[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (9,043)

Search Parameters:
Keywords = amelioration

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 6224 KiB  
Article
Polysaccharides from Trametes versicolor as a Potential Prebiotic to Improve the Gut Microbiota in High-Fat Diet Mice
by Ming Bai, Zhenfeng Huang, Xiaoya Zheng, Mingyong Hou and Song Zhang
Microorganisms 2024, 12(8), 1654; https://doi.org/10.3390/microorganisms12081654 (registering DOI) - 13 Aug 2024
Abstract
Polysaccharides derived from Trametes versicolor have been found to exhibit hypolipidemic activity in hyperlipidemic mice, but the mechanism by which they modulate intestinal flora is still unclear. Currently, this study aimed to investigate the regulatory effects of extracellular (EPTV) and intracellular polysaccharides from [...] Read more.
Polysaccharides derived from Trametes versicolor have been found to exhibit hypolipidemic activity in hyperlipidemic mice, but the mechanism by which they modulate intestinal flora is still unclear. Currently, this study aimed to investigate the regulatory effects of extracellular (EPTV) and intracellular polysaccharides from T. versicolor (IPTV) on the dysbiosis of intestinal flora in mice fed a high-fat diet (HFD). The results showed that the oral administration of T. versicolor polysaccharides significantly ameliorated lipid accumulation and steatosis in hepatocytes. The gut dysbiosis in the HFD mice was characterized by a decrease in abundance and diversity of bacteria and an increase in the Firmicutes/Bacteroidetes ratio. However, T. versicolor polysaccharides attenuated these changes and reduced the relative abundance of bile-salt-hydrolase (BSH)-producing bacteria, such as Bacillus, Enterococcus, Bifidobacterium, and Lactococcus. It is noteworthy that T. versicolor polysaccharides also restored the disorganization of intestinal fungi in HFD mice, with EPTV treatment leading to a higher relative abundance of Basidiomycota and Ascomycota compared to IPTV. Additionally, T. versicolor polysaccharides enhanced the growth of butyrate-producing bacteria via the buk and but pathways, accompanied by an increase in short-chain fatty acids (SCFAs), especially butyrate. IPTV also increased the expression of G-protein-coupled receptors 41 (GPR41) and 43 (GPR43) by 40.52% and 113.24% each, as compared to 62.42% and 110.28%, respectively, for EPTV. It is suggested that IPTV and EPTV have the potential to counteract hyperlipidemia-associated intestinal flora disorders and improve lipid metabolism. Full article
(This article belongs to the Section Gut Microbiota)
Show Figures

Figure 1

Figure 1
<p>The procedure of animal experiment.</p>
Full article ">Figure 2
<p>Effects of PTVs on lipid profiles of high-fat-diet-fed mice. (<b>A</b>) Serum lipid profiles including total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C). Data are presented as means ± SD, <span class="html-italic">n</span> = 10. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, compared with NC group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01, compared with HC group. (<b>B</b>) HE staining of liver was performed to observe the morphology of liver (×400). Yellow arrow indicates fat pathological changes.</p>
Full article ">Figure 3
<p>PTVs restore the high-fat-diet-induced gut bacterial dysbiosis at different taxonomic levels in HFD-fed mice. (<b>A</b>) The relative abundance of gut microbes at phylum levels; (<b>B</b>) the ratio of Firmicutes–Bacteroidetes at phylum levels; (<b>C</b>) heat map summarizing the relative abundance of gut microbes at genus levels; (<b>D</b>) evolutionary clustering analysis of bacterial community structure; (<b>E</b>) PCoA analysis of bacterial community structure; (<b>F</b>) NMDS analysis of bacterial community structure. Data are presented as means ± SD, <span class="html-italic">n</span> = 6. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, compared with NC group; ** <span class="html-italic">p</span> &lt; 0.01, compared with HC group.</p>
Full article ">Figure 4
<p>Dynamics of the intestinal microorganism in HFD-fed mice. (<b>A</b>) The cladogram of intestinal bacteria with significant differences between NC, HC, IPTV (200 mg/kg), and EPTV (200 mg/kg); (<b>B</b>) the cladogram of intestinal fungi with significant differences between NC, HC, IPTV (200 mg/kg), and EPTV (200 mg/kg); (<b>C</b>) bacterium–bacterium co-occurrence network; (<b>D</b>) fungus–fungus co-occurrence network.</p>
Full article ">Figure 5
<p>Effects of PTVs on specific gut microbial population at genus and species levels in HFD-fed mice. (<b>A</b>) The relative abundance of gut BSH-producing bacteria; (<b>B</b>) the relative abundance of gut SCFA-producing bacteria. Data are presented as means ± SD, <span class="html-italic">n</span> = 6. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, compared with NC group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01, compared with HC group.</p>
Full article ">Figure 6
<p>PTVs restore the high-fat-diet-induced gut fungal dysbiosis at different taxonomic levels in HFD-fed mice. (<b>A</b>) The relative abundance of gut fungal microbes at phylum levels; (<b>B</b>) the relative abundance of gut fungal microbes at family levels; (<b>C</b>) heat map summarizing the relative abundance of gut fungal microbes at genus levels; (<b>D</b>) evolutionary clustering analysis of fungal microbe community structure; (<b>E</b>) PCoA analysis of fungal microbe community structure; (<b>F</b>) NMDS analysis of fungal microbe community structure. Data are presented as means ± SD, <span class="html-italic">n</span> = 6.</p>
Full article ">Figure 7
<p>Prediction of functional pathway using PICRUSt analysis. (<b>A</b>) Total KEGG pathways changed in all groups. (<b>B</b>) COG pathways changed in all groups.</p>
Full article ">Figure 8
<p>PTVs upregulates butyrate production by <span class="html-italic">buk</span> and <span class="html-italic">but</span> genes accompanied by an increase in SCFA receptor expression in HFD-fed mice. (<b>A</b>) GC–MS chromatograms of SCFA in feces. (<b>B</b>) SCFA concentration in feces measured by GC–MS. (<b>C</b>) Predominant butyrate-producing genes: relative abundance of butyrate kinase (<span class="html-italic">buk</span>) and butyryl-CoA (<span class="html-italic">but</span>) bacterial genes in feces. (<b>D</b>) Protein expression of SCFA receptors (GPR41, GPR43) in colon. Data are presented as means ± SD, <span class="html-italic">n</span> = 5. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, compared with NC group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01, compared with HC group.</p>
Full article ">Figure 9
<p>Spearman’s correlations between the intestinal microbe and lipid metabolic parameters. (<b>A</b>) Heat map describing Spearman’s correlations between the intestinal bacterial community and lipid metabolic parameters. (<b>B</b>) Heat map describing Spearman’s correlations between the intestinal fungal community and lipid metabolic parameters. (<b>C</b>) Heat map describing Spearman’s correlations between the intestinal bacterial and fungal communities. * <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 ">
13 pages, 3371 KiB  
Article
Safflower Yellow Injection Alleviates Myocardial Ischemia/Reperfusion Injury by Reducing Oxidative and Endoplasmic Reticulum Stress
by Wulin Liang, Mingqian Zhang, Jiahui Gao, Rikang Huang, Lu Cheng, Liyuan Zhang, Zhishan Huang, Zhanhong Jia and Shuofeng Zhang
Pharmaceuticals 2024, 17(8), 1058; https://doi.org/10.3390/ph17081058 - 12 Aug 2024
Abstract
Safflower yellow is an extract of the famous Chinese medicine Carthamus tinctorious L, and safflower yellow injection (SYI) is widely used clinically to treat angina pectoris. However, there are few studies on the anti-myocardial ischemia/reperfusion (I/R) injury effect of SYI, and its mechanisms [...] Read more.
Safflower yellow is an extract of the famous Chinese medicine Carthamus tinctorious L, and safflower yellow injection (SYI) is widely used clinically to treat angina pectoris. However, there are few studies on the anti-myocardial ischemia/reperfusion (I/R) injury effect of SYI, and its mechanisms are unclear. In the present study, we aimed to investigate the protective effect of SYI on myocardial I/R injury and explore its underlying mechanisms. Male Sprague Dawley rats were randomly divided into a control group, sham group, model group, and SYI group (20 mg/kg, femoral vein injection 1 h before modeling). The left anterior descending coronary artery was ligated to establish a myocardial I/R model. H9c2 cells were exposed to oxygen–glucose deprivation/reoxygenation (OGD/R) after incubation with 80 μg/mL SYI for 24 h. In vivo, TsTC, HE, and TUNEL staining were performed to evaluate myocardial injury and apoptosis. A kit was used to detect superoxide dismutase (SOD) and malondialdehyde (MDA) to assess oxidative stress. In vitro, flow cytometry was used to detect the reactive oxygen species (ROS) content and apoptosis rate. Protein levels were determined via Western blotting. Pretreatment with SYI significantly reduced infarct size and pathological damage in rat hearts and suppressed cardiomyocyte apoptosis in vivo and in vitro. In addition, SYI inhibited oxidative stress by increasing SOD activity and decreasing MDA content and ROS production. Myocardial I/R and OGD/R activate endoplasmic reticulum (ER) stress, as evidenced by increased expression of activating transcription factor 6 (ATF6), glucose-regulated protein 78 (GRP78), cysteinyl aspartate-specific proteinase caspase-12, and C/EBP-homologous protein (CHOP), which were all inhibited by SYI. SYI ameliorated myocardial I/R injury by attenuating apoptosis, oxidative damage, and ER stress, which revealed new mechanistic insights into its application. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>SYI reduced myocardial I/R injury in rats. (<b>A</b>) Representative photographs of TTC-stained heart slices. (<b>B</b>) Quantitative analysis of the infarct area (<span class="html-italic">n</span> = 6). (<b>C</b>) LDH activity in serum (<span class="html-italic">n</span> = 6). (<b>D</b>) HE staining shows pathological changes in the myocardium (200× magnification; scale bar is 100 μm). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the sham group. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 compared with the model group.</p>
Full article ">Figure 2
<p>SYI attenuated I/R-induced myocardial apoptosis in rats. (<b>A</b>) Representative TUNEL staining (400× magnification; scale bar is 20 μm). (<b>B</b>) Quantitative analysis of apoptosis (<span class="html-italic">n</span> = 3). (<b>C</b>) The expression levels of Bax and Bcl−2 were analyzed via Western blotting. (<b>D</b>,<b>E</b>) The relative protein expression of Bax and Bcl−2 (<span class="html-italic">n</span> = 3). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the sham group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with the model group.</p>
Full article ">Figure 3
<p>SYI attenuated I/R-induced oxidative and ER stress in rat hearts. (<b>A</b>,<b>B</b>) The SOD activity and MDA content in the myocardium (<span class="html-italic">n</span> = 6). (<b>C</b>) The expression levels of ER stress-related proteins (ATF6, GRP78, caspase-12, and CHOP) were analyzed via Western blotting. (<b>D</b>–<b>G</b>) The relative protein expression of ATF6, GRP78, caspase-12, and CHOP (<span class="html-italic">n</span> = 3). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the sham group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with the model group.</p>
Full article ">Figure 4
<p>SYI reduced OGD/R-induced injury in H9c2 cells. (<b>A</b>) Cell viability of H9c2 cells incubated with different concentrations of SYI for 24 h. (<b>B</b>) Effects of SYI on cell viability after OGD/R (<span class="html-italic">n</span> = 6). (<b>C</b>) Effects of SYI on OGD/R-induced LDH leakage (<span class="html-italic">n</span> = 3). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the sham group. * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.01 compared with the model group.</p>
Full article ">Figure 5
<p>SYI reduced OGD/R-induced apoptosis in H9c2 cells. (<b>A</b>) Hoechst 33342 staining showing the anti-apoptotic potential of SYI. (<b>B</b>) Apoptosis in H9c2 cells was analyzed via flow cytometry. (<b>C</b>) Quantitative analysis of the apoptotic rate (<span class="html-italic">n</span> = 3). (<b>D</b>) Caspase-3 activity (<span class="html-italic">n</span> = 3). (<b>E</b>) The expression levels of Bax and Bcl-2 were analyzed via Western blotting. (<b>F</b>,<b>G</b>) The relative protein expression of Bax and Bcl-2 (<span class="html-italic">n</span> = 3). <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control group. * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 compared with the model group.</p>
Full article ">Figure 6
<p>SYI attenuated OGD/R-induced oxidative and ER stress in H9c2 cells. (<b>A</b>) The ROS expression level was observed. (<b>B</b>) Quantitative analysis of ROS fluorescence intensity. (<b>C</b>,<b>D</b>) SOD activity and MDA contents in cells (<span class="html-italic">n</span> = 3). (<b>E</b>) The level of ER stress was assessed. (<b>F</b>) Quantitative analysis of ER-Tracker Red staining (<span class="html-italic">n</span> = 3). (<b>G</b>) The expression levels of ER stress-related proteins (ATF6, GRP78, caspase-12, and CHOP) were analyzed via Western blotting. (<b>H</b>–<b>K</b>) The relative protein expression of ATF6, GRP78, caspase-12, and CHOP (<span class="html-italic">n</span> = 3). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the sham group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the model group.</p>
Full article ">
20 pages, 14588 KiB  
Article
Biomineralized MnO2 Nanoparticle-Constituted Hydrogels Promote Spinal Cord Injury Repair by Modulating Redox Microenvironment and Inhibiting Ferroptosis
by Yuyu Sun, Jinlong Zhang, Yong Gu, Tianqing Liu and Liang Chen
Pharmaceutics 2024, 16(8), 1057; https://doi.org/10.3390/pharmaceutics16081057 - 12 Aug 2024
Abstract
Spinal cord injury (SCI) is one of the most severe injuries, characterized by multiple positive feedback regulatory signaling networks formed by oxidative stress and inflammation in the injury microenvironment, leading to neuronal cell damage and even death. Here, astragaloside IV (AS), known for [...] Read more.
Spinal cord injury (SCI) is one of the most severe injuries, characterized by multiple positive feedback regulatory signaling networks formed by oxidative stress and inflammation in the injury microenvironment, leading to neuronal cell damage and even death. Here, astragaloside IV (AS), known for its regulatory role in ferroptosis, was encapsulated in the cavity of apoferritin (HFn) after an in situ biomineralization process involving MnO2, resulting in the synthesis of HFn@MnO2/AS nanoparticles. These nanoparticles were then dispersed in chitosan/polyvinyl alcohol/glutaraldehyde/sodium β-glycerophosphate (CGPG) hydrogels to form CGPG-HFn@MnO2/AS injectable thermosensitive hydrogels that can scavenge reactive oxygen species (ROS) in the microenvironment. Our findings indicated that the prepared CGPG-HFn@MnO2/AS hydrogel exhibited remarkable efficacy in scavenging ROS in vitro, effectively ameliorating the oxidative stress microenvironment post-SCI. Furthermore, it inhibited oxidative stress-induced ferroptosis in vitro and in vivo by regulating SIRT1 signaling, thereby promoting neuronal cell migration and repair. Hence, the developed hydrogel combining MnO2 and AS exhibited multifaceted abilities to modulate the pathological microenvironment, providing a promising therapeutic strategy for central nervous system (CNS) diseases. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Hydrodynamic particle size distribution of HFn detected by DLS. (<b>B</b>) The morphology image of the prepared HFn observed by TEM with negative staining by using 2% phosphotungstic acid (Scale bar: 50 nm). (<b>C</b>) Hydrodynamic particle size distribution of HFn@MnO<sub>2</sub>/AS nanoparticles detected by DLS. (<b>D</b>) The morphology image of HFn@MnO<sub>2</sub>/AS nanoparticles observed by TEM without negative staining (Scale bar: 100 nm). (<b>E</b>) Native page image of HFn and HFn@MnO<sub>2</sub>/AS nanoparticles. (<b>F</b>) CD spectra of HFn and HFn@MnO<sub>2</sub>/AS nanoparticles. (<b>G</b>) XPS analysis of HFn@MnO<sub>2</sub>/AS nanoparticles with full scan. (<b>H</b>) Mn 2p core-level spectra of HFn@MnO<sub>2</sub>/AS nanoparticles detected by XPS. (<b>I</b>) The hydrodynamic diameters of HFn@MnO<sub>2</sub>/AS NPs stored in PBS and PBS contained 10% FBS solutions at 4 °C for 7 days.</p>
Full article ">Figure 2
<p>(<b>A</b>) The mechanical properties of CS, CGP, CGG, CGPG, and CGPG-HFn@MnO<sub>2</sub>/AS hydrogels. n = 3, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p &lt;</span> 0.001. Rheological G′ and G″ against temperatures of GC (<b>B</b>), GCPC (<b>C</b>), and GCPC-HFn@MnO<sub>2</sub>/AS (<b>D</b>) hydrogels. (<b>E</b>) The gel formation image of blank hydrogels at 37 °C and the SEM micrograph of blank CGPG hydrogels (Scale bar: 50 μm). (<b>F</b>) The gel formation image of CGPG-HFn@MnO<sub>2</sub>/AS hydrogels at 37 °C and the SEM micrograph of CGPG-HFn@MnO<sub>2</sub>/AS hydrogels (Scale bar: 200 μm). (<b>G</b>) The in vitro degradation analysis of GC hydrogels. (<b>H</b>) The in vitro degradation analysis of CGP, CGG, and CGPG hydrogels. (<b>I</b>) The in vitro cumulative release of AS from HFn@MnO<sub>2</sub>/AS nanoparticles. (<b>J</b>) The in vitro cumulative release of AS from CGPG-HFn@MnO<sub>2</sub>/AS hydrogels.</p>
Full article ">Figure 3
<p>(<b>A</b>) The degradation behavior of HFn@MnO<sub>2</sub> incubated in different pH solutions (pH 7.4 and 6.5) with or without H<sub>2</sub>O<sub>2</sub>. (<b>B</b>) O<sub>2</sub>-bubbles image observed after incubation with H<sub>2</sub>O<sub>2</sub> in an acidic condition to confirm the formation of MnO<sub>2</sub>. (<b>C</b>) Evaluation of in vitro antioxidant effects of blank CGPG hydrogels and CGPG-HFn@MnO<sub>2</sub>/AS hydrogels in the absence of cells; ** <span class="html-italic">p &lt;</span> 0.01. (<b>D</b>) Evaluation of antioxidant effects of HFn@MnO<sub>2</sub>/AS in PC12 cells after being stimulated by H<sub>2</sub>O<sub>2</sub> (Scale bar: 500 μm) (<b>E</b>) Analysis of the ROS fluorescence intensity according to section D by Image J (version v1.53t). (<b>F</b>) Cellular uptake of FITC-HFn@MnO<sub>2</sub>/AS in CGPG hydrogels by PC12 cells detected by CLSM after incubation for 8, 12, and 24 h. (Scale bar: 75 μm).</p>
Full article ">Figure 4
<p>Effects of CGPG-HFn@MnO<sub>2</sub>/AS on H<sub>2</sub>O<sub>2</sub>-treated PC12 cells. (<b>A</b>) Cell viability was detected using the CCK-8 assay kit. (<b>B</b>) Cellular iron levels were detected using a kit. (<b>C</b>) Cellular GSH levels were detected using a kit. (<b>D</b>) Cellular MDA levels were detected using a kit. (<b>E</b>) Cellular SOD activity was detected using a kit. (<b>F</b>,<b>G</b>) Cellular ROS levels were detected using the fluorescent probe DCFH-DA. (Scale bar of <b>G</b>: 500 μm) (<b>H</b>–<b>M</b>) The protein levels of SIRT1, XCT, GPX4, 4-HNE, and TFR1 were detected by western blot assay. G1: control group. G2: H<sub>2</sub>O<sub>2</sub> group. G3: HFn@MnO<sub>2</sub>/AS group. G4:CGPG group. G5: CGPG-HFn@MnO<sub>2</sub>/AS. (<b>N</b>,<b>O</b>) Cell migration was evaluated using the wound healing assay. (Scale bar of <b>N</b>: 400 μm)Statistical analyses were performed using a one-way analysis of variance (ANOVA), followed by Tukey’s post-hoc test (n ≥ 3). # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 vs. the control group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. the H<sub>2</sub>O<sub>2</sub> group.</p>
Full article ">Figure 5
<p>Effects of CGPG-HFn@MnO<sub>2</sub>/AS on rats undergoing SCI. (<b>A</b>,<b>B</b>) The hind-limb motor function recovery of SCI rats was evaluated using footprint analysis. ## <span class="html-italic">p</span> &lt; 0.01 vs. the control group; ** <span class="html-italic">p</span> &lt; 0.01 vs. the SCI group. (<b>C</b>) The degree of hind-limb recovery was evaluated by the BBB score. ## <span class="html-italic">p</span> &lt; 0.01 vs. the sham group; ** <span class="html-italic">p</span> &lt; 0.01 vs. the SCI group. (<b>D</b>) The histological lesion was evaluated using H&amp;E staining. (<b>E</b>–<b>J</b>) The protein levels of SIRT1, XCT, GPX4, 4-HNE, and TFR1 were detected using a western blot assay. G1: sham group. G2: SCI group. G3: CGPG-HFn@MnO<sub>2</sub>/AS group. G4:CGPG group. G5: CGPG-HFn@MnO<sub>2</sub>/AS. (<b>K</b>) The level of iron in the spinal cord was detected using a kit. (<b>L</b>) The level of GSH in the spinal cord was detected using a kit. (<b>M</b>) The level of MDA in the spinal cord was detected using a kit. (<b>N</b>) The activity of SOD in the spinal cord was detected using a kit. Statistical analyses were performed using a one-way analysis of variance (ANOVA), followed by Tukey’s post-hoc test (n ≥ 3). ## <span class="html-italic">p</span> &lt; 0.01 vs. the sham group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. the SCI group.</p>
Full article ">Figure 6
<p>Safety analysis of CGPG-HFn@MnO<sub>2</sub>/AS hydrogels. (<b>A</b>) H&amp;E staining images of different organs (heart, liver, spleen, lung, and kidney) in healthy SD rats after administration (n = 3) (Scale bar: 200 μm). (<b>B</b>) Hematology analysis of RBCs, HGBs, PLTs, and WBCs in different groups (n = 3). (<b>C</b>) Serum biochemical analysis of BUN, ALT, AST, and ALP in different groups (n = 3).</p>
Full article ">
19 pages, 5690 KiB  
Article
Oxidative Stress, Oxidative Damage, and Cell Apoptosis: Toxicity Induced by Arecoline in Caenorhabditis elegans and Screening of Mitigating Agents
by Kaiping Xiang, Bing Wang, Lanying Wang, Yunfei Zhang, Hanzeng Li and Yanping Luo
Toxins 2024, 16(8), 352; https://doi.org/10.3390/toxins16080352 - 12 Aug 2024
Abstract
As the areca nut market is expanding, there is a growing concern regarding areca nut toxicity. Areca nut alkaloids are the major risky components in betel nuts, and their toxic effects are not fully understood. Here, we investigated the parental and transgenerational toxicity [...] Read more.
As the areca nut market is expanding, there is a growing concern regarding areca nut toxicity. Areca nut alkaloids are the major risky components in betel nuts, and their toxic effects are not fully understood. Here, we investigated the parental and transgenerational toxicity of varied doses of areca nut alkaloids in Caenorhabditis elegans. The results showed that the minimal effective concentration of arecoline is 0.2–0.4 mM. First, arecoline exhibited transgenerational toxicity on the worms’ longevity, oviposition, and reproduction. Second, the redox homeostasis of C. elegans was markedly altered under exposure to 0.2–0.4 mM arecoline. The mitochondrial membrane potential was thereafter impaired, which was also associated with the induction of apoptosis. Moreover, antioxidant treatments such as lycopene could significantly ameliorate the toxic effects caused by arecoline. In conclusion, arecoline enhances the ROS levels, inducing neurotoxicity, developmental toxicity, and reproductive toxicity in C. elegans through dysregulated oxidative stress, cell apoptosis, and DNA damage-related gene expression. Therefore, the drug-induced production of reactive oxygen species (ROS) may be crucial for its toxic effects, which could be mitigated by antioxidants. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of arecoline on the toxicity to <span class="html-italic">C. elegans.</span> (<b>A</b>) F0 generation lifespan; (<b>B</b>) F1 generation lifespan; (<b>C</b>) average longevity; (<b>D</b>) body length; (<b>E</b>) body width; (<b>F</b>) average egg production; (<b>G</b>) head thrashes; (<b>H</b>) body bends; (<b>I</b>) muscarinic acetylcholine receptor content. Data are presented as mean ± SEM. Values followed by the different lowercase letters within a row represent the significant difference (Tukey tests, <span class="html-italic">p</span> &lt; 0.05). One-way ANOVA with Tukey post hoc test.</p>
Full article ">Figure 2
<p>Effects of arecoline on ROS, lipofuscin accumulation, and gonad cell corpses in <span class="html-italic">C. elegans</span>. (<b>A</b>) Fluorescence pictures of ROS and lipofuscin accumulation and the gonad cell corpses assay. (<b>B</b>) Quantification of ROS accumulation. (<b>C</b>) Quantification of lipofuscin accumulation. (<b>D</b>) Quantification of gonad cell corpses. (<b>E</b>,<b>F</b>) Cell corpses were counted in N2. (<b>G</b>,<b>H</b>) Cell corpses were counted in the <span class="html-italic">ced-1(e1735)</span> strains. Values followed by the different lowercase letters within a row represent the significant difference (Tukey tests, <span class="html-italic">p &lt;</span> 0.05). One-way ANOVA with Tukey post hoc test.</p>
Full article ">Figure 3
<p>Effect of arecoline on the antioxidant enzyme system and non-enzymatic antioxidant system of <span class="html-italic">C. elegans</span>. (<b>A</b>) Oxidizing enzyme; (<b>B</b>) GSSG and GSH content; (<b>C</b>) MDA content, 8-OHdG content and protein carbonyl content; (<b>D</b>) UV stress; (<b>E</b>) mean survival time of UV stress. Values followed by the different lowercase letters within a row represent the significant difference (Tukey tests, <span class="html-italic">p &lt;</span> 0.05). *** Statistical significance at <span class="html-italic">p</span> &lt; 0.001. One-way ANOVA with Tukey post hoc test.</p>
Full article ">Figure 4
<p>Effect of arecoline on mitochondria in <span class="html-italic">C. elegans.</span> (<b>A</b>) Electron transport chain complex I; (<b>B</b>) electron transport chain complex III. (<b>C</b>) MMP. (<b>D</b>) Quantitative analysis of green/red fluorescence intensity ratio. Values followed by the different lowercase letters within a row represent the significant difference (Tukey tests, <span class="html-italic">p &lt;</span> 0.05). One-way ANOVA with Tukey post hoc test.</p>
Full article ">Figure 5
<p>Effect of arecoline on gene expression in <span class="html-italic">C. elegans</span>. (<b>A</b>) <span class="html-italic">daf-16</span> gene, <span class="html-italic">skn-1</span> gene and their downstream genes. (<b>B</b>) DNA damage genes. (<b>C</b>) Apoptosis genes.</p>
Full article ">Figure 6
<p>Effect of arecoline + antioxidants on ROS levels in <span class="html-italic">C. elegans</span>. (<b>A</b>) Fluorescence pictures of ROS. (<b>B</b>) Quantification of ROS accumulation. CK: 0 mM; NC: 0.04 mM antioxidants: curcumin; phillyrin; vitamin C; melatonin; lycopene; PC: 0.4 mM arecoline. Values followed by the different lowercase letters within a row represent the significant difference (Tukey tests, <span class="html-italic">p &lt;</span> 0.05). One-way ANOVA with Tukey post hoc test.</p>
Full article ">Figure 7
<p>Effect of arecoline + lycopene on the toxicity in <span class="html-italic">C. elegans</span>. (<b>A</b>) Lifespan. (<b>B</b>) Mean lifespan. (<b>C</b>) Body length. (<b>D</b>) Body width. (<b>E</b>) Head thrashes. (<b>F</b>) Body bends. (<b>G</b>) Average number of progent. CK: 0 mM; NC: 0.04 mM lycopene; PC: 0.4 mM arecoline. Values followed by the different lowercase letters within a row represent the significant difference (Tukey tests, <span class="html-italic">p &lt;</span> 0.05). One-way ANOVA with Tukey post hoc test.</p>
Full article ">Figure 8
<p>Effect of arecoline + lycopene on apoptosis. (<b>A</b>) Gonad apoptosis, the arrows indicate the number of apoptotic cells. (<b>B</b>) The gonad apoptosis in the worms was quantified using ImageJ 1.51 software. (<b>C</b>,<b>D</b>) Cell corpses were counted in N2. (<b>E</b>,<b>F</b>) Cell corpses were counted in <span class="html-italic">ced-1</span>(e1735) strains in <span class="html-italic">C. elegans.</span> CK: 0 mM; NC: 0.04 mM lycopene; PC: 0.4 mM arecoline, a &lt; 0.05, b &lt; 0.01, c &lt; 0.001. One-way ANOVA with Tukey post hoc test.</p>
Full article ">
22 pages, 21022 KiB  
Article
Forest Fire Detection Based on Spatial Characteristics of Surface Temperature
by Houzhi Yao, Zhigao Yang, Gui Zhang and Feng Liu
Remote Sens. 2024, 16(16), 2945; https://doi.org/10.3390/rs16162945 - 12 Aug 2024
Viewed by 150
Abstract
Amidst the escalating threat of global warming, which manifests in more frequent forest fires, the prompt and accurate detection of forest fires has ascended to paramount importance. The current surveillance algorithms employed for forest fire monitoring—including, but not limited to, fixed threshold algorithms, [...] Read more.
Amidst the escalating threat of global warming, which manifests in more frequent forest fires, the prompt and accurate detection of forest fires has ascended to paramount importance. The current surveillance algorithms employed for forest fire monitoring—including, but not limited to, fixed threshold algorithms, multi-channel threshold algorithms, and contextual algorithms—rely primarily upon the degree of deviation between the pixel temperature and the background temperature to discern pyric events. Notwithstanding, these algorithms typically fail to account for the spatial heterogeneity of the background temperature, precipitating the consequential oversight of low-temperature fire point pixels, thus impeding the expedited detection of fires in their initial stages. For the amelioration of this deficiency, the present study introduces a spatial feature-based (STF) method for forest fire detection, leveraging Himawari-8/9 imagery as the main data source, complemented by the Shuttle Radar Topography Mission (SRTM) DEM data inputs. Our proposed modality reconstructs the surface temperature information via selecting the optimally designated machine learning model, subsequently identifying the fire point through utilizing the difference between the reconstructed surface temperatures and empirical observations, in tandem with the spatial contextual algorithm. The results confirm that the random forest model demonstrates superior efficacy in the reconstruction of the surface temperature. Benchmarking the STF method against both the fire point datasets disseminated by the China Forest and Grassland Fire Prevention and Suppression Network (CFGFPN) and the Wild Land Fire (WLF) fire point product validation datasets from Himawari-8/9 yielded a zero rate of omission errors and a comprehensive evaluative index, predominantly surpassing 0.74. These findings show that the STF method proposed herein significantly augments the identification of lower-temperature fire point pixels, thereby amplifying the sensitivity of forest surveillance. Full article
Show Figures

Figure 1

Figure 1
<p>Overview map of the study area.</p>
Full article ">Figure 2
<p>Vegetation area and DEM in Hunan Province.</p>
Full article ">Figure 3
<p>Histogram of the frequency distribution of the surface temperatures in vegetation areas in Hunan Province on different dates.</p>
Full article ">Figure 4
<p>Flowchart of fire point detection algorithm.</p>
Full article ">Figure 5
<p>Feature correlation heatmap at different moments during the daytime.</p>
Full article ">Figure 6
<p>Scatter density plot of validation data for RF at different moments of the day.</p>
Full article ">Figure 7
<p>Scatter density plot of reconstructed LST versus original LST.</p>
Full article ">Figure 8
<p>LST of original vs. reconstructed vegetation area during daytime.</p>
Full article ">Figure 9
<p>LST of original vs. reconstructed area at nighttime.</p>
Full article ">Figure 10
<p>The result of fire point identification at 15:30 on 18 October 2022.</p>
Full article ">Figure 11
<p>The result of fire point identification at 10:30 on 19 October 2022.</p>
Full article ">Figure 12
<p>The result of fire point identification at 15:20 on 23 October 2022.</p>
Full article ">Figure 13
<p>The results of fire detection.</p>
Full article ">Figure 14
<p>Identification results of fire point image elements in Xintian County, Hunan Province, at four moments on 18 and 19 October 2022. (<b>a</b>) Mid-infrared 7th band of Himawari-9 image and its bright temperature. (<b>b</b>) Identification results of the algorithm of this study. (<b>c</b>) Results of WLF fire point product.</p>
Full article ">
25 pages, 7897 KiB  
Article
Fufang Muji Granules Ameliorate Liver Fibrosis by Reducing Oxidative Stress and Inflammation, Inhibiting Apoptosis, and Modulating Overall Metabolism
by Lei Men, Zhihong Gu, Enhua Wang, Jiwen Li, Zhongyu Li, Keke Li, Chunbin Li and Xiaojie Gong
Metabolites 2024, 14(8), 446; https://doi.org/10.3390/metabo14080446 - 11 Aug 2024
Viewed by 288
Abstract
Fufang Muji granules (FMGs) are a prominent modern prescription Chinese patent formulation derived from the Muji decoction. Utilized in clinical practice for nearly four decades, FMGs have demonstrated efficacy in treating liver diseases. However, the precise mechanism of action remains unclear. This study [...] Read more.
Fufang Muji granules (FMGs) are a prominent modern prescription Chinese patent formulation derived from the Muji decoction. Utilized in clinical practice for nearly four decades, FMGs have demonstrated efficacy in treating liver diseases. However, the precise mechanism of action remains unclear. This study investigates the hepatoprotective effects of FMGs against liver fibrosis in rats based on untargeted metabolomics and elucidates their underlying mechanisms. A comprehensive model of liver fibrosis was established with 30% CCl4 (2 mL/kg) injected intraperitoneally, and a fat and sugar diet combined with high temperatures and humidity. Rats were orally administered FMGs (3.12 g/kg/d) once daily for six weeks. FMG administration resulted in improved liver fibrosis and attenuated hepatic oxidative stress and apoptosis. Furthermore, FMGs inhibited hepatic stellate cell activation and modulated transforming growth factor β1/Smad signaling. Additionally, FMG treatment influenced the expression levels of interleukin-6, interleukin-1β, and tumour necrosis factor alpha in the injured liver. Metabolic pathways involving taurine and hypotaurine metabolism, as well as primary bile acid biosynthesis, were identified as mechanisms of action for FMGs. Immunohistochemistry, quantitative reverse transcription polymerase chain reaction (RT-qPCR), and quantitative analysis also revealed that FMGs regulated taurine and hypotaurine metabolism and bile acid metabolism. These findings provide a valuable understanding of the role of FMGs in liver fibrosis management. Full article
19 pages, 777 KiB  
Review
Regulation of Intestinal Inflammation by Walnut-Derived Bioactive Compounds
by Kexin Dai, Neel Agarwal, Alexander Rodriguez-Palacios and Abigail Raffner Basson
Nutrients 2024, 16(16), 2643; https://doi.org/10.3390/nu16162643 - 10 Aug 2024
Viewed by 624
Abstract
Walnuts (Juglans regia L.) have shown promising effects in terms of ameliorating inflammatory bowel disease (IBD), attributed to their abundant bioactive compounds. This review comprehensively illustrates the key mechanisms underlying the therapeutic potential of walnuts in IBD management, including the modulation of [...] Read more.
Walnuts (Juglans regia L.) have shown promising effects in terms of ameliorating inflammatory bowel disease (IBD), attributed to their abundant bioactive compounds. This review comprehensively illustrates the key mechanisms underlying the therapeutic potential of walnuts in IBD management, including the modulation of intestinal mucosa permeability, the regulation of inflammatory pathways (such as NF-kB, COX/COX2, MAPCK/MAPK, and iNOS/NOS), relieving oxidative stress, and the modulation of gut microbiota. Furthermore, we highlight walnut-derived anti-inflammatory compounds, such as polyunsaturated fatty acids (PUFA; e.g., ω-3 PUFA), tocopherols, phytosterols, sphingolipids, phospholipids, phenolic compounds, flavonoids, and tannins. We also discuss unique anti-inflammatory compounds such as peptides and polysaccharides, including their extraction and preparation methods. Our review provides a theoretical foundation for dietary walnut supplementation in IBD management and provides guidance for academia and industry. In future, research should focus on the targeted isolation and purification of walnut-derived anti-inflammatory compounds or optimizing extraction methods to enhance their yields, thereby helping the food industry to develop dietary supplements or walnut-derived functional foods tailored for IBD patients. Full article
Show Figures

Figure 1

Figure 1
<p>The mechanism of walnuts regulating IBD. (1) An illustration of the intestinal mucosal barrier and the effect of walnuts on permeability. (2) A depiction of the antioxidant effects of walnuts on ROS. (3) A pathway map showing NF-κB, COX/COX-2 and MAPK signaling modulation by walnuts. (4) Diagram showing changes in gut microbiota composition due to walnut consumption.</p>
Full article ">
22 pages, 3214 KiB  
Review
Plant-Based Nanovesicular Gel Formulations Applied to Skin for Ameliorating the Anti-Inflammatory Efficiency
by Hanan Abdelmawgoud Atia, Mona M. Shahien, Somaia Ibrahim, Enas Haridy Ahmed, Hemat A. Elariny and Marwa H. Abdallah
Gels 2024, 10(8), 525; https://doi.org/10.3390/gels10080525 - 10 Aug 2024
Viewed by 192
Abstract
Inflammation is a vascular response that occurs when the immune system responds to a range of stimuli including viruses, allergens, damaged cells, and toxic substances. Inflammation is accompanied by redness, heat, swelling, discomfort, and loss of function. Natural products have been shown to [...] Read more.
Inflammation is a vascular response that occurs when the immune system responds to a range of stimuli including viruses, allergens, damaged cells, and toxic substances. Inflammation is accompanied by redness, heat, swelling, discomfort, and loss of function. Natural products have been shown to have considerable therapeutic benefits, and they are increasingly being regarded as feasible alternatives for clinical preventative, diagnostic, and treatment techniques. Natural products, in contrast to developed medications, not only contain a wide variety of structures, they also display a wide range of biological activities against a variety of disease states and molecular targets. This makes natural products appealing for development in the field of medicine. In spite of the progress that has been made in the application of natural products for clinical reasons, there are still factors that prevent them from reaching their full potential, including poor solubility and stability, as well limited efficacy and bioavailability. In order to address these problems, transdermal nanovesicular gel systems have emerged as a viable way to overcome the hurdles that are encountered in the therapeutic use of natural products. These systems have a number of significant advantages, including the ability to provide sustained and controlled release, a large specific surface area, improved solubility, stability, increased targeting capabilities and therapeutic effectiveness. Further data confirming the efficacy and safety of nanovesicles–gel systems in delivering natural products in preclinical models has been supplied by extensive investigations conducted both in vitro and in vivo. This study provides a summary of previous research as well as the development of novel nanovesicular gel formulations and their application through the skin with a particular emphasis on natural products used for treatment of inflammation. Full article
Show Figures

Figure 1

Figure 1
<p>Arachidonic acid pathway and its inhibition by several drugs.</p>
Full article ">Figure 2
<p>Schematic diagram represents the structure of the conventional nanovesicles (liposomes and niosomes).</p>
Full article ">Figure 3
<p>The advantages and disadvantages of traditional nanovesicular systems [<a href="#B25-gels-10-00525" class="html-bibr">25</a>,<a href="#B43-gels-10-00525" class="html-bibr">43</a>].</p>
Full article ">Figure 4
<p>Schematic representation of the structure of various innovative nanovesicular carriers.</p>
Full article ">Figure 5
<p>The transdermal pathway of transethosomes through the skin [<a href="#B84-gels-10-00525" class="html-bibr">84</a>].</p>
Full article ">Figure 6
<p>Proposed concept for the production of phytosomes. The creation of hydrogen bonds between phytochemicals and the polar head of phospholipids is illustrated through schematic and structural images. Dashed lines symbolize the hydrogen bonds [<a href="#B105-gels-10-00525" class="html-bibr">105</a>].</p>
Full article ">
18 pages, 2619 KiB  
Review
Pathophysiological Relationship between Type 2 Diabetes Mellitus and Metabolic Dysfunction-Associated Steatotic Liver Disease: Novel Therapeutic Approaches
by Shifat-E Ferdous and Jessica M. Ferrell
Int. J. Mol. Sci. 2024, 25(16), 8731; https://doi.org/10.3390/ijms25168731 (registering DOI) - 10 Aug 2024
Viewed by 207
Abstract
Type 2 diabetes mellitus (T2DM), often featuring hyperglycemia or insulin resistance, is a global health concern that is increasing in prevalence in the United States and worldwide. A common complication is metabolic dysfunction-associated steatotic liver disease (MASLD), the hepatic manifestation of metabolic syndrome [...] Read more.
Type 2 diabetes mellitus (T2DM), often featuring hyperglycemia or insulin resistance, is a global health concern that is increasing in prevalence in the United States and worldwide. A common complication is metabolic dysfunction-associated steatotic liver disease (MASLD), the hepatic manifestation of metabolic syndrome that is also rapidly increasing in prevalence. The majority of patients with T2DM will experience MASLD, and likewise, individuals with MASLD are at an increased risk for developing T2DM. These two disorders may act synergistically, in part due to increased lipotoxicity and inflammation within the liver, among other causes. However, the pathophysiological mechanisms by which this occurs are unclear, as is how the improvement of one disorder can ameliorate the other. This review aims to discuss the pathogenic interactions between T2D and MASLD, and will highlight novel therapeutic targets and ongoing clinical trials for the treatment of these diseases. Full article
(This article belongs to the Special Issue New Advances in Type 2 Diabetes and Its Complications)
Show Figures

Figure 1

Figure 1
<p>Spectrum of MASLD progression. Due to obesity, lifestyle or other factors, a healthy liver may become steatotic (TG present in &gt;5% of hepatocytes). Steatohepatitis can develop from steatosis with the addition of inflammatory insults. Both MASLD and MASH are often reversible, while cirrhosis may partially regress with treatment. Otherwise, this can lead to hepatocellular carcinoma and may ultimately require liver transplantation [<a href="#B9-ijms-25-08731" class="html-bibr">9</a>,<a href="#B10-ijms-25-08731" class="html-bibr">10</a>].</p>
Full article ">Figure 2
<p>Inflammation, lipotoxicity and fibrosis contribute to the pathophysiological relationships between T2DM and MASLD. β-cell dysfunction causes IR in T2DM, followed by organ-specific pathology. (<b>A</b>) A chronic high-fat diet (HFD) can lead to IR, with increased glucose and insulin but blunted downstream receptor signaling, resulting in sustained elevated blood glucose. Glucose and insulin promote de novo lipogenesis through the activation of ChREBP and SREBP1c, respectively, to produce free fatty acids (FFAs). (<b>B</b>) Increased hepatic FFA causes mitochondrial dysfunction and ER stress followed by the rise of reactive oxygen species (ROS) and inflammatory mediators. The activation of JNK signaling by these mediators can cause hepatic cell damage and lead to inflammation; JNK signaling also decreases insulin sensitivity. (<b>C</b>) The development of IR is also related to alterations in adipokines secreted from adipose tissue. Decreased adiponectin induces lipotoxicity by increasing FFA, while increased inflammatory TNF-α drives hepatic satellite cell activation, leading to hepatic fibrosis due to excess collagen deposition. (<b>D</b>) Finally, chronic HFD affects the gut microbiota. Increased gut permeability and bacterial cellular lipopolysaccharide (LPS) or ethanol production due to dysbiosis leads to an increase in hepatic toll-like receptor 4 activation and subsequent inflammation and fibrogenesis in the liver. Red: hepatic pathways; green: adipocyte pathways; purple: intestinal pathways.</p>
Full article ">Figure 3
<p>Novel treatment approaches common to T2DM and MASLD. PPAR agonists and THR agonists act to reduce hepatic free fatty acids and subsequent lipogenesis. AMPK activation is targeted via direct agonists or indirectly through THR agonists and GPR119 agonists to reduce lipogenesis. Mitochondrial targeting increases thermogenesis and reduces gluconeogenesis to ultimately reduce lipogenesis and steatosis in the liver. In the pancreas, PPAR and THR agonists act to increase insulin secretion and reduce hepatic lipogenesis. In the distal small intestine, GRP119 agonists as well as incretin mimetics potently stimulate insulin release and promote weight loss.</p>
Full article ">Figure 4
<p>Bile-acid-based treatments for MASLD. Bile acids are synthesized from cholesterol and are released into the duodenum post-prandially to aid in nutrient and fat digestion. Bile acids also activate the receptors FXR and TGR5 to regulate metabolism. In enterocytes, the activation of FXR by bile acids or agonists induces FGF19 release which suppresses bile acid synthesis and has been shown to reduce steatosis. In hepatocytes, the activation of FXR suppresses bile acid synthesis and promotes glucose homeostasis and β-oxidation. Also in the liver, FGF21 promotes insulin sensitivity and reduces lipogenesis. In intestinal L-cells, the activation of TGR5 promotes insulin sensitivity through the release of GLP-1.</p>
Full article ">
17 pages, 9976 KiB  
Article
Nifedipine Improves the Ketogenic Diet Effect on Insulin-Resistance-Induced Cognitive Dysfunction in Rats
by Nancy M. Abdel-Kareem, Shimaa M. Elshazly, May A. Abd El Fattah, Afaf A. Aldahish, Sawsan A. Zaitone, Sahar K. Ali and Enas A. Abd El-Haleim
Pharmaceuticals 2024, 17(8), 1054; https://doi.org/10.3390/ph17081054 - 10 Aug 2024
Viewed by 260
Abstract
Insulin resistance, induced by high fructose consumption, affects cognitive function negatively. Nifedipine may be suggested for neurological disorders. This study aimed to assess the effect of nifedipine with either a normal diet (ND) or a ketogenic diet (KD) in cognitive dysfunction. Male Wistar [...] Read more.
Insulin resistance, induced by high fructose consumption, affects cognitive function negatively. Nifedipine may be suggested for neurological disorders. This study aimed to assess the effect of nifedipine with either a normal diet (ND) or a ketogenic diet (KD) in cognitive dysfunction. Male Wistar rats received 10% fructose in drinking water for 8 weeks to induce insulin resistance. Rats received nifedipine (5.2 mg/kg/day; p.o.) later with ND or KD for an additional five weeks. One and two-way ANOVAs were used in analyzing the data. Reversion to the ND improved insulin resistance and lipid profile, besides brain-derived neurotrophic factor (BDNF), glycogen synthase kinase-3 beta (GSK3β), and insulin-degrading enzyme (IDE) levels. Rats fed KD alone and those that received nifedipine with KD did not show similar improvement in the previously mentioned parameters as the ND group. However, nifedipine-ND rats showed improvement in cognitive behavior and insulin resistance. Treatment with nifedipine-KD ameliorated GSK3β, amyloid β (Aβ), and tau protein levels. As the nifedipine-KD combination succeeded in diminishing the accumulated Aβ and tau protein, KD may be used for a while due to its side effects, then nifedipine treatment could be continued with an ND. This conclusion is based on the finding that this combination mitigated insulin resistance with the associated improved behavior. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of the normal diet (ND) and the ketogenic diet (KD) with or without nifedipine (5.2 mg/kg/day, p.o.) on the body weight difference, Final weight of IR at the end of the experiment— their weights after 8 weeks’ treatment with 10% fructose in insulin-resistant (IR) rats. Data are represented as median and interquartile percent (25–75%) and analyzed using one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet; and + <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND + nifedipine group.</p>
Full article ">Figure 2
<p>Effect of 10% fructose treatment after 8 weeks on the oral glucose tolerance test (OGTT). Blood glucose levels were evaluated before glucose administration and after administration of 2.5 mg/kg glucose at 30, 60, and 90 min. Results are represented as the median and interquartile range (min. to max.) (<span class="html-italic">n</span> = 6) and analyzed using two-way ANOVA followed by the Bonferroni test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats.</p>
Full article ">Figure 3
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on the behavioral response in IR rats. Results are represented as median and interquartile range (min. to max.) (<span class="html-italic">n</span> = 6) and analyzed using two-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats.</p>
Full article ">Figure 4
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on serum lipid profile: (<b>a</b>) Cholesterol, (<b>b</b>) triglycerides, (<b>c</b>) high-density lipoprotein (HDL), and (<b>d</b>) low-density lipoprotein (LDL) (<b>d</b>) in (IR) rats. Results are represented as median and interquartile range (min. to max. with all points) (<span class="html-italic">n</span> = 6) and analyzed using one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; and @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet.</p>
Full article ">Figure 5
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on the brain-derived neurotrophic factor (BDNF) in the brain hippocampus of IR rats. Results are represented as median and interquartile range (25–75%) (<span class="html-italic">n</span> = 6) and analyzed using one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats.</p>
Full article ">Figure 6
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on (<b>a</b>) glycogen synthase kinase-3 beta (GSK3β) and (<b>b</b>) insulin-degrading enzyme (IDE) in the brain hippocampus of IR rats. Results are represented as median and interquartile range (25–75%) (<span class="html-italic">n</span> = 6) and analyzed using one-way ANOVA followed by the Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet; and + <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND + nifedipine group.</p>
Full article ">Figure 7
<p>Effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on histopathological examinations of insulin-resistant rats. Photomicrographs of hematoxylin-and-eosin-stained (<b>a</b>): for cerebral cortex and (<b>b</b>): for hippocampus slices from various groups. (<b>A</b>) Brain tissue that is uniform and without any neuronal damage. Grade 0 for both tissues. (<b>B</b>) The perineuronal edema and neurons with pyknotic nuclei (black arrows), Rosenthal fibers may be seen in gliosis (black arrowheads) regions, and the red arrows represent red neurons for both cerebral cortex and hippocampus, 75% of brain tissue. Grade 3 for both tissues. (<b>C</b>) Red arrows indicate that there are a few scattered red neurons, for both the cerebral cortex and hippocampus. For the cerebral cortex only, tissue edema (red arrowheads) is seen. The red neurons are sparsely distributed (red arrow). Perineuronal edema (black arrows), tissue edema (red arrowheads), and gliosis (black arrowheads) are all seen, 25% of brain tissue. Grade 2 for both tissues. (<b>D</b>) The red neurons are sparsely distributed (red arrow). Perineuronal edema (black arrows), tissue edema (red arrowheads), and gliosis in cerebral cortex only (black arrowheads) are all seen in both tissues, 45% of brain tissue. Grade 3 for cerebral cortex and 15% of brain tissue for hippocampus. Grade 2. (<b>E</b>) Neurons in cerebral cortex and hippocampus with pyknotic nuclei and perineuronal edema are seen (black arrows), 10% of brain tissue. Grade 1 for both tissues. There are gliosis regions with Rosenthal fibers in cerebral cortex only (black arrowheads). There are red neurons (shown by red arrows). Pictures captured at magnification power ×40 for cerebral cortex and ×20 for hippocampus.</p>
Full article ">Figure 8
<p>(<b>a</b>,<b>b</b>) (<b>A</b>) Weak focal levels of Aβ (black arrows). (<b>B</b>) Significant increase in levels of Aβ (black arrows). (<b>C</b>) Significant reduction in levels of Aβ (black arrows). (<b>D</b>) Significant increase in levels of Aβ (black arrows). (<b>E</b>) Weak focal levels of Aβ (black arrows). Pictures captured at magnification power ×40 for cerebral cortex and 20× for hippocampus. (<b>c</b>,<b>d</b>) (<b>A</b>) Weak levels of tau in the cytoplasm of a few neuron cells (black arrows). (<b>B</b>) There is an increase in the levels of tau in many neurons (black arrows). (<b>C</b>) There is a reduction in the levels of tau in many neurons (black arrows). (<b>D</b>) There is an increase in the levels of tau in many neurons (black arrows). (<b>E</b>) There are weak levels of tau in cytoplasm of some neurons (black arrows) in cerebral cortex while a significant increase in the levels of tau in cytoplasm of some neurons (black arrows). Pictures captured at magnification power ×40 for cerebral cortex and 20× for hippocampus. Statistical analysis showed the effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on immunohistochemical staining of cerebral cortex and hippocampal levels of amyloid β (Aβ) (<b>e</b>,<b>f</b>) and tau protein (<b>g</b>,<b>h</b>) of insulin-resistant rats. Results are represented as median and interquartile range (25–75%) (<span class="html-italic">n</span> = 6) and analyzed by one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet; and + <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND + nifedipine group.</p>
Full article ">Figure 8 Cont.
<p>(<b>a</b>,<b>b</b>) (<b>A</b>) Weak focal levels of Aβ (black arrows). (<b>B</b>) Significant increase in levels of Aβ (black arrows). (<b>C</b>) Significant reduction in levels of Aβ (black arrows). (<b>D</b>) Significant increase in levels of Aβ (black arrows). (<b>E</b>) Weak focal levels of Aβ (black arrows). Pictures captured at magnification power ×40 for cerebral cortex and 20× for hippocampus. (<b>c</b>,<b>d</b>) (<b>A</b>) Weak levels of tau in the cytoplasm of a few neuron cells (black arrows). (<b>B</b>) There is an increase in the levels of tau in many neurons (black arrows). (<b>C</b>) There is a reduction in the levels of tau in many neurons (black arrows). (<b>D</b>) There is an increase in the levels of tau in many neurons (black arrows). (<b>E</b>) There are weak levels of tau in cytoplasm of some neurons (black arrows) in cerebral cortex while a significant increase in the levels of tau in cytoplasm of some neurons (black arrows). Pictures captured at magnification power ×40 for cerebral cortex and 20× for hippocampus. Statistical analysis showed the effect of the ND and KD with or without nifedipine (5.2 mg/kg/day, p.o.) on immunohistochemical staining of cerebral cortex and hippocampal levels of amyloid β (Aβ) (<b>e</b>,<b>f</b>) and tau protein (<b>g</b>,<b>h</b>) of insulin-resistant rats. Results are represented as median and interquartile range (25–75%) (<span class="html-italic">n</span> = 6) and analyzed by one-way ANOVA followed by Tukey–Kramer test, with * <span class="html-italic">p</span> ≤ 0.05 vs. normal rats; # <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND; @ <span class="html-italic">p</span> ≤ 0.05 vs. IR + ketogenic diet; and + <span class="html-italic">p</span> ≤ 0.05 vs. IR + ND + nifedipine group.</p>
Full article ">Figure 9
<p>A schematic presentation for the results of the study.</p>
Full article ">
8 pages, 684 KiB  
Brief Report
Acetylcholinesterase Inhibitor Ameliorates Early Cardiometabolic Disorders in Fructose-Overloaded Rat Offspring
by Victor Hugo Martins de Miranda, Camila Paixão Dos Santos, Pietra Petrica Neves, Antonio Viana Nascimento-Filho, Marina Rascio Henriques Dutra, Nathalia Bernardes, Maria Claúdia Irigoyen and Kátia De Angelis
Pharmaceuticals 2024, 17(8), 1055; https://doi.org/10.3390/ph17081055 - 10 Aug 2024
Viewed by 228
Abstract
Background: We investigate the role of galantamine on autonomic dysfunction associated with early cardiometabolic dysfunction in the offspring of fructose-overloaded rats. Methods: Wistar rats received fructose diluted in drinking water (10%) or water for 60 days prior to mating. Fructose overload was maintained [...] Read more.
Background: We investigate the role of galantamine on autonomic dysfunction associated with early cardiometabolic dysfunction in the offspring of fructose-overloaded rats. Methods: Wistar rats received fructose diluted in drinking water (10%) or water for 60 days prior to mating. Fructose overload was maintained until the end of lactation. The offspring (21 days after birth) of control and fructose-overloaded animals were divided into three groups: control (C), fructose (F) and fructose + galantamine (GAL). GAL (5 mg/kg) was administered orally until the offspring were 51 days old. Metabolic, hemodynamic and cardiovascular autonomic modulation were evaluated. Results: The F group showed decreased insulin tolerance (KITT) compared to the C and GAL groups. The F group, in comparison to the C group, had increased arterial blood pressure, heart rate and sympathovagal balance (LF/HF ratio) and a low-frequency band of systolic arterial pressure (LF-SAP). The GAL group, in comparison to the F group, showed increased vagally mediated RMSSD index, a high-frequency band (HF-PI) and decreased LF/HF ratio and variance in SAP (VAR-SAP) and LF-SAP. Correlations were found between HF-PI and KITT (r = 0.60), heart rate (r = −0.65) and MAP (r = −0.71). Conclusions: GAL treatment significantly improved cardiovascular autonomic modulation, which was associated with the amelioration of cardiometabolic dysfunction in offspring of parents exposed to chronic fructose consumption. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Rate constant of glucose disappearance (KIIT). (<b>B</b>) Mean arterial pressure (mmHg). (<b>C</b>) Heart rate (bpm). * <span class="html-italic">p</span> &lt; 0.05 vs. C; &amp; <span class="html-italic">p</span> &lt; 0.05 vs. F. Control (C, <span class="html-italic">n</span> = 6), fructose (F, <span class="html-italic">n</span> = 6) and fructose treated with galantamine (GAL, <span class="html-italic">n</span> = 6).</p>
Full article ">Figure 2
<p>Pearson correlations involving offspring of fructose-fed and fructose + galantamine-fed rats. Positive correlation: (<b>A</b>) HF–PI and KITT. Negative correlations: (<b>B</b>) HF–PI and heart rate; and (<b>C</b>) HF–PI and MAP.</p>
Full article ">
18 pages, 4917 KiB  
Article
The Beneficial Effects of Lacticaseibacillus paracasei subsp. paracasei DSM 27449 in a Letrozole-Induced Polycystic Ovary Syndrome Rat Model
by Yan Zhang Lee, Shih-Hsuan Cheng, Yu-Fen Lin, Chien-Chen Wu and Ying-Chieh Tsai
Int. J. Mol. Sci. 2024, 25(16), 8706; https://doi.org/10.3390/ijms25168706 - 9 Aug 2024
Viewed by 237
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age globally. Emerging evidence suggests that the dysregulation of microRNAs (miRNAs) and gut dysbiosis are linked to the development of PCOS. In this study, the effects of Lacticaseibacillus paracasei subsp. [...] Read more.
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age globally. Emerging evidence suggests that the dysregulation of microRNAs (miRNAs) and gut dysbiosis are linked to the development of PCOS. In this study, the effects of Lacticaseibacillus paracasei subsp. paracasei DSM 27449 (DSM 27449) were investigated in a rat model of PCOS induced by letrozole. The administration of DSM 27449 resulted in improved ovarian function, reduced cystic follicles, and lower serum testosterone levels. Alterations in miRNA expressions and increased levels of the pro-apoptotic protein Bax in ovarian tissues were observed in PCOS-like rats. Notably, the administration of DSM 27449 restored the expression of miRNAs, including miR-30a-5p, miR-93-5p, and miR-223-3p, leading to enhanced ovarian function through the downregulation of Bax expressions in ovarian tissues. Additionally, 16S rRNA sequencing showed changes in the gut microbiome composition after letrozole induction. The strong correlation between specific bacterial genera and PCOS-related parameters suggested that the modulation of the gut microbiome by DSM 27449 was associated with the improvement of PCOS symptoms. These findings demonstrate the beneficial effects of DSM 27449 in ameliorating PCOS symptoms in letrozole-induced PCOS-like rats, suggesting that DSM 27449 may serve as a beneficial dietary supplement with the therapeutic potential for alleviating PCOS. Full article
(This article belongs to the Special Issue Molecular Research in Prebiotics, Probiotics and Postbiotics)
Show Figures

Figure 1

Figure 1
<p>Effects of letrozole treatment on the weight of rats. (<b>A</b>) A detailed animal treatment scheme. Female Sprague-Dawley rats were randomly divided into four groups. The control group received 1% carboxymethyl cellulose (CMC), whereas the letrozole, Diane-35, and DSM 27449 groups received 1 mg/kg body weight of letrozole during polycystic ovary syndrome (PCOS) induction. Letrozole-induced PCOS-like rats received phosphate-buffered saline (PBS), Diane-35, or DSM 27449 by oral gavage. Estrous cycle determination was performed during the experiment, and an oral glucose tolerance test (OGTT) was performed at the end of the experiment. (<b>B</b>) Growth curves of rats from day 1–42. (<b>C</b>) Average weight gain at the end of the study between the groups. N = 8 per 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, and **** <span class="html-italic">p</span> &lt; 0.0001 compared with the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared with the letrozole group.</p>
Full article ">Figure 2
<p>Effects of <span class="html-italic">L. paracasei</span> subsp. <span class="html-italic">paracasei</span> DSM 27449 on ovarian function in letrozole-induced polycystic ovary syndrome-like rats. (<b>A</b>) Representative estrous cycles in rats (P, proestrus; E, estrus; M/D, metestrus/diestrus). (<b>B</b>) Percentage of time spent in different phases of the estrous cycle for the last five days of the experiment. (<b>C</b>) Hematoxylin and eosin staining of representative ovaries. The number of (<b>D</b>) cystic follicles and (<b>E</b>) corpora lutea of the experimental groups. N = 8 per 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, and **** <span class="html-italic">p</span> &lt; 0.0001 compared with the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared with the letrozole group. Scale bar = 100 μm in (<b>C</b>).</p>
Full article ">Figure 3
<p><span class="html-italic">L. paracasei</span> subsp. <span class="html-italic">paracasei</span> DSM 27449 attenuated an increase in serum testosterone levels and androgen-receptor (AR) expression in letrozole-induced polycystic ovary syndrome-like rats. (<b>A</b>) Serum testosterone levels. N = 6–8 per group. (<b>B</b>) AR immunohistochemical staining of representative ovarian sections. (<b>C</b>) Quantitative analysis of AR-positive areas in the ovarian sections. N = 8 per 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, and **** <span class="html-italic">p</span> &lt; 0.0001 compared with the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the letrozole group. Scale bar = 100 μm in (<b>B</b>).</p>
Full article ">Figure 4
<p><span class="html-italic">L. paracasei</span> subsp. <span class="html-italic">paracasei</span> DSM 27449 downregulated the expression of the pro-apoptotic protein Bax in the ovarian tissues of letrozole-induced polycystic ovary syndrome (PCOS)-like rats. (<b>A</b>) Heat map of the Spearman’s rank correlation test visualizing the correlation between PCOS-related parameters and microRNA expressions. N = 6–8 per group, * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01. (<b>B</b>) Immunohistochemical staining of Bax (top), Bcl-2 (middle), and Beclin-1 (bottom) on representative ovarian sections. Quantitative analysis of the expression of (<b>C</b>) Bax, (<b>D</b>) Bcl-2, and (<b>E</b>) Beclin-1 in the ovarian sections. N = 8 per group; ** <span class="html-italic">p</span> &lt; 0.01 compared with the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared with the letrozole group. Scale bar = 100 μm in (<b>B</b>).</p>
Full article ">Figure 5
<p>Effects of <span class="html-italic">L. paracasei</span> subsp. <span class="html-italic">paracasei</span> DSM 27449 on the gut microbiome in letrozole-induced polycystic ovary syndrome-like rats. Alpha diversity is represented by (<b>A</b>) the Chao1 index, (<b>B</b>) the Shannon index, and (<b>C</b>) the Simpson index. N = 8 per group; <sup><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01 compared with the Diane-35 group. (<b>D</b>) Beta diversity is represented by a non-metric multi-dimensional scaling (NMDS) plot and the analysis of similarities (ANOSIMs). (<b>E</b>) Composition of the gut microbiome at the phylum level. (<b>F</b>–<b>K</b>) Relative abundance of differential bacteria at the genus level between the control and letrozole groups and between the control and DSM 27449 groups. N = 8 per group; the Bonferroni method was applied to correct for Type I errors, with statistical significance considered at * <span class="html-italic">p</span> &lt; 0.05/2.</p>
Full article ">Figure 5 Cont.
<p>Effects of <span class="html-italic">L. paracasei</span> subsp. <span class="html-italic">paracasei</span> DSM 27449 on the gut microbiome in letrozole-induced polycystic ovary syndrome-like rats. Alpha diversity is represented by (<b>A</b>) the Chao1 index, (<b>B</b>) the Shannon index, and (<b>C</b>) the Simpson index. N = 8 per group; <sup><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01 compared with the Diane-35 group. (<b>D</b>) Beta diversity is represented by a non-metric multi-dimensional scaling (NMDS) plot and the analysis of similarities (ANOSIMs). (<b>E</b>) Composition of the gut microbiome at the phylum level. (<b>F</b>–<b>K</b>) Relative abundance of differential bacteria at the genus level between the control and letrozole groups and between the control and DSM 27449 groups. N = 8 per group; the Bonferroni method was applied to correct for Type I errors, with statistical significance considered at * <span class="html-italic">p</span> &lt; 0.05/2.</p>
Full article ">Figure 6
<p>Heat maps of the Spearman’s rank correlation coefficient. (<b>A</b>) Heat map representing the correlation between differential genera and polycystic ovary syndrome-related parameters. (<b>B</b>) Heat map representing the correlation between differential genera and microRNA expressions. N = 6–8 per 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, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
16 pages, 24226 KiB  
Article
Protective Effects of Recombined Mussel Adhesive Protein against AD Skin Inflammation in Mice
by Yu Wu, Feng Li, Yan Gong, Xing Wan and Li-Ming Zhou
Cosmetics 2024, 11(4), 134; https://doi.org/10.3390/cosmetics11040134 - 9 Aug 2024
Viewed by 308
Abstract
(1) Background: Atopic dermatitis (AD) is characterized as a chronic inflammatory skin disease with a significant incidence rate. The pathophysiological mechanisms underlying AD remain incompletely understood. However, extensive research demonstrates that a complex interplay among genetic, immune, and environmental factors contributes to the [...] Read more.
(1) Background: Atopic dermatitis (AD) is characterized as a chronic inflammatory skin disease with a significant incidence rate. The pathophysiological mechanisms underlying AD remain incompletely understood. However, extensive research demonstrates that a complex interplay among genetic, immune, and environmental factors contributes to the disruption of skin barrier function. Inflammation is identified as one of the pathological mechanisms in AD. Recombined mussel adhesive protein exhibits anti-inflammatory properties. However, recombinant mussel adhesive protein has been used less frequently for AD, so we explored the therapeutic effect of recombinant mussel adhesive protein for AD and the potential mechanism. (2) Methods: We established a mice model of AD in vivo and an LPS-induced inflammation model in HaCaT cells in vitro. Through assessment of skin lesion scores, itch frequency, transepidermal water loss, skin microcirculation, HE staining, Elisa assays for IL-6, IL-12, IL-13, IL-4, IL-5, IFN-γ, IgE, and TNF-α, immunohistochemical staining for filaggrin and CK14, Masson staining, and Western blot analysis of NF-κB p65, P-P65, Keap1, and Nrf2, the effects of recombined mussel adhesive protein on AD symptoms, pathology, inflammation, and its mechanisms are investigated. (3) Results: The recombined mussel adhesive protein significantly improved the compromised skin barrier, reduced scratching frequency in mice, decreased transepidermal water loss, and lowered the expression of inflammatory factors, thus ameliorating skin inflammation damage. Mechanistically, recombined mussel adhesive protein downregulated the expression of P-p65/p65 and Keap1 while upregulating the level of Nrf2. (4) Conclusions: Overall, our results demonstrate the effectiveness of recombined mussel adhesive protein in attenuating DNFB-induced AD by inhibiting NF-κB and activating the Keap1/Nrf2 signaling pathway. Thus, recombined mussel adhesive protein is a promising therapeutic candidate for the treatment of AD. Full article
Show Figures

Figure 1

Figure 1
<p>The effect of recombined mussel adhesive protein on the skin lesions in mice with AD. (<b>A</b>) Skin lesion scores: #: compared with the control group, <span class="html-italic">p</span> &lt; 0.05; *: compared with the experimental group, <span class="html-italic">p</span> &lt; 0.05. (<b>B</b>) H&amp;E staining of the skin lesion: scale bar = 200 μm. (<b>C</b>) Masson staining of the skin lesion: scale bar = 200 μm.</p>
Full article ">Figure 2
<p>The effect of recombined mussel adhesive protein on the skin barrier function in mice with AD. (<b>A</b>) Scratching times. (<b>B</b>) Transcutaneous water loss rates. (<b>C</b>) Microcirculation index. (<b>D</b>) Statistical results of microcirculation index. Values represent the mean ± SD; a <span class="html-italic">t</span>-test was used for significance analysis. #: compared with the control group, <span class="html-italic">p</span> &lt; 0.05; *: compared with the model group, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>The effect of recombined mussel adhesive protein on the skin keratin in mice with AD. (<b>A</b>–<b>C</b>) Immunohistochemical results of CK14 in the skin of mice with specific dermatitis after drug intervention, ((<b>A</b>)—4×), ((<b>B</b>)—10×); (<b>D</b>–<b>F</b>) Immunohistochemical results of filaggrin in the skin of mice with specific dermatitis after drug intervention, ((<b>D</b>)—4×), ((<b>F</b>)—10×). Values represent the mean ± SD; a <span class="html-italic">t</span>-test was used for significance analysis. *: compared with the model group, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>The effect of recombined mussel adhesive protein on inflammation in mice with AD. (<b>A</b>–<b>G</b>) Results of Elisa assay for inflammatory factor: TNF-α, INF-γ, IL-12, IL-13, IL-5, IL-4, and IgE. Values represent the mean ± SD; a <span class="html-italic">t</span>-test was used for significance analysis. #: compared with the control group, <span class="html-italic">p</span> &lt; 0.05; *: compared with the model group, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>The effect of recombined mussel adhesive protein on inflammation in HaCaT cells. (<b>A</b>) Effect of recombined mussel adhesive protein on the cell viability of HaCaT cells. (<b>B</b>–<b>E</b>) Effect of recombined mussel adhesive protein on the LPS-induced expression of inflammatory factors in HaCaT cells: TNF-α, INF-γ, IL-6, IL-13. Values represent the mean ± SD; a <span class="html-italic">t</span>-test was used for significance analysis. #: compared with the control group, <span class="html-italic">p</span> &lt; 0.05; *: compared with the model group, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>The effect of recombined mussel adhesive protein on the NF-κB and Keap1/Nrf2 pathways. (<b>A</b>,<b>B</b>) Protein levels of NF-κB p6 and P-p65. (<b>C</b>,<b>D</b>) Protein levels of Keap1 and Nrf2. Values represent the mean ± SD; a <span class="html-italic">t</span>-test was used for significance analysis. #: compared with the control group, <span class="html-italic">p</span> &lt; 0.05; *: compared with the model group, <span class="html-italic">p</span> &lt; 0.05; @: compared with the recombined mussel adhesive protein group, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
17 pages, 2535 KiB  
Article
Association between Dietary Inflammatory Index and Hyperemesis Gravidarum
by Shihan Zhi, Lan Zhang, Wenjie Cheng, Yuan Jin, Zhaoqing Long, Wei Gu, Le Ma, Shunming Zhang and Jing Lin
Nutrients 2024, 16(16), 2618; https://doi.org/10.3390/nu16162618 - 8 Aug 2024
Viewed by 430
Abstract
(1) Background: Diet holds a pivotal position in exacerbating or ameliorating chronic inflammation, which has been implicated in the pathogenesis of hyperemesis gravidarum (HG). However, no study has explored the association between dietary inflammatory potential and HG. This study aimed to investigate the [...] Read more.
(1) Background: Diet holds a pivotal position in exacerbating or ameliorating chronic inflammation, which has been implicated in the pathogenesis of hyperemesis gravidarum (HG). However, no study has explored the association between dietary inflammatory potential and HG. This study aimed to investigate the potential correlation between following a pro-inflammatory diet and the likelihood of developing HG. (2) Methods: A total of 2033 Chinese pregnant women (mean age: 31.3 ± 3.4 years) were included in this cross-sectional study from April 2021 to September 2022 as part of the China Birth Cohort Study (CBCS). Dietary inflammatory index (DII) scores with 23 food components were constructed through dietary intakes collected via a reliable 108-item semi-quantitative food frequency questionnaire. HG was defined as a pregnancy-unique quantification of emesis (PUQE) score ≥13 points, severe nausea and vomiting leading to weight loss ≥5%, or being hospitalized for treatment due to the disease. The relationship between DII and HG was conducted utilizing binary logistic regression and restricted cubic spline regression. (3) Results: Overall, 8.2% (n = 167) of study participants had HG. The DII scores ranged from −4.04 to 3.82. After adjusting for potential confounders, individuals with the highest tertile of DII score had a higher risk of HG (OR = 1.65, 95% CI: 1.04, 2.62, Ptrend = 0.032). Such an association was stronger in those with pre-pregnancy overweight/obesity (Pinteraction = 0.018). (4) Conclusions: A higher DII score, which serves as a marker for a diet promoting inflammation, is correlated with an elevated risk of developing HG. This finding suggests that dietary recommendations for HG should focus on minimizing the DII through incorporating foods abundant in anti-inflammatory components. Full article
Show Figures

Figure 1

Figure 1
<p>The restricted cubic spline for the association of dietary inflammatory index with hyperemesis gravidarum. Knots were placed at the 25th, 50th, and 75th percentiles of the dietary inflammatory index distribution. The solid red line represents the point estimate of the OR, and the red area represents the 95% confidence interval. Results were adjusted for age, gestational weeks, parity, total energy intake, physical activity, pre-pregnancy body mass index, annual household income, educational level, employment status, smoking, alcohol drinking, and use of nutritional supplements. OR: odds ratio, CI: confidence interval, HG: hyperemesis gravidarum.</p>
Full article ">Figure 2
<p>Subgroup analyses for the association of dietary inflammatory index with the risk of hyperemesis gravidarum. The association of dietary inflammatory index with hyperemesis gravidarum was linear, and thus we modeled DII as a continuous variable (per standard deviation increase) in the subgroup analysis. The left side shows the variables and their grouping, the gray vertical line indicates that the OR is 1, the diamond indicates the point estimate of the OR, the horizontal line represents the corresponding 95% CI, purple indicates no statistically significant difference, and green indicates that the difference is significant. We did not conduct a subgroup analysis of smoking and alcohol drinking due to the small number of smokers and alcohol drinkers. Multivariable binary logistic analyses were conducted by adjusting for age, gestational weeks, parity, total energy intake, physical activity, pre-pregnancy body mass index, annual household income, educational level, employment status, smoking, alcohol drinking, and use of nutritional supplements. OR: odds ratio, CI: confidence interval.</p>
Full article ">Figure A1
<p>Distribution of the dietary inflammatory index among the study participants (<span class="html-italic">n</span> = 2033).</p>
Full article ">
20 pages, 655 KiB  
Review
Biomaterials Designed to Modulate Reactive Oxygen Species for Enhanced Bone Regeneration in Diabetic Conditions
by Mingshan Li, Zhihe Zhao and Jianru Yi
J. Funct. Biomater. 2024, 15(8), 220; https://doi.org/10.3390/jfb15080220 - 8 Aug 2024
Viewed by 288
Abstract
Diabetes mellitus, characterized by enduring hyperglycemia, precipitates oxidative stress, engendering a spectrum of complications, notably increased bone vulnerability. The genesis of reactive oxygen species (ROS), a byproduct of oxygen metabolism, instigates oxidative detriment and impairs bone metabolism in diabetic conditions. This review delves [...] Read more.
Diabetes mellitus, characterized by enduring hyperglycemia, precipitates oxidative stress, engendering a spectrum of complications, notably increased bone vulnerability. The genesis of reactive oxygen species (ROS), a byproduct of oxygen metabolism, instigates oxidative detriment and impairs bone metabolism in diabetic conditions. This review delves into the mechanisms of ROS generation and its impact on bone homeostasis within the context of diabetes. Furthermore, the review summarizes the cutting-edge progress in the development of ROS-neutralizing biomaterials tailored for the amelioration of diabetic osteopathy. These biomaterials are engineered to modulate ROS dynamics, thereby mitigating inflammatory responses and facilitating bone repair. Additionally, the challenges and therapeutic prospects of ROS-targeted biomaterials in clinical application of diabetic bone disease treatment is addressed. Full article
Show Figures

Figure 1

Figure 1
<p>Out-of-balance bone regeneration in diabetic states. The left half depicts the inhibitory effects of diabetic condition on bone formation. The right half highlights the promotion of bone resorption.</p>
Full article ">
Back to TopTop