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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (246)

Search Parameters:
Keywords = mast cell tumors

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 5396 KiB  
Article
Regulation of Age-Related Lipid Metabolism in Ovarian Cancer
by Jihua Feng, Clay Douglas Rouse, Lila Taylor, Santiago Garcia, Ethan Nguyen, Isabella Coogan, Olivia Byrd, Andrew Berchuck, Susan K. Murphy and Zhiqing Huang
Int. J. Mol. Sci. 2025, 26(1), 320; https://doi.org/10.3390/ijms26010320 - 1 Jan 2025
Viewed by 479
Abstract
The mortality rate of ovarian cancer (OC) remains the highest among female gynecological malignancies. Advanced age is the highest risk factor for OC development and progression, yet little is known about the role of the aged tumor microenvironment (TME). We conducted RNA sequencing [...] Read more.
The mortality rate of ovarian cancer (OC) remains the highest among female gynecological malignancies. Advanced age is the highest risk factor for OC development and progression, yet little is known about the role of the aged tumor microenvironment (TME). We conducted RNA sequencing and lipidomic analysis of young and aged gonadal adipose tissue from rat xenografts before and after OC formation. The rates of tumor formation (p = 0.047) and tumor volume (p = 0.002) were significantly higher in the aged rats than in their young counterparts. RNA sequencing data showed significant differences in gene expression profiles between the groups of young and aged rat adipose tissues (p < 0.05), including S100a8, S100a9, Il1rl1, Lcn2, C3, Hba-a1, Fcna, and Pnpla3. At the time of tumor generation, there were also changes in the lipid components within the gonadal adipose tissues of young and aged rats, with higher levels of free fatty acids (FFAs) and triglycerides (TGs) in aged rats. Furthermore, the aged TME showed changes in immune cell composition, especially inflammation-related cells, including neutrophils, myeloid dendritic cells, CD4+ T cells (non-regulatory), and mast cell activation (p < 0.05). The correlation between S100a8, S100a9, neutrophil, and omega-5, FFA 18:3 levels was also determined. Additionally, omega-5, which is downregulated in aged rats, inhibited OC cell proliferation in vitro (p < 0.001). Our study suggests that the aged TME promotes OC proliferation resulting from age-related changes in gene/pathway expression, lipid metabolism, and immune cell distribution. Targeting the aging adipose microenvironment, particularly lipid metabolism, is a promising therapeutic strategy for OC and warrants further investigation. Significance: The aging microenvironment contributes to OC development and progression because of changes in the immune response regulatory genes S100a8 and S100a9, secreted by adipocytes, preadipocytes, or neutrophils, and by altering omega-5 metabolism. Full article
(This article belongs to the Special Issue Molecular Diagnostics and Genomics of Tumors)
Show Figures

Figure 1

Figure 1
<p>Workflow for xenograft rat model. OC: ovarian cancer. PTA-A: pre-tumor adipose from aged rats. PTA-Y: pre-tumor adipose from young rats. TSA-A: tumor surrounding adipose from aged rats. TSA-Y: tumor surrounding adipose from young rats.</p>
Full article ">Figure 2
<p>More tumor formation in aged rat group. (<b>A</b>). Comparison of the tumor formation rate between aged and young rat groups. Thirteen out of thirteen rats in the aged group formed tumors by day 26 post-ovarian cancer (OC) cell implantation. Eleven out of sixteen rats (68.75%) in the young group formed tumors by day 26 post-OC cell implantation. (<b>B</b>). By day 26 post-OC cell implantation, the average tumor volume from the aged rat group was significantly bigger than that from the young rat group, aged tumor median = 17.64 cm<sup>3</sup>, young tumor median = 1.14 cm<sup>3</sup>.</p>
Full article ">Figure 3
<p><b>Principal component analysis (PCA) in PTA-A&amp;Y, TSA-A&amp;Y, and PTA&amp;TSA-A.</b> PCA was performed with the 29 biological samples including 16 fat tissues from young rats and 13 from aged rats. The analysis showed the similarities of differential gene expressions within the aged groups and the clear separations of gene expression between aged and young groups for all 3 panels of datasets, PTA-A&amp;Y (<b>A</b>), TSA-A&amp;Y (<b>B</b>), and PTA&amp;TSA-A (<b>C</b>). PTA-A: pre-tumor adipose from aged rats. PTA-Y: pre-tumor adipose from young rats. TSA-A: tumor surrounding adipose from aged rats. TSA-Y: tumor surrounding adipose from young rats.</p>
Full article ">Figure 4
<p>Volcano plots of transcriptome analysis show differential gene expression in PTA-A&amp;Y (<b>A</b>), TSA-A&amp;Y (<b>B</b>), and PTA&amp;TSA-A (<b>C</b>). Comparisons by Volcano plots of unique differential expressed genes (DEGs) in PTA-A&amp;Y, TSA-A&amp;Y, and PTA&amp;TSA-A groups. The x and y axes show the log2 FC and -log10(adjusted <span class="html-italic">p</span>-value), respectively. Types down, not, and up indicate downregulated DEGs, no difference DEGs, and upregulated DEGs in the aged group. PTA-A: pre-tumor adipose from aged rats. PTA-Y: pre-tumor adipose from young rats. TSA-A: tumor surrounding adipose from aged rats. TSA-Y: tumor surrounding adipose from young rats.</p>
Full article ">Figure 5
<p><b>Transcriptome analysis shows differential gene expression in PTA-A&amp;Y, TSA-A&amp;Y, and PTA&amp;TSA-A.</b> (<b>A</b>). Venn plot of upregulated and downregulated differential expressed genes (DEGs) in PTA-A&amp;Y, TSA-A&amp;Y, and PTA&amp;TSA-A groups. Up-before: upregulated genes in PTA-A&amp;Y; down-before: downregulated genes in PTA-A&amp;Y; up-tumor: upregulated genes in TSA-A&amp;Y; down-tumor: downregulated genes in TSA-A&amp;Y; up-aged: upregulated genes in PTA&amp;TSA-A; down-aged: downregulated genes in PTA&amp;TSA-A. The bar graph shows the number of DEGs in each adipose group. The numbers are included in each group. (<b>B</b>). The heat map of DEGs in PTA-A&amp;Y, TSA-A&amp;Y, and PTA&amp;TSA-A. The differentially expressed genes are shown on the right of the heat map. PTA-Y: pre-tumor adipose from young rats. PTA-A: pre-tumor adipose from aged rats. TSA-Y: tumor surrounding adipose from young rats. TSA-A: tumor surrounding adipose from aged rats. A-BT: aged rats before tumor formation. A-T: aged rats when tumor formed.</p>
Full article ">Figure 6
<p><b>Enrichment of KEGG Pathway in PTA-A&amp;Y, TSA-A&amp;Y, and PTA&amp;TSA-A groups.</b> Twenty-six KEGG pathways were enriched in all 3 of the PTA-A&amp;Y, TSA-A&amp;Y, and PTA&amp;TSA-A groups, including cytokine receptors, cytokines and growth factors, viral protein interaction with cytokine and cytokine receptor, IL-17 signaling pathway, glycerolipid metabolism. PTA-A&amp;Y: comparison of pre-tumor adipose from aged rats and young rats. TSA-A&amp;Y: comparison of tumor surrounding adipose from aged rats and young rats. PTA-A: pre-tumor adipose from aged rats. TSA-A: tumor surrounding adipose from aged rats.</p>
Full article ">Figure 7
<p><b>Differential expression of metabolites in TSA-A&amp;Y adipose.</b> (<b>A</b>). Orthogonal Partial Least Square Discriminant Analysis (OPLS-DA) was carried out with 28 biological samples including 13 adipose tissues from aged rats and 15 adipose tissues from young rats at primary tumor formation (5 young rats without tumors). The green plot shows the lipid components in young rats and the red shows the lipids in aged rats. The lipid samples within each group were closely related, while the separation of lipids between the groups was far, indicating the reliability of the metabolome data from the aged rat adipose groups. (<b>B</b>). The top 25 pathways of Relational Database of Metabolomics Pathways (RaMP) functional annotation. (<b>C</b>). Volcano plots showed differential expression of metabolites (DEMs) between TSA-A and TSA-Y. The x axes show the log2 FC and the y axes show -log10 (<span class="html-italic">p</span>-value), respectively. The dots represent the lipid components in each group. (<b>D</b>–<b>F</b>). The bar graphs show scaled intensity values of altered glycerolipids (GLs, <b>D</b>), glycerophospholipids (GPs, <b>E</b>) and fatty acyls (FAs, <b>F</b>) between TSA-A (orange bar, aged) and TSA-Y (blue bars, young).</p>
Full article ">Figure 8
<p><b>Transcriptome–metabolome correlative regulation.</b> (<b>A</b>). The gene expression profiling from RNA-seq analysis and lipid profiling from lipidomic analysis were conducted for their correlation in groups of tumor-surrounding adipose from aged rats and young rats (TSA-A&amp;Y), the adipose from primary tumor surrounding tissues from aged and young rats. The gene–lipid, immune cell–gene, and immune cell–lipid correlations were evaluated in TSA-A&amp;Y using Spearman’s correlation. With the thresholds of correlation ≥0.5, absolute log2 FC ≥ 1, VIP ≥ 1, <span class="html-italic">p</span> &lt; 0.05, we found 25 genes and 20 lipids that are closely related. The darker the color, the higher the correlation coefficient between gene expression and lipids. (<b>B</b>,<b>C</b>). The correlation between immune infiltration and gene expression from RNA-seq and lipid metabolites from Lipidomic in groups of TSA-A&amp;Y. (<b>B</b>). The correlation between 4 immune cells (Neutrophil, Myeloid dendritic cell, T cell CD4+ (non-regulatory), and Mast cell activated) and 5 genes (<span class="html-italic">S100a8</span>, <span class="html-italic">S100a9</span>, <span class="html-italic">C3</span>, <span class="html-italic">Fcna</span>, <span class="html-italic">Pnpla3</span>). (<b>C</b>). The correlation between 4 immune cells and 13 lipids.</p>
Full article ">Figure 9
<p><b>The in vitro studies confirmed the in vivo findings from rat xenograft model</b>. (<b>A</b>). Aging was induced in preadipocytes, 3T3-L1, with carboplatin treatment at 2 μM for 72 h. The blue color represents senescent cells. (<b>B</b>). Ovarian cancer (OC) cell line, A2780, showed higher cell proliferation when exposed in an aging conditioned medium from aged preadipocyte 3T3-L1 cells. <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001. (<b>C</b>). Gene expression was validated using RT-PCR. Eight genes showed differential expression in groups before tumor formation (PTA-A&amp;Y), when tumor formed (TSA-A&amp;Y), and PTA&amp;TSA-A, and their involvement in lipid regulation was validated using RT-PCR with the gene-specific probes for Taqman assay and 6 adipose tissues from each of the aged rats and young rats. As shown with RNA-seq analysis, the genes of <span class="html-italic">S100a8</span>, <span class="html-italic">S100a9</span>, <span class="html-italic">C3</span>, <span class="html-italic">Il1rl1</span>, <span class="html-italic">Lon2</span>, <span class="html-italic">Hba-a1</span> and <span class="html-italic">Fcna</span> showed upregulation in aged rats and gene <span class="html-italic">Pnpla3</span> showed downregulation in aged rats. <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. (<b>D</b>). OC cell line, A2780, HEYA8, and CAOV2 showed lower proliferation when exposed in FFA (18:3) containing medium in a dose-dependent manner. <sup>ns</sup> <span class="html-italic">p</span> &gt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001. OC: ovarian cancer.</p>
Full article ">
22 pages, 15766 KiB  
Article
Pharmacological Effects of Antioxidant Mycosporine-Glycine in Alleviating Ultraviolet B-Induced Skin Photodamage: Insights from Metabolomic and Transcriptomic Analyses
by Kai Wang, Ling Qin, Huan Lin, Mengke Yao, Junhan Cao, Qing Zhang, Changfeng Qu, Yingying He, Jinlai Miao and Ming Liu
Antioxidants 2025, 14(1), 30; https://doi.org/10.3390/antiox14010030 - 29 Dec 2024
Viewed by 462
Abstract
Mycosporine-glycine (M-Gly), a member of the mycosporine-like amino acid (MAA) family, is known for its potent antioxidant and anti-inflammatory properties. However, its in vivo efficacy in alleviating acute skin photodamage, primarily caused by oxidative stress, has not been well explored. In this investigation, [...] Read more.
Mycosporine-glycine (M-Gly), a member of the mycosporine-like amino acid (MAA) family, is known for its potent antioxidant and anti-inflammatory properties. However, its in vivo efficacy in alleviating acute skin photodamage, primarily caused by oxidative stress, has not been well explored. In this investigation, 30 female ICR mice were divided into four groups: a control group and three Ultraviolet B (UVB)-exposed groups treated with saline or M-Gly via intraperitoneal injection for 30 days. At the end of the experiment, UVB exposure caused erythema, wrinkling, collagen degradation, and mast cell infiltration in mouse dorsal skin. M-Gly treatment improved skin appearance and reduced mast cell numbers, while also elevating antioxidant levels, including superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH). Furthermore, M-Gly reduced inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and IL-1β, typically upregulated after UVB exposure. M-Gly also protected skin collagen by upregulating type I procollagen and decreasing MMP-1 levels. Skin metabolomic profiling identified 34 differentially abundant metabolites, and transcriptomic analysis revealed 752 differentially expressed genes. The combined metabolomic and transcriptomic data indicate that M-Gly’s protective effects may involve the regulation of ion transport, cellular repair, metabolic stability, collagen preservation, and the Nrf2/HO-1 pathway. These findings highlight M-Gly’s potential as an endogenous antioxidant for protecting skin from UVB-induced damage. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>ABTS radical scavenging capacity of M-Gly (<b>A</b>) and vitamin C (<b>B</b>).</p>
Full article ">Figure 2
<p>The body weight and food intake records of mice during the modeling process and the results of M-Gly biosafety detection of mice. (<b>A</b>) Line chart of body weight change in mice. (<b>B</b>) The diet condition of mice. (<b>C</b>) Liver weight was recorded in each group. (<b>D</b>) Kidney weight was recorded in each group. (<b>E</b>) Spleen weight was recorded in each group. (<b>F</b>) Serum ALT levels were detected in each group. (<b>G</b>) Serum AST levels were detected in each group. NC represents the normal control group, MC represents the model negative group, H-MG represents the high-dose experimental group, and L-MG represents the low-dose experimental group.</p>
Full article ">Figure 3
<p>Hematoxylin and eosin (H&amp;E) staining section images of the liver, kidney, and spleen of mice in each group. NC represents the normal control group, MC represents the model negative group, H-MG represents the high-dose experimental group, and L-MG represents the low-dose experimental group.</p>
Full article ">Figure 4
<p>The preventive and therapeutic effects of M-Gly administration on UVB-induced skin damage in mice. (<b>A</b>) Images of the back skin of mice, <span class="html-italic">n</span> = 6 or <span class="html-italic">n</span> = 8. (<b>B</b>) Histological staining of mouse dorsal skin (H&amp;E, Masson staining, and Toluidine blue staining), <span class="html-italic">n</span> = 3. In the Masson staining results, blue represents collagen fibers.</p>
Full article ">Figure 5
<p>Indices of skin damage induced by UVB radiation in mice. (<b>A</b>) Statistical analysis of the skin thickness. (<b>B</b>) Statistical analysis of the density of collagen fibers in the dermis of mouse dorsal skin. (<b>C</b>) MMP-1 levels in mice skin. (<b>D</b>) Type I procollagen levels in mice skin. (<b>E</b>) Statistical analysis of the mast cells number in the mouse dorsal skin. NC represents the normal control group, MC represents the model negative group, H-MG represents the high-dose experimental group, and L-MG represents the low-dose experimental group. # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001, compared with the NC group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, compared with the MC group; ns represents no statistically significant difference between the two groups.</p>
Full article ">Figure 6
<p>(<b>A</b>–<b>D</b>) Effects of M-Gly on levels of MDA, SOD, CAT, and GSH in UVB damaged skin of mice. (<b>E</b>–<b>G</b>) Effects of M-Gly on the inflammatory factors (TNF-α, IL-6, and IL-1β) in UVB damaged skin of mice. NC represents the normal control group, MC represents the model negative group, H-MG represents the high-dose experimental group, and L-MG represents the low-dose experimental group. # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001, compared with the NC group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001, compared with the MC group; ns represents no statistically significant difference between the two groups.</p>
Full article ">Figure 7
<p>The PCA and PLS-DA analysis results of the samples from each group in the skin metabolomics. (<b>A</b>) PCA score plot of NC group, MC group, M-Gly group, and QC samples. (<b>B</b>) PLS-DA score plot of NC group vs. MC group. (<b>C</b>) PLS-DA score plot of MC group vs. M-Gly group. The confidence ellipse indicates that the set of “real” samples is distributed in this region with 95% confidence.</p>
Full article ">Figure 8
<p>The volcano plot results of metabolites in different experimental groups. (<b>A</b>) Volcano plot of significant differential metabolites of NC group vs. MC group. (<b>B</b>) Volcano plot of significant differential metabolites of MC group vs. M-Gly group.</p>
Full article ">Figure 9
<p>Specific differential metabolite analysis of M-Gly intervention. (<b>A</b>) Venn diagram of differential metabolites between the two comparison groups (NC vs. MC and MC vs. M-Gly). (<b>B</b>) The content of prolylhydroxyproline in skin metabolites between NC group and MC group. (<b>C</b>) The content of prolylhydroxyproline in skin metabolites between the MC group and M-Gly group. * Represents <span class="html-italic">p</span> &lt; 0.05, *** represents <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 10
<p>Metabolic pathway enrichment analysis of identified differential metabolites. The size of the bubbles in the figure represents the number of metabolites enriched in the pathway, while the color of the bubbles indicates the significance level of the enrichment, as represented by the <span class="html-italic">p</span>-value.</p>
Full article ">Figure 11
<p>Effects of M-Gly intervention on skin transcriptome. (<b>A</b>) PCA score plot of NC group, MC group, and M-Gly group samples. The confidence ellipse indicates that the set of "real" samples is distributed in this region with 95% confidence. (<b>B</b>) Volcano plot of significant differential genes (DEGs) of MC group vs. NC group. (<b>C</b>) Volcano plot of significant DEGs of M-Gly group vs. MC group. (<b>D</b>) Venn diagram of DEGs between the two comparison groups (NC vs. MC and MC vs. M-Gly).</p>
Full article ">Figure 12
<p>Functional enrichment analysis of target genes. (<b>A</b>) GO enrichment analysis of identified DEGs. (<b>B</b>) KEGG enrichment analysis of identified DEGs.</p>
Full article ">Figure 13
<p>Effect of M-Gly intervention on Nrf2/HO-1 pathway. (<b>A</b>) GSEA enrichment plot of the WP_NRF2_PATHWAY gene set in the M-Gly group and MC group. (<b>B</b>) The expression heatmap of genes involved in the GSEA. Red represents a higher expression level of the gene in the sample, while blue represents a lower expression level. (<b>C</b>) The expression level of Nrf2 mRNA. (<b>D</b>) The expression level of HO-1 mRNA. NC represents the normal control group, MC represents the model negative group, H-MG represents the high-dose experimental group, and L-MG represents the low-dose experimental group. ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001, compared with the NC group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001, compared with the MC group.</p>
Full article ">
24 pages, 5776 KiB  
Article
Garcinia brasiliensis Leaves Extracts Inhibit the Development of Ascitic and Solid Ehrlich Tumors
by Lucas Sylvestre Silva, Eduardo Cavallini, Rafael André da Silva, Monielle Sant’Ana, Ariane Harumi Yoshikawa, Thiago Salomão, Bianca Huang, Paula Craice, Luiz Philipe de Souza Ferreira, Heitor Pedro Della Matta, Cristiane Damas Gil, Maria de Lourdes Gomes Pereira and Ana Paula Girol
Pharmaceuticals 2025, 18(1), 24; https://doi.org/10.3390/ph18010024 - 28 Dec 2024
Viewed by 366
Abstract
Background: Garcinia brasiliensis is traditionally known for its medicinal properties. Objectives: Here, we investigated the effects of crude extract (CE) and ethyl acetate fraction (EAF) obtained from G. brasiliensis leaves on the ascitic (EA) and solid (ES) forms of Ehrlich tumors. Methods [...] Read more.
Background: Garcinia brasiliensis is traditionally known for its medicinal properties. Objectives: Here, we investigated the effects of crude extract (CE) and ethyl acetate fraction (EAF) obtained from G. brasiliensis leaves on the ascitic (EA) and solid (ES) forms of Ehrlich tumors. Methods: Induced and uninduced BALB/c mice were treated intramuscularly, for 7 or 14 days, with saline solution or CE and EAF, both at a 10% concentration, based on in vitro cytotoxicity assessment. Biochemical analyses were also performed to evaluate in vivo cytotoxicity. In relation to tumor-induced animals, morphological changes, plasma enzymes, inflammatory mediators and the induction of apoptosis were analyzed, in addition to histopathological studies, to evaluate the inhibition of tumor growth. Results: Alanine aminotransferase (ALT), aspartate aminotransferase (AST) and gamma glutamyl transferase (GGT) were regulated by CE and EAF administration. Furthermore, both treatments were effective in inhibiting tumor growth in EA and ES by modulating the levels of interleukin (IL)-6 and tumor necrosis factor (TNF)-α, decreasing mast cells numbers and inducing apoptosis. Conclusions: This research indicates that both CE and EAF from G. brasiliensis leaves have potential antitumor effects with low cytotoxicity. Full article
(This article belongs to the Special Issue Bioactive Compounds Derived from Plants and Their Medicinal Potential)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Standard curves for phytochemical analyses. (<b>A</b>) Standard curve for total flavonoid content, constructed using quercetin as standard, from a serial dilution of 25 μg/mL to 1.56 μg/mL (R<sup>2</sup> = 0.9943). (<b>B</b>) Standard curve for total polyphenol content, constructed using gallic acid as standard, from different concentrations (2; 1.5; 1.0; 0.5; and 0.25 μg/mL—R<sup>2</sup> = 0.9957). Solid lines represent measurements and the dashed lines represent the linear trend line.</p>
Full article ">Figure 2
<p>Scavenging capacity against DPPH. (<b>A</b>) DPPH assay visual result. C+: positive control (standard antioxidant, butylhydroxytoluene-BHT); C−: negative control; CE10: crude extract at a 10% concentration; and EAF10: ethyl acetate fraction at a 10% concentration. (<b>B</b>) Comparisons of total antioxidant activity among controls and CE at a 10% concentration and EAF at a 10% concentration. Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated as **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>In vitro cytotoxicity test by hemolysis. (<b>A</b>) Visual result of the hemolysis test for the CE. Negative control (C−), positive control (C+), CE samples at 5% concentration (CE5), 10% concentration (CE10), 20% concentration (CE20) and 50% concentration (CE50). (<b>B</b>) Graphic of cytotoxicity in red blood cells from the CE samples. Solid line represents measurements and the dashed line represents the linear trend line. (<b>C</b>) Comparisons of cytotoxicity among controls and samples of CE. (<b>D</b>) Visual result of the hemolysis test for the EAF. Negative control (C−), positive control (C+), CE samples at 5% concentration (CE5), 10% concentration (CE10), 20% concentration (CE20) and 50% concentration (CE50). (<b>E</b>) Graphic of cytotoxicity in red blood cells from the EAF samples. Solid line represents measurements and the dashed line represents the linear trend line (<b>F</b>) Comparisons of cytotoxicity among controls and samples of EAF. Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated as **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Blood biochemical analyses of ascitic Ehrlich tumor-induced animals and non-induced animals treated with CE at 10% concentration and EAF at 10% concentration for 7 days. The dosages of (<b>A</b>) ALT, (<b>B</b>) AST, (<b>C</b>) Gamma GT, (<b>D</b>) creatinine, (<b>E</b>) urea, (<b>F</b>) albumin, (<b>G</b>) alkaline phosphatase, (<b>H</b>) uric acid and (<b>I</b>) blood glucose levels were measured in blood plasma. Results presented as mean ± S.E.M (<span class="html-italic">n</span> = 8/group). Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated 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; and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Blood biochemical analyses of solid Ehrlich tumor-induced animals and non-induced animals treated with CE and EAF for 14 days. The dosages of (<b>A</b>) ALT, (<b>B</b>) AST, (<b>C</b>) Gamma GT, (<b>D</b>) creatinine, (<b>E</b>) urea, (<b>F</b>) albumin, (<b>G</b>) alkaline phosphatase, (<b>H</b>) uric acid and (<b>I</b>) blood glucose levels were measured in blood plasma. Results presented as mean ± S.E.M (<span class="html-italic">n</span> = 8/group. Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated as * <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; 0.0001.</p>
Full article ">Figure 6
<p>Analysis of the development of ascitic Ehrlich tumors and effects of treatments. Animals induced with ascitic Ehrlich tumors: (<b>A</b>) untreated. Dashed line represents the abdominal circumference measurement, (<b>B</b>) treated with CE for 7 days and (<b>C</b>) treated with EAF for 7 days. (<b>D</b>) Abdominal circumference (cm), (<b>E</b>) body weight (g), (<b>F</b>) ascitic liquid total (mL), (<b>G</b>) ascitic tumor cells after centrifugation (%) and (<b>H</b>) cell count × 10<sup>5</sup>/mL. Results presented as mean ± S.E.M. (<span class="html-italic">n</span> = 8/group). Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated as * <span class="html-italic">p</span> &lt; 0.05; and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>Analysis of solid Ehrlich tumor development and effects of treatments. Macroscopic analysis showing the tumors in the animals’ bodies (<b>A</b>–<b>C</b>) and after excision (<b>D</b>–<b>F</b>), with the measured axes (major D and minor d), (<b>G</b>) <math display="inline"><semantics> <mrow> <mo>∆</mo> <mtext> </mtext> </mrow> </semantics></math>body weight, (<b>H</b>) major tumor diameter and (<b>I</b>) minor tumor diameter of untreated solid Ehrlich tumor-induced mice (ES) indicated in induced mice treated with crude extract (ES/CE) and induced mice treated with ethyl acetate fraction (ES/EAF). Results are presented as means ± S.E.M. (<span class="html-italic">n</span> = 8/group). Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated as ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 8
<p>Histopathological analysis of solid Ehrlich tumors. Tumor masses of untreated animals (<b>A</b>–<b>C</b>), treated with CE (<b>D</b>–<b>F</b>) and treated with EAF (<b>G</b>–<b>I</b>). Dashed lines indicate regions of necrosis. Yellow arrows show mitosis figures. White asterisks indicate skeletal striated muscle. The detailed magnifications show mitosis figures (<b>C</b>) and necrosis regions (<b>F</b>). Bars of 500 μm (<b>A</b>,<b>D</b>,<b>G</b>), 200 μm (<b>B</b>,<b>E</b>,<b>H</b>) and 50 μm (<b>C</b>,<b>F</b>). Staining, Hematoxylin–Eosin. Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test.</p>
Full article ">Figure 9
<p>Analysis of mast cells and Mcpt6 levels in animals induced with solid Ehrlich tumor. (<b>A</b>) Various mast cells were present in animals induced with tumors without treatment. But fewer mast cells were present in animals induced with tumors and treated (<b>B</b>) with CE or (<b>C</b>) EAF. The insets represent enlargements of the dashed areas. Bars of 50 μm. Staining, Toluidine blue. (<b>D</b>) Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated as * <span class="html-italic">p</span> &lt; 0.05. (<b>E</b>) Western blotting for Mcpt6 expression in animals treated with CE and EAF. (<b>F</b>) Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated as * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 10
<p>Analysis of Bax levels. Animals induced with ascitic Ehrlich tumors and treated with CE 10% concentration and EAF 10% concentration for 7 days (<b>A</b>,<b>C</b>). Animals induced with solid Ehrlich tumors and treated with CE 10% concentration and EAF 10% concentration for 14 days (<b>B</b>,<b>D</b>). Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated as * <span class="html-italic">p</span> &lt; 0.05. and ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 11
<p>Measurement of IL-6, IFN-γ and TNF-α cytokine levels in ascitic (<b>A</b>–<b>C</b>) and solid (<b>D</b>–<b>F</b>) Ehrlich tumors. Statistical analysis was conducted using one-way ANOVA, followed by Tukey’s multiple comparison test. Significance levels are indicated as * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 12
<p>Representative scheme of the results obtained by the administration of CE at 10% concentration and EAF 10% concentration in mice non-induced and induced with ascitic and solid Ehrlich tumors. Both extractive solutions showed potential for tumor growth inhibition. Upward-facing red arrows indicate increased levels. Downward-facing green arrows indicate reduced levels.</p>
Full article ">Figure 13
<p>Groups of animals that were non-induced and induced with Ehrlich ascitic and solid tumors and were treated for 7 days and 14 days.</p>
Full article ">Figure 14
<p>Schematic summary of the assessments used in this investigation.</p>
Full article ">
13 pages, 712 KiB  
Article
Oxidative Status and Lipid Metabolism Analytes in Dogs with Mast Cell Tumors: A Preliminary Study
by Argyrios Ginoudis, Dimitra Pardali, Mathios E. Mylonakis, Androniki Tamvakis, Asta Tvarijonaviciute, Evgenia Lymperaki, Jose Joaquin Ceron and Zoe Polizopoulou
Antioxidants 2024, 13(12), 1473; https://doi.org/10.3390/antiox13121473 - 29 Nov 2024
Viewed by 538
Abstract
Mast cell tumors (MCTs) are common skin neoplasms in dogs. Prognostic indicators include histologic grade, clinical stage, high Ki-67 index, elevated argyrophilic nucleolus organizer regions (AgNOR) index, c-kit mutations, and recurrence after surgery. Blood serum redox status has been shown to correlate with [...] Read more.
Mast cell tumors (MCTs) are common skin neoplasms in dogs. Prognostic indicators include histologic grade, clinical stage, high Ki-67 index, elevated argyrophilic nucleolus organizer regions (AgNOR) index, c-kit mutations, and recurrence after surgery. Blood serum redox status has been shown to correlate with prognostic factors in canine lymphoma and mammary tumors. This study aimed to assess the correlation between established prognostic factors and serum redox status and lipid metabolism analytes in dogs with MCTs. Dogs with cutaneous (n = 33) or subcutaneous (n = 6) MCTs, without comorbidities, were studied. Staging was evaluated based on cytology of regional lymph nodes and ultrasound-guided liver and spleen aspiration cytology. Histologic grading and immunohistochemical staining for Ki-67 and KIT patterns were performed on excised tumor specimens. Dogs were categorized by Patnaik grading (1–3), Kiupel grading (low/high), metastatic status, Ki-67 positive nuclei per cm2 (>23 or ≤23), and KIT pattern (I, II–III). Paraoxonase-1, Butyrylcholinesterase, Cupric Reducing Antioxidant Capacity (CUPRAC), Diacron Reactive Oxygen Metabolites (d-ROMs), and oxy-adsorbent levels were measured before any therapeutic intervention. ANOVA and independent t-tests were used to detect differences in the mean values among groups. Paraoxonase-1 activity was significantly lower in Patnaik grade 3 (p = 0.003) and Kiupel high-grade (p = 0.022) MCTs. No significant differences were found in CUPRAC, d-ROMs, or oxy-adsorbent levels across different prognostic groups. This study found a significant correlation between histologic grading and Paraoxonase-1 activity, suggesting a potential role of Paraoxonase-1 as a prognostic biomarker in canine MCTs. Full article
(This article belongs to the Special Issue Antioxidant Role of High-Density Lipoprotein)
Show Figures

Figure 1

Figure 1
<p>Boxplot illustrating Paraoxonase-1 (PON-1) activity in dogs with cutaneous (<span class="html-italic">n</span> = 33) mast cell tumors (MCTs) categorized by Kiupel histologic grading. A total of 25 dogs had low-grade MCTs, and 8 had high-grade MCTs. The mean PON-1 activity was significantly lower in high-grade MCTs compared to low-grade MCTs (<span class="html-italic">p =</span> 0.022).</p>
Full article ">Figure 2
<p>Boxplot illustrating Paraoxonase-1 (PON-1) activity in dogs with cutaneous (<span class="html-italic">n</span> = 33) mast cell tumors (MCTs) categorized by Patnaik histologic grading. In total, 1 dog had a grade 1 MCT, 25 had grade 2 MCTs, and 7 had grade 3 MCTs. The mean PON-1 activity was significantly lower in grade 3 MCTs (<span class="html-italic">p</span> = 0.003).</p>
Full article ">
21 pages, 4736 KiB  
Article
Anti-Atopic Dermatitis Effect of Azalomycin F on 2,4-Dinitrofluorobenzene-Induced Mice and Potential Mechanism
by Wenjia Zhao, Jianping Zhu, Xinrong Luo, Fengxian Lian, Yanli Yang, Su He, Jinzhou Zhu and Ganjun Yuan
Int. J. Mol. Sci. 2024, 25(23), 12846; https://doi.org/10.3390/ijms252312846 - 29 Nov 2024
Viewed by 592
Abstract
Azalomycin F (AZF) is a kind of antibiotic with antifungal and antibacterial activities, as well as anti-inflammatory and anti-tumor activities. In this study, we evaluated the effects of AZF on atopic dermatitis (AD) and its possible molecular mechanisms. Mice with 2,4-dinitrofluorobenzene-induced AD-like skin [...] Read more.
Azalomycin F (AZF) is a kind of antibiotic with antifungal and antibacterial activities, as well as anti-inflammatory and anti-tumor activities. In this study, we evaluated the effects of AZF on atopic dermatitis (AD) and its possible molecular mechanisms. Mice with 2,4-dinitrofluorobenzene-induced AD-like skin lesions were topically treated with 10–30 mg/kg AZF on their dorsal skin for 12 days. Observations focused on skin lesion scores, the frequency of scratching, and histopathological alterations in the skin. In addition, IgE and inflammatory cytokine levels in serum were assessed. The results indicated that topical application of 10–20 mg/kg AZF could reduce skin lesion scores and scratching frequencies in AD mice, while 15–20 mg/kg AZF decreased epidermal thickness and mast cell infiltration. Additionally, the serum levels of IgE, IFN-γ, IL-4, TSLP and IL-1β were reduced with 10–20 mg/kg AZF treatment. Moreover, RNA-Seq was employed to reveal the potential molecular mechanisms underlying anti-AD effects of AZF. KEGG enrichment analysis revealed that the most significantly differentially expressed genes are predominantly enriched in signaling pathways such as NF-κB and TNF. Protein–protein interaction network analysis identifies the key genes including Il1b, Tnf, and Cxcl1. In summary, 15 mg/kg AZF effectively alleviates the inflammatory response in AD mice, and the potential mechanism may involve the regulation of key signaling pathways like NF-κB and TNF, thereby reducing inflammatory factor levels and eliciting an anti-inflammatory effect. These findings provide valuable scientific evidence for the development of novel natural drugs for the treatment of AD. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>Effects of Azalomycin F (AZF) on atopic dermatitis (AD)-like clinical symptoms in 2,4-dinitrofluorobenzene (DNFB)-induced Balc/c mice. (<b>A</b>) Schematic diagram of the experimental protocol. (<b>B</b>) Images of the dorsal skin of mice in each group. (<b>C</b>) Dermatitis score. (<b>D</b>) The number of scratches by mice within 15 min. The results are expressed as the mean ± SD (<span class="html-italic">n</span> = 12). (<b>E</b>) spleen index and (<b>F</b>) thymus index. The results are expressed as the mean ± SD (n = 6). Differences were assessed by analysis of variance (ANOVA) and denoted as follows: <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, compared to AD group. Tac, tacrolimus.</p>
Full article ">Figure 2
<p>Effects of Azalomycin F (AZF) on DNFB-induced AD-like histopathological alterations in Balb/c mice. (<b>A</b>) Representative images of hematoxylin eosin (H&amp;E) and toluidine blue (TB) staining in each group. H&amp;E-stained specimens were observed under a microscope at 40× and 200× magnification (Scale bar = 500 and 100 μm). TB-stained specimens were observed under a microscope at 200× magnification (Scale bar = 100 μm). Black lines indicate the thickness of the epidermis, yellow arrows denote epidermal thickening, red arrows point to neovascularization, blue arrows represent inflammatory cells, green arrows indicate connective tissue hyperplasia, and black arrows denote mast cells. (<b>B</b>) The epidermal thickness of each group. (<b>C</b>) The number of mast cells in each group. The results are expressed as the mean ± SD (n = 3). Differences were assessed by analysis of variance (ANOVA) and denoted as follows: <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, compared to AD group.</p>
Full article ">Figure 3
<p>Effects of azalomycin F (AZF) on IgE and inflammatory mediator levels in the serum of DNFB-induced Balb/c mice. (<b>A</b>) IgE, (<b>B</b>) IFN-γ, (<b>C</b>) IL-4, (<b>D</b>) TSLP, and (<b>E</b>) IL-1β. The results were expressed as the mean ± SD (n = 5~6). Differences were assessed by analysis of variance (ANOVA) and denoted as follows: <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, compared to AD group; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, compared to AD group.</p>
Full article ">Figure 4
<p>The bioinformatics analyses identified differentially expressed genes (DEGs) and enriched pathways. (<b>A</b>–<b>C</b>) Volcano plot of DEGs in the Con_vs_AD, Con_vs_AZF and AD_vs_AZF group. (<b>D</b>–<b>F</b>) Gene Ontology (GO) annotation of DEGs in the Con_vs_AD, Con_vs_AZF and AD_vs_AZF group. The top 10 functional classified GO terms of DEGs annotated by the subontology of GO analysis including Biological Process (BP), Molecular Function (MF), and Cellular Components (CC). (<b>G</b>,<b>I</b>,<b>J</b>) The top 30 pathways with the most significant Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment in the Con_vs_AD, Con_vs_AZF and AD_vs_AZF group. (<b>H</b>) Venn diagrams of DEGs. CON: control group; AD: atopic dermatitis model group; AZF: 15 mg/kg AZF group.</p>
Full article ">Figure 5
<p>Protein–protein interaction (PPI) network construction of DEGs. (<b>A</b>) The PPI network of DEGs identified from Con_vs_AD. (<b>B</b>) The top 30 highly connected DEGs and the top 3 significant modules extracted from the PPI network (MCODE scores are 15 and 13). (<b>C</b>) PPI network constructed from 137 co-expressed genes between Con_vs_AD and AD_vs._AZF. Red represents gene expression upregulated in (<b>A</b>,<b>B</b>). Node size indicates the number of genes interacted with in (<b>A</b>,<b>C</b>). Line thickness indicates the strength of data support in (<b>C</b>).</p>
Full article ">
17 pages, 1227 KiB  
Article
Mutational Landscape of KIT Proto-Oncogene Coding Sequence in 62 Canine Cutaneous and Subcutaneous Mast Cell Tumors
by Ludovica Montanucci, Elena Guidolin, Rosa Maria Lopparelli, Greta Mucignat, Marianna Pauletto, Mery Giantin and Mauro Dacasto
Vet. Sci. 2024, 11(12), 593; https://doi.org/10.3390/vetsci11120593 - 25 Nov 2024
Viewed by 1010
Abstract
Canine mast cell tumors (MCTs) are common skin neoplasms with varying biological behaviors. The KIT proto-oncogene plays a key role in the development of these tumors, and internal tandem duplications on exon 11 are usually associated with more aggressive behavior, increased local recurrence, [...] Read more.
Canine mast cell tumors (MCTs) are common skin neoplasms with varying biological behaviors. The KIT proto-oncogene plays a key role in the development of these tumors, and internal tandem duplications on exon 11 are usually associated with more aggressive behavior, increased local recurrence, and decreased survival time. However, apart from exons 8–11 and 17, there is limited understanding of the overall KIT mutational landscape in canine MCTs. This work aims to analyze the entire KIT coding sequence (21 exons) in a cohort of 62 MCTs, which included 38 cutaneous and 24 subcutaneous tumors, and potentially identify new variants. In addition to confirming previously reported activating KIT mutations in exons 8, 9, and 11, we identified new variants in exons 2, 3, 5, 16, and the 3′ untranslated region (UTR). Notably, these last variants include an amino acid change (Asp/His) in exon 16. Additionally, we confirmed a differential prevalence of KIT variants in cutaneous and subcutaneous MCTs. These findings enhance our understanding of the KIT proto-oncogene coding sequence and provide valuable information for future confirmatory studies. Full article
(This article belongs to the Special Issue Genetic Diseases and Gene Mutation-Related Tumors in Small Animals)
Show Figures

Figure 1

Figure 1
<p>Graphical representation of the Type III tyrosine kinase receptor encoded by the <span class="html-italic">KIT</span> gene. Ig1–5 indicates the extracellular immunoglobulin-like domains; TMD indicates the transmembrane domain; JMD indicates the intracellular juxtamembrane domain; TK1 and TK2 indicate the tyrosine kinase domains. The PDB (Protein Data Bank) structures used for the visualizations are the structures of the human tyrosine kinase: 8DFP for the extracellular region (exons 1–9, top) and 7ZW8 for the intracellular region (exons 11–21, bottom).</p>
Full article ">Figure 2
<p>cDNA sequence of exons 11 and 12 and its protein translation. The black cDNA sequence indicates exon 11, while the blue cDNA sequence indicates exon 12. The individual codons are highlighted with alternating clear and pink shadows. The ITDs and the complex insertion–duplication are represented in each subsequent line. For each variant, the sequence of only the duplicated region is reported (bold, green). The sequence highlighted in yellow corresponds to the 7 bp insertion of INS-DUP579.</p>
Full article ">
18 pages, 1628 KiB  
Review
The Immunomodulatory Mechanisms of BTK Inhibition in CLL and Beyond
by Qu Jiang, Yayi Peng, Carmen Diana Herling and Marco Herling
Cancers 2024, 16(21), 3574; https://doi.org/10.3390/cancers16213574 - 23 Oct 2024
Viewed by 1488
Abstract
Bruton’s tyrosine kinase (BTK), a cytoplasmic tyrosine kinase, plays a pivotal role in B cell biology and function. As an essential component of the B cell receptor (BCR) signaling pathway, BTK is expressed not only in B cells but also in myeloid cells, [...] Read more.
Bruton’s tyrosine kinase (BTK), a cytoplasmic tyrosine kinase, plays a pivotal role in B cell biology and function. As an essential component of the B cell receptor (BCR) signaling pathway, BTK is expressed not only in B cells but also in myeloid cells, including monocytes/macrophages, dendritic cells, neutrophils, and mast cells. BTK inhibitors (BTKis) have revolutionized the treatment of chronic lymphocytic leukemia (CLL) and other B cell malignancies. Besides their well-characterized role in inhibiting BCR signaling, BTKis also exert significant immunological influences outside the tumor cell that extend their therapeutic potential and impact on the immune system in different ways. This work elucidates the immunomodulatory mechanisms associated with BTK inhibition, focusing on CLL and other clinical contexts. We discuss how BTK inhibition affects various immune cells, including B cells, T cells, and macrophages. The effects of BTKis on the profiles of cytokines, also fundamental parts of the tumor microenvironment (TME), are summarized here as well. This review also appraises the implications of these immunomodulatory actions in the management of autoimmune diseases and infections. Summarizing the dual role of BTK inhibition in modulating malignant lymphocyte and immune cell functions, this paper highlights the broader potential clinical use of compounds targeting BTK. Full article
(This article belongs to the Special Issue Ibrutinib in Chronic Lymphocytic Leukemia)
Show Figures

Figure 1

Figure 1
<p>B cell receptor signaling pathway [<a href="#B5-cancers-16-03574" class="html-bibr">5</a>]. Abbreviations: BCR = B cell receptor; SYK = spleen tyrosine kinase; LYN = Lyn kinase; ITAM = immuno-receptor tyrosine-based activation motif; BTK = Bruton’s tyrosine kinase; PI3K = phosphoinositide 3-kinase; AKT = protein kinase B; PLCγ2 = phospholipase C gamma 2; P = phosphorylation; IP3 = inositol 1,4,5-trisphosphate; CaM = calmodulin; NFAT = nuclear factor of activated T cells; DAG = diacylglycerol; PKC = protein kinase C; NFκB = nuclear factor kappa-light-chain-enhancer of activated B cells; ERK = extracellular signal-regulated kinase; mTOR = mechanistic target of rapamycin. Image created in BioRender.com (accessed date 1 Oct. 2024). A list of abbreviations is included in <a href="#app1-cancers-16-03574" class="html-app">Supplementary Table S1</a>.</p>
Full article ">Figure 2
<p>The role of BTK in myeloid cells; details are in the main text. (I) BTK in neutrophils (green box); (II) BTK in conventional dendritic cells (blue box); (III) BTK in macrophages (gray box); (IV) BTK in mast cells (orange box). Abbreviations: BTK = Bruton’s tyrosine kinase; TLR = Toll-like receptor; GM-CSFR = granulocyte–macrophage colony-stimulating factor receptor; FcεRI = high-affinity IgE receptor; SHIP1 = SH2-containing inositol phosphatase 1; IL-10 = interleukin-10; STAT3 = signal transducer and activator of transcription 3; P = phosphorylation; MYD88 = myeloid differentiation primary response 88; MHC-II = major histocompatibility complex class II; TEC = tyrosine kinase expressed in hepatocellular carcinoma; LPS = lipopolysaccharide; FcγR = Fc gamma receptor; pink X = deficient. Image created in BioRender.com (access date 1 October 2024).</p>
Full article ">Figure 3
<p>BTK inhibition in CLL cells and their environment; details are in the main text. Abbreviations: BCR = B cell receptor; BTK = Bruton’s tyrosine kinase; SYK = spleen tyrosine kinase; LYN = Lyn kinase; PLCγ2 = phospholipase C gamma 2; PKC-βII = protein kinase C β II; NFκB = nuclear factor kappa-light-chain-enhancer of activated B cells; VLA-4 = very late activation antigen 4; VCAM-1 = vascular cell adhesion molecule 1; MSC = mesenchymal stromal cells; IL-6 = interleukin-6; TNF-α = tumor necrosis factor-alpha; PD-L1 = programmed cell death ligand 1; PD-1 = programmed cell death protein 1; MDSCs = myeloid-derived suppressor cells; CAR = chimeric antigen receptor; Treg = regulatory T cells; Th = T helper. Image created in BioRender.com (access date 1 October 2024).</p>
Full article ">
15 pages, 949 KiB  
Article
Regional Variations in and Key Predictors of Feline Tumor Malignancy: A Decade-Long Retrospective Study in Korea
by Byung-Joon Seung, Min-Kyung Bae and Jung-Hyang Sur
Animals 2024, 14(20), 2989; https://doi.org/10.3390/ani14202989 - 16 Oct 2024
Viewed by 865
Abstract
Feline cancer is increasingly recognized as a major cause of mortality, yet data on tumor prevalence and behavior in cats, particularly in non-Western regions, remain limited. This study analyzed a decade of feline tumor data in Korea from 2012 to 2022, focusing on [...] Read more.
Feline cancer is increasingly recognized as a major cause of mortality, yet data on tumor prevalence and behavior in cats, particularly in non-Western regions, remain limited. This study analyzed a decade of feline tumor data in Korea from 2012 to 2022, focusing on age, breed, and anatomical location as predictors of malignancy. Data were collected from 683 cats, with regression analysis applied to determine significant associations. Older cats exhibited a markedly higher risk of malignancy, particularly in mast cell and mammary tumors. Tumors in the mammary gland and alimentary tract had malignancy rates exceeding 90%, underscoring the need for early detection in these regions. Interestingly, squamous cell carcinoma was rare in the skin, in stark contrast to Western studies, likely reflecting differences in environmental exposure. While breed was not a statistically significant predictor, certain breeds, including Persians and Russian Blues, showed a higher frequency of malignancy. These findings highlight the importance of regional tumor research in cats and the need for larger, multicenter datasets that incorporate environmental, genetic, and lifestyle factors. Understanding these influences will help refine veterinary care and improve cancer treatment outcomes in feline populations. Full article
(This article belongs to the Section Companion Animals)
Show Figures

Figure 1

Figure 1
<p>Distribution of benign and malignant tumors in cats. (<b>A</b>) Distribution by age, with error bars representing standard deviations (SDs) to show variability. Asterisks (****) indicate statistically significant differences between groups (<span class="html-italic">p</span> &lt; 0.0001). (<b>B</b>) Distribution by sex, illustrating differences in tumor prevalence between males and females.</p>
Full article ">Figure 2
<p>Patterns of tumor distribution by breed and anatomical location in cats. (<b>A</b>) Distribution by breed, highlighting the comparison of benign and malignant tumor cases among various cat breeds. (<b>B</b>) Distribution by anatomical location, illustrating the prevalence of tumor types in different body regions.</p>
Full article ">
11 pages, 803 KiB  
Article
Circulating Endocannabinoids in Canine Cutaneous Mast Cell Tumor
by Valentina Rinaldi, Fabiana Piscitelli, Andrea Boari, Roberta Verde, Paolo Emidio Crisi and Tiziana Bisogno
Animals 2024, 14(20), 2986; https://doi.org/10.3390/ani14202986 - 16 Oct 2024
Viewed by 1005
Abstract
A cutaneous mast cell tumor (cMCT) is among the most common tumors in dogs. Endocannabinoids (eCBs) belong to the endocannabinoid system (ECS), which involves also cannabinoid receptors and an enzymatic system of biosynthesis and degradation. In this study, plasma levels of N-arachidonoylethanolamine [...] Read more.
A cutaneous mast cell tumor (cMCT) is among the most common tumors in dogs. Endocannabinoids (eCBs) belong to the endocannabinoid system (ECS), which involves also cannabinoid receptors and an enzymatic system of biosynthesis and degradation. In this study, plasma levels of N-arachidonoylethanolamine (AEA), 2-arachidonoylglycerol (2-AG), N-palmitoylethanolamine (PEA), and N-oleoylethanolamine (OEA) were evaluated in 17 dogs with MCTs of varying histological grades and clinical stages, as well as in a control group of 11 dogs. Dogs affected by cMCT had higher plasma levels of 2-AG (p = 0.0001) and lower levels of AEA (p = 0.0012) and PEA (p = 0.0075) compared to the control group, while no differences were observed at the OEA level between healthy and cMCT dogs (p = 0.9264). The ability of eCBs to help discriminate between healthy and cMCT dogs was interrogated through the area under the ROC curve (AUC). An accuracy of 0.98 (95% confidence interval [CI], 0.94–1.02) was found for 2-AG, of 0.85 (95% CI, 0.71–0.99) for AEA, and of 0.81% for PEA (95% CI, 0.64–0.69). Values > 52.75 pmol/mL for 2-AG showed 94% sensitivity and 90% specificity in distinguishing cMCT. This is the first study to demonstrate alterations in plasmatic levels of eCBs in dogs affected by MCTs, suggesting the significance of these biomarkers in the tumorigenic process and their potential use as biomarkers in the future. Full article
(This article belongs to the Section Companion Animals)
Show Figures

Figure 1

Figure 1
<p>Scatter dot plot of plasmatic concentrations expressed in pmol/mL of 2-arachidonoylglycerol (2-AG), <span class="html-italic">N</span>-arachidonoylethanolamine (AEA), <span class="html-italic">N</span>-palmitoylethanolamine (PEA), and N-oleoylethanolamine (OEA) in dogs affected by cutaneous mast cell tumor (cMCT, n = 17) and in healthy dogs (n = 11). Bold font denotes statistical significance.</p>
Full article ">Figure 2
<p>Receiver operating characteristic (ROC) curves of 2-arachidonoylglycerol (2-AG), <span class="html-italic">N</span>-arachidonoylethanolamine (AEA), and <span class="html-italic">N</span>-palmitoylethanolamine (PEA).</p>
Full article ">
14 pages, 3697 KiB  
Article
Efficacy and Potential Mechanisms of Naringin in Atopic Dermatitis
by Seung-Ah Yoo, Ki-Chan Kim and Ji-Hyun Lee
Int. J. Mol. Sci. 2024, 25(20), 11064; https://doi.org/10.3390/ijms252011064 - 15 Oct 2024
Cited by 1 | Viewed by 1145
Abstract
Atopic dermatitis (AD) is one of the most prevalent chronic inflammatory skin diseases. Topical treatments are recommended for all patients regardless of severity, making it essential to develop an effective topical AD treatment with minimal side effects; We investigated the efficacy of topical [...] Read more.
Atopic dermatitis (AD) is one of the most prevalent chronic inflammatory skin diseases. Topical treatments are recommended for all patients regardless of severity, making it essential to develop an effective topical AD treatment with minimal side effects; We investigated the efficacy of topical application of naringin in AD and explored the possible mechanisms using an AD mouse model induced by 1-chloro-2,4-dinitrobenzene (DNCB). Clinical, histological, and immunological changes related to AD and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling proteins in the skin tissues were measured as outcomes; Naringin treatment resulted in a significant improvement in dermatitis severity score and reduced epidermal thickness and mast cell count in the skin (p < 0.05). Naringin also demonstrated the ability to inhibit DNCB-induced changes in interleukin (IL) 4, chemokine (C-C motif) ligand (CCL) 17, CCL22, IL1β, interferon-gamma (IFN-γ), and tumor necrosis factor-alpha (TNF-α) levels by quantitative real-time polymerase chain reaction (qRT-PCR) and IL13 by enzyme-linked immunosorbent assay (ELISA) (p < 0.05). Western blot results exhibited the decreased JAK1, JAK2, STAT1, STAT3, phospho-STAT3, and STAT6 expression in the naringin-treated groups (p < 0.05); The findings of this study suggest that topical naringin may effectively improve the symptoms of AD and could be used as a therapeutic agent for AD. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structures of (<b>A</b>) flavonoid and (<b>B</b>) naringin.</p>
Full article ">Figure 2
<p>Schematic description of the experiment (n = 6 per group). DNCB, 1-chloro-2,4-dinitrobenzene.</p>
Full article ">Figure 3
<p>(<b>A</b>) Clinical images of the ear and back at the end of the challenge period (day 16). (<b>B</b>) Graphs representing dermatitis severity and ear thickness assessment results. Values represent the mean ± SEM (n = 6). Data compared among multiple groups were analyzed using one-way analysis of variance. * <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 compared to the NC or DNCB-only group. DNCB, 1-chloro-2,4-dinitrobenzene; NC, normal control.</p>
Full article ">Figure 4
<p>(<b>A</b>) Pathology images of H&amp;E and toluidine blue staining in dorsal skin tissue samples. Mast cells are stained purple with toluidine blue. Original magnification = ×200, scale bar = 100 μm. (<b>B</b>) Graphs representing the epidermal thickness and the number of mast cells. Values are mean ± SEM (n = 6). Data compared among multiple groups were analyzed using one-way analysis of variance. *** <span class="html-italic">p</span> &lt; 0.001 compared to the NC or DNCB-only group. H&amp;E, hematoxylin and eosin; NC, normal control; DNCB, 1-chloro-2,4-dinitrobenzene.</p>
Full article ">Figure 5
<p>Expressed mRNA level of (<b>A</b>) TNF-α, (<b>B</b>) IFN-γ, (<b>C</b>) IL4, (<b>D</b>) CCL17, (<b>E</b>) CCL22, (<b>F</b>) IL1β, (<b>G</b>) IL31, and (<b>H</b>) TSLP quantified by qRT-PCR and (<b>I</b>) IL13 by ELISA in dorsal skin tissues. The expression of each gene was normalized to that of Actb. Each qRT-PCR reaction was performed in triplicate. Values are mean ± SEM (n = 6). Data compared among multiple groups were analyzed using one-way analysis of variance. * <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 compared to the NC or DNCB-only group. CCL, chemokine C-C motif ligand; DNCB, 1-chloro-2,4-dinitrobenzene; IL, interleukin; INF-γ, interferon-gamma; NC, normal control; TNF-α, tumor necrosis factor-alpha; TSLP, thymic stromal lymphopoietin.</p>
Full article ">Figure 6
<p>Expression of JAK–STAT proteins in dorsal skin tissues. Immunoblotting intensities were calculated with ImageJ software (version 1.8.0). Values are mean ± SEM (n = 6). Data compared among multiple groups were analyzed using one-way analysis of variance. * <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 compared to the NC or DNCB-only group. DNCB, 1-chloro-2,4-dinitrobenzene; JAK-STAT, Janus kinase-signal transducer and activator of transcription; NC, normal control; P-STAT3, phospho-STAT3.</p>
Full article ">
11 pages, 1512 KiB  
Article
Investigation of the Theragnostic Role of KIT Expression for the Treatment of Canine Mast Cell Tumors with Tyrosine Kinase Inhibitors
by Davide De Biase, Marcello De Leo, Giuseppe Piegari, Ilaria d’Aquino, Evaristo Di Napoli, Carmela Mercogliano, Alfonso Calabria, Agata Pula, Luigi Navas, Valeria Russo and Orlando Paciello
Vet. Sci. 2024, 11(10), 492; https://doi.org/10.3390/vetsci11100492 - 10 Oct 2024
Viewed by 1343
Abstract
Several reports have indicated that canine MCTs express a mutated form of a tyrosine kinase receptor, namely KIT, that is involved in abnormal mast cell growth and differentiation. Currently, the post-surgical prognosis for MCTs is related to three different KIT immunohistochemical expression patterns. [...] Read more.
Several reports have indicated that canine MCTs express a mutated form of a tyrosine kinase receptor, namely KIT, that is involved in abnormal mast cell growth and differentiation. Currently, the post-surgical prognosis for MCTs is related to three different KIT immunohistochemical expression patterns. However, to our knowledge, there are few studies specifically exploring the efficacy of treatment with tyrosine kinase inhibitors related to KIT staining pattern. The purpose of this study was to investigate the potential theragnostic role of KIT expression patterns by studying their correlation to the overall survival and progression-free survival in dogs treated with only tyrosine kinase inhibitors immediately after surgery. We selected 66 cases of canine cutaneous MCTs with complete clinical background. A statistical analysis was performed to assess the overall survival status. Our data suggest an important role of KIT in the etiopathogenesis of canine MCTs and indicate that the anomalous cytoplasmatic distribution of KIT is potentially related to a lower efficacy of tyrosine kinase inhibitors, thus providing a significant prognostic information about the treatment outcome. Full article
Show Figures

Figure 1

Figure 1
<p>KIT immunohistochemical staining patterns. (<b>A</b>) Pattern 1, identified by membranous labeling in more than 90% of neoplastic cells. (<b>B</b>) Pattern 2, identified by focal, perinuclear or stippled cytoplasmic labeling with loss of perimembranous labeling in more than 10% of neoplastic cells. (<b>C</b>) Pattern 3, identified by diffuse cytoplasmic labeling in more than 10% of neoplastic cells.</p>
Full article ">Figure 2
<p>Kaplan–Meier survival graph comparing the cumulative survival times in the three KIT expression subclasses: (<b>a</b>) pattern 1, (<b>b</b>) pattern 2 and (<b>c</b>) pattern 3.</p>
Full article ">Figure 3
<p>Kaplan–Meier survival graph comparing the progression-free survival times in the three KIT expression subclasses: (<b>a</b>) pattern 1, (<b>b</b>) pattern 2 and (<b>c</b>) pattern 3.</p>
Full article ">Figure 4
<p>Kaplan–Meier survival graph comparing the three KIT staining patterns in term of cumulative survival times in dogs with MCTs treated post-surgery with TKI therapy. (<b>a</b>) KIT pattern 1 vs. KIT pattern 3; (<b>b</b>) KIT pattern 1 vs. KIT pattern 2; (<b>c</b>) KIT pattern 2 vs. KIT pattern 3.</p>
Full article ">Figure 5
<p>Kaplan–Meier survival graph comparing the three KIT staining patterns in term of progression-free survival times in dogs with MCTs treated post-surgery with TKI therapy. (<b>a</b>) KIT pattern 1 vs. KIT pattern 3; (<b>b</b>) KIT pattern 1 vs. KIT pattern 2; (<b>c</b>) KIT pattern 2 vs. KIT pattern 3.</p>
Full article ">Figure 6
<p>Kaplan–Meier survival graph comparing the three KIT staining patterns in term of cumulative survival times in dogs with MCTs treated with surgery alone. (<b>a</b>) KIT pattern 1 vs. KIT pattern 3; (<b>b</b>) KIT pattern 1 vs. KIT pattern 2; (<b>c</b>) KIT pattern 2 vs. KIT pattern 3.</p>
Full article ">Figure 7
<p>Kaplan–Meier survival graph comparing the three KIT staining patterns in term of progression-free survival times in dogs with MCTs treated with surgery alone. (<b>a</b>) KIT pattern 1 vs. KIT pattern 3; (<b>b</b>) KIT pattern 1 vs. KIT pattern 2; (<b>c</b>) KIT pattern 2 vs. KIT pattern 3.</p>
Full article ">
17 pages, 6521 KiB  
Article
DNA Methylation Negatively Regulates Gene Expression of Key Cytokines Secreted by BMMCs Recognizing FMDV-VLPs
by Mingzhu Li, Peng Ning, Ruoman Bai, Zhanyun Tian, Shujia Liu and Limin Li
Int. J. Mol. Sci. 2024, 25(19), 10849; https://doi.org/10.3390/ijms251910849 - 9 Oct 2024
Viewed by 1099
Abstract
Virus-like particles (VLPs) have been studied and used as vaccines to control foot-and-mouth disease (FMD). Mast cells (MCs) express various pattern recognition receptors that recognize pathogens and secrete numerous cytokines to initiate and modulate immune responses. Our previous study showed that bone marrow-derived [...] Read more.
Virus-like particles (VLPs) have been studied and used as vaccines to control foot-and-mouth disease (FMD). Mast cells (MCs) express various pattern recognition receptors that recognize pathogens and secrete numerous cytokines to initiate and modulate immune responses. Our previous study showed that bone marrow-derived mast cells (BMMCs) can recognize foot-and-mouth disease virus-like particles (FMDV-VLPs) to differentially express various cytokines and that histone acetylation can regulate the cytokines secreted during BMMC recognition of FMDV-VLPs. To demonstrate the role of DNA methylation in this response process, BMMCs that recognize FMDV-VLPs were treated with azacytidine (5-AZA), an inhibitor of DNA methylation transferase. We prepared FMDV-VLPs as described previously and cultured the BMMCs. The transcription and expression of key cytokines and transcription factors were determined using real-time quantitative PCR (RT-qPCR) and Western blotting. Results showed that pre-treatment with AZA resulted in the increased transcription and expression of tumor necrosis factor α (TNF-α), interleukin (IL)-6, IL-13, and IL-10, while the changes in IL-13 transcription and IL-6 expression were irrelevant to mannose receptors (MRs). Furthermore, analysis of the transcription factors indicated that both the transcription and expression of nuclear factor-kappa B (NF-κB) increased significantly in the AZA pre-treated group, indicating that DNA methylation may also regulate NF-κB expression to modulate TNF-α, IL-13, and IL-6. However, pre-treatment with AZA did not alter the expression of microphthalmia-associated transcription factor (MITF) or GATA-2. All the data demonstrate that DNA methylation negatively regulates the transcription and expression of TNF-α, IL-13, IL-10, and IL-6 secreted by recognizing FMDV-VLPs. These results provide new ideas for the mast cell-based design of more effective vaccine adjuvants and targeted therapies in the future. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>Expression and identification of FMDV-VLPs. (<b>A</b>) CHO-K1 cells transfected with recombinant plasmid. (<b>B</b>) CHO-K1 cells without being transfected. (<b>C</b>) Purification and identification of FMDV-VLPs by SDS-PAGE. M, prestained protein ladder; 1, unpurified supernatants of CHO-K1 cells transfected with recombinant plasmid; 2, purified supernatants of CHO-K1 cells transfected with recombinant plasmid. (<b>D</b>) Purification and identification of FMDV-VLPs by Western blot. M, prestained protein ladder; 1, purified supernatants of CHO-K1 cells transfected with recombinant plasmid. (<b>E</b>) Identification of FMDV-VLPs by transmission microscopy. Virus-like particles approximately 30 nm in diameter were observed (indicated by red arrows). Full-length blots are presented in <a href="#app1-ijms-25-10849" class="html-app">Figure S1</a>.</p>
Full article ">Figure 2
<p>Identification of BMMCs by flow cytometry. BMMCs were identified by flow cytometry at 8 weeks post-culture. (<b>A</b>) Allophycocyanin (APC)Armenian Hamster IgG Isotype control antibodies (BioLegend, San Diego, CA, USA) and fluorescein isothiocyanate (FITC) Rat IgG2b, κ isotype control antibodies were used as isotype controls (BioLegend, San Diego, CA, USA). (<b>B</b>) BMMCs were directly labeled with APC Armenian Hamster anti-mouse FcεRIα antibodies and FITC Rat anti-mouse CD117 (c-kit) antibodies and identified using flow cytometry. BMMCs, bone marrow-derived mast cells.</p>
Full article ">Figure 3
<p>Determination of 5-AZA dose and effect on BMMC viability. The relative expression of IL-6 was detected by real-time quantitative PCR after 5-AZA treatment for 24 h (<b>A</b>) and 48 h (<b>B</b>), respectively. Subfigures (<b>C</b>,<b>D</b>) represent the BMMC viability of 5-AZA treatment for 24 h and 48 h, respectively. For cell viability, there were no significances between any two groups. The BMMCs were incubated with FMDV-VLPs for 6 h before 5-AZA treatment. Data are representative of three experiments. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Determination of cytokine transcription by RT-qPCR in different treatment groups. BMMCs were treated with 20 nmol/mL 5-AZA for 24 h. Bone marrow-derived mast cells were harvested for mRNA analysis. The mock group represents pure BMMC culture, while iMR + AZA + VLP represents BMMCs inoculated with mannan, then treated with 5-AZA, and finally added with VLPs. iMR + VLP represents BMMCs inoculated with mannan and then inoculated with VLPs. VLP represents BMMCs inoculated with VLPs. AZA + VLPs represents BMMCs treated with 5-AZA and then inoculated with VLPs. Mannan + AZA represents BMMCs treated with mannan and then inoculated with 5-AZA. Mannan represents BMMCs inoculated with mannan. AZA refers to 5-AZA. Data are presented as Mean ± SD from three independent experiments and normalized to GAPDH. Standard curves of the real-time quantitative PCR are summarized in <a href="#app1-ijms-25-10849" class="html-app">Figure S2</a>. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Determination of cytokines expression by ELISA in different treatment groups. BMMCs were treated with 20 nmol/mL 5-AZA for 24 h. The supernatants were collected from the corresponding groups and determined the expression of IL-6, TNF-α, IL-13, and IL-10, respectively. BMMCs without any pre-treatment group are indicated as MOCK. iMR + AZA + VLP represents BMMCs inoculated with mannan, then treated with 5-AZA, finally added with VLPs. iMR + VLP represents BMMCs inoculated with mannan and then inoculated with VLPs. VLP represents BMMCs inoculated with VLPs. AZA + VLPs represents BMMCs treated with 5-AZA and then inoculated with VLPs. Mannan +AZA represents BMMCs treated with mannan and then inoculated with 5-AZA. Mannan represents BMMCs inoculated with mannan. AZA refers to 5-AZA. Data are presented as Mean ± SD from three independent experiments, each with triplicate samples. * <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 6
<p>Determination of transcription factors mRNA expression. RT-qPCR was utilized to determine the expression of <span class="html-italic">NF-κB</span>, <span class="html-italic">MITF</span>, and <span class="html-italic">GATA-2</span> in different BMMC groups at different times. BMMCs without any pre-treatment group are indicated as MOCK. iMR + AZA + VLP represents BMMCs inoculated with mannan, then treated with 5-AZA, and finally added with VLPs. iMR + VLP represents BMMCs inoculated with mannan and then inoculated with VLPs. VLP represents BMMCs inoculated with VLPs. AZA + VLPs represents BMMCs treated with 5-AZA and then inoculated with VLPs. Mannan + AZA represents BMMCs treated with mannan and then inoculated with 5-AZA. Mannan represents BMMCs inoculated with mannan. AZA refers to 5-AZA. Data are presented as Mean ± SD from three independent experiments and normalized to <span class="html-italic">GAPDH</span>. * <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 7
<p>Determination of transcription factors mRNA expression by Western blot. BMMCs without any pre-treatment group are indicated as MOCK. iMR + AZA + VLP represents BMMCs inoculated with mannan, then treated with 5-AZA, and finally added with VLPs. iMR + VLP represents BMMCs inoculated with mannan and then inoculated with VLPs. VLP represents BMMCs inoculated with VLPs. AZA + VLPs represents BMMCs treated with 5-AZA and then inoculated VLPs. Mannan + AZA represents BMMCs treated with mannan and then inoculated with 5-AZA. Mannan represents BMMCs inoculated with mannan. AZA refers to 5-AZA. Data are presented as Mean ± SD from three independent experiments. (<b>A</b>,<b>B</b>) represents the Western blot results and quantitative analysis of NF-κB p65, respectively; (<b>C</b>,<b>D</b>) represents the Western blot results and quantitative analysis of MITF, respectively; (<b>E</b>,<b>F</b>), represents the Western blot results and quantitative analysis of GATA-2, respectively. Full-length blots are presented in <a href="#app1-ijms-25-10849" class="html-app">Figure S1</a>. * <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 8
<p>Flow chart of methylation sequencing and statistical analysis of methylation levels in cytokine promoter regions between groups. (<b>A</b>) The flow chart of methylation sequencing. (<b>B</b>–<b>E</b>) represents methylation levels in the <span class="html-italic">IL-6</span>, <span class="html-italic">IL-13</span>, <span class="html-italic">TNF-α</span>, and <span class="html-italic">IL-10</span> promotor regions, respectively. BMMCs without any pre-treatment group are indicated as MOCK. iMR + AZA + VLP represents BMMCs inoculated with mannan, then treated with 5-AZA, and finally added with VLPs. iMR + VLP represents BMMCs inoculated with mannan and then inoculated with VLPs. VLP represents BMMCs inoculated with VLPs. AZA + VLPs represents BMMCs treated with 5-AZA and then inoculated VLPs. AZA refers to 5-AZA. Each datapoint represents the average of DNA methylation rate in each locus. DNA methylation rate in each locus = number of methylations at that locus/number of sequences measured. A one-way ANOVA was carried out. *** <span class="html-italic">p</span> &lt; 0.001, ns indicates no significance.</p>
Full article ">Figure 9
<p>Methylation rate of each locus in the <span class="html-italic">IL-6</span>, <span class="html-italic">IL-13</span>, <span class="html-italic">TNF-α</span>, and <span class="html-italic">IL-10</span> promotor regions. (<b>A</b>) <span class="html-italic">IL-6</span>. (<b>B</b>) <span class="html-italic">IL-13</span>. (<b>C</b>) <span class="html-italic">TNF-α</span>. (<b>D</b>) <span class="html-italic">IL-10</span>. BMMCs without any pre-treatment group are indicated as MOCK. iMR + AZA + VLP represents BMMCs inoculated with mannan, then treated with 5-AZA, and finally added with VLPs. iMR + VLP represents BMMCs inoculated with mannan and then inoculated with VLPs. VLP represents BMMCs inoculated with VLPs. AZA + VLPs represents BMMCs treated with 5-AZA and then inoculated VLPs. AZA refers to 5-AZA. The abscissa number indicates the location of each CpG locus relative to the 5′-UTR. DNA methylation rate in each locus = number of methylations at that locus/number of sequences measured. Z-test was carried out. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
23 pages, 1950 KiB  
Review
Beyond Cancer Cells: How the Tumor Microenvironment Drives Cancer Progression
by Hussein Sabit, Borros Arneth, Shaimaa Abdel-Ghany, Engy F. Madyan, Ashraf H. Ghaleb, Periasamy Selvaraj, Dong M. Shin, Ramireddy Bommireddy and Ahmed Elhashash
Cells 2024, 13(19), 1666; https://doi.org/10.3390/cells13191666 - 9 Oct 2024
Viewed by 2886
Abstract
Liver cancer represents a substantial global health challenge, contributing significantly to worldwide morbidity and mortality. It has long been understood that tumors are not composed solely of cancerous cells, but also include a variety of normal cells within their structure. These tumor-associated normal [...] Read more.
Liver cancer represents a substantial global health challenge, contributing significantly to worldwide morbidity and mortality. It has long been understood that tumors are not composed solely of cancerous cells, but also include a variety of normal cells within their structure. These tumor-associated normal cells encompass vascular endothelial cells, fibroblasts, and various inflammatory cells, including neutrophils, monocytes, macrophages, mast cells, eosinophils, and lymphocytes. Additionally, tumor cells engage in complex interactions with stromal cells and elements of the extracellular matrix (ECM). Initially, the components of what is now known as the tumor microenvironment (TME) were thought to be passive bystanders in the processes of tumor proliferation and local invasion. However, recent research has significantly advanced our understanding of the TME’s active role in tumor growth and metastasis. Tumor progression is now known to be driven by an intricate imbalance of positive and negative regulatory signals, primarily influenced by specific growth factors produced by both inflammatory and neoplastic cells. This review article explores the latest developments and future directions in understanding how the TME modulates liver cancer, with the aim of informing the design of novel therapies that target critical components of the TME. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Figure 1

Figure 1
<p>Tumor microenvironment components. Cancer cells are surrounded by numerous non-cancerous cells including those related to the immune system such as B cells, T-cells, dendritic cells, monocytes, eosinophils, and basophils, among others. Cancer-associated fibroblasts are also common in the TME.</p>
Full article ">Figure 2
<p>Tumor cell interaction with the microenvironment. The interplay between cancer cells and The TME is the main factor in cancer progression. Some inducers are released from the tumor cells to affect other components such as the FGF family, VEGF, and IL-8 among others which induce angiogenesis, and other factors including TGF-b1 and FASL induce immunosuppression. Tumor cells also release IL-4 and IL-19 to induce immune cell recruitment and education and release CCL2, IL-1, and IL-6 to activate bone marrow to take part in the production of myeloid cell recruitment. GM-CSF: Granulocyte-macrophage colony-stimulating factor, FGF: Fibroblast growth factor, Ang2: Angiopoietin-2, PDGF: Platelet-derived growth factor, PD-L1: Programmed Cell Death Ligand-1, TIM3: T-cell immunoglobulin domain and mucin domain 3, FASL: Fas ligand, CXCL: chemokines, CAF: cancer-associated fibroblasts, IL: Interleukin, M2: M2 macrophage, N2: Neutrophil, Treg: T-regulatory lymphocyte, MDSC: myeloid-derived suppressor cell, MMP: Matrix metalloproteinase, GF: Growth factor, IDO: Indolamine 2,3 dioxygenase.</p>
Full article ">Figure 3
<p>Immunological mechanisms regulating tumor growth. (<b>A</b>) Tumor suppressive microenvironment. Tumor cell proliferation is inhibited by activated CD4+, CD8+, NK, M1 macrophages, and neutrophils. (<b>B</b>) Immunosuppressive microenvironment. Tumor cells that secrete factors such as TGFβ1, G-CSF, etc. promote MDSC, Treg cells, and M2 macrophages, which inhibit anti-tumor T-cells and NK cells.</p>
Full article ">
16 pages, 2184 KiB  
Article
Age at Tumor Diagnosis in 14,636 Canine Cases from the Pathology-Based UNIPI Animal Cancer Registry, Italy: One Size Doesn’t Fit All
by Niccolò Fonti, Francesca Parisi, Alessio Lachi, Elena Sophie Dhein, Franco Guscetti, Alessandro Poli and Francesca Millanta
Vet. Sci. 2024, 11(10), 485; https://doi.org/10.3390/vetsci11100485 - 8 Oct 2024
Viewed by 2056
Abstract
Cancer is the most common cause of death in adult dogs. All dogs would benefit from early diagnosis, but there are no specific guidelines regarding the schedule of cancer screening in companion animals. The aim of this study was to retrospectively evaluate the [...] Read more.
Cancer is the most common cause of death in adult dogs. All dogs would benefit from early diagnosis, but there are no specific guidelines regarding the schedule of cancer screening in companion animals. The aim of this study was to retrospectively evaluate the age at diagnosis in Italian oncological canine patients. A total of 14,636 canine histologically confirmed neoplastic cases were coded according to the Vet-ICD-O-canine-1 and stratified by malignancy, sex, neutering status, breed, cephalic index, body size, and tumor type. Differences in age distribution were analyzed and the influence of these variables on the time of first malignancy diagnosis was assessed using an event history analysis model. The median age at diagnosis for benign and malignant tumors was 9 and 10 years, respectively. Intact and purebred dogs were diagnosed earlier, but the median age differed significantly by breed. The earliest age at diagnosis was recorded for lymphomas and mast cell tumors. The model showed an accelerating effect of large size, brachy- and dolichocephaly, and sexual integrity in female dogs on the time of malignancy diagnosis. Our results confirm that a “one-size-fits-all” approach to cancer screening is not accurate in dogs and provide relevant data that may lead to the establishment of breed-based screening schedules. Full article
(This article belongs to the Special Issue Focus on Tumours in Pet Animals)
Show Figures

Figure 1

Figure 1
<p>Age distribution of benign and malignant tumors. In brackets: mean; median age at diagnosis in years. *** <span class="html-italic">p</span> &lt; 0.001, Mann–Whitney U test. n = number.</p>
Full article ">Figure 2
<p>Age at malignant tumor diagnosis by sex and neutering status in 8486 cases. In brackets: mean; median age at diagnosis in years. *** <span class="html-italic">p</span> &lt; 0.001, ns = not significant (<span class="html-italic">p</span> &gt; 0.05), Pairwise Wilcoxon rank sum test. n = number.</p>
Full article ">Figure 3
<p>Age at malignant tumor diagnosis for the most common breeds (&gt;50 cases) in 7671 cases. In brackets on the right: mean; median age at diagnosis in years. * <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, Dunn’s test for “purebred” vs. “mixed-breed” comparison. n = number.</p>
Full article ">Figure 4
<p>Age at tumor diagnosis for the 10 most common malignant histotypes in 8143 cases. In brackets on the right: mean; median age at diagnosis in years for the overall dog population (mixed-breed and purebred). The following Vet-ICD-O-canine-1 codes were used for histotype classification: “Skin adnexal neoplasms” = [839–842], “Hard tissue sarcomas” = [918–924], “Blood vessel neoplasms” = [912–916], “Soft tissue sarcomas” = [880–892, 954–957], “Melanomas” = [972–892], “Adenocarcinomas” = [814–838], “SCCs” (Squamous cell carcinomas) = [805–808], “Lymphomas” = [959–972], “Complex neoplasms” = [893–899], “MCTs” (Mast cell tumors) = [974]. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, Pairwise Wilcoxon rank sum test for “purebred” vs. “mixed-breed” comparison for each histotype. n = number.</p>
Full article ">Figure 5
<p>Accelerated Failure Time (AFT) model-generated estimated hazard function. The hazard rate (<span class="html-italic">y</span> axis) expresses the instantaneous risk that the event of malignant tumor diagnosis occurs at time <span class="html-italic">t’</span>, given that the event did not occur before time <span class="html-italic">t</span> (<span class="html-italic">x</span> axis).</p>
Full article ">Figure 6
<p>Accelerated Failure Time (AFT) model-generated estimated survival functions in respect to malignant tumor diagnosis in female dogs, providing a comparison of intact (blue line) vs. neutered (red line) female dogs.</p>
Full article ">
11 pages, 2202 KiB  
Case Report
Long-Term Survival in Canine Hepatosplenic T-Cell Lymphoma Treated with Toceranib Phosphate Following Splenectomy: A Case of Atypical Lymphoma
by Makoto Akiyoshi, Masaharu Hisasue, Midori Goto Asakawa and Sakurako Neo
Vet. Sci. 2024, 11(10), 458; https://doi.org/10.3390/vetsci11100458 - 1 Oct 2024
Viewed by 1703
Abstract
Toceranib phosphate (toceranib) is approved for canine mast cell tumor treatment. However, no long-term response to toceranib in canine HSTCL has been reported. Here, we describe a case of a 10-year-old castrated mixed-breed dog that presented with a 3-month history of weight loss, [...] Read more.
Toceranib phosphate (toceranib) is approved for canine mast cell tumor treatment. However, no long-term response to toceranib in canine HSTCL has been reported. Here, we describe a case of a 10-year-old castrated mixed-breed dog that presented with a 3-month history of weight loss, polydipsia, and polyuria. The clinicopathological and imaging abnormalities included icterus, biliary obstruction, and splenomegaly with multiple diffuse splenic hypoechoic nodules. On day 21, a cholecystectomy was performed to remove the obstruction, followed by a liver biopsy and splenectomy. Cytology of the spleen and liver showed many small lymphocytes with intracytoplasmic granules (sGLs). Splenic and hepatic infiltration of neoplastic CD3/granzyme B-positive small cells and lymphocytic cholecystitis with granzyme B-negative small cells were noted. T-cell receptor gene clonal rearrangements were observed in the liver tissues. The dog was diagnosed with a hepatosplenic T-cell lymphoma (HSTCL) of sGLs concurrent with lymphocytic cholecystitis. The icterus resolved after surgery, but there was progressive elevation of liver enzyme levels. Toceranib was administered from day 39, resulting in decreased liver enzyme levels, and the dog remained in good condition. The dog stayed in remission after toceranib administration and survived for 460 days. Toceranib should be considered an effective treatment option for canine HSTCL. Full article
(This article belongs to the Special Issue Histopathology and Therapy in Small Animals Oncology)
Show Figures

Figure 1

Figure 1
<p>Photomicrographs of liver (<b>A</b>) and spleen (<b>B</b>) cytology, and Wright and Giemsa staining. Cytological analysis of the liver and spleen revealed a moderate number of small lymphocytes with clusters of well-differentiated hepatocytes. The lymphocytes had small (up to 1.5 × RBC) nuclei with indistinct nucleoli and small amounts of light blue cytoplasm with small amounts of fine, red to magenta granules.</p>
Full article ">Figure 2
<p>Photomicrograph of histopathology (<b>A</b>,<b>D</b>,<b>G</b>) and immunohistochemistry of anti CD3 antibody (<b>B</b>,<b>E</b>,<b>H</b>) and anti-granzyme B antibody (<b>C</b>,<b>F</b>,<b>I</b>) of the liver (<b>A</b>–<b>C</b>), spleen (<b>D</b>–<b>F</b>), and gallbladder (<b>G</b>–<b>I</b>). Histopathology revealed infiltration of moderate numbers of small lymphocytes with relatively spared hepatic lobular architecture. These neoplastic lymphocytes are stained positively for anti-CD3 antibody (<b>B</b>) and anti-granzyme B antibody (<b>C</b>) and negative for anti-CD20 antibody. Histopathology of the spleen showed diffuse congestion and enlarged red pulps with small lymphocytes. Approximately 50% of the lymphocytes are stained positively for anti-CD3 antibody (<b>E</b>) and anti-granzyme B antibody (<b>F</b>) and negative for anti-CD20 antibody. In the gallbladder, approximately 30% of infiltrating small lymphocytes are stained positively for anti-CD3 antibody (<b>H</b>), but unlike those in the spleen and liver, negative for anti-granzyme B antibody (<b>I</b>).</p>
Full article ">Figure 3
<p>Clinical course of this case. The graph shows ALT (solid lines) and ALP (dotted lines) transition, and each drug’s dose transition and administration date. L: L-asparaginase, 400 U/kg, SC, on day 341. V: vinblastine, 2.0 mg/m<sup>2</sup>, IV, on day 348. M: Masitinib, 11.1 mg/kg/day, PO, from day 355 until day 361. C: carboplatin, 200 mg/m<sup>2</sup>, IV, on day 362. D: doxorubicin, 30 mg/m<sup>2</sup>, IV, on day 383. H: chlorambucil, 2 mg/head/each other day, PO, from day 398 until 404. A: nimustine, 25 mg/m<sup>2</sup>, IV, on day 405, 426 and 447.</p>
Full article ">
Back to TopTop