Figure 3
<p>Schematic illustration showing the anti-inflammatory and antioxidant effects of GA. On the left, GA inhibits the binding of HMGB1 to TLR4 and RAGE receptors, blocking downstream signaling cascades such as MyD88/TRAF6 and NF-κB activation, thereby reducing pro-inflammatory cytokine production. On the right, GA activates the Keap1/Nrf2 pathway, leading to increased expression of antioxidant enzymes (e.g., HO-1 and SOD), which reduce ROS and mitigate oxidative stress. Red T-shapes represent the inhibition of molecule expression, red and black arrows represent the promotion of molecule expression, and dashed black arrows indicate molecules with potential promotive influence.</p>
Full article ">Figure 4
<p>Schematic representation of GA’s role in regulating apoptosis and autophagy. On the left, GA inhibits mitochondrial cytochrome c release and caspase-3 activation by modulating the Bax/Bcl-2 ratio, thus reducing neuronal apoptosis. On the right, GA promotes autophagy via the Keap1/Nrf2 and PINK1/Parkin pathway, facilitating the clearance of damaged mitochondria and reducing oxidative stress, contributing to enhanced neuronal survival. Red and black T-shapes represent the inhibition of molecule expression, red and black arrows indicate the promotion of molecule expression, and dotted red arrows represent molecules that may promote additional effects.</p>
Full article ">
25 pages, 2416 KiB  
Review
The Impact of Genetic Mutations on the Efficacy of Immunotherapies in Lung Cancer
by Ki Lui, Kwok-Kuen Cheung, Winnie Wing-Man Ng, Yanping Wang, Doreen W. H. Au and William C. Cho
Int. J. Mol. Sci. 2024, 25(22), 11954; https://doi.org/10.3390/ijms252211954 - 7 Nov 2024
Viewed by 802
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide, primarily driven by genetic mutations. The most common genetic alterations implicated in lung cancer include mutations in TP53, KRAS, KEAP1, NF1, EGFR, NRF2, ATM, ALK, [...] Read more.
Lung cancer is the leading cause of cancer-related mortality worldwide, primarily driven by genetic mutations. The most common genetic alterations implicated in lung cancer include mutations in TP53, KRAS, KEAP1, NF1, EGFR, NRF2, ATM, ALK, Rb1, BRAF, MET, and ERBB2. Targeted therapies have been developed to inhibit cancer growth by focusing on these specific genetic mutations. However, either the mutations are undruggable or the efficacy of these therapies is often compromised over time due to the emergence of drug resistance, which can occur through additional mutations in the targeted protein or alternative growth signaling pathways. In recent years, immunotherapy has emerged as a promising approach to enhance the effectiveness of cancer treatment by leveraging the body’s immune system. Notable advancements include immune checkpoint inhibitors, monoclonal antibodies targeting cell surface receptors, antibody–drug conjugates, and bispecific antibodies. This review provides an overview of the mechanisms of FDA-approved immunotherapeutic drugs, offering an updated perspective on the current state and future developments in lung cancer therapy. More importantly, the factors that positively and negatively impact the immunotherapy’s efficacy will also be discussed. Full article
(This article belongs to the Special Issue Challenges and Future Perspectives in Treatment for Lung Cancer)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The mechanism of immunosuppression is mediated by PD-1/PD-L1 binding, and checkpoint inhibitors block PD-1/PD-L1 interaction and induce immuno-activation. The T cell receptor (TCR) identifies non-self-antigens and is activated by the antigen–MHC complex displayed on cancer cells. However, as depicted in (<b>A</b>), the T cell is suppressed when the PD-1 receptor on the T cell interacts with the PD-L1 antigen on the cancer cell. Checkpoint inhibitors can attenuate this suppression, which binds to the PD-1 receptor (represented by the blue antibody) or the PD-L1 antigen (represented by the green antibody). The binding masks the interaction sites, preventing the association between PD-1 and PD-L1. Consequently, the inhibitory effect on the T cell is lifted, allowing the T cell to initiate an attack on the cancer cell, as illustrated in (<b>B</b>).</p>
Full article ">Figure 2
<p>Mechanism of monoclonal antibody targeting in cancer therapy. The monoclonal antibody binds to the extracellular domain of cancer cell surface receptors, forming a complex that is subsequently internalized into the lysosome for degradation. Additionally, the antibody–receptor complex activates an immune response involving T cells and dendritic cells.</p>
Full article ">Figure 3
<p><b>Schematic of an antibody–drug conjugate (ADC).</b> The ADC is composed of a monoclonal antibody linked to a cytotoxic drug. The monoclonal antibody specifically targets a cell surface receptor. Upon binding to the receptor, the antibody–receptor complex is internalized into the cell and directed to lysosomes. The complex undergoes degradation within the lysosome, releasing the cytotoxic drug, which subsequently kills cancer.</p>
Full article ">Figure 4
<p><b>Working principle of a bispecific antibody.</b> Bispecific antibodies are engineered in the form of Immunoglobulin G (IgG) through genetic engineering. One-half of the IgG is designed to recognize the T cell’s antigen on cytotoxic T cells, while the other half is tailored to identify a specific antigen in cancer cells. This specificity allows the bispecific antibody to target any antigen of interest specific to cancer antigen in a specific patient. The function of the bispecific antibody is to bridge the T cell and cancer cell, facilitating close proximity for T cell-mediated cancer destruction. Additionally, the innate immune system, such as macrophages or NK cells, uses the Fc receptor to recognize the bispecific antibody and subsequently releases cytokines to enhance cancer elimination or by direct cancer attack.</p>
Full article ">
14 pages, 2475 KiB  
Article
PTD-FNK Alleviated LPS-Induced Oxidative Stress of Boar Testicular Sertoli Cells via Keap1-Nrf2 Pathway
by Weixia Ji, Qiuyan Huang, Qiqi Ma, Xingxing Song, Xin Zhang, Xun Li, Xiaoye Wang, Sutian Wang, Yanling Wang, Zhengzhong Xiao and Chuanhuo Hu
Vet. Sci. 2024, 11(11), 543; https://doi.org/10.3390/vetsci11110543 - 6 Nov 2024
Viewed by 652
Abstract
PTD-FNK, a synthetic anti-apoptotic protein, has been shown to potently alleviate cellular injuries. However, the effects of PTD-FNK on oxidative defense in boar testicular Sertoli cells (SCs) against oxidative injury has not been explored. In this study, we show that exposure of SCs [...] Read more.
PTD-FNK, a synthetic anti-apoptotic protein, has been shown to potently alleviate cellular injuries. However, the effects of PTD-FNK on oxidative defense in boar testicular Sertoli cells (SCs) against oxidative injury has not been explored. In this study, we show that exposure of SCs to 100 mg/L lipopolysaccharide (LPS) for 12 h leads to decreased survival rate, superoxide dismutase (SOD) activity, and increased malondialdehyde (MDA). Treatment with 0.01 nmol/L PTD-FNK for 4 h significantly enhanced the activity of SOD, catalase (CAT), glutathione peroxidase (GSH-Px), and total antioxidant capacity (T-AOC) in SCs. Concurrently, PTD-FNK treatment effectively reduced the production of reactive oxygen species (ROS) and the levels of 8-hydroxy-2′-deoxyguanosine (8-OHdG) in SCs. Moreover, using His pull-down and LC-MS techniques, we identified PTD-FNK-interacting proteins and confirmed that this protective effect may be mediated by the regulation of the Keap1-Nrf2 signaling pathway by PTD-FNK. Therefore, PTD-FNK alleviates LPS-induced oxidative stress via the Keap1/Nrf2 pathway, providing novel insights for the development of therapeutic agents targeting testicular oxidative damage. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>,<b>B</b>) The morphology of SCs was identified by Oil Red O staining; (<b>C</b>) arrows indicate the bipolar corpuscles in the nucleus. The SC marker gene was detected by PCR. 1.<span class="html-italic">GATA4</span>; 2.<span class="html-italic">SOX9</span>; M.DL2000 Marker.</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) Effects of PTD-FNK on ROS production of LPS-induced boar SCs; (<b>C</b>) CCK8 assays of the PTD-FNK effect on cell viability; (<b>D</b>–<b>G</b>) effects of PTD-FNK on GSH-Px, CAT, 8-OHdG, and T-AOC contents of LPS-induced boar SCs; (<b>H</b>–<b>J</b>) effects of PTD-FNK on mRNA expression of GSH-Px, CAT, and SOD in LPS-induced boar SCs. <span class="html-italic">n</span> ≥ 3 (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt;0.001; **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 3
<p>(<b>A</b>) His recombinant protein was detected by CBB; (<b>B</b>) His recombinant protein was detected by WB; (<b>C</b>) the expression of SCs His antibody was detected by WB; (<b>D</b>) “silver stain” detection result after pull-down; the red arrow represents the bait protein; the black arrows indicate the PTD-FNK interacting proteins; (<b>E</b>) Venn diagram of PTD-FNK differential protein sets in boar SCs; (<b>F</b>,<b>G</b>) PTD-FNK effects on <span class="html-italic">HSPA5</span> and <span class="html-italic">VIM</span> mRNA expression in boar SCs. <span class="html-italic">n</span> ≥ 3 ****<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 4
<p>(<b>A</b>–<b>E</b>) The relative protein level of Nrf2, Keap1, HO-1, and <span class="html-italic">NQO1</span>; (<b>F</b>–<b>G</b>) Nrf2 and Keap1 protein levels were measured using ELISA; (<b>H</b>–<b>K</b>) <span class="html-italic">Nrf2</span>, <span class="html-italic">Keap1</span>, <span class="html-italic">HO-1</span>, and <span class="html-italic">NQO1</span> mRNA levels; control, no drug added; LPS, induced by 100 mg/L LPS for 12 h; LPS + PTD-FNK, protected by 0.01 mmol/L PTD-FNK for 4 h and then induced by 100 mg/L LPS for 12 h; LPS+ML385, induced by 5μM ML385 for 2 h, followed induced by 100 mg/L LPS for 12 h; LPS + PTD-FNK + ML385, induced by 5μM ML385 for 2 h, protected by 0.01 mmol /L PTD-FNK for 4 h, and then induced by 100 mg/L LPS for 12 h. <span class="html-italic">n</span> ≥ 3 (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
19 pages, 4911 KiB  
Article
Artemisia Ordosica Polysaccharides Enhance Antioxidant Capacity of Peripheral Blood Lymphocytes in Poultry Through Nrf2/Keap1 and TLR4/NF-κB Signal Pathway
by Yuanyuan Xing, Yankai Zheng, Lu Chen, Yuanqing Xu, Xiao Jin, Li Hong, Sumei Yan and Binlin Shi
Antioxidants 2024, 13(11), 1308; https://doi.org/10.3390/antiox13111308 - 28 Oct 2024
Viewed by 398
Abstract
Artemisia ordosica polysaccharides (AOP) can promote animal growth, improve intestinal morphology, regulate immunity, and enhance antioxidant capacity. To investigate the antioxidant capacity of AOP, three experiments were conducted. (1) Different concentrations of AOP (0, 50, 100, 150, 200, and 250 μg/mL) and 1 [...] Read more.
Artemisia ordosica polysaccharides (AOP) can promote animal growth, improve intestinal morphology, regulate immunity, and enhance antioxidant capacity. To investigate the antioxidant capacity of AOP, three experiments were conducted. (1) Different concentrations of AOP (0, 50, 100, 150, 200, and 250 μg/mL) and 1 µg/mL VA on peripheral blood lymphocytes (PBLs) treated with/without lipopolysaccharides (LPS) were investigated to select the optimum concentration. The results showed that 150 μg/mL AOP had significant antioxidation activity. (2) The PBLs was randomly divided into eight treatments with six replicates, namely CON, AOP, LPS, ML385 (Nrf2 inhibitor), AOP + LPS, AOP + ML385, LPS + ML385 and LPS + ML385 + AOP. The results showed that under a normal condition or stress condition, AOP presented antioxidation activity via upregulating Nrf2/Keap1 pathway-related gene expression. (3) The PBLs was randomly divided into eight treatments with six replicates, namely CON, AOP, LPS, PDTC (NF-κB inhibitor), AOP + LPS, AOP + PDTC, LPS + PDTC and LPS + PDTC + AOP. The results showed that under a normal condition, AOP presented antioxidation activity via increasing TLR4/NF-κB pathway-related gene expression; under a stress condition, AOP alleviated oxidative damage caused by LPS via suppressing TLR4/NF-κB pathway-related gene expression. Full article
(This article belongs to the Special Issue Antioxidant and Protective Effects of Plant Extracts—2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Effects of AOP on the cell viability of PBLs (%). Note: Different concentration of AOP (0, 50, 100, 150, 200, and 250 μg/mL) and 1 µg/mL VA were supplemented to the culture medium for 24 h. Afterward, each treatment was further divided into two groups: one group with the addition of 10 μg/mL LPS as the stress group, and the other as the non-stress group, with continued cultivation for 6 h. Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">Figure 2
<p>Effects of AOP on the antioxidant enzyme activity and oxidative stress metabolites in PBLs. Note: (<b>A</b>–<b>H</b>): SOD, superoxide dismutase; GPx, glutathione peroxidase; CAT, catalase; T-AOC, total antioxidant capacity; MDA, malondialdehyde; PC, protein carbonyl; ROS, reactive oxygen species; 8-OHdG, 8-hydroxy-2′-deoxyguanosine. Different concentrations of AOP (0, 50, 100, 150, 200, and 250 μg/mL) and 1 µg/mL VA were supplemented to the culture medium for 24 h. Afterward, each treatment was further divided into two groups: one group with the addition of 10 μg/mL LPS as the stress group, and the other as the non-stress group, with continued cultivation for 6 h. Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">Figure 3
<p>Effects of AOP on the Nrf2 signaling pathway-related gene expression in PBLs. Note: (<b>A</b>–<b>E</b>): <span class="html-italic">Nrf2</span>, nuclear factor erythroid-2-related factor 2; <span class="html-italic">Keap1</span>, Kelch-like ECH-associated protein 1; <span class="html-italic">GPx</span>, glutathione peroxidase; <span class="html-italic">SOD</span>, superoxide dismutase; <span class="html-italic">CAT</span>, catalase. Different concentrations of AOP (0, 50, 100, 150, 200, and 250 μg/mL) and 1 µg/mL VA were supplemented to the culture medium for 24 h. Afterward, each treatment was further divided into two groups: one group with the addition of 10 μg/mL LPS as the stress group, and the other as the non-stress group, with continued cultivation for 6 h. The gene expression for β-actin was used as a housekeeping gene. The relative expression levels from the control group were used as reference values. Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">Figure 4
<p>Effects of AOP on the NF-κB signaling pathway-related gene expression in PBLs. Note: (<b>A</b>–<b>G</b>): <span class="html-italic">TLR4</span>, toll-like receptor 4; <span class="html-italic">MyD88</span>, myeloid differentiation primary response 88; <span class="html-italic">IKKβ</span>, inhibitory kappa B kinase beta; <span class="html-italic">IκBα</span>, inhibitor of NF-κB alpha; <span class="html-italic">NF-κB p65</span>, nuclear factor kappa-B p65; <span class="html-italic">IL-1β</span>, interleukin-1β and <span class="html-italic">IL-6</span>, interleukin-6. Different concentrations of AOP (0, 50, 100, 150, 200, and 250 μg/mL) and 1 µg/mL VA were supplemented to the culture medium for 24 h. Afterward, each treatment was further divided into two groups: one group with the addition of 10 μg/mL LPS as the stress group, and the other as the non-stress group, with continued cultivation for 6 h. The gene expression for β-actin was used as a housekeeping gene. The relative expression levels from the control group were used as reference values. Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">Figure 5
<p>Effects of AOP on the antioxidative enzymes and oxidative stress metabolites in PBLs challenged by LPS and blocked by ML385. Note: (<b>A</b>–<b>H</b>): GPx, glutathione peroxidase; SOD, superoxide dismutase; CAT, catalase; T-AOC, total antioxidant capacity; MDA, malondialdehyde; PC, protein carbonyl; ROS, reactive oxygen species; 8-OHdG, 8-hydroxy-2′-deoxyguanosine. CON (control group), LPS (lipopolysaccharide), AOP (Artemisia ordosica polysaccharides), and ML385 (Nrf2 inhibitor). Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">Figure 6
<p>Effects of AOP on the gene expression of the Nrf2 signaling pathway in PBLs challenged by LPS and blocked by ML385. Note: (<b>A</b>–<b>E</b>): <span class="html-italic">Nrf2</span>, nuclear factor erythroid-2-related factor 2; <span class="html-italic">Keap1</span>, Kelch-like ECH-associated protein 1; <span class="html-italic">GPx</span>, glutathione peroxidase; <span class="html-italic">SOD</span>, superoxide dismutase; <span class="html-italic">CAT</span>, catalase. CON (control group), LPS (lipopolysaccharide), AOP (Artemisia ordosica polysaccharides), and ML385 (Nrf2 inhibitor). The gene expression for β-actin was used as a housekeeping gene. The relative expression levels from the control group were used as reference values. Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">Figure 7
<p>Effects of AOP on the gene expression of the NF-κB signaling pathway in PBLs challenged by LPS and blocked by ML385. Note: (<b>A</b>–<b>G</b>): <span class="html-italic">TLR4</span>, toll-like receptor 4; <span class="html-italic">MyD88</span>, myeloid differentiation primary response 88; <span class="html-italic">IKKβ</span>, inhibitory kappa B kinase beta; <span class="html-italic">IκBα</span>, inhibitor of NF-κB alpha; <span class="html-italic">NF-κB p65</span>, nuclear factor kappa-B p65; <span class="html-italic">IL-1β</span>, interleukin-1β; <span class="html-italic">IL-6</span>, interleukin-6. CON (control group), LPS (lipopolysaccharide), AOP (Artemisia ordosica polysaccharides), and ML385 (Nrf2 inhibitor). β-actin was used as a housekeeping gene. The relative expression levels from the control group were used as reference values. Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">Figure 8
<p>Effects of AOP on the antioxidative enzymes and oxidative stress metabolites in PBLs challenged by LPS and blocked by PDTC. Note: (<b>A</b>–<b>H</b>): GPx, glutathione peroxidase; SOD, superoxide dismutase; CAT, catalase; T-AOC, total antioxidant capacity; MDA, malondialdehyde; PC, protein carbonyl; ROS, reactive oxygen species; 8-OHdG, 8-hydroxy-2′-deoxyguanosine. CON (control group), LPS (lipopolysaccharide), AOP (Artemisia ordosica polysaccharides), and PDTC (NF-κB inhibitor). Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">Figure 9
<p>Effects of AOP on the gene expression of the Nrf2 Signaling pathway in PBLs challenged by LPS and blocked by PDTC. Note: (<b>A</b>–<b>E</b>): <span class="html-italic">Nrf2</span>, nuclear factor erythroid-2-related factor 2; <span class="html-italic">Keap1</span>, Kelch-like ECH-associated protein 1; <span class="html-italic">GPx</span>, glutathione peroxidase; <span class="html-italic">SOD</span>, superoxide dismutase; <span class="html-italic">CAT</span>, catalase. CON (control group), LPS (lipopolysaccharide), AOP (Artemisia ordosica polysaccharides), and PDTC (NF-κB inhibitor). β-actin was used as a housekeeping gene. The relative expression levels from the control group were used as reference values. Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">Figure 10
<p>Effects of AOP on the gene expression of the NF-κB signaling pathway in PBLs challenged by LPS and blocked by PDTC. Note: (<b>A</b>–<b>G</b>): <span class="html-italic">TLR4</span>, toll-like receptor 4; <span class="html-italic">MyD88</span>, myeloid differentiation primary response 88; <span class="html-italic">IKKβ</span>, inhibitory kappa B kinase beta; <span class="html-italic">IκBα</span>, inhibitor of NF-κB alpha; <span class="html-italic">NF-κB p65</span>, nuclear factor kappa-B p65; <span class="html-italic">IL-1β</span>, interleukin-1β; <span class="html-italic">IL-6</span>, interleukin-6. CON (control group), LPS (lipopolysaccharide), AOP (Artemisia ordosica polysaccharides), and PDTC (NF-κB inhibitor). β-actin was used as a housekeeping gene. The relative expression levels from the control group were used as reference values. Each value is shown as the mean ± SEM (n = 6); * <span class="html-italic">p</span> &lt; 0.05 or vs. control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. LPS group.</p>
Full article ">
21 pages, 5821 KiB  
Review
Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis
by Giovanni Tossetta, Sonia Fantone, Lucrezia Togni, Andrea Santarelli, Fabiola Olivieri, Daniela Marzioni and Maria Rita Rippo
Antioxidants 2024, 13(10), 1270; https://doi.org/10.3390/antiox13101270 - 18 Oct 2024
Viewed by 1065
Abstract
Periodontitis affects up to 40% of adults over 60 years old and is a consequence of gingivitis. Periodontitis is characterized by a chronic inflammation, periodontal damage, and alveolar bone resorption. The nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2)/Kelch-like ECH-Associated Protein 1 [...] Read more.
Periodontitis affects up to 40% of adults over 60 years old and is a consequence of gingivitis. Periodontitis is characterized by a chronic inflammation, periodontal damage, and alveolar bone resorption. The nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2)/Kelch-like ECH-Associated Protein 1 (KEAP1) (NRF2/KEAP1) signaling pathway plays a key role in periodontitis by modulating redox balance and inflammation of the periodontium. However, NRF2 expression is decreased in gingival tissues of patients with periodontitis while oxidative stress is significantly increased in this pathology. Oxidative stress and lipopolysaccharide (LPS) produced by gram-negative bacteria favor the production of inflammatory causing periodontal inflammation and favoring alveolar bone. In this review, we analyzed the current literature regarding the role of natural and synthetic compounds in modulating the NRF2/KEAP1 pathway in in vitro and in vivo models of periodontitis in order to evaluate new potential treatments of periodontitis that can improve the outcome of this disease. Full article
Show Figures

Figure 1

Figure 1
<p>Pathogenesis of periodontitis. If gingivitis remains unresolved, there is a persistence of pathogens and inflammation that leads to different grades of periodontitis. CAL = clinical attachment loss; PD = probing depth.</p>
Full article ">Figure 2
<p>NRF2/KEAP1 signaling regulation. Under the basal condition, NRF2 is bound to the KEAP1/CUL3/RBX1 E3-Ub ligase complex that induces NRF2 proteasomal degradation. Under oxidant stimuli, ROS oxidate the cysteine residues of KEAP1, causing a conformational change that inhibits NRF2 ubiquitination/degradation. Since NRF2 avoids proteasomal degradation, it can migrate into the nucleus and bind ARE regions present in the upstream regulatory region (promoter) of several antioxidant genes, causing their transcription. ARE, antioxidant response element; Cul3, Cullin 3; E3, Ubiquitin ligase 3; KEAP1, Kelch Like ECH Associated Protein 1; NRF2, Nuclear Factor Erythroid 2-Related Factor 2; RBX1, RING box protein 1; Ub, Ubiquitin.</p>
Full article ">Figure 3
<p>Molecular structures of dehydrocostus lactone and hesperetin. The chemical structures of the compounds illustrated in this table have been taken from ChemSpider free database (<a href="https://www.chemspider.com" target="_blank">https://www.chemspider.com</a> (accessed on 30 September 2024)).</p>
Full article ">Figure 4
<p>Molecular structures of baicalein and magnolol. The chemical structures of the compounds illustrated in this figure have been taken from ChemSpider free database (<a href="https://www.chemspider.com" target="_blank">https://www.chemspider.com</a> (accessed on 30 September 2024)).</p>
Full article ">Figure 5
<p>Modulation of NRF2/KEAP1 signaling by phytotherapeutics in periodontitis. Phytotherapeutics (in light blue) increase NRF2 expression, favoring its nuclear translocation and binding to the ARE regions present in the promoter of antioxidant genes (NQO1, CAT, HO-1, SOD, etc.), inducing their transcription. The increased expression of HO-1 inhibits NF-κB activation, thus reducing inflammatory cytokines production (in green). The activation of NRF2 also increases GSH levels, reducing ROS levels and DNA damage (in blue). Moreover, the activation of NRF2 reduces cell senescence, osteoclast differentiation, and alveolar bone loss while favoring osteoblast differentiation, osteogenesis, and periodontal tissue repair (in red). ARE, antioxidant response element; CAT, catalase; GSH, glutathione; HO-1, Heme-oxygenase 1; IL, interleukin; NQO1, NAD(P)H:quinone oxidoreductase; NRF2, Nuclear Factor Erythroid 2-Related Factor 2; SOD, superoxide dismutase; TNF-α, Tumor Necrosis Factor-α.</p>
Full article ">
21 pages, 3907 KiB  
Article
Synthesis and Bioevaluation of New Stable Derivatives of Chrysin-8-C-Glucoside That Modulate the Antioxidant Keap1/Nrf2/HO-1 Pathway in Human Macrophages
by Javier Ávila-Román, Lirenny Quevedo-Tinoco, Antonio J. Oliveros-Ortiz, Sara García-Gil, Gabriela Rodríguez-García, Virginia Motilva, Mario A. Gómez-Hurtado and Elena Talero
Pharmaceuticals 2024, 17(10), 1388; https://doi.org/10.3390/ph17101388 - 17 Oct 2024
Viewed by 675
Abstract
Background/Objectives: The beneficial effects of the flavonoid chrysin can be reduced by its poor oral bioavailability. It has been shown that chrysin-8-C-glucoside (1) has a better absorption capability. The aim of this study was to evaluate the antioxidant and anti-inflammatory [...] Read more.
Background/Objectives: The beneficial effects of the flavonoid chrysin can be reduced by its poor oral bioavailability. It has been shown that chrysin-8-C-glucoside (1) has a better absorption capability. The aim of this study was to evaluate the antioxidant and anti-inflammatory activity of this glucoside, as well as the respective hexa-acetate derivative 1a and the hexa-ethyl carbonate derivative 1b since the inclusion of moieties in bioactive molecules may increase or modify their biological effects. Methods: THP-1 macrophages were used to determine the viability in the presence of chrysin derivatives, and non-cytotoxic concentrations were selected. Subsequently, lipopolysaccharide (LPS)-induced reactive oxygen species (ROS) production and inflammatory mediators were examined. The involvement of chrysin derivatives with the Keap1 and Nrf2 antioxidant system was determined by docking and Western blotting studies. Results: Our data demonstrated, for the first time, that pretreatment with the three compounds caused a significant reduction in LPS-induced reactive oxygen species (ROS) production and pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and interleukin 1β (IL-1β) levels, as well as in cyclooxygenase 2 (COX-2) expression. The mechanisms underlying these protective effects were related, at least in part, to the competitive molecular interactions of these phenolic compounds with Kelch-like ECH-associated protein 1 (Keap1)–nuclear factor erythroid 2-related factor 2 (Nrf2), which would allow the dissociation of Nrf2 and its translocation into the nucleus and the subsequent up-regulation of hemo-oxygenase 1 (HO-1) expression. Conclusions: Compared to the 8-C-glucoside parent chrysin, compound 1a exhibited the strongest antioxidant and anti-inflammatory activity. We hypothesized that the incorporation of an acetate group (1a) may reduce its polarity and, thus, increase membrane permeability, leading to better pharmacological activity. These findings support the potential use of these phenolic compounds as Nrf2 activators against oxidative-stress-related inflammatory diseases. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Formulas of chrysin-8-<span class="html-italic">C</span>-glucoside (<b>1</b>) and the respective hexa-acetate derivative (<b>1a</b>) and the hexa-ethyl carbonate derivative (<b>1b</b>).</p>
Full article ">Figure 2
<p>Effect of phenolics <b>1</b>, <b>1a</b>, and <b>1b</b> on the intracellular ROS production in THP-1 macrophages stimulated by LPS. These cells were treated with concentrations of 50 and 100 µM for compounds <b>1</b> and <b>1b</b> and 5 and 10 µM for compound <b>1a</b> for 1 h and then stimulated with 1 µg/mL LPS for 24 h. The positive reference drug dexamethasone (Dex) was used at 1 µM. Phenolics reduced the intracellular ROS production in a concentration-dependent manner even below Control levels at the higher tested concentrations. Values are representative of four independent experiments (<span class="html-italic">n</span> = 4). Data are arithmetic means ± SEM plotted by vertical bars. Mean value was significantly different vs. Control (++ <span class="html-italic">p</span> &lt; 0.01; Student’s <span class="html-italic">t</span> test). Arithmetic mean values were significantly different in comparison with LPS (* <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001; Kruskal–Wallis test followed by Dunn’s Multiple Comparison test.</p>
Full article ">Figure 3
<p>Effect of phenolics <b>1</b>, <b>1a,</b> and <b>1b</b> on LPS-induced TNF-α (<b>A</b>) and IL-1β (<b>B</b>) production in THP-1 macrophages. These cells were treated with the concentrations of 10, 50 and 100 µM for the compounds <b>1</b> and <b>1b</b> and 1, 5, and 10 µM for the compound <b>1a</b>, for 1 h and then stimulated with 1 µg/mL LPS for 24 h. Cytokines were quantified in cellular supernatants by using ELISA assay. The positive reference drug dexamethasone (Dex) was used at 1 μM. Values are arithmetic means ± SEM from six independent experiments (<span class="html-italic">n</span> = 6). Data are arithmetic means ± SEM plotted by vertical bars. Mean value was significantly different vs. Control (+++ <span class="html-italic">p</span> &lt; 0.001; Student’s <span class="html-italic">t</span> test). Arithmetic mean values were significantly different in comparison with LPS group (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; one-way ANOVA followed by Bonferroni’s Multiple Comparison test).</p>
Full article ">Figure 4
<p>Effect of phenolics <b>1</b>, <b>1a</b>, and <b>1b</b> on the pro-inflammatory <span class="html-italic">COX-2</span> expression and the <span class="html-italic">Nrf2/HO-1</span> signaling pathway in THP-1 macrophages stimulated by LPS. These cells were treated with concentrations of 10, 50, and 100 µM for compounds <b>1</b> and <b>1b</b> and 1, 5, and 10 µM for compound <b>1a</b> for 1 h and then stimulated with 1 µg/mL LPS for 24 h. The positive reference drug dexamethasone (Dex) was used at 1 μM. (<b>A</b>) Representative Western blotting analysis of COX-2, Nrf2, and HO-1 proteins from the same membrane. Densitometric analysis of COX-2 (<b>B</b>), Nrf2 (<b>C</b>), and HO-1 (<b>D</b>) carrying out normalization to β-actin. Results are representative of four independent experiments (<span class="html-italic">n</span> = 4). Values are arithmetic means ± SEM plotted by vertical bars. Arithmetic mean value was significantly different vs. Control (+++ <span class="html-italic">p</span> &lt; 0.001, ++ <span class="html-italic">p</span> &lt; 0.01; Student’s <span class="html-italic">t</span> test). Arithmetic mean values were significantly different in comparison with the LPS group (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; Kruskal–Wallis test followed by Dunn’s Multiple Comparison test).</p>
Full article ">Figure 5
<p>Molecular interactions of compounds <b>1</b>, <b>1a</b>, and <b>1b</b> with Keap1 calculated by docking protocols. The protein data (PDB ID: 4ZY3) were extracted from the Protein Data Bank website (<a href="https://www.rcsb.org/" target="_blank">https://www.rcsb.org/</a>). The docking protocol included the UCSF Quimera 1.15, Autodock Tools 1.5.6, and AutoDock Vina software. The molecular models of ligands <b>1</b>, <b>1a</b>, and <b>1b</b> involved in the docking study were geometry-optimized global minimum conformers, which were found by the Monte Carlo search protocol in the MMFF94 force field, as executed in the Spartan’04 program, followed by geometry optimization by the DFT B3LYP/DGDZVP level of theory in the Gaussian 16 software. (<b>A</b>) One pose was determined for glucoside flavonoid <b>1</b>, while two poses were found for derivative <b>1a</b> represented by (<b>B</b>) <b>1a</b>P1 and (<b>C</b>) <b>1a</b>P2. Compound <b>1b</b> showed three poses including (<b>D</b>) <b>1b</b>P1, (<b>E</b>) <b>1b</b>P2, and (<b>F</b>) <b>1b</b>P3.</p>
Full article ">Figure 6
<p>Docking models of compound <b>1</b>. The <b>left</b> image shows the Kelch domain of Keap1, which is a segment of the protein constituted by the 6-bladed <span class="html-italic">β</span>-propeller structure interacting with compound <b>1</b>. The <b>right</b> image shows the rings A and B from the flavonoid skeleton providing recurrent molecular interactions with Ala556, Arg415, and Asn382 amino acids from Keap1.</p>
Full article ">Figure 7
<p>Proposed diagram for the anti-inflammatory activity of phenolics <b>1</b>, <b>1a</b>, and <b>1b</b> through the interaction with the Keap1/Nrf2/HO-1 signaling pathway. In homeostatic conditions, the Keap1–Nrf2 system is placed joined to the cell’s internal hemimembrane. In these conditions, inactive Nrf2 is occasionally ubiquitinated and is degraded in the proteosome. In oxidative conditions, the intracellular ROS production and the inflammatory process are triggered, and the Nrf2 signaling pathway may be activated to control ROS-induced damage. Then, Nrf2 is dissociated from the Keap1 dimer and migrates into the nucleus to provide the transcription of its target gene, HO-1. This fact leads to the down-regulation of ROS production and inflammatory processes by inhibiting TNF-α, IL-1β, and COX-2 levels. In oxidative stress conditions, phenolics <b>1</b>, <b>1a</b>, and <b>1b</b> may be incorporated into the cell and act as Keap1 ligands, causing Nrf2 not to bind to or dissociate from Keap1, leading to further Nrf2 activation.</p>
Full article ">Figure 8
<p>Methodology for the obtention and chemical characterization of <b>1</b>, <b>1a</b>, and <b>1b</b>.</p>
Full article ">
15 pages, 6127 KiB  
Article
Deer Blood Hydrolysate Protects against D-Galactose-Induced Premature Ovarian Failure in Mice by Inhibiting Oxidative Stress and Apoptosis
by Yu Wang, Hongyan Pei, Weijia Chen, Rui Du, Jianming Li and Zhongmei He
Nutrients 2024, 16(20), 3473; https://doi.org/10.3390/nu16203473 - 14 Oct 2024
Viewed by 1030
Abstract
Background: Premature ovarian failure (POF) is a common disease among women, which can cause many complications and seriously threaten women’s physical and mental health. Currently, hormone replacement therapy is the primary treatment for premature ovarian failure. However, the side effects are serious and [...] Read more.
Background: Premature ovarian failure (POF) is a common disease among women, which can cause many complications and seriously threaten women’s physical and mental health. Currently, hormone replacement therapy is the primary treatment for premature ovarian failure. However, the side effects are serious and will increase the chance of breast cancer and endometrial cancer. Deer blood hydrolysate (DBH) is the product of enzymatic hydrolysis of deer blood, has antioxidant, anti-ageing, and anti-fatigue effects, and has the potential to improve premature ovarian failure. Methods: In our experiment, a mouse model of premature ovarian failure was established through intraperitoneal injection of 400 mg/kg/d of D-gal for 42 days. At the same time, different doses of DBH were gavaged to observe its ameliorative effect on premature ovarian failure. Results: The experimental findings indicated that DBH could restore the irregular oestrus cycle of POF mice, improve the abnormal amounts in serum hormones follicle-stimulating hormone (FSH), luteinising hormone (LH), progesterone (P) and estradiol (E2), increase the number of primordial follicles and decrease the number of atretic follicles. In addition, DBH also raised the level of superoxide dismutase (SOD) and reduced the level of malondialdehyde (MDA) and reduced the apoptosis of ovarian granulosa cells in mice. The WB assay results showed that gavage of DBH restored the decrease in the indication of nuclear factor erythroid 2-related factor 2 (Nrf2), Heme Oxygenase-1 (Ho-1), and B-cell lymphoma-2 (Bcl-2) proteins and reduced the elevated expression of Kelch-like ECH-associated protein 1 (Keap1), Bcl-2 associated X protein (Bax), and Cysteinyl aspartate specific proteinase-3 (Caspase-3) proteins that were induced by D-gal. Conclusions: To sum up, the present research indicated that DBH can ameliorate D-gal-induced oxidative stress and apoptosis by regulating the Nrf2/HO-1 signalling pathway and the Bcl-2/Bax/caspase-3 apoptosis pathway, which can be used for further development as a nutraceutical product to improve premature ovarian failure. Full article
(This article belongs to the Section Proteins and Amino Acids)
Show Figures

Figure 1

Figure 1
<p>Molecular weight distribution of DBH.</p>
Full article ">Figure 2
<p>The experimental procedure and the impact of DBH on weight and ovarian and uterine indices in mice. (<b>A</b>) Diagrammatic illustration of the experimental procedure. (<b>B</b>) Changes in body weight of mice during the experiment. (<b>C</b>,<b>D</b>) Effect of DBH on ovarian and uterine indices. (mean ± SEM, <span class="html-italic">n</span> = 8, * <span class="html-italic">p</span> &lt; 0.05 vs. control group, ** <span class="html-italic">p</span> &lt; 0.01 vs. control group, *** <span class="html-italic">p</span> &lt; 0.001 vs. control group, # <span class="html-italic">p</span> &lt; 0.05 vs. POF group, ns <span class="html-italic">p</span> &gt; 0.05 vs. POF group, ### <span class="html-italic">p</span> &lt; 0.001 vs. POF group).</p>
Full article ">Figure 3
<p>Effect of DBH on the oestrous cycle in POF mice. (<b>A</b>) Vaginal smears stained with methylene blue solution from different oestrus periods (P is preestrus, E is estrus, M is metestrus and D is diestrus). (<b>B</b>) Frequency of occurrence of cycle stages during the 27 days. (mean ± SEM, <span class="html-italic">n</span> = 8, * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. the control group; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 vs. the POF group). (<b>C</b>) Common patterns of regular and irregular estrus cycles.</p>
Full article ">Figure 4
<p>The impact of DBH on serum sexual hormones. (<b>A</b>–<b>D</b>) The FSH, LH, P and E2 levels of serum in different treatment groups. (mean ± SEM, <span class="html-italic">n</span> = 8, * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. the control group; # <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Effects of DBH on oxidative stress. (<b>A</b>) The SOD activity and (<b>B</b>) the MDA content were measured in the ovarian tissues. (mean ± SEM, <span class="html-italic">n</span> = 8, * <span class="html-italic">p</span> &lt; 0.05 vs. the control group; # <span class="html-italic">p</span> &lt; 0.05 vs. the POF group).</p>
Full article ">Figure 6
<p>The ovaries were stained using H&amp;E stain (100×) (<span class="html-italic">n</span> = 8).</p>
Full article ">Figure 7
<p>The impact of DBH on D-gal-induced ovarian cell apoptosis. (<b>A</b>): In situ TUNEL was used to analyse apoptosis. (200×) (<b>B</b>): Proportion of TUNEL-positive granulocytes to total cell count in different treatment groups. (mean ± SEM, <span class="html-italic">n</span> = 8, ** <span class="html-italic">p</span> &lt; 0.01 vs. the control group; ## <span class="html-italic">p</span> &lt; 0.01 vs. the POF group).</p>
Full article ">Figure 8
<p>DBH’s effect on the expression of Nrf2/HO-1 and Bcl-2/Bax/caspase-3 within ovarian tissues. (<b>A</b>,<b>E</b>) Images that are representative of western blotting results. (<b>B</b>–<b>D</b>) Quantification of the expression of Nrf2, HO-1, and Keap1. (<b>F</b>–<b>H</b>) Quantification of the protein expression of Bcl-2, Bax, and caspase-3. (mean ± SEM, <span class="html-italic">n</span> = 3, ** <span class="html-italic">p</span> &lt; 0.01 vs. the control group; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 vs. the POF group).</p>
Full article ">
12 pages, 5458 KiB  
Article
Anti-Photoaging Effects of Antioxidant Peptide from Seahorse (Hippocampus abdominalis) in In Vivo and In Vitro Models
by Fengqi Yang, Yang Yang, Dandan Xiao, Poongho Kim, Jihee Lee, You-Jin Jeon and Lei Wang
Mar. Drugs 2024, 22(10), 471; https://doi.org/10.3390/md22100471 - 14 Oct 2024
Viewed by 1185
Abstract
Overexposure to ultraviolet (UV) radiation can lead to photoaging, which contributes to skin damage. The objective of this study was to evaluate the effects of an antioxidant peptide (SHP2) purified from seahorse (Hippocampus abdominalis) alcalase hydrolysate on UVB-irradiated skin damage in [...] Read more.
Overexposure to ultraviolet (UV) radiation can lead to photoaging, which contributes to skin damage. The objective of this study was to evaluate the effects of an antioxidant peptide (SHP2) purified from seahorse (Hippocampus abdominalis) alcalase hydrolysate on UVB-irradiated skin damage in human keratinocyte (HaCaT) and human dermal fibroblast (HDF) cells and a zebrafish model. The data revealed that SHP2 significantly enhanced cell viability by attenuating apoptosis through the reduction of intracellular reactive oxygen species (ROS) levels in UVB-stimulated HaCaT cells. Moreover, SHP2 effectively inhibited ROS, improved collagen synthesis, and suppressed the secretion of matrix metalloproteinases (MMPs) in UVB-irradiated HDF cells. SHP2 restored the protein levels of HO-1, Nrf2, and SOD, while decreasing Keap1 expression in UVB-treated HDF, indicating stimulation of the Keap1/Nrf2/HO-1 signaling pathway. Furthermore, an in vivo study conducted in zebrafish confirmed that SHP2 inhibited photoaging by reducing cell death through the suppression of ROS generation and lipid peroxidation. Particularly, 200 µg/mL of SHP2 exerted a remarkable anti-photoaging effect on both in vitro and in vivo models. These results demonstrate that SHP2 possesses antioxidant properties and regulates skin photoaging activities, suggesting that SHP2 may have the potential for use in the development of cosmetic products. Full article
(This article belongs to the Special Issue Marine Anti-Inflammatory and Antioxidant Agents, 4th Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>SHP2 protects HaCaT cells against UVB-induced photodamage. (<b>A</b>) Cytotoxicity of SHP2 on HaCaT cells; (<b>B</b>) intracellular ROS scavenging effect of SHP2 in UVB-irradiated HaCaT cells; (<b>C</b>) protective effect of SHP2 against UVB-induced cell death in HaCaT cells. The experiments were conducted in triplicate and the data are expressed as the mean ± SE. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 as compared to the UVB-irradiated group and ## <span class="html-italic">p</span> &lt; 0.01 as compared to the control group.</p>
Full article ">Figure 2
<p>SHP2 protects HaCaT cells against UVB-induced apoptosis. (<b>A</b>) Morphology of the normal cells; (<b>B</b>) morphology of the cells irradiated by UVB; (<b>C</b>) morphology of the cells treated with 50 μg/mL SHP2 and irradiated by UVB; (<b>D</b>) morphology of the cells treated with 100 μg/mL SHP2 and irradiated by UVB; (<b>E</b>) morphology of the cells treated with 200 μg/mL SHP2 and irradiated by UVB; (<b>F</b>) quantification of apoptotic cells. The apoptotic body formation was evaluated by Hoechst 33342 staining assay. White arrows indicate apoptotic bodies. **** <span class="html-italic">p</span> &lt; 0.0001 as compared to the UVB-irradiated group and #### <span class="html-italic">p</span> &lt; 0.0001 as compared to the control group.</p>
Full article ">Figure 3
<p>SHP2 protects HDF cells against UVB-induced damage. (<b>A</b>) Cytotoxicity of SHP2 on HDF cells; (<b>B</b>) intracellular ROS scavenging effect of SHP2 in UVB-irradiated HDF cells; (<b>C</b>) protective effect of SHP2 against UVB-induced cell death in HDF cells. The experiments were conducted in triplicate and the data are expressed as the mean ± SE. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 as compared to the UVB-irradiated group and ## <span class="html-italic">p</span> &lt; 0.01 as compared to the control group.</p>
Full article ">Figure 4
<p>SHP2 improves collagen synthesis and inhibits MMPs’ expression in UVB-irradiated HDF cells. (<b>A</b>) Collagen contents in UVB-irradiated HDF cells; (<b>B</b>) MMP-1 expression levels in UVB-irradiated HDF cells; (<b>C</b>) MMP-2 expression levels in UVB-irradiated HDF cells; (<b>D</b>) MMP-8 expression levels in UVB-irradiated HDF cells; (<b>E</b>) MMP-9 expression levels in UVB-irradiated HDF cells; (<b>F</b>) MMP-13 expression levels in UVB-irradiated HDF cells. The experiments were conducted in triplicate and the data are expressed as the mean ± SE. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 as compared to the UVB-irradiated group and ## <span class="html-italic">p</span> &lt; 0.01 as compared to the control group.</p>
Full article ">Figure 5
<p>Protective effects of SHP2 on Keap1/Nrf2/HO-1 signaling pathway in UVB-irradiated HDF cells. (<b>A</b>) Western blot bands; (<b>B</b>) HO-1 protein expression; (<b>C</b>) Keap1 protein expression; (<b>D</b>) Nrf2 protein expression; (<b>E</b>) SOD protein expression. The experiments were conducted in triplicate and the data are expressed as the mean ± SE. **** <span class="html-italic">p</span> &lt; 0.0001 as compared to the UVB-irradiated group and ## <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 as compared to the control group.</p>
Full article ">Figure 6
<p>SHP2 protects zebrafish against UVB-induced oxidative stress in the zebrafish. (<b>A</b>) ROS generation in zebrafish; (<b>B</b>) cell death level in zebrafish; (<b>C</b>) lipid peroxidation level in zebrafish. ROS, cell death, and lipid peroxidation levels were measured by Image J software (v.1.8.0). The data are expressed as means ± SE. ** <span class="html-italic">p</span> &lt; 0.01 as compared to the UVB-treated group and ## <span class="html-italic">p</span> &lt; 0.01 as compared to the control group.</p>
Full article ">
18 pages, 5725 KiB  
Review
Protective and Detoxifying Effects of Resveratrol on Zearalenone-Mediated Toxicity: A Review
by Qiongxia Lv, Wenjing Xu, Fan Yang, Jiahui Li, Wenjuan Wei, Xiaoguang Chen, Yumei Liu and Ziqiang Zhang
Int. J. Mol. Sci. 2024, 25(20), 11003; https://doi.org/10.3390/ijms252011003 - 13 Oct 2024
Viewed by 1335
Abstract
Zearalenone (ZEA) is a mycotoxin produced by Fusarium spp. fungi and is widely found in moldy corn, wheat, barley, and other grains. ZEA is distributed to the whole body via blood circulation after metabolic transformation in animals. Through oxidative stress, immunosuppression, apoptosis, autophagy, [...] Read more.
Zearalenone (ZEA) is a mycotoxin produced by Fusarium spp. fungi and is widely found in moldy corn, wheat, barley, and other grains. ZEA is distributed to the whole body via blood circulation after metabolic transformation in animals. Through oxidative stress, immunosuppression, apoptosis, autophagy, and mitochondrial dysfunction, ZEA leads to hepatitis, neurodegenerative diseases, cancer, abortion, and stillbirth in female animals, and decreased sperm motility in male animals. In recent years, due to the influence of climate, storage facilities, and other factors, the problem of ZEA pollution in global food crops has become particularly prominent, resulting in serious problems for the animal husbandry and feed industries, and threatening human health. Resveratrol (RSV) is a natural product with therapeutic activities such as anti-inflammatory, antioxidant, and anticancer properties. RSV can alleviate ZEA-induced toxic effects by targeting signaling pathways such as NF-κB, Nrf2/Keap1, and PI3K/AKT/mTOR via attenuating oxidative damage, inflammatory response, and apoptosis, and regulating cellular autophagy. Therefore, this paper provides a review of the protective effect of RSV against ZEA-induced toxicity and its molecular mechanism, and discusses the safety and potential clinical applications of RSV in the search for natural mycotoxin detoxification agents. Full article
(This article belongs to the Section Molecular Toxicology)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of ZEA and RSV [<a href="#B35-ijms-25-11003" class="html-bibr">35</a>,<a href="#B36-ijms-25-11003" class="html-bibr">36</a>].</p>
Full article ">Figure 2
<p>Toxic effects of ZEA.</p>
Full article ">Figure 3
<p>Schematic representation of the biological effects of RSV.</p>
Full article ">Figure 4
<p>RSV ameliorates ZEA-induced toxicity by inhibiting ROS and OS production and activating Nrf2-mediated antioxidant defenses.</p>
Full article ">Figure 5
<p>RSV alleviates ZEA-induced inflammatory response by regulating NLRP3, TLR4, and NF-κB signaling pathways.</p>
Full article ">Figure 6
<p>RSV attenuates ZEA-induced mitochondrial dysfunction and apoptosis by activating the SIRT1/FOXO3a signaling pathway and regulating mitochondrial function.</p>
Full article ">Figure 7
<p>RSV regulates ZEA-induced mitochondrial autophagy and cellular autophagy through PI3K/Akt/mTOR and AMPK signaling pathways.</p>
Full article ">
28 pages, 3321 KiB  
Article
Interplay of Cellular Nrf2/NF-κB Signalling after Plasma Stimulation of Malignant vs. Non-Malignant Dermal Cells
by Kristina Manzhula, Alexander Rebl, Kai Budde-Sagert and Henrike Rebl
Int. J. Mol. Sci. 2024, 25(20), 10967; https://doi.org/10.3390/ijms252010967 - 11 Oct 2024
Viewed by 1429
Abstract
Skin cancer is one of the most common malignancies worldwide. Cold atmospheric pressure Plasma (CAP) is increasingly successful in skin cancer therapy, but further research is needed to understand its selective effects on cancer cells at the molecular level. In this study, A431 [...] Read more.
Skin cancer is one of the most common malignancies worldwide. Cold atmospheric pressure Plasma (CAP) is increasingly successful in skin cancer therapy, but further research is needed to understand its selective effects on cancer cells at the molecular level. In this study, A431 (squamous cell carcinoma) and HaCaT (non-malignant) cells cultured under identical conditions revealed similar ROS levels but significantly higher antioxidant levels in unstimulated A431 cells, indicating a higher metabolic turnover typical of tumour cells. HaCaT cells, in contrast, showed increased antioxidant levels upon CAP stimulation, reflecting a robust redox adaptation. Specifically, proteins involved in antioxidant pathways, including NF-κB, IκBα, Nrf2, Keap1, IKK, and pIKK, were quantified, and their translocation level upon stimulation was evaluated. CAP treatment significantly elevated Nrf2 nuclear translocation in non-malignant HaCaT cells, indicating a strong protection against oxidative stress, while selectively inducing NF-κB activation in A431 cells, potentially leading to apoptosis. The expression of pro-inflammatory genes like IL-1B, IL-6, and CXCL8 was downregulated in A431 cells upon CAP treatment. Notably, CAP enhanced the expression of antioxidant response genes HMOX1 and GPX1 in non-malignant cells. The differential response between HaCaT and A431 cells underscores the varied antioxidative capacities, contributing to their distinct molecular responses to CAP-induced oxidative stress. Full article
(This article belongs to the Special Issue NRF2/KEAP1 Signalling in Cancer)
Show Figures

Figure 1

Figure 1
<p>The Nrf2/Keap1/ARE and NF-κB/IκB signalling pathways. Proteins/genes investigated in this study are marked with a star. (<b>A</b>) Under basal conditions, Keap1 molecules bind Nrf2, facilitating its labelling with ubiquitin and targeting it for proteasomal degradation, while Keap1 is regenerated. During oxidative stress, the Keap1 cysteine thiols react to a broad range of oxidative and electrophile molecules leading to conformational changes interrupting the interactions between Nrf2 and Keap1. Thus, Nrf2 is released to translocate into the nucleus, where it binds to the ARE present in the promotors of numerous cytoprotective genes. (<b>B</b>) Regulation of NF-κB signalling occurs via activation of the trimeric kinase complex (IKKα, IKKβ, IKKγ) via the upstream TAK1. IKK-mediated IκBα phosphorylation is followed by IκBα degradation and rapid nuclear translocation of NF-κB, where it activates genes for immune regulation and apoptosis pathways. [ARE: antioxidant response element, Cul3: Cullin 3, <span class="html-italic">CXCL8</span>: chemokine (C-X-C motif) ligand 8, <span class="html-italic">GPX1</span>: glutathione peroxidase 1, <span class="html-italic">HMOX1</span>: heme oxygenase 1, IκBα: inhibitor of κBα, IKKβ: IκB kinase β, IL-1R: interleukin 1 receptor, NF-κB: nuclear factor-kappa B, Nrf2: nuclear factor-erythroid 2-related factor 2, P: phosphate, RBX1: RING-box protein 1, ROS: reactive oxygen species, sMaf: small Maf, TLR: toll-like receptor, TNFR: tumour necrosis factor receptor, TRAF6: TNF receptor-associated factor 6, Ub: ubiquitin] (created in BioRender. Rebl, H. (2024) BioRender.com/s71v052 accessed on 1 October 2024).</p>
Full article ">Figure 2
<p>Characterisation of unstimulated dermal cells. (<b>A</b>) Intracellular levels of reactive oxygen species (ROS) are comparable in both cell lines (n = 2), where 0.5 µg/mL H<sub>2</sub>O<sub>2</sub> served as a positive control. (<b>B</b>) The basal cellular antioxidant level (displayed as Trolox equivalents) is significantly lower in HaCaT (orange) cells relative to A431 (blue) cells (n = 4). (<b>C</b>) A431 cells show a slightly higher nuclear/cytosolic ratio of Nrf2 in a basal state compared to HaCaT cells (dotted line indicates the basal level of HaCaT; n = 3), where 15 µg/mL H<sub>2</sub>O<sub>2</sub> served as positive control. (<b>D</b>) Protein abundances of NF-κB, IκBα, Nrf2, Keap1, IKK, and pIKK (n = 3). HaCaT cells show significantly higher protein levels of Keap1 and IKK compared to A431 cells. (<b>E</b>) Cell viability of A431 cells significantly and dose-dependently decreases after a CAP treatment durations of ≥45 s, whereas HaCaT cells display a slower decline with significance appearing after ≥90 s of treatment (n = 3). * Compared to the respective control (0 s) (* <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; **** <span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 3
<p>Oxygen consumption rates of A431 (<b>left</b>) and HaCaT (<b>right</b>) cells after treatment with cold atmospheric pressure plasma (CAP). Values starting at 3 h after treatment were analysed, and unstimulated cells were normalised to a value of 1. CAP treatment of 60 s led to a significant reduction (black arrow) from tA= 5.5 h only for A431, whereas 90 s exposure showed a significant decrease (grey arrows) from tA= 5.5 h (A431) and tH = 4.5 h (HaCaT) (lines show mean values from n = 4). Arrows mark the timepoints where alterations in the oxygen consumption rate start to be significant compared to the respective untreated control (0 s). Statistical analyses were performed using multiple <span class="html-italic">t</span>-tests, and statistical significance was determined with the Holm–Sidak method (alpha = 0.05).</p>
Full article ">Figure 4
<p>(<b>A</b>) Measurement of ROS in media treated with cold atmospheric pressure plasma (CAP). Hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>) concentrations significantly increase in plasma-activated media in a dose-dependent manner. The H<sub>2</sub>O<sub>2</sub> concentration used as a positive control in all experiments (441 µM, red dotted line) is nearly equivalent to the hydrogen peroxide content in 60 s CAP treatment (492 µM) (n = 3). (<b>B</b>) Antioxidative capacity displayed as Trolox equivalents after CAP exposure. The antioxidative capacity of untreated tumour cells is significantly higher than in HaCaT cells; however, the antioxidant level drops in these cells post CAP treatment. In contrast, plasma treatment leads to elevated antioxidant levels in HaCaT cells. p # compared to A431 negative control, * compared to the respective negative control (n = 4). (<b>C</b>) Determination of RNS in media after CAP treatment. Nitrite (left, blue, NO<sub>2</sub><sup>−</sup>) and nitrate (right, beige, NO<sub>3</sub><sup>−</sup>) concentrations significantly increase with a longer treatment duration (**** <span class="html-italic">p</span> ≤ 0.0001, n = 3).</p>
Full article ">Figure 5
<p>Translocation dynamics of Nrf2 (<b>A</b>), Keap1 (<b>B</b>), NF-κB (<b>C</b>), and IκBα (<b>D</b>) in A431 (blue) and HaCaT (orange) cells after cold atmospheric pressure plasma (CAP) exposure. (<b>A</b>) In HaCaT cells, Nrf2 translocation is significantly increased after CAP treatment—independent of the dose, whereas A431 cells show a marginal upward trend (n = 3). (<b>B</b>) No significant changes in the Keap1 localisation are apparent in either cell line (n = 3). (<b>C</b>) In A431 cells, NF-κB translocation is significantly increased after 180 s of CAP treatment, whereas HaCaT cells show no noteworthy alterations (n = 4). (<b>D</b>) Generally, IκBα translocation increases dose-dependently in both cell lines. However, A431 cells display a significant decrease after 90 and 120 s of CAP treatment, gradually giving way to an increase with an escalating exposure time, ultimately converging to the control (white filling) ratio of 1 after 180 s. Dark grey filling: treatment with argon gas only; light grey filling: positive ctrl; (* <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤ 0.001; **** <span class="html-italic">p</span> ≤ 0.0001, n = 3).</p>
Full article ">Figure 6
<p>Mean intensities of proteins associated with Nrf2 and NF-κB signalling in A431 and HaCaT cells. (<b>A</b>, right) In HaCaT cells, the Keap1 abundance was significantly increased after 180 s exposure to cold atmospheric pressure plasma (CAP). (<b>B</b>, left) Also, A431 cells exhibited significantly reduced levels of NF-κB after CAP treatment. (<b>C</b>, left) The abundance of IKK was significantly reduced in HaCaT cells relative to A431 cells. For positive controls (grey filling), 15 µg/mL H<sub>2</sub>O<sub>2</sub> and 20 μg/mL Poly(I:C) were used, respectively (* <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01, n = 3).</p>
Full article ">Figure 7
<p>Expression analyses of Nrf2- and NF-κB -associated genes in A431 and HaCaT cells treated with cold atmospheric pressure plasma (CAP). (<b>A</b>) Log2-fold change levels of the indicated genes relative to the respective untreated control. Positive controls are indicated in grey. CAP leads to an activation of <span class="html-italic">IL-6</span>, <span class="html-italic">TNF</span>, and <span class="html-italic">HMOX</span> expression in HaCaT, while A431 expression levels are moderately affected (* <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤ 0.001; **** <span class="html-italic">p</span> ≤ 0.0001). (<b>B</b>) Transcript numbers/µg RNA show the low copy genes <span class="html-italic">TNF</span>, <span class="html-italic">HMOX1</span>, and <span class="html-italic">IL-6</span> in both unstimulated dermal cell lines, while the other genes examined are referred to as high copy genes. (<b>C</b>) The factor maps indicate the contribution of individual genes to the overall separation of log10-transformed expression data in A431 and HaCaT cells. The lengths of the blue and orange vectors radiating from the centroid (0.0) indicate the contribution of each gene to the overall variance of data in A431 and HaCaT cells, respectively. The majority of variance is described by the dimensions Dim1 and Dim2 given in parentheses on the axis labelling. The coloured clusters (ctrl, grey; plasma, yellow; argon, brown; H<sub>2</sub>O<sub>2</sub>, green; poly[I:C], purple) indicate samples treated in the same way.</p>
Full article ">
20 pages, 4516 KiB  
Article
By-Products Valorization: Peptide Fractions from Milk Permeate Exert Antioxidant Activity in Cellular and In Vivo Models
by Valeria Scalcon, Federico Fiorese, Marica Albanesi, Alessandra Folda, Gianfranco Betti, Marco Bellamio, Emiliano Feller, Claudia Lodovichi, Giorgio Arrigoni, Oriano Marin and Maria Pia Rigobello
Antioxidants 2024, 13(10), 1221; https://doi.org/10.3390/antiox13101221 - 10 Oct 2024
Viewed by 657
Abstract
The discarding of agri-food by-products is a stringent problem due to their high environmental impact. Recovery strategies can lead to a reduction of waste and result in new applications. Agri-food waste represents a source of bioactive molecules, which could promote health benefits. The [...] Read more.
The discarding of agri-food by-products is a stringent problem due to their high environmental impact. Recovery strategies can lead to a reduction of waste and result in new applications. Agri-food waste represents a source of bioactive molecules, which could promote health benefits. The primary goal of this research has been the assessment of the antioxidant activity of milk permeate, a dairy farm by-product, and the isolation and identification of peptide fractions endowed with antioxidant activity. The chromatographic extraction of the peptide fractions was carried out, and the peptides were identified by mass spectrometry. The fractions showed radical scavenging activity in vitro. Moreover, the results in the Caco-2 cell model demonstrated that the peptide fractions were able to protect from oxidative stress by stimulating the Keap1/Nrf2 antioxidant signaling pathway, increasing the transcription of antioxidant enzymes. In addition, the bioactive peptides can affect cellular metabolism, increasing mitochondrial respiration. The action of the peptide fractions was also assessed in vivo on a zebrafish model and resulted in the protection of the whole organism from the adverse effects of acute cold stress, highlighting their strong capability to protect from an oxidative insult. Altogether, the results unveil novel recovery strategies for food by-products as sources of antioxidant bioactive peptides that might be utilized for the development of functional foods. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Antioxidant activity of PFs obtained from MP. (<b>A</b>) ABTS scavenging activity is reported as Trolox C equivalent antioxidant capacity (TEAC); (<b>B</b>) DPPH scavenging assay; results are reported as percentage with respect to the control (Cnt); (<b>C</b>) estimation of total phenolic content expressed as gallic acid equivalent (GAE). Results are the mean ± SD of three replicates.</p>
Full article ">Figure 2
<p>Protective effect of PFs obtained from milk permeate on oxidative stress induction in Caco-2 cells. (<b>A</b>) Effects of the PFs on cell viability. Caco-2 cells were treated with the indicated fractions for 24 h and oxidative stress was induced by 180 µM TbOOH (for 18 h). Results are shown as percentage of cell viability with respect to the Cnt; (<b>B</b>) Estimation of ROS production in Caco-2 cells treated with the indicated PFs for 24 h in the absence (grey) or presence (red) of 300 µM TbOOH and expressed as percentage with respect to the Cnt. Results are the mean ± SD of three replicates ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Nrf2 and NF-κB levels in the nuclear fraction of Caco-2 cells treated with 5–30% ACN PF from MP. Nuclear fractions of cells treated with 0.05 mg/mL of 5–30% ACN PF and/or with 2 mM NAC for 24 h were extracted, and Western blot analysis was carried out to estimate Nrf2, NF-κB, and PCNA levels. (<b>A</b>) Representative WB of protein expression in Caco-2 cells in the different conditions. (<b>Aˈ</b>) Quantitative analysis of the WB after normalization using PCNA as a nuclear loading control. Results are the mean ± SD of three replicates * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Expression of antioxidant enzymes in Caco-2 cells treated with 5–30% ACN PF from MP. Cells were treated with 0.05 mg/mL of 5–30% ACN PF for 24 h. Afterwards, cells were lysed, and WB analysis was carried out. (<b>A</b>) Representative images of protein expression of the various enzymes in Caco-2 cells via WB; (<b>B</b>) Ponceau S staining reporting the protein loading; (<b>C</b>) Quantitative analysis of the Western blot after normalization using GAPDH and β-actin as loading controls. Results are the mean ± SD of three replicates * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Oxygen consumption rates and glycolytic activity of Caco-2 cells treated with the PF. Caco-2 cells were treated with 0.05 mg/mL of the 5–30% ACN PF for 24 h. (<b>A</b>) The oxygen consumption rates (OCRs) were assessed using the Seahorse Xfe24 analyzer as described in the <a href="#sec2-antioxidants-13-01221" class="html-sec">Section 2</a>. Basal respiration and respiratory capacity in the presence of sequential addition of 1 µM oligomycin, 0.5 µM FCCP and the combination of 1 μM antimycin A + 1 µM rotenone, was measured. (<b>Aˈ</b>) Basal, ATP-linked and maximal respirations are shown as the mean ± SD of 5 experiments, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>B</b>) Cellular glycolytic activity was determined in the presence of sequential addition of 10 mM glucose, 1 μM antimycin A + 1 µM rotenone, and 2-deoxy-D-glucose. (<b>Bˈ</b>) Glycolysis, maximal glycolytic capacity, and glycolytic reserve are reported as the mean ± SD of 3 experiments.</p>
Full article ">Figure 6
<p>Effects of the 5–30% ACN PF in vivo on zebrafish larvae under cold stress conditions. Zebrafish larvae were divided into four groups: control (placed in 28 °C water) (blue box), acute cold stressed (placed in 10 °C water for 5 min) (pink box), acute cold stressed + 30 min recovery (green box), and acute cold stressed + 30 min recovery in the presence of the PF (yellow box). The motility of the four groups of larvae was assessed using the DanioVision system. (<b>A</b>) Total swimming distance; (<b>B</b>) Average swimming speed. (<span class="html-italic">n</span> = 75, distributed as 25 controls, 13 cold stress, 19 cold stress + recovery, and 18 cold stress + recovery + 5–30% ACN PF), * <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 ">
20 pages, 4704 KiB  
Article
Extraction, Characterization, and Antioxidant Activity of Eucommia ulmoides Polysaccharides
by Yuqing Peng, Yulin Yang, Yitong Tian, Meng Zhang, Kang Cheng, Xuelei Zhang, Mengqing Zhou, Ming Hui and Yong Zhang
Molecules 2024, 29(20), 4793; https://doi.org/10.3390/molecules29204793 - 10 Oct 2024
Cited by 1 | Viewed by 718
Abstract
Herein, the ultrasound-assisted extraction conditions affecting the yield of EUPS (Eucommia ulmoides polysaccharide) were analyzed using a Box-Behnken response surface design. The alleviation effect of EUPS on diquat-induced oxidative stress in mice was also studied. A maximum EUPS yield of 2.60% was [...] Read more.
Herein, the ultrasound-assisted extraction conditions affecting the yield of EUPS (Eucommia ulmoides polysaccharide) were analyzed using a Box-Behnken response surface design. The alleviation effect of EUPS on diquat-induced oxidative stress in mice was also studied. A maximum EUPS yield of 2.60% was obtained under the following optimized conditions: an extraction temperature of 63 °C, extraction time of 1 h, and ratio of liquid to raw materials of 22:1. EUPS exhibited strong 1,1-diphenyl-2-picryl-hydrazyl (DPPH) radical-scavenging ability (87.05%), 2′-Azinobis-(3-ethylbenzthiazoline-6-sulphonate) (ABTS) radical-scavenging ability (101.17%), and hydroxyl radical-scavenging ability (62.92%). The administration of EUPS increased the activities of superoxide dismutase, catalase, and glutathione peroxidase and decreased malondialdehyde levels in the livers of mice exposed to diquat. EUPS may inhibit the downregulation of NAD(P)H:quinoneoxidoreductase 1 and heme oxygenase 1 mRNA expression in the livers of diquat-administered mice through the Nrf2-Keap1 signaling pathway. Moreover, the abundance of Firmicutes and Ligilactobacillus was enhanced, whereas that of Helicobacter decreased in the gut of the remaining groups of mice compared with that of the diquat-treated mice. Therefore, EUPS exhibited an antioxidant effect and improved oxidative stress and intestinal flora abundance in mice. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of ratio of water to material (<b>a</b>), extraction temperature (<b>b</b>), and extraction time (<b>c</b>) on extraction yield of EUPS.</p>
Full article ">Figure 2
<p>Three-dimensional surface plots showing the interaction effects of (<b>a</b>) extraction temperature and ratio of water to materials, (<b>b</b>) extraction time and ratio of water to materials, and (<b>c</b>) extraction time and extraction temperature on the extracted yield of EUPS.</p>
Full article ">Figure 3
<p>FI-IR spectra of EUPS.</p>
Full article ">Figure 4
<p>HPLC spectra of EUPS: 1. mannose, 2. ribose, 3. rhamnose, 4. glucuronic acid, 5. galacturonic acid, 6. glucose, 7. galactose, 8. xylose, 9. arabinose, and 10. fucose.</p>
Full article ">Figure 5
<p>Particle size distribution of EUPS.</p>
Full article ">Figure 6
<p>Scavenging ability of EUPS and Vc for DPPH radicals.</p>
Full article ">Figure 7
<p>Scavenging ability of EUPS and Vc for ABTS radicals.</p>
Full article ">Figure 8
<p>Scavenging ability of EUPS and Vc to hydroxyl radicals.</p>
Full article ">Figure 9
<p>Reduction ability of EUPS and Vc.</p>
Full article ">Figure 10
<p>Effect of EUPS on antioxidant activities in the serum of mice. (<b>A</b>) Serum CAT activity; (<b>B</b>) serum MDA activity; (<b>C</b>) serum SOD activity; (<b>D</b>) serum GSH-Px activity; (<b>E</b>) serum T-AOC activity. The data are expressed as the means ± SEM (<span class="html-italic">n</span> = 10); the different letters indicate the statistically significant differences in all of the treatment groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 11
<p>Expression levels of Nrf2 genes (<b>A</b>), Keap1genes (<b>B</b>), NQO1 genes (<b>C</b>), and HO-1 genes (<b>D</b>) in mice liver. The data are expressed as the means ± SEM (<span class="html-italic">n</span> = 10); the different letters indicate the statistically significant differences in all of the treatment groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 12
<p>Effects of EUPS on species composition. (<b>a</b>) Histogram of relative abundance at the phylum level. (<b>b</b>) Histogram of relative abundance at the genus level.</p>
Full article ">Figure 13
<p>Alpha diversity of gut microbiota. (<b>a</b>) Chao1 index. (<b>b</b>) Shannon index. Each dot in the graph represents a sample, and dots of different colors indicate different groups.</p>
Full article ">Figure 14
<p>Beta diversity analysis. (<b>a</b>) PCoA analysis. (<b>b</b>) NMDS analysis. (<b>c</b>) UPGMA.</p>
Full article ">
Back to TopTop