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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (724)

Search Parameters:
Keywords = Keap1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 10000 KiB  
Article
Impacts of Copper Deficiency on Oxidative Stress and Immune Function in Mouse Spleen
by Xiaocong Li, Xin Zeng, Wanqin Yang, Peng Ren, Hengxiao Zhai and Heng Yin
Nutrients 2025, 17(1), 117; https://doi.org/10.3390/nu17010117 - 30 Dec 2024
Viewed by 310
Abstract
Introduction: Copper is an essential trace element crucial for enzyme synthesis and metabolism. Adequate copper levels are beneficial for maintaining the normal immune function of the spleen. Copper deficiency disrupts the metabolic processes within the spleen and impairs its immune function. This research [...] Read more.
Introduction: Copper is an essential trace element crucial for enzyme synthesis and metabolism. Adequate copper levels are beneficial for maintaining the normal immune function of the spleen. Copper deficiency disrupts the metabolic processes within the spleen and impairs its immune function. This research examines the impact of copper deficiency on the spleen and the potential recovery following copper supplementation. Methods: Weaned mice underwent a 4-week copper-deficient diet, succeeded by 1-week of copper repletion via intraperitoneal copper sulfate injection. Histological examination was used to assess pathological changes in the spleen. Biochemical assays were performed to measure oxidative stress levels in the spleen. ELISA, qPCR, and Western blot were employed to examine alterations in inflammatory markers, immune indicators, and oxidative regulatory factors across various levels. Results: Copper deficiency caused histological damage to the spleen, altered the expression of oxidative stress regulatory pathways (Nrf2, Keap1, and HO-1), and affected the expression of key inflammatory enzymes (iNOS, COX2) and transcription factor NF-κB, leading to oxidative damage. This was reflected by decreased levels of SOD, GSH, and T-AOC, along with increased levels of CAT and MDA. The levels of inflammatory cytokines IL-1β, TNF-α, IL-6, and IL-8 were notably increased. Copper supplementation significantly improved these changes. Conclusions: Copper deficiency leads to spleen tissue damage in mice, affecting the Nrf2 regulatory pathway and inducing oxidative damage. Subsequent copper supplementation with copper sulfate effectively ameliorates the damage caused by copper deficiency. Full article
(This article belongs to the Section Nutritional Immunology)
Show Figures

Figure 1

Figure 1
<p>Effects of Cu deficiency and supplementation on spleen index and histological morphology in mice. (<b>A</b>) Spleen microscopic structure of mice, H&amp;E, 100×. (<b>B</b>) Appearance and size of spleen. (<b>C</b>) Spleen index of the groups of mice. * <span class="html-italic">p</span> &lt; 0.05, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 2
<p>Effects of Cu deficiency and supplementation on oxidative damage of spleen in mice. (<b>A</b>–<b>E</b>) represent the CAT activity, SOD activity, GSH concentration, total antioxidant capacity, and MDA content in the spleens of mice from each group, respectively. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 3
<p>Effects of Cu deficiency and supplementation on cytokine of serum in mice. (<b>A</b>–<b>D</b>) represent concentrations of IL-1β, IL-6, IL-8, and TNF-α in mouse serum, respectively. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 4
<p>Effects of Cu deficiency and supplementation on mRNA expression of spleen inflammation-related genes. (<b>A</b>–<b>G</b>) represent the mRNA relative expression levels of IL-1β, IL-6, IL-8, TNF-α, NF-κB, iNOS, and COX2 in the spleens of mice, respectively. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 5
<p>Effects of Cu deficiency and supplementation on mRNA expression of spleen nrf2 signaling pathway. (<b>A</b>–<b>C</b>) represent the mRNA relative expression levels of Nrf2, Keap1, and HO-1 in the spleens of mice, respectively. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 6
<p>Impact of Cu deficiency and supplementation on spleen inflammation-related protein expression. (<b>A</b>) Protein levels of p-NF-κB, NF-κB, TNF-α, IL-1β, and IL-6 in the spleen, using β-actin as the loading control. (<b>B</b>) The ratio of p-NF-κB to NF-κB. (<b>C</b>–<b>E</b>) The relative expression levels of TNF-α, IL-1β, and IL-6 were normalized relative to β-actin. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>Effects of Cu deficiency and supplementation on protein expression of nrf2 signaling pathway of spleen. (<b>A</b>) Protein levels of Nrf2, Keap1, and HO-1 in spleen, using β-actin as loading control. (<b>B</b>–<b>D</b>) The relative expression levels of Nrf2, Keap1, and HO-1 were normalized to β-actin expression. *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">
15 pages, 7166 KiB  
Article
Ameliorative Effects of Raisin Polyphenol Extract on Oxidative Stress and Aging In Vitro and In Vivo via Regulation of Sirt1–Nrf2 Signaling Pathway
by Wenjing Gao, Caiyun Zhao, Xin Shang, Bin Li, Jintian Guo, Jingteng Wang, Bin Wu and Yinghua Fu
Foods 2025, 14(1), 71; https://doi.org/10.3390/foods14010071 - 30 Dec 2024
Viewed by 376
Abstract
Raisins are an important source of polyphenolic compounds in plant foods, and polyphenols are associated with antioxidant and anti-aging activity. In this work, 628 polyphenols in raisin extracts were characterized using UPLC-MS/MS, mainly including tricetin 3′-glucuronide, diisobutyl phthalate, butyl isobutyl phthalate, isoquercitrin and [...] Read more.
Raisins are an important source of polyphenolic compounds in plant foods, and polyphenols are associated with antioxidant and anti-aging activity. In this work, 628 polyphenols in raisin extracts were characterized using UPLC-MS/MS, mainly including tricetin 3′-glucuronide, diisobutyl phthalate, butyl isobutyl phthalate, isoquercitrin and 6-hydroxykaempferol-7-O-glucoside. The oxidative stress in H2O2-induced HepG2 cells and D-gal-induced aging mice was alleviated by raisin polyphenols (RPs) via increases in the cellular levels of superoxide dismutase (SOD), catalase (CAT) and glutathione (GSH), along with decreases in malonaldehyde (MDA), reactive oxygen species (ROS) and advanced glycosylation end-products (AGEs) levels. In addition, it was observed that RPs enhanced Sirt1 and Sirt3 expression, initiating the Keap1-Nrf2 signaling pathway, by upregulating the levels of nuclear Nrf2, facilitating the expressions of the antioxidant proteins NQO1 and HO-1, and downregulating Keap1 and cytoplasmic Nrf2 protein levels in H2O2-induced HepG2 cells and D-gal-induced aging mice. In summary, RP exerted antioxidant and anti-aging effects via regulating the Sirt1–Nrf2 signaling pathway in vitro and in vivo. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Figure 1

Figure 1
<p>Animal experimental protocol.</p>
Full article ">Figure 2
<p>Total ion flow diagram of RP: (<b>A</b>) positive ion mode; (<b>B</b>) negative ion mode.</p>
Full article ">Figure 2 Cont.
<p>Total ion flow diagram of RP: (<b>A</b>) positive ion mode; (<b>B</b>) negative ion mode.</p>
Full article ">Figure 3
<p>Effect of RPs on H<sub>2</sub>O<sub>2</sub>-induced HepG2 cells: (<b>A</b>) Cell viability; (<b>B</b>) HepG2 cells were stained with DCFH-DA. Scale bar: 50 μm; The fluorescence values were determined using a microplate reader for (<b>C</b>) ROS; (<b>D</b>) MDA; (<b>E</b>): SOD; (<b>F</b>) CAT; and (<b>G</b>) GSH. All the data are represented as the means ± SDs. <span class="html-italic">n</span> = 3 for each group. ### <span class="html-italic">p</span> &lt; 0.001 vs. control group. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 vs. H<sub>2</sub>O<sub>2</sub> group.</p>
Full article ">Figure 4
<p>Effect of RPs on the Sirt1–Nrf2 signaling pathway in H<sub>2</sub>O<sub>2</sub> -induced HepG2 cells: (<b>A</b>) Sirt1; (<b>B</b>) Sirt3; (<b>C</b>) Keap1; (<b>D</b>) cytoplasmic Nrf2; (<b>E</b>) nuclear Nrf2; (<b>F</b>) NQO1; (<b>G</b>) HO-1. All the data are represented as the means ± SDs. <span class="html-italic">n</span> = 3 for each group. # <span class="html-italic">p</span> &lt; 0.05; ### <span class="html-italic">p</span> &lt; 0.001 vs. control group. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 vs. H<sub>2</sub>O<sub>2</sub> group.</p>
Full article ">Figure 5
<p>Effects of RPs on body weight (<b>A</b>) and organ index (<b>B</b>) in aging mice (<span class="html-italic">n</span> = 8 for each group). ### <span class="html-italic">p</span> &lt; 0.001 vs. NC 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 vs. MC group. NC: normal control group; MC: D-gal induced aging group; VE: vitamin E-treated group; RP-L: RP group (50 mg/kg); RP-M: RP group (100 mg/kg); RP-H: RP group (200 mg/kg).</p>
Full article ">Figure 6
<p>Effect of RPs on the histopathology in D-gal-induced aging mice: (<b>A</b>) hippocampus; (<b>B</b>) cerebral cortex; (<b>C</b>) liver. Scale bar: 50 µm. NC: normal control group; MC: D-gal induced aging group; VE: vitamin E-treated group; RP-L: RP group (50 mg/kg); RP-M: RP group (100 mg/kg); RP-H: RP group (200 mg/kg).</p>
Full article ">Figure 7
<p>Effect of RPs on oxidative stress in aging mice: (<b>A</b>) SOD; (<b>B</b>) CAT; (<b>C</b>) GSH; (<b>D</b>) MDA; (<b>E</b>) ROS; (<b>F</b>) AGEs. All the data are represented as means ± SDs. <span class="html-italic">n</span> = 6 for each group. ### <span class="html-italic">p</span> &lt; 0.001 vs. NC 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 vs. MC group. NC: normal control group; MC: D-gal induced aging group; VE: vitamin E-treated group; RP-L: RP group (50 mg/kg); RP-M: RP group (100 mg/kg); RP-H: RP group (200 mg/kg).</p>
Full article ">Figure 8
<p>Effect of RPs on the Sirt1–Nrf2 signaling pathway in aging mice: (<b>A</b>) Sirt1; (<b>B</b>) Sirt3; (<b>C</b>) Keap1; (<b>D</b>) cytoplasmic Nrf2; (<b>E</b>) nuclear Nrf2; (<b>F</b>) NQO1; (<b>G</b>) HO-1. All the data are represented as the means ± SDs. <span class="html-italic">n</span> = 6 for each group. ### <span class="html-italic">p</span> &lt; 0.001 vs. NC 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 vs. MC group. NC: normal control group; MC: D-gal induced aging group; VE: vitamin E-treated group; RP-L: RP group (50 mg/kg); RP-M: RP group (100 mg/kg); RP-H: RP group (200 mg/kg).</p>
Full article ">
14 pages, 2763 KiB  
Article
Identification of Protein Hydrolysates from Sesame Meal and In Vivo Study of Their Gastric Mucosal Protective Effects
by Yutong Yuan, Xinyi Wang, Nan Ling, Jingxuan Zhou, Lei Zhao, Baoping Ji, Feng Zhou and Liang Zhao
Foods 2024, 13(24), 4178; https://doi.org/10.3390/foods13244178 - 23 Dec 2024
Viewed by 397
Abstract
This study aimed to investigate the protective effects and defense mechanisms of a sesame meal protein hydrolysate against ethanol-induced acute gastric mucosal injury in mice. The target peptides in the hydrolysate were identified by LC-MS/MS, the activity was predicted by PeptideRanker, and the [...] Read more.
This study aimed to investigate the protective effects and defense mechanisms of a sesame meal protein hydrolysate against ethanol-induced acute gastric mucosal injury in mice. The target peptides in the hydrolysate were identified by LC-MS/MS, the activity was predicted by PeptideRanker, and the KM mice were orally administered distilled water, a sesame peptide, and omeprazole for 24 consecutive days. Acute gastric mucosal injury was then induced in mice with 70% ethanol, except for the CK group. The sesame peptide significantly inhibited the over-accumulation of ALT, AST, MDA, TNF-α, IL-1β, and MPO, while promoting the reduction in GSH, T-AOC, GSSG, and EGF expression. In addition, a Western blotting analysis showed that sesame peptide significantly up-regulated the expression of HO-1 and NQO1 proteins in the Nrf2/Keap1 signaling pathway, and down-regulated Keap1 protein. The defense effect of a sesame peptide on gastric mucosa may be achieved by alleviating the overproduction of lipid peroxides and improving the antioxidant activity. Full article
(This article belongs to the Special Issue Bioactive Peptides: Preparation, Characterization and Properties)
Show Figures

Figure 1

Figure 1
<p>Experimental schedule of animal treatment (<b>A</b>): gastric mucosal damage was induced by ethanol in mice after gavage of different doses of SPs (200 and 400 mg/kg) for 21 days. Effect of SPs on body weight in mice (<b>B</b>), macroscopic map of mouse stomach tissue (<b>C</b>), injury index and injury inhibition rate (<b>D</b>). CK: blank control group; MG: model group; PG: omeprazole positive control group (20 mg/kg bw); LSG: low-dose sesame peptide group (200 mg/kg bw); and HSG: high-dose sesame peptide group (400 mg/kg bw). Obvious areas of damage are marked by red circles. Different letters represent significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Histologic evaluation of gastric tissues for H&amp;E staining (<b>A</b>) and histologic scoring (<b>B</b>). CK: blank control group; MG: model group; PG: omeprazole positive control group (20 mg/kg bw); LSG: low-dose sesame peptide group (200 mg/kg bw); and HSG: high-dose sesame peptide group (400 mg/kg bw). Localized epithelial cell necrosis and detachment of gastric mucosa are marked by black arrows, and localized mucosal hemorrhage is marked by red arrows. Different letters represent groups with significant differences (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Effects of SPs on serum ALT (<b>A</b>) and AST (<b>B</b>), oxidative stress markers in gastric tissues (<b>C</b>–<b>F</b>), and serum levels of inflammatory factors (<b>G</b>–<b>J</b>) in mice. CK group: blank control group; MG group: model group; PG group: positive omeprazole control group (20 mg/kg bw); LSG group: low-dose sesame peptide group (200 mg/kg bw); HSG group: high-dose sesame peptide group (400 mg/kg bw). Different letters represent significant differences between groups at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Effects of SPs on Nrf2/Keap1 antioxidant signaling pathways. Protein blot analysis of Nrf2, NQO1, Keap1, and HO-1. (<b>A</b>,<b>B</b>) Protein content analysis of Nrf2, Keap1, HO-1, and NQO1 (<b>C</b>–<b>F</b>). CK group: blank control group; MG group: model group; PG group: positive omeprazole control group (20 mg/kg bw); LSG group: low-dose sesame peptide group (200 mg/kg bw); HSG group: high-dose sesame peptide group (400 mg/kg bw). Different letters represent significant differences between groups at <span class="html-italic">p</span> &lt; 0.05. Summary of schematic diagram of SPs protecting against ethanol-induced gastric mucosal injury (<b>G</b>). SPs enhance gastric mucosal barrier by regulating antioxidant factors, inflammatory factors, and activating Nrf2 signaling pathway, thereby alleviating ethanol-induced gastric mucosal injury in mice. Black arrows represent changes in indicator levels in the control and administered groups relative to the model group.</p>
Full article ">
24 pages, 17809 KiB  
Article
Transcriptomic Characterization Reveals Mitochondrial Involvement in Nrf2/Keap1-Mediated Osteoclastogenesis
by Eiko Sakai and Takayuki Tsukuba
Antioxidants 2024, 13(12), 1575; https://doi.org/10.3390/antiox13121575 - 20 Dec 2024
Viewed by 310
Abstract
Although osteoclasts play crucial roles in the skeletal system, the mechanisms that underlie oxidative stress during osteoclastogenesis remain unclear. The transcription factor Nrf2 and its suppressor, Keap1, function as central mediators of oxidative stress. To further elucidate the function of Nrf2/Keap1-mediated oxidative stress [...] Read more.
Although osteoclasts play crucial roles in the skeletal system, the mechanisms that underlie oxidative stress during osteoclastogenesis remain unclear. The transcription factor Nrf2 and its suppressor, Keap1, function as central mediators of oxidative stress. To further elucidate the function of Nrf2/Keap1-mediated oxidative stress regulation in osteoclastogenesis, DNA microarray analysis was conducted in this study using wild-type (WT), Keap1 knockout (Keap1 KO), and Nrf2 knockout (Nrf2 KO) osteoclasts. Principal component analysis showed that 403 genes, including Nqo1, Il1f9, and Mmp12, were upregulated in Keap1 KO compared with WT osteoclasts, whereas 24 genes, including Snhg6, Ccdc109b, and Wfdc17, were upregulated in Nrf2 KO compared with WT osteoclasts. Moreover, 683 genes, including Car2, Calcr, and Pate4, were upregulated in Nrf2 KO cells compared to Keap1 KO cells. Functional analysis by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis showed upregulated genes in Nrf2 KO osteoclasts were mostly enriched in oxidative phosphorylation. Furthermore, GeneMANIA predicted the protein–protein interaction network of novel molecules such as Rufy4 from genes upregulated in Nrf2 KO osteoclasts. Understanding the complex interactions between these molecules may pave the way for developing promising therapeutic strategies against bone metabolic diseases caused by increased osteoclast differentiation under oxidative stress. Full article
(This article belongs to the Special Issue Role of Nrf2 and ROS in Bone Metabolism)
Show Figures

Figure 1

Figure 1
<p>Microarray analysis of WT, <span class="html-italic">Nrf2</span> KO, and <span class="html-italic">Keap1</span> KO cells. (<b>A</b>) Splenic macrophages from WT, <span class="html-italic">Keap1</span> KO, and <span class="html-italic">Nrf2</span> KO mice were cultured with 30 ng/mL M-CSF and 50 ng/mL RANKL for three days, followed by TRAP staining. Representative photographs showing red-colored osteoclasts. (<b>a</b>) WT, (<b>b</b>) <span class="html-italic">Keap1</span> KO, and (<b>c</b>) <span class="html-italic">Nrf2</span> KO mice. Scale bars: 100 μm. (<b>B</b>) Splenic macrophages from two mice each of WT, <span class="html-italic">Keap1</span> KO, and <span class="html-italic">Nrf2</span> KO were cultured with 30 ng/mL M-CSF and 50 ng/mL RANKL for three days, and RNA was collected from each cell for DNA microarray analysis (single microarray analysis for each cell). Graphs showing scatter plots of (<b>a</b>) <span class="html-italic">Keap1</span> KO cells vs. WT osteoclasts, (<b>b</b>) <span class="html-italic">Nrf2</span> KO osteoclasts vs. WT osteoclasts, and (<b>c</b>) <span class="html-italic">Nrf2</span> KO osteoclasts vs. <span class="html-italic">Keap1</span> KO cells. Green lines indicate log<sub>2</sub>2 or log<sub>2</sub>0.5.</p>
Full article ">Figure 2
<p>Validation of microarray data. Upregulated genes in <a href="#antioxidants-13-01575-t001" class="html-table">Table 1</a> were confirmed by qRT-PCR. The relative mRNA levels of <span class="html-italic">Nqo1</span>, <span class="html-italic">Il1f9</span>, <span class="html-italic">Mmp12</span>, <span class="html-italic">Slc39a4</span>, <span class="html-italic">Fabp7</span>, <span class="html-italic">Cxcl14</span>, <span class="html-italic">Gsta3</span>, <span class="html-italic">Rnf128</span>, <span class="html-italic">Ly6g</span>, <span class="html-italic">Tanc2</span>, and <span class="html-italic">Gclm</span> in <span class="html-italic">Keap1</span> KO were confirmed. Data are presented as the mean ± SD from three independent experiments (* <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>Validation of microarray data. Downregulated genes in <a href="#antioxidants-13-01575-t002" class="html-table">Table 2</a> were confirmed by qRT-PCR. The relative mRNA levels of <span class="html-italic">Calcr</span>, <span class="html-italic">Scin</span>, <span class="html-italic">Ctsk</span>, <span class="html-italic">Pate4</span>, <span class="html-italic">Ocstamp</span>, <span class="html-italic">Ccr3</span>, <span class="html-italic">Tm4sf19</span>, and <span class="html-italic">Steap4</span> in <span class="html-italic">Keap1</span> KO were confirmed. Data are presented as the mean ± SD from three independent experiments (* <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>Validation of microarray data. Upregulated genes in <a href="#antioxidants-13-01575-t003" class="html-table">Table 3</a> were confirmed by qRT-PCR. (<b>A</b>) Significant upregulation of <span class="html-italic">Snhg6</span> and <span class="html-italic">ccdc109b</span> in <span class="html-italic">Nrf2</span> KO osteoclasts derived from splenocyte were confirmed. <span class="html-italic">Ppbp</span> gene expression tended to increase. (<b>B</b>) Significant upregulation of <span class="html-italic">Snhg6</span>, <span class="html-italic">Wfdc17</span>, <span class="html-italic">Ppbp</span>, and <span class="html-italic">Ctsk</span> in <span class="html-italic">Nrf2</span> KO osteoclasts derived from BMMs were confirmed. Data are presented as the mean ± SD from three independent experiments (* <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 5
<p>Validation of microarray data. Downregulated genes in <a href="#antioxidants-13-01575-t004" class="html-table">Table 4</a> were confirmed by qRT-PCR. Significant downregulation of <span class="html-italic">Ctse</span>, <span class="html-italic">Ifi202b</span>, <span class="html-italic">Me1</span>, <span class="html-italic">Cbr3</span>, <span class="html-italic">Thy1</span>, <span class="html-italic">Lrrc32</span>, <span class="html-italic">Rnf128</span>, <span class="html-italic">Cxcl14</span>, <span class="html-italic">Slc7a11</span>, and <span class="html-italic">Nqo1</span> in <span class="html-italic">Nrf2</span> KO osteoclasts were confirmed. Data are presented as the mean ± SD from three independent experiments (* <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>Validation of microarray data. Upregulated genes in <a href="#antioxidants-13-01575-t005" class="html-table">Table 5</a> were confirmed by qRT-PCR. Significant upregulation of <span class="html-italic">Calcr</span>, <span class="html-italic">Pate4</span>, <span class="html-italic">Oscar</span>, <span class="html-italic">Scin</span>, <span class="html-italic">Akr1c18</span>, <span class="html-italic">Ctsk</span>, <span class="html-italic">Steap4</span>, <span class="html-italic">Adck3</span>, <span class="html-italic">Tm4sf19</span>, <span class="html-italic">Atp6v0d2</span>, and <span class="html-italic">Ccr3</span> in <span class="html-italic">Nrf2</span> KO osteoclasts were confirmed. Data are presented as the mean ± SD from three independent experiments (** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 7
<p>Validation of microarray data. Downregulated genes in <a href="#antioxidants-13-01575-t006" class="html-table">Table 6</a> were confirmed by qRT-PCR. Significant downregulation of <span class="html-italic">Nqo1</span>, <span class="html-italic">Ctse</span>, <span class="html-italic">Cxcl14</span>, <span class="html-italic">Rnf128</span>, <span class="html-italic">Me1</span>, <span class="html-italic">Mmp12</span>, <span class="html-italic">Slc39a4</span>, <span class="html-italic">Gclm</span>, <span class="html-italic">Slc7a11</span>, <span class="html-italic">Cbr3</span>, and <span class="html-italic">Fabp7</span> in <span class="html-italic">Nrf2</span> KO osteoclasts were confirmed. Data are presented as the mean ± SD from three independent experiments (* <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 8
<p>GO enrichment analysis. (<b>A</b>) Up- or downregulated genes were analyzed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) for GO enrichment analysis in <span class="html-italic">Keap1</span> KO cells compared with WT osteoclasts. (<b>B</b>) Up- or downregulated genes were analyzed using DAVID for GO enrichment analysis in <span class="html-italic">Nrf2</span> KO osteoclasts compared with WT osteoclasts. (<b>C</b>) Up- or downregulated genes were analyzed using DAVID for GO enrichment analysis in <span class="html-italic">Nrf2</span> KO osteoclasts compared with <span class="html-italic">Keap1</span> KO cells.</p>
Full article ">Figure 8 Cont.
<p>GO enrichment analysis. (<b>A</b>) Up- or downregulated genes were analyzed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) for GO enrichment analysis in <span class="html-italic">Keap1</span> KO cells compared with WT osteoclasts. (<b>B</b>) Up- or downregulated genes were analyzed using DAVID for GO enrichment analysis in <span class="html-italic">Nrf2</span> KO osteoclasts compared with WT osteoclasts. (<b>C</b>) Up- or downregulated genes were analyzed using DAVID for GO enrichment analysis in <span class="html-italic">Nrf2</span> KO osteoclasts compared with <span class="html-italic">Keap1</span> KO cells.</p>
Full article ">Figure 8 Cont.
<p>GO enrichment analysis. (<b>A</b>) Up- or downregulated genes were analyzed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) for GO enrichment analysis in <span class="html-italic">Keap1</span> KO cells compared with WT osteoclasts. (<b>B</b>) Up- or downregulated genes were analyzed using DAVID for GO enrichment analysis in <span class="html-italic">Nrf2</span> KO osteoclasts compared with WT osteoclasts. (<b>C</b>) Up- or downregulated genes were analyzed using DAVID for GO enrichment analysis in <span class="html-italic">Nrf2</span> KO osteoclasts compared with <span class="html-italic">Keap1</span> KO cells.</p>
Full article ">Figure 9
<p>KEGG pathway enrichment analysis. Compared to <span class="html-italic">Keap1</span> KO cells, <span class="html-italic">Nrf2</span> KO osteoclasts exhibited a marked increase in expression of genes (surrounded by red lines) involved in oxidative phosphorylation (<b>A</b>) and osteoclast differentiation (<b>B</b>), whereas marked decreased in expression of genes (surrounded by blue lines) involved in focal adhesion (<b>C</b>) and ECM–receptor interaction (<b>D</b>).</p>
Full article ">Figure 9 Cont.
<p>KEGG pathway enrichment analysis. Compared to <span class="html-italic">Keap1</span> KO cells, <span class="html-italic">Nrf2</span> KO osteoclasts exhibited a marked increase in expression of genes (surrounded by red lines) involved in oxidative phosphorylation (<b>A</b>) and osteoclast differentiation (<b>B</b>), whereas marked decreased in expression of genes (surrounded by blue lines) involved in focal adhesion (<b>C</b>) and ECM–receptor interaction (<b>D</b>).</p>
Full article ">Figure 9 Cont.
<p>KEGG pathway enrichment analysis. Compared to <span class="html-italic">Keap1</span> KO cells, <span class="html-italic">Nrf2</span> KO osteoclasts exhibited a marked increase in expression of genes (surrounded by red lines) involved in oxidative phosphorylation (<b>A</b>) and osteoclast differentiation (<b>B</b>), whereas marked decreased in expression of genes (surrounded by blue lines) involved in focal adhesion (<b>C</b>) and ECM–receptor interaction (<b>D</b>).</p>
Full article ">Figure 10
<p>Protein–protein interaction network analysis by GeneMANIA. (<b>A</b>) Predicted network of proteins that interact with proteins encoded by the top 40 upregulated genes in <span class="html-italic">Nrf2</span> KO osteoclast against <span class="html-italic">Keap1</span> KO cells. (<b>B</b>) Predicted network of proteins interacting with proteins encoded by genes upregulated in Nrf2 KO osteoclast compared with WT osteoclasts.</p>
Full article ">Figure 10 Cont.
<p>Protein–protein interaction network analysis by GeneMANIA. (<b>A</b>) Predicted network of proteins that interact with proteins encoded by the top 40 upregulated genes in <span class="html-italic">Nrf2</span> KO osteoclast against <span class="html-italic">Keap1</span> KO cells. (<b>B</b>) Predicted network of proteins interacting with proteins encoded by genes upregulated in Nrf2 KO osteoclast compared with WT osteoclasts.</p>
Full article ">
14 pages, 4733 KiB  
Article
Rice Straw-Derived Biochar Mitigates Microcystin-LR-Induced Hepatic Histopathological Injury and Oxidative Damage in Male Zebrafish via the Nrf2 Signaling Pathway
by Wang Lin, Fen Hu, Wansheng Zou, Suqin Wang, Pengling Shi, Li Li, Jifeng Yang and Pinhong Yang
Toxins 2024, 16(12), 549; https://doi.org/10.3390/toxins16120549 - 18 Dec 2024
Viewed by 628
Abstract
Microcystin-leucine arginine (MC-LR) poses a serious threat to aquatic animals during cyanobacterial blooms. Recently, biochar (BC), derived from rice straw, has emerged as a potent adsorbent for eliminating hazardous contaminants from water. To assess the joint hepatotoxic effects of environmentally relevant concentrations of [...] Read more.
Microcystin-leucine arginine (MC-LR) poses a serious threat to aquatic animals during cyanobacterial blooms. Recently, biochar (BC), derived from rice straw, has emerged as a potent adsorbent for eliminating hazardous contaminants from water. To assess the joint hepatotoxic effects of environmentally relevant concentrations of MC-LR and BC on fish, male adult zebrafish (Danio rerio) were sub-chronically co-exposed to varying concentrations of MC-LR (0, 1, 5, and 25 μg/L) and BC (0 and 100 μg/L) in a fully factorial experiment. After 30 days exposure, our findings suggested that the existence of BC significantly decreased MC-LR bioavailability in liver. Furthermore, histopathological analysis revealed that BC mitigated MC-LR-induced hepatic lesions, which were characterized by mild damage, such as vacuolization, pyknotic nuclei, and swollen mitochondria. Compared to the groups exposed solely to MC-LR, decreased malondialdehyde (MDA) and increased catalase (CAT) and superoxide dismutase (SOD) were noticed in the mixture groups. Concurrently, significant changes in the mRNA expression levels of Nrf2 pathway genes (cat, sod1, gstr, keap1a, nrf2a, and gclc) further proved that BC reduces the oxidative damage induced by MC-LR. These findings demonstrate that BC decreases MC-LR bioavailability in the liver, thereby alleviating MC-LR-induced hepatotoxicity through the Nrf2 signaling pathway in zebrafish. Our results also imply that BC could serve as a potentially environmentally friendly material for mitigating the detrimental effects of MC-LR on fish. Full article
(This article belongs to the Special Issue Toxic Cyanobacterial Bloom Detection and Removal: What's New?)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>SEM images (<b>A</b>,<b>B</b>), size distribution (<b>C</b>), and zeta potential (<b>D</b>) of BC.</p>
Full article ">Figure 2
<p>Hepatic H&amp;E-stained sections of zebrafish. Control (<b>A</b>); 1 μg/L MC-LR (<b>B</b>); 5 μg/L MC-LR (<b>C</b>); 25 μg/L MC-LR (<b>D</b>); 100 μg/L BC (<b>E</b>); 1 μg/L MC-LR+ BC (<b>F</b>); 5 μg/L MC-LR + BC (<b>G</b>); and 25 μg/L MC-LR + BC (<b>H</b>). Pyknotic nuclei (black arrow), swollen hepatocytes (white arrowhead), and vacuolization (black arrowhead). Bar = 50 um.</p>
Full article ">Figure 3
<p>Hepatic ultrastructural alterations in zebrafish. Control (<b>A</b>); 1 μg/L MC-LR (<b>B</b>); 5 μg/L MC-LR (<b>C</b>); 25 μg/L MC-LR (<b>D</b>); 100 μg/L BC (<b>E</b>); 1 μg/L MC-LR + BC (<b>F</b>); 5 μg/L MC-LR+ BC (<b>G</b>); and 25 μg/L MC-LR + BC (<b>H</b>). Swollen mitochondria (black arrowhead); deformed nuclei (white arrowhead); vacuolization (white arrow); and dilated endoplasmic reticulum (black box). Mt, mitochondria; Nu, nuclei; ER, endoplasmic reticulum. Bar = 1 μm.</p>
Full article ">Figure 4
<p>Changes in MDA content (<b>A</b>), SOD activity (<b>B</b>), CAT activity (<b>C</b>), GPx activity (<b>D</b>), GST activity (<b>E</b>), and GSH content (<b>F</b>) in the liver of zebrafish. Asterisks (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01) indicate significant differences between the treatments and control. Hashes (# <span class="html-italic">p</span> &lt; 0.05) indicate significant differences between the MC-LR + BC groups and the MC-LR-only groups, respectively. The values are presented as mean ± SD (n = 3).</p>
Full article ">Figure 5
<p>Heatmap of Nrf2 signaling pathway gene expression. Asterisks (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01) indicate significant differences between the treatments and control. The dotted box with hashes (# <span class="html-italic">p</span> &lt; 0.05; ## <span class="html-italic">p</span> &lt; 0.01) indicate significant differences between the MC-LR + BC groups and the MC-LR-only groups, respectively. The values are presented as mean ± SD (n = 6).</p>
Full article ">
15 pages, 2659 KiB  
Article
Bacillus amyloliquefaciens Regulates the Keap1/Nrf2 Signaling Pathway to Improve the Intestinal (Caco-2 Cells and Chicken Jejunum) Oxidative Stress Response Induced by Lipopolysaccharide (LPS)
by Xing Chen, Aijuan Zheng, Shuzhen Li, Zedong Wang, Zhimin Chen, Jiang Chen, Zhiheng Zou, Haijun Liang and Guohua Liu
Antioxidants 2024, 13(12), 1550; https://doi.org/10.3390/antiox13121550 - 17 Dec 2024
Viewed by 569
Abstract
This article aims to investigate the mechanism by which Bacillus amyloliquefaciens alleviates lipopolysaccharide (LPS)-induced intestinal oxidative stress. The study involved two experimental subjects: human colorectal adenocarcinoma (Caco-2) cells and Arbor Acres broiler chickens. The experiment involving two samples was designed with the same [...] Read more.
This article aims to investigate the mechanism by which Bacillus amyloliquefaciens alleviates lipopolysaccharide (LPS)-induced intestinal oxidative stress. The study involved two experimental subjects: human colorectal adenocarcinoma (Caco-2) cells and Arbor Acres broiler chickens. The experiment involving two samples was designed with the same treatment groups, specifically the control (CK) group, lipopolysaccharide (LPS) group, Bacillus amyloliquefaciens (JF) group, and JF+LPS group. In the Caco-2 experiment, we administered 2 μg/mL of LPS and 1 × 106 CFU/mL of JF to the LPS and JF groups, respectively. In the broiler experiment, the LPS group (19–21 d) received an abdominal injection of 0.5 mg/kg BW of LPS, whereas the JF group was fed 1 × 107 CFU/g of JF throughout the entire duration of the experiment (1–21 d). The results indicated the following: (1) JF significantly decreased the DPPH free radical clearance rate and hydrogen peroxide levels (p < 0.05). (2) JF significantly enhanced the total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and glutathione peroxidase (GSH Px) activity in Caco-2 cells (p < 0.05), while concurrently reducing malondialdehyde (MDA) content (p < 0.05). (3) Compared to the CK group, JF significantly increased the mRNA expression levels of nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), SOD, catalase (CAT), GSH-Px, interleukin-4 (IL-4), interleukin-10 (IL-10), Claudin, Occludin1, zonula occludens-1 (ZO-1), and mucin 2 (MUC2) in Caco-2 cells (p < 0.05), while concurrently reducing the mRNA expression of Kelch-like ECH-associated protein 1 (Keap1), tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-8 (IL-8) (p < 0.05). In comparison to the LPS group, the JF+LPS group demonstrated a significant increase in the mRNA expression of Nrf2, SOD, GSH-Px, and IL-4, as well as Occludin1, ZO-1, and MUC2 in Caco-2 cells (p < 0.05), alongside a decrease in the mRNA expression of Keap1, TNF-α, and IL-1β (p < 0.05). (4) In broiler chickens, the JF group significantly elevated the levels of T-AOC, CAT, and GSH-Px in the jejunum while reducing MDA content (p < 0.05). Furthermore, the CAT level in the JF+LPS group was significantly higher than that observed in the LPS group, and the levels of MDA, TNF-α, and IL-1β were significantly decreased (p < 0.05). (5) In comparison to the CK group, the JF group exhibited a significant increase in Nrf2 levels in the jejunum of broiler chickens (p < 0.05). Notably, the mRNA expression levels of IL-4, IL-10, Claudin, Occludin1, ZO-1, and MUC2 were reduced (p < 0.05), while the mRNA expression levels of Keap1, TNF-α, and IL-1β also showed a decrease (p < 0.05). Furthermore, the mRNA expression levels of Nrf2, Occludin1, ZO-1, and MUC2 in the JF+LPS group were significantly elevated compared to those in the LPS group (p < 0.05), whereas the mRNA expression levels of Keap1 and TNF-α were significantly diminished (p < 0.05). In summary, JF can enhance the intestinal oxidative stress response, improve antioxidant capacity and intestinal barrier function, and decrease the expression of inflammatory factors by regulating the Keap1/Nrf2 signaling pathway. Full article
Show Figures

Figure 1

Figure 1
<p>Cellular antioxidant capacity in Caco-2 cells. (<b>A</b>) DPPH free radical clearance rate; (<b>B</b>) hydrogen peroxide content; (<b>C</b>) T-AOC, total antioxidant capacity; (<b>D</b>) SOD, superoxide dismutase; (<b>E</b>) MDA, malondialdehyde; (<b>F</b>) GSH-Px, glutathione peroxidase. CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 2
<p>mRNA expression of cellular antioxidant in Caco-2 cells. (<b>A</b>) mRNA relative expression (<span class="html-italic">Keap1</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">Nrf2</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">HO-1</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">SOD</span>/<span class="html-italic">GAPDH</span>); (<b>E</b>) mRNA relative expression (<span class="html-italic">CAT</span>/<span class="html-italic">GAPDH</span>); (<b>F</b>) mRNA relative expression (<span class="html-italic">GSH-Px</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 3
<p>mRNA expression of cellular inflammatory factors in Caco-2 cells. (<b>A</b>) mRNA relative expression (<span class="html-italic">TNF-α</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">IL-1β</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">IL-6</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">IL-8</span>/<span class="html-italic">GAPDH</span>); (<b>E</b>) mRNA relative expression (<span class="html-italic">IL-4</span>/<span class="html-italic">GAPDH</span>); (<b>F</b>) mRNA relative expression (<span class="html-italic">IL-10</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 4
<p>mRNA expression of cellular intestinal barrier function in Caco-2 cells. (<b>A</b>) mRNA relative expression (<span class="html-italic">Claudin</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">Occludin1</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">ZO-1</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">MUC2</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 5
<p>Antioxidant index of 21 d broiler jejunum. (<b>A</b>) T-AOC, total antioxidant capacity; (<b>B</b>) SOD, superoxide dismutase; (<b>C</b>) CAT, catalase; (<b>D</b>) MDA, malondialdehyde; (<b>E</b>) GSH-Px, glutathione peroxidase. CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 6.</p>
Full article ">Figure 6
<p>Inflammatory factors of 21 d broiler jejunum. (<b>A</b>) TNF-α, tumor necrosis factor-alpha; (<b>B</b>) IL-1β, interleukin-1β; (<b>C</b>) IL-6, interleukin-6; (<b>D</b>) IL-8, interleukin-8; (<b>E</b>) IL-4, interleukin-4; (<b>F</b>) IL-10, interleukin-10. CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 6.</p>
Full article ">Figure 7
<p>Expression of antioxidant-related mRNA in 21 d broiler jejunum. (<b>A</b>) mRNA relative expression (Keap1/GAPDH); (<b>B</b>) mRNA relative expression (Nrf2/GAPDH); (<b>C</b>) mRNA relative expression (HO-1/GAPDH). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 8
<p>Expression of inflammatory factor-related mRNA in 21 d broiler jejunum. (<b>A</b>) mRNA relative expression (<span class="html-italic">TNF-α</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">IL-1β</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">IL-4</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">IL-10</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 9
<p>Expression of intestinal barrier function-related mRNA in 21 d broiler jejunum. (<b>A</b>) mRNA relative expression (<span class="html-italic">Claudin</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">Occludin1</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">ZO-1</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">MUC2</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">
22 pages, 5873 KiB  
Article
Quality by Design Approach for the Formulation and Evaluation of Stem Cells Derived Rosmarinic Acid-Loaded Nanofibers as an Anti-Wrinkle Patch: In Vitro and In Vivo Characterizations
by Rehab Abdelmonem, Ahmed Bakr, Ingy Badawy, Ahmed Ibrahim Abd El Maksoud and Reem T. Attia
Pharmaceutics 2024, 16(12), 1598; https://doi.org/10.3390/pharmaceutics16121598 - 16 Dec 2024
Viewed by 515
Abstract
Background/Objectives: Skin wrinkles result from a myriad of multifaceted processes involving intrinsic and extrinsic aging. To combat this effect, plant stem cells offer a renewable and eco-friendly source for various industries, including cosmeceuticals. Salvia miltiorrhiza (SM), which contains the bioactive compound Rosmarinic acid [...] Read more.
Background/Objectives: Skin wrinkles result from a myriad of multifaceted processes involving intrinsic and extrinsic aging. To combat this effect, plant stem cells offer a renewable and eco-friendly source for various industries, including cosmeceuticals. Salvia miltiorrhiza (SM), which contains the bioactive compound Rosmarinic acid (RA) and has been proposed for its anti-wrinkle effect. Methods: In the present study, calli from SM were cultured and Quality by Design (QbD) was implemented to investigate the effect of different types and concentrations of elicitors; jasmonic acid (JA) and salicylic acid (SA). Both raised RA levels yet, jasmonic acid (50 µM) has resulted in the highest yield for RA, at 16 mg/g. A nanofiber patch was prepared and characterized in-vitro by the release percentage, drug content, swelling degree, scanning electron microscope, and surface roughness. Then, the anti-wrinkle effect of the patch was tested in a UV wrinkle-induced mouse model. Results: Interestingly, after treatment, there were visibly fewer wrinkles, and the skin was softer than in the untreated control group. This study suggests that the treatment exerted its effect through the Nrf2/Keap1 pathway, which plays a crucial role in cellular antioxidant protective processes. By activating this pathway through boosting Nrf2 and diminishing Keap1 cellular content, the nanofiber patch enhances the production of antioxidant enzymes, such as superoxide dismutase and glutathione peroxidase, enhancesglutathione, and reduces the skin lipid peroxidation, collectively indicating enhanced skin quality. Conclusions: In conclusion, this study highlights the importance of this formula as an anti-wrinkle treatment, and future clinical studies are recommended to further unveil the potential of this formula. Full article
Show Figures

Figure 1

Figure 1
<p>A chart showing the effect of different concentrations of Jasmonic acid and Salicylic acid on Rosmarinic acid (RA) yield in the <span class="html-italic">Salvia miltiorrhiza</span> stem cell extract compared with the (0) control: RA yield without elicitors, which is indicated with the green column.</p>
Full article ">Figure 2
<p>The effect of Jasmonic acid and Salicylic acid elicitor concentrations on Rosmarinic acid level, solid lines represent the predicted responses from the statistical model and dashed lines Indicate observed experimental responses.</p>
Full article ">Figure 3
<p>Desirability ramps for the ideal elicitor concentration where red dots represent jasmonic acid concentration and blue dots represent rosmarinic acid concentration.</p>
Full article ">Figure 4
<p>In vitro release profile of Rosmarinic acid from nanofiber patch in acetate buffer pH 5.5. Data are presented as mean+/− SD at <span class="html-italic">p</span> &lt; 0.05 <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 5
<p>FTIR spectrum of Polyurethane/Rosmarinic acid electrospun nanofiber.</p>
Full article ">Figure 6
<p>In vitro release profile of Rosmarinic acid electrospun nanofiber in acetate buffer pH 5.5 showing the patch’s stability after 3 months of preparation. Data are presented as mean+/− SD at <span class="html-italic">p</span> &lt; 0.05 <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 7
<p>SEM images of electrospun RA/PU nanofiber patch, polymer solution 10%wt with flow rate 0.8 mL/h and 25 kV, (<b>A</b>) 30 µM, (<b>B</b>) 10 µM, (<b>C</b>) Free Polyurethane. Green arrows sign to the nanospheres of extracted rosmarinic acid, yellow arrows sign to the uniformly loaded nanofibers.</p>
Full article ">Figure 8
<p>Surface roughness images for Rosmarinic acid/Polyurethane nanofiber patch.</p>
Full article ">Figure 9
<p>Photograph of a sample of the mice’s back after treatment. (<b>A</b>) Normal control (NC) group, (<b>B</b>) positive control (PC) group, (<b>C</b>) free patch group (Free-P), (<b>D</b>) liquid RA group, (<b>E</b>) medicated nanofiber patch-treated group (Mediated P).</p>
Full article ">Figure 10
<p>Skin histopathological images. (<b>A</b>) Normal control (NC) group, (<b>B</b>) positive control (PC) group, (<b>C</b>) free patch group (Free-P), (<b>D</b>) liquid RA group, (<b>E</b>) medicated nanofiber patch-treated group (Medicated P). Arrows represent the variation in epidermal thickening and the presence of necrotic tissues.</p>
Full article ">Figure 11
<p>Effect of different treatments on skin Nrf2 and Keap1 content. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6). Statistical analysis was carried out using one-way ANOVA followed by Tukey’s Multiple Comparison test, and the used <span class="html-italic">p</span> value is shown for each comparison in the graph. Each shape over the bar-chart represents a sample that was tested.</p>
Full article ">Figure 12
<p>Effect of different treatments on skin GSH and GPx, SOD, MDA. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6). Statistical analysis was carried out using one-way ANOVA followed by Tukey’s Multiple Comparison test, and the used <span class="html-italic">p</span> value is shown for each comparison in the graph. Each shape over the bar-chart represents a sample that was tested.</p>
Full article ">
17 pages, 2160 KiB  
Article
Harshly Oxidized Activated Charcoal Enhances Protein Persulfidation with Implications for Neurodegeneration as Exemplified by Friedreich’s Ataxia
by Anh T. T. Vo, Uffaf Khan, Anton V. Liopo, Karthik Mouli, Kenneth R. Olson, Emily A. McHugh, James M. Tour, Madhavan Pooparayil Manoj, Paul J. Derry and Thomas A. Kent
Nanomaterials 2024, 14(24), 2007; https://doi.org/10.3390/nano14242007 - 13 Dec 2024
Viewed by 513
Abstract
Harsh acid oxidation of activated charcoal transforms an insoluble carbon-rich source into water-soluble, disc structures of graphene decorated with multiple oxygen-containing functionalities. We term these pleiotropic nano-enzymes as “pleozymes”. A broad redox potential spans many crucial redox reactions including the oxidation of hydrogen [...] Read more.
Harsh acid oxidation of activated charcoal transforms an insoluble carbon-rich source into water-soluble, disc structures of graphene decorated with multiple oxygen-containing functionalities. We term these pleiotropic nano-enzymes as “pleozymes”. A broad redox potential spans many crucial redox reactions including the oxidation of hydrogen sulfide (H2S) to polysulfides and thiosulfate, dismutation of the superoxide radical (O2*), and oxidation of NADH to NAD+. The oxidation of H2S is predicted to enhance protein persulfidation—the attachment of sulfur to cysteine residues. Persulfidated proteins act as redox intermediates, and persulfidation protects proteins from irreversible oxidation and ubiquitination, providing an important means of signaling. Protein persulfidation is believed to decline in several neurological disorders and aging. Importantly, and consistent with the role of persulfidation in signaling, the master antioxidant transcription factor Nrf2 is regulated by Keap1’s persulfidation. Here, we demonstrate that pleozymes increased overall protein persulfidation in cells from apparently healthy individuals and from individuals with the mitochondrial protein mutation responsible for Friedreich’s ataxia. We further find that pleozymes specifically enhanced Keap1 persulfidation, with subsequent increased accumulation of Nrf2 and Nrf2’s antioxidant targets. Full article
(This article belongs to the Special Issue Carbon-Based Nanomaterials for Biomedicine Applications)
Show Figures

Figure 1

Figure 1
<p>The diagram of the dimedone-switch method to detect persulfidation. NBF-Cl forms a bond with a sulfur atom in thiol (-SH), persulfide (-SSH), and nitrogen atoms in the amino group (-NH<sub>2</sub>) and tags the protein with a green fluorescence (first reactions in top and bottom panels). A dimedone switch selectively replaces the NBF-binding on persulfide and labels persulfide with a red fluorescence (second reaction in bottom panel). The dimedone switch cannot displace the NBF-binding on the thiol or amino group (second reaction in top panel). Figure adapted from Živanović et al., 2020 [<a href="#B20-nanomaterials-14-02007" class="html-bibr">20</a>].</p>
Full article ">Figure 2
<p>Pleozymes induced global protein persulfidation in endothelial cells and fibroblasts, with a focus on high- and low-molecular-weight proteins. (<b>A</b>) Persulfidation signals relative to total protein increased with pleozyme treatment at therapeutically relevant dosages of 4 µg/mL versus untreated control in cultured brain endothelial (bEnd.3) cells. The high-molecular-weight (MW) proteins from 40 to 140 kDa (white box) expressed less persulfidation in the control and enriched persulfidation/total protein with pleozyme treatment and were less consistent from the H<sub>2</sub>S donor, GYY4137. One representative gel image/experiment from N = 4 independent experiments. (<b>B</b>,<b>C</b>) Pleozymes also increased persulfidation/total protein ratio in fibroblasts from apparently healthy individuals (AHI). Fibroblasts exhibited two persulfidation-rich regions with pleozymes: high MW from 40 to 140 kDa (white box) and low molecular weight from 12 to 13 kDa (white, dash box). (<b>D</b>) The persulfidation-rich region/total protein region in endothelial cells quantified with N = 4 independent experiments was significantly different for the PEG-OAC pleozymes. Student’s <span class="html-italic">t</span>-test, paired, two-tailed, * represented <span class="html-italic">p</span> &lt; 0.05. Individual values for high MW and low MW persulfidation-rich region/total protein region quantification are shown in (<b>E</b>,<b>F</b>), including the H<sub>2</sub>S donor, GYY41237, whose effects were less consistent than the pleozymes.</p>
Full article ">Figure 3
<p>Pleozymes increased Keap1 persulfidation and induced the expression of Nrf2 and its antioxidant target genes. (<b>A</b>) Pleozymes increased KEAP1 persulfidation in brain endothelial (bEnd.3) cells at t = 1 h. N = 4 technical replicates. One-way ANOVA followed by Tukey’s multiple comparisons test, ** represented <span class="html-italic">p</span> &lt; 0.01, ns represented not significant. Error bars represented standard deviation from 4 technical replicates. (<b>B</b>) Pleozymes induced Nrf2 with the presence of lipoic acid, an H<sub>2</sub>S donor. (<b>C</b>) Co-treating pleozymes with lipoic acid prolonged the pleozyme-induced Nrf2 accumulation (3 h). This was seen in all 3 independent experiments. (<b>D</b>) The expression of Nrf2 targets increased after treatment with pleozymes and/or lipoic acid.</p>
Full article ">Figure 4
<p>Co-treatment of pleozymes and lipoic acid, a H<sub>2</sub>S donor, induced the formation of the disulfide intramolecular (OxIM) bond of Keap1 after 3 h. Brain endothelial (bEnd.3) cells were treated with pleozymes and/or lipoic acid for three hours, following which cell lysates were collected and prepared for a redox Western blot to visualize oxidative modifications of Keap1. Keap1 existed in its reduced form (reduced band) and two oxidative forms (OxIR1 and OxIR2 bands with oxidized intermolecular disulfide bonds) at basal conditions. In the pleozyme-treated group, pleozymes maintained Keap1 oxidative expressions similarly to the control group. Exposure to lipoic acid, a H<sub>2</sub>S donor, induced the appearance of the oxidized intramolecular (OxIM) disulfide bonds and decreased the intensity of the reduced band, regardless of pleozymes treatment.</p>
Full article ">Figure 5
<p>Treatment of FRDA skin fibroblasts (N = 4) with pleozymes (t = 1 h) significantly increased the persulfidation of both higher- and lower-molecular-weight proteins. In skin fibroblasts, cultured cells derived from Friedreich’s ataxia individuals (FRDA), persulfidation signals relative to total protein increased with pleozyme treatment at therapeutically relevant dosages of 4 µg/mL versus untreated (N = 4 biological replicates, 2 females and 2 males). Two representative examples (<b>A</b>,<b>B</b>) demonstrated enriched persulfidation in high-molecular-weight (MW) proteins from 40 to 140 kDa (white box) and lower-molecular-weight proteins from 12 to 13 kDa (dashed white box) in these FRDA fibroblasts compared to the untreated group. (<b>C</b>) Quantification of the intensity ratio of the -SSH (persulfidation) intensities/total protein intensities. Statistical analysis between untreated and treated groups was performed with the paired <span class="html-italic">t</span>-test, * represented <span class="html-italic">p</span> &lt; 0.05, and ** represented <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
27 pages, 2646 KiB  
Review
Role of NRF2 in Pathogenesis of Alzheimer’s Disease
by Ching-Tung Chu, Akira Uruno, Fumiki Katsuoka and Masayuki Yamamoto
Antioxidants 2024, 13(12), 1529; https://doi.org/10.3390/antiox13121529 - 13 Dec 2024
Viewed by 513
Abstract
Alzheimer’s disease (AD) is a polygenic, multifactorial neurodegenerative disorder and remains the most prevalent form of dementia, globally. Despite decades of research efforts, there is still no effective cure for this debilitating condition. AD research has increasingly focused on transcription factor NRF2 (nuclear [...] Read more.
Alzheimer’s disease (AD) is a polygenic, multifactorial neurodegenerative disorder and remains the most prevalent form of dementia, globally. Despite decades of research efforts, there is still no effective cure for this debilitating condition. AD research has increasingly focused on transcription factor NRF2 (nuclear factor erythroid 2-related factor 2) as a potential therapeutic target. NRF2 plays a crucial role in protecting cells and tissues from environmental stressors, such as electrophiles and reactive oxygen species. Recently, an increasing number of studies have demonstrated that NRF2 is a key regulator in AD pathology. NRF2 is highly expressed in microglia, resident macrophages in the central nervous system, and contributes to neuroinflammation, phagocytosis and neurodegeneration in AD. NRF2 has been reported to modulate microglia-induced inflammation and facilitate the transition from homeostatic microglia to a disease-associated microglia subset. Genetic and pharmacological activation of NRF2 has been demonstrated to improve cognitive function. Here, we review the current understanding of the involvement of NRF2 in AD and the critical role that NRF2 plays in microglia in the context of AD. Our aim is to highlight the potential of targeting NRF2 in the microglia as a promising therapeutic strategy for mitigating the progression of AD. Full article
(This article belongs to the Special Issue Role of NRF2 Pathway in Neurodegenerative Diseases)
Show Figures

Figure 1

Figure 1
<p>Domain architectures of NRF2 and KEAP1. (<b>A</b>). NRF2 (nuclear actor erythroid 2-related factor 2) consists of seven domains, referred to as Neh1 to Neh7, each identified based on their specific biological functions and their homology to other protein domains. Neh stands for Nrf2-Ech homology domain. These domains play the crucial regulatory roles of NRF2 in cellular defense mechanisms, including oxidative stress response and detoxification processes, and anti-inflammation. (<b>B</b>). KEAP1 (Kelch-like ECH-associated protein) is composed of five domains. BTB domain and DGR domain are protein–protein interaction domains, separated by an intervening region (IVR). The BTB domain regulates homodimerization of KEAP1 and binding to CUL3 (Cullin3), while the DGR domain and the C-terminal region facilitate the binding with Neh2 domain of NRF2.</p>
Full article ">Figure 2
<p>Schematic presentation of the KEAP1-NRF2 system. Under normal conditions (black arrows), NRF2 is bound to KEAP1 in the cytoplasm. KEAP1 targets NRF2 for ubiquitination, leading to its degradation via the proteasomal pathway. Upon exposure to oxidative stress or NRF2 inducers (orange arrows), the KEAP1 undergoes modifications that reduce its ubiquitin ligase activity, resulting in weakening of KEAP1 for ubiquitinating NRF2. Subsequently, newly synthesized NRF2 translocates to the nucleus. In the nucleus, NRF2 binds to CNC-sMaf-binding elements (CsMBEs) or antioxidant response elements (AREs) in the promoter/enhancer regions of target genes. This binding activates the transcription of genes involved in the synthesis of reduced glutathione (GSH), antioxidant defense, detoxification processes, iron metabolism and inflammation.</p>
Full article ">Figure 3
<p>Expression levels of NRF2 in three major central nervous system (CNS) cell types. Microglia show high NRF2 expression, astrocytes exhibit medium NRF2 expression, and neurons display low NRF2 expression.</p>
Full article ">Figure 4
<p>NRF2 regulation of microglial states in AD. The downregulation of NRF2 in AD microglia enhances the expression of neurodegenerative phenotype markers, such as <span class="html-italic">CD11b</span>, <span class="html-italic">CD86</span>, and <span class="html-italic">iNOS</span>. This phenomenon has been observed in AD mini-brains, AD mouse models, and AD patients [<a href="#B190-antioxidants-13-01529" class="html-bibr">190</a>]. Conversely, the activation of NRF2 in AD mice leads to a reduction in the expression of disease-associated microglia (DAM) markers, including <span class="html-italic">TREM2</span>, <span class="html-italic">TYROBP</span>, <span class="html-italic">CST7</span>, and <span class="html-italic">ITGAX</span> [<a href="#B76-antioxidants-13-01529" class="html-bibr">76</a>]. These findings collectively underscore the crucial role of NRF2 in regulating the phenotypic transition of microglia within the context of AD, suggesting that enhancing NRF2 signaling could be a potential therapeutic strategy to modulate neuroinflammation and improve outcomes in AD.</p>
Full article ">Figure 5
<p>Schematic representation of NRF2-mediated neuroprotection and ferroptosis in AD. The relationship between ferroptosis and AD pathologies is increasingly being recognized. Activation of the NRF2 pathway is associated with neuroprotection in the brain, primarily through the upregulation of antioxidant genes such as GPX4 and NQO1, as well as genes involved in iron metabolism. Oxidative stress and iron deposition in the brain can promote ferroptosis, which exacerbates AD pathologies. The correlation between NRF2 activation and reduced ferroptosis activity suggests that NRF2 may serve as a potential therapeutic target for mitigating neurodegeneration in AD, in part by preventing ferroptosis.</p>
Full article ">Figure 6
<p>Schematic representation of NRF2 functions in CNS. (<b>A</b>) Cell type-specific functions of NRF2 in CNS. Diverse roles of NRF2 in three distinct cell types within the CNS and vasculature are shown. Note that in microglia, NRF2 reduces inflammation, enhances antioxidant defenses, and regulates phagocytosis, contributing to immune homeostasis and neuroprotection. In astrocytes, NRF2 promotes glutathione (GSH) production, alleviates metabolic stress, and reduces inflammation, supporting neuronal survival and CNS health. In neurons, NRF2 improves mitochondrial function and resistance to oxidative damage, protecting against neurodegeneration. In the vasculature, NRF2 enhances endothelial barrier function and mitigates inflammatory responses, maintaining vascular integrity and reducing neurovascular dysfunction. Collectively, NRF2 serves as a central regulator of oxidative stress and inflammation across multiple CNS cell types and the vascular system, highlighting its therapeutic potential in neurodegenerative diseases such as AD. (<b>B</b>) Schematic representation of NRF2 regulation in the CNS. When NRF2 activated in the CNS, NRF2 regulates the activation of microglia and astrocytes, leading to a decrease in inflammatory responses and a reduction in oxidative stress. Additionally, NRF2 plays a vital role in maintaining iron homeostasis, enhancing mitochondrial function and protection of cerebrovascular health. Together, these effects of NRF2 activation underscore its significant influence in mitigating AD pathology by modulating cellular responses and processes.</p>
Full article ">
16 pages, 6036 KiB  
Article
Ganoderma lucidum Spore Powder Alleviates Metabolic-Associated Fatty Liver Disease by Improving Lipid Accumulation and Oxidative Stress via Autophagy
by Yuxuan Zhang, Jiali Zhou, Lan Yang, Hang Xiao, Dongbo Liu and Xincong Kang
Antioxidants 2024, 13(12), 1501; https://doi.org/10.3390/antiox13121501 - 9 Dec 2024
Viewed by 964
Abstract
Lipid accumulation and oxidative stress, which could be improved by autophagy, are the “hits” of metabolic-associated fatty liver disease (MAFLD). Ganoderma lucidum spore powder (GLSP) has the effect of improving liver function. However, there are few reports about its effects on and mechanisms [...] Read more.
Lipid accumulation and oxidative stress, which could be improved by autophagy, are the “hits” of metabolic-associated fatty liver disease (MAFLD). Ganoderma lucidum spore powder (GLSP) has the effect of improving liver function. However, there are few reports about its effects on and mechanisms impacting MAFLD alleviation. This study investigated the effect of GLSP on hepatic lipid accumulation and oxidative stress and explored the role that autophagy played in this effect. The results showed that GLSP effectively reduced lipid accumulation and activated autophagy in the livers of mice with high-fat-diet-induced disease and palmitic acid-induced hepatocytes. GLSP reduced the lipid accumulation by reducing lipogenesis and promoting lipid oxidation in HepG2 cells. It decreased the production of ROS, increased the activity of SOD and CAT, and improved the mitochondrial membrane potential via the Keap1/Nrf2 pathway. The alleviating effects of GLSP on the lipid accumulation and oxidative stress was reversed by 3-methyladenine (3-MA), an autophagy inhibitor. GLSP activated autophagy via the AMPK pathway in HepG2 cells. In conclusion, GLSP could attenuate MAFLD by the improvement of lipid accumulation and oxidative stress via autophagy. This paper is the first to report the improvement of MAFLD through autophagy promotion. It will shed novel light on the discovery of therapeutic strategies targeting autophagy for MAFLD. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>GLSP alleviated high-fat-induced MAFLD in mice. (<b>A</b>) Food intake (<span class="html-italic">n</span> = 12 in each group), (<b>B</b>) body weight (<span class="html-italic">n</span> = 12 in each group), and (<b>C</b>) body weight gain (<span class="html-italic">n</span> = 12 in each group); (<b>D</b>) representative images of livers; (<b>E</b>) liver weight (<span class="html-italic">n</span> = 12 in each group); (<b>F</b>) liver index (weight of liver/weight of body; <span class="html-italic">n</span> = 12 in each group); (<b>G</b>) representative images of H&amp;E and Oil Red O staining of liver; (<b>H</b>) TG and TC levels in liver and serum (<span class="html-italic">n</span> = 6 in each group). NCD: control group; HFD: high-fat diet group; SIM: simvastatin (15 mg/kg b.w/day) group; GLSP: GLSP (3%) group. Data are expressed as mean ± SEM, and different letters indicate significant differences (<span class="html-italic">p</span> &lt; 0.05). Statistical differences were assessed by Tukey’s test of one-way ANOVA.</p>
Full article ">Figure 2
<p>GLSP reduced palmitic acid (PA)-induced lipid accumulation in HepG2 cells. (<b>A</b>) Effects of 0, 200, 400, 800, 1600, and 3200 μg/mL of GLSP on cell viability after 12 h treatment. (<b>B</b>) Intracellular lipid accumulation was analyzed by Oil red O staining; scale bars, 50 μm. (<b>C</b>,<b>D</b>) Intracellular TG and TC levels in HepG2 cells. (<b>E</b>) Western blot analysis of ACC, FASN, SREBP1, ACOX1, CPT1A, PPARα, and GAPDH protein levels. HepG2 cells treated with or without PA (0.6 mM) and with or without GLSP (50, 200, 800 μg/mL). Data are expressed as mean ± SEM, and different letters indicate significant differences (<span class="html-italic">p</span> &lt; 0.05). Statistical differences were assessed by Tukey’s test of one-way ANOVA.</p>
Full article ">Figure 3
<p>Effect of GLSP on PA-induced oxidative stress in HepG2 cells. (<b>A</b>) ROS production detected by DCFH-DA detector; scale bars, 50 μm. (<b>B</b>) SOD activity was measured with Total Superoxide Dismutase Assay Kit with WST-8. (<b>C</b>) CAT activity was measured with Micro CAT Assay Kit. (<b>D</b>) Effect of GLSP on mitochondrial damage. GLSP was added into HepG2 cells for 12 h. Representative images of JC-1-derived red and green fluorescence; scale bars, 50 μm. (<b>E</b>) Western blot analysis of Keap1, Nrf2, and HO-1 protein levels with GAPDH as control. HepG2 cells treated with or without PA (0.6 mM) and with or without GLSP (50, 200, 800 μg/mL). Data are expressed as mean ± S.E., and different letters indicate significant differences (<span class="html-italic">p</span> &lt; 0.05). Statistical differences were assessed by Tukey’s test of one-way ANOVA.</p>
Full article ">Figure 4
<p>GLSP induced autophagy in vivo and in vitro and activated autophagy via AMPK signal pathway in HepG2 cells. (<b>A</b>) Representative images of LC3 immunohistochemistry of liver sections. (<b>B</b>) LC3 immunofluorescence staining in HepG2 cells; scale bars, 10 μm. Liver sections or HepG2 cells were stained with anti-LC3 antibody and observed with laser confocal microscope. (<b>C</b>) Effects of GLSP on protein expressions of autophagic indicators LC3, P-AMPK/AMPK, and P-mTOR/mTOR in HepG2 cells, with GAPDH as control. HepG2 cells were treated with or without PA (0.6 mM) and with or without GLSP (50, 200, 800 μg/mL).</p>
Full article ">Figure 5
<p>GLSP improved lipid accumulation in HepG2 cells by inducing autophagy. HepG2 cells were treated with 800 µg/mL of GLSP and 3 mM of 3-MA for 12 h and then treated with PA (0.6 mM) for 12 h. (<b>A</b>) LC3 immunofluorescence staining showed endogenous LC3 level of HepG2 cells; scale bars, 10 μm. (<b>B</b>) Western blot analysis of LC3, P-AMPK/AMPK, and P-mTOR/mTOR protein levels, with GAPDH as control. (<b>C</b>) Intracellular lipid accumulation was analyzed by Oil Red O staining; scale bars, 50 μm. (<b>D</b>,<b>E</b>) Intracellular TG and TC levels in HepG2 cells. (<b>F</b>) Western blot analysis of P-ACC/ACC, FASN, SREBP1, ACOX1, CPT1A, and PPARα protein levels, with GAPDH as control. Data are expressed as mean ± SEM, and different letters indicate significant differences (<span class="html-italic">p</span> &lt; 0.05). Statistical differences were assessed by Tukey’s test of one-way ANOVA.</p>
Full article ">Figure 6
<p>Autophagy contributed to effect of GLSP protecting HepG2 cells from oxidative stress under PA stress. HepG2 cells were treated with 800 µg/mL of GLSP and/or 3 mM of 3-MA for 12 h and then treated with 0.6 mM PA for 12 h. (<b>A</b>) Intracellular ROS in HepG2 cells after fluorescence staining is exhibited; scale bars, 50 µm. (<b>B</b>,<b>C</b>) SOD and CAT were measured with Total Superoxide Dismutase Assay Kit with WST-8 and Micro Catalase (CAT) Assay Kit in HepG2 cells. (<b>D</b>) Representative images of JC-1-derived red and green fluorescence of HepG2 cells treated with or without 3-MA; scale bars, 50 µm. (<b>E</b>) Effect of GLSP on protein expression of Keap1, Nrf2, and HO-1 protein levels with or without autophagy inhibitor 3-MA. GAPDH was used as control. Data are expressed as mean ± S.E., and different letters indicate significant differences (<span class="html-italic">p</span> &lt; 0.05). Statistical differences were assessed by Tukey’s test of one-way ANOVA.</p>
Full article ">
16 pages, 7143 KiB  
Article
Novel Insight into the Composition Differences Between Buffalo and Holstein Milk and Potential Anti-Inflammation and Antioxidant Effect on Caco-2 Cells
by Luyao Wang, Xinyue Hu, Jiaqi Jiang, Dong Wang, Chaobin Qin, Ling Li, Deshun Shi, Qingyou Liu, Jian Wang, Hui Li, Jieping Huang and Zhipeng Li
Foods 2024, 13(23), 3915; https://doi.org/10.3390/foods13233915 - 4 Dec 2024
Viewed by 826
Abstract
Milk is one of the most common sources of nutrients in humans, however, the composition and healthy value of the milk derived from different animals are very different. Here, we systemically compared the protein and lipid profiles and evaluated the anti-inflammation and antioxidant [...] Read more.
Milk is one of the most common sources of nutrients in humans, however, the composition and healthy value of the milk derived from different animals are very different. Here, we systemically compared the protein and lipid profiles and evaluated the anti-inflammation and antioxidant effect of buffalo and Holstein-derived milk on Caco-2 cells. Results showed that 906 proteins and 1899 lipids were identified in the buffalo milk and Holstein milk samples including 161 significantly different proteins (DEPs) and 49 significantly different lipids. The DEPs were mainly enriched in defense response-related terms, while the differential lipids were mainly included in fat digestion and absorption and cholesterol metabolism pathways. In addition, the Caco-2 cells co-cultured with buffalo and Holstein milk components showed significant benefits in being resistant to LPS-induced inflammation stress and H2O2-induced ROS stress. The qRT-PCR and ELISA results showed that the expression of TNF-α, IL-1β, and IL-6 was significantly lower (p < 0.05) in the cells co-cultured with milk components. Further analysis showed that, after H2O2 treatment, the expression of keap1 and Nrf-2 in the Caco-2 cells co-cultured with milk components was significantly lower (p < 0.05). In addition, being co-cultured with milk components significantly decreased the SOD, MDA, CAT, and GSH-Px content (p < 0.05) in the Caco-2 cells induced by H2O2. This study provides a novel insight into the differences in proteins and lipids between buffalo milk and Holstein milk, and a reference understanding of the anti-inflammation and antioxidant effect of the consumption of milk on the intestines. Full article
(This article belongs to the Section Dairy)
Show Figures

Figure 1

Figure 1
<p>Proteome analysis of the buffalo and Holstein milk. (<b>A</b>) Principal coordinates (PCoA) analysis of data from different milk samples. (<b>B</b>) Identification of proteins in buffalo milk and Holstein milk. (<b>C</b>) Identification of differentially expressed proteins (DEPs) in the buffalo milk and Holstein milk. (<b>D</b>) Subcellular localization analysis of the DEPs. (<b>E</b>) GO analysis of the DEPs.</p>
Full article ">Figure 2
<p>Lipidome analysis of the buffalo and Holstein milk. (<b>A</b>) Correlation analysis of QC samples. (<b>B</b>) Analysis of total lipid in the milk samples. (<b>C</b>) Statistics of lipid subclasses and molecular numbers in the milk samples. (<b>D</b>) The proportion of lipid subclasses in different milk samples. (<b>E</b>) Differential analysis of the lipids. (<b>F</b>) Pathway enrichment analysis of the differential lipids.</p>
Full article ">Figure 3
<p>Anti-inflammatory effect of the buffalo and Holstein milk components on Caco-2 cells. (<b>A</b>) Caco-2 cells co-cultured with buffalo and Holstein milk components. (<b>B</b>) Caco-2 cells treated with LPS (after co-culturing with buffalo and Holstein milk components).</p>
Full article ">Figure 4
<p>qRT-PCR detection of TNF-α (<b>A</b>), IL-1β (<b>B</b>), and IL-6 (<b>C</b>) in the Caco-2 cells treated with LPS. ELISA detection of TNF-α (<b>D</b>), IL-1β (<b>E</b>), and IL-6 (<b>F</b>) in the Caco-2 cells treated with LPS. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Antioxidant effect of buffalo and Holstein milk components on Caco-2 cells. (<b>A</b>) Caco-2 cells were treated with H<sub>2</sub>O<sub>2</sub> (after co-cultured with buffalo and Holstein milk components). (<b>B</b>) qRT-PCR detection of Nrf-2 in Caco-2 cells treated with H<sub>2</sub>O<sub>2</sub>. (<b>C</b>) qRT-PCR detection of keap1 in Caco-2 cells treated with H<sub>2</sub>O<sub>2</sub>. (<b>D</b>) Flow cytometry detection of ROS in Caco-2 cells treated with H<sub>2</sub>O<sub>2</sub>. (<b>E</b>) SOD content in Caco-2 cells treated with H<sub>2</sub>O<sub>2</sub>. (<b>F</b>) MDA content in Caco-2 cells treated with H<sub>2</sub>O<sub>2</sub>. (<b>G</b>) CAT content in Caco-2 cells treated with H<sub>2</sub>O<sub>2</sub>. (<b>H</b>) GSH-Px content in Caco-2 cells treated with H<sub>2</sub>O<sub>2</sub>. * <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 ">
21 pages, 12975 KiB  
Article
Fecal Microbiota Transplantation Activity of Floccularia luteovirens Polysaccharides and Their Protective Effect on Cyclophosphamide-Induced Immunosuppression and Intestinal Injury in Mice
by He Ma, Abdul Mueed, Yanxu Ma, Muhammad Ibrahim, Ling Su and Qi Wang
Foods 2024, 13(23), 3881; https://doi.org/10.3390/foods13233881 - 30 Nov 2024
Viewed by 891
Abstract
Floccularia luteovirens polysaccharides (FLP1s) have potential biological activities. Our previous study showed that FLP1s positively regulated gut immunity and microbiota. However, it is still unclear whether FLP1s mediate gut microbiota in immunosuppressed mice. This research aims to explore the relationship between FLP1-mediated gut [...] Read more.
Floccularia luteovirens polysaccharides (FLP1s) have potential biological activities. Our previous study showed that FLP1s positively regulated gut immunity and microbiota. However, it is still unclear whether FLP1s mediate gut microbiota in immunosuppressed mice. This research aims to explore the relationship between FLP1-mediated gut microbes and intestinal immunity in immunosuppressed mice through fecal microbiota transplantation (FMT). The results demonstrated that FLP1s exhibited prebiotic and anti-immunosuppressive effects on CTX-induced immunosuppressed mice. FFLP1 treatment (microbiota transplantation from the fecal sample) remarkably elevated the production of sIgA and secretion of the anti-inflammatory cytokines IL-4, TNF-α, and IFN-γ in the intestine of CTX-treated mice, inducing activation of the MAPK pathway. Moreover, FFLP1s mitigated oxidative stress by activating the Nrf2/Keap1 signaling pathway and strengthened the intestinal barrier function by upregulating the expression level of tight junction proteins (occludin, claudin-1, MUC-2, and ZO-1). Furthermore, FFPL1s restored gut dysbiosis in CTX-treated immunosuppressed mice by increasing the abundance of Alloprevotella, Lachnospiraceae, and Bacteroides. They also modified the composition of fecal metabolites, leading to enhanced regulation of lipolysis in adipocytes, the cGMP-PKG pathway, the Rap1 signaling pathway, and ovarian steroidogenesis, as indicated by KEGG pathway analysis. These findings indicate that FLP1s could modulate the response of the intestinal immune system through regulation of the gut microbiota, thus promoting immune activation in CTX-treated immunosuppressed mice. FLP1s can serve as a natural protective agent against CTX-induced immune injury. Full article
Show Figures

Figure 1

Figure 1
<p>Animal experiments of different groups. (<b>A</b>) Donor groups, (<b>B</b>) ABX groups, and (<b>C</b>) FMT recipient groups.</p>
Full article ">Figure 2
<p>The influence of FLP1 gavage in ABX groups. (<b>A</b>) Body weight. (<b>B</b>) Thymus index. (<b>C</b>) Spleen index. (<b>D</b>) H&amp;E staining of colon sections for different groups (scale bar = 200 μm). Data are presented as mean ± SD (n = 6). Different letters over bars denote the statistical significance between two groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>The regulated secretion of immunomodulatory and antioxidant effects in ABX groups. (<b>A</b>–<b>E</b>) Small intestinal oxidative enzyme (T-AOC, MDA, CAT, SOD, and GSH-Px) levels in mice. (<b>F</b>–<b>J</b>) Small intestinal cytokine (sIgA, IFN-γ, TNF-α, IL-6, and IL-4) levels in mice. Data are presented as mean ± SD (n = 6). Different letters over bars denote the statistical significance between two groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>The influence of FFLP1 in immunosuppressed mice. (<b>A</b>) Body weight. (<b>B</b>) Thymus index. (<b>C</b>) Spleen index. (<b>D</b>) H&amp;E staining for colon tissues in different groups (scale bar = 200 μm). (<b>E</b>) Bands of occludin, claudin, ZO-1, and MUC-2 expression in the intestinal mucosa, with β-actin used as a standard. Data are presented as mean ± SD (n = 6). Different letters over bars denote the statistical significance between two groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>The regulated secretion of immunomodulatory and antioxidant effects in FMT groups. (<b>A</b>–<b>E</b>) Small intestinal oxidative enzyme (T-AOC, CAT, SOD, GSH-Px, and MDA) levels in mice. (<b>F</b>–<b>J</b>) Small intestinal cytokine (sIgA, IL-6, IL-4, IFN-γ, and TNF-α) levels in mice. (<b>K</b>) Bands of Nrf2 and Keap1 expression, with β-actin used as a standard. (<b>L</b>) Bands of p-38, p-p38, ERK, p-ERK, JNK, and p-JNK in intestinal mucosa, with β-actin used as a standard. Data are presented as mean ± SD (n = 6). Different letters over bars denote the statistical significance between two groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Regulation of gut microbiota of immunosuppressed mice by FMT treatment. (<b>A</b>) ACE index, (<b>B</b>) Chao1 index, and (<b>C</b>) rank–abundance curves for all samples. (<b>D</b>) PCoA analysis and (<b>E</b>) PCA analysis of each sample’s microbial composition. Community bar plot at the (<b>F</b>) phylum level and (<b>G</b>) family level. Data are presented with n = 5.</p>
Full article ">Figure 7
<p>FMT treatment modulated the composition of gut microbiota in immunosuppressed mice. (<b>A</b>) Histogram of the LDA scores. (<b>B</b>) Bar chart of LEfSe analysis. Data are presented with n = 5.</p>
Full article ">Figure 8
<p>FMT treatment alters the level of fecal metabolites in Cy-treated immunosuppressed mice. (<b>A</b>) Score plot of the PCA model. (<b>B</b>) Score plot of the PSL-DA model. (<b>C</b>) Permutations test of the PSL-DA model. (<b>D</b>) Volcano plot. Data are presented with n = 5.</p>
Full article ">Figure 9
<p>(<b>A</b>) Heatmap of different metabolites in FFLP1 and FMC groups. (<b>B</b>) Metabolomic pathway enrichment analysis. (<b>C</b>) Metabolic pathway analysis of cyclic AMP; red indicates an increase in metabolite levels, blue indicates a decrease, solid lines represent direct interactions, dashed lines represent indirect interactions, single arrows indicate unidirectional regulation, and double arrows indicate bidirectional regulation. Data are presented with n = 5.</p>
Full article ">Figure 10
<p>Correlation between different gut microbiota, gut metabolites, and biomarkers. (<b>A</b>) Correlation between different gut microbiota and biomarkers related to immunity. (<b>B</b>) Correlation between different gut metabolites and biomarkers related to immunity. (<b>C</b>) Correlation between different gut microbiota and metabolites. Spearman analysis was used for the matrix (* <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 ">
12 pages, 3585 KiB  
Article
Melatonin Improves H2O2-Induced Oxidative Stress in Sertoli Cells Through Nrf2-Keap1 Signaling Pathway
by Ying Tang, Ziming Wang, Yanru Chen, Junying Wang, Hongzhan Wang, Bo Li, Bojing Liu and Peng Zheng
Genes 2024, 15(12), 1544; https://doi.org/10.3390/genes15121544 - 28 Nov 2024
Viewed by 520
Abstract
Background: Oxidative stress in the testicles of male livestock can cause reduced fertility. Melatonin is a natural product with antioxidant effects, but its specific antioxidant mechanism is still unclear. This study used calf testicular Sertoli cells as materials to explore the mechanism [...] Read more.
Background: Oxidative stress in the testicles of male livestock can cause reduced fertility. Melatonin is a natural product with antioxidant effects, but its specific antioxidant mechanism is still unclear. This study used calf testicular Sertoli cells as materials to explore the mechanism by which melatonin alleviates the oxidative stress of Sertoli cells, laying a foundation for improving the fertility of bulls. Methods: The optimal treatment concentrations of H2O2 and melatonin (MLT) were screened out using a CCK8 kit and MDA kit. Then, the cells were divided into four groups for treatment: control group, H2O2 treatment group, MLT treatment group, and H2O2 and MLT co-treatment group, then the MDA, ROS, GSH, and SOD contents were detected. Real-time quantitative PCR analysis and Western blot analysis were used to detect genes and proteins related to the Nrf2-Keap1 pathway. Immunofluorescence staining was used to analyze changes in Nrf2. Results: Research results show that the MDA content of cells in the group treated with H2O2 and MLT combined was significantly lower than that in the group treated with H2O2 alone, but there was no difference from the control group. Compared with the control group, the ROS level of cells in the H2O2-treated group significantly increased, and the content of GSH and SOD significantly decreased. Compared with the H2O2-treated group, the ROS level of cells in the H2O2 and MLT co-treated group significantly decreased, and the content of GSH and SOD increased significantly, but no difference from the control group. Similarly, MTL can alleviate the changes in cellular Nrf2, Keap1, HO-1, and NQO1 expression caused by H2O2. Conclusions: Melatonin activates the Nrf2-Keap1 signaling pathway in Sertoli cells, elevating the expression of HO-1 and NQO1, and thereby exerting its antioxidant capabilities. Full article
(This article belongs to the Special Issue Genetic Regulation Mechanisms of Animal Reproduction)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effects of different concentrations of H<sub>2</sub>O<sub>2</sub> and MLT on cell viability and MDA. (<b>A</b>) Effect of H<sub>2</sub>O<sub>2</sub> on cell viability; (<b>B</b>) Effect of H<sub>2</sub>O<sub>2</sub> on the content of MDA in cells; (<b>C</b>) Effects of MLT on cell viability; (<b>D</b>) Effect of MLT on the content of MDA in cells; (<b>E</b>) Changes in cell viability; (<b>F</b>) Changes in MDA content. Control group, H<sub>2</sub>O<sub>2</sub> group (300 µmol/L H<sub>2</sub>O<sub>2</sub>), MLT group (1 µmol/L MLT), H<sub>2</sub>O<sub>2</sub> + MLT group (300 µmol/L H<sub>2</sub>O<sub>2</sub> + 1 µmol/L MLT). Different letters on the shoulder mark indicate significant differences (<span class="html-italic">p</span> &lt; 0.05); Markings with the same letter on the shoulder mark indicate that the difference is not significant (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 2
<p>Effects of different concentrations of H<sub>2</sub>O<sub>2</sub> and MLT on ROS, GSH, and SOD. (<b>A</b>–<b>D</b>) ROS staining results, the arrow indicates positive cells; (<b>A</b>) Control group; (<b>B</b>) H<sub>2</sub>O<sub>2</sub> group; (<b>C</b>) MLT group; (<b>D</b>) H<sub>2</sub>O<sub>2</sub> + MLT group. (<b>E</b>) ROS results analysis; (<b>F</b>) Changes in GSH activity; (<b>G</b>) Changes in SOD activity. Different letters on the shoulder mark indicate significant differences (<span class="html-italic">p</span> &lt; 0.05); Markings with the same letter on the shoulder mark indicate that the difference is not significant (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 3
<p>The level of expression of Keap1, HO-1, and NQO1 gene mRNA. (<b>A</b>–<b>C</b>) effects on Keap1, HO-1 and NQO1 mRNA expression in SCs. (<b>D</b>–<b>F</b>) effects on Keap1, HO-1 and NQO1 protein expression in SCs. Different letters on the shoulder mark indicate significant differences (<span class="html-italic">p</span> &lt; 0.05); Markings with the same letter on the shoulder mark indicate that the difference is not significant (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 4
<p>Effect of melatonin on nuclear translocation of Nrf2 protein in calf Sertoli cells under oxidative stress. Different letters on the shoulder mark indicate significant differences (<span class="html-italic">p</span> &lt; 0.05); Markings with the same letter on the shoulder mark indicate that the difference is not significant (<span class="html-italic">p</span> &gt;0.05).</p>
Full article ">Figure 5
<p>Effects of melatonin on Nrf2 protein expression. (<b>A</b>) Control group, (<b>B</b>) H<sub>2</sub>O<sub>2</sub> group, (<b>C</b>) MLT + H<sub>2</sub>O<sub>2</sub> group.</p>
Full article ">
30 pages, 16811 KiB  
Article
Unveiling Smyd-2’s Role in Cytoplasmic Nrf-2 Sequestration and Ferroptosis Induction in Hippocampal Neurons After Cerebral Ischemia/Reperfusion
by Daohang Liu and Yizhun Zhu
Cells 2024, 13(23), 1969; https://doi.org/10.3390/cells13231969 - 28 Nov 2024
Viewed by 736
Abstract
SET and MYND Domain-Containing 2 (Smyd-2), a specific protein lysine methyltransferase (PKMT), influences both histones and non-histones. Its role in cerebral ischemia/reperfusion (CIR), particularly in ferroptosis—a regulated form of cell death driven by lipid peroxidation—remains poorly understood. This study identifies the expression of [...] Read more.
SET and MYND Domain-Containing 2 (Smyd-2), a specific protein lysine methyltransferase (PKMT), influences both histones and non-histones. Its role in cerebral ischemia/reperfusion (CIR), particularly in ferroptosis—a regulated form of cell death driven by lipid peroxidation—remains poorly understood. This study identifies the expression of Smyd-2 in the brain and investigates its relationship with neuronal programmed cell death (PCD). We specifically investigated how Smyd-2 regulates ferroptosis in CIR through its interaction with the Nuclear Factor Erythroid-2-related Factor-2 (Nrf-2)/Kelch-like ECH-associated protein (Keap-1) pathway. Smyd-2 knockout protects HT-22 cells from Erastin-induced ferroptosis but not TNF-α + Smac-mimetic-induced apoptosis/necroptosis. This neuroprotective effect of Smyd-2 knockout in HT-22 cells after Oxygen–Glucose Deprivation/Reperfusion (OGD/R) was reversed by Erastin. Smyd-2 knockout in HT-22 cells shows neuroprotection primarily via the Nuclear Factor Erythroid-2-related Factor-2 (Nrf-2)/Kelch-like ECH-associated protein (Keap-1) pathway, despite the concurrent upregulation of Smyd-2 and Nrf-2 observed in both the middle cerebral artery occlusion (MCAO) and OGD/R models. Interestingly, vivo experiments demonstrated that Smyd-2 knockout significantly reduced ferroptosis and lipid peroxidation in hippocampal neurons following CIR. Moreover, the Nrf-2 inhibitor ML-385 abolished the neuroprotective effects of Smyd-2 knockout, confirming the pivotal role of Nrf-2 in ferroptosis regulation. Cycloheximide (CHX) fails to reduce Nrf-2 expression in Smyd-2 knockout HT-22 cells. Smyd-2 knockout suppresses Nrf-2 lysine methylation, thereby promoting the Nrf-2/Keap-1 pathway without affecting the PKC-δ/Nrf-2 pathway. Conversely, Smyd-2 overexpression disrupts Nrf-2 nuclear translocation, exacerbating ferroptosis and oxidative stress, highlighting its dual regulatory role. This study underscores Smyd-2’s potential for ischemic stroke treatment by disrupting the Smyd-2/Nrf-2-driven antioxidant capacity, leading to hippocampal neuronal ferroptosis. By clarifying the intricate interplay between ferroptosis and oxidative stress via the Nrf-2/Keap-1 pathway, our findings provide new insights into the molecular mechanisms of CIR and identify Smyd-2 as a promising therapeutic target. Full article
Show Figures

Figure 1

Figure 1
<p>MCAO and OGD/R conduce to Smyd-2 activation in the hippocampus and HT-22 cells. (<b>A</b>). Representative images of TTC staining, quantitative analysis of infarct volume, asymmetrical test scores, and the adhesive removal scores in MCAO mice (<span class="html-italic">n</span> = 5). (<b>B</b>). Smyd-2 expression in mouse brain after CIR (<span class="html-italic">n</span> = 5). (<b>C</b>). Smyd-2 expression in Ht-22 cells challenged with OGD/R (<span class="html-italic">n</span> = 5). * <span class="html-italic">p</span> &lt; 0.05 vs. sham group, *** <span class="html-italic">p</span> &lt; 0.001 vs. sham group; *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group.</p>
Full article ">Figure 2
<p>The inhibition of Smyd-2 expression delays the progression of CIR impairment. (<b>A</b>–<b>C</b>) Effects of Smyd-2 on cerebral infarction volume and neurobehavioral function in MCAO mice (<span class="html-italic">n</span> = 5). (<b>D</b>) Representative images of FJB staining slices of the hippocampus and cortex; scale bars = 40 μm (<span class="html-italic">n</span> = 5). (<b>E</b>) Representative images of LFB staining slices of the hippocampus and cortex and quantitative analysis of the breakdown products of myelin sheathes in the hippocampus and cortex; scale bars = 40 μm (<span class="html-italic">n</span> = 5). (<b>F</b>) The transfection effects of Smyd-2-overexpressing adenovirus and siRNA in Ht-22 cells (<span class="html-italic">n</span> = 5). (<b>G</b>) The neuronal viability, LDH release, SOD level, and MDA level of Ht-22 cells challenged with Smyd-2 siRNA and Smyd-2-overexpressing adenovirus after OGD/R (<span class="html-italic">n</span> = 5). ** <span class="html-italic">p</span> &lt; 0.01 vs. sham group, *** <span class="html-italic">p</span> &lt; 0.001 vs. sham group; ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001 vs. CIR group and CIR + IVC-NC group; *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + Si-NC group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + AD-NC group.</p>
Full article ">Figure 3
<p>Smyd-2 knockout counteracts the effect of OGD/R on lipid peroxidation in Ht-22 cells. (<b>A</b>) BODIPY-581/591-C11 staining was applied to analyze and quantify the effect of Smyd-2 siRNA and adenovirus-mediated Smyd-2 on HT-22 cell ferroptosis challenged with OGD/R. The representative images were obtained with an optical microscope at 400× magnification; scale bars = 50 μm. The GSH level of HT-22 cells challenged with AD-Smyd-2 and Si-Smyd-2 after OGD/R. (<b>B</b>) DCFH-DA staining was applied to analyze and quantify the effect of Smyd-2 siRNA and adenovirus-mediated Smyd-2 on HT-22 cell ferroptosis challenged with OGD/R. The representative images were obtained with a confocal microscope at 200× magnification; scale bars = 100 μm (<span class="html-italic">n</span> = 6). (<b>C</b>) Immunofluorescence method was applied to investigate Smyd-2 and GPX-4 protein expression and localization in HT-22 cells and their relation to neuronal ferroptosis after OGD/R. The representative images were obtained with a confocal microscope at 400× magnification; scale bars = 50 μm. *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + Si-NC group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + AD-NC group.</p>
Full article ">Figure 4
<p>Smyd-2 regulates the abnormality in neuronal ferroptosis caused by CIR. (<b>A</b>) Representative images of Perls staining slices of the hippocampus and cortex. Scale bars = 40 μm (<span class="html-italic">n</span> = 5). (<b>B</b>–<b>D</b>) Representative images of Western blot and quantitative analysis of the expression of Smyd-2, GPX-4, FTH-1, SLC7A11, ACSL-4, 15-LOX, COX-2, NQO-1, Keap-1, HO-1, nucleus Nrf-2, <span class="html-italic">p</span>-Nrf-2, and Nrf-2 (TP) in the hippocampus. *** <span class="html-italic">p</span> &lt; 0.001 vs. sham group; ### <span class="html-italic">p</span> &lt; 0.001 vs. CIR group and CIR + ICV-NC group.</p>
Full article ">Figure 5
<p>The effect of Smyd-2 overexpression on programmed cell death of HT-22 cells induced by Erastin and Smac mimetic + TNF-α. (<b>A</b>) The neuronal viability and LDH release of HT-22 cells challenged with Erastin. (<b>B</b>,<b>C</b>) The neuronal viability and LDH release of HT-22 cells challenged with Erastin. (c,d,e,f,g,h,i,j) The neuronal viability, LDH release, SOD level, and MDA level of Si-Smyd-2 HT-22 cells challenged with Erastin and Smac mimetic (<span class="html-italic">n</span> = 6). (<b>D</b>) Annexin V/PI staining was applied to observe and analyze the effect of Erastin and Smac mimetic on different types of programmed cell death in HT-22 cells induced by OGD/R. The representative images were obtained with an optical microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 6). *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group, * <span class="html-italic">p</span> &lt; 0.05 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + Si-NC group, # <span class="html-italic">p</span> &lt; 0.05 vs. OGD/R + Si-NC group.</p>
Full article ">Figure 6
<p>The effect of Si-Smyd-2 combined with Erastin on the ferroptosis of HT-22 cells induced by OGD/R. (<b>A</b>) The neuronal viability, LDH release, SOD, and MDA of HT-22 cells challenged with Si-Smyd-2 and Erastin after OGD/R. (<b>B</b>) GSH kit was applied to analyze and quantify the effect of the Si-RNA-mediated Smyd-2 and Erastin on HT-22 cell ferroptosis challenged with OGD/R (<span class="html-italic">n</span> = 5). (<b>C</b>) Pathophysiological and physiological morphologies of mitochondria in each HT-22 cell group were observed by transmission electron microscopy. The red arrows mark the increased electron density of the matrix and fractured and vague cristae. The blue arrows mark vacuoles in mitochondria. The enlarged region bounded by a rectangular dotted box conduces to obtaining a more detailed view of the mitochondria for each experimental condition. The representative images were obtained with an optical microscope at 8k× magnification; scale bars = 1 μm. The representative enlarged images were obtained with an optical microscope at 20k× magnification; scale bars = 500 nm (<span class="html-italic">n</span> = 5). (<b>D</b>–<b>F</b>) Representative Western blots and quantitative evaluation of Smyd-2, SLC7A11, ACSL-4, FTH-1, GPX-4, Nrf-2, Keap-1, p-Nrf-2, and nucleus Nrf-2 expression levels in each HT-22 cell group. Data normalized to the loading control GAPDH are expressed as % of control (<span class="html-italic">n</span> = 5). *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + Si-NC group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R + Si-Smyd-2 group.</p>
Full article ">Figure 7
<p>Effects of Smyd-2 (KO) combined with Nrf-2 inhibitor ML-385 on ferroptosis and lipid peroxidation in MCAO mice and OGD/R-induced HT-22 cells. (<b>A</b>) The neuronal viability, LDH release, SOD, and MDA of HT-22 cells were challenged with Si-Smyd-2 and ML-385 after OGD/R (<span class="html-italic">n</span> = 5). (<b>B</b>) BODIPY-581/591-C11 staining, GSH kit, and DCFH-DA staining were used to analyze and quantify the effect of Si-RNA-mediated Smyd-2 and ML-385 on lipid peroxidation of HT-22 cells after OGD/R. The representative images of BODIPY-581/591-C11 staining were obtained with an optical microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 5). The representative images of DCFH-DA staining were obtained with an optical microscope at 200× magnification; scale bars = 100 μm (<span class="html-italic">n</span> = 5). (<b>C</b>) Annexin V/PI double fluorescence staining was used to study the effect of Si-RNA-mediated Smyd-2 and ML-385 on a different form of programmed cell death in HT-22 cells induced by OGD/R. The representative images were obtained with a confocal microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 5). (<b>D</b>) The effect of Si-RNA-mediated Smyd-2 combined with ML-385 on ferroptosis-related proteins in HT-22 cells induced by OGD/R. Representative Western blots and quantitative evaluation of ACSL-4, Keap-1, SLC7A11, Smyd-2, FTH-1, GPX-4, p-Nrf-2, Nrf-2 (TP), nucleus Nrf-2, PGC-1α, COX-2, 15-LOX, NQO-1, and HO-1 expression levels in each HT-22 cell group. Data normalized to the loading control GAPDH are expressed as % of control (<span class="html-italic">n</span> = 5). (<b>E</b>) Pathophysiological and physiological morphologies of mitochondria in each HT-22 cell group were observed by TEM. The red arrows mark the increased electron density of the matrix and fractured and vague cristae. The blue arrows mark vacuoles in mitochondria. The zoom region bounded by a rectangular dotted box allows a more detailed view of mitochondria for each experimental condition. The representative images were obtained with an optical microscope at 8k× magnification; scale bars = 1 μm (<span class="html-italic">n</span> = 5). The representative enlarged images were obtained with an optical microscope at 20k× magnification; scale bars = 500 nm. *** <span class="html-italic">p</span> &lt; 0.001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.001 vs. OGD/R group and Si-NC + OGD/R group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.001 vs. Si-Smyd-2 + OGD/R group.</p>
Full article ">Figure 7 Cont.
<p>Effects of Smyd-2 (KO) combined with Nrf-2 inhibitor ML-385 on ferroptosis and lipid peroxidation in MCAO mice and OGD/R-induced HT-22 cells. (<b>A</b>) The neuronal viability, LDH release, SOD, and MDA of HT-22 cells were challenged with Si-Smyd-2 and ML-385 after OGD/R (<span class="html-italic">n</span> = 5). (<b>B</b>) BODIPY-581/591-C11 staining, GSH kit, and DCFH-DA staining were used to analyze and quantify the effect of Si-RNA-mediated Smyd-2 and ML-385 on lipid peroxidation of HT-22 cells after OGD/R. The representative images of BODIPY-581/591-C11 staining were obtained with an optical microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 5). The representative images of DCFH-DA staining were obtained with an optical microscope at 200× magnification; scale bars = 100 μm (<span class="html-italic">n</span> = 5). (<b>C</b>) Annexin V/PI double fluorescence staining was used to study the effect of Si-RNA-mediated Smyd-2 and ML-385 on a different form of programmed cell death in HT-22 cells induced by OGD/R. The representative images were obtained with a confocal microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 5). (<b>D</b>) The effect of Si-RNA-mediated Smyd-2 combined with ML-385 on ferroptosis-related proteins in HT-22 cells induced by OGD/R. Representative Western blots and quantitative evaluation of ACSL-4, Keap-1, SLC7A11, Smyd-2, FTH-1, GPX-4, p-Nrf-2, Nrf-2 (TP), nucleus Nrf-2, PGC-1α, COX-2, 15-LOX, NQO-1, and HO-1 expression levels in each HT-22 cell group. Data normalized to the loading control GAPDH are expressed as % of control (<span class="html-italic">n</span> = 5). (<b>E</b>) Pathophysiological and physiological morphologies of mitochondria in each HT-22 cell group were observed by TEM. The red arrows mark the increased electron density of the matrix and fractured and vague cristae. The blue arrows mark vacuoles in mitochondria. The zoom region bounded by a rectangular dotted box allows a more detailed view of mitochondria for each experimental condition. The representative images were obtained with an optical microscope at 8k× magnification; scale bars = 1 μm (<span class="html-italic">n</span> = 5). The representative enlarged images were obtained with an optical microscope at 20k× magnification; scale bars = 500 nm. *** <span class="html-italic">p</span> &lt; 0.001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.001 vs. OGD/R group and Si-NC + OGD/R group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.001 vs. Si-Smyd-2 + OGD/R group.</p>
Full article ">Figure 8
<p>Smyd-2 methylates Nrf-2 (Lys-508) to inhibit OGD/R-induced Nrf-2 (Ser-40) phosphorylation and nuclear translocation. (<b>A</b>) Representative Western blot and quantitative evaluation of p-Nrf-2, Nrf-2 (TP), and nucleus Nrf-2 expression levels in each HT-22 cell group. Data normalized to the loading control GAPDH are expressed as % of control. Data normalized to the loading control GAPDH and histone H-3 are expressed as % of control (<span class="html-italic">n</span> = 5). (<b>B</b>) Confocal images of the localization with immunofluorescence-stained Smyd-2 (green) and immunofluorescence-stained Nrf-2 (red) in various HT-22 cell groups after OGD/R. The zoom region bounded by rectangular boxes represents Smyd-2-Nrf-2 binding in the cytoplasm of HT-22 cells, and the Nrf-2 was transported to the cell nucleus. Scale bars = 10 μm (<span class="html-italic">n</span> = 5). (<b>C</b>) Representative Western blots and quantitative evaluation of Nrf-2 (TP), p-Nrf-2, nucleus Nrf-2, PKC-δ, p-PKC-δ, Smyd-2, and Keap-1 expression levels in each HT-22 cell group. Data normalized to the loading control histone H-3 are expressed as % of GAPDH (<span class="html-italic">n</span> = 5). (<b>D</b>) Quantitative analysis of the expression of the methylation level of Nrf-2 (<span class="html-italic">n</span> = 5). (<b>E</b>) The possible docking sites of two target proteins, Smyd-2/Nrf-2. The binding mode of the complex Nrf-2 with Smyd-2. *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R + Si-NC group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group; ^^^ <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R + CHX group. *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + AD-Smyd-2 group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R + AD-Smyd-2 group.</p>
Full article ">
15 pages, 11994 KiB  
Article
Oxidative Stress and Keap1-Nrf2 Pathway Involvement in Bisphenol A-Induced Liver Damage in Rats
by Juan Tang, Kai Wang, Dan Shen and Chunmei Li
Toxics 2024, 12(12), 864; https://doi.org/10.3390/toxics12120864 - 28 Nov 2024
Viewed by 510
Abstract
Bisphenol A (BPA), extensively utilized in the manufacture of epoxy resins and polycarbonate plastics, is prevalent in the environment. Its exposure has been associated with an increased risk of hepatic lesions; however, the underlying mechanisms and the spectrum of its effects remain poorly [...] Read more.
Bisphenol A (BPA), extensively utilized in the manufacture of epoxy resins and polycarbonate plastics, is prevalent in the environment. Its exposure has been associated with an increased risk of hepatic lesions; however, the underlying mechanisms and the spectrum of its effects remain poorly understood. This study investigates the role of the Keap1-Nrf2 signaling pathway in regulating BPA-induced hepatotoxicity in vivo using a rat model. Over a 30-day period, rats were orally administered either corn oil or BPA (0.5, 5, and 50 mg/kg). Changes in hepatic and kidney histology were assessed via transmission electron microscopy and HE staining. Oxidative stress levels in the liver tissue and serum were quantified, while the mRNA expression of Nrf2, Keap1, GPX2, HO-1, and caspase-3 was evaluated using qRT-PCR. Additionally, the expression of Nrf2 and cleaved caspase-3 in the liver tissue was measured through immunohistochemistry and Western blotting. Results indicated that BPA exposure significantly reduced the liver and adrenal coefficients in the treated rats compared to controls. Notable histomorphological alterations were observed in the liver and kidney tissues of the BPA-treated rats. The serum levels of GOT and TNF-α were significantly elevated in the BPA group relative to the controls. Evidence of oxidative stress was supported by increased malondialdehyde levels and decreased total superoxide dismutase activity in the liver and kidney, alongside a reduction in glutathione peroxidase activity in the liver tissue. Furthermore, BPA exposure enhanced the mRNA expression levels of Nrf2, Keap1, GPX2, HO-1, and caspase-3 in the liver tissue. Concurrently, Nrf2 and cleaved caspase-3 expression levels were elevated in the BPA-treated group compared to the controls. These findings suggest that BPA may contribute to metabolic disorders of liver function and poses a hepatotoxicity risk. Moreover, the activation of the Keap1-Nrf2 pathway may offer protective effects against hepatotoxicity, with potential implications for human liver disease. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Histopathological changes in the liver and kidney tissues of male rats following oral BPA administration through hematoxylin and eosin staining at a 200× magnification. (<b>A</b>) A normal liver section. (<b>B</b>–<b>D</b>) BPA-treated groups (0.5, 5, and 50 mg/kg). (<b>E</b>) A normal kidney section. (<b>F</b>–<b>H</b>) BPA-treated groups (0.5, 5, and 50 mg/kg). (<b>I</b>,<b>J</b>) The hepatic and renal damages histologic score evaluation compared to the control; ** <span class="html-italic">p</span> &lt; 0.01. Electron micrographs of liver tissues from control and BPA-treated rats. (<b>a</b>,<b>e</b>) A normal liver section. (<b>b</b>–<b>d</b>,<b>f</b>–<b>h</b>) BPA-treated groups (0.5, 5, and 50 mg/kg).</p>
Full article ">Figure 2
<p>Effect of BPA treatment on GPT (<b>A</b>), GOT (<b>B</b>) enzyme activities, IL-1β (<b>C</b>), IL-6 (<b>D</b>), and TNF-α (<b>E</b>) levels in serum. Level of IL-1β (<b>F</b>), IL-6 (<b>G</b>), and TNF-α (<b>H</b>) in liver tissue. Data shown are mean ± SD of eight animals in each group compared to control; * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Immunohistochemical analysis of Nrf2 and cleaved caspase-3 protein in liver. Nrf2: (<b>A</b>) Negative control. (<b>B</b>) Normal liver section. (<b>C</b>–<b>E</b>) BPA-treated groups (0.5, 5, and 50 mg/kg). (<b>F</b>) Positive cells (%). Cleaved caspase-3: (<b>a</b>) Negative control. (<b>b</b>) Normal liver section. (<b>c</b>–<b>e</b>) BPA-treated groups (0.5, 5, and 50 mg/kg). (<b>f</b>) Positive cells (%). Data shown are mean ± SD of eight animals in each group compared to control; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span>&lt; 0.01.</p>
Full article ">Figure 4
<p>Protein expression of Nrf2 and cleaved caspase-3 in liver samples of rats. Western blot analysis of Nrf2 and cleaved caspase-3 protein in liver (<b>A</b>–<b>C</b>). RT-PCR analyses of Nrf2, Keap1, GPX2, HO-1, and caspase-3 mRNA of liver (<b>D</b>). RT-PCR analyses of IL-1, IL-6, NF-κB, TNF-α, and MAPK3 mRNA of liver (<b>E</b>). RT-PCR analyses of PPARα, SCD1, SREBP1c, DGAT, and FAS (<b>F</b>). Data shown are mean ± SEM of eight animals in each group compared to control; * <span class="html-italic">p</span> &lt; 0.05, and ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
Back to TopTop