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

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (480)

Search Parameters:
Keywords = Beclin-1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 4882 KiB  
Article
Tat-Beclin-1 Peptide Ameliorates Metabolic Dysfunction-Associated Steatotic Liver Disease by Enhancing Hepatic Autophagy
by Chun-Liang Chen, Fen-Fen Huang, Hsueh-Fang Lin, Chi-Chien Wu, Yen-Hsuan Ni and Yu-Cheng Lin
Int. J. Mol. Sci. 2024, 25(22), 12372; https://doi.org/10.3390/ijms252212372 (registering DOI) - 18 Nov 2024
Abstract
Autophagy plays a crucial role in hepatic lipid metabolism, making it a key therapeutic target for addressing metabolic dysfunction-associated steatotic liver disease (MASLD). This study evaluates the efficacy of the Tat-Beclin-1 (TB-1) peptide, a specific autophagy inducer, in mitigating MASLD. Initially, we examined [...] Read more.
Autophagy plays a crucial role in hepatic lipid metabolism, making it a key therapeutic target for addressing metabolic dysfunction-associated steatotic liver disease (MASLD). This study evaluates the efficacy of the Tat-Beclin-1 (TB-1) peptide, a specific autophagy inducer, in mitigating MASLD. Initially, we examined the impact of the TB-1 peptide on autophagic activity and intracellular lipid metabolism in HepG2 cells treated with oleic acid, using a Tat scrambled (TS) control peptide for comparison. Subsequently, we established a MASLD mouse model by feeding a high-fat diet (HFD) for 16 weeks, followed by intraperitoneal administration of TB-1 or TS. Assessments included liver histopathology, serum biochemistry, and autophagy marker analysis. Our findings indicate that the TB-1 peptide significantly increased the LC3II/β-actin ratio in a dose- and time-dependent manner while promoting the expression of key autophagy markers Beclin-1 and ATG5-12. Furthermore, TB-1 treatment led to a marked reduction in both the size and number of lipid droplets in HepG2 cells. In vivo, HFD-fed mice exhibited increased liver weight, elevated serum alanine aminotransferase levels, and impaired oral glucose tolerance. TB-1 administration effectively mitigated these hepatic and metabolic disturbances. Histological analysis further revealed a substantial reduction in the severity of hepatic steatosis and fibrosis in TB-1-treated mice compared to TS controls. In conclusion, the TB-1 peptide shows significant potential in reducing the severity of MASLD in both HepG2 cell models and HFD-induced MASLD mouse models. Enhancing autophagy through TB-1 represents a promising therapeutic strategy for treating MASLD. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

Figure 1
<p>Induction of autophagy marker expression in HepG2 cells by TB-1 peptide. HepG2 cells were treated with TB-1 peptide or TS control peptide at various concentrations (<b>A</b>,<b>B</b>) and for different durations (<b>C</b>,<b>D</b>). B+ and B− represent conditions with or without bafilomycin A1 treatment, respectively. Immunoblot analyses were conducted to quantify the levels of autophagy marker proteins. The fold change for each target protein is shown relative to the TS control sample. Statistical significance: * <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. TS: Tat-Beclin-1 scrambled control peptide; TB: Tat-Beclin-1 peptide.</p>
Full article ">Figure 2
<p>Reduction in intracellular lipid droplet content in HepG2 cells by TB-1 peptide. The upper panel shows Oil Red O staining of HepG2 cells treated with medium alone or TS control peptide. Treatment with TB-1 peptide at concentrations of 10, 30, and 50 μM resulted in a significant, dose-dependent decrease in lipid droplet content (red) (<b>A</b>). The lower panel presents the quantification of the average number and size of lipid droplets per cell (<b>B</b>). Lipid droplets were quantified using ImageJ software (version 1.53k), with at least 20 cells analyzed per experiment to determine the average number and size. Data are presented as the mean ± SD of three independent experiments. The scale bar represents 50 μm. Statistical significance: *** <span class="html-italic">p</span> &lt; 0.001. TS: Tat-Beclin-1 scrambled control peptide; TB: Tat-Beclin-1 peptide.</p>
Full article ">Figure 3
<p>Reduction in liver steatosis and fibrosis in the HFD-induced murine MASLD model by TB-1 peptide. Liver histology was evaluated using H and E staining to assess liver steatosis, indicated by black arrow heads (<b>A</b>) and Masson’s trichrome staining to evaluate liver fibrosis, indicated by black arrows (<b>B</b>) following treatment with the TB-1 peptide. Liver steatosis (<b>C</b>) and fibrosis (<b>D</b>) were quantified in all samples using ImageJ software (version 1.53k) and collagen proportionate area (CPA) measurement. Data are presented as mean ± SD (<span class="html-italic">n</span> = 10 per group). The scale bar represents 100 μm, with a magnification of 200×. Statistical significance: *** <span class="html-italic">p</span> &lt; 0.001. Abbreviations: HFD, high-fat diet; CD, chow diet; H and E, hematoxylin and eosin; CPA, collagen proportionate area; LD, lipid droplet; TS, Tat scrambled control peptide; TB, Tat-Beclin-1 peptide.</p>
Full article ">Figure 4
<p>Induction of autophagy in the liver of HFD-induced murine MASLD model by TB-1 peptide. (<b>A</b>) Protein expression levels in the liver of mice were analyzed using Western blotting. (<b>B</b>) The immunoblots were quantified using Image J software (version 1.53k), with results presented as mean ± SD (<span class="html-italic">n</span> = 10 per group). The blots show the effects of TB-1 peptide on the levels of autophagy-related proteins, including LC3-II, SQSTM1/p62, Beclin-1, SIRT1, and the phospho-ULK1 (Ser 317) to total ULK1 ratio. Statistical significance: * <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. Abbreviations: CD, chow diet; HFD, high fat diet; TS, Tat scrambled control peptide; TB, Tat-Beclin-1 peptide; SIRT1, Sirtuin 1; ULK1, Unc-51-like kinase 1.</p>
Full article ">Figure 5
<p>The effects of TB-1 peptide on hepatic fatty acid oxidation. (<b>A</b>) Expression levels of fatty acid oxidation-related genes in the liver of the HFD-induced murine MASLD model. (<b>B</b>) Oxygen consumption rate (OCR), (<b>C</b>) basal respiration, (<b>D</b>) maximal respiration, and (<b>E</b>) ATP production measured in oleic acid-loaded HepG2 cells following TB-1 peptide treatment. All data are presented as fold changes compared to the expression level of the CD + TS group (<span class="html-italic">n</span> = 10 per group). Statistical significance: * <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. Abbreviations: CD, chow diet; HFD, high-fat diet; TS, Tat scrambled control peptide; TB, Tat-Beclin-1 peptide; <span class="html-italic">Ucp2</span>, uncoupling protein 2; <span class="html-italic">Lcad</span>, long-chain acyl-CoA dehydrogenase; <span class="html-italic">Cpt-1α</span>, carnitine palmitoyl-transferase 1α; <span class="html-italic">Acox</span>, acyl-CoA oxidase; <span class="html-italic">Ppar-α</span>, peroxisome proliferator-activated receptor α; OCR, oxygen consumption rate.</p>
Full article ">Figure 6
<p>Proposed mechanism of action of TB-1 peptide in promoting autophagy, leading to reduced lipid droplet accumulation. GAPR-1: Golgi-Associated Plant Pathogenesis-Related Protein 1. Red arrow means increase and blue arrow means decrease.</p>
Full article ">
12 pages, 9628 KiB  
Article
Selenized Yeast Protects Against Cadmium-Induced Follicular Atresia in Laying Hens by Reducing Autophagy in Granulosa Cells
by Caimei Wu, Yuxuan Jiang, Ziyun Zhou, Yuwei Zhang, Yixuan Zhou, Shiping Bai, Jian Li, Fali Wu, Jianping Wang and Yang Lyu
Curr. Issues Mol. Biol. 2024, 46(11), 13119-13130; https://doi.org/10.3390/cimb46110782 (registering DOI) - 18 Nov 2024
Viewed by 76
Abstract
Cadmium (Cd) exposure can induce follicular atresia and laying performance reduction in hens, which is linked to autophagy within the granulosa cells. Selenium (Se) can influence autophagy and counteract Cd toxicity. This study aimed to investigate the protective effect of Se on Cd-induced [...] Read more.
Cadmium (Cd) exposure can induce follicular atresia and laying performance reduction in hens, which is linked to autophagy within the granulosa cells. Selenium (Se) can influence autophagy and counteract Cd toxicity. This study aimed to investigate the protective effect of Se on Cd-induced follicular atresia in laying hens. Sixty-four laying hens were randomly allocated into 4 treatments: control group: basal diet; Se group: basal diet + 0.4 mg/kg Se from selenized yeast; Cd group: basal diet + 25 mg/kg Cd from CdCl2; and Cd+Se group: basal diet + 25 mg/kg Cd + 0.4 mg/kg Se. Compared to the Cd group, Se supplementation alleviated the ovarian pathological changes and oxidative stress in the follicles, serum, liver, and ovary, increased daily laying production, ovarian weight and F5–F1 follicle amounts, serum levels of progesterone and oestradiol, and up-regulated mTOR expression (p < 0.05), while decreasing the count of autophagic vacuoles, ovarian atresia follicle numbers, and Cd deposition, and down-regulated expression levels of autophagy-related mRNAs, including ATG5, LC3-I, and LC3-II, Beclin1, and Dynein in the follicles (p < 0.05). In conclusion, 0.4 mg/kg Se supplementation protected against Cd-induced laying performance reduction and follicular atresia, which were achieved via decreasing oxidative stress and inhibiting mTOR pathways of autophagy. Full article
(This article belongs to the Special Issue Molecular Research in Food Science)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effects of Cd and Se supplementation on the morphological changes in follicles and ovarian weight in laying hens. (<b>A</b>) Ovarian relative weight; (<b>B</b>) number of F1–F5 follicles; (<b>C</b>) ovarian morphology: a—the control (Con) group, b—the Se group, c—the Cd group, and d—the Cd+Se group. Different letters (a and b) on the bars represent significant differences (<span class="html-italic">p</span> &lt; 0.05), data are means ± SEM (<span class="html-italic">n</span> = 8).</p>
Full article ">Figure 2
<p>Effects of Cd and Se supplementation on the counts of primordial follicles, primary follicles, and atretic follicles (<b>A</b>), as well as on ovarian histomorphology (<b>B</b>) in laying hens aged 63 to 70 weeks. (<b>B</b>) The histological images were magnified 200 times, and the yellow arrow indicates that the ovarian vacuolar cells are clearly visible. Different letters (a, b, and c) on the bars represent significant differences (<span class="html-italic">p</span> &lt; 0.05), data are means ± SEM (<span class="html-italic">n</span> = 8).</p>
Full article ">Figure 3
<p>Effect of Cd and Se supplementation on the antioxidant capacity in the liver (<b>A</b>), ovarian (<b>B</b>), follicular (<b>C</b>) and serum (<b>D</b>). Different letters (a, b, c, and d) on the bars represent significant differences (<span class="html-italic">p</span> &lt; 0.05), data are means ± SEM (<span class="html-italic">n</span> = 8).</p>
Full article ">Figure 4
<p>Effects of Cd and Se supplementation on serum and follicular hormones, ovarian and follicular deposition of Cd and Se, and follicular autophagy-related mRNA expression levels. (<b>A</b>) E2 levels in the serum; (<b>B</b>) P4 levels in the serum; (<b>C</b>) E2 levels in the follicles; (<b>D</b>) P4 levels in the follicles; (<b>E</b>) ovarian Cd deposition; (<b>F</b>) follicular Cd deposition; (<b>G</b>) ovarian Se deposition; (<b>H</b>) follicular Se deposition; (<b>I</b>) follicular autophagy-related mRNA expression levels. Different letters (a, b and c) on the bars represent significant differences (<span class="html-italic">p</span> &lt; 0.05), data are means ± SEM (<span class="html-italic">n</span> = 8).</p>
Full article ">Figure 5
<p>Effects of Cd and Se supplementation on ultrastructure of granulosa cell layer of follicles. (<b>A</b>) The CON; (<b>B</b>) Se supplementation; (<b>C</b>) Cd+Se supplementation; (<b>D</b>,<b>E</b>) Cd treatment. The red box and lines in the first picture of (<b>D</b>) indicate that the next picture is an enlarged part of the boxed area. Blue arrows represent autophagosomes, yellow arrows represent lipid droplets, red arrows represent mitochondria that have swollen, the green arrow represents mitochondrial autophagy, and orange arrows represent apoptosis. N, nucleus, Mi, mitochondria, RER, rough endoplasmic reticulum.</p>
Full article ">
18 pages, 3068 KiB  
Article
Manganese Exposure Enhances the Release of Misfolded α-Synuclein via Exosomes by Impairing Endosomal Trafficking and Protein Degradation Mechanisms
by Dharmin Rokad, Dilshan S. Harischandra, Manikandan Samidurai, Yuan-Teng Chang, Jie Luo, Vivek Lawana, Souvarish Sarkar, Bharathi N. Palanisamy, Sireesha Manne, Dongsuk Kim, Gary Zenitsky, Huajun Jin, Vellareddy Anantharam, Auriel Willette, Arthi Kanthasamy and Anumantha G. Kanthasamy
Int. J. Mol. Sci. 2024, 25(22), 12207; https://doi.org/10.3390/ijms252212207 - 14 Nov 2024
Viewed by 353
Abstract
Excessive exposure to manganese (Mn) increases the risk of chronic neurological diseases, including Parkinson’s disease (PD) and other related Parkinsonisms. Aggregated α-synuclein (αSyn), a hallmark of PD, can spread to neighboring cells by exosomal release from neurons. We previously discovered that Mn enhances [...] Read more.
Excessive exposure to manganese (Mn) increases the risk of chronic neurological diseases, including Parkinson’s disease (PD) and other related Parkinsonisms. Aggregated α-synuclein (αSyn), a hallmark of PD, can spread to neighboring cells by exosomal release from neurons. We previously discovered that Mn enhances its spread, triggering neuroinflammatory and neurodegenerative processes. To better understand the Mn-induced release of exosomal αSyn, we examined the effect of Mn on endosomal trafficking and misfolded protein degradation. Exposing MN9D dopaminergic neuronal cells stably expressing human wild-type (WT) αSyn to 300 μM Mn for 24 h significantly suppressed protein and mRNA expression of Rab11a, thereby downregulating endosomal recycling, forcing late endosomes to mature into multivesicular bodies (MVBs). Ectopic expression of WT Rab11a significantly mitigated exosome release, whereas ectopic mutant Rab11a (S25N) increased it. Our in vitro and in vivo studies reveal that Mn exposure upregulated (1) mRNA and protein levels of endosomal Rab27a, which mediates the fusion of MVBs with the plasma membrane; and (2) expression of the autophagosomal markers Beclin-1 and p62, but downregulated the lysosomal marker LAMP2, thereby impairing autophagolysosome formation as confirmed by LysoTracker, cathepsin, and acridine orange assays. Our novel findings demonstrate that Mn promotes the exosomal release of misfolded αSyn by impairing endosomal trafficking and protein degradation. Full article
Show Figures

Figure 1

Figure 1
<p>Generation of GFP-tagged MN9D dopaminergic cells stably expressing human αSyn and characterization of exosomes. (<b>A</b>) Immunofluorescence of stably expressed GFP-fused human αSyn (red) in MN9D_αSynGFP cells, and GFP fluorescence (green) in both vector (control) MN9D_EVGFP cells and human αSyn-expressing MN9D_αSynGFP cells. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar, 100 µm. (<b>B</b>) Nanoparticle tracking analysis showing the concentration of exosomes from MN9D_αSynGFP cells from vehicle-stimulated (red) and Mn-stimulated (black) cells. (<b>C</b>) Nanoparticle tracking analysis showing the size distribution of exosomes from MN9D_αSynGFP cells from vehicle- (red) and Mn-stimulated (blue) cells. (<b>D</b>) TEM image of exosomes secreted from MN9D_αSynGFP cells displays distinctive morphology. Scale bar, 200 nm. (<b>E</b>) Western blots of GFP-fused human αSyn in exosomes from MN9D_αSynGFP cells compared to MN9D_EVGFP cells relative to exosome-positive markers Flotillin-1 and AIP-1/Alix in both cell types. (<b>F</b>) Average Thioflavin T (ThT) fluorescence in exosome samples isolated from Mn-treated and untreated αSyn-expressing and EV_MN9D cells, showing more aggregated αSyn in Mn-treated αSyn-expressing cells. (<b>G</b>) Protein aggregation rates (PAR) in exosome samples. Each trace and dot represent the average of 4 technical replicates. Statistically significant differences are denoted as * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and **** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">n</span> = 2–9.</p>
Full article ">Figure 2
<p>Mn exposure downregulates Rab11a expression both in vitro and in vivo. (<b>A</b>) Western blot of Rab11a from MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>B</b>) Densitometry analysis of Rab11a in Mn-exposed and control MN9D_αSynGFP cells. (<b>C</b>) ICC analysis of MN9D_αSynGFP cells (Rab11a; red) with and without Mn exposure for 24 h. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar, 50 µm. (<b>D</b>) Quantitative analysis of Rab11a integrated raw density from immunofluorescence analysis. (<b>E</b>) RT-qPCR analysis of <span class="html-italic">Rab11a</span> mRNA expression in MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>F</b>) Schematic illustration of Mn exposure in mice (30 mg/kg, male Swiss Webster and C57BL/6 mice) via oral gavage for 30 d. (<b>G</b>) Western blot of Rab11a from substantia nigral tissues from vehicle-treated and Mn-treated mice (30 mg/kg, male Swiss Webster). (<b>H</b>) Densitometry of Rab11a from substantia nigral tissues of vehicle- and Mn-exposed mice. (<b>I</b>) Western blot of control MN9D_αSynGFP (Vec) cells compared to the ectopic expression of wild-type (WT) Rab11a and dominant negative (DN) mutant Rab11a, respectively, in MN9D_αSynGFP cells transfected with WT Rab11a and DN Rab11 plasmids. (<b>J</b>) Concentration of exosomes from control and Mn-stimulated MN9D_αSynGFP cells (Vec), MN9D_αSynGFP cells expressing WT Rab11a, and MN9D_αSynGFP cells expressing DN Rab11a. (<b>K</b>) Nanoparticle tracking analysis showing the size distribution of exosome samples from exosome count, control and Mn-stimulated MN9D_αSynGFP cells (Vec; blue), MN9D_αSynGFP cells expressing WT Rab11a (green) and MN9D_αSynGFP cells expressing DN Rab11a (orange). Each group is represented by the mean ± S.E.M. from ≥3 separate measurements from vehicle- or Mn-treated groups. Statistically significant differences are denoted as * <span class="html-italic">p</span> ≤ 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. <span class="html-italic">n</span> = 2–7.</p>
Full article ">Figure 3
<p>Mn exposure upregulates Rab27a expression both in vitro and in vivo. (<b>A</b>) Western blot of Rab27a in MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>B</b>) Densitometry of Rab27a in Mn-exposed and control MN9D_αSynGFP cells. (<b>C</b>) Immunofluorescence analysis of MN9D_αSynGFP cells (Rab27a; red) with and without Mn exposure for 24 h. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar, 50 µm. (<b>D</b>) Quantitative analysis of Rab27a integrated raw density from immunofluorescence analysis. (<b>E</b>) RT-qPCR analysis of <span class="html-italic">Rab27a</span> mRNA expression in MN9D_αSynGFP cells treated with and without Mn exposure for 24 h. (<b>F</b>) Western blot of Rab27a from substantia nigral tissues from vehicle- and Mn-treated mice (30 mg/kg for 30 days, male Swiss Webster). (<b>G</b>) Densitometry of Rab27a in substantia nigral tissues of vehicle- and Mn-exposed mice. Each group is represented by the mean ± S.E.M. from ≥6 separate measurements from vehicle- and Mn-treated groups. Statistically significant differences are denoted as * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. <span class="html-italic">n</span> = 4–10.</p>
Full article ">Figure 4
<p>Mn exposure impairs the autophagic/lysosomal system in MN9D_αSynGFP cells. (<b>A</b>) Western blot of Beclin 1, p62, and LAMP2 from MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>B</b>) Densitometry of Beclin 1 (<b>C</b>) p62 and (<b>D</b>) LAMP2 in vehicle- and Mn-exposed MN9D_αSynGFP cells. (<b>E</b>) Western blot of LC3B-I and II from MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>F</b>) Densitometry of LC3B-II in vehicle- and Mn-exposed MN9D_αSynGFP cells. (<b>G</b>) Immunofluorescence analysis of MN9D_αSynGFP cells (LAMP2; red) with and without Mn exposure for 24 h. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar, 50 µm. (<b>H</b>) Immunofluorescence analysis showing reduced LysoTracker intensity in MN9D_αSynGFP cells following Mn treatment. Scale bar, 50 µm. (<b>I</b>) Cathepsin assay showing increased activity following Mn treatment. (<b>J</b>) Acridine orange assay showing Mn exposure reduced signal intensities. (<b>K</b>) Western blot analysis of lysosomal marker LAMP2 and autophagic markers Beclin 1 and p62 in the substantia nigra of mice with and without Mn exposure (30 mg/kg for 30 days, male Swiss Webster). (<b>L</b>) Densitometry of Beclin 1, (<b>M</b>) p62, and (<b>N</b>) LAMP2 in the substantia nigra of mice with and without Mn exposure. Statistically significant differences are denoted as * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. <span class="html-italic">n</span> = 3–9.</p>
Full article ">Figure 5
<p>Mn exposure downregulates VPS35 expression in vitro. (<b>A</b>) Western blot of VPS35 from MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>B</b>) Densitometry analysis of VPS35 in Mn-exposed and control MN9D_αSynGFP cells. (<b>C</b>) ICC analysis of MN9D_αSynGFP cells (VPS35; red) with and without Mn exposure for 24 h. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar 50 µm. (<b>D</b>) Quantitative analysis of VPS35 integrated raw density from immunofluorescence analysis. (<b>E</b>) RT-qPCR analysis of VPS35 mRNA expression in MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>F</b>) Western blot analysis of VPS35 in the substantia nigra of mice with and without Mn exposure (30 mg/kg for 30 days, male Swiss Webster). (<b>G</b>) Densitometry of VPS35 in the substantia nigra of mice with and without Mn exposure. Statistically significant differences are denoted as ** <span class="html-italic">p</span> &lt; 0.01. <span class="html-italic">n</span> = 2–8.</p>
Full article ">Figure 6
<p>Schematic representation of Mn’s potential to disrupt the endosomal trafficking and lysosomal pathway. Under normal, stress-free conditions, early endosomes form and are converted into late endosomes. Late endosomes can either be recycled back to the plasma membrane, as mediated by Rab11a, or they can mature into multivesicular bodies. MVBs either fuse with the plasma membrane, as mediated by Rab27a, to release exosomes, or MVBs can also merge with a lysosome to degrade its contents through the autophagolysosome pathway. Our study supports a model in which, when cells are exposed to Mn, key mediators of this endosomal trafficking pathway, such as Rab11a and Rab27a, as well as the lysosomal pathway, become dysregulated, leading to increased exosome release. Rab5 and Rab7 are involved in the maturation of early endosomes to late endosomes. Certain graphical representations in the above schematic depict proteins, such as divalent metal-ion transporter-1 (DMT-1), which are involved in Mn transport and homeostasis and are shown here in the shape of ion channels. The skull markings represent toxic conditions from Mn exposure.</p>
Full article ">
18 pages, 6525 KiB  
Article
SIRT1-Dependent Neuroprotection by Resveratrol in TOCP-Induced Spinal Cord Injury: Modulation of ER Stress and Autophagic Flux
by Xiangsheng Tian, Yiquan Ou, Shengyuan Shi, Qiuhua Zhou, Sihong Long, Yao Xiang, Weichao Zhao and Dingxin Long
Toxics 2024, 12(11), 810; https://doi.org/10.3390/toxics12110810 - 11 Nov 2024
Viewed by 399
Abstract
This study explores the neuroprotective effects of resveratrol (Resv) against tri-o-cresyl phosphate (TOCP)-induced neurotoxicity in the spinal cord of adult hens. It is well documented that TOCP exposure causes significant neurodegeneration via mechanisms that involve endoplasmic reticulum (ER) stress and impaired autophagy. In [...] Read more.
This study explores the neuroprotective effects of resveratrol (Resv) against tri-o-cresyl phosphate (TOCP)-induced neurotoxicity in the spinal cord of adult hens. It is well documented that TOCP exposure causes significant neurodegeneration via mechanisms that involve endoplasmic reticulum (ER) stress and impaired autophagy. In this experiment, adult hens were assigned to one of four groups: Control, Resv, TOCP, and TOCP + Resv. The spinal cord tissues were examined through transmission electron microscopy, hematoxylin and eosin (HE) staining, Nissl staining, and Western blotting to evaluate key proteins associated with ER stress and autophagy. Additionally, RT-qPCR and immunofluorescence were employed to measure sirtuin1 (SIRT1) expression. The findings revealed that TOCP induced severe ultrastructural damage, including disrupted myelin sheaths, dilated ER, and extensive neurodegeneration, as confirmed by histological evaluations. The expression levels of GRP78, p-PERK, p-eIF2α, ATF4, CHOP, Beclin-1, P62, and LC3-II were also significantly elevated by TOCP. However, Resv treatment markedly attenuated these pathological changes by reducing ER stress, restoring autophagic flux, and upregulating SIRT1 expression, preserving spinal cord integrity. These results indicate that Resv can effectively counteract TOCP-induced neurotoxicity by modulating ER stress and autophagy, underscoring its potential as a therapeutic agent for neuroprotection. Full article
(This article belongs to the Section Neurotoxicity)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of TOCP and resveratrol. (<b>A</b>) TOCP (PubChem CID: 86003503). (<b>B</b>) Resveratrol (PubChem CID: 5056).</p>
Full article ">Figure 2
<p>Schematic diagram of the experimental procedures.</p>
Full article ">Figure 3
<p>Effect of TOCP on ultrastructure of spinal cord in hens. Electron microscopy of spinal cord. Scale bar = 1.0 µm. The Control group represents a healthy nerve structure, the TOCP group indicates neurotoxicity, as evidenced by the disrupted myelin sheaths (blue arrow), the mitochondria exhibited disrupted cristae (red arrow) and dilated and disorganized ER (yellow arrow).</p>
Full article ">Figure 4
<p>Resv can improve the spinal cord structural injury caused by TOCP. (<b>A</b>): HE staining of spinal cord (scale bar = 100 µm) and enlarged view of some areas (scale bar = 40 µm). (<b>B</b>): Histopathological score (nuclear pyknosis), <span class="html-italic">n</span> = 3. (<b>C</b>): Histopathological score (vacuolation), <span class="html-italic">n</span> = 3. Data are presented as the mean ± SEM. Statistical analyses were performed with one-way ANOVA. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Resv has a protective effect on TOCP-induced neuronal injury. (<b>A</b>) Nissl staining of spinal cord. Scale bar = 100 µm. (<b>B</b>) Quantitative analysis of neuronal density (neurons/mm<sup>2</sup>), <span class="html-italic">n</span> = 3. Data are presented as the mean ± SEM. Statistical analyses were performed with one-way ANOVA. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, ns = not significant.</p>
Full article ">Figure 6
<p>Resv can relieve ER stress induced by TOCP. (<b>A</b>) Western blotting analysis of ER stress-related proteins including GRP78, P-PERK, P-eIF2α, ATF4, and CHOP in spinal cord tissue. (<b>B</b>–<b>F</b>) Image J 1.50d was applied to analyze the gray scale of the protein bands of GRP78 (<b>B</b>), p-PERK (<b>C</b>), p-eIF2α (<b>D</b>), ATF4 (<b>E</b>) and CHOP (<b>F</b>) (<span class="html-italic">n</span> = 3). (<b>G</b>–<b>K</b>) The relative mRNA levels of GRP78 (<b>G</b>), PERK (<b>H</b>), eIF2α (<b>I</b>), ATF4 (<b>J</b>), and CHOP (<b>K</b>) in spinal cord tissue were detected using RT-qPCR (<span class="html-italic">n</span> = 4). Data are presented as the mean ± SEM. Statistical analyses were performed with one-way ANOVA. * <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, ns = not significant.</p>
Full article ">Figure 7
<p>Resv modulates abnormal autophagy flux induced by TOCP. (<b>A</b>) Western blotting analysis of autophagy-related proteins including LC3-II, Beclin-1 and P62 in spinal cord tissue. (<b>B</b>–<b>D</b>) Image J was applied to analyze the gray scale of the protein bands of LC3-II (<b>B</b>), Beclin-1 (<b>C</b>), and P62 (<b>D</b>) (<span class="html-italic">n</span> = 3). (<b>E</b>,<b>F</b>) The relative mRNA levels of Beclin-1 (<b>E</b>), and P62 (<b>F</b>) in spinal cord tissue were detected using RT-qPCR (<span class="html-italic">n</span> = 4). Data are presented as the mean ± SEM. Statistical analyses were performed with one-way ANOVA. * <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, ns = not significant.</p>
Full article ">Figure 8
<p>Resv can activate SIRT1 and reduce the inhibition of SIRT1 by TOCP. (<b>A</b>) Western blotting analysis of SIRT1 protein in spinal cord tissue. (<b>B</b>) Image J was applied to analyze the gray scale of the protein bands of SIRT1 (<span class="html-italic">n</span> = 3). (<b>C</b>) The relative mRNA levels of SIRT1 in spinal cord tissue were detected using RT-qPCR (<span class="html-italic">n</span> = 4). Data are presented as the mean ± SEM. Statistical analyses were performed with one-way ANOVA. * <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, ns = not significant.</p>
Full article ">Figure 9
<p>The expression and localization of SIRT1 in spinal cord were detected by immunofluorescence. (<b>A</b>) Immunofluorescence staining for SIRT1. Scale bar = 50 µm. (<b>B</b>) Quantitative analysis of SIRT1 fluorescence intensity (<span class="html-italic">n</span> = 3). Data are presented as the mean ± SEM. Statistical analyses were performed with one-way ANOVA. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, ns = not significant.</p>
Full article ">
59 pages, 4829 KiB  
Article
FAAH Inhibition Counteracts Neuroinflammation via Autophagy Recovery in AD Models
by Federica Armeli, Roberto Coccurello, Giacomo Giacovazzo, Beatrice Mengoni, Ilaria Paoletti, Sergio Oddi, Mauro Maccarrone and Rita Businaro
Int. J. Mol. Sci. 2024, 25(22), 12044; https://doi.org/10.3390/ijms252212044 - 9 Nov 2024
Viewed by 329
Abstract
Endocannabinoids have attracted great interest for their ability to counteract the neuroinflammation underlying Alzheimer’s disease (AD). Our study aimed at evaluating whether this activity was also due to a rebalance of autophagic mechanisms in cellular and animal models of AD. We supplied URB597, [...] Read more.
Endocannabinoids have attracted great interest for their ability to counteract the neuroinflammation underlying Alzheimer’s disease (AD). Our study aimed at evaluating whether this activity was also due to a rebalance of autophagic mechanisms in cellular and animal models of AD. We supplied URB597, an inhibitor of Fatty-Acid Amide Hydrolase (FAAH), the degradation enzyme of anandamide, to microglial cultures treated with Aβ25-35, and to Tg2576 transgenic mice, thus increasing the endocannabinoid tone. The addition of URB597 did not alter cell viability and induced microglia polarization toward an anti-inflammatory phenotype, as shown by the modulation of pro- and anti-inflammatory cytokines, as well as M1 and M2 markers; moreover microglia, after URB597 treatment released higher levels of Bdnf and Nrf2, confirming the protective role underlying endocannabinoids increase, as shown by RT-PCR and immunofluorescence experiments. We assessed the number and area of amyloid plaques in animals administered with URB597 compared to untreated animals and the expression of autophagy key markers in the hippocampus and prefrontal cortex from both groups of mice, via immunohistochemistry and ELISA. After URB597 supply, we detected a reduction in the number and areas of amyloid plaques, as detected by Congo Red staining and a reshaping of microglia activation as shown by M1 and M2 markers’ modulation. URB597 administration restored autophagy in Tg2576 mice via an increase in BECN1 (Beclin1), ATG7 (Autophagy Related 7), LC3 (light chain 3) and SQSTM1/p62 (sequestrome 1) as well as via the activation of the ULK1 (Unc-51 Like Autophagy Activating Kinase 1) signaling pathway, suggesting that it targets mTOR/ULK1-dependent autophagy pathway. The potential of endocannabinoids to rebalance autophagy machinery may be considered as a new perspective for therapeutic intervention in AD. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

Figure 1
<p>The mRNA expression of different inflammatory cytokines. mRNA of IL-1 β, IL-6, <span class="html-italic">Tgf-β</span>, and IL-10, monitored by qPCR and normalized to 18 S ribosome subunit. Data are shown as mean ± SD from three independent experiments performed in triplicate. Expression profiles were determined using the 2<sup>−ΔΔCT</sup> method. * <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 2
<p>mRNA expression of <span class="html-italic">Nrf2</span> was evaluated by qRT-PCR at 3 h and 6 h. Data are shown as mean ± SD from three independent experiments performed in triplicate. Expression profiles were determined using the 2<sup>−ΔΔCT</sup> method. ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>mRNA expression of <span class="html-italic">Bdnf</span> was evaluated by qRT-PCR at 1 h and 6 h. Data are shown as mean ± SD from three independent experiments performed in triplicate. Expression profiles were determined using the 2<sup>−ΔΔCT</sup> method. * <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.</p>
Full article ">Figure 4
<p>Immunohistochemical analysis (Ematoxillin and DAB chromogen) in sagittal sections of PFC and hippocampus. (<b>A</b>) shows decreased iNOS marking in Tg2576 mice treated with URB597; (<b>B</b>) shows increased ARG-1 marking in Tg2576 mice treated with URB597; these results were confirmed by quantification shown in graphs. Analysis was performed by considering 4 10× magnification fields for both hippocampus and cortex, and values were expressed as mean ± SD of total positive cells expressed in both brain regions from 6 independent experiments for each experimental group using unpaired Student <span class="html-italic">t</span> test. Significance levels were denoted as follows: * <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 5
<p>Congo Red analysis in sagittal sections including either vehicle or URB597-treated Tg2576 mice. Images show amyloid plaques. Analysis was performed by considering 4 magnification fields; area was expressed in μm<sup>2</sup> and values were expressed as mean ± SD of total positive cells expressed in both brain regions from 6 independent experiments for each experimental group, using unpaired Student <span class="html-italic">t</span> test. Significance levels were marked as follows: *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001. Magnification 10× and 40×.</p>
Full article ">Figure 6
<p>Analysis of mRNA expression of factors involved in the autophagic process. (<b>A</b>) mRNA expressions were evaluated by qRT-PCR at 6 h and 24 h of <span class="html-italic">Atg7</span>, BECN1, <span class="html-italic">Lc3</span>, and <span class="html-italic">SQSTM1/p62</span>. Data are shown as the mean ± SD from three independent experiments performed in triplicate. Expression profiles were determined using the 2<sup>−ΔΔCT</sup> method. Significance levels were denoted as follows: * <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. (<b>B</b>) Immunohistochemical analysis (Ematoxillin and DAB chromogen) in sagittal sections of vehicle-treated WT and Tg2576 experimental groups and WT and Tg2576 treated with URB597. The images show ATG7- and BECN1-positive cells in PFC and hippocampus. Analysis was performed by considering 4 10× magnification fields for both hippocampus and PFC, and the values were expressed as the mean ± SD of the total positive cells expressed in both of the brain regions from 6 independent experiments for each experimental group using an unpaired Student <span class="html-italic">t</span> test. Significance levels are indicated as follows: * <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. (<b>C</b>) Immunohistochemical analysis (Ematoxillin and DAB chromogen) in sagittal sections of vehicle-treated WT and Tg2576 groups and WT and Tg2576 administered with URB597. The images show the LC3I-II- and SQSTM1/p62-positive cells in the PFC and hippocampus. Analysis was performed by considering four 10× magnification fields from the hippocampus and PFC, and the values were expressed as the mean ± SD of the total positive cells expressed in both of the brain regions from 6 independent experiments for each experimental group, using an unpaired Student <span class="html-italic">t</span> test. Significance levels are indicated as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6 Cont.
<p>Analysis of mRNA expression of factors involved in the autophagic process. (<b>A</b>) mRNA expressions were evaluated by qRT-PCR at 6 h and 24 h of <span class="html-italic">Atg7</span>, BECN1, <span class="html-italic">Lc3</span>, and <span class="html-italic">SQSTM1/p62</span>. Data are shown as the mean ± SD from three independent experiments performed in triplicate. Expression profiles were determined using the 2<sup>−ΔΔCT</sup> method. Significance levels were denoted as follows: * <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. (<b>B</b>) Immunohistochemical analysis (Ematoxillin and DAB chromogen) in sagittal sections of vehicle-treated WT and Tg2576 experimental groups and WT and Tg2576 treated with URB597. The images show ATG7- and BECN1-positive cells in PFC and hippocampus. Analysis was performed by considering 4 10× magnification fields for both hippocampus and PFC, and the values were expressed as the mean ± SD of the total positive cells expressed in both of the brain regions from 6 independent experiments for each experimental group using an unpaired Student <span class="html-italic">t</span> test. Significance levels are indicated as follows: * <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. (<b>C</b>) Immunohistochemical analysis (Ematoxillin and DAB chromogen) in sagittal sections of vehicle-treated WT and Tg2576 groups and WT and Tg2576 administered with URB597. The images show the LC3I-II- and SQSTM1/p62-positive cells in the PFC and hippocampus. Analysis was performed by considering four 10× magnification fields from the hippocampus and PFC, and the values were expressed as the mean ± SD of the total positive cells expressed in both of the brain regions from 6 independent experiments for each experimental group, using an unpaired Student <span class="html-italic">t</span> test. Significance levels are indicated as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>LC3-II ELISA assay on homogenates of PFC and hippocampus from untreated and vehicle/URB597-treated Tg2576 and WT mice. Data are shown as mean ± SD from three independent experiments performed in duplicate using unpaired Student <span class="html-italic">t</span> test. Significance levels were denoted as follows: ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Immunohistochemical analysis (Ematoxillin and DAB chromogen) in sagittal sections of vehicle-treated WT and Tg2576 experimental groups and WT and Tg2576 treated with URB597. (<b>A</b>) The images show mTOR positive cells in the PFC and the hippocampus. Analysis was performed by considering 4 10× magnification fields for both hippocampus and PFC, and the values were expressed as the mean ± SD of the total positive cells expressed in both the brain regions from 6 independent experiments for each experimental group using an unpaired Student <span class="html-italic">t</span> test. Significance levels were denoted as follows: * <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. (<b>B</b>) The images show ULK1-positive cells in the PFC and the hippocampus. Analysis was performed by considering 4 10× magnification fields for both the hippocampus and the PFC, and the values were expressed as mean ± SD of the total positive cells expressed in both of the brain regions from 6 independent experiments for each experimental group using an unpaired Student <span class="html-italic">t</span> test. Significance levels were denoted as follows: * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
8 pages, 442 KiB  
Brief Report
Quercetin’s Potential in MASLD: Investigating the Role of Autophagy and Key Molecular Pathways in Liver Steatosis and Inflammation
by Ioannis Katsaros, Maria Sotiropoulou, Michail Vailas, Emmanouil Ioannis Kapetanakis, Georgia Valsami, Alexandra Tsaroucha and Dimitrios Schizas
Nutrients 2024, 16(22), 3789; https://doi.org/10.3390/nu16223789 - 5 Nov 2024
Viewed by 527
Abstract
Metabolic dysfunction-associated fatty liver disease (MASLD) is a widespread liver disorder characterized by excessive fat accumulation in the liver, commonly associated with metabolic syndrome components such as obesity, diabetes, and dyslipidemia. With a global prevalence of up to 30%, MASLD is projected to [...] Read more.
Metabolic dysfunction-associated fatty liver disease (MASLD) is a widespread liver disorder characterized by excessive fat accumulation in the liver, commonly associated with metabolic syndrome components such as obesity, diabetes, and dyslipidemia. With a global prevalence of up to 30%, MASLD is projected to affect over 100 million people in the U.S. and 20 million in Europe by 2030. The disease ranges from Steatotic Lived Disease (SLD) to more severe forms like metabolic dysfunction-associated steatohepatitis (MASH), which can progress to cirrhosis and hepatocellular carcinoma. Autophagy, a cellular process crucial for lipid metabolism and homeostasis, is often impaired in MASLD, leading to increased hepatic lipid accumulation and inflammation. Key autophagy-related proteins, such as Beclin1, LC3A, SQSTM1 (p62), CD36, and Perilipin 3, play significant roles in regulating this process. Disruption in these proteins contributes to the pathogenesis of MASLD. Quercetin, a natural polyphenolic flavonoid with antioxidant and anti-inflammatory properties, has promising results in mitigating MASLD. It may reduce hepatic lipid accumulation, improve mitochondrial function, and enhance autophagy. However, further research is needed to elucidate its mechanisms and validate its therapeutic potential in clinical settings. This underscores the need for continued investigation into autophagy and novel treatments for MASLD. Full article
(This article belongs to the Special Issue Nutrition in the Liver Damage)
Show Figures

Figure 1

Figure 1
<p>The effect of Quercetin on MASLD (metabolic dysfunction-associated fatty liver disease).</p>
Full article ">
13 pages, 2838 KiB  
Article
Whole Blood Transcriptome Analysis in Dairy Ewes Fed a Dietary Grape Pomace Supplementation
by Andrea Ianni, Francesca Bennato, Camillo Martino, Maria Antonietta Saletti, Francesco Pomilio and Giuseppe Martino
Vet. Sci. 2024, 11(11), 536; https://doi.org/10.3390/vetsci11110536 - 1 Nov 2024
Viewed by 588
Abstract
The present study aims to evaluate the effect of a dietary supplementation with 10% grape pomace (GP) on the whole blood transcriptome of lactating ewes. By applying a log2FC higher than 0.5 or lower than −0.5 and a false discovery rate [...] Read more.
The present study aims to evaluate the effect of a dietary supplementation with 10% grape pomace (GP) on the whole blood transcriptome of lactating ewes. By applying a log2FC higher than 0.5 or lower than −0.5 and a false discovery rate (FDR) <0.05, the down-regulation of genes coding for plexin C1, ethanolamine kinase 1, tax1-binding protein 1, transmembrane 9 superfamily member 2, and Beclin-1 was observed in animals that received the dietary supplementation. This aspect was also accompanied by a reduction in the blood activity of matrix metalloproteinase 9 (MMP-9; p < 0.05), a gelatinase commonly involved in both acute and chronic pathological events. The ELISA test on other factors involved in inflammatory processes, interleukin 1 (IL-1) and tumor necrosis factor α (TNF-α), as well as in the antioxidant response, glutathione peroxidase (GPx), and catalase (CAT), did not reveal any significant changes (p > 0.05). Overall, the introduction of GP in the diet of ewes gave indications of greater efficacy in preserving animal welfare, with interesting cues regarding the valorization of a by-product with a high biological value. Full article
(This article belongs to the Special Issue Effects of Nutrition on Ruminants Production Performance and Health)
Show Figures

Figure 1

Figure 1
<p>Principal component analysis (PCA) of genes expressed in whole blood samples obtained from ewes fed a standard diet (control group; CG) and the grape pomace dietary supplementation (experimental group; EG).</p>
Full article ">Figure 2
<p>Interaction network of proteins corresponding to the differentially expressed genes (DEGs) identified through the blood transcriptome characterization. By making reference to the <span class="html-italic">Ovis aries</span> database, the software STRING 12.0 was effective in building the interaction network for each protein identified. Interactions are reported with different colors: cyan is from curated databases, magenta is experimentally determined, dark green is gene neighborhood, red is gene fusion, blue is gene co-occurrence, light green is text-mining, black is co-expression, and light blue is protein homology.</p>
Full article ">Figure 3
<p>ELISA evaluation of the activity of two enzymes of the antioxidant response, glutathione peroxidase (GPx) and catalase (CAT), and the concentration of two pro-inflammatory cytokines, inteleukin-1 (IL-1) and tumor necrosis factor-α (TNF-α). The analysis was performed on blood serum samples obtained from ewes fed a standard diet (CG; n = 5) and the grape pomace dietary supplementation (EG; n = 5).</p>
Full article ">Figure 4
<p>Representative zymography on serum blood samples obtained from ewes fed a standard diet (CG; lanes 1–3; n = 5) and the grape pomace dietary supplementation (EG; lanes 4–6; n = 5). In the histogram, the data are reported as a percentage of density of the band of interest, referring to the total calculated in the reference lane. The evaluation was effective in highlighting the activity of matrix metalloproteinases 2 (MMP-2) and 9 (MMP-9).</p>
Full article ">
19 pages, 4902 KiB  
Article
HSP90 Enhances Mitophagy to Improve the Resistance of Car-Diomyocytes to Heat Stress in Wenchang Chickens
by Jiachen Shi, Zeping Ji, Xu Yao, Yujie Yao, Chengyun Li, Qijun Liang and Xiaohui Zhang
Int. J. Mol. Sci. 2024, 25(21), 11695; https://doi.org/10.3390/ijms252111695 - 30 Oct 2024
Viewed by 491
Abstract
Heat shock protein 90 (HSP90) is recognized for its protective effects against heat stress damage; however, the specific functions and underlying molecular mechanisms of HSP90 in heat-stressed cardiomyocytes remain largely unexplored, particularly in tropical species. In our study, Wenchang chickens (WCCs) were classified [...] Read more.
Heat shock protein 90 (HSP90) is recognized for its protective effects against heat stress damage; however, the specific functions and underlying molecular mechanisms of HSP90 in heat-stressed cardiomyocytes remain largely unexplored, particularly in tropical species. In our study, Wenchang chickens (WCCs) were classified into two groups: the heat stress survival (HSS) group and the heat stress death (HSD) group, based on their survival following exposure to heat stress. Heat stress resulted in significant cardiomyocyte damage, mitochondrial dysfunction, and apoptosis in the HSD group, while the damage was less pronounced in the HSS group. We further validated these findings in primary cardiomyocytes derived from Wenchang chickens (PCWs). Additionally, heat stress was found to upregulate Pink1/Parkin-mediated mitophagy, which was accompanied by an increase in HSP90 expression in both cardiomyocytes and PCWs. Our results demonstrated that HSP90 overexpression enhances PINK1/Parkin-mediated mitophagy, ultimately inhibiting apoptosis and oxidative stress in heat-stressed PCWs. However, the application of Geldanamycin (GA) reversed these effects. Notably, we discovered that HSP90 interacts with Beclin-1 through mitochondrial translocation and directly regulates mitophagy levels in PCWs. In summary, we have elucidated a novel role for HSP90 and mitophagy in regulating heat stress-induced acute cardiomyocyte injury. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Establishment of heat-stressed cardiomyocyte models in vitro and in vivo in WCCs. (<b>A</b>) Myocardial tissues in WCCs were stained by H&amp;E staining. Scale bars: 2 μm (<b>top</b>) and 500 nm (<b>bottom</b>), n = 3. Vertical yellow arrow: swelling of cardiac fibers, horizontal yellow arrow: myocardial fiber rupture. (<b>B</b>) The levels of CK-MB in the serum of heat-stressed WCCs were detected. Data are presented as the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>C</b>) The levels of LDH in the myocardial tissue of heat-stressed WCCs were detected. Data are presented as the mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>D</b>) Heat-stressed PCWs for 3 h (HS) were stained by H&amp;E (scale bar: 20 μm). In each group, the image on the right is a partial enlargement of the image on the left. (<b>E</b>) The levels of CK-MB in the supernatant of heat-stressed PCWs cultures were measured. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>F</b>) The levels of LDH in the supernatant of heat-stressed PCWs cultures were measured (n = 3). The differences in the data of cells heat-stressed for different times vs. those heat-stressed for 0 h are indicated by * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01, n = 3.</p>
Full article ">Figure 2
<p>Heat stress induces mitochondrial dysfunction in cardiomyocytes and PCWs, leading to oxidative stress. (<b>A</b>) TEM images showing mitochondrial morphology and autophagosomes in heat-stressed cardiomyocytes. M indicates mitochondria; blue arrows point to mitochondrial autophagosomes. Scale bars: 2 μm (<b>top</b>) and 500 nm (<b>bottom</b>). (<b>B</b>) The mitochondrial membrane potential (MMP) in the myocardial tissue of heat-stressed WCCs were detected; data are presented as the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. The levels of ROS (<b>C</b>), GSH (<b>D</b>), and MDA (<b>E</b>) in the myocardial tissue of heat-stressed WCCs were detected. Data are presented as the mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>F</b>) TEM images showing the presence of abnormally shaped mitochondria and autophagosomes in the Con group and heat-stressed PCWs for 3 h (HS). Blue arrows point to autophagosomes, and yellow arrows point to mitochondria with disrupted cristae. Scale bars: 2 μm (<b>top</b>) and 500 nm (<b>bottom</b>), n = 3. (<b>G</b>) PCWs were treated with heat stress for 3 h and then treated with a JC-1 probe and observed under a fluorescence microscope. The representative images for each condition are shown. Scale bars: 20 μm. Red fluorescence represents JC-1 aggregates, and green fluorescence represents JC-1 monomers, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>H</b>) The ROS levels of PCWs were determined after exposure to heat stress for 3 h. * <span class="html-italic">p</span> &lt; 0.05, n = 3. The GSH (<b>I</b>) and MDA (<b>J</b>) levels of PCWs were determined after exposure to heat stress for different times. The differences in the data of cells heat-stressed for different times vs. those heat-stressed for 0 h are indicated by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3.</p>
Full article ">Figure 3
<p>Heat stress induces apoptosis in cardiomyocytes and PCWs. (<b>A</b>) TUNEL staining was performed in the myocardial tissue of WCCs following treatment with heat stress. Apoptosis was determined by TUNEL staining (red) and the nuclei were stained with DAPI (blue). TUNEL positive ratio is defined as the number of TUNEL positive cells divided by the total number of cells. Values are presented as the mean ± SEM of three individual experiments, Scale bars: 50 μm, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>B</b>) Measurement of protein expression of apoptosis-related proteins Bcl-2, Bax, Caspase-3, and cleaved caspase-3 in myocardial tissues of WCCs treated with heat stress by Western blot analysis, normalized to GAPDH. Measurement data are expressed as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>C</b>) Hoechst 33342 staining was performed in heat-stressed PCWs for 3 h, scale bars: 50 μm, * <span class="html-italic">p</span> &lt; 0.05, n = 3. (<b>D</b>) After exposure to heat stress for different times, Bcl-2, Bax, Caspase-3, and Cleaved caspase-3 in whole cells were detected via Western blot analysis with the indicated antibodies. Representative bands are shown. The differences in the data of cells heat-stressed for different times vs. those heat-stressed for 0 h are indicated by * <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>Heat stress induces mitophagy and high HSP90 expression in cardiomyocytes and PCWs. (<b>A</b>) Measurement of protein expression of mitophagy-related proteins Pink1, Parkin, LC3, Beclin-1, and P62 in myocardial tissues of WCCs treated with heat stress by Western blot analysis, normalized to GAPDH. Measurement data are expressed as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>B</b>) After exposure to heat stress for different times, Pink1, Parkin, LC3, Beclin-1, and P62 in whole cells were detected via Western blot analysis with the indicated antibodies. Representative bands are shown. The differences in the data of cells heat-stressed for different times vs. those heat-stressed for 0 h are indicated by * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01. (<b>C</b>) Immunofluorescence staining of LC3 dots (LC3, green) and Mito Tracker (Mito, red) in heat-stressed PCWs for 3 h. Scale bars: 10 μm, n = 3. (<b>D</b>) Measurement of protein expression of HSP90 in myocardial tissues of WCCs treated with heat stress by Western blot analysis, normalized to GAPDH. Measurement data are expressed as mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>E</b>) After exposure to heat stress for different times, HSP90 in whole cells were detected via Western blot analysis with the indicated antibodies. Representative bands are shown. The differences in the data of cells heat-stressed for different times vs. those heat-stressed for 0 h are indicated by * <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>HSP90 overexpression reduces heat stress-induced PCWs damage and apoptosis, while enhancing mitophagy. (<b>A</b>,<b>B</b>) The levels of CK-MB and LDH in PCWs with or without HSP90 overexpression after heat stress. Measurement data are expressed as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>C</b>,<b>D</b>) The levels of ROS and GSH in PCWs with or without HSP90 overexpression after heat stress. Measurement data are expressed as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>E</b>) HSP90, Pink1, Parkin, LC3, Beclin-1, and P62 in PCWs with or without HSP90 overexpression after heat stress were detected via Western blot analysis with the indicated antibodies. Representative bands are shown. Data are expressed as the means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>F</b>) Bcl-2, Bax, Caspase-3, and cleaved caspase-3 in PCWs with or without HSP90 overexpression after heat stress were detected via Western blot analysis with the indicated antibodies. Representative bands are shown. Data are expressed as the means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3.</p>
Full article ">Figure 6
<p>Inhibition of HSP90 function leads to increased apoptosis and damage, with downregulated mitophagy in heat-stressed PCWs. (<b>A</b>,<b>B</b>) The levels of CK-MB and LDH in PCWs treated with or without GA after heat stress. Measurement data are expressed as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>C</b>,<b>D</b>) The levels of ROS and GSH in PCWs treated with or without GA after heat stress. Measurement data are expressed as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>E</b>) HSP90, Pink1, Parkin, LC3, Beclin-1, and P62 in PCWs treated with or without GA after heat stress were detected via Western blot analysis with the indicated antibodies. Representative bands are shown. Data are expressed as the means ± SEM * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3. (<b>F</b>) Bcl-2, Bax, caspase-3, and cleaved-caspase-3 in PCWs treated with or without GA after heat stress were detected via Western blot analysis with the indicated antibodies. Representative bands are shown. Data are expressed as the means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 3.</p>
Full article ">Figure 7
<p>Effect of heat stress on the interaction between HSP90 and Beclin-1. (<b>A</b>) Coimmunoprecipitation (Co-IP) of HSP90 and Beclin-1 in the myocardial tissue of WCCs following treatment with heat stress. (<b>B</b>) Immunofluorescence staining of HSP90 (HSP90, green) and Beclin-1 (Beclin-1, red) in PCWs heat-stressed for 3 h, scale bars: 10 μm, n = 3. (<b>C</b>) Immunofluorescence staining of HSP90 (HSP90, green) and Mito Tracker (Mito, red) in PCWs heat-stressed for 3 h, scale bars: 10 μm, n = 3.</p>
Full article ">
12 pages, 2847 KiB  
Article
Anti-Inflammatory and Autophagy Activation Effects of 7-Methylsulfonylheptyl Isothiocyanate Could Suppress Skin Aging: In Vitro Evidence
by Yeong Hee Cho and Jung Eun Park
Antioxidants 2024, 13(11), 1282; https://doi.org/10.3390/antiox13111282 - 23 Oct 2024
Viewed by 562
Abstract
Skin inflammation, characterized by redness, swelling, and discomfort, is exacerbated by oxidative stress, where compounds like 7-methylsulfonylheptyl isothiocyanate (7-MSI) from cruciferous plants exhibit promising antioxidant and anti-inflammatory properties, though their effects on skin aging and underlying mechanisms involving the NLRP3 inflammasome and autophagy [...] Read more.
Skin inflammation, characterized by redness, swelling, and discomfort, is exacerbated by oxidative stress, where compounds like 7-methylsulfonylheptyl isothiocyanate (7-MSI) from cruciferous plants exhibit promising antioxidant and anti-inflammatory properties, though their effects on skin aging and underlying mechanisms involving the NLRP3 inflammasome and autophagy are not fully elucidated. NLRP3 is a crucial inflammasome involved in regulating inflammatory responses, and our study addresses its activation and associated physiological effects. Using biochemical assays such as ELISA, RT-qPCR, Western blotting, confocal microscopy, and RNA interference, we evaluated 7-MSI’s impact on cytokine production, protein expression, and genetic regulation in Raw 264.7 and RAW-ASC cells. 7-MSI significantly reduced TNF-α, IL-1β, IL-6, COX-2, and PGE transcription levels in LPS-stimulated Raw 264.7 cells, indicating potent anti-inflammatory effects. It also inhibited NF-κB signaling and NLRP3 inflammasome activity, demonstrating its role in preventing the nuclear translocation of NF-κB and reducing caspase-1 and IL-1β production. In terms of autophagy, 7-MSI enhanced the expression of Beclin-1, LC3, and Atg12 while reducing phospho-mTOR levels, suggesting an activation of autophagy. Moreover, it effectively decreased ROS production induced by LPS. The interaction between autophagy and inflammasome regulation was further confirmed through experiments showing that interference with autophagy-related genes altered the effects of 7-MSI on cytokine production. Collectively, this study demonstrates that 7-MSI promotes autophagy, including ROS removal, and to suppress inflammation, we suggest the potential use of 7-MSI as a skin care and disease treatment agent. Full article
Show Figures

Figure 1

Figure 1
<p>Structure of 7-MSI.</p>
Full article ">Figure 2
<p>Anti-inflammatory effect of 7-MSI. (<b>A</b>) Raw 264.7 cells were exposed to varying concentrations of 7-MSI (0-4 μg/mL) in the presence of LPS (1 μg/mL) for 1 h. TNF-α levels in the culture supernatants were quantified using a specific ELISA kit for TNF-α and were compared to a standard curve for accuracy. (<b>B</b>) Raw 264.7 cells were exposed to 7-MSI at concentrations of 0.5, 1, and 2 μg/mL, alongside LPS at 1 μg/mL, for a duration of 3 h. Total RNA was extracted, and RT-qPCR was performed using primers that specifically target IL-1β, IL-6, COX-2, and PGEs. (<b>C</b>) The histograms display the relative expression rates of IL-1β, IL-6, COX-2, and PGEs, calculated as the ratio of signal intensity to GAPDH. (<b>D</b>) RAW 264.7 cells were exposed to 7-MSI (1 μg/mL) alongside LPS (1 μg/mL) for durations of 0.5, 1, and 3 h. Protein expression levels of IκBα, p-IκBα, and GAPDH were assessed through Western blotting. (<b>E</b>,<b>F</b>) The histograms illustrate the relative expression rates of IκBα and p-IκBα, normalized to the signal intensity of GAPDH. The data represent mean ± SD from triplicate Western blot experiments. Statistical significance was analyzed using Student’s <span class="html-italic">t</span>-test with the following significance levels: * <span class="html-italic">p</span> &lt; 0.00001; ** <span class="html-italic">p</span> &lt; 0.00005; *** <span class="html-italic">p</span> &lt; 0.0001; <sup><tt>ǂ</tt></sup> <span class="html-italic">p</span> &lt; 0.0005; <sup><tt>ǂǂ</tt></sup> <span class="html-italic">p</span> &lt; 0.001; <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05, compared to the “LPS only” group. (<b>G</b>) Cells were treated with LPS (1 μg/mL) alone for 1 h or in combination with 7-MSI (1 μg/mL) for 1 h, and then stained with a fluorescein-conjugated antibody targeting NF-κB p65. The green fluorescence was subsequently observed using a confocal microscope.</p>
Full article ">Figure 3
<p>7-MSI suppresses NLRP3 inflammasome activity in RAW-ASC. (<b>A</b>) RAW-ASC cells were first primed with LPS (0.01 μg/mL) for 3 h, followed by treatment with 7-MSI (2 μg/mL) and ATP (1 mM) for an additional 3 h. Western blotting was performed to assess the expression levels of ASC, NLRP3, pro-caspase-1, and GAPDH. (<b>B</b>) The histograms display the relative expression levels of ASC, NLRP3, and pro-caspase-1 proteins, with each value calculated as the signal intensity ratio to GAPDH. (<b>C</b>) The expression levels of NLRP3, ASC, caspase-1, IL-1β, and GAPDH were measured using RT-qPCR. The cells were primed with LPS (0.01 μg/mL) for 3 h and subsequently treated with 7-MSI (2 μg/mL) and ATP (1 mM) for another 3 h. (<b>D</b>) The histograms depicting the relative expression rates from the RT-qPCR analysis. Each value was determined by comparing the signal intensity ratio to that of GAPDH. The symbols “+” and “-” indicate the presence or absence of the respective treatments. The histograms show the means ± S.D. from two independent experiments. Statistical significance was evaluated using Student’s <span class="html-italic">t</span>-test with the following significance levels: * <span class="html-italic">p</span> &lt; 0.0001; ** <span class="html-italic">p</span> &lt; 0.0005; *** <span class="html-italic">p</span> &lt; 0.005, compared to the “LPS-primed + ATP” group. (<b>E</b>) RAW-ASC cells treated with LPS (0.01 μg/mL) for 3 h and then with 7-MSI (2 μg/mL) and ATP (1 mM) for an additional 3 h were stained with fluorescein-labeled anti-ASC and anti-Caspase-1 antibodies. The green and red fluorescences emitted were observed using a confocal microscope.</p>
Full article ">Figure 4
<p>Inhibitory effects of 7-MSI on caspase-1 activity and IL-1β production in RAW-ASC cells. (<b>A</b>) Caspase-1 activity measured in RAW-ASC cells. The cells were first primed with LPS (0.01 μg/mL) for 3 h, followed by co-treatment with 7-MSI (2 μg/mL) and ATP (1 mM) for an additional 3 h. Caspase-1 activity was assessed using a specific assay kit. (<b>B</b>) IL-1β concentrations in the culture supernatants. Following the same treatment protocol as in Panel A, IL-1β levels were quantified using an ELISA kit specific for IL-1β. The results represent the means ± standard deviation (SD) of duplicate determinations from three independent experiments. Statistical significance was assessed using a Student’s <span class="html-italic">t</span>-test, with significance levels indicated by the following symbols: * for <span class="html-italic">p</span> &lt; 0.005 and ǂ for <span class="html-italic">p</span> &lt; 0.5, both compared to the “LPS-primed + ATP” group. The data indicate that 7-MSI potentially inhibits both caspase-1 activity and IL-1β production under the conditions tested.</p>
Full article ">Figure 5
<p>7-MSI induces activation autophagy in Raw 264.7 cells. (<b>A</b>) Raw 264.7 cells were exposed to 7-MSI (1 μg/mL) along with LPS (1 μg/mL) for either 30 min or 1 h. Western blotting was utilized to assess the expression levels of mTOR, phosphorylated mTOR (p-mTOR), Beclin-1, Atg12, LC3, and GAPDH. TGF-β (10 ng/mL) was used as a positive control for the experiment. (<b>B</b>) The histograms show the relative expression levels of the proteins from the 1 h treatment group in panel A. Each value was calculated as the ratio of signal intensity to that of GAPDH, ensuring accurate normalization and comparison. The symbols “+” and “−” denote the inclusion and omission of the respective treatments. The data represent the means ± S.D. values of the results of two separate experiments. The statistical significance of the data was determined using a Student’s <span class="html-italic">t</span>-test, with significance levels indicated as follows: * <span class="html-italic">p</span> &lt; 0.0001; ** <span class="html-italic">p</span> &lt; 0.0005; ǂ <span class="html-italic">p</span> &lt; 0.005, in comparison to the “none” group, which was treated with LPS only. (<b>C</b>) Cells treated with LPS (1 μg/mL) for 1 h or co-treated with LPS (1 μg/mL) and 7-MSI (1 μg/mL) for 1 h were stained with a fluorescein-labeled antibody against LC3-II. Green fluorescence, indicating the presence of LC3-II, was visualized using a confocal microscope, allowing for the observation of autophagy-related processes in the cells. (<b>D</b>) To measure ROS levels, the cells were then further incubated with DCFH-DA (20 μM) for 30 min at 37 °C and the fluorescence intensities were measured using flow cytometry. * <span class="html-italic">p</span> &lt; 0.0005, in comparison to the “LPS” group.</p>
Full article ">Figure 6
<p>Effect of ATG5 and Beclin-1 knockdowns on inflammatory response in RAW-ASC cells. (<b>A</b>) Quantification of TNF-α levels in culture supernatants of RAW-ASC cells transfected with siRNAs targeting none, mock, Beclin-1, mTOR, and ATG-5. Post-transfection, cells were primed with LPS (0.01 μg/mL) for 3 h and subsequently treated with 7-MSI (2 μg/mL) and ATP (1 mM) for an additional 3 h. TNF-α levels were measured using a specific ELISA kit. (<b>B</b>) Measurement of IL-1β levels in the culture supernatants following the same treatment as described for Panel A. IL-1β was quantified using an IL-1β-specific ELISA kit. Statistical analyses were performed using a Student’s <span class="html-italic">t</span>-test, with the significance levels denoted as * for <span class="html-italic">p</span> &lt; 0.005 and ǂ for <span class="html-italic">p</span> &lt; 0.05, both compared to the “ATP” group (LPS-primed group). The “None” label refers to the non-transfected control group. This figure demonstrates how the knockdown of specific autophagy-related genes influences the production of inflammatory cytokines in response to LPS and ATP treatments, adjusted with 7-MSI.</p>
Full article ">Figure 7
<p>Schematic summary of 7-MSI on anti-inflammation and autophagy activation. (<b>A</b>) summarizes the effect of 7-MSI on autophagy and reactive oxygen species (ROS) clearance. 7-MSI activates autophagy and reduces ROS, which in turn can lead to general inflammation or specifically trigger the NLRP3 inflammasome, contributing to skin inflammation. (<b>B</b>) illustrates the molecular pathways influenced by LPS through TLR4, highlighting how 7-MSI intersects with these pathways. In the priming stage, the expression of NLRP3 and pro-IL-1β is increased, while the activation stage involves the formation of the ASC and caspase-1 complex, leading to the production of active caspase-1 and IL-1β. 7-MSI modulates these pathways at various points, potentially reducing the inflammatory response by inhibiting the activation of key inflammatory mediators.</p>
Full article ">
19 pages, 6124 KiB  
Article
HIV Protein Nef Induces Cardiomyopathy Through Induction of Bcl2 and p21
by Olena Kondrachuck, Pierce Ciccone, Nicole Ford, Kim Hong, Yuka Kimura, Jorgo Zi, Sumaya Yusuf, Aya Alkousa, Nishit Tailor, Rithvik Rajkumar, Jay Rappaport and Manish K. Gupta
Int. J. Mol. Sci. 2024, 25(21), 11401; https://doi.org/10.3390/ijms252111401 - 23 Oct 2024
Viewed by 566
Abstract
HIV-associated cardiovascular diseases remain a leading cause of death in people living with HIV/AIDS (PLWHA). Although antiretroviral drugs suppress the viral load, they fail to remove the virus entirely. HIV-1 Nef protein is known to play a role in viral virulence and HIV [...] Read more.
HIV-associated cardiovascular diseases remain a leading cause of death in people living with HIV/AIDS (PLWHA). Although antiretroviral drugs suppress the viral load, they fail to remove the virus entirely. HIV-1 Nef protein is known to play a role in viral virulence and HIV latency. Expression of Nef protein can be detected in different organs, including cardiac tissue. Despite the established role of Nef protein in HIV-1 replication, its impact on organ function inside the human body is not clear. To understand the effect of Nef at the organ level, we created a new Nef-transgenic (Nef-TG) mouse that expresses Nef protein in the heart. Our study found that Nef expression caused inhibition of cardiac function and pathological changes in the heart with increased fibrosis, leading to heart failure and early mortality. Further, we found that cellular autophagy is significantly inhibited in the cardiac tissue of Nef-TG mice. Mechanistically, we found that Nef protein causes the accumulation of Bcl2 and Beclin-1 proteins in the tissue, which may affect the cellular autophagy system. Additionally, we found Nef expression causes upregulation of the cellular senescence marker p21 and senescence-associated β-galactosidase expression. Our findings suggest that the Nef-mediated inhibition of autophagy and induction of senescence markers may promote aging in PLWHA. Our mouse model could help us to understand the effect of Nef protein on organ function during latent HIV infection. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Nef protein expression alters cardiac morphology and induces fibrosis in the heart. For histological analysis, heart tissues from WT and TG-33 mice were collected at 10–13 weeks. (<b>A</b>) Representative images show whole heart and butterfly sections of the heart stained with H&amp;E. (<b>B</b>) Graph shows the heart-to-body weight ratio (<span class="html-italic">n</span> = 38 WT (20 males, 18 females) and 11 TG-33 (5 males, 6 females)). Representative images show the paraffin tissue section stained with (<b>C</b>) H&amp;E, (<b>D</b>) Masson’s trichrome, and (<b>F</b>) WGA stains. The graphs show quantification of (<b>E</b>) the cross-section area of cells and (<b>G</b>) the fibrotic area (<span class="html-italic">n</span> = 7 WT (5 males, 2 females) and 6 TG-33 (2 males, 4 females)). Data are presented with standard deviation. Statistical significances were calculated between WT and TG-33 mice (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, **** <span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 2
<p><b>Nef-transgenic mice exhibit cardiac dysfunction.</b> (<b>A</b>) Representative images show M-mode echocardiography of the wild-type (WT) and TG-33 mice at 10–13 weeks of age. The images of the left ventricle were captured at the mid-papillary level (the parasternal short-axis view). The graphs show quantification of (<b>B</b>) fractional shortening and (<b>C</b>) diameter of the left ventricle during systole (diameter, s) and (<b>D</b>) during diastole (diameter, d) (<span class="html-italic">n</span> = 32 WT (16 males, 16 females) and 24 TG-33 (13 males, 11 females)). Data are presented with standard deviation. Statistical significances were calculated between WT and TG-33 mice (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01).</p>
Full article ">Figure 3
<p><b>Nef expression alters autophagic markers in the heart of TG-33 mice.</b> The Western blots and graphs show expression of autophagy marker proteins (<b>A</b>,<b>B</b>) LC3, (<b>C</b>,<b>D</b>) Beclin-1, and (<b>E</b>,<b>F</b>) p62 in the left ventricular tissue of adult mice hearts (10–13 weeks old) (<span class="html-italic">n</span> = 11 WT (6 males, 5 females) and 7 TG-33 (3 males, 4 females)). GAPDH was used as a loading control. Data are presented with standard deviation. Statistical significances were calculated between WT and TG-33 mice (* <span class="html-italic">p</span> ≤ 0.05, ns = non-significant).</p>
Full article ">Figure 4
<p><b>Nef expression decreases autophagy flux in the hearts of TG-33 mice.</b> TG-33 mice were crossed with autophagy reporter mice tfL-C3 to monitor the autophagy process in the heart. Age-matched transgenic tfL-C3 mice were used as a control group. (<b>A</b>) Representative fluorescence microscopic images show autophagosome (yellow puncta) and autophagosome–lysosome fused (red puncta) formation in the adult mouse tissue of control and TG-33 mice. (<b>B</b>) The graph shows the quantification of the mean number of yellow and red puncta per cardiomyocyte (<span class="html-italic">n</span> = 7 tfL-C3 (WT) mice (3 males, 4 females) and 6 TG-33 (3 males, 3 females)). (<b>C</b>,<b>D</b>) Western blot and graph show autophagy of Nef-transgenic mice during non-fasting and fasting conditions (<span class="html-italic">n</span> = 10 WT non-fasting mice (4 males, 6 females), 9 WT fasting mice (5 males, 4 females), 10 TG-33 non-fasting mice (5 males, 5 females), and 7 TG-33 fasting mice (3 males, 4 females)). Data are presented with standard deviation. Statistical significances were calculated between WT and TG-33 mice (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001, ns = non-significant).</p>
Full article ">Figure 5
<p><b>Nef protein upregulates Bcl2 protein expression in the cardiac tissue of TG-33 mice and HEK293 cells.</b> (<b>A</b>) Western blot shows the expression of Bcl2 protein in the heart tissue. Western blot was performed in cardiac tissue protein lysate of 10 -13-week-old WT and TG-33 mice. GAPDH was used as a loading control. (<b>B</b>) The graph shows quantification of Bcl2 protein in the heart (<span class="html-italic">n</span>= 11 WT (6 males, 5 females) and 7 TG-33 (3 males, 4 females)). Data are presented with standard deviation. Statistical significances were calculated between WT and Nef-TG mice (**** <span class="html-italic">p</span> ≤ 0.0001). Western blots show the expression of (<b>C</b>) Bcl2 and (<b>E</b>) Nef proteins in HEK293 cells transfected with the Nef and the Bcl2 plasmids. GAPDH was used as a loading control. The graphs show the quantification of (<b>D</b>) Bcl2 and (<b>F</b>) Nef proteins in HEK293 cells. The experiment was repeated three times. Data are presented with standard deviation. Statistical significances were calculated between the control group and cells expressing both Bcl2 and Nef proteins (*** <span class="html-italic">p</span> ≤ 0.001, **** <span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 6
<p>Nef protein expression is associated with increased p21 protein but not p16 and p53 in HEK293 cells and in the heart tissue. The representative Western blots show the expression of (<b>A</b>) p21, (<b>C</b>) p16, and (<b>E</b>) p53 proteins in the HEK293 cells transfected with the Nef plasmid or GFP. GAPDH was used as a loading control. The graphs show the quantification of (<b>B</b>) p21, (<b>D</b>) p16, and (<b>F</b>) p53 proteins in the transfected HEK293 cells. The experiment was repeated three times. The Western blots show the expression of (<b>G</b>) p21, (<b>I</b>) p16, and (<b>K</b>) p53 proteins in the cardiac tissue protein lysate of 10–13-week-old WT and TG-33 mice. GAPDH was used as a loading control. The graphs show the quantification of (<b>H</b>) p21, (<b>J</b>) p16, and (<b>L</b>) p53 proteins in the heart tissue (<span class="html-italic">n</span> = 11 WT (6 males, 5 females) and 7 TG-33 (3 males, 4 females)). Data are presented with standard deviation. Statistical significances were calculated between GFP and Nef-transfected HEK293 cells and WT and TG-33 mice (*** <span class="html-italic">p</span> ≤ 0.001, **** <span class="html-italic">p</span> ≤ 0.0001, ns = non-significant).</p>
Full article ">
17 pages, 3267 KiB  
Article
Dietary Probiotic Pediococcus acidilactici GKA4, Dead Probiotic GKA4, and Postbiotic GKA4 Improves Cisplatin-Induced AKI by Autophagy and Endoplasmic Reticulum Stress and Organic Ion Transporters
by Jaung-Geng Lin, Wen-Ping Jiang, You-Shan Tsai, Shih-Wei Lin, Yen-Lien Chen, Chin-Chu Chen and Guan-Jhong Huang
Nutrients 2024, 16(20), 3532; https://doi.org/10.3390/nu16203532 - 18 Oct 2024
Viewed by 822
Abstract
Background/Objectives: Acute kidney injury (AKI) syndrome is distinguished by a quick decline in renal excretory capacity and usually diagnosed by the presence of elevated nitrogen metabolism end products and/or diminished urine output. AKI frequently occurs in hospital patients, and there are no existing [...] Read more.
Background/Objectives: Acute kidney injury (AKI) syndrome is distinguished by a quick decline in renal excretory capacity and usually diagnosed by the presence of elevated nitrogen metabolism end products and/or diminished urine output. AKI frequently occurs in hospital patients, and there are no existing specific treatments available to diminish its occurrence or expedite recovery. For an extended period in the food industry, Pediococcus acidilactici has been distinguished by its robust bacteriocin production, effectively inhibiting pathogen growth during fermentation and storage. Methods: In this study, the aim is to assess the effectiveness of P. acidilactici GKA4, dead probiotic GKA4, and postbiotic GKA4 against cisplatin-induced AKI in an animal model. The experimental protocol involves a ten-day oral administration of GKA4, dead probiotic GKA4, and postbiotic GKA4 to mice, with a cisplatin intraperitoneal injection being given on the seventh day to induce AKI. Results: The findings indicated the significant alleviation of the renal histopathological changes and serum biomarkers of GKA4, dead probiotic GKA4, and postbiotic GKA4 in cisplatin-induced nephrotoxicity. GKA4, dead probiotic GKA4, and postbiotic GKA4 elevated the expression levels of HO-1 and decreased the expression levels of Nrf-2 proteins. In addition, the administration of GKA4, dead probiotic GKA4, and postbiotic GKA4 significantly reduced the expression of apoptosis-related proteins (Bax, Bcl-2, and caspase 3), autophagy-related proteins (LC3B, p62, and Beclin1), and endoplasmic reticulum (ER) stress-related proteins (GRP78, PERK, ATF-6, IRE1, CHOP, and Caspase 12) in kidney tissues. Notably, GKA4, dead probiotic GKA4, and postbiotic GKA4 also upregulated the levels of proteins related to organic anion transporters and organic cation transporters. Conclusions: Overall, the potential therapeutic benefits of GKA4, dead probiotic GKA4, and postbiotic GKA4 are significant, particularly after cisplatin treatment. This is achieved by modulating apoptosis, autophagy, ER stress, and transporter proteins to alleviate oxidative stress. Full article
(This article belongs to the Special Issue What Is Proper Nutrition for Kidney Diseases?)
Show Figures

Figure 1

Figure 1
<p>The experimental framework (<b>A</b>) and the renoprotective efficacy of GKA4, dead probiotic GKA4, and postbiotic GKA4 alleviates neurotoxicity induced by cisplatin treatment. Oral administration of GKA4, dead probiotic GKA4, and postbiotic GKA4 at 250 mg/kg was carried out daily for 10 consecutive days, with cisplatin being administered one hour after the seventh dose. The sacrifice of the mice occurred on the eleventh day. The levels of BUN (<b>B</b>) and CRE in the serum (<b>C</b>), the renal sections stained with H&amp;E (400×) (<b>D</b>), and the renal injury scale (<b>E</b>) were assessed. Means ± S.E.M (<span class="html-italic">n</span> = 5) are shown in the presentation of the data. Statistical significance (<span class="html-italic">p</span> &lt; 0.001) is indicated by <sup>###</sup> when compared with the control group sample. Statistical significance at *** <span class="html-italic">p</span> &lt; 0.001 was evident in contrast to the cisplatin group.</p>
Full article ">Figure 1 Cont.
<p>The experimental framework (<b>A</b>) and the renoprotective efficacy of GKA4, dead probiotic GKA4, and postbiotic GKA4 alleviates neurotoxicity induced by cisplatin treatment. Oral administration of GKA4, dead probiotic GKA4, and postbiotic GKA4 at 250 mg/kg was carried out daily for 10 consecutive days, with cisplatin being administered one hour after the seventh dose. The sacrifice of the mice occurred on the eleventh day. The levels of BUN (<b>B</b>) and CRE in the serum (<b>C</b>), the renal sections stained with H&amp;E (400×) (<b>D</b>), and the renal injury scale (<b>E</b>) were assessed. Means ± S.E.M (<span class="html-italic">n</span> = 5) are shown in the presentation of the data. Statistical significance (<span class="html-italic">p</span> &lt; 0.001) is indicated by <sup>###</sup> when compared with the control group sample. Statistical significance at *** <span class="html-italic">p</span> &lt; 0.001 was evident in contrast to the cisplatin group.</p>
Full article ">Figure 2
<p>GKA4, dead probiotic GKA4, and postbiotic GKA4 alleviate oxidative stress in cisplatin-challenged AKI. The levels of MDA (<b>A</b>) and GSH (<b>B</b>) were assessed through specific assays for MDA and GSH. Means ± S.E.M (<span class="html-italic">n</span> = 5) are shown in the presentation of the data. Statistical significance (<span class="html-italic">p</span> &lt; 0.001) is indicated by <sup>###</sup> when compared with the control group sample. *** <span class="html-italic">p</span> &lt; 0.001 compared with the cisplatin group.</p>
Full article ">Figure 3
<p>The effects of GKA4, dead probiotic GKA4, and postbiotic GKA4 on cisplatin-induced protein expression, including that of HO-1 and Nrf2, were investigated in kidney tissues. The expression of HO-1 and Nrf2 proteins in renal homogenates was assessed via Western blot analysis after exposure to cisplatin. Densitometric analysis was employed to assess the protein bands. The experiments were conducted independently at least three times and representative images were presented.</p>
Full article ">Figure 4
<p>GKA4, dead probiotic GKA4, and postbiotic GKA4 administration resulted in changes in the expression levels of Bax, Bcl-2, and caspase-3 proteins after exposure to cisplatin. Antibodies specific to Bax, Bcl-2, caspase-3, and β-actin were used to conduct a Western blot analysis on kidney tissue lysates. A densitometric analysis was employed to assess the protein bands. The experiments were conducted independently at least three times, and representative images were presented.</p>
Full article ">Figure 5
<p>GKA4, dead probiotic GKA4, and postbiotic GKA4 resulted in a reduction in the levels of LC3B, P62, and Beclin 1 protein following exposure to cisplatin. Antibodies specific to LC3B, P62, Beclin 1, and β-actin were employed for a Western blot analysis of kidney tissue lysates. Protein bands were analyzed via densitometric analysis. The experiments were replicated at least three times, and representative images were displayed.</p>
Full article ">Figure 6
<p>GKA4, dead probiotic GKA4, and postbiotic GKA4 modulated the expression of ER stress proteins in mice with cisplatin-induced AKI. A Western blot analysis was performed on kidney tissue lysates to evaluate protein expression using antibodies specific to GRP78, PERK, ATF-6, IRE1, CHOP, and caspase 12. Densitometric analysis was employed to assess the protein bands. The experiments were conducted independently at least three times, and representative images were presented.</p>
Full article ">Figure 7
<p>GKA4, dead probiotic GKA4, and postbiotic GKA4 regulated renal transporter expressions following exposure to cisplatin in mice. A Western blot analysis was performed on kidney tissue lysates to evaluate protein expression using antibodies specific to OAT1, OAT3, OCT3, and MATE1. A densitometric analysis was employed to assess the protein bands. The experiments were conducted independently at least three times, and representative images were presented.</p>
Full article ">
22 pages, 5813 KiB  
Article
Intracellular Iron Deficiency and Abnormal Metabolism, Not Ferroptosis, Contributes to Homocysteine-Induced Vascular Endothelial Cell Death
by Wenting Shi, Jing Zhang, Wairong Zhao, Meiyan Yue, Jie Ma, Silu Zeng, Jingyi Tang, Yu Wang and Zhongyan Zhou
Biomedicines 2024, 12(10), 2301; https://doi.org/10.3390/biomedicines12102301 - 10 Oct 2024
Viewed by 861
Abstract
Background/Objectives: Homocysteine (Hcy) and iron are factors co-related with the progression of cardiovascular diseases. The vascular endothelium is an important barrier for physiological homeostasis, and its impairment initiates cardiovascular injury. However, the mechanism underlying Hcy-caused vascular endothelial cell injury and the participation of [...] Read more.
Background/Objectives: Homocysteine (Hcy) and iron are factors co-related with the progression of cardiovascular diseases. The vascular endothelium is an important barrier for physiological homeostasis, and its impairment initiates cardiovascular injury. However, the mechanism underlying Hcy-caused vascular endothelial cell injury and the participation of iron are not fully elucidated. This study aims to investigate the Hcy-induced vascular endothelial injury and iron metabolism dysfunction as well as the underlying molecular mechanism. Methods: Human umbilical vein endothelial cells (HUVECs) were employed as the experimental model to examine the Hcy-induced endothelial injury and its underlying mechanism via various biochemical assays. Results: Hcy suppressed the cell viability and proliferation and caused cell death in a concentration-dependent manner. Hcy induced cell cycle arrest, apoptosis, and autophagy as well as impairment of intracellular energy metabolism. Hcy disrupted the intracellular antioxidant system and mitochondrial function by increasing intracellular ROS, MDA and mitochondrial content, and decreasing the SOD activity and mitochondrial membrane potential. Hcy significantly reduced the GSH-Px activity along with the accumulation of intracellular GSH in a concentration-dependent manner. Ferroptosis inhibitors, Ferrostatin-1 (Fer-1), and Deferoxamine (DFO) significantly decreased the Hcy-caused cytotoxicity accompanied by a reduction in dysregulated mitochondria content, but only DFO ameliorated the elevation of intracellular ROS, and neither Fer-1 nor DFO affected the Hcy-caused reduction in intracellular ATP. In addition, Hcy decreased the intracellular concentration of iron, and supplementing Hcy with various concentrations of Fe3+ increased the cell viability and decreased the LDH release in a concentration-dependent manner. Hcy dramatically decreased the mRNA expression level of transferrin receptor while increasing the mRNA expression levels of transferrin, ferritin light chain, ferritin heavy chain, ferroportin, and SLC7A11. Moreover, Hcy suppressed the protein expression of phospho-Akt, phospho-mTOR, Beclin-1, LC3A/B, Nrf2, HO-1, phospho-MEK1/2, phospho-ERK1/2, and Caspase-3 in concentration- and time-dependent manners. Conclusions: Hcy-induced vascular endothelial injury is likely to be associated with apoptosis and autophagy, but not ferroptosis. The key underlying mechanisms are involved in the disruption of the intracellular antioxidant system and iron metabolism via regulation of PI3K/Akt/mTOR, MAPKs, Nrf2/HO-1, and iron metabolism. Full article
Show Figures

Figure 1

Figure 1
<p>The effect of Hcy on endothelial cell morphology, cytotoxicity, and cell viability in HUVECs<b>.</b> (<b>A</b>) The cell morphology of HUVECs treated with indicated concentrations of Hcy for 24 h, n = 3. (<b>B</b>) The cytotoxicity and cell viability of Hcy were examined by LDH release and MTT assays, respectively. Data are presented as folds or percentages of the control group, n = 4. (<b>C</b>) HUVECs were suspended with various concentrations (2, 4, and 8 mM) of Hcy and cultured in an RTCA system for 24 h, and the anti-proliferation effect of Hcy is presented as the Cell Index. The Cell Index of the Hcy-treated group and the control group were also summarized at 24 h, n = 3. (<b>D</b>) HUVECs were cultured and attached for 24 h and then treated with various concentrations (2, 4, and 8 mM) of Hcy for another 24 h. The toxicity effect of Hcy was recorded in real time by RTCA. The Cell Index was normalized to the folds of the value at the time point of adding Hcy. The normalized Cell Index of the Hcy-treated group and control group were also summarized after treatment with Hcy for 24 h, n = 3. (<b>E</b>,<b>F</b>) Presentive images and analysis of live and dead cell staining. The green (calcein-AM) and red (PI) fluorescence demonstrate the live and dead cells, respectively, n = 5. Data are presented as mean ± S.E.M. # <span class="html-italic">p</span> ˂ 0.05, ## <span class="html-italic">p</span> ˂ 0.01, and ### <span class="html-italic">p</span> ˂ 0.001 vs. the control group.</p>
Full article ">Figure 2
<p>The effects of Hcy on cell cycle, apoptosis, autophagy, and energy metabolism in HUVECs. (<b>A</b>) The cell cycle was analyzed by PI staining, followed by flow cytometry. The cell population percentages of the G0/G1, S, and G2/M phases were summarized in both the control group and the Hcy-treated group, n = 3. (<b>B</b>) The apoptosis cells were detected by annexin V-FITC and PI double-staining using flow cytometry. The percentages of early (LR) and late (UR) apoptotic cells were calculated and summarized, n = 3. (<b>C</b>–<b>G</b>) HUVECs were co-treated with Hcy (8 mM) with various indicated concentrations of Z-VDA-FMK (n = 3), 3-MA (n = 3), Wort (n = 3), LY294002 (n = 3), or Rapa (n = 4) for 24 h. The cytotoxicity was examined by LDH release assay. Results are presented as folds of the control group. (<b>H</b>) The intracellular ATP concentration (µmol/mg protein) was quantified by the commercially available kit, n = 4. Data are presented as mean ± S.E.M. # <span class="html-italic">p</span> ˂ 0.05, ## <span class="html-italic">p</span> ˂ 0.01, and ### <span class="html-italic">p</span> ˂ 0.001 vs. the control group. * <span class="html-italic">p</span> ˂ 0.05, ** <span class="html-italic">p</span> ˂ 0.01, and *** <span class="html-italic">p</span> ˂ 0.001 vs. the Hcy-treated group.</p>
Full article ">Figure 3
<p>The effect of Hcy on intracellular ROS and lipid peroxidation in HUVECs. (<b>A</b>,<b>B</b>) The HUVECs were treated with Hcy (8 mM) for 24 h, and then the intracellular ROS were indicated by DHE staining. The fluorescence intensity was calculated using ImageJ software (1.49 V), n = 3. (<b>C</b>) The HUVECs were treated with various indicated concentrations of Hcy for 24 h and the SOD activity was measured using a commercially available kit, n = 3. (<b>D</b>–<b>F</b>) Co-treatment of Hcy (8 mM) with indicated concentrations of DPI (n = 3), NAC (n = 5), VitE (n = 3), and Liproxtatin-1 (n = 3) for 24 h, followed by cytotoxicity detection using LDH release kit. (<b>G</b>) The HUVECs were treated with various indicated concentrations of Hcy for 24 h. The intracellular level of MDA was measured using a commercially available kit, n = 8. (<b>H</b>) Co-treatment of Hcy (8 mM) with various indicated concentrations of Liproxtatin-1 for 24 h followed by LDH release assay, n = 3. The results were normalized to folds of the control group. Data are presented as mean ± S.E.M. # <span class="html-italic">p</span> ˂ 0.05, ## <span class="html-italic">p</span> ˂ 0.01, and ### <span class="html-italic">p</span> ˂ 0.001 vs. the control group. * <span class="html-italic">p</span> ˂ 0.05, and *** <span class="html-italic">p</span> ˂ 0.001 vs. the Hcy-treated group.</p>
Full article ">Figure 4
<p>The role of ferroptosis on Hcy-induced vascular endothelial cell toxicity in HUVECs. (<b>A</b>,<b>B</b>) The HUVECs were treated with various concentrations (2, 4, and 8 mM) of Hcy for 24 h. The intracellular GSH-Px activity and GSH were measured by commercially available kits, n = 5. (<b>C</b>,<b>D</b>) The HUVECs were treated with Hcy (8 mM) for 24 h. The mRNA expression of GPX4 (n = 7) and SLC7A11 (n = 5) genes was examined by real-time PCR. (<b>E</b>,<b>F</b>) The HUVECs were co-treated with Hcy (8 mM), with various indicated concentrations of Fer-1 or DFO for 24 h, followed by the LDH release assay, n = 3. (<b>G</b>,<b>H</b>) HUVECs were suspended with Hcy (8 mM), with or without Fer-1 (80 µM) or DFO (80 µM), for 24 h. The cell proliferation was recorded in real time by RTCA, n = 4. The results were normalized to folds of the control group. Data are presented as mean ± S.E.M. # <span class="html-italic">p</span> ˂ 0.05, ## <span class="html-italic">p</span> ˂ 0.01, and ### <span class="html-italic">p</span> ˂ 0.001 vs. the control group. * <span class="html-italic">p</span> ˂ 0.05, ** <span class="html-italic">p</span> ˂ 0.01, and *** <span class="html-italic">p</span> ˂ 0.001 vs. the Hcy-treated group.</p>
Full article ">Figure 5
<p>The effect of ferroptosis inhibitors on Hcy-induced intracellular ROS production and mitochondrial dysfunction in HUVECs. (<b>A</b>–<b>C</b>) The HUVECs were treated with Hcy (8 mM), with or without Fer-1 (80 µM) or DFO (80 µM), for 24 h. Then, the intracellular ROS, number of mitochondria, and mitochondria membrane potential were indicated by DHE (<b>A</b>), Mito-tracker (<b>B</b>), and JC-1 (<b>C</b>) staining, respectively, n = 3. (<b>D</b>–<b>F</b>) The fluorescence intensity was calculated using ImageJ software. The results were normalized to folds of the control group. (<b>G</b>) The HUVECs were treated with Hcy (8 mM), with or without Fer-1 (80 µM) or DFO (80 µM), for 24 h. Then, the intracellular concentration of ATP was detected by a commercially available kit, n = 4. Data are presented as mean ± S.E.M. # <span class="html-italic">p</span> ˂ 0.05 and ## <span class="html-italic">p</span> ˂ 0.01 vs. the control group. * <span class="html-italic">p</span> ˂ 0.05, and *** <span class="html-italic">p</span> ˂ 0.001 vs. the Hcy-treated group.</p>
Full article ">Figure 6
<p>The role of iron metabolism in Hcy-induced vascular endothelial cell toxicity in HUVECs. (<b>A</b>) HUVECs were treated with Hcy (8 mM) for 24 h. The intracellular level of iron was measured by a commercially available kit according to its manual, n = 3. (<b>B</b>,<b>C</b>) Co-treatment of Hcy (8 mM) with indicated concentrations of Fe<sup>3+</sup> for 24 h, followed by cell viability and cytotoxicity detections using MTT and LDH release assays, respectively, n = 3. (<b>D</b>) The cell morphology was observed by an inverted microscope equipped with 10× and 20× objective lenses. (<b>E</b>) The HUVECs were treated with or without Hcy (8 mM) for 24 h. The mRNA expressions of transferrin receptor (n = 6), transferrin (n = 5), ferritin light chain (n = 5), ferritin heavy chain (n = 6), and ferriportin (n = 6) genes were examined by real-time PCR. Results are presented as folds or percentages of the control group. Data are presented as mean ± S.E.M. # <span class="html-italic">p</span> ˂ 0.05, ## <span class="html-italic">p</span> ˂ 0.01, and ### <span class="html-italic">p</span> ˂ 0.001 vs. the control group. * <span class="html-italic">p</span> ˂ 0.05, ** <span class="html-italic">p</span> ˂ 0.01, and *** <span class="html-italic">p</span> ˂ 0.001 vs. the Hcy-treated group.</p>
Full article ">Figure 7
<p>The effect of Hcy on Akt/mTOR autophagy signaling in HUVECs. (<b>A</b>) The HUVECs were treated with various concentrations (2, 4, and 8 mM) of Hcy for 24 h. The representative bands in Western blotting analysis. (<b>B</b>–<b>E</b>) The quantitative protein expressions of phospho-mTOR (n = 3), mTOR (n = 3), phospho-Akt (n = 4), Akt (n = 4), Beclin-1 (n = 4), LC3A/B (n = 4), and GAPDH (n = 4) were detected by Western blotting analysis. GAPDH served as the internal control. The results were normalized to folds of the control group. Data are presented as mean ± S.E.M. # <span class="html-italic">p</span> ˂ 0.05, ## <span class="html-italic">p</span> ˂ 0.01, and ### <span class="html-italic">p</span> ˂ 0.001 vs. the control group.</p>
Full article ">Figure 8
<p>The effect of Hcy on MAPKs and Nrf2/HO-1 signaling in HUVECs. (<b>A</b>) The HUVECs were treated with various concentrations (2, 4, and 8 mM) of Hcy for 24 h. The representative bands in Western blotting analysis. (<b>B</b>–<b>F</b>) The quantitative protein expressions of Nrf2 (n = 4), HO-1 (n = 4), phospho-MEK1/2 (n = 4), MEK1/2 (n = 4), phospho-ERK1/2 (n = 3), ERK1/2 (n = 3), Caspase-3 (n = 4), and GAPDH (n = 3) were detected by Western blotting analysis. GAPDH served as the internal control. The results were normalized to folds of the control group. Data are presented as mean ± S.E.M. # <span class="html-italic">p</span> ˂ 0.05, ## <span class="html-italic">p</span> ˂ 0.01, and ### <span class="html-italic">p</span> ˂ 0.001 vs. the control group.</p>
Full article ">Figure 9
<p>Schematic overview of the underlying mechanism of Hcy-induced endothelial death.</p>
Full article ">
23 pages, 4823 KiB  
Article
Alleviation of Autophagic Deficits and Neuroinflammation by Histamine H3 Receptor Antagonist E159 Ameliorates Autism-Related Behaviors in BTBR Mice
by Shilu Deepa Thomas, Petrilla Jayaprakash, Nurfirzana Z. H. J. Marwan, Ezzatul A. B. A. Aziz, Kamil Kuder, Dorota Łażewska, Katarzyna Kieć-Kononowicz and Bassem Sadek
Pharmaceuticals 2024, 17(10), 1293; https://doi.org/10.3390/ph17101293 - 28 Sep 2024
Viewed by 972
Abstract
Background/Objectives: Autism spectrum disorder (ASD) is a neurodevelopmental condition marked by social interaction difficulties, repetitive behaviors, and immune dysregulation with elevated pro-inflammatory markers. Autophagic deficiency also contributes to social behavior deficits in ASD. Histamine H3 receptor (H3R) antagonism is a potential treatment strategy [...] Read more.
Background/Objectives: Autism spectrum disorder (ASD) is a neurodevelopmental condition marked by social interaction difficulties, repetitive behaviors, and immune dysregulation with elevated pro-inflammatory markers. Autophagic deficiency also contributes to social behavior deficits in ASD. Histamine H3 receptor (H3R) antagonism is a potential treatment strategy for brain disorders with features overlapping ASD, such as schizophrenia and Alzheimer’s disease. Methods: This study investigated the effects of sub-chronic systemic treatment with the H3R antagonist E159 on social deficits, repetitive behaviors, neuroinflammation, and autophagic disruption in male BTBR mice. Results: E159 (2.5, 5, and 10 mg/kg, i.p.) improved stereotypic repetitive behavior by reducing self-grooming time and enhancing spontaneous alternation in addition to attenuating social deficits. It also decreased pro-inflammatory cytokines in the cerebellum and hippocampus of treated BTBR mice. In BTBR mice, reduced expression of autophagy-related proteins LC3A/B and Beclin 1 was observed, which was elevated following treatment with E159, attenuating the disruption in autophagy. The co-administration with the H3R agonist MHA (10 mg/kg, i.p.) reversed these effects, highlighting the role of histaminergic neurotransmission in observed behavioral improvements. Conclusions: These preliminary findings suggest the therapeutic potential of H3R antagonists in targeting neuroinflammation and autophagic disruption to improve ASD-like behaviors. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Chemical structure and pharmacological in vitro binding affinity profile of E159 on selected human histamine receptor subtypes.</p>
Full article ">Figure 2
<p><b>Left</b> panel: Predicted binding mode of E159 (<b>left</b>) within histamine H3R receptor binding site. Hydrogen bonds are shown with yellow dashed lines, salt bridges with magenta lines, cation-π interactions with green lines, and π−π interactions with blue lines. Roman numerals indicate the respective TMs; <b>Right</b> panel: Summary of ligand-protein contacts from MD simulation (<b>top</b>; hydrogen bond is shown with a purple dashed line, π−π interactions as green and cation-π as red lines), and contacts histogram (<b>bottom</b>; green for hydrogen bonds, violet for hydrophobic contacts, blue for water bridges; X-axis represents interaction fraction (1.0 = 100% simulation time), Y-axis represents particular interacting amino acids.</p>
Full article ">Figure 3
<p>(<b>Left</b> panel): Orientation of E159 during the 250 ns MD simulation. Different colors represent distinct frames: 0 ns is shown in blue, transitioning through the violet spectrum (dark to light: 25–100 ns) to grey (125 ns) and vice versa through the orange spectrum (light to dark: 150–225 ns) to red (250 ns). (<b>Right</b> panel): Time evolution of RMSD for ligand (magenta) and protein (grey blue and dark red) for specific frames relative to the reference frame at 0 ns.</p>
Full article ">Figure 4
<p>E159 mitigated compulsive grooming in <span class="html-italic">BTBR</span> mice. Both <span class="html-italic">B6</span> and <span class="html-italic">BTBR</span> mice, were administered an intraperitoneal injection of a vehicle, E159 or ARP before the assessment of self-grooming. <span class="html-italic">BTBR</span> mice exhibited a significantly higher grooming duration in comparison to <span class="html-italic">B6</span> mice. E159 as well as ARP, significantly decreased self-grooming in the autistic model. Additionally, the impact of co-injection of (R)-α-methylhistamine (MHA) on E159 (2.5 mg/kg)-induced reduction in grooming duration in <span class="html-italic">BTBR</span> mice was evaluated. * <span class="html-italic">p</span> &lt; 0.001 relative to <span class="html-italic">B6</span> mice treated with vehicle, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.001 relative to <span class="html-italic">BTBR</span> mice treated with vehicle, <sup><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01 relative to E159 (2.5 mg)-treated autistic mice, (n = 6). (mean ± SEM, n = 6/group).</p>
Full article ">Figure 5
<p>E159 treatment enhanced the alternation behavior in autistic mice. A significantly lower alternation behavior was seen in <span class="html-italic">BTBR</span> mice compared to <span class="html-italic">B6</span> mice. However, E159 or ARP considerably improved the alternation in <span class="html-italic">BTBR</span> mice. The impact of MHA (10 mg/kg) co-administration on the enhancement of alternation behavior induced by E159 (2.5 mg) in autistic mice was evaluated. * <span class="html-italic">p</span> &lt; 0.001 relative to <span class="html-italic">B6</span> mice treated with vehicle. ** <span class="html-italic">p</span> &lt; 0.01, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.05 relative to <span class="html-italic">BTBR</span> mice treated with vehicle. <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01 relative to E159 (2.5 mg/kg, i.p.)-treated <span class="html-italic">BTBR</span> mice. (mean ± SEM, n = 6/group).</p>
Full article ">Figure 6
<p>E159 improved impaired sociability in the autistic mice. Mice explored all three chambers for two consecutive 10 min sessions. Results measured included (<b>A</b>) the Sociability Index (SI) and (<b>B</b>) the Social Novelty Index (SNI). <span class="html-italic">BTBR</span> mice were administered E159 or ARP. The impact of co-administering MHA (10 mg/kg) on the enhancement of SI and SNI induced by 2.5 mg of E159 in the autistic model was evaluated. (<b>A</b>) SI: * <span class="html-italic">p</span> &lt; 0.001 relative to <span class="html-italic">B6</span> mice treated with vehicle. ** <span class="html-italic">p</span> &lt; 0.001, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.01 relative to <span class="html-italic">BTBR</span> mice treated with vehicle, <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05 relative to E159 (2.5 mg)-treated <span class="html-italic">BTBR</span> mice. (<b>B</b>) SNI: * <span class="html-italic">p</span> &lt; 0.01 relative to <span class="html-italic">B6</span> mice treated with vehicle, ** <span class="html-italic">p</span> &lt; 0.01, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.05 relative to <span class="html-italic">BTBR</span> mice treated with vehicle, <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01 relative to E159 (2.5 mg)-treated <span class="html-italic">BTBR</span> mice, (n = 6). (mean ± SEM, n = 6/group).</p>
Full article ">Figure 6 Cont.
<p>E159 improved impaired sociability in the autistic mice. Mice explored all three chambers for two consecutive 10 min sessions. Results measured included (<b>A</b>) the Sociability Index (SI) and (<b>B</b>) the Social Novelty Index (SNI). <span class="html-italic">BTBR</span> mice were administered E159 or ARP. The impact of co-administering MHA (10 mg/kg) on the enhancement of SI and SNI induced by 2.5 mg of E159 in the autistic model was evaluated. (<b>A</b>) SI: * <span class="html-italic">p</span> &lt; 0.001 relative to <span class="html-italic">B6</span> mice treated with vehicle. ** <span class="html-italic">p</span> &lt; 0.001, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.01 relative to <span class="html-italic">BTBR</span> mice treated with vehicle, <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05 relative to E159 (2.5 mg)-treated <span class="html-italic">BTBR</span> mice. (<b>B</b>) SNI: * <span class="html-italic">p</span> &lt; 0.01 relative to <span class="html-italic">B6</span> mice treated with vehicle, ** <span class="html-italic">p</span> &lt; 0.01, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.05 relative to <span class="html-italic">BTBR</span> mice treated with vehicle, <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01 relative to E159 (2.5 mg)-treated <span class="html-italic">BTBR</span> mice, (n = 6). (mean ± SEM, n = 6/group).</p>
Full article ">Figure 7
<p>Treatment with E159 had no discernible impact on locomotor ability in autistic mice. (<b>A</b>) <span class="html-italic">BTBR</span> mice showed significantly greater distances travelled relative to <span class="html-italic">B6</span> mice. (<b>B</b>) Pretreatment with E159 or ARP did not significantly affect the duration spent in periphery in autistic mice. (<b>C</b>) Additionally, <span class="html-italic">BTBR</span> mice spent less duration in the center relative to <span class="html-italic">B6</span> mice. Data are shown as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 relative to <span class="html-italic">B6</span> mice treated with vehicle. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 relative to <span class="html-italic">BTBR</span> mice treated with vehicle. <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.01 versus <span class="html-italic">BTBR</span> mice treated with E159 (2.5 mg) (n = 6).</p>
Full article ">Figure 8
<p>The proteins p-mTOR, mTOR, LC3A/B and Beclin-1 were determined by Western blotting (<b>A</b>). <span class="html-italic">BTBR</span> mice showed decreased expression level of Beclin-1 and LC3A/B in the cerebellum with increase in the levels of p-mTOR/ mTOR which further suggests autophagic deficiency. E159 ameliorates autophagic deficits with reduced levels of p-mTOR/ mTOR (<b>B</b>), and increased Beclin-1 (<b>C</b>) and LC3A/B (<b>D</b>). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus <span class="html-italic">B6</span> mice treated with vehicle. * <span class="html-italic">p</span> &lt; 0.05 versus <span class="html-italic">BTBR</span> mice treated with vehicle. ** <span class="html-italic">p</span> &lt; 0.05,*** <span class="html-italic">p</span> &lt; 0.01 versus E159 (2.5 mg)-treated <span class="html-italic">BTBR</span> mice (n = 3).</p>
Full article ">Figure 8 Cont.
<p>The proteins p-mTOR, mTOR, LC3A/B and Beclin-1 were determined by Western blotting (<b>A</b>). <span class="html-italic">BTBR</span> mice showed decreased expression level of Beclin-1 and LC3A/B in the cerebellum with increase in the levels of p-mTOR/ mTOR which further suggests autophagic deficiency. E159 ameliorates autophagic deficits with reduced levels of p-mTOR/ mTOR (<b>B</b>), and increased Beclin-1 (<b>C</b>) and LC3A/B (<b>D</b>). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus <span class="html-italic">B6</span> mice treated with vehicle. * <span class="html-italic">p</span> &lt; 0.05 versus <span class="html-italic">BTBR</span> mice treated with vehicle. ** <span class="html-italic">p</span> &lt; 0.05,*** <span class="html-italic">p</span> &lt; 0.01 versus E159 (2.5 mg)-treated <span class="html-italic">BTBR</span> mice (n = 3).</p>
Full article ">
16 pages, 4063 KiB  
Article
Hyperbaric Treatment Stimulates Chaperone-Mediated Macroautophagy and Autophagy in the Liver Cells of Healthy Female Rats
by Agnieszka Pedrycz, Mariusz Kozakiewicz, Mansur Rahnama, Marek Kos, Ewelina Grywalska, Marietta Bracha, Anna Grzywacz and Iwona Bojar
Int. J. Mol. Sci. 2024, 25(19), 10476; https://doi.org/10.3390/ijms251910476 - 28 Sep 2024
Viewed by 708
Abstract
The role of autophagy goes far beyond the elimination of damaged cellular components and the quality control of proteins. It also cleanses cells from inclusions, including pathogenic viruses, and provides energy-forming components. The liver, which is an organ with increased metabolism, is made [...] Read more.
The role of autophagy goes far beyond the elimination of damaged cellular components and the quality control of proteins. It also cleanses cells from inclusions, including pathogenic viruses, and provides energy-forming components. The liver, which is an organ with increased metabolism, is made up of cells that are particularly vulnerable to damage. Therefore, detoxification of liver cells in the process of autophagy has become a very important issue clinically. The aim of this study was an immunohistochemical evaluation of proteins activated in rat liver cells at different stages of hyperbaric autophagy. The rats used for the study were randomly divided into six equivalent groups—three control groups and three experimental groups. Animals from the experimental groups were subjected to hyperbaric treatment in a hyperbaric chamber, with a pressure of 1.6 ATA for 120 min. They breathed atmospheric air. Rats were decapitated within 5 or 10 days after removal from the chamber. Immunohistochemical reactions with beclin 1, LC3B, RAB7, and HSC73 proteins were carried out on preparations made from liver slices. A three-step labeled streptavidin–biotin detection method of paraffin blocks (LSAB three-step) was used for immunohistochemical research. The results were evaluated using computer programs for morphometric analysis of microscopic images by calculating the mean surface areas occupied by a positive immunohistochemical reaction in individual groups for all antibodies tested. Increased closure of substrates in the autophagosome (beclin 1) induced late endosome transport and accelerated autophagosome maturation process (RAB7). Furthermore, a larger number of autophagosomes (LC3B) was observed in liver cells immediately after the cessation of hyperbaric activity; however, this decreased after 5 days. During this time, chaperone-mediated autophagy (HSC73) was observed on a larger scale. This means that increased macroautophagy induced by hyperbaric treatment weakens with time that has elapsed since the cessation of high pressure, whereas similarly induced chaperone-mediated autophagy intensifies over time. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Novel Therapies of Liver Diseases)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) KI control group. Invisible immunohistochemical reaction for beclin 1 in the liver fragment of a female rat that was decapitated on the 1st day of the experiment. H + AEC dyeing. (<b>B</b>) KII control group. Invisible immunohistochemical reaction for beclin 1 in a fragment of the liver of a female rat that was decapitated on the 5th day of the experiment. Dyeing H + AEC. (<b>C</b>) KIII control group. Invisible immunohistochemical reaction for beclin 1 in a fragment of the liver of a female rat that was decapitated on the 10th day of the experiment. Dyeing H + AEC.</p>
Full article ">Figure 2
<p>(<b>A</b>) DHI experimental group. Moderately severe immunohistochemical reaction for beclin 1 in a fragment of the liver of a female rat that underwent hyperbaric treatment and was decapitated immediately after removal from the hyperbaric chamber. H+AEC dyeing. (<b>B</b>) Experimental group DHII. Moderately severe immunohistochemical reaction for beclin 1 in a fragment of the liver of a female rat that underwent hyperbaric treatment and was decapitated 5 days after removal from the hyperbaric chamber. H + AEC dyeing. (<b>C</b>) Experimental group DHIII. Absent immunohistochemical reaction for beclin 1 in a liver fragment of a female rat that underwent hyperbaric treatment and was decapitated 10 days after removal from the hyperbaric chamber. Dyeing H + AEC.</p>
Full article ">Figure 3
<p>(<b>A</b>) Control group KI. Invisible immunohistochemical reaction for LC3B in a fragment of the liver of a female rat that was decapitated on 1st day of the experiment. H + AEC staining. (<b>B</b>) KII control group. Invisible immunohistochemical reaction for LC3B in a fragment of the liver of a female rat that was decapitated on the 5th day of the experiment. Magnification approx. 180×. Dyeing H + AEC. (<b>C</b>) KIII control group. Invisible immunohistochemical reaction for LC3B in a fragment of the liver of a female rat that was decapitated on the 10th day of the experiment. Magnification approx. 180×. H + AEC staining.</p>
Full article ">Figure 4
<p>(<b>A</b>) Experimental group DHI. Moderately severe immunohistochemical reaction for LC3B in a liver fragment of a female rat that underwent hyperbaric treatment and was decapitated immediately after removal from the hyperbaric chamber. H+AEC dyeing. (<b>B</b>) Experimental group DHII. Moderately severe immunohistochemical reaction for LC3B in a liver fragment of a female rat that underwent hyperbaric treatment and was decapitated 5 days after removal from the hyperbaric chamber. Dyeing H + AEC. (<b>C</b>) Experimental group DHIII. Absent immunohistochemical reaction for LC3B in a liver fragment of a female rat that underwent hyperbaric treatment and was decapitated 10 days after removal from the hyperbaric chamber. Magnification approx. 360×. H + AEC staining.</p>
Full article ">Figure 5
<p>(<b>A</b>) Control group KI. Invisible immunohistochemical reaction for RAB7 in the liver fragment of a female rat that was decapitated on the 1st day of the experiment. Magnification approx. 300×. H + AEC staining. (<b>B</b>) KII control group. Invisible immunohistochemical reaction for RAB7 in a fragment of the liver of a female rat that was decapitated on the 5th day of the experiment. Magnification approx. 300×. H+AEC staining. (<b>C</b>) KIII control group. Invisible immunohistochemical reaction for RAB7 in a fragment of the liver of a female rat that was decapitated on the 10th day of the experiment. Magnification approx. 500×. H + AEC staining.</p>
Full article ">Figure 6
<p>(<b>A</b>) Experimental group DHI. Moderately severe immunohistochemical reaction for RAB7 in a fragment of the liver of a female rat that underwent hyperbaric treatment and was decapitated immediately after removal from the hyperbaric chamber. Magnification approx. 300×. H + AEC staining. (<b>B</b>) Experimental group DHII. Moderately severe immunohistochemical reaction for RAB7 in a fragment of the liver of a female rat that underwent hyperbaric treatment and was decapitated 5 days after removal from the hyperbaric chamber. Magnification approx. 300×. H + AEC staining. (<b>C</b>) Experimental group DHIII. Absent immunohistochemical reaction for RAB7 in a liver fragment of a female rat that underwent hyperbaric treatment and was decapitated 10 days after removal from the hyperbaric chamber. Magnification approx. 500×. H + AEC staining.</p>
Full article ">Figure 7
<p>(<b>A</b>) Control group KI. Invisible immunohistochemical reaction for HSC73 in a fragment of the liver of a female rat that was decapitated on the first day of the experiment. H + AEC dyeing. (<b>B</b>) KII control group. Invisible immunohistochemical reaction for HSC73 in a fragment of the liver of a female rat that was decapitated on the 5th day of the experiment. Magnification approx. 300×. H + AEC staining. (<b>C</b>) KIII control group. Invisible immunohistochemical reaction for HSC73 in a fragment of the liver of a female rat that was decapitated on the 10th day of the experiment. Magnification approx. 500×. H + AEC staining.</p>
Full article ">Figure 8
<p>(<b>A</b>) DHI experimental group. Invisible immunohistochemical reaction for HSC73 in a fragment of the liver of a female rat that underwent hyperbaric treatment and was decapitated on the first day of the experiment, i.e., immediately after removal from the hyperbaric chamber. Magnification approx. 500×. H + AEC staining. (<b>B</b>) Experimental group DHII. Moderately severe immunohistochemical reaction for HSC73 in a fragment of the liver of a female rat that underwent hyperbaric treatment and was decapitated on the 5th day of the experiment, i.e., 5 days after removal from the hyperbaric chamber. H + AEC dyeing. (<b>C</b>) Experimental group DHIII. A strongly intensified immunohistochemical reaction for HSC73 in a fragment of the liver of a female rat, which was subjected to hyperbaric treatment and decapitated on the 10th day of the experiment, i.e., 10 days after removal from the hyperbaric chamber. H + AEC dyeing.</p>
Full article ">
15 pages, 6890 KiB  
Article
c-Jun N-terminal Kinase Supports Autophagy in Testicular Ischemia but Triggers Apoptosis in Ischemia-Reperfusion Injury
by Sarah R. Alotaibi, Waleed M. Renno and May Al-Maghrebi
Int. J. Mol. Sci. 2024, 25(19), 10446; https://doi.org/10.3390/ijms251910446 - 27 Sep 2024
Viewed by 611
Abstract
Oxidative stress triggered by testicular torsion and detorsion in young males could negatively impact future fertility. Using a rat animal model for testicular IRI (tIRI), we aim to study the induction of autophagy (ATG) during testicular ischemia and tIRI and the role of [...] Read more.
Oxidative stress triggered by testicular torsion and detorsion in young males could negatively impact future fertility. Using a rat animal model for testicular IRI (tIRI), we aim to study the induction of autophagy (ATG) during testicular ischemia and tIRI and the role of oxidative-stress-induced c-Jun N-terminal Kinase (JNK) as a cytoprotective mechanism. Sixty male Sprague-Dawley rats were divided into five groups: sham, ischemia only, ischemia+SP600125 (a JNK inhibitor), tIRI only, and tIRI+SP600125. The tIRI rats underwent an ischemic injury for 1 h followed by 4 h of reperfusion, while ischemic rats were subjected to 1 h of ischemia only without reperfusion. Testicular-ischemia-induced Beclin 1 and LC3B expression was associated with decreased p62/SQSTM1 expression, increased ATP and alkaline phosphatase (AP) activity, and slightly impaired spermatogenesis. SP600125 treatment improved p62 expression and reduced the levels of Beclin 1 and LC3B but did not affect ATP or AP levels. The tIRI-induced apoptosis lowered the expression of the three ATG proteins and AP activity, activated caspase 3, and caused spermatogenic arrest. SP600125-inhibited JNK during tIRI restored sham levels to all investigated parameters. This study emphasizes the regulatory role of JNK in balancing autophagy and apoptosis during testicular oxidative injuries. Full article
Show Figures

Figure 1

Figure 1
<p>JNK phosphorylation is associated with testicular ischemia and tIRI. (<b>a</b>) Representative Western blots (WB) of JNK and p-JNK (T183 and Y185) in ipsilateral testes of experimental groups: sham, ischemia, tIRI, ischemia+SP600125, and tIRI+SP600125. SP600125 is injected intraperitoneally 30 min prior to ischemia and tIRI at 15 mg/Kg. (<b>b</b>) WB band intensity of JNK and p-JNK normalized to GAPDH. (<b>c</b>) Ratio of p-JNK to JNK expression. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6/group), <span class="html-italic">p</span>-value &lt; 0.05. * Ischemia compared to sham and # tIRI compared to sham.</p>
Full article ">Figure 2
<p>JNK modulates the gene expression of autophagy markers: Beclin 1, LC3B, and p62. (<b>a</b>) Representative Western blots and (<b>b</b>) bar graphs of Beclin 1, LC3B, and p62/SQSTM1 protein expression in all 5 experimental groups. (<b>c</b>) The relative mRNA expression of autophagy markers Becn1, Lc3b, and Sqstm1 was calculated using the 2<sup>−ΔΔCt</sup> method. The fold change in gene expression in ischemia-, tIRI-, and SP600125-treated groups was calculated relative to the sham group. * Ischemia compared to sham and # tIRI compared to sham.</p>
Full article ">Figure 3
<p>JNK modulates LC3B expression. LC3B was detected in seminiferous tubules using fluorescence confocal microscopy. Representative immunofluorescence (IF)-stained testicular tissue sections for (<b>a</b>) sham, (<b>b</b>) ischemia, (<b>c</b>) ischemia+SP600125, (<b>d</b>) tIRI, and (<b>e</b>) tIRI+SP600125 groups. (<b>f</b>) Quantification of the average IF intensity and statistical analysis. Images were taken at 40× magnification with a scale bar of 50 μm. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6/group), <span class="html-italic">p</span>-value &lt; 0.05. * Ischemia compared to sham.</p>
Full article ">Figure 4
<p>JNK modulates lysosomal alkaline phosphatase (AP) activity but not ATP levels. Biochemical colorimetric assay kits measured AP activity and ATP concentration in the 5 experimental groups. The data were analyzed using one-way analysis of variance (ANOVA) followed by the Holm–Sidak multiple comparisons test and presented as mean values ± SD (<span class="html-italic">n</span> = 6/group). * Ischemia compared to sham and # tIRI compared to sham.</p>
Full article ">Figure 5
<p>JNK modulates caspase 3 activity. Caspase 3 activity was measured using a colorimetric assay, and the fold change in ischemia-, tIRI-, and SP600125-treated groups was calibrated to sham activity. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6/group), <span class="html-italic">p</span>-value &lt; 0.05. * tIRI compared to ischemia and # tIRI compared to sham.</p>
Full article ">Figure 6
<p>JNK modulates spermatogenesis activity. H&amp;E-stained testicular tissue sections for (<b>a</b>) sham, (<b>b</b>) ischemia, (<b>c</b>) ischemia+SP600125, (<b>d</b>) tIRI, and (<b>e</b>) tIRI+SP600125 groups. (<b>f</b>) Spermatogenesis analysis using the Johnsen score. Images were taken at 40× magnification with a scale bar of 200 μm. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6/group), <span class="html-italic">p</span>-value &lt; 0.05. * Ischemia compared to sham and # tIRI compared to sham.</p>
Full article ">
Back to TopTop