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

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,347)

Search Parameters:
Keywords = TNF-alpha

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 490 KiB  
Article
The Impact of Probiotics on Clinical Symptoms and Peripheral Cytokines Levels in Parkinson’s Disease: Preliminary In Vivo Data
by Luca Magistrelli, Elena Contaldi, Annalisa Visciglia, Giovanni Deusebio, Marco Pane and Angela Amoruso
Brain Sci. 2024, 14(11), 1147; https://doi.org/10.3390/brainsci14111147 - 15 Nov 2024
Viewed by 304
Abstract
Introduction. Previous studies have shown that probiotics have positive effects on both motor and non-motor symptoms in Parkinson’s disease (PD). Additionally, in preclinical settings, probiotics have demonstrated the ability to counteract neuronal loss and alpha-synuclein aggregation, important pathological hallmarks of PD. Notably, preliminary [...] Read more.
Introduction. Previous studies have shown that probiotics have positive effects on both motor and non-motor symptoms in Parkinson’s disease (PD). Additionally, in preclinical settings, probiotics have demonstrated the ability to counteract neuronal loss and alpha-synuclein aggregation, important pathological hallmarks of PD. Notably, preliminary in vitro studies have revealed the immunomodulatory properties of probiotics. This study aims to evaluate the impact of probiotics on symptoms and peripheral cytokines levels in PD patients compared to placebo. Methods. Patients were enrolled and blindly randomized to receive either active probiotics (comprising Bifidobacterium animalis subsp. lactis BS01 LMG P-21384, Bifidobacterium longum BL03 DSM 16603, Bifidobacterium adolescentis BA02 DSM 18351, Fructo-oligosaccharides and Maltodextrin-Group A) or placebo (Maltodextrin-Group B). Clinical evaluations and plasma levels cytokines (TNF-α, IFN-γ, IL-6, and TGF-β) were also assessed at enrollment and after 12 weeks. Anti-parkinsonian therapy remained stable throughout the study. Results. Forty PD patients were recruited. After 12 weeks, Group A showed significant improvement in motor symptoms (UPDRS III: 13.89 ± 4.08 vs. 12.74 ± 4.57, p = 0.028) and non-motor symptoms (NMSS: 34.32 ± 21.41 vs. 30.11 ± 19.89, p = 0.041), with notable improvement in the gastrointestinal sub-item (3.79 ± 4.14 vs. 1.89 ± 2.54, p = 0.021). A reduction of IFN-γ levels was observed in both groups, but group A also showed a significant decrease in IL-6 and a slight increase in the anti-inflammatory cytokine TGF-β. Conclusions. Our data suggest that probiotics may modulate peripheral cytokines levels and improve clinical symptoms in PD patients. Probiotics may, therefore, represent a valuable adjunctive therapy to conventional anti-parkinsonian drugs. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the study protocol.</p>
Full article ">
15 pages, 964 KiB  
Systematic Review
Purslane Ameliorates Inflammation and Oxidative Stress in Diabetes Mellitus: A Systematic Review
by Zikho Nkhumeleni, Wendy N. Phoswa and Kabelo Mokgalaboni
Int. J. Mol. Sci. 2024, 25(22), 12276; https://doi.org/10.3390/ijms252212276 - 15 Nov 2024
Viewed by 309
Abstract
Type 2 diabetes (T2D) is characterised by insulin resistance and leads to hyperglycaemia. Its prevalence and associated complications continue to rise exponentially, despite the existence of pharmaceutical drugs, and this has prompted research into exploring safer herbal remedies. Portulaca oleracea (purslane) has been [...] Read more.
Type 2 diabetes (T2D) is characterised by insulin resistance and leads to hyperglycaemia. Its prevalence and associated complications continue to rise exponentially, despite the existence of pharmaceutical drugs, and this has prompted research into exploring safer herbal remedies. Portulaca oleracea (purslane) has been investigated in animal and clinical trials to explore its effects on diabetes, yielding conflicting results. This study aimed to evaluate the effects of purslane on inflammation and oxidative stress in diabetes mellitus. We conducted a comprehensive literature search on Scopus PubMed, and through a manual bibliographical search to find relevant studies from inception to 13 September 2024. The search terms included purslane, portulaca oleracea, and type 2 diabetes mellitus. Of the 38 retrieved studies, 12 were considered relevant and underwent critical review. Evidence from rodent studies showed decreased inflammatory markers such as interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-α), nuclear factor kappa-beta (NF-κβ), and C-reactive (CRP), while interleukin-10 (IL-10) was increased after intervention with purslane. The markers of oxidative stress such as superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), glutathione peroxidase (GPx), and total antioxidant capacity (TAC) levels increased, thiobarbituric acid reactive substances (TBARS), reactive oxygen species (ROS) and malondialdehyde (MDA) decreased. Notably, the evidence from clinical trials showed a significant reduction in NF-κβ and CRP after purslane treatment; however, no effect was observed on MDA and TAC. The evidence gathered in this study suggests that purslane exerts anti-inflammatory properties by downregulating NF-κβ, thus suppressing the production of associated pro-inflammatory cytokines. Therefore, purslane may be used as an antioxidant and inflammatory agent for diabetes. However, further clinical evidence with a broader population is required to validate the therapeutic properties of purslane in diabetes. Full article
(This article belongs to the Special Issue Oxidative Stress and Inflammation in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Purslane and its active compounds [<a href="#B26-ijms-25-12276" class="html-bibr">26</a>,<a href="#B29-ijms-25-12276" class="html-bibr">29</a>,<a href="#B31-ijms-25-12276" class="html-bibr">31</a>]. An active compound is found in purslane leaves and stems.</p>
Full article ">Figure 2
<p>PRISMA flow chart depicting study selection, screening and inclusion.</p>
Full article ">
13 pages, 2313 KiB  
Article
Lactobacillus helveticus HY7801 Improves Premenstrual Syndrome Symptoms by Regulating Sex Hormones and Inflammatory Cytokines in a Mouse Model of Metoclopramide-Induced Hyperprolactinemia
by Hyeon-Ji Kim, Ji-Woong Jeong, Joo-Yun Kim, Jae-Jung Shim and Jae-Hwan Lee
Nutrients 2024, 16(22), 3889; https://doi.org/10.3390/nu16223889 - 14 Nov 2024
Viewed by 399
Abstract
Background/Objectives: Premenstrual syndrome (PMS), a clinical condition that manifests in the form of various physical and psychological symptoms, occurs periodically during the luteal phase of the menstrual cycle and reduces quality of life. Methods: Here, we conducted in vitro and in vivo experiments [...] Read more.
Background/Objectives: Premenstrual syndrome (PMS), a clinical condition that manifests in the form of various physical and psychological symptoms, occurs periodically during the luteal phase of the menstrual cycle and reduces quality of life. Methods: Here, we conducted in vitro and in vivo experiments to investigate the effects of Lactobacillus helveticus HY7801 (HY7801) on PMS symptoms. Results: Data from the in vitro experiments showed that HY7801 inhibits prolactin secretion by estradiol-induced GH3 cells, as well as the secretion of pro-inflammatory cytokines by LPS-induced Raw 264.7 cells. Additionally, the oral administration of HY7801 (109 colony-forming units/kg/day) to mice with metoclopramide-induced hyperprolactinemia reduced uterine tissue mass and endometrial thickness, both of which were increased excessively in the presence of prolactin. HY7801 also regulated the serum levels of follicle-stimulating hormone and prostaglandin E1/E2, as well as recovering the progesterone/estradiol ratio. HY7801 also downregulated the serum levels of prolactin and pro-inflammatory cytokines such as interleukin (Il)-6, tumor necrosis factor-alpha (Tnf), and IL-1β. Finally, HY7801 reduced the expression of genes encoding inflammatory cytokines (i.e., Tnf and Il-6), cyclooxygenase-2 (Cox-2), and inducible nitric oxide synthase (iNOS) in mice with hyperprolactinemia. Conclusion: In summary, HY7801 may be a functional bacterium that alleviates PMS symptoms by modulating hormones and inflammatory markers. Full article
(This article belongs to the Special Issue Eating Behavior and Women's Health)
Show Figures

Figure 1

Figure 1
<p>Effect of HY7801 on (<b>A</b>) the viability of GH3 cells and (<b>B</b>) the secretion of prolactin by estradiol (E2)-treated GH3 cells. Data are presented as the mean ± SE. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. untreated group; * <span class="html-italic">p</span> &lt; 0.05 vs. E2-treated group. E2: estradiol; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 + E2.</p>
Full article ">Figure 2
<p>Effects of HY7801 on pro-inflammatory cytokines secreted by LPS-induced Raw 264.7 cells. (<b>A</b>) IL-6; (<b>B</b>) TNF-α. Data are presented as the mean ± SE. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. untreated group; *** <span class="html-italic">p</span> &lt; 0.001 vs. LPS-treated group. IL-6: interleukin-6; TNF-α: tumor necrosis factor-alpha; LPS: lipopolysaccharide; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 + LPS.</p>
Full article ">Figure 3
<p>Effect of HY7801 on physiological indicators in mice with MCP-induced hyperprolactinemia. (<b>A</b>) Dietary intake; (<b>B</b>) water intake; (<b>C</b>) change in body weight; (<b>D</b>) mass of uterine tissue; and (<b>E</b>) mass of spleen tissue. Data are presented as the mean ± SE. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. CON group; * <span class="html-italic">p</span> &lt; 0.05 vs. MCP group. CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">Figure 4
<p>Histological analysis of uterine tissue from mice with MCP-induced hyperprolactinemia. (<b>A</b>) Hematoxylin and eosin-stained sections; 200× magnification; arrows point to the endometrium. (<b>B</b>) Endometrial thickness. Data are presented as the mean ± SE. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. CON group; *** <span class="html-italic">p</span> &lt; 0.001 vs. MCP group. CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">Figure 5
<p>Effect of HY7801 on serum levels of sex hormones in mice with MCP-induced hyperprolactinemia. (<b>A</b>) Prolactin; (<b>B</b>) FSH; and (<b>C</b>) the progesterone/estradiol ratio. Data are presented as the mean ± SE. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. CON group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. MCP group. FSH: follicle-stimulating hormone; CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">Figure 6
<p>Effect of HY7801 on serum levels of pro-inflammatory cytokines and prostaglandin in mice with MCP-induced hyperprolactinemia. (<b>A</b>) IL-6; (<b>B</b>) TNF-α; (<b>C</b>) IL-1β; (<b>D</b>) PGE1; (<b>E</b>) PGE2 levels; and (<b>F</b>) the PGE1/PGE2 ratio. Data are presented as the mean ± SE. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. CON group; * <span class="html-italic">p</span> &lt; 0.05 vs. MCP group. IL-6: interleukin-6; TNF: tumor necrosis factor-alpha; IL-1β: interleukin-1β; PGE1: prostaglandin E1; PGE2: prostaglandin E2; CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">Figure 7
<p>Effect of HY7801 on the expression of mRNA encoding inflammation-related genes in uterine tissues from MCP-induced hyperprolactinemia mice. (<b>A</b>) <span class="html-italic">Il-6</span>; (<b>B</b>) <span class="html-italic">Tnf</span>; (<b>C</b>) <span class="html-italic">Cox-2</span>; and (<b>D</b>) <span class="html-italic">iNOS</span>. Data are presented as the mean ± SE. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. CON group; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. MCP group. <span class="html-italic">Il-6</span>: interleukin-6; <span class="html-italic">Tnf</span>: tumor necrosis factor-alpha; <span class="html-italic">Cox-2</span>: cyclooxygenase-2; <span class="html-italic">iNOS</span>: inducible nitric oxide synthase; CON: non-treatment group; MCP: metoclopramide-induced mice; PFM: prefemin (100 mg/kg/day) + MCP; HY7801: <span class="html-italic">Lactobacillus helveticus</span> HY7801 (10<sup>9</sup> CFU/kg/day) + MCP.</p>
Full article ">
13 pages, 1531 KiB  
Article
Effect of Subconjunctival Injection of Canine Adipose-Derived Mesenchymal Stem Cells on Canine Spontaneous Corneal Epithelial Defects
by Pechchalee Kengkla, Yaowalak Panyasing, Aree Thayananuphat and Nalinee Tuntivanich
Animals 2024, 14(22), 3270; https://doi.org/10.3390/ani14223270 - 13 Nov 2024
Viewed by 360
Abstract
Spontaneous chronic corneal epithelial defects (SCCEDs) are characterized by nonadherent corneal epithelium leading to poor attachment to the corneal stroma. The objective of this study was to characterize corneal outcomes concurrently with the quantification of tumor necrosis factor-alpha (TNF-α) and vascular endothelial growth [...] Read more.
Spontaneous chronic corneal epithelial defects (SCCEDs) are characterized by nonadherent corneal epithelium leading to poor attachment to the corneal stroma. The objective of this study was to characterize corneal outcomes concurrently with the quantification of tumor necrosis factor-alpha (TNF-α) and vascular endothelial growth factor-A (VEGF-A) in tear fluid after the subconjunctival injection of canine adipose-derived mesenchymal stem cells (cAD-MSCs) in canine SCCEDs. Ten eyes with SCCEDs, which were nonresponsive to two rounds of diamond burr debridement, were included in this study. All eyes received a single subconjunctival injection of 1 × 106 cAD-MSCs. Ophthalmic examinations were performed before treatment and on day 7, 14, and 21 after treatment. Tear samples were collected for the quantification of TNF-α and VEGF-A concentrations by a canine multiplex immunoassay. Nine out of ten eyes revealed complete healing by day 21. The mean healing time was 10.89 ± 1.7 days. All eyes showed a decreased degree of ocular discomfort, in accordance with the degree of corneal characteristics. The concentrations of VEGF-A significantly reduced from pre-treatment (4334.91 ± 1275.92 pg/mL) to day 21 post-treatment (3064.61 ± 1028.66 pg/mL). No significant difference in TNF-α concentration was observed before/after treatment. In conclusion, the single use of a subconjunctival injection of cAD-MSCs could be used as an alternative treatment for canine SCCEDs. Full article
(This article belongs to the Section Companion Animals)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Corneal characteristics of 10 canine eyes with SCCEDs before and after treatment with the subconjunctival injection of cAD-MSCs.</p>
Full article ">Figure 2
<p>Bar graph illustrating percentage of corneal epithelial defect (<b>a</b>), corneal neovascularization (<b>b</b>), and corneal opacification area (<b>c</b>) before and after subconjunctival injection of cAD-MSCs; * <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 3
<p>Bar graph illustrating concentrations of TNF-α (<b>a</b>) and VEGF-A (<b>b</b>) (pg/mL) in tear fluid before and after subconjunctival injection of cAD-MSCs; * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
23 pages, 6216 KiB  
Article
Intraperitoneal Administration of S100A8 Ameliorates Experimental Acute Colitis in Rats
by Kano Matsuo, Masaki Ikemoto and Kohki Okada
Biology 2024, 13(11), 916; https://doi.org/10.3390/biology13110916 - 11 Nov 2024
Viewed by 359
Abstract
S100A8 is a protein that is abundant in neutrophils and macrophages (MΦ), but its role in inflammation remains unclear. This study aimed to assess the immunological role(s) of S100A8 in acute intestinal inflammation in rats and its role in MΦ. Rat recombinant S100A8 [...] Read more.
S100A8 is a protein that is abundant in neutrophils and macrophages (MΦ), but its role in inflammation remains unclear. This study aimed to assess the immunological role(s) of S100A8 in acute intestinal inflammation in rats and its role in MΦ. Rat recombinant S100A8 (rr-S100A8, 1.0 mg/kg) was intraperitoneally administered daily to rats with 3% dextran sulfate sodium (DSS) (DSS + A8 group)-induced experimental acute colitis. The histological severity score (6.50 ± 0.51, p = 0.038) in the DSS + A8 group rats remained lower than that (9.75 ± 1.48) of the rats without S100A8 (DSS group) administration. The tumor necrosis factor-alpha (TNF-α) production in the colon tissues of the rats in the DSS + A8 group (4.76 ± 0.90 pg/mL/g, p = 0.042) was significantly suppressed, compared with that of the DSS group (10.45 ± 2.04 pg/mL/g). To stimulate rat peritoneal MΦ, rr-S100A8, the anti-rat S100A8 antibody, and a lipopolysaccharide (LPS) were used in the in vitro experiments. In the MΦ stimulated with rr-S100A8 for 2 h, the mRNA level of intracellular S100A8 (47.41 ± 24.44, p = 0.002) increased in an autocrine manner, whereas that of S100A9 (0.24 ± 0.43, p = 0.782) was not significant. The TNF-α mRNA level in the MΦ treated with LPS and the anti-rat S100A8 antibody significantly increased (102.26 ± 18.60, p = 0.001) compared to that with LPS alone (16.9 ± 8.56). These results indicate that S100A8 can serve as an anti-inflammatory protein in acute inflammation by negatively regulating S100A9 and TNF-α production through inflammatory signaling pathways in MΦ. Full article
(This article belongs to the Section Medical Biology)
Show Figures

Figure 1

Figure 1
<p>In vivo experimental protocol. Five groups are as follows: (1) Normal; (2) DSS; (3) DSS + A8; (4) DSS/Normal; and (5) DSS/A8 (see <a href="#sec2-biology-13-00916" class="html-sec">Section 2</a>).</p>
Full article ">Figure 2
<p>In vitro experimental protocol. Peritoneal MΦ of Wistar rats (<span class="html-italic">n</span> = 10) are isolated as previously described [<a href="#B13-biology-13-00916" class="html-bibr">13</a>]. In vitro experiments were performed under six conditions (1)–(6). After stimulation, mRNAs of S100A8, S100A9, interleukin (IL)-1β, tumor necrosis factor-alpha (TNF-α), IL-10, and tumor growth factor (TGF)-β in macrophages (MΦ) were measured using real-time polymerase chain reaction (PCR) (ΔΔCT method).</p>
Full article ">Figure 3
<p>Alterations in the body weight and colon length of rats. (<b>A</b>) The <span class="html-italic">X</span>-axis represents the experimental period, whereas the <span class="html-italic">Y</span>-axis represents the percentage of fluctuation in the body weight of the three rat groups. (<b>B</b>) The <span class="html-italic">X</span>-axis represents three groups of rats, and the <span class="html-italic">Y</span>-axis represents the colon length (cm) of the three rat groups. Values are presented as the mean ± standard deviation (SD). * <span class="html-italic">p</span> &lt; 0.05 (Normal vs. DSS), ** <span class="html-italic">p</span> &lt; 0.05 (Normal vs. DSS + A8). Normal group (gray), DSS group (red), and DSS + A8 group (blue).</p>
Full article ">Figure 4
<p>Alterations in the disease activity index (DAI) scores of rats. DAI scores in each group were assessed based on established criteria (<a href="#biology-13-00916-t002" class="html-table">Table 2</a>) [<a href="#B31-biology-13-00916" class="html-bibr">31</a>]. The <span class="html-italic">X</span>-axis represents the days after the start of the experiment, and the <span class="html-italic">Y</span>-axis represents the DAI scores [0–4]. Values are presented as the mean ± standard deviation (SD). * <span class="html-italic">p</span> &lt; 0.05 (Normal vs. DSS). Normal group (gray); DSS group (red); and DSS + A8 group (blue).</p>
Full article ">Figure 5
<p>Histological severity (HIS) in the colonic tissues of rats. Panels (<b>A</b>–<b>C</b>) demonstrate microscopic images of the colon tissues in the Normal, DSS, and DSS + A8 groups, respectively, based on the hematoxylin and eosin (H&amp;E) staining, in which all panels were observed with high power magnitude (×200). All microscopic images were observed using a BIOREVO BZ-9000 microscope (Keyence Co., Ltd., Osaka, Japan). Black arrows; non-inflammatory regions, red arrows; inflammatory regions, and light blue arrows; mild inflammatory regions. Black scale bar = 50 μm. Panel (<b>D</b>) demonstrates HIS scores, which are assessed based on established criteria (<a href="#biology-13-00916-t003" class="html-table">Table 3</a>) on day 10 after initiating the experiment [<a href="#B32-biology-13-00916" class="html-bibr">32</a>]. The <span class="html-italic">y</span>-axis represents the HIS scores [0–14]. Values are presented as the mean ± standard deviation (SD). * <span class="html-italic">p</span> &lt; 0.05 (Normal vs. DSS), ** <span class="html-italic">p</span> &lt; 0.05 (Normal vs. DSS + A8). Normal group (gray); DSS group (red); and DSS + A8 group (blue).</p>
Full article ">Figure 6
<p>Analysis of S100A8 and S100A9 expressions in colon tissue using enzyme-linked immunosorbent assay (ELISA). Panels (<b>A</b>,<b>B</b>) demonstrate the relative amount of S100A8 and S100A9, respectively. The <span class="html-italic">Y</span>-axis represents the relative amount of the expression level (absorbance) of S100A8 and S100A9 in the colon tissue of the Normal group as “1”. Values are presented as the mean ± standard deviation (SD). * <span class="html-italic">p</span> &lt; 0.05 (Normal vs. DSS), ** <span class="html-italic">p</span> &lt; 0.05 (Normal vs. DSS + A8). DSS group (red) and DSS + A8 group (blue). Panel (<b>C</b>) demonstrates the mRNA expression levels of S100A8 (upper panel) and S100A9 (middle panel) in the colon. The expression level of β-actin (lower panel) in the colon is used as an internal control. Images are captured using a ChemiDoc XRS+ (Bio-Rad Laboratories, Inc.). The images of the original PCR and agarose gel electrophoresis results are shown in <a href="#app1-biology-13-00916" class="html-app">Figure S1</a>.</p>
Full article ">Figure 7
<p>Fluorescent immunohistochemical staining (FICS) of S100A8 and S100A9 in rat colon tissues. Expression of S100A8 (red, tetramethylrhodamine [TRITC]) and S100A9 (green, FITC) in colon tissues were assessed, in which nuclei (blue, 4′,6-diamidino-2-phenylindole [DAPI]) are counterstained. Panels (<b>A</b>–<b>D</b>) (Normal); panels (<b>E</b>–<b>H</b>) (DSS); and panels (<b>I</b>–<b>L</b>) (DSS + A8). Panels (<b>A</b>,<b>E</b>,<b>I</b>) (DAPI); panels (<b>B</b>,<b>F</b>,<b>J</b>) (TRITC); panels (<b>C</b>,<b>G</b>,<b>K</b>) (FITC); and panels (<b>D</b>,<b>H</b>,<b>L</b>) (merged). All microscopic images were obtained using a BIOREVO BZ-9000 microscope (Keyence Co., Ltd., Osaka, Japan) with high power magnitude (×100). White scale bar = 50 μm.</p>
Full article ">Figure 8
<p>Immunohistochemical (IHC) staining of neutrophil elastase (NE) in colon tissues. The localization of neutrophils in the colon tissues of Normal (<b>A</b>), DSS (<b>B</b>), and DSS + A8 (<b>C</b>) groups was observed by detecting NE expression in neutrophils. Positive reactions (brown, red arrows) are stained with diaminobenzidine (DAB). All microscopic images are observed using a BIOREVO BZ-9000 microscope (Keyence Co., Ltd., Osaka, Japan) with high power magnitude (×100). Black scale bar = 50 μm.</p>
Full article ">Figure 9
<p>Measurement of inflammatory cytokines in the serum and colon tissue of rats using enzyme-linked immunosorbent assay (ELISA). Inflammatory cytokines (tumor necrosis factor [TNF]-α, interleukin [IL]-6, and IL-1β) in the serum (panel (<b>A</b>)) and colon tissue (panel (<b>B</b>)) were measured using ELISA. The <span class="html-italic">Y</span>-axis represents the concentration of each cytokine (pg/mL or pg/mL/g). Values are presented as the mean ± standard deviation (SD). * <span class="html-italic">p</span> &lt; 0.05 (Normal vs. DSS), ** <span class="html-italic">p</span> &lt; 0.05 (Normal vs. DSS + A8). Normal group (gray); DSS group (red); and DSS + A8 group (blue).</p>
Full article ">Figure 10
<p>Alterations in the body weight and colon length of rats. (<b>A</b>) The <span class="html-italic">X</span>-axis represents the experimental period, and the <span class="html-italic">Y</span>-axis represents the percentage of alteration in the body weight of rats in the two groups. (<b>B</b>) Pictures of representative colon tissues from the DSS/Normal (upper panel) and DSS/A8 (lower panel) groups. (<b>C</b>) The <span class="html-italic">Y</span>-axis represents the colon length (cm) of the rats in the two groups. Values are presented as the mean ± standard deviation (SD). DSS/Normal group (pink) and DSS/A8 group (green).</p>
Full article ">Figure 11
<p>Alterations in the disease activity index (DAI) scores of rats. DAI scores were assessed based on established criteria (<a href="#biology-13-00916-t002" class="html-table">Table 2</a>) [<a href="#B31-biology-13-00916" class="html-bibr">31</a>]. The <span class="html-italic">Y</span>-axis represents the DAI scores [0–4] on every experimental day. Values are presented as the mean ± standard deviation (SD). * <span class="html-italic">p</span> &lt; 0.05 (DSS/Normal vs. DSS/A8). DSS/Normal group (pink) and DSS/A8 group (green).</p>
Full article ">Figure 12
<p>Histological assessment of the severity in colonic tissues of rats based on hematoxylin and eosin (H&amp;E) staining. Panels (<b>A</b>,<b>B</b>) demonstrate microscopic images of the colon tissues in DSS/Normal and DSS/A8 groups, respectively. Red arrows; inflammatory regions and light blue arrows; mild inflammatory regions. All the microscopic images are observed using a BIOREVO BZ-9000 microscope (Keyence Co., Ltd., Osaka, Japan). All the panels are observed at high magnification (×200). Black scale bar = 50 μm. Panel (<b>C</b>) demonstrates histological severity (HIS) scores, which are assessed based on the established criteria, on day 20 after initiating the experiment [<a href="#B32-biology-13-00916" class="html-bibr">32</a>]. The <span class="html-italic">Y</span>-axis represents the HIS scores [0–14]. Values are presented as the mean ± standard deviation (SD). DSS/Normal group (pink) and DSS/A8 group (green).</p>
Full article ">Figure 13
<p>Analysis of S100A8 and S100A9 mRNA expression in macrophages (MΦ) using real-time polymerase chain reaction (PCR). The mRNA expression levels of the S100A8 and S100A9 are indicated by panels (<b>A</b>,<b>B</b>), respectively. <span class="html-italic">Y</span>-axis represents the relative amount of the expression level of S100A8 and S100A9 mRNAs in the MΦ as the expression in the cells with no stimulation as “1”. Values are presented as the mean ± standard deviation (SD). * <span class="html-italic">p</span> &lt; 0.05 (No stimulation vs. each stimulus condition).</p>
Full article ">Figure 14
<p>Analysis of cytokine mRNA expression in macrophages (MΦ) using real-time polymerase chain reaction (PCR). Panels (<b>A</b>–<b>D</b>) demonstrate interleukin (IL)-1β, tumor necrosis factor (TNF)-α, IL-10, and tumor growth factor (TGF)-β mRNA levels, respectively. The <span class="html-italic">Y</span>-axis represents the relative amount against the expression level of each cytokine in MΦ with no stimulation as referred to as “1”. Values are presented as the mean ± standard deviation (SD). * <span class="html-italic">p</span> &lt; 0.05 (no stimulation vs. each stimulus condition).</p>
Full article ">
14 pages, 1802 KiB  
Article
Iota-Carrageenan from Marine Alga Solieria filiformis Prevents Naproxen-Induced Gastrointestinal Injury via Its Antioxidant and Anti-Inflammatory Activities
by João L. S. Pinheiro, Willer M. Sousa, Lucas H. M. Rodrigues, Francisco F. Bezerra, Cecília L. O. A. Cunha, Victória M. R. Santos, Samara R. B. D. Oliveira, Rudy D. Bingana, André Luiz. R. Barbosa, Marcellus H. L. P. Souza, Ana Lúcia P. Freitas and Renan O. S. Damasceno
Biomedicines 2024, 12(11), 2574; https://doi.org/10.3390/biomedicines12112574 - 10 Nov 2024
Viewed by 594
Abstract
Background: Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used in therapy due to their anti-inflammatory and analgesic properties. However, their clinical use is often associated with gastrointestinal complications. Thus, this study aimed to investigate the protective effect of a sulfated iota-carrageenan isolated from the [...] Read more.
Background: Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used in therapy due to their anti-inflammatory and analgesic properties. However, their clinical use is often associated with gastrointestinal complications. Thus, this study aimed to investigate the protective effect of a sulfated iota-carrageenan isolated from the marine alga Solieria filiformis (IC-Sf) against naproxen-induced gastrointestinal injury. Methods: Parameters of gastrointestinal injury, secretory and motor functions, and toxicity were evaluated. Results: The results demonstrated that IC-Sf significantly reduced naproxen-induced gastrointestinal macroscopic injury, with a maximum effect observed at 30 mg/kg. IC-Sf also preserved gastrointestinal antioxidant defense and prevented lipid peroxidation, with a reduction in the non-protein sulfhydryl group (NP-SH) and malondialdehyde (MDA) concentrations induced by naproxen. Additionally, IC-Sf mitigated naproxen-induced gastrointestinal inflammation, as evidenced by reduced myeloperoxidase (MPO) activity, tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β). IC-Sf did not alter gastric secretion or gastrointestinal motility. In addition, the animals treated with IC-Sf did not present toxic effects. Conclusions: In conclusion, IC-Sf protected the gastrointestinal tract against the harmful effects of naproxen by inhibiting the inflammatory response and lipid peroxidation, suggesting its potential as a new therapeutic agent or food additive. Full article
(This article belongs to the Special Issue Health-Related Applications of Natural Molecule Derived Structures)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Biochemical structure of iota-carrageenan from <span class="html-italic">S. filiformis</span> and its effect against naproxen-induced gastrointestinal macroscopic injury. (<b>A</b>) Structure of IC-Sf; (<b>B</b>) Gastric and (<b>C</b>) Intestinal injury. Different letters (a–c) on the bar represent significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Effect of IC-Sf on naproxen-induced gastrointestinal oxidative stress. (<b>A</b>) NP-SH levels and (<b>B</b>) MDA concentration in the gastric mucosa; and (<b>C</b>) NP-SH levels and (<b>D</b>) MDA concentration in small intestine mucosa. Different letters (a–c) on the bar represent significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Effect of IC-Sf on naproxen-induced gastrointestinal inflammation. (<b>A</b>,<b>B</b>) MPO activity; (<b>C</b>,<b>D</b>) IL-1β levels; (<b>E</b>,<b>F</b>) TNF-α levels. Different letters (a,b) on the bar represent significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Photomicrograph of organs. Heart (<b>A</b>), spleen (<b>B</b>), kidney (<b>C</b>), liver (<b>D</b>), and lung (<b>E</b>) of control (<b>A1</b>–<b>E1</b>) or IC-Sf-treated (<b>A2</b>–<b>E2</b>) animals stained with hematoxylin and eosin (HE). The analysis revealed no differences between groups. The heart presented normal cardiac muscle fibers (arrows), and it was possible to distinguish the white pulp (black arrows) and the red pulp (red arrows) in the spleen. The renal glomeruli (black arrows) and Bowman’s space (red arrows) presented the expected conformation. In the liver, there were no morphological alterations in hepatocytes (black arrows) or Kupffer cells (red arrows), while well-preserved alveolar structures were observed in the lungs (arrows). Tissue sections were observed under a light microscope at 400×.</p>
Full article ">
14 pages, 577 KiB  
Review
Infusion Reactions to Infliximab in Pediatric Patients with Inflammatory Bowel Disease
by Rajmohan Dharmaraj, Tess Pei Lemon, Rasha Elmaoued, Ricardo Orlando Castillo and Razan Alkhouri
Children 2024, 11(11), 1366; https://doi.org/10.3390/children11111366 - 10 Nov 2024
Viewed by 486
Abstract
Infliximab (IFX) is a recombinant DNA-derived chimeric IgG monoclonal antibody protein that inhibits tumor necrosis factor alpha (TNF-α). IFX, like other agents derived from foreign proteins, can cause infusion reactions both during and after the infusion. The incidence of infusion reactions ranges between [...] Read more.
Infliximab (IFX) is a recombinant DNA-derived chimeric IgG monoclonal antibody protein that inhibits tumor necrosis factor alpha (TNF-α). IFX, like other agents derived from foreign proteins, can cause infusion reactions both during and after the infusion. The incidence of infusion reactions ranges between 0% and 15% in pediatric patients. The potential underlying mechanisms for these reactions may include anaphylaxis and anaphylactoid reactions, cytokine release syndrome, serum sickness-like reactions, and the development of antibodies against IFX. Several precautions can help reduce the risk of a new infusion reaction, such as a gradual increase in the infusion rate, scheduled infusions, and administering premedication or immunomodulators alongside IFX. Acute mild to moderate reactions often resolve spontaneously after a temporary cessation of the infusion or reduction in the infusion rate. Strategies like graded dose challenges and premedication can be utilized to prevent recurrence. In cases of severe reactions, desensitization or switching to an alternative biologic may be considered. This article aims to review the most recent guidelines for managing IFX-related infusion reactions in pediatric patients with inflammatory bowel disease (IBD), relying on the best available evidence. Full article
Show Figures

Figure 1

Figure 1
<p>Algorithm for management of infliximab infusion reactions.</p>
Full article ">
17 pages, 11413 KiB  
Article
Levamisole Ameliorates Rheumatoid Arthritis by Downregulating the PI3K/Akt Pathway in SD Rats
by Mu Guo, Xiangbin Yu, Zesheng Yang, Hanlu Zheng, Jiahui Zhang, Junxiang Wang, Yiqi Liao, Weirui Huang, Zhaolong Lin, Yingxue Yan, Nengfu Qiu, Jianmin Chen and Yue Yu
Pharmaceuticals 2024, 17(11), 1504; https://doi.org/10.3390/ph17111504 - 8 Nov 2024
Viewed by 473
Abstract
Background/Objectives: Rheumatoid arthritis (RA) is a systemic chronic autoimmune disease characterized by a protracted course, high rates of morbidity, and disability yet lacks effective therapeutic modalities. Levamisole (LVM), an immunomodulatory drug, has been clinically reported for its potential in RA treatment, while its [...] Read more.
Background/Objectives: Rheumatoid arthritis (RA) is a systemic chronic autoimmune disease characterized by a protracted course, high rates of morbidity, and disability yet lacks effective therapeutic modalities. Levamisole (LVM), an immunomodulatory drug, has been clinically reported for its potential in RA treatment, while its therapeutic mechanism toward RA remains to be elucidated. Hence, this study provides theoretical support for the application of LVM in the treatment of RA. Methods: This study employed male Sprague–Dawley (SD) rats to construct the adjuvant-induced arthritis (AIA) model, administering LVM orally (5 mg/kg, 15 mg/kg, and 45 mg/kg) for 25 days. An evaluation of LVM’s therapeutic effects on RA was conducted through arthritis index scores, paw pad thickness, paw volume, hematoxylin and eosin (H&E) staining, 3D microcomputed tomography (micro-CT) scans, serum levels of pro-/anti-inflammatory cytokines, and serum biochemical indicators. Western blotting and immunohistochemistry staining were utilized to measure the expression levels of phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) proteins in synovial and ankle joint tissues. Results: Treatment with the median dose of LVM (15 mg/kg, M-LVM) significantly reduced the arthritis index (p < 0.01), paw pad thickness (p < 0.001), and paw volume (p < 0.01) without affecting body weight. Additionally, M-LVM alleviated inflammatory lesions in the synovium and ankle joints and also normalized serum levels of interleukin-1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-beta (TGF-β). The Model group exhibited significant increases in serum levels of alkaline phosphatase (ALP) (p < 0.01), creatine kinase (CK) (p < 0.05), and glucose (GLU) (p < 0.001) compared with the Control group; however, M-LVM effectively regulated these parameters to normal levels. Western blotting and immunohistochemistry staining revealed that PI3K-/Akt-related proteins were highly expressed in the synovial and ankle joint tissues of rats in the Model group, while treatment with M-LVM significantly reduced the expression of these proteins. Furthermore, histological examination of major organs (heart, liver, lungs, kidneys, and thymus) showed no significant pathological changes, with the exception of the spleen, where M-LVM ameliorated splenic lesions. Conclusions: We demonstrate that LVM at an optimal dose substantially relieves synovitis and bone erosion in AIA rats by inhibiting the PI3K/Akt signaling pathway. Full article
(This article belongs to the Special Issue Pyrazole and Thiazole Derivatives in Medicinal Chemistry)
Show Figures

Figure 1

Figure 1
<p>The chemical structures of (<b>A</b>) MTX and (<b>B</b>) LVM.</p>
Full article ">Figure 2
<p>LVM alleviated RA symptoms in AIA rat model in a dose-dependent manner. (<b>A</b>) The body weight, (<b>B</b>) arthritis index, (<b>C</b>) foot pad thickness, and (<b>D</b>) paw volume were statistically analyzed. (<b>E</b>) Direct view of swelling in the right hind foot of rats. (<b>F</b>) Representative three-dimensional reconstruction images of rats’ paws from different treated groups in micro-CT. (<b>G</b>) Histological images depicting the H&amp;E staining of ankle joint tissues (scale bar, 100 μm). (<b>H</b>) Histological images depicting the H&amp;E staining of synovial membrane (scale bar, 100 μm). The red arrows indicate changes in the organizational structure. All data are shown as mean ± SEM. *** <span class="html-italic">p</span> &lt; 0.001 when comparing with the Control group using an unpaired Student’s <span class="html-italic">t</span>-test; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 when comparing with the Model group using one-way ANOVA followed by Dunnett’s post hoc test. Definitions of abbreviations for each dosing group: Control, blank control group; Model, model group; MTX, positive drug group; L-LVM, 5 mg/kg LVM group; M-LVM, 15 mg/kg LVM group; H-LVM, 45 mg/kg LVM group. <span class="html-italic">n</span> = 6 in each group.</p>
Full article ">Figure 3
<p>The levels of IL-1β, TNF-α, and TGF-β in the serum were measured by enzyme-linked immunosorbent assay (ELISA). All data are shown as mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 when comparing with the Control group using an unpaired Student’s <span class="html-italic">t</span>-test; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 when comparing with the Model group using one-way ANOVA followed by Dunnett’s post hoc test. Definitions of abbreviations for each dosing group: Control, blank control group; Model, model group; MTX, positive drug group; L-LVM, 5 mg/kg LVM group; M-LVM, 15 mg/kg LVM group; H-LVM, 45 mg/kg LVM group. <span class="html-italic">n</span> = 6 in each group.</p>
Full article ">Figure 4
<p>The levels of serum biochemical markers were measured by automatic biochemical analyzer. All data are shown as mean ± SEM. * <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 when comparing with the Control group using an unpaired Student’s <span class="html-italic">t</span>-test; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 when comparing with the Model group using one-way ANOVA followed by Dunnett’s post hoc test. Definitions of abbreviations for each dosing group: Control, blank control group; Model, model group; MTX, positive drug group; L-LVM, 5 mg/kg LVM group; M-LVM, 15 mg/kg LVM group; H-LVM, 45 mg/kg LVM group. <span class="html-italic">n</span> = 6 in each group.</p>
Full article ">Figure 5
<p>M-LVM inhibited PI3k/Akt pathway in ankle joint tissues of AIA rats. (<b>A</b>) The protein levels of p-PI3K, PI3K, p-Akt, and Akt in ankle joint tissues were detected by Western blotting. (<b>B</b>) The protein levels of PI3K and Akt in ankle joint tissues were analyzed by immunostaining (scale bar, 100 μm). All data are shown as mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 when comparing with the Control group using an unpaired Student’s <span class="html-italic">t</span>-test; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 when comparing with the Model group using one-way ANOVA followed by Dunnett’s post hoc test. Definitions of abbreviations for each dosing group: Control, blank control group; Model, model group; MTX, positive drug group; L-LVM, 5 mg/kg LVM group; M-LVM, 15 mg/kg LVM group; H-LVM, 45 mg/kg LVM group. <span class="html-italic">n</span> = 3 in each group.</p>
Full article ">Figure 6
<p>M-LVM inhibited PI3k/Akt pathway in synovial tissues of AIA rats. (<b>A</b>) The protein levels of p-PI3K, PI3K, p-Akt, and Akt in synovial tissues were detected by Western blotting. (<b>B</b>) The protein levels of PI3K and Akt in synovial tissues were analyzed by immunostaining (scale bar, 100 μm). All data are shown as mean ± SEM. * <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 when comparing with the Control group using an unpaired Student’s <span class="html-italic">t</span>-test; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 when comparing with the Model group using one-way ANOVA followed by Dunnett’s post hoc test. Definitions of abbreviations for each dosing group: Control, blank control group; Model, model group; MTX, positive drug group; L-LVM, 5 mg/kg LVM group; M-LVM, 15 mg/kg LVM group; H-LVM, 45 mg/kg LVM group. <span class="html-italic">n</span> = 3 in each group.</p>
Full article ">Figure 7
<p>Evaluation of potential adverse effects of LVM in AIA rats. (<b>A</b>) The results of H&amp;E staining (scale bar, 100 μm). (<b>B</b>) Organ coefficient of major organs. All data are shown as mean ± SEM. *** <span class="html-italic">p</span> &lt; 0.001 when comparing with the Control group using an unpaired Student’s <span class="html-italic">t</span>-test; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 when comparing with the Model group using one-way ANOVA followed by Dunnett’s post hoc test. Definitions of abbreviations for each dosing group: Control, blank control group; Model, model group; MTX, positive drug group; L-LVM, 5 mg/kg LVM group; M-LVM, 15 mg/kg LVM group; H-LVM, 45 mg/kg LVM group. <span class="html-italic">n</span> = 6 in each group.</p>
Full article ">Figure 8
<p>The experimental schematic.</p>
Full article ">
17 pages, 4942 KiB  
Article
Anti-Inflammatory Effects and Metabolomic Analysis of Ilex Rotunda Extracted by Supercritical Fluid Extraction
by Duc Dat Le, Young Su Jang, Vinhquang Truong, Thientam Dinh, Thinhulinh Dang, Soojung Yu and Mina Lee
Int. J. Mol. Sci. 2024, 25(22), 11965; https://doi.org/10.3390/ijms252211965 - 7 Nov 2024
Viewed by 327
Abstract
Ilex rotunda is a famous medicinal plant with many ethnopharmacological uses. It is traditionally employed for treating inflammation and cardiovascular diseases. In this study, we established green technology to extract the leaves and twigs of I. rotunda. The obtained extracts and [...] Read more.
Ilex rotunda is a famous medicinal plant with many ethnopharmacological uses. It is traditionally employed for treating inflammation and cardiovascular diseases. In this study, we established green technology to extract the leaves and twigs of I. rotunda. The obtained extracts and their fractions were evaluated for their anti-inflammatory potential. In cytokine assays, the extract, n-hexane (H), methylene chloride (MC), and EtOAc (E) fractions of the twigs of I. rotunda significantly inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO), interleukin (IL)-6, and tumor necrosis factor (TNF)-α production in RAW264.7 macrophages. Furthermore, the extract, H, and MC fractions of the leaves of I. rotunda modulated cytokine expression by downregulating LPS-induced NO, IL-6, and TNF-α production in RAW264.7 macrophages. Western blotting analysis revealed that the extracts and fractions of the leaves and twigs of I. rotunda inhibited inflammatory cytokines by inactivating nuclear factor kappa B (NFκB) action by reducing the phosphorylation of transcript factor (p65) and nuclear factor-kappa B inhibitor alpha (IκBα) degradation, or by inactivating mitogen-activated protein kinase (MAPK) through the p38 or ERK signaling pathways via the active ingredients of the leaves and twigs of I. rotunda. Ultra-high-resolution liquid chromatography–Orbitrap mass analysis (UHPLC–ESI-Orbitrap-MS/MS)-based molecular networking, in cooperation with social open platform-guided isolation and dereplication, led to the identification of metabolites in this plant. Our findings indicate that the leaves and twigs of I. rotunda could be promising candidates for developing therapeutic strategies to treat anti-inflammatory diseases. Full article
(This article belongs to the Special Issue Molecular Pharmacology of Medicinal Plants)
Show Figures

Figure 1

Figure 1
<p>Antioxidative effects of leaf extract and fractions (<b>A</b>,<b>C</b>) and twig extract and fractions (<b>B</b>,<b>D</b>) were accessed by scavenging activity of DPPH and ABTS radicals. Samples were assayed at 10 and 100 µg/mL in triplicates. Values are expressed as means ± SD. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to positive control (Ascorbic acid, AA).</p>
Full article ">Figure 2
<p>Cytotoxic effects of leaf (<b>A</b>,<b>B</b>) and twig (<b>C</b>,<b>D</b>) extracts and their fractions on cell survival at different concentrations. Cell viability was assessed using the MTT assay. RAW264.7 cells were treated with LPS or samples in a dose-dependent manner at concentrations of 10, 20, 30, 50, and 100 µg/mL and repeated three times. Values are expressed as means ± SD. Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01, compared to the control (CTL).</p>
Full article ">Figure 3
<p>The inhibitory effects of the leaf and twig extracts and their fractions on NO (<b>A</b>,<b>B</b>), IL-6 (<b>C</b>,<b>D</b>), and TNF-α (<b>E</b>,<b>F</b>) production induced by LPS-activated RAW264.7 cells. Cells were pretreated with tested samples (10, 20, and 30 µg/mL) for 1 h and then stimulated with LPS (5 ng/mL) for 24 h. NO, IL-6, and TNF-α production in the culture media were quantified using the Griess assay and an enzyme immunoassay (EIAISA) kit, respectively. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to controls.</p>
Full article ">Figure 4
<p>Western blot analysis of the effects of leaf extract and fractions (<b>A</b>) and twig extract and fractions (<b>B</b>) on iNOS, COX-2 expression levels. LPS-induced RAW264.7 macrophages were pretreated with samples at concentrations of 30 μg/mL and incubated for 6 h. Total proteins were then isolated, separated by SDS-PAGE, and immunoblotted using specific p-p65, p-IκBα, and IκBα antibodies. β-Actin served as an internal control. Relative optical density ratio vs. β-actin or total form was determined using a densitometric analysis program (Bio-Rad Quantity One Software, version 4.6.3 (Basic), Bio-Rad Laboratories Inc., Hercules, CA, USA), normalized to internal control. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 compared to non-sample in LPS stimulation.</p>
Full article ">Figure 5
<p>Inhibitory effects of leaf extract and fractions (<b>A</b>) and twig extract and fractions (<b>B</b>) on LPS-induced NF-κB signaling pathway in RAW264.7 macrophages. Cells were pretreated with MC fraction from twigs (20 μg/mL) and other samples (30 μg/mL) for 1 h, followed by LPS stimulation for 15 min. Proteins were isolated, separated using SDS-PAGE, and immunoblotted with antibodies specific to p-p65, p-IκBα, IκBα. β-actin was served as internal control. Relative optical density ratio versus β-actin or the total form was determined using densitometric analysis software (Bio-Rad Quantity One Software, version 4.6.3 (Basic), Bio-Rad Laboratories Inc., Hercules, CA, USA) and normalized against internal control. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, compared to non-sample under LPS stimulation.</p>
Full article ">Figure 6
<p>The inhibitory effects of leaf extract and fractions (<b>A</b>) and twig extract and fractions (<b>B</b>) on the LPS-induced MAPK signaling pathway in RAW264.7 macrophages. After pretreating cells with the MC fraction of twigs (20 μg/mL) and other samples (30 μg/mL) for 1 h, cells were stimulated with LPS for 15 min. Total proteins were then isolated, separated by SDS-PAGE, and immunoblotted using specific antibodies for p-p38, p-ERK, and ERK. β-Actin was served as an internal control. The relative optical density ratio vs. β-actin or total form was determined using a densitometric analysis program (Bio-Rad Quantity One Software, version 4.6.3 (Basic), Bio-Rad Laboratories Inc., Hercules, CA, USA) normalized to the internal control. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to the control in LPS stimulation.</p>
Full article ">Figure 7
<p>The cell viability (<b>A</b>,<b>B</b>) and inhibitory effects of leaf and twig extracts and their fractions on IL-2 production (<b>C</b>,<b>D</b>) induced by CD3-CD28 Jurkat T cells. Cells were pretreated with the samples (10, 50, and 100 µg/mL) for 1 h and subsequently stimulated with CD3-CD28 (7 µg/mL and 2 µg/mL CD3 and CD28, respectively) for 24 h. IL-2 production in the culture media was quantified using an enzyme-linked immunosorbent assay (ELISA) kit. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to the non-sample control in CD3-CD28 stimulation.</p>
Full article ">Figure 8
<p>The chromatograms of H (A, black), E (B, red), B (C, green), MC (D, blue), and W fractions (E, yellow) and extracts (F, magenta) of leaves of <span class="html-italic">I</span>. <span class="html-italic">rotunda</span> detected in TIC for negative ion mode.</p>
Full article ">Figure 9
<p>Feature-based molecular network of extract (Red) and fractions [H: Blue; MC: Orange; E: Violet; B: Dark Green; W: Yellow] of twigs (GNPS molecular network with job ID = 96ad1b5bbed84c8bb073d8c882f3897b) of <span class="html-italic">I</span>. <span class="html-italic">rotunda</span>.</p>
Full article ">Figure 10
<p>Feature-based molecular network of extract (Neon Blue) and fractions [H: Black; MC: Purple; E: Magenta; B: Yellow; W: Blue] of leaves (GNPS molecular network with job ID = cfa6443e13824edf8132a3f391ad91f3) of <span class="html-italic">I</span>. <span class="html-italic">rotunda</span>.</p>
Full article ">
25 pages, 21810 KiB  
Article
Morphofunctional Features of Glomeruli and Nephrons After Exposure to Electrons at Different Doses: Oxidative Stress, Inflammation, Apoptosis
by Grigory Demyashkin, Sergey Koryakin, Mikhail Parshenkov, Polina Skovorodko, Matvey Vadyukhin, Zhanna Uruskhanova, Yulia Stepanova, Vladimir Shchekin, Artem Mirontsev, Vera Rostovskaya, Sergey Ivanov, Petr Shegay and Andrei Kaprin
Curr. Issues Mol. Biol. 2024, 46(11), 12608-12632; https://doi.org/10.3390/cimb46110748 - 6 Nov 2024
Viewed by 467
Abstract
Kidney disease has emerged as a significant global health issue, projected to become the fifth-leading cause of years of life lost by 2040. The kidneys, being highly radiosensitive, are vulnerable to damage from various forms of radiation, including gamma (γ) and X-rays. However, [...] Read more.
Kidney disease has emerged as a significant global health issue, projected to become the fifth-leading cause of years of life lost by 2040. The kidneys, being highly radiosensitive, are vulnerable to damage from various forms of radiation, including gamma (γ) and X-rays. However, the effects of electron radiation on renal tissues remain poorly understood. Given the localized energy deposition of electron beams, this study seeks to investigate the dose-dependent morphological and molecular changes in the kidneys following electron irradiation, aiming to address the gap in knowledge regarding its impact on renal structures. The primary aim of this study is to conduct a detailed morphological and molecular analysis of the kidneys following localized electron irradiation at different doses, to better understand the dose-dependent effects on renal tissue structure and function in an experimental model. Male Wistar rats (n = 75) were divided into five groups, including a control group and four experimental groups receiving 2, 4, 6, or 8 Gray (Gy) of localized electron irradiation to the kidneys. Biochemical markers of inflammation (interleukin-1 beta [IL-1β], interleukin-6 [IL-6], interleukin-10 [IL-10], tumor necrosis factor-alpha [TNF-α]) and oxidative stress (malondialdehyde [MDA], superoxide dismutase [SOD], glutathione [GSH]) were measured, and morphological changes were assessed using histological and immunohistochemical techniques (TUNEL assay, caspase-3). The study revealed a significant dose-dependent increase in oxidative stress, inflammation, and renal tissue damage. Higher doses of irradiation resulted in increased apoptosis, early stages of fibrosis (at high doses), and morphological changes in renal tissue. This study highlights the dose-dependent effects of electrons on renal structures, emphasizing the need for careful consideration of the dosage in clinical use to minimize adverse effects on renal function. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

Figure 1
<p>Design of the experiment. Special notations: (<b>A</b>)—Male Wistar rats (9–10 weeks old) were randomly assigned to five groups after a 7-day quarantine period. These groups included one control group (intact) and four experimental groups, each receiving a different dose of electron irradiation (2 Gy, 4 Gy, 6 Gy, and 8 Gy) targeted at the abdomino-pelvic region; (<b>B</b>)—irradiation was performed using a NOVAC-11 pulsed electron accelerator. Specific doses were administered with careful shielding to protect surrounding tissues; (<b>C</b>)—following irradiation, blood samples were collected from the animals for biochemical analysis (7 days post-irradiation). The evaluation of blood biochemical parameters was conducted according to the established research methodology; (<b>D</b>)—morphological examinations and organ homogenate studies were performed post-irradiation, following the procedures detailed in the research methodology (7 days post-irradiation).</p>
Full article ">Figure 2
<p>Specialized patented restraint devices (sleds), developed by the Laboratory of Radiation Pathomorphology of the A.F. Tsyb Medical Radiological Research Center.</p>
Full article ">Figure 3
<p>Comparison of body weight and kidney mass across experimental groups measured at 7 days post-irradiation. All data are presented as mean ± SD. Statistically significant differences are indicated by symbols: *—comparison with control group (<span class="html-italic">p</span> &lt; 0.05). (<b>A</b>) Body weight of animals in experimental groups: the body weight of animals decreased progressively with increasing doses of electron irradiation, with the most significant reduction observed at 8 Gy. (<b>B</b>) Kidney mass in experimental groups: kidney mass showed a dose-dependent decrease, with the highest reduction occurring at 8 Gy compared to the control group.</p>
Full article ">Figure 4
<p>Levels of different cytokines in blood of experimental groups measured at 7 days post-irradiation: (<b>A</b>)—data for IL-1β; (<b>B</b>)—data for IL-6; (<b>C</b>)—data for TNF-α; (<b>D</b>)—data for IL-10. Data are presented as mean ± SD. Experimental groups are numbered according to the study design. Statistically significant differences are indicated by symbols: *—comparison with control group (<span class="html-italic">p</span> &lt; 0.05); **—comparison with control group (<span class="html-italic">p</span> &lt; 0.01); ***—comparison with control group (<span class="html-italic">p</span> &lt; 0.001); †—comparison between Group II (2 Gy) and Group IV (8 Gy) (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4 Cont.
<p>Levels of different cytokines in blood of experimental groups measured at 7 days post-irradiation: (<b>A</b>)—data for IL-1β; (<b>B</b>)—data for IL-6; (<b>C</b>)—data for TNF-α; (<b>D</b>)—data for IL-10. Data are presented as mean ± SD. Experimental groups are numbered according to the study design. Statistically significant differences are indicated by symbols: *—comparison with control group (<span class="html-italic">p</span> &lt; 0.05); **—comparison with control group (<span class="html-italic">p</span> &lt; 0.01); ***—comparison with control group (<span class="html-italic">p</span> &lt; 0.001); †—comparison between Group II (2 Gy) and Group IV (8 Gy) (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 5
<p>Levels of different markers of oxidative stress in kidney homogenate of experimental groups (7 days post-irradiation): (<b>A</b>)—data for MDA; (<b>B</b>)—data for SOD; (<b>C</b>)—data for GSH. Data are presented as mean ± SD. Experimental groups are numbered according to the study design. Statistically significant differences are indicated by symbols: *—comparison with control group (<span class="html-italic">p</span> &lt; 0.05); **—comparison with control group (<span class="html-italic">p</span> &lt; 0.01); ***—comparison with control group (<span class="html-italic">p</span> &lt; 0.001); †—comparison between group II (2 Gy) and group IV (8 Gy) (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>A glomerulus of a rat from the control group; stain—hematoxylin and eosin, magnified ×400.</p>
Full article ">Figure 7
<p>Kidneys of rats from experimental groups at different radiation doses, evaluated 7 days post-irradiation; stain—hematoxylin and eosin; different magnification. On the slides: dilation of Bowman’s capsule (*), vacuolization (∆), dystrophic changes in nephron tubules (<b>□</b>), perivascular and periglomerular edema (◊), mild inflammatory (●).</p>
Full article ">Figure 7 Cont.
<p>Kidneys of rats from experimental groups at different radiation doses, evaluated 7 days post-irradiation; stain—hematoxylin and eosin; different magnification. On the slides: dilation of Bowman’s capsule (*), vacuolization (∆), dystrophic changes in nephron tubules (<b>□</b>), perivascular and periglomerular edema (◊), mild inflammatory (●).</p>
Full article ">Figure 8
<p>Kidneys of rats from experimental groups at different radiation doses, evaluated 7 days post-irradiation; stain—Masson’s trichrome; magn. ×40.</p>
Full article ">Figure 9
<p>The kidney of a rat from 8 Gy group (7 days post-irradiation); stain—Masson’s trichrome; magn.: left ×100, right ×200. On the slides: mild fibrosis (*).</p>
Full article ">Figure 10
<p>TUNEL staining of kidney tissue of all experiment groups (7 days post-irradiation): TUNEL-positive cells (green, pointers are green arrows); DAPI-positive cells (blue cells); scale bar = 50 μm, 70 μm and 80 μm.</p>
Full article ">Figure 10 Cont.
<p>TUNEL staining of kidney tissue of all experiment groups (7 days post-irradiation): TUNEL-positive cells (green, pointers are green arrows); DAPI-positive cells (blue cells); scale bar = 50 μm, 70 μm and 80 μm.</p>
Full article ">Figure 11
<p>Quantitative distribution of TUNEL-positive cells in kidney tissue sections after electron irradiation (7 days post-irradiation). Data are presented as mean ± SD. Experimental groups are numbered according to the study design. Statistically significant differences are indicated by symbols: *—comparison with control group (<span class="html-italic">p</span> &lt; 0.05); **—comparison with control group (<span class="html-italic">p</span> &lt; 0.01); ***—comparison with control group (<span class="html-italic">p</span> &lt; 0.001); ø—comparison between group II (2 Gy electron dose) and group IV (8 Gy electron dose) (<span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 12
<p>Kidneys from the control and experimental groups: (<b>A</b>)—immunohistochemical reactions with antibodies to caspase-3, magnification ×40; scale bar—45 μm, 50 μm, 65 μm; (<b>B</b>)—quantification of caspase-3-positive cells in renal tissue according to the immunohistochemical analysis, graph: (<b>a</b>)—caspase-3-positive cells in the renal medulla; (<b>b</b>)—caspase-3-positive cells in the proximal and distal tubules of nephrons; (<b>c</b>)—in the tubules of the loop of Henle and the collecting ducts. Experimental groups are numbered according to the study design. All data are presented as mean ± SD. Statistically significant differences are indicated by symbols: *—comparison with control group (<span class="html-italic">p</span> &lt; 0.05); **—comparison with control group (<span class="html-italic">p</span> &lt; 0.01); ***—comparison with control group (<span class="html-italic">p</span> &lt; 0.001); †—comparison between group II (2 Gy) and group IV (8 Gy) (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 13
<p>Illustration of the mechanism of radiation-induced nephropathy after electron irradiation (based on the specific literature): (<b>A</b>)—selection of animals for the study; (<b>B</b>)—irradiation of experimental animals using specialized facilities (different irradiation modes are possible); (<b>C</b>)—initiation of DNA double-strand breakdown; (<b>D</b>)—cascade of molecular and cellular reactions leading to direct disease formation (of varying severity).</p>
Full article ">
25 pages, 5726 KiB  
Article
The Combined Administration of Eicosapentaenoic Acid (EPA) and Gamma-Linolenic Acid (GLA) in Experimentally Induced Colitis: An Experimental Study in Rats
by Orestis Ioannidis, Angeliki Cheva, Ioannis Varnalidis, Ioannis Koutelidakis, Vasileios Papaziogas, Panagiotis Christidis, Elissavet Anestiadou, Konstantinos Aggelopoulos, Ioannis Mantzoros, Manousos George Pramateftakis, Efstathios Kotidis, Barbara Driagka, Stamatios Aggelopoulos and Evangelos J. Giamarellos-Bourboulis
J. Clin. Med. 2024, 13(22), 6661; https://doi.org/10.3390/jcm13226661 - 6 Nov 2024
Viewed by 390
Abstract
Background/Objectives: Ulcerative colitis (UC) is a chronic inflammatory bowel disease with limited effective treatments, prompting the need for investigation of novel therapeutic approaches. Eicosapentaenoic acid (EPA) and gamma-linolenic acid (GLA) have demonstrated potential anti-inflammatory properties, but their combined effects on UC have not [...] Read more.
Background/Objectives: Ulcerative colitis (UC) is a chronic inflammatory bowel disease with limited effective treatments, prompting the need for investigation of novel therapeutic approaches. Eicosapentaenoic acid (EPA) and gamma-linolenic acid (GLA) have demonstrated potential anti-inflammatory properties, but their combined effects on UC have not been thoroughly investigated. This study aimed to evaluate the effect of the combined administration of EPA and GLA on clinical and histopathologic features of experimental UC models. Methods: Thirty-six male Wistar rats were randomized in three groups (DSS group, Ensure Plus group, and Oxepa group), with twelve rats in each group. Experimental colitis was induced by administrating dextran sulfate sodium (DSS) 8%. The DSS group received tap water, the Ensure Plus group was given a high caloric diet, and the Oxepa group received a special diet containing high levels of EPA and GLA. Disease activity index (DAI) and microscopic activity index (MAI) were measured. Inflammatory markers were calculated both in blood and large intestine, liver, spleen, and lung tissue samples. Neutrophil and macrophage populations were assessed with immunohistochemistry. Results: No significant differences in the DAI index were found between the groups, but the MAI revealed statistically significant differences (p < 0.001). While no significant differences were observed in tumor necrosis factor-alpha (TNF-α) levels, interleukin-17 (IL-17) levels in the large intestine showed statistically significant differences (p = 0.05), with the Ensure Plus and Oxepa groups displaying lower levels compared to the DSS group (p = 0.021 and p = 0.043, respectively). Significant differences in neutrophil infiltration were found in both the large intestine (p < 0.001) and lungs (p = 0.002), with the Oxepa group showing fewer cells. Similarly, significant differences in macrophage infiltration were observed in the large intestine (p = 0.038) and spleen (p < 0.001), with the Oxepa group having lower macrophage counts. Conclusions: In conclusion, the combination of EPA and GLA demonstrates local anti-inflammatory effects and improves the histopathological outcomes in UC. Full article
(This article belongs to the Special Issue Targeted Treatment in Inflammatory Bowel Diseases (IBD))
Show Figures

Figure 1

Figure 1
<p>The disease activity index (DAI) of the different groups at the end of the experimental procedure. There was no statistically significant difference among groups.</p>
Full article ">Figure 2
<p>Microscopic findings of large intestine. (<b>A1</b>–<b>A4</b>) Microscopic findings of large intestine in DSS group. (<b>Α1</b>–<b>A3</b>) Mucosal fibrosis, inflammatory cell infiltrates in the presence of abundant neutrophilic and eosinophilic polymorphonuclear leukocytes, extensive loss of calycal cells, extensive loss of crypts, severe swelling, reactive and regenerative type of glandular epithelial lesions, extensive infiltration of the submucosal tissue by inflammatory cells with presence of multiple or large lymph nodes and vascular destruction [(<b>A1</b>) ×20, (<b>Α2</b>) ×40, (<b>Α3</b>) ×200]. (<b>A4</b>) Presence of multiple lymph nodes in the submucosa (×20). (<b>B1</b>–<b>B4</b>) Microscopic findings of large intestine in Ensure plus group. (<b>B1</b>–<b>B3</b>) Moderate loss of crypts with disarrangement of architecture and reduction of calyceal cells, extensive inflammatory cell infiltrates of the basement membrane, thickening of mucosal tissue due to edema, and limited ulceration with presence individual lymph nodes [(<b>B1</b>) ×20, (<b>B2</b>) ×40, (<b>B3</b>) ×100]. (<b>B4</b>): Presence of small ulcer and single lymph node in the submucosa (×40). (<b>C1</b>–<b>C4</b>) Microscopic small bowel findings in Oxepa group. (<b>C1</b>–<b>C3</b>) Limited loss of crypts and good maintenance of mucosal thickness and number of calyceal cells, limited inflammatory infiltration or absence there of submucosa and the mucosa mainly between the crypts and not inside it epithelium, and ulceration in the presence of fewer and smaller ulcers, with evidence of regeneration, absence of edema in the mucosa and submucosa, and absence of occasional presence of small lymphocytes. Even more rare ulcers with local regeneration; preservation of mucosal thickness; absence of edema and inflammation of the mucosa; and good preservation of crypts, goblet cells, and glands are observed, as well as mild inflammation between the crypts and not within the endothelium [(<b>C1</b>) ×40, (<b>C2</b>) ×100, (<b>C3</b>) ×100, (<b>C4</b>) ×200].</p>
Full article ">Figure 3
<p>The microscopic activity index (MAI) of the different groups at the end of the experimental procedure. An asterisk (*) indicates a significant difference from DSS group, and hash (#) a significant difference from Ensure Plus group. Statistically significant differences were observed between the DSS and Ensure Plus groups (<span class="html-italic">p</span> = 0.011), DSS and Oxepa groups (<span class="html-italic">p</span> &lt; 0.001), and Ensure Plus and Oxepa groups (<span class="html-italic">p</span> = 0.028).</p>
Full article ">Figure 4
<p>(<b>A</b>) The number of MPO positive cells (neutrophils) in the colon (per mm<sup>2</sup>) in the different groups at the end of the experimental procedure. An asterisk (*) indicates a significant difference from DSS group, and a hash (#) a significant difference from Ensure Plus group. (<b>B</b>) The number of CD68-positive cells (macrophages) in the colon (per mm<sup>2</sup>) in the different groups at the end of the experiment process. An asterisk (*) indicates a significant difference from DSS group.</p>
Full article ">Figure 5
<p>(<b>MPO A</b>–<b>C</b>) MPO immunohistochemical staining of colon tissue sections. Numerous positive cells (neutrophils) were observed in both DSS ((<b>MPO-A</b>) ×200) and Ensure Plus groups ((<b>MPO-B</b>) ×200), whereas in Oxepa group statistically significant fewer positive cells ((<b>MPO-C</b>) ×200) were observed. (<b>CD68 A</b>–<b>C</b>) Immunohistochemical staining of CD68 of colon tissue. A small number of positive cells (macrophages) are observed in the DSS group ((<b>CD68-A</b>) ×200), whereas in the Oxepa group, there are statistically significant more positive cells ((<b>CD68-C</b>) ×200). Ensure Plus group B showed an intermediate number of positive cells that did not differ significantly from the other 2 groups ((<b>CD68-B</b>) ×200).</p>
Full article ">Figure 6
<p>Values of TNF-α (pg/mL) and IL-17 (pg/mL) in the inferior vena cava (IVC) and portal vein (PV) in the different groups at the end of the experimental procedure. Despite the obvious differences, <span class="html-italic">p</span> is not statistically significant.</p>
Full article ">Figure 7
<p>TNF-α (pg/g) values in tissues in the different groups at the end of the experimental procedure. Despite the obvious differences in some tissues, <span class="html-italic">p</span> is not statistically significant. IL-17 (pg/g) values in tissues in the different groups at the end of the experimental procedure. An asterisk (*) indicates a significant difference between groups.</p>
Full article ">Figure 8
<p>The presence of MPO-positive cells (neutrophils) in the liver, spleen, and lungs in the different groups at the end of the experimental procedure. An asterisk (*) indicates a significant difference from DSS group, and a hash (#) a significant difference from Ensure Plus group. Values of the degree of presence of CD68-positive cells (macrophages) in the liver, spleen, and lungs in the different groups at the end of the experimental process. An asterisk (*) indicates a significant difference from DSS group, and a hash (#) a significant difference from Ensure Plus group.</p>
Full article ">Figure 9
<p>MPO and immunohistochemical staining of MPO and CD68 among various tissues. MPO immunohistochemical staining of liver tissue sections. A few neutrophils are depicted ((<b>A</b>) ×200, (<b>B</b>) ×400). Findings are similar across groups. Immunohistochemical staining of CD68 of liver tissue sections. The findings are similar in all groups, without the presence of positive cells. Rarely are macrophages illustrated ((<b>A</b>) ×200, (<b>B</b>) ×400). MPO immunohistochemical staining of spleen tissue sections. A few neutrophils in small groups are shown ((<b>A</b>) ×200, (<b>B</b>) ×400). Findings are similar across groups. CD68 immunohistochemical staining of spleen tissue sections. (<b>A</b>) Few macrophages (×200) are depicted, findings observed in group C. (<b>B</b>) A few to confluent macrophages are depicted (×200); findings observed in DSS and Ensure Plus groups. MPO immunohistochemical staining of lung tissue sections. (<b>A</b>) Rare neutrophils (×200), findings observed in Oxepa group. (<b>B</b>) Few neutrophils (×200), findings observed in DSS and Ensure Plus groups. CD68 immunohistochemical staining of lung tissue sections. Rare macrophages are depicted ((<b>A</b>) ×200, (<b>B</b>) ×400). Findings are similar across groups.</p>
Full article ">
18 pages, 4784 KiB  
Article
An Investigation of RNA Methylations with Biophysical Approaches in a Cervical Cancer Cell Model
by Buket Sağlam, Onur Akkuş, Azime Akçaöz-Alasar, Çağatay Ceylan, Günnur Güler and Bünyamin Akgül
Cells 2024, 13(22), 1832; https://doi.org/10.3390/cells13221832 - 6 Nov 2024
Viewed by 612
Abstract
RNA methylation adds a second layer of genetic information that dictates the post-transcriptional fate of RNAs. Although various methods exist that enable the analysis of RNA methylation in a site-specific or transcriptome-wide manner, whether biophysical approaches can be employed to such analyses is [...] Read more.
RNA methylation adds a second layer of genetic information that dictates the post-transcriptional fate of RNAs. Although various methods exist that enable the analysis of RNA methylation in a site-specific or transcriptome-wide manner, whether biophysical approaches can be employed to such analyses is unexplored. In this study, Fourier-transform infrared (FT-IR) and circular dichroism (CD) spectroscopy are employed to examine the methylation status of both synthetic and cellular RNAs. The results show that FT-IR spectroscopy is perfectly capable of quantitatively distinguishing synthetic m6A-methylated RNAs from un-methylated ones. Subsequently, FT-IR spectroscopy is successfully employed to assess the changes in the extent of total RNA methylation upon the knockdown of the m6A writer, METTL3, in HeLa cells. In addition, the same approach is shown to accurately detect reduction in total RNA methylation upon the treatment of HeLa cells with tumor necrosis factor alpha (TNF-α). It is also demonstrated that m1A and m6A methylation induce quite a distinct secondary structure on RNAs, as evident from CD spectra. These results strongly suggest that both FT-IR and CD spectroscopy methods can be exploited to uncover biophysical properties impinged on RNAs by methyl moieties, providing a fast, convenient and cheap alternative to the existing methods. Full article
(This article belongs to the Section Cellular Biophysics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The FT-IR average spectrum of synthetic RNA samples (both at 2 μg/µL concentration). (<b>A</b>) The sequence of synthetic RNAs and their estimated secondary structure. (<b>B</b>) FT-IR absorbance spectra in the 3600–800 cm<sup>−1</sup> spectral range. A magnification the FT-IR (<b>C</b>,<b>D</b>) absorbance spectra and (<b>E</b>,<b>F</b>) second derivative spectra are also shown, representing the CH stretching and bending vibrations, respectively. The synthetic RNA samples without methylation (black) and with m<sup>6</sup>A methylation (red) are displayed. The peaks are color coded.</p>
Full article ">Figure 2
<p>The FT-IR spectrum of mixtures of synthetic RNA samples. (<b>A</b>) The FT-IR average spectra of synthetic RNA (0% methylation), m<sup>6</sup>A RNA (100% methylation) and their mixtures with the ratio of 1:3, 1:1, 3:1 (25%, 50%, 75% methylation, respectively), showing the spectral region of stretching and bending vibrations of CH molecules. Methylation concentration was plotted versus the intensities (<b>B</b>) at 2984 cm<sup>−1</sup> with R<sup>2</sup>: 0.98, (<b>C</b>) at 2883 cm<sup>−1</sup> with R<sup>2</sup>: 0.76, (<b>D</b>) at 1478 cm<sup>−1</sup> with R<sup>2</sup>: 0.96 and (<b>E</b>) at 1363 cm<sup>−1</sup> with R<sup>2</sup>: 0.66. To prepare the RNA mixtures at various ratios, 2 μg of m<sup>6</sup>A synthetic RNAs (100% methylated) and 2 μg of un-methylated synthetic RNAs (so-called 0% methylated) were mixed at appropriate amounts of volume.</p>
Full article ">Figure 3
<p>The CD spectra of synthetic RNA samples (both at 26.8 ng/µL concentration). (<b>A</b>) The CD absorbance spectra and (<b>B</b>) its 1st derivative spectra without methylation (black) and with m<sup>6</sup>A methylation (red) are displayed. The peaks are color coded.</p>
Full article ">Figure 4
<p>The effects of the knockdown of methylation writers on cellular m<sup>6</sup>A status. (<b>A</b>) Western blotting results to verify METTL3 knockdown in HeLa cells (fold change). (<b>B</b>) Western blot results of TRMT61A knocked-down HeLa cells (fold change). (<b>C</b>) The percentage change in m<sup>6</sup>A modification in the total RNA after METTL3 knockdown in HeLa cells (N = 2 for all time points) (<b>D</b>) SELECT results for METTL3 knocked-down HeLa cells by operating a site-specific detection of m<sup>6</sup>A modification on the MALAT1 gene. Data were normalized by MALAT1 A2511. Error bars indicate mean ± SD of three biological replicates with two technical replicates, unless indicated otherwise. The statistical significance was demonstrated as * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001 by two-tailed Student’s <span class="html-italic">t</span> test.</p>
Full article ">Figure 5
<p>The FT-IR average spectra of cell-derived RNA samples from HeLa si-METTL3 and HeLa si-NC (both at 2.5 μg/µL concentration). (<b>A</b>) FT-IR absorbance spectra in the 3600–800 cm<sup>−1</sup> spectral range. (<b>B</b>,<b>C</b>) FT-IR absorbance spectra and (<b>D</b>,<b>E</b>) second derivative spectra, representing the CH stretching and bending vibrations. The IR peaks for RNA samples of HeLa si-METTL3 (black) and HeLa si-NC (red) are color coded.</p>
Full article ">Figure 6
<p>The CD spectra of RNA samples (all at 114 ng/µL concentration) isolated from HeLa cells (HeLa si-NC, HeLa si-METTL3, HeLa si-TRMT61A). (<b>A</b>) The CD absorbance spectra and (<b>B</b>) the 1st derivative spectra are displayed. The peaks are color coded.</p>
Full article ">Figure 7
<p>An analysis of the total RNAs from HeLa cells after CHX/TNF-α treatment. (<b>A</b>) Apoptosis rates of CHX/TNF-α treatment of HeLa cells. (<b>B</b>) Pro-caspase 3, Cleaved caspase 3, pro-caspase 8 and cleaved caspase 8 expression levels after CHX/TNF-α treatment. (<b>C</b>) METTL3 and TRMT61A expression levels after CHX/TNF-α treatment. (<b>D</b>) The change in the global m<sup>6</sup>A methylation amount after CHX/TNF-α treatment. (<b>E</b>) CD spectra and (<b>F</b>) FT-IR average absorbance spectra of total RNA from CHX/TNF-α treated HeLa cells. The peaks are color coded. Error bars indicate mean ± SD of three biological replicates. The statistical significance was demonstrated as * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; **** <span class="html-italic">p</span> &lt; 0.0001 by two-tailed Student’s <span class="html-italic">t</span> test.</p>
Full article ">
17 pages, 2694 KiB  
Article
Maternal Dietary Improvement or Leptin Supplementation During Suckling Mitigates the Long-Term Impact of Maternal Obesogenic Conditions on Inflammatory and Oxidative Stress Biomarkers in the Offspring of Diet-Induced Obese Rats
by Catalina Amadora Pomar, Jenifer Trepiana, Irene Besné-Eseverri, Pedro Castillo, Andreu Palou, Mariona Palou, Maria P. Portillo and Catalina Picó
Int. J. Mol. Sci. 2024, 25(22), 11876; https://doi.org/10.3390/ijms252211876 - 5 Nov 2024
Viewed by 403
Abstract
This study investigates the impact of maternal nutrition during lactation on inflammation and oxidative stress in the offspring of diet-induced obese rats, along with the potential benefits of leptin supplementation during suckling. Dams were fed either a standard diet (SD), a western diet [...] Read more.
This study investigates the impact of maternal nutrition during lactation on inflammation and oxidative stress in the offspring of diet-induced obese rats, along with the potential benefits of leptin supplementation during suckling. Dams were fed either a standard diet (SD), a western diet (WD) before and during gestation and lactation (WD-dams), or a WD switched to an SD during lactation (Rev-dams). Offspring were supplemented with leptin or vehicle during suckling and then fed an SD or WD until four months. Offspring of the Rev-dams exhibited improved metabolic indicators, including lower body weight, reduced plasma levels of TNF-alpha, a higher adiponectin/leptin (A/L) ratio, enhanced liver antioxidant defenses, and decreased inflammation markers in white adipose tissue (WAT) compared to WD-dams, with sex differences. Leptin supplementation further modulated these markers, reducing oxidative stress in liver and inflammation in WAT and liver (e.g., hepatic Tnfa expression decreased by 45% (males) and 41% (females) in the WD group on an SD), and improving the A/L ratio, with effects varying by maternal conditions and sex. In conclusion, this study underscores the importance of maternal nutrition and leptin intake during suckling in shaping long-term metabolic and inflammatory health in offspring, offering strategies to mitigate the adverse effects of maternal obesity on future generations. Full article
Show Figures

Figure 1

Figure 1
<p>Circulating parameters of O-C, O-WD, and O-Rev male and female animals treated with vehicle or leptin during suckling and fed an SD or WD from weaning, at four months of age. Data are presented as the mean ± SEM (n = 8–12). Statistics: After data separation depending on post-weaning diet, three-way ANOVA was performed to analyze the effects of sex, maternal diet, and/or leptin treatment. In each sex, two-way ANOVA was performed to analyze the effects of leptin treatment and/or maternal diet. Single comparisons between leptin- and vehicle-treated rats of all experimental groups were carried out using the Mann–Whitney U test. Symbols: sex (S), maternal diet (MD), leptin treatment (L); Data that do not share a letter are significantly different, A ≠ B (<span class="html-italic">p</span> &lt; 0.05, LSD post hoc, two- or three-way ANOVA); *, different from their vehicle-treated equal (<span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). Abbreviations: offspring of C-dams (O-C), offspring of WD-dams (O-WD), offspring of Rev-dams (O-Rev), vehicle (Veh), leptin (Lep).</p>
Full article ">Figure 2
<p>Expression levels of genes related to inflammation in retroperitoneal white adipose tissue of O-C, O-WD, and O-Rev male and female animals treated with vehicle or leptin during suckling and fed an SD or WD from weaning, at four months of age. Data are presented as the mean ± SEM (n = 8–12) and are expressed as a percentage of the value for the O-C male rats. Statistics: After data separation depending on post-weaning diet, three-way ANOVA was performed to analyze the effects of sex, maternal diet, and/or leptin treatment. In each sex, two-way ANOVA was performed to analyze the effects of leptin treatment and/or maternal diet. Single comparisons between leptin- and vehicle-treated rats of all experimental groups were carried out using the Mann–Whitney U test. Symbols: sex (S), maternal diet (MD), leptin treatment (L); Data that do not share a letter are significantly different, A ≠ B (<span class="html-italic">p</span> &lt; 0.05, LSD post hoc, two- or three-way ANOVA); *, different from their vehicle-treated equal (<span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). Abbreviations: offspring of C-dams (O-C), offspring of WD-dams (O-WD), offspring of Rev-dams (O-Rev), vehicle (Veh), leptin (Lep).</p>
Full article ">Figure 3
<p>Expression levels of genes related to inflammation in liver of O-C, O-WD, and O-Rev male and female animals treated with vehicle or leptin during suckling and fed an SD or WD from weaning, at four months of age. Data are presented as the mean ± SEM (n = 8–12) and are expressed as a percentage of the value for the O-C male rats. Statistics: After data separation depending on post-weaning diet, three-way ANOVA was performed to analyze the effects of sex, maternal diet, and/or leptin treatment. In each sex, two-way ANOVA was performed to analyze the effects of leptin treatment and/or maternal diet. Single comparisons between leptin- and vehicle-treated rats of all experimental groups were carried out using the Mann–Whitney U test. Symbols: sex (S), maternal diet (MD), leptin treatment (L); Data that do not share a letter are significantly different, A ≠ B (<span class="html-italic">p</span> &lt; 0.05, LSD post hoc, two- or three-way ANOVA); *, different from their vehicle-treated equal (<span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). Abbreviations: offspring of C-dams (O-C), offspring of WD-dams (O-WD), offspring of Rev-dams (O-Rev), vehicle (Veh), leptin (Lep).</p>
Full article ">Figure 4
<p>Markers of antioxidant defenses in the liver of O-C, O-WD, and O-Rev male and female animals treated with vehicle or leptin during suckling and fed an SD or WD from weaning, at four months of age. Data are presented as the mean ± SEM (n = 8–12). Statistics: After data separation, depending on the post-weaning diet, three-way ANOVA was performed to analyze the effects of sex, maternal diet, and/or leptin treatment. In each sex, two-way ANOVA was performed to analyze the effects of leptin treatment and/or maternal diet. Single comparisons between leptin- and vehicle-treated rats for all experimental groups were carried out using the Mann–Whitney U test. Sex (S), maternal diet (MD), and leptin treatment (L); Data that do not share a letter are significantly different, A ≠ B ≠ C (<span class="html-italic">p</span> &lt; 0.05, LSD post hoc and two- or three-way ANOVA); * different from their vehicle-treated equal (<span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). Offspring of C-dams (O-C), offspring of WD-dams (O-WD), offspring of Rev-dams (O-Rev), vehicle (Veh), and leptin (Lep).</p>
Full article ">Figure 5
<p>Summary of the main long-term effects of maternal conditions during suckling on O-C, O-WD, and O-Rev male and female animals weaned onto an SD or WD. Offspring of C-dams (O-C), offspring of WD-dams (O-WD), and offspring of Rev-dams (O-Rev). Arrows indicate increases (↑) or decreases (↓) according to a two-way or three-way ANOVA. Red arrows represent negative effects, while blue arrows indicate positive effects.</p>
Full article ">Figure 6
<p>Summary of the main long-term effects of leptin supplementation during suckling on O-C, O-WD, and O-Rev male and female animals weaned onto an SD or WD. Offspring of C-dams (O-C), offspring of WD-dams (O-WD), and offspring of Rev-dams (O-Rev). Arrows indicate increases (↑) or decreases (↓) according to a Mann–Whitney U test, two-way or three-way ANOVA. Red arrows represent negative effects, while blue arrows indicate positive effects.</p>
Full article ">
20 pages, 15219 KiB  
Article
Regulatory Effect of Fucoidan Hydrolysates on Lipopolysaccharide-Induced Inflammation and Intestinal Barrier Dysfunction in Caco-2 and RAW264.7 Cells Co-Cultures
by Xiaodan Fu, Xinru Huang, Huizi Tan, Xiaojun Huang and Shaoping Nie
Foods 2024, 13(22), 3532; https://doi.org/10.3390/foods13223532 - 5 Nov 2024
Viewed by 526
Abstract
Fucoidan, a sulfated polysaccharide rich in fucose, is derived from brown algae and marine invertebrates. Multiple bioactivities have been shown with fucoidan, while growing attraction has emerged in its low-molecular-weight (Mw) hydrolysates. Here, the anti-inflammatory effect of fucoidan, low-Mw acidolyzed fucoidan (LMAF, <1.5 [...] Read more.
Fucoidan, a sulfated polysaccharide rich in fucose, is derived from brown algae and marine invertebrates. Multiple bioactivities have been shown with fucoidan, while growing attraction has emerged in its low-molecular-weight (Mw) hydrolysates. Here, the anti-inflammatory effect of fucoidan, low-Mw acidolyzed fucoidan (LMAF, <1.5 kDa), and high-Mw acidolyzed fucoidan (HMAF, 1.5–20 kDa) were investigated in vitro using lipopolysaccharide (LPS)-stimulated Caco-2 and RAW264.7 co-cultures. Fucoidan, LMAF, and HMAF with different structures exhibited varied anti-inflammatory effects. LMAF and HMAF effectively decreased the nitric oxide release of RAW264.7 cells. LMAF exhibited a competitive effect in reducing tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6 levels compared to HMAF and fucoidan. Transcriptome of RAW264.7 revealed that LPS and LMAF mainly regulated the transcriptional expression of genes, including Tnf, Il6, Il1b, Junb, and Nfkb1 in the TNF signaling pathway, NF-kappa B signaling pathway, and cytokine–cytokine receptor interaction. RT-PCR results indicated that LMAF markedly reduced the LPS-elevated expression of Cxcl2, Tnf, Ccl2, Il1b, and Csf2. Moreover, LMAF effectively increased the proteins expression of Claudin-1, Occludin, and Zonula occluden-1 in Caco-2 cells. This study highlights the potential of LMAF to improve inflammation and intestinal barrier integrity, offering a foundation for further application of low-Mw fucoidan hydrolysates. Full article
(This article belongs to the Special Issue Marine Food: Development, Quality and Functionality)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effects of fucoidan and its hydrolysates on cell viability. Sample toxicity in Caco-2 (<b>A</b>) and RAW264.7 (<b>B</b>) cells. Protective effect in LPS (1 μg/mL)-induced Caco-2 (<b>C</b>) and RAW264.7 (<b>D</b>) cells. Cells were treated with fucoidan, LMAF, and HMAF for 24 h, respectively, followed by assessment using the CCK8 testing method. Data are expressed as the mean ± SD (<span class="html-italic">n</span> = 5). <sup>a–e</sup> Significant differences among varied concentrations for each fucoidan and its hydrolysates were analyzed using one-way ANOVA with a post hoc Tukey’s HSD test (<span class="html-italic">p</span> &lt; 0.05). Statistical significance between LPS-treated group and other groups was performed using Student’s <span class="html-italic">t</span>-test (* <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). NC, normal control.</p>
Full article ">Figure 2
<p>The effect of fucoidan and its acidolyzed hydrolysates on intestinal epithelial integrity and cytokines production in Caco-2/RAW264.7 co-cultures with LPS stimulation. (<b>A</b>) Transepithelial electrical resistance (TEER). (<b>B</b>) Nitric oxide production. Inflammatory cytokine production of (<b>C</b>) TNF-α, (<b>D</b>) IL-1β, (<b>E</b>) IL-6, and (<b>F</b>) IL-10 with treatment of fucoidan, LMAF, and HMAF, respectively. Data are presented as the mean ± SD (<span class="html-italic">n</span> = 5). Statistical significance between LPS-treated group and other groups was performed using Student’s <span class="html-italic">t</span>-test (* <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). NC, normal control.</p>
Full article ">Figure 3
<p>Impact of LMAF on the transcriptome of RAW264.7 cells in co-cultures. (<b>A</b>) PCA plot of LPS versus the normal control (NC) group. (<b>B</b>) PCA plot comparing the LMAF and LPS group. (<b>C</b>) Volcano plot highlighting significant DEGs between LPS and NC. (<b>D</b>) Volcano plot of significant DEGs between LMAF and LPS. Red dots represent upregulated DEGs; blue dots represent downregulated DEGs (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 4
<p>Functional annotation analysis of differentially expressed genes. GO annotations analysis of LPS versus the normal control (NC) group (<b>A</b>) and LMAF versus the LPS group (<b>B</b>). KEGG annotations analysis of LPS versus the NC group (<b>C</b>) and LMAF versus the LPS group (<b>D</b>) (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 5
<p>Enrichment analysis of differentially expressed gene. (<b>A</b>) GO enrichment analysis. (<b>B</b>) KEGG enrichment analysis. Multidimensional GO enrichment circle diagram of LPS versus the NC group (<b>C</b>) and LMAF versus the LPS group (<b>D</b>). Multidimensional KEGG enrichment circle diagram of LPS versus the NC group (<b>E</b>) and LMAF versus the LPS group (<b>F</b>) (<span class="html-italic">n</span> = 3). NC, normal control.</p>
Full article ">Figure 6
<p>Analysis of key differentially expressed gene. (<b>A</b>) Venn diagram of shared DEGs expressed in two libraries of LPS versus the NC group and LMAF versus the LPS group. (<b>B</b>) KEGG enrichment analysis of shared DEGs. (<b>C</b>) Heatmap plot of shared DEGs. The red and blue colors indicate the upregulated and downregulated DEGs, respectively. (<b>D</b>) The protein–protein interaction network of shared DEGs. Increasing node indicates a stronger core. (<b>E</b>) The gene–gene interaction network among the shared DEGs. (<b>F</b>) KEGG enrichment analysis of DEGs expressed in shared enriched KEGG pathway in two libraries of LPS versus the NC group and LMAF versus the LPS group. Circle heatmap plot (<b>G</b>), protein–protein interaction network (<b>H</b>), and gene–gene interaction network (<b>I</b>) of DEGs expressed in shared enriched KEGG pathway. NC, normal control.</p>
Full article ">Figure 7
<p>Effects of LMAF on modulating inflammation and intestinal barrier dysfunction in LPS-stimulated Caco-2 and RAW264.7 co-cultures. (<b>A</b>) Possible mechanisms of LMAF on ameliorating inflammation in RAW264.7 cells. (<b>B</b>) Experssion levels of transcripts in TNF signaling pathway involved in ameliorating inflammation (<span class="html-italic">n</span> = 3). Relative mRNA expression changes across all groups in RNA-seq analysis were analyzed by one-way ANOVA followed by Tukey’s HSD post hoc test (* <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>) The effects of fucoidan and acidolyzed hydrolysates on RT-PCR expression of inflammatory related genes (<span class="html-italic">n</span> = 3). Significant differences of each other groups compared to LPS group were analyzed by one-way ANOVA followed by Tukey’s HSD post hoc test (* <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>D</b>) The effects of fucoidan and acidolyzed hydrolysates on expression of Claudin-1, Occludin, ZO-1, and proliferating cell nuclear antigen (PCNA) in Caco-2 cells. Protein levels (green) of ZO-1, Occludin, Claudin-1, and PCNA (red) were detected by immunofluorescence. Nuclei were counterstained with DAPI. Scale bar = 100 µm.</p>
Full article ">
14 pages, 2532 KiB  
Article
Piperine as an Herbal Alternative for the Prevention of Drug-Induced Liver Damage Caused by Paracetamol
by Aline Meireles Coelho, Isabela Ferreira Queiroz, Luiza Oliveira Perucci, Tatiana Prata Menezes, Wanderson Geraldo Lima, André Talvani and Daniela Caldeira Costa
Pharmaceuticals 2024, 17(11), 1477; https://doi.org/10.3390/ph17111477 - 2 Nov 2024
Viewed by 704
Abstract
Background/Objective: Hepatic drug intoxication is becoming increasingly common with the increasing use of chronic medications. Piperine has emerged as a promising alternative for protecting the liver against drug-induced injury. We evaluated the prophylactic effects of piperine in C57BL/6 mice with an acute liver [...] Read more.
Background/Objective: Hepatic drug intoxication is becoming increasingly common with the increasing use of chronic medications. Piperine has emerged as a promising alternative for protecting the liver against drug-induced injury. We evaluated the prophylactic effects of piperine in C57BL/6 mice with an acute liver injury induced by a paracetamol (APAP) overdose. Methods: Piperine was administered at a dose of 20 mg/kg (P20) or 40 mg/kg (P40) for eight consecutive days before the animals were exposed to a hepatotoxic dose of paracetamol (500 mg/kg). The animals were euthanized 3 h after the paracetamol overdose. Results: The prophylactic treatment with piperine (P20 and P40) maintained the levels of alanine aminotransferase (ALT) and the biomarkers of oxidative damage (TBARS and carbonylated proteins), which were statistically similar to those for the control group. The extent of hepatocyte necrosis and TNF-α (tumor necrosis factor-alpha) levels were lower than those in the group exposed to liver injury (APAP group). Piperine modulated the gene expression of CYP2E1 (cytochrome P4502E1) and the inflammasome pathway (NLRP3, CASP-1, IL-1β, and IL-18), which play a crucial role in the inflammatory response. In the P40 group, the degree of hepatic hyperemia was similar to that in the control group, as was the increase in metalloproteinase 9 (MMP-9) activity. Conclusion: Piperine has demonstrated beneficial and promising effects for the prevention of liver injury resulting from paracetamol-induced drug intoxication. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>An evaluation of liver injury via ALT (<b>A</b>) and AST (<b>B</b>) levels in the serum of C57BL/6 mice subjected to prophylactic treatment with piperine (20 mg/kg and 40 mg/kg) for 8 days, followed by APAP intoxication. The different letters indicate statistical differences between the groups, where any letter other than “a” signifies a difference from the control group, and any letter other than “b” signifies a difference from the APAP group, as determined by a one-way analysis of variance followed by Tukey’s post hoc test. Statistical significance is considered at <span class="html-italic">p</span> &lt; 0.05. ALT: alanine aminotransferase; AST: aspartate aminotransferase.</p>
Full article ">Figure 2
<p>Histopathological evaluation and representative photomicrographs of the liver in C57BL/6 mice subjected to prophylactic piperine treatment for 8 days, followed by APAP intoxication. (<b>A</b>) The group treated with saline solution, showing normal liver architecture (control group); (<b>B</b>) the APAP group; and (<b>C</b>,<b>D</b>) the groups receiving piperine pretreatment of 20 mg/kg and 40 mg/kg, respectively. Staining: hematoxylin and eosin (HE). Magnification: 40x objective. The arrows indicate areas of necrosis. Panel (<b>E</b>) displays the percentage area of liver necrosis in the experimental groups, (<b>F</b>) shows the hepatocyte binucleation index, and (<b>G</b>) shows the hyperemia index. The different letters indicate statistical differences between the groups: any letter other than “a” signifies a difference from the control group; any letter other than “b” signifies a difference from the APAP group; and “c” signifies a significant difference from both the control group (“a”) and the APAP group (“b”), as determined by a one-way analysis of variance followed by Tukey’s post hoc test, with the significance set at <span class="html-italic">p</span> &lt; 0.05. The statistical contingency analysis in panels (<b>F</b>) and (<b>G</b>) was assessed using the chi-square test and Fisher’s exact test, with the different letters indicating statistical differences between the groups. Black arrows indicate area of necrosis.</p>
Full article ">Figure 3
<p>Assessment of oxidative damage biomarkers. Liver damage was evaluated by measuring protein carbonylation (<b>A</b>) and thiobarbituric acid reactive substances (TBARS) (<b>B</b>), while matrix remodeling was assessed via matrix metalloproteinase 9 (MMP-9) activity in zymography assays (<b>C</b>) of the livers of C57BL/6 mice subjected to prophylactic piperine treatment for 8 days, followed by APAP intoxication. The different letters indicate statistical differences between the groups. In panels A and B, any letter other than “a” signifies a difference from the control group, and any letter other than “b” signifies a difference from the APAP group. In panel C, the letter “b” indicates that the P40 group is different from the other experimental groups, as determined by a one-way analysis of variance followed by Tukey’s post hoc test. Statistical significance is considered at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Evaluation of gene expression in the inflammasome pathway (<span class="html-italic">NLRP3</span> (<b>A</b>)<span class="html-italic">, CASP-1</span> (<b>B</b>)<span class="html-italic">, IL-1β</span> (<b>C</b>), and <span class="html-italic">IL-18</span> (<b>D</b>)) and the isoenzymes <span class="html-italic">CYP2E1</span> (<b>E</b>) and <span class="html-italic">CYP1A2</span> (<b>F</b>) in the mice subjected to prophylactic treatment with piperine for 8 days, followed by APAP intoxication. The different letters indicate statistical differences between the groups: any letter other than “a” signifies a difference from the control group, and any letter other than “b” signifies a difference from the APAP group. The notation “ab” is used when the statistical analysis indicates that a group is statistically equal to both the control and APAP groups, as determined by a one-way analysis of variance followed by Tukey’s post hoc test. Abbreviations: <span class="html-italic">NLRP3</span>: NLR family pyrin domain-containing protein 3; <span class="html-italic">CASP-1</span>: caspase-1; <span class="html-italic">IL-1β</span>: interleukin-1β; <span class="html-italic">IL-18</span>: interleukin-18; <span class="html-italic">CYP1A2</span>: cytochrome P450 1A2; <span class="html-italic">CYP2E1</span>: cytochrome P450 2E1.</p>
Full article ">Figure 5
<p>Evaluation of the cytokines TNF-α (<b>A</b>) and IL-6 (<b>B</b>) in the serum, and TNF-α (<b>C</b>) and IL-1β (<b>D</b>) in the liver of mice subjected to prophylactic treatment with piperine for 8 days, followed by APAP intoxication. The different letters indicate statistical differences between the groups: any letter other than “a” signifies a difference from the control group, any letter other than “b” signifies a difference from the APAP group, and “c” indicates that the group is significantly different from both the control group (“a”) and the APAP group (“b”). The notation “ac” is used when the statistical analysis shows that the group is statistically similar to both the control and P20 groups. The notation “ab” is used when the statistical analysis indicates that the group is statistically equal to both the control and APAP groups, as determined by a one-way analysis of variance followed by Tukey’s post hoc test. Significant differences are considered at <span class="html-italic">p</span> &lt; 0.05. Abbreviations: TNF-α: tumor necrosis factor-alpha; IL-6: interleukin-6; IL-1β: interleukin-1 beta.</p>
Full article ">Figure 6
<p>Piperine administered preventively for 8 days protected against drug intoxication induced by paracetamol. Parameters, including biochemical, histological, redox status, and inflammatory, were preserved in piperine-treated groups, indicating liver protection. The red arrows represent the effects of paracetamol on the different parameters evaluated, while the black arrows represent the effects of the P20 and P40 groups on these parameters.</p>
Full article ">
Back to TopTop