[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,213)

Search Parameters:
Keywords = mast cells

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 23025 KiB  
Article
Disulfidptosis: A New Target for Parkinson’s Disease and Cancer
by Tingting Liu, Xiangrui Kong and Jianshe Wei
Curr. Issues Mol. Biol. 2024, 46(9), 10038-10064; https://doi.org/10.3390/cimb46090600 - 12 Sep 2024
Viewed by 311
Abstract
Recent studies have uncovered intriguing connections between Parkinson’s disease (PD) and cancer, two seemingly distinct disease categories. Disulfidptosis has garnered attention as a novel form of regulated cell death that is implicated in various pathological conditions, including neurodegenerative disorders and cancer. Disulfidptosis involves [...] Read more.
Recent studies have uncovered intriguing connections between Parkinson’s disease (PD) and cancer, two seemingly distinct disease categories. Disulfidptosis has garnered attention as a novel form of regulated cell death that is implicated in various pathological conditions, including neurodegenerative disorders and cancer. Disulfidptosis involves the dysregulation of intracellular redox homeostasis, leading to the accumulation of disulfide bonds and subsequent cell demise. This has sparked our interest in exploring common molecular mechanisms and genetic factors that may be involved in the relationship between neurodegenerative diseases and tumorigenesis. The Gene4PD database was used to retrieve PD differentially expressed genes (DEGs), the biological functions of differential expression disulfidptosis-related genes (DEDRGs) were analyzed, the ROCs of DEDRGs were analyzed using the GEO database, and the expression of DEDRGs was verified by an MPTP-induced PD mouse model in vivo. Then, the DEDRGs in more than 9000 samples of more than 30 cancers were comprehensively and systematically characterized by using multi-omics analysis data. In PD, we obtained a total of four DEDRGs, including ACTB, ACTN4, INF2, and MYL6. The enriched biological functions include the regulation of the NF-κB signaling pathway, mitochondrial function, apoptosis, and tumor necrosis factor, and these genes are rich in different brain regions. In the MPTP-induced PD mouse model, the expression of ACTB was decreased, while the expression of ACTN4, INF2, and MYL6 was increased. In pan-cancer, the high expression of ACTB, ACTN4, and MYL6 in GBMLGG, LGG, MESO, and LAML had a poor prognosis, and the high expression of INF2 in LIHC, LUAD, UVM, HNSC, GBM, LAML, and KIPAN had a poor prognosis. Our study showed that these genes were more highly infiltrated in Macrophages, NK cells, Neutrophils, Eosinophils, CD8 T cells, T cells, T helper cells, B cells, dendritic cells, and mast cells in pan-cancer patients. Most substitution mutations were G-to-A transitions and C-to-T transitions. We also found that miR-4298, miR-296-3p, miR-150-3p, miR-493-5p, and miR-6742-5p play important roles in cancer and PD. Cyclophosphamide and ethinyl estradiol may be potential drugs affected by DEDRGs for future research. This study found that ACTB, ACTN4, INF2, and MYL6 are closely related to PD and pan-cancer and can be used as candidate genes for the diagnosis, prognosis, and therapeutic biomarkers of neurodegenerative diseases and cancers. Full article
(This article belongs to the Section Bioinformatics and Systems Biology)
Show Figures

Figure 1

Figure 1
<p>The DEDRG-enriched GO terms and KEGG pathways. (<b>A</b>) GOBP, GOCC, and GOMF analysis. (<b>B</b>) Signaling pathway enrichment analysis. Red represents DEDRGs, green represents biological process, purple represents molecular function, orange represents cellular component, and blue represents signaling pathways.</p>
Full article ">Figure 2
<p>Spatio-temporal expression profiles of (<b>A</b>) ACTB, (<b>B</b>) ACTN4, (<b>C</b>) INF2, and (<b>D</b>) MYL6 retrieved from BrainSpan. The darker the blue color, the higher the protein expression level in the brain region.</p>
Full article ">Figure 3
<p>Diagnostic value of DEDRGs in (<b>A</b>) 16 PD and 9 control subjects from the substantia nigra postmortem brain from the GSE7621 dataset; (<b>B</b>) 8 PD and 9 control subjects from the substantia nigra of postmortem brains from the GSE20163 dataset; (<b>C</b>) 6 PD and 5 control subjects from substantia nigra samples from the GSE20164 dataset; (<b>D</b>) control Braak α-synuclein Stage 0: 8 samples; Braak α-synuclein stages 1–2: 5 samples; Braak α-synuclein stages 3–4: 7 samples; Braak α-synuclein stages 5–6: 8 samples from the GSE49036 dataset; (<b>E</b>) 8 PD and 8 control subjects from peripheral mononuclear blood cells from the GSE22491 dataset; (<b>F</b>) 233 healthy controls and 205 idiopathic PD patients from whole blood from the GSE99039 dataset. (<b>G</b>) The expression of DEDRGs from the GSE49036 dataset at different stages. Gene ID: 200801_x_at, 213867_x_at, 224594_x_at, AFFX-HSAC07/X00351_3_at, AFFX-HSAC07/X00351_5_at, AFFX-HSAC07/X00351_M_at, 200601_at, 218144_s_at, 222534_s_at, 222535_at, 224469_s_at, 212082_s_at, 214002_at.</p>
Full article ">Figure 4
<p>Validity verification of DEDRGs. (<b>A</b>) Validation of DEDRGs by Western blotting. (<b>B</b>) Statistical plots of SLC7A11, ACTB, ACTN4, INF2, and MYL6. Compared with the saline group, ns = no significance, * <span class="html-italic">p &lt;</span> 0.05, ** <span class="html-italic">p</span> &lt; 0.01. <span class="html-italic">n</span> = 3. (<b>C</b>) Location of ACTN4 and INF2 proteins in cells from the HPA database: green represents the target protein, red represents microtubules, yellow represents the endoplasmic reticulum, and blue represents the nucleus (scale bar, 20 µm).</p>
Full article ">Figure 5
<p>Box plot of differential expression of DEDRGs between normal and tumor samples. (<b>A</b>) The differential expression of ACTB in pan-cancer. (<b>B</b>) The differential expression of ACTN4 in pan-cancer. (<b>C</b>) The differential expression of INF2 in pan-cancer. (<b>D</b>) The differential expression of MYL6 in pan-cancer. Compared with the normal 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, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>Pan-cancer prognostic analysis of DEDRGs using univariate Cox regression, including (<b>A</b>) ACTB, (<b>B</b>) ACTN4, (<b>C</b>) INF2, and (<b>D</b>) MYL6.</p>
Full article ">Figure 7
<p>Survival analysis of DEDRG expression in pan-cancer. (<b>A</b>–<b>H</b>) Survival curves of ACTB in GBMLGG, LGG, MESO, KIRC, UVM, HNSC, LIHC, LUAD. (<b>I</b>–<b>N</b>) Survival curves of ACTN4 in GBMLGG, LGG, MESO, PAAD, LUAD, KIRC. (<b>O</b>–<b>R</b>) Survival curves of INF2 in LIHC, HNSC, GBM, LAML. (<b>S</b>–<b>X</b>) Survival curves of MYL6 in GBMLGG, LGG, ACC, UVM, LAML, SARC.</p>
Full article ">Figure 8
<p>Pan-cancer immune infiltration analysis: (<b>A</b>) Immunoinfiltration analysis of ACTB in GBMLGG, LGG, MESO, KIRC, UVM, HNSC, LIHC, LUAD, and GBM. (<b>B</b>) Immunoinfiltration analysis of ACTN4 in GBMLGG, LGG, MESO, PAAD, LUAD, and KIRC. (<b>C</b>) Immunoinfiltration analysis of INF2 in LIHC, HNSC, GBM, and LAML. (<b>D</b>) Immunoinfiltration analysis of MYL6 in GBMLGG, ACC, LGG, UVM, LAML, and SARC. The correlation coefficient being positive indicates a positive correlation between two variables; a negative correlation coefficient indicates a negative correlation between two variables. The absolute value of the correlation coefficient represents the degree of correlation: 0–0.3 indicates weak or no correlation; 0.3–0.5 indicates weak correlation; 0.5–0.8 indicates moderate correlation; 0.8–1 indicates strong correlation. * <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 9
<p>Single-cell type analysis of DEDRGs, including ACTB, ACTN4, INF2, and MYL6, mainly from glandular epithelial cells, squamous epithelial cells, specialized epithelial cells, endocrine cells, neuronal cells, glial cells, germ cells, trophoblast cells, endothelial cells, muscle cells, adipocytes, pigment cells, mesenchymal cells, undifferentiated cells, and blood and immune cells.</p>
Full article ">Figure 10
<p>Gene mutation analysis of DEDRGs. (<b>A</b>) The pan-cancer mutation status of ACTB was determined using the cBioPortal tool. (<b>B</b>) ACTB base mutation frequency. (<b>C</b>) The pan-cancer mutation status of ACTN4 was determined using the cBioPortal tool. (<b>D</b>) ACTN4 base mutation frequency. (<b>E</b>) The pan-cancer mutation status of INF2 was determined using the cBioPortal tool. (<b>F</b>) INF2 base mutation frequency. (<b>G</b>) The pan-cancer mutation status of MYL6 was determined using the cBioPortal tool. (<b>H</b>) MYL6 base mutation frequency.</p>
Full article ">Figure 11
<p>Tumor pathological staining of ACTB in glioma, renal cancer, head and neck cancer, liver cancer, and lung cancer; ACTN4 in glioma, pancreatic cancer, lung cancer, and renal cancer; INF2 in liver cancer, head and neck cancer, and glioma; MYL6 in glioma (scale bar, 20 µm).</p>
Full article ">Figure 12
<p>Coexpression network of DEDRGs and target miRNAs.</p>
Full article ">Figure 13
<p>Binding mode of screened drugs to their targets by molecular docking. (<b>A</b>) The structure of cyclophosphamide. (<b>B</b>) The structure of ethinyl estradiol. (<b>C</b>) The structure of ACTB (3byh). (<b>D</b>) Molecular docking results of ACTB and cyclophosphamide. (<b>E</b>) Molecular docking results of ACTB and ethinyl estradiol.</p>
Full article ">
21 pages, 1728 KiB  
Review
Mammalian Inner Ear-Resident Immune Cells—A Scoping Review
by Betül Karayay, Heidi Olze and Agnieszka J. Szczepek
Cells 2024, 13(18), 1528; https://doi.org/10.3390/cells13181528 - 12 Sep 2024
Viewed by 249
Abstract
Background: Several studies have demonstrated the presence of resident immune cells in the healthy inner ear. Aim: This scoping review aimed to systematize this knowledge by collecting the data on resident immune cells in the inner ear of different species under steady-state conditions. [...] Read more.
Background: Several studies have demonstrated the presence of resident immune cells in the healthy inner ear. Aim: This scoping review aimed to systematize this knowledge by collecting the data on resident immune cells in the inner ear of different species under steady-state conditions. Methods: The databases PubMed, MEDLINE (Ovid), CINAHL (EBSCO), and LIVIVO were used to identify articles. Systematic reviews, experimental studies, and clinical data in English and German were included without time limitations. Results: The search yielded 49 eligible articles published between 1979 and 2022. Resident immune cells, including macrophages, lymphocytes, leukocytes, and mast cells, have been observed in various mammalian inner ear structures under steady-state conditions. However, the physiological function of these cells in the healthy cochlea remains unclear, providing an opportunity for basic research in inner ear biology. Conclusions: This review highlights the need for further investigation into the role of these cells, which is crucial for advancing the development of therapeutic methods for treating inner ear disorders, potentially transforming the field of otolaryngology and immunology. Full article
(This article belongs to the Section Cells of the Nervous System)
Show Figures

Figure 1

Figure 1
<p>Study flow diagram, according to PRISMA extension for scoping reviews [<a href="#B18-cells-13-01528" class="html-bibr">18</a>].</p>
Full article ">Figure 2
<p>Schematic presentation of the main findings of this scoping review. ES, endolymphatic sac; OC, organ of Corti. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 3
<p>The location of macrophages, leukocytes, lymphocytes, and mast cells at a steady state in the human inner ear with emphasis on the cochlea. ST, scala tympani, SM, scala media, SV, scala vestibuli. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
22 pages, 14482 KiB  
Article
Key Disease-Related Genes and Immune Cell Infiltration Landscape in Inflammatory Bowel Disease: A Bioinformatics Investigation
by Kawthar S. Alghamdi, Rahaf H. Kassar, Wesam F. Farrash, Ahmad A. Obaid, Shakir Idris, Alaa Siddig, Afnan M. Shakoori, Sallwa M. Alshehre, Faisal Minshawi and Abdulrahman Mujalli
Int. J. Mol. Sci. 2024, 25(17), 9751; https://doi.org/10.3390/ijms25179751 - 9 Sep 2024
Viewed by 474
Abstract
Inflammatory Bowel Diseases (IBD), which encompass ulcerative colitis (UC) and Crohn’s disease (CD), are characterized by chronic inflammation and tissue damage of the gastrointestinal tract. This study aimed to uncover novel disease-gene signatures, dysregulated pathways, and the immune cell infiltration landscape of inflamed [...] Read more.
Inflammatory Bowel Diseases (IBD), which encompass ulcerative colitis (UC) and Crohn’s disease (CD), are characterized by chronic inflammation and tissue damage of the gastrointestinal tract. This study aimed to uncover novel disease-gene signatures, dysregulated pathways, and the immune cell infiltration landscape of inflamed tissues. Eight publicly available transcriptomic datasets, including inflamed and non-inflamed tissues from CD and UC patients were analyzed. Common differentially expressed genes (DEGs) were identified through meta-analysis, revealing 180 DEGs. DEGs were implicated in leukocyte transendothelial migration, PI3K-Akt, chemokine, NOD-like receptors, TNF signaling pathways, and pathways in cancer. Protein–protein interaction network and cluster analysis identified 14 central IBD players, which were validated using eight external datasets. Disease module construction using the NeDRex platform identified nine out of 14 disease-associated genes (CYBB, RAC2, GNAI2, ITGA4, CYBA, NCF4, CPT1A, NCF2, and PCK1). Immune infiltration profile assessment revealed a significantly higher degree of infiltration of neutrophils, activated dendritic cells, plasma cells, mast cells (resting/activated), B cells (memory/naïve), regulatory T cells, and M0 and M1 macrophages in inflamed IBD tissue. Collectively, this study identified the immune infiltration profile and nine disease-associated genes as potential modulators of IBD pathogenesis, offering insights into disease molecular mechanisms, and highlighting potential disease modulators and immune cell dynamics. Full article
(This article belongs to the Special Issue Immunoanalytical and Bioinformatics Methods in Immunology Research)
Show Figures

Figure 1

Figure 1
<p>Differential expression analysis of Crohn’s disease (CD). (<b>a</b>) Volcano plots across four CD datasets. The colored dots represent the significant DEGs identified at |log2FC| ≥ 1 and an adjusted <span class="html-italic">p</span>-value of ≤0.05. The top 10 genes within each dataset are shown. (<b>b</b>) The upset plots depict DEG distribution across CD datasets. (<b>c</b>) Heatmap of top 10 DEGs identified by meta-analysis. The expression heatmap depicts expression levels of significantly different upregulated and downregulated genes. The color indicates high expression (red) and low expression (green).</p>
Full article ">Figure 2
<p>Differential expression analysis of ulcerative colitis (UC). (<b>a</b>) Volcano plots across four UC datasets. The colored dots represent the significant DEGs identified at |log2FC| ≥ 1 and an adjusted <span class="html-italic">p</span>-value of ≤0.05. The top 10 genes within each dataset are shown. (<b>b</b>) The upset plots depict DEG distribution across UC datasets. (<b>c</b>) Heatmap of top 10 DEGs identified by meta-analysis. The expression heatmap depicts expression levels of significantly different upregulated and downregulated genes. The color indicates high expression (red) and low expression (green).</p>
Full article ">Figure 3
<p>Enrichment analysis of DEGs. Bubble plot of top 20 gene ontology (GO) and KEGG signaling pathways for (<b>a</b>) CD and (<b>b</b>) UC. The bubble color scaled the enrichment score. The size of the bubbles represents the level of DEG enrichment within each pathway. (<b>c</b>) The Sankey plot represents both shared and distinct pathways between CD and UC.</p>
Full article ">Figure 4
<p>Characterization of IBD-DEGs. (<b>a</b>) Venn diagram illustrates shared DEGs between CD and UC, identifying 180 IBD-DEGs with 106 upregulated and 74 downregulated. (<b>b</b>) Tissue specificity enrichment analysis of IBD-DEGs shows predominant enrichment in tissues such as the small intestine, appendix, colon, duodenum, and rectum. (<b>c</b>) PPI network of IBD-DEGs, displaying top genes with their centrality parameters obtained from network analysis. Node size corresponds to the degree of connectivity. (<b>d</b>) Hub genes identified from the PPI network were ranked based on their degree of connectivity.</p>
Full article ">Figure 5
<p>Potential key IB-DEGs. (<b>a</b>) PPI network with clusters identified using MCODE and BiCoN clustering methods in Cytoscape. Nodes with lavender color indicate the genes in the clusters and the cluster regions in the PPI network (<b>b</b>) 14 key IBD-DEGs identified through the intersection of clusters identified by both methods. (<b>c</b>) Heatmap showing the validation of the candidate IBD-DEGs across eight external validation datasets (GSE117993, GSE4183, GSE13367, GSE16879, GSE36807, GSE38713, GSE6731, and GSE59071). Consistent expression patterns of the identified genes were observed across these datasets. Red signifies upregulation, and blue signifies downregulation of expression levels.</p>
Full article ">Figure 6
<p>Disease modules of key IBD-DEGs and mechanistic pathways. (<b>a</b>) An illustration of IBD-related genes recovered through the Get Disease Genes function in NeDRex platform. (<b>b</b>) Disease module obtained from IBD-related genes and potential key IBD genes using DIAMOnD algorithm. Nine out of 14 key IBD-DEGs are present in the subnetwork highlighted in yellow. (<b>c</b>) Enriched pathways for key IBD-DEGs.</p>
Full article ">Figure 7
<p>Immune cell infiltration fraction between inflamed and non-inflamed IBD tissues. Violin plots of the proportion of 22 immune cells in inflamed (<span class="html-italic">n</span> = 397, red) vs. non-inflamed (<span class="html-italic">n</span> = 370, blue) IBD tissues. The red boxplot represents inflamed, and the blue boxplot represents non-inflamed tissues. Significance levels are indicated as follows: ns = non-significant, * <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.</p>
Full article ">Figure 8
<p>Workflow used in this study.</p>
Full article ">
15 pages, 3057 KiB  
Article
Changes in Anxiety-Related Behaviors, Voiding Patterns, and Urinary Bladder Contractile Properties in Male Mice Exposed to Water Avoidance Stress for 1 Day and 28 Days
by Sarunnuch Sattayachiti, Panida Chumpong, Seree Niyomdecha, Dania Cheaha and Nipaporn Konthapakdee
Biology 2024, 13(9), 707; https://doi.org/10.3390/biology13090707 - 9 Sep 2024
Viewed by 359
Abstract
Repeated water avoidance stress (WAS) for 10 days is a common rodent model to mimic the effect of chronic psychological stress on urinary bladder dysfunction. However, it remains obscure whether changes in the stress exposure period impact urinary bladder impairment differently. Therefore, this [...] Read more.
Repeated water avoidance stress (WAS) for 10 days is a common rodent model to mimic the effect of chronic psychological stress on urinary bladder dysfunction. However, it remains obscure whether changes in the stress exposure period impact urinary bladder impairment differently. Therefore, this study aimed to investigate the effect of 1 (acute), 10 (chronic), and 28 (prolonged) days of WAS on anxiety-related behavior, voiding pattern, urinary bladder mast cells, and bladder contractility in C57BL/6J male mice. Mice exposed to 1 and 10 days of WAS showed decreased unsupported rearing. A decreased total void area after 1 and 10 days of the WAS was observed, which was reversed in the 28-day-WAS group. There was an increased number of degranulated mast cells in the bladder of the 10-day-WAS group. The 1-day WAS exposure enhanced tonic contractile response to a muscarinic agonist, carbachol, which was reversed by 5-HT3 receptor antagonist pre-incubation. Interestingly, the 28-day WAS group showed a similar tonic contractile response to the control group. Our findings provide more insightful information about using 1-day WAS as an acute psychological stress model, and stress exposure longer than 10 days did not produce anxiety-like behavior and urinary bladder impairment. Full article
(This article belongs to the Section Physiology)
Show Figures

Figure 1

Figure 1
<p>A scheme demonstrating the experimental timeline of this study.</p>
Full article ">Figure 2
<p>Changes in anxiety-related behavior of control and water avoidance stress (WAS) on days 1, 10, and 28. The total duration (<b>A</b>) and the total number (<b>B</b>) of unsupported rearing. The total duration (<b>C</b>) and the total number (<b>D</b>) of supported rearing. The total duration of grooming behavior (<b>E</b>). (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, Unpaired <span class="html-italic">t</span>-test, ns = not significant, N = 4–7 in each group.)</p>
Full article ">Figure 2 Cont.
<p>Changes in anxiety-related behavior of control and water avoidance stress (WAS) on days 1, 10, and 28. The total duration (<b>A</b>) and the total number (<b>B</b>) of unsupported rearing. The total duration (<b>C</b>) and the total number (<b>D</b>) of supported rearing. The total duration of grooming behavior (<b>E</b>). (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, Unpaired <span class="html-italic">t</span>-test, ns = not significant, N = 4–7 in each group.)</p>
Full article ">Figure 3
<p>Representative images of urine patterns on filter papers on days 1, 10, and 28 of control (<b>A</b>,<b>C</b>,<b>E</b>) and water avoidance stress (WAS) (<b>B</b>,<b>D</b>,<b>F</b>) groups.</p>
Full article ">Figure 4
<p>Changes in the number of urine spots (<b>A</b>,<b>B</b>) and total voided area (<b>C</b>,<b>D</b>) from voiding spot analysis of the control and water avoidance stress (WAS) groups on days 1, 10, and 28. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, Unpaired <span class="html-italic">t</span>-test, ns = not significant, N = 8–10 in each group). Pie graphs representing the proportion of large urine spot (L) (size ≥ 6.00 cm<sup>2</sup>) and small spot size (S) (size 0.4–5.99 cm<sup>2</sup>) from the voiding spot analysis of the control and water avoidance stress (WAS) groups on days 1, 10, and 28 (<b>E</b>) (N = 8–10 in each group).</p>
Full article ">Figure 5
<p>Representative images of degranulated mast cells (<b>A</b>) in the urinary bladder as indicated with the arrows. Bar graphs representing the total number of mast cells (<b>B</b>) and the number of degranulated mast cells (<b>C</b>). The stacked bar graph represents percentages of degranulated and non- degranulated mast cells in the bladder tissues of the control and water avoidance stress (WAS) groups on days 1, 10, and 28 (<b>D</b>). Scale bar = 10 µm, 40× magnification, U = Urothelium (* <span class="html-italic">p</span> &lt; 0.05, Unpaired <span class="html-italic">t</span>-test, ns = not significant, N = 3 in each group).</p>
Full article ">Figure 6
<p>Tonic contraction of bladder strips from the control (<b>A</b>) and 1-day WAS groups in response to 0.1, 0.3, 1.0, 3.0, 10, and 30 µM carbachol with (<b>C</b>) or without ondansetron pre-incubation (<b>B</b>) (* <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, One-way ANOVA with Dunnett’s multiple comparison vs. baseline, N = 6–8).</p>
Full article ">Figure 7
<p>Tonic contraction of bladder strips from the control (<b>A</b>) and 28-day WAS groups in response to 0.1, 0.3, 1.0, 3.0, 10, and 30 µM carbachol with (<b>C</b>) or without ondansetron pre-incubation (<b>B</b>) (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, One-way ANOVA with Dunnett’s multiple comparisons vs. baseline, N = 6–8).</p>
Full article ">
13 pages, 10036 KiB  
Article
Antibacterial and Antiallergic Effects of Three Tea Extracts on Histamine-Induced Dermatitis
by Zeting Huang, Lanyue Zhang, Jie Xuan, Tiantian Zhao and Weihua Peng
Pharmaceuticals 2024, 17(9), 1181; https://doi.org/10.3390/ph17091181 - 7 Sep 2024
Viewed by 510
Abstract
Atopic dermatitis (AD) is a persistent and recurrent inflammatory skin condition with a genetic basis. However, the fundamental reasons and mechanisms behind this phenomenon remain incompletely understood. While tea extracts are known to reduce histamine-induced skin allergies and inflammation, the specific mechanisms by [...] Read more.
Atopic dermatitis (AD) is a persistent and recurrent inflammatory skin condition with a genetic basis. However, the fundamental reasons and mechanisms behind this phenomenon remain incompletely understood. While tea extracts are known to reduce histamine-induced skin allergies and inflammation, the specific mechanisms by which various types of Chinese tea provide their protective effects are still not fully elucidated. In this study, a model of skin itching induced by histamine is used to explore the functions and mechanisms of three types of tea extract (Keemun black tea (HC), Hangzhou green tea (LC), and Fujian white tea (BC)) in alleviating histamine-induced dermatitis. The components of three tea extracts are identified by UPLC-Q-TOF-MS, and we found that their main components are alkaloids, fatty acyls, flavonoids, organic acids, and phenols. The inhibitory effects of three types of tea extract on Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) in skin injury are investigated by MIC and flow cytometry. The three types of tea extract have an inhibitory effect on the growth of bacterial flora, with HC showing the best inhibitory activity. The effect of the three types of tea extract on histamine-induced dermatitis is also evaluated. Furthermore, itchy skin experiments, HE staining, toluidine blue staining, and immunohistochemical staining of mouse skin tissues were performed to determine the variations of scratching, epidermal thickness, mast cell number, IL-1β, and NGF content after the administration of the tea extracts. The three types of tea extracts all alleviate and inhibit skin itching, epidermal hyperplasia, and allergic dermatitis. BC effectively alleviates epidermal hyperplasia caused by skin allergies, and LC significantly downregulates NGF. HC reduces histamine-induced mast cell infiltration and downregulates IL-1β to alleviate skin itching. Consequently, tea emerges a potent natural product that can inhibit the growth of skin wound bacterial flora and exhibit skin repair effects on histamine-induced allergic dermatitis. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

Figure 1
<p>LC extract chromatograms in positive (<b>A</b>) and negative (<b>B</b>) modes. HC extract chromatograms in positive (<b>C</b>) and negative (<b>D</b>) modes. BC extract chromatograms in positive (<b>E</b>) and negative (<b>F</b>) modes.</p>
Full article ">Figure 2
<p>Chemical structures of main constituents of the three tea extracts.</p>
Full article ">Figure 3
<p>Inhibitory effect of three tea extracts on <span class="html-italic">E. coli</span> and <span class="html-italic">S. aureus</span>. MICs of three tea extracts on (<b>A</b>) <span class="html-italic">E. coli</span> and (<b>B</b>) <span class="html-italic">S. aureus</span>, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Antibacterial effects of three tea extracts on <span class="html-italic">S. aureus.</span> (<b>A</b>) Results of flow pattern of <span class="html-italic">S. aureus</span>. (<b>B</b>) Histogram results of <span class="html-italic">S. aureus</span> (areas Q1, Q2, Q3, and Q4 in this Figure are dead, damaged, alive, and unstained bacterial areas, respectively).</p>
Full article ">Figure 5
<p>Number of scratches. (<b>A</b>) Pictures of mice backs. (<b>B</b>) Number of scratches (<span class="html-italic">n</span> = 10, compared with model group, ** <span class="html-italic">p</span> &lt; 0.01). The red arrow represents the scratch area of the mouse.</p>
Full article ">Figure 6
<p>HE staining results, (<b>A</b>) pictures of staining for mice, and (<b>B</b>) epidermal thickness (<span class="html-italic">n</span> = 10). The red arrow represents the epidermis of mouse skin.</p>
Full article ">Figure 7
<p>Toluidine blue staining results, (<b>A</b>) pictures of staining for mice, and (<b>B</b>) mast cell number (<span class="html-italic">n</span> = 10, compared with model group, ** <span class="html-italic">p</span> &lt; 0.01). The red arrow indicates mast cells in the skin of mice.</p>
Full article ">Figure 8
<p>(<b>A</b>) Immunohistochemical test results for IL-1β. (<b>B</b>) Integrated optical density of IL-1β (<span class="html-italic">n</span> = 10, compared with model group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01). The red arrow indicates the IL-1β cytokine in the mouse skin.</p>
Full article ">Figure 9
<p>(<b>A</b>) Immunohistochemical test results for NGF. (<b>B</b>) Integrated optical density of NGF (<span class="html-italic">n</span> = 10, compared with model group, ** <span class="html-italic">p</span> &lt; 0.01). The red arrow indicates the NGF cytokine in the mouse skin.</p>
Full article ">
13 pages, 3110 KiB  
Article
Preventive and Therapeutic Effects of Lactiplantibacillus plantarum HD02 and MD159 through Mast Cell Degranulation Inhibition in Mouse Models of Atopic Dermatitis
by A-Ram Kim, Seong-Gak Jeon, Hyung-Ran Kim, Heeji Hong, Yong Won Yoon, Byung-Min Lee, Chung Hoo Yoon, Soo Jin Choi, Myoung Ho Jang and Bo-Gie Yang
Nutrients 2024, 16(17), 3021; https://doi.org/10.3390/nu16173021 - 6 Sep 2024
Viewed by 586
Abstract
As the relationship between the gut microbiome and allergies becomes better understood, targeted strategies to prevent and treat allergies through gut microbiome modulation are being increasingly developed. In the study presented herein, we screened various probiotics for their ability to inhibit mast cell [...] Read more.
As the relationship between the gut microbiome and allergies becomes better understood, targeted strategies to prevent and treat allergies through gut microbiome modulation are being increasingly developed. In the study presented herein, we screened various probiotics for their ability to inhibit mast cell degranulation and identified Lactiplatibacillus plantarum HD02 and MD159 as effective candidates. The two strains significantly attenuated vascular permeability induced by mast cell degranulation in a passive cutaneous anaphylaxis (PCA) model and, in the MC903-induced murine atopic dermatitis (AD) model, demonstrated comparable preventive effects against allergies, reducing blood levels of MCPT-1 (mast cell protease-1) and total IgE. In the house dust mite (HDM)-induced murine AD model, both L. plantarum HD02 and MD159 showed therapeutic effects, with L. plantarum HD02 demonstrating superior efficacy. Nevertheless, L. plantarum MD159 better suppressed transepidermal water loss (TEWL). Furthermore, L. plantarum HD02 and MD159 significantly increased the number of splenic Foxp3+ regulatory T cells, with L. plantarum MD159 having a more pronounced effect. However, only L. plantarum HD02 achieved a reduction in immune cells in the draining lymph nodes. Our findings highlight L. plantarum HD02 and MD159 as promising candidates for the prevention and treatment of allergies, demonstrating significant efficacy in suppressing mast cell degranulation, reducing the number of allergy biomarkers, and modulating immune responses in experimental models of AD. Their distinct mechanisms of action suggest potential complementary roles in addressing allergic diseases, underscoring their therapeutic promise in clinical applications. Full article
(This article belongs to the Section Prebiotics and Probiotics)
Show Figures

Figure 1

Figure 1
<p>Screening of probiotic strains for the inhibition of mast cell degranulation via the β-hexosaminidase assay. Various probiotic strains were tested to identify those capable of inhibiting IgE-induced mast cell degranulation using a mast cell line RBL-2H3 and β-hexosaminidase assay. Each probiotic strain was assessed for inhibitory effects in the form of (<b>A</b>) the bacteria and (<b>B</b>) their culture supernatants. Statistical analysis was performed only on probiotic strains that inhibited mast cell degranulation by 40% or less in (<b>A</b>). Data are represented as the mean ± SD. **, <span class="html-italic">p</span> &lt; 0.01 and ***, <span class="html-italic">p</span> &lt; 0.001 versus the MRS group.</p>
Full article ">Figure 2
<p>Inhibition of IgE-induced mast cell degranulation by <span class="html-italic">L. plantarum</span> HD02 and MD159 in a PCA mouse model. To investigate the ability of probiotic strains to inhibit mast cell degranulation in vivo, a murine model of PCA was created according to the experimental scheme displayed in (<b>A</b>). Degranulation of mast cells was measured by the extent to which Evans blue, administered through the tail vein along with the antigen DNP-HSA, leaked into the ear skin. Representative photographs are shown in (<b>B</b>), and the amount of Evans blue extracted from both ears is presented in dot plots in (<b>C</b>). i.d., intradermal; i.v., intravenous; p.o., oral; Ag, antigen; QD, once a day; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Prevention of allergic response by <span class="html-italic">L. plantarum</span> HD02 and MD159 in an MC-903-induced AD mouse model. To assess the preventive effects of <span class="html-italic">L. plantarum</span> HD02 and MD159, the mice were induced with AD based on the experimental scheme presented in (<b>A</b>). Representative images of the ear skin are shown in (<b>B</b>), and changes in ear thickness are shown graphically in (<b>C</b>). Blood levels of total IgE (<b>D</b>) and MCPT-1 (<b>E</b>) are also shown. Data are presented as the mean ± SEM. QD, once a day; p.o., oral; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Therapeutic effects of <span class="html-italic">L. plantarum</span> HD02 and MD159 in an HDM-induced AD mouse model. The mice were induced with AD using HDM and treated with HD02 or MD159 according to the experimental scheme shown in (<b>A</b>). Representative images of dorsal skin are shown in (<b>B</b>), and changes in dermatitis scores are presented graphically in (<b>C</b>). The number of scratches (<b>D</b>), total IgE levels in the blood (<b>E</b>), and TEWL on the dorsal skin (<b>F</b>) were measured. Data are presented as the mean ± SEM. BIW, twice weekly; QW, once weekly; QD, once a day; p.o., oral; * <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>Inhibition of epidermal layer thickness and mast cell infiltration by <span class="html-italic">L. plantarum</span> HD02 and MD159 in an HDM-induce AD mouse model. In a mouse model of HDM-induced AD, skin legions were analyzed. Paraffin sections were stained with H&amp;E to measure the thickness of the epidermal layer (<b>A</b>,<b>B</b>) and toluidine blue to observe mast cell infiltration (<b>C</b>,<b>D</b>). The graphs display the thickness of the epidermal layer (<b>B</b>) and the number of infiltrated mast cells (<b>D</b>). Data are presented as the mean ± SEM *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Regulation of immune responses by <span class="html-italic">L. plantarum</span> HD02 and MD159 in an HDM-induced AD mouse model. The involvement of <span class="html-italic">L. plantarum</span> HD02 and MD159 in regulating immune responses was investigated in a mouse model of HDM-induced AD by analyzing the draining lymph node, ALN, and spleen. The expressions of <span class="html-italic">il-4</span> and <span class="html-italic">ifng</span> in the ALN were examined using qRT-PCR (<b>A</b>) The numbers of total cells, T cells, and B cells in the ALN were counted (<b>B</b>). In the spleen, the ratio of Foxp3+ Treg cells among total T cells was measured via flow cytometric analysis (<b>C</b>). Data are presented as mean ± SEM. Dexa, dexamethasone; * <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 ">
7 pages, 189 KiB  
Article
Anaphylactic Shock Caused by Eating Buckwheat
by Dan Orga-Dumitriu, Dana M. Harris and Corina Porr
J. Clin. Med. 2024, 13(17), 5243; https://doi.org/10.3390/jcm13175243 - 4 Sep 2024
Viewed by 300
Abstract
Background: Urticaria is a common disease with a marked influence on quality of life. The key cell involved is the mast cell, which can be activated by a vast variety of stimuli, and the major mediator is histamine. Allergic urticaria is a [...] Read more.
Background: Urticaria is a common disease with a marked influence on quality of life. The key cell involved is the mast cell, which can be activated by a vast variety of stimuli, and the major mediator is histamine. Allergic urticaria is a disorder with a large variety of causes: food, drugs, insect venom, skin contact with allergens, and physical exercise. Buckwheat consumption has increased in European countries and the USA because it is gluten-free. It can trigger anaphylactic shock if ingested, inhaled, or handled with the hands. Five common buckwheat allergens named Fag e1 to 5 (Fag e1, 2, and 3 are considered the major allergens) and two tartary buckwheat allergens named Fag t1 and Fag t2 have been described. Method: We present the case of a patient who experienced two anaphylactic shocks and in whom the etiological factor was buckwheat. The patient presented to the Allergology department for the evaluation of two episodes of severe allergic reactions that required emergency therapy, episodes that involved the loss of consciousness and were of major severity. At each anaphylactic shock, an ambulance was requested, and emergency therapy was administered, leading to the patient’s recovery within a few hours. Diagnosis: Since each episode occurred a few minutes after eating, the diagnosis was established based on a detailed anamnesis and prick skin tests, followed by specific IgE dosages. Other foods consumed by the patient, assessed by prick skin testing and specific IgE dosages of suspected foods, were excluded as the etiological cause. Increased levels of buckwheat-specific immunoglobulin E were highlighted, thus identifying the etiological agent. The treatment of anaphylactic shock was performed urgently by the ambulance crew with adrenaline, infusion solutions, cortisone preparations, and antihistamines. Result: Following the treatment that was initiated, there was a partial remission of the lesions after a few hours. Conclusions: Buckwheat allergy is rare, but it produces symptoms that affect the skin, gastrointestinal tract, and respiratory tract, as well as anaphylaxis. In a professional environment, it can trigger allergic rhinitis, asthma, and hives. Although buckwheat allergens have been described, their clinical relevance has only been studied in a small number cases. In current practice, the only commercially available allergen is Beech e2 per the ImmunoCAP ISAC microarray. Diagnosis can be difficult in clinical practice. This reported case suggests the need for a thorough anamnesis, since buckwheat is consumed as a hidden allergen, and in Europe, it is not necessary to label foods containing this allergen. Full article
16 pages, 1982 KiB  
Article
Peripheral Inflammation Featuring Eosinophilia or Neutrophilia Is Associated with the Survival and Infiltration of Eosinophils within the Tumor among Various Histological Subgroups of Patients with NSCLC
by Bilal Alashkar Alhamwe, Kadriya Yuskaeva, Friederike Wulf, Frederik Trinkmann, Mark Kriegsmann, Michael Thomas, Corinna Ulrike Keber, Elke Pogge von Strandmann, Felix J. Herth, Saeed Kolahian, Harald Renz and Thomas Muley
Int. J. Mol. Sci. 2024, 25(17), 9552; https://doi.org/10.3390/ijms25179552 - 3 Sep 2024
Viewed by 698
Abstract
Immune activation status determines non-small cell lung cancer (NSCLC) prognosis, with reported positive/negative associations for T helper type 2 (TH2) responses, including allergen-specific IgE and eosinophils. Our study seeks to explore the potential impact of these comorbid immune responses on the survival rates [...] Read more.
Immune activation status determines non-small cell lung cancer (NSCLC) prognosis, with reported positive/negative associations for T helper type 2 (TH2) responses, including allergen-specific IgE and eosinophils. Our study seeks to explore the potential impact of these comorbid immune responses on the survival rates of patients with NSCLC. Our retrospective study used data from the Data Warehouse of the German Center for Lung Research (DZL) and Lung Biobank at Thoraxklinik Heidelberg. We estimated the association of blood eosinophilia and neutrophilia on survival rates in an inflammatory cohort of 3143 patients with NSCLC. We also tested sensitization to food and inhalants and high-sensitivity C-reactive protein (hs-CRP) in a comorbidity cohort of 212 patients with NSCLC. Finally, we estimated the infiltration of immune-relevant cells including eosinophils, T-cells, and mast cells in a tissue inflammatory sub-cohort of 60 patients with NSCLC. Sensitization to at least one food or inhalant (sIgE) was higher in patients with adenocarcinoma (adeno-LC) than the non-adenocarcinoma (non-adeno-LC). Furthermore, hs-CRP was higher in non-adeno-LC compared with adeno-LC. Peripheral inflammation, particularly eosinophilia and neutrophilia, was associated with poor survival outcomes in NSCLC with a clear difference between histological subgroups. Finally, blood eosinophilia was paralleled by significant eosinophil infiltration into the peritumoral tissue in the lung. This study provides novel perspectives on the crucial role of peripheral inflammation, featuring eosinophilia and neutrophilia, with overall survival, underscoring distinctions between NSCLC subgroups (adeno-LC vs. non-adeno-LC). Peripheral eosinophilia enhances eosinophil infiltration into tumors. This sheds light on the complex interplay between inflammation, eosinophil infiltration, and NSCLC prognosis among various histological subtypes. Further studies are required to underscore the role of eosinophils in NSCLC among different histological subgroups and their role in shaping the tumor microenvironment. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Allergy and Asthma: 3rd Edition)
Show Figures

Figure 1

Figure 1
<p><b>Chronic inflammatory biomarkers in adeno-LC vs. non-adeno-LC</b>. (<b>A</b>) Patients sensitized to at least one inhalant or food allergen (sIgE &gt; 0.35 kU/L). Specific IgE was measured for a representative panel of inhalant allergens (n = 8) and food allergens (n = 6) in the sera of NSCLC patients (n = 212), of which n = 115 had adeno-LC and n = 97 non-adeno-LC. Significant differences were tested using the Mann–Whitney U-test. (<b>B</b>) Distribution of hs-CRP concentrations in adeno-LC and non-adeno-LC. Significant differences were tested using the Mann–Whitney U-test. (<b>C</b>) Distribution of hs-CRP concentrations considering the histology (adeno-LC vs. non-adeno-LC) and lung comorbidities (asthma, COPD, and neither). Both stars and cycles describe outliers. Among these outliers, stars specifically mark extreme values. The numbers of patients in each group are shown. Significant differences were tested using the Kruskal–Wallis test.</p>
Full article ">Figure 2
<p><b>Impact of peripheral inflammation on cumulative survival.</b> (<b>A</b>–<b>C</b>) Cumulative survival in patients with NSCLC with respect to peripheral blood eosinophil and neutrophil counts. Blue line: blood eosinophil ≤ 500 cells/µL and blood neutrophil ≤ 7700 cells/µL. Green line: neutrophilia (blood neutrophil &gt; 7700 cells/µL). Red line: eosinophilia (blood eosinophil &gt; 500 cells/µL). Purple line: both eosinophilia and neutrophilia (blood eosinophil &gt; 500 cells/µL and blood neutrophil &gt; 7700 cells/µL). Cumulative survival was tested for all patients with NSCLC (<b>A</b>); those with adeno-LC (<b>B</b>); and those with non-adeno-LC (<b>C</b>). Univariate results of pairwise comparison (log-rank test) were listed in the figure. For results of multivariate regression analyses adjusting for age, gender, stage, BMI, and smoking status, refer to <a href="#ijms-25-09552-t002" class="html-table">Table 2</a> and <a href="#ijms-25-09552-t003" class="html-table">Table 3</a>.</p>
Full article ">Figure 3
<p><b>Hazard ratios using Cox regression analysis model.</b> (<b>A</b>,<b>B</b>) The hazard ratio and overall survival were calculated according to subgroups (adeno-LC and vs. non-adeno-LC) controlling for tumor stage (<b>A</b>) and blood eosinophilia and neutrophilia (<b>B</b>). Hazard ratios were derived from a Cox regression analysis model.</p>
Full article ">Figure 4
<p><b>Distribution of eosinophils in lung cancer tissue</b>. (<b>A</b>,<b>B</b>) The distribution of infiltrated eosinophils in adeno-LC vs. non-adeno-LC, regardless of eosinophilia or neutrophilia status, in peritumoral (<b>A</b>) and intratumoral (<b>B</b>) regions. (<b>C</b>,<b>D</b>) The infiltration of eosinophils between adeno-LC and non-adeno-LC with respect to peripheral eosinophilia (blood eosinophils &gt; 500 or ≤500 cells/µL), neutrophilia (blood neutrophils &gt; 7700 or ≤7700 cells/µL), and CRP concentration (&gt;5 or ≤5 mg/L). Giemsa-stained eosinophils were counted per square millimeter in the peritumoral and intratumoral regions of the lung cancer tissue as described in the Materials and Methods. Mean ± SEM values are shown. Significant differences between and within the groups were tested using the Mann–Whitney U-test. Only significant differences are shown: <span class="html-italic">* p</span> &lt; 0.05; <span class="html-italic">*** p</span> &lt; 0.001. The colored dots in (<b>C</b>,<b>D</b>) represent the number of patients and the representative tumor stage.</p>
Full article ">
12 pages, 3025 KiB  
Article
The Skin Histopathology of Pro- and Parabiotics in a Mouse Model of Atopic Dermatitis
by Hun Hwan Kim, Se Hyo Jeong, Min Yeong Park, Pritam Bhagwan Bhosale, Abuyaseer Abusaliya, Jeong Doo Heo, Hyun Wook Kim, Je Kyung Seong, Tae Yang Kim, Jeong Woo Park, Byeong Soo Kim and Gon Sup Kim
Nutrients 2024, 16(17), 2903; https://doi.org/10.3390/nu16172903 - 30 Aug 2024
Viewed by 560
Abstract
As it has been revealed that the activation of human immune cells through the activity of intestinal microorganisms such as pro- and prebiotics plays a vital role, controlling the proliferation of beneficial bacteria and suppressing harmful bacteria in the intestine has become essential. [...] Read more.
As it has been revealed that the activation of human immune cells through the activity of intestinal microorganisms such as pro- and prebiotics plays a vital role, controlling the proliferation of beneficial bacteria and suppressing harmful bacteria in the intestine has become essential. The importance of probiotics, especially for skin health and the immune system, has led to the emergence of products in various forms, including probiotics, prebiotics, and parabiotics. In particular, atopic dermatitis (AD) produces hypersensitive immunosuppressive substances by promoting the differentiation and activity of immune regulatory T cells. As a result, it has been in the Th1 and Th2 immune balance through a mechanism that suppresses skin inflammation or allergic immune responses caused by bacteria. Furthermore, an immune mechanism has recently emerged that simultaneously controls the expression of IL-17 produced by Th17. Therefore, the anti-atopic effect was investigated by administering doses of anti-atopic candidate substances (Lactobacilus sakei CVL-001, Lactobacilus casei MCL, and Lactobacilus sakei CVL-001 Lactobacilus casei MCL mixed at a ratio of 4:3) in an atopy model using 2,4-dinitrochlorobenzene and observing symptom changes for 2 weeks to confirm the effect of pro-, para-, and mixed biotics on AD. First, the body weight and feed intake of the experimental animals were investigated, and total IgG and IgM were confirmed through blood biochemical tests. Afterward, histopathological staining was performed using H&E staining, Toluidine blue staining, Filaggrin staining, and CD8 antibody staining. In the treatment group, the hyperproliferation of the epidermal layer, the inflammatory cell infiltration of the dermal layer, the expression of CD8, the expression of filaggrin, and the secretion of mast cells were confirmed to be significantly reduced. Lastly, small intestine villi were observed through a scanning microscope, and scoring evaluation was performed through skin damage. Through these results, it was confirmed that AD was reduced when treated with pro-, para-, and mixed biotics containing probiotics and parabiotics. Full article
(This article belongs to the Special Issue The Role of Probiotics and Prebiotics in Immunomodulation)
Show Figures

Figure 1

Figure 1
<p>Histologic features of DNCB-induced AD-like skin damage were evaluated using H&amp;E staining. (<b>A</b>) H&amp;E staining of experimental animal skin for anti-atopic effects of pro-, para-, and mixed biotics (H&amp;E × 200). (<b>B</b>) Histopathologic score in AD-like skin lesions. H&amp;E staining showed that pro-, para-, and mixed biotics all alleviated DNCB-induced inflammation in the epidermal layer, with a histologically significant reduction in the T3 group. The red line in the figure is the relative width of the tissue stain. The data represent the mean ± SD of three independent experiments. (## <span class="html-italic">p</span> &lt; 0.01 vs. VC, * <span class="html-italic">p</span> &lt; 0.05 vs. NC, ** <span class="html-italic">p</span> &lt; 0.01 vs. NC).</p>
Full article ">Figure 2
<p>Histologic features of CD8 antibody staining and Filaggrin staining of AD-like skin lesions with DNCB. (<b>A</b>) CD8 antibody staining of experimental animal skin for anti-atopic effects of pro-, para-, and mixed biotics. Increased expression of CD8 in the epidermal and dermal layers was observed in the DNCB-induced NC, but relatively decreased CD8 expression was observed in the T1, T2, and T3 groups. Similarly, (<b>B</b>) Filaggrin staining in AD showed an increase in the thickness of the epithelial layer in the DNCB-induced NC, but a decrease in T1, T2, and T3. The data represent the mean ± SD of three independent experiments. (# <span class="html-italic">p</span> &lt; 0.05 vs. VC, ## <span class="html-italic">p</span> &lt; 0.01 vs. VC, * <span class="html-italic">p</span> &lt; 0.05 vs. NC, ** <span class="html-italic">p</span> &lt; 0.01 vs. NC, *** <span class="html-italic">p</span> &lt; 0.001 vs. NC).</p>
Full article ">Figure 3
<p>Toluidine blue staining histologic features of AD-like skin lesions with DNCB. (<b>A</b>) Toluidine blue staining of experimental animal skin for anti-atopic effects of pro-, para-, and mixed biotics. (<b>B</b>) Mast cell count in AD-like skin lesions. Mast cells increased dramatically in the DNCB-induced group, but decreased in the T1, T2, and T3 groups, with a significant decrease in the T3 group. The red arrow is the estimated number of mast cells. Data represent the mean ± SD of three independent experiments. (## <span class="html-italic">p</span> &lt; 0.01 vs. VC, * <span class="html-italic">p</span> &lt; 0.05 vs. NC, ** <span class="html-italic">p</span> &lt; 0.01 vs. NC).</p>
Full article ">Figure 4
<p>(<b>A</b>) Visual assessment and average atopic score for each symptom between days 1–15 of atopy-induced skin damage with DNCB and scratching behavior scoring results. (<b>B</b>) Enlarged the condition of atopic dermatitis in each group. (<b>C</b>) Each item is scored as no symptoms (0), mild (1), moderate (2), or severe (3). Means with different superscripts in the same row are significantly different at <span class="html-italic">p</span> &lt; 0.05 via Duncan’s multiple range tests. The data represent the mean ± SD of three independent experiments. (# <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 vs. VC).</p>
Full article ">
11 pages, 1505 KiB  
Article
Evaluation of a Novel Detection Method for Allergen-Specific IgE Antibodies with IgE Receptor Crosslinking Using Rat Food Allergy Model
by Soichiro Ishii, Yuki Koga, Tomoharu Yokooji, Misaki Kakino, Ryohei Ogino, Takanori Taogoshi and Hiroaki Matsuo
Foods 2024, 13(17), 2713; https://doi.org/10.3390/foods13172713 - 27 Aug 2024
Viewed by 426
Abstract
The specific detection of serum IgE antibodies specific to allergens (sIgE Abs) that can crosslink the plural high-affinity IgE receptor (FcεRIα) molecules on the surface of mast cells or basophils with a multivalent allergen can reduce the false-positive diagnoses observed in chemiluminescent and [...] Read more.
The specific detection of serum IgE antibodies specific to allergens (sIgE Abs) that can crosslink the plural high-affinity IgE receptor (FcεRIα) molecules on the surface of mast cells or basophils with a multivalent allergen can reduce the false-positive diagnoses observed in chemiluminescent and fluorescence enzyme immunoassays for type-I allergic patients. In this study, we detected sIgE Abs to the egg-allergen ovalbumin (OVA) and the wheat-allergen gluten in the sera of rats sensitized with each allergen using an amplified luminescence proximity homogeneous assay by crosslinking (AlphaCL). OVA and gluten were reacted with each sIgE Ab in the sera. Then, acceptor and donor beads labeled with the human FcεRIα were added to the reacted solution. The luminescence intensity for anti-OVA IgE Abs in the sera with the removal of IgG Abs was observed in five of seven (71.4%) of the sensitized rats, whereas no signals were observed in any of the unsensitized rats. The AlphaCL could also detect anti-gluten sIgE Abs in the sera of sensitized rats, but not of unsensitized rats. In conclusion, we successfully detected sIgE Abs in the sera of rats sensitized to two allergens using the AlphaCL. This detection method has the potential to be used as a new diagnostic tool for type-I allergic patients. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram showing the principle of AlphaCL for detection of sIgE Abs. When the sIgE Abs bound to FcεRIα on each bead are brought into proximity by crosslinking with the allergen, the acceptor beads can receive singlet oxygen emitted from the donor beads by excitation at 680 nm. A sharp emission peak at 615 nm is due to the energy generated by the singlet oxygen within the acceptor beads. sIgE, allergen-specific IgE; FcεRI, high-affinity IgE receptors.</p>
Full article ">Figure 2
<p>Effect of IgG removal from serum on the detection for serum anti-DNP (<b>A</b>) and anti-OVA (<b>B</b>) sIgE Abs by AlphaCL. Diluted human serum containing anti-DNP sIgE Abs and sera of rats unsensitized and sensitized to OVA were processed on the protein G HP SpinTrap™ column. Net counts were calculated from the total counts in each sample by subtracting the counts in buffer without sIgE Abs. ** <span class="html-italic">p</span> &lt; 0.01 was considered significantly different from buffer solution. <sup>††</sup> <span class="html-italic">p</span> &lt; 0.01 was considered significantly different from serum without IgG removal. All experiments were conducted in duplicate. N.D., not detected.</p>
Full article ">Figure 3
<p>Optimization of serum dilutions (<b>A</b>), OVA concentrations (<b>B</b>), and the incubation time (<b>C</b>), and concentration-dependent crosslinking of each bead by serum anti-OVA sIgE Abs (<b>D</b>). Net counts were calculated from the total counts in each sample by subtracting the counts in buffer without sIgE Abs. The cut-off value was set as the mean plus two standard deviations of buffer alone. All experiments were conducted in duplicate.</p>
Full article ">Figure 4
<p>Detection of anti-OVA sIgE Abs (<b>A</b>) and anti-gluten IgE Abs (<b>B</b>) in sera of rats unsensitized (open column) and sensitized (closed column) to OVA (<span class="html-italic">n</span> = 7) and gluten (<span class="html-italic">n</span> = 3) by ELISA and AlphaCL. The scatter plots of serum sIgE levels of sensitized rat to OVA (open circle) and gluten (closed circle) detected between ELISA and AlphaCL (<b>C</b>). Statistical correlations were obtained by Spearman’s correlation coefficient and rank test. Net counts were calculated from the total counts in each sample by subtracting the counts in buffer without sIgE Abs. All experiments were conducted in duplicate. Dashed lines represent cut-off values. N.D., not detected.</p>
Full article ">
11 pages, 1949 KiB  
Article
Autoimmune Mast Cell Activation Test as a Diagnostic Tool in Chronic Spontaneous Urticaria
by Ana Koren, Luka Dejanović, Matija Rijavec, Peter Kopač, Mojca Bizjak, Mihaela Zidarn, Mitja Košnik and Peter Korošec
Int. J. Mol. Sci. 2024, 25(17), 9281; https://doi.org/10.3390/ijms25179281 - 27 Aug 2024
Viewed by 369
Abstract
Chronic spontaneous urticaria (CSU) is associated with skin mast cell activation, and its triggering mechanisms are not completely elucidated. Evidence suggests an autoimmune component of CSU. Our aim was to assess the usefulness of an autoimmune mast cell activation test (aiMAT) for diagnosing [...] Read more.
Chronic spontaneous urticaria (CSU) is associated with skin mast cell activation, and its triggering mechanisms are not completely elucidated. Evidence suggests an autoimmune component of CSU. Our aim was to assess the usefulness of an autoimmune mast cell activation test (aiMAT) for diagnosing and differentiating CSU into different subtypes. We enrolled 43 patients with active, uncontrolled CSU before starting treatment with omalizumab and 15 controls. Patients were evaluated based on omalizumab response. aiMATs were performed using non-IgE-sensitized (NS) or myeloma IgE-sensitized (S) LAD2 cells, which were then stimulated with CSU/control sera (25 µL and 10 µL). The expression of CD63 was assessed with flow cytometry. CD63 response on NS-LAD2 was significantly increased in CSU patients compared to controls after the stimulation with 25 µL CSU/control sera (p = 0.0007) and with 10 µL CSU/control sera (p = 0.0001). The ROC curve analysis demonstrated an area under the curve (AUC) of 0.82. The cutoff for autoimmune-non-IgE-sensitized-MAT was 40.3% CD63+ LAD2, which resulted in 73.3% sensitivity and 81.4% specificity. CD63 response on S-LAD2 was significantly increased in CSU patients compared to controls after the stimulation with 25 µL CSU/control sera (p = 0.03). The ROC curve analysis demonstrated an AUC of 0.66. The cutoff for the autoimmune-myeloma IgE-sensitized-MAT was 58.4% CD63+ cells, which resulted in 62.8% sensitivity and 66.7% specificity. Overall, 36 out of 43 (84%) patients responded to omalizumab, and 7 (16%) were nonresponders. We found no differences between LAD2 CD63 response and response to omalizumab. In conclusion, aiMAT could represent a new diagnostic tool in CSU. Additional studies are needed to evaluate the potential benefits during omalizumab therapy. Full article
(This article belongs to the Special Issue Progression of Allergy and Immune Response)
Show Figures

Figure 1

Figure 1
<p>LAD2 immunophenotyping. Expression of CD117, FcɛRI, IgE, and IgE after stimulation with patients’/control sera on (<b>A</b>) non-IgE sensitized or (<b>B</b>) myeloma IgE sensitized LAD2 cells. APC-Cy7, Allophycocyanin/Cyanine7; PE, phycoerythrin; FITC, fluorescein isothiocyanate.</p>
Full article ">Figure 2
<p>The diagnostic utility of autoimmune MAT in CSU. (<b>A</b>) Comparison in LAD2 CD63 response between 43 CSU patients and 15 controls after the stimulation of non-IgE sensitized LAD2 cells with 25 µL and 10 µL of sera and the corresponding receiver operator characteristic (ROC) curve. (<b>B</b>) Comparison in LAD2 CD63 response between CSU patients and controls after the stimulation of myeloma IgE sensitized LAD2 cells with 25 µL and 10 µL of sera and the corresponding ROC curve. The presented values are the median ± interquartile range.</p>
Full article ">Figure 3
<p>Spearman’s coefficient correlation analysis between LAD2 CD63 response after the stimulation with 25 µL CSU sera and the stimulation with 10 µL CSU sera using (<b>A</b>) non-IgE sensitized LAD2 cells (NS-LAD2) or (<b>B</b>) myeloma IgE sensitized LAD2 cells (S-LAD2) in 43 CSU patients.</p>
Full article ">Figure 4
<p>Differences in LAD2 CD63 response between nonresponders (OMA-NR) and responders (OMA-R) to omalizumab therapy and controls (C) after the stimulation of (<b>A</b>) non-IgE sensitized LAD2 or (<b>B</b>) myeloma IgE sensitized LAD2 cells with 25 µL (upper row) and 10 µL (lower row) of sera. The presented values are the median ± interquartile range.</p>
Full article ">
20 pages, 18298 KiB  
Article
The Contribution of Mast Cells to the Regulation of Elastic Fiber Tensometry in the Skin Dermis of Children with Marfan Syndrome
by Dmitrii Atiakshin, Ekaterina Nikolaeva, Alla Semyachkina, Andrey Kostin, Artem Volodkin, Sergey Morozov, Michael Ignatyuk, Liudmila Mikhaleva, Grigory Demyashkin, Daniel Elieh-Ali-Komi, Igor Buchwalow and Markus Tiemann
Int. J. Mol. Sci. 2024, 25(17), 9191; https://doi.org/10.3390/ijms25179191 - 24 Aug 2024
Viewed by 481
Abstract
Marfan syndrome (MFS) is a hereditary condition accompanied by disorders in the structural and regulatory properties of connective tissue, including elastic fibers, due to a mutation in the gene encodes for fibrillin-1 protein (FBN1 gene) and the synthesis of abnormal fibrillin-1 glycoprotein. Despite [...] Read more.
Marfan syndrome (MFS) is a hereditary condition accompanied by disorders in the structural and regulatory properties of connective tissue, including elastic fibers, due to a mutation in the gene encodes for fibrillin-1 protein (FBN1 gene) and the synthesis of abnormal fibrillin-1 glycoprotein. Despite the high potential of mast cells (MCs) to remodel the extracellular matrix (ECM), their pathogenetic significance in MFS has not been considered yet. The group of patients with Marfan syndrome included two mothers and five children (three girls aged 4, 11, and 11 and two boys aged 12 and 13). Normal skin was examined in two children aged 11 and 12. Histochemical, monoplex, and multiplex immunohistochemical techniques; combined protocols of simultaneous histochemical and immunohistochemical staining (the results of staining were assessed using light, epifluorescence, and confocal microscopy); and bioinformatics algorithms for the quantitative analysis of detected targets were used to evaluate mast cells and their relationship with other cells from extracellular structures in the skin dermis. Analysis of the skin MC population in children with Marfan syndrome revealed a considerably increased number of intra-organic populations with the preservation of the specific Tryptase+Chymase+CPA3+ protease profile typical of the skin. The features of the MC histotopography phenotype in MFS consisted of closer colocalization with elastic fibers, smooth muscle cells, and fibroblasts. MCs formed many intradermal clusters that synchronized the activity of cell functions in the stromal landscape of the tissue microenvironment with the help of spatial architectonics, including the formation of cell chains and the creation of fibrous niches. In MCs, the expression of specific proteases, TGF-β, and heparin increased, with targeted secretion of biologically active substances relative to the dermal elastic fibers, which had specific structural features in MFS, including abnormal variability in thickness along their entire length, alternating thickened and thinned areas, and uneven surface topography. This paper discusses the potential role of MCs in strain analysis (tensometry) of the tissue microenvironment in MFS. Thus, the quantitative and qualitative rearrangements of the skin MC population in MFS are aimed at altering the stromal landscape of the connective tissue. The results obtained should be taken into account when managing clinical signs of MFS manifested in other pathogenetically critical structures of internal organs, including the aorta, tendons, cartilage, and parenchymal organs. Full article
(This article belongs to the Special Issue Mast Cells in Immunity and Disease: Second Edition)
Show Figures

Figure 1

Figure 1
<p>Mast cell content in the skin of patients with Marfan syndrome (in %, relative content to other cells in the dermis). Notes: *—mother of patient No. 2; **—mother of patients No. 6 and No. 7; CPA3—Carboxypeptidase A3.</p>
Full article ">Figure 2
<p>Histotopography of tryptase-positive mast cells in the skin dermis of patients with Marfan syndrome. Frequency of colocalization of MCs (%) with a fibrous component, αSMA-positive cells, and fibroblast. Staining technique: <sup>1</sup> multiplex detection of tryptase, elastic, and collagen fibers; <sup>2</sup> multiplex immunohistochemical detection of tryptase and α-SMA; <sup>3</sup> Giemsa staining. α-SMA: alpha-smooth muscle actin. Notes: *—mother of patient No. 2; **—mother of patients No. 6 and No. 7.</p>
Full article ">Figure 3
<p>Skin mast cells in children without pathology. Technique: Giemsa staining. (<b>a</b>) Two contacting MCs with signs of secretion (arrowed). (<b>b</b>) MCs in the skin dermis: one of the cells has a long, narrow process (arrowed). (<b>c</b>,<b>d</b>) MC colocalization with fibroblasts (arrowed). (<b>e</b>) Adjacency of an MC to the postcapillary wall (arrowed). (<b>f</b>) A group of MCs in contact with each other and fibroblasts (arrowed). (<b>g</b>) MCs and a nuclear-free MC fragment located between bundles of collagen fibers (arrowed). (<b>h</b>,<b>i</b>) Elongated MCs in contact with fibrocytes (presumably arrowed) and collagen fibers (double arrowed). (<b>j</b>) MC that forms an outgrowth in the skin dermis. (<b>k</b>) An elongated MC adjacent to two fibroblasts. (<b>l</b>) A nuclear-free MC fragment among bundles of collagen fibers. (<b>m</b>) A degranulated MC with a predominantly peripheral localization of secretory granules. (<b>n</b>) MCs in contact with each other (arrowed), forming a functional chain throughout. Scale: 5 µm.</p>
Full article ">Figure 4
<p>Mast cells in the fibrous landscape of the skin dermis in children without pathology. Technique: (<b>a</b>–<b>j</b>) Weigert stain combined with tryptase immunohistochemical detection. (<b>k</b>–<b>m</b>) Weigert–Van Gieson stain combined with immunohistochemical tryptase detection. Notes: (<b>a</b>) general view of the elastic network in the papillary (arrowed) and reticular (double arrowed) layers of the skin dermis, with MCs. (<b>b</b>,<b>c</b>) MC colocalization with thin elastic fibers in the papillary layer of the skin dermis (arrowed). (<b>h</b>–<b>m</b>) Interaction of MCs with elastic fibers in the reticular dermis. (<b>h</b>) MC in contact with several small-caliber (arrowed) and large-caliber (double arrowed) elastic fibers. (<b>i</b>,<b>j</b>) Tryptase-positive MC granules surrounding thick elastic fibers (arrowed). (<b>k</b>) Predominant localization of MC secretory granules in the area of elastic fibers. (<b>l</b>,<b>m</b>) Target tryptase secretion towards elastic fibers (arrowed) and collagen fibers (double arrowed). Scale: (<b>a</b>) 50 µm; the rest—5 µm.</p>
Full article ">Figure 5
<p>Mast cells in the skin dermis in Marfan syndrome. Technique: (<b>a</b>–<b>h</b>) staining with Giemsa solution, (<b>i</b>–<b>t</b>) immunohistochemical staining for tryptase (<b>i</b>–<b>l</b>), carboxypeptidase A3 (<b>m</b>–<b>p</b>), chymase (<b>q</b>,<b>r</b>), and CD117 (<b>s</b>,<b>t</b>). Notes: (<b>a</b>–<b>e</b>) options for various MC colocalizations with fibroblasts (arrowed). (<b>f</b>) A nuclear-free area in the cytoplasm of an elongated MC. (<b>g</b>,<b>h</b>) Secretion of heparin-containing granules onto elastic fibers. (<b>i</b>) A group of MCs closely localized to fibroblasts. (<b>j</b>) Directed distribution of tryptase over long distances in the skin dermis (arrowed). (<b>k</b>) Active tryptase secretion onto the elastic fiber (arrowed). (<b>l</b>) Group of interacting CPA3<sup>+</sup> MCs. (<b>m</b>) An elongated MC with a predominantly peripheral localization of CPA3 in the cytoplasm. (<b>n</b>–<b>p</b>) Active targeted secretion of CPA3 onto elastic fibers. MC granules adjacent to elastic fibers. (<b>q</b>,<b>r</b>) Active chymase degranulation onto fibers and cells of the skin dermis. (<b>s</b>,<b>t</b>) Predominantly peripheral localization of CD117 in the MC cytoplasm (arrowed). Scale: 5 µm. CPA3—Carboxypeptidase A3.</p>
Full article ">Figure 6
<p>Target secretion of mast cell tryptase to the structural components of the elastic landscape of the skin dermis in Marfan syndrome. Technique: Combined Weigert staining with immunohistochemical detection of MC tryptase. Notes: (<b>a</b>,<b>b</b>) MC migration to areas with abnormal elastic fibers. (<b>c</b>) Intensive degranulation of MC with the development of elastic fiber degradation. (<b>d</b>) Spatial localization of MCs between three directions of elastic fibers. (<b>e</b>–<b>g</b>) Targeted MC degranulation onto selected elastic fibers in the skin dermis (arrowed). (<b>h</b>) Accumulation of several MCs in areas of elastic fiber remodeling (arrowed). (<b>i</b>–<b>m</b>) Targeted tryptase secretion to the selective loci on elastic fibers of various thicknesses (arrowed). (<b>n</b>,<b>o</b>) Interaction of individual (<b>n</b>) or grouped (<b>o</b>) secretory granules (<b>n</b>) and the external component of elastic fibers from abnormal microfibrils with tryptase accumulation on the surface (arrowed). Scale: 5 µm.</p>
Full article ">Figure 7
<p>Mast cell mapping in the fibrous landscape of the skin dermis in MFS. Technique: Combined Weigert–Van Gieson staining with immunohistochemical tryptase detection. Notes: (<b>a</b>) Simultaneous tryptase secretion onto the collagen and elastic fibers in the skin dermis (arrowed). (<b>b</b>) Formation of a pericellular zone of extracellular matrix remodeling (arrowed) and its impact on elastic fibers (double arrowed). (<b>c</b>) Active effect of tryptase on the collagen ECM (double arrowed) and extensive areas on the surface of elastic fibers (arrowed). (<b>d</b>) Selective localization of autonomous secretory MC granules in the elastic fiber area in the skin dermis (arrowed). (<b>e</b>) Selective tryptase secretion onto elastic fibers with a difference in their histochemical properties (arrowed). (<b>f</b>,<b>g</b>) Active entry of tryptase into areas of contact with collagen (arrowed) and elastic fibers (double arrowed). (<b>h</b>) Evident remodeling of the skin dermis by mast cells, with visible signs of elastic fiber destruction (indicated by an arrow). (<b>i</b>,<b>j</b>) Variants in MC localization in the tensometric areas of the elastic network of the skin dermis (arrowed), with intense tryptase secretion on the surface of the fibers. Scale: 5 µm.</p>
Full article ">Figure 8
<p>Secretory profile of skin MCs in Marfan syndrome. Technique: Multiplex immunohistochemistry with simultaneous detection of tryptase, chymase, and CPA3 (<b>a</b>) and tryptase with αSMA (<b>b</b>,<b>c</b>) and TGFβ (<b>d</b>). Notes: (<b>a</b>) MCs in the skin dermis with a predominant Tryptase<sup>+</sup>Chymase<sup>+</sup>CPA3<sup>+</sup> profile. (<b>b</b>,<b>c</b>) Colocalization of MCs with smooth myocytes in the microvasculature (double arrowed) and α-SMA-positive cells (arrowed), with active tryptase secretion. (<b>d</b>,<b>e</b>) High level of TGF-β expression in MCs in the skin dermis (arrowed). Scale: (<b>c’</b>) 2 µm; (<b>e</b>) 5 µm; the rest—10 µm. CPA3: carboxypeptidase A3. TGF-β: transforming growth factor beta.</p>
Full article ">
20 pages, 3949 KiB  
Article
The Complement System Is Essential for Arteriogenesis by Enhancing Sterile Inflammation as a Relevant Step in Collateral Artery Growth
by Amanda Zhu, Carolin Baur, Philipp Götz, Katharina Elbs, Manuel Lasch, Anna Faro, Klaus T. Preissner and Elisabeth Deindl
Cells 2024, 13(17), 1405; https://doi.org/10.3390/cells13171405 - 23 Aug 2024
Viewed by 409
Abstract
Arteriogenesis is an inflammatory driven mechanism, describing the growth of a natural bypass from pre-existing collateral arteries to compensate for an occluded artery. The complement system component C3 is a potent natural inflammatory activator. Here, we investigated its impact on the process of [...] Read more.
Arteriogenesis is an inflammatory driven mechanism, describing the growth of a natural bypass from pre-existing collateral arteries to compensate for an occluded artery. The complement system component C3 is a potent natural inflammatory activator. Here, we investigated its impact on the process of collateral artery growth using C3-deficient (C3 −/−) and wildtype control mice in a murine hindlimb model of arteriogenesis. Induction of arteriogenesis by unilateral femoral artery ligation resulted in decreased perfusion recovery in C3 −/− mice on day 7 as shown by Laser Doppler imaging. Immunofluorescence staining revealed a reduced vascular cell proliferation in C3 −/− mice. Gene expression analysis displayed a significant reduction in monocyte chemoattractant protein-1 (MCP-1) expression in C3 −/− mice. Interestingly, 3 days after induction of arteriogenesis, the number of macrophages (CD68+) recruited to growing collaterals was not affected by C3 deficiency. However, a significant reduction in inflammatory M1-like polarized macrophages (CD68+/MRC1) was noted. Forced mast cell activation by Compound 48/80 as well as exogenous MCP-1 application rescued the number of M1-like polarized macrophages along with perfusion recovery in C3 −/− mice. In summary, this study demonstrates that complement C3 influences arteriogenesis by mediating MCP-1 expression, which is essential for the induction and enhancement of sterile inflammation. Full article
Show Figures

Figure 1

Figure 1
<p>C3 −/− mice show reduced perfusion recovery after femoral artery ligation (FAL). (<b>a</b>) Representative photographs of superficial collateral arteries (arrows) in the adductor muscles of wildtype (WT, upper panels) and C3 −/− (lower panels) mice. Left panels: Grown collateral arteries of the occluded leg (Occ) have increased caliber size and display a corkscrew-like pattern. Right panels: Pre-existing collaterals on the sham-operated leg (Sham) appear as thin linear vessels. Photographs were taken 7 days after FAL. (<b>b</b>) Representative flux images of Laser Doppler measurements of wildtype (WT, left images) and C3 −/− mice (right images) 3 days and 7 days after FAL. Flux scale bar shown below (red = high flow, blue = low flow). (<b>c</b>) Line graph displaying the relative perfusion (Occ/Sham) of WT and C3 −/− mice before FAL (baseline), directly after FAL (aFAL), at day 3 and day 7 after FAL. Data shown are means ± SEM, <span class="html-italic">n</span> = 5 mice per group, * <span class="html-italic">p</span> &lt; 0.05 WT compared to C3 −/− by two-way ANOVA with Bonferroni’s multiple comparison test.</p>
Full article ">Figure 2
<p>C3 deficiency reduces vascular cell proliferation. The scatter plots with bars show (<b>a</b>) the inner luminal diameter of growing collaterals and (<b>b</b>) the percentage of proliferating vascular cells (BrdU<sup>+</sup> cells) per growing collateral of wildtype (WT) and C3 −/− mice 7 days after femoral artery ligation (FAL). The dashed horizontal line in (<b>a</b>) represents the mean sham value. Data shown are means ± SEM, <span class="html-italic">n</span> = 6 mice per group with 3 slices, 2 collaterals each, * <span class="html-italic">p</span> &lt; 0.05 WT compared to C3 −/− by unpaired student’s <span class="html-italic">t</span>-test. (<b>c</b>) Representative images of immunofluorescence staining of growing collateral arteries showing merged images (left) and single stain (right) of WT (upper panels) and C3 −/− (lower panels) mice 7 days after FAL. BrdU (red) served as a proliferation marker, ACTA2 (green) served as a smooth muscle cell marker, CD31 (white) visualized the endothelial cell layer and DAPI (blue) labeled nuclei. Scale bar: 20 µm.</p>
Full article ">Figure 3
<p>C3 deficiency impairs M1-like macrophage polarization after induction of arteriogenesis. The scatter plots with bars display (<b>a</b>) the total number of perivascular macrophages (CD68<sup>+</sup> cells) 3 days and (<b>b</b>) 7 days after femoral artery ligation (FAL), the number of M1-like macrophages (CD68<sup>+</sup>/MRC1<sup>−</sup> cells) (<b>c</b>) 3 days and (<b>d</b>) 7 days after FAL, as well as (<b>e</b>) the number of M2-like macrophages (CD68<sup>+</sup>/MRC1<sup>+</sup> cells) per collateral artery on day 3 and (<b>f</b>) day 7 after induction of arteriogenesis of WT and C3 −/− mice. The dashed horizontal line represents the mean sham value. Data shown are means ± SEM, <span class="html-italic">n</span> = 3 mice per group with 3 slices, 2 collaterals each on day 3, and <span class="html-italic">n</span> = 6 mice per group with 3 slices, 2 collaterals each on day 7 after FAL, * <span class="html-italic">p</span> &lt; 0.05 and ns ≥ 0.05 WT compared to C3 −/− by unpaired student’s <span class="html-italic">t</span>-test. (<b>g</b>) Representative images of immunofluorescence stains of growing collateral arteries of WT (upper panel) and C3 −/− (lower panel) mice 3 days and (<b>h</b>) 7 days after FAL. CD68 (green) served as a marker for macrophages, MRC1 (red) served as a marker for M2-like macrophages, CD31 (white) labeled the endothelial cell layer and DAPI (blue) labeled nuclei. Scale bar: 20 µm.</p>
Full article ">Figure 4
<p>C3 is essential for MCP-1 expression in arteriogenesis. The scatter plots with bars show (<b>a</b>) the mRNA expression level of MCP-1 in collaterals of untreated wildtype (WT) and C3 −/− mice as well as (<b>b</b>) WT and C3 −/− mice treated with C48/80, as quantified by qRT-PCR 12 h after femoral artery occlusion (occ, orange bars) or sham operation (sham, grey bars). The data were normalized to the expression level of the 18S rRNA and shown are means ± SEM, <span class="html-italic">n</span> = 3 in triplicates per group, * <span class="html-italic">p</span> &lt; 0.05 and ns ≥ 0.05, compared by two-way ANOVA.</p>
Full article ">Figure 5
<p>Additional MCP-1 availability increases the number of M1-like macrophages in C3 −/− mice 3 days after induction of arteriogenesis. Scatter plots with bars representing (<b>a</b>) the total number of perivascular macrophages (CD68<sup>+</sup> cells) of wildtype (WT, grey bars), C3 −/− (pink bars) and C3 −/− after treatment with C48/80 (green bars) or (<b>b</b>) MCP-1 (blue bars), the number of M1-like macrophages (CD68<sup>+</sup>/MRC1<sup>−</sup> cells) after treatment with (<b>c</b>) C48/80 or (<b>d</b>) MCP-1 as well as the number of M2-like macrophages (CD68<sup>+</sup>/MRC1<sup>+</sup> cells) after treatment with (<b>e</b>) C48/80 or (<b>f</b>) MCP-1 per collateral artery on day 3 after femoral artery ligation (FAL). The dashed horizontal line represents the mean sham value. Data shown are means ± SEM, <span class="html-italic">n</span> = 3 mice per group with 3 slices, 2 collaterals each, * <span class="html-italic">p</span> &lt; 0.05 and ns ≥ 0.05, compared by one-way ANOVA with Bonferroni’s multiple comparison test.</p>
Full article ">Figure 6
<p>Treatment with C48/80 as well as with MCP-1 rescues arteriogenesis in C3 −/− mice. Scatter plots with bars show the percentage of proliferating vascular cells (BrdU<sup>+</sup> cells) in WT, C3 −/− and C3 −/− treated with (<b>a</b>) C48/80 or (<b>b</b>) MCP-1 collateral arteries 7 days after FAL. <span class="html-italic">n</span> = 4 mice per group, 3 slices with 2 collaterals each were analyzed per mouse. * <span class="html-italic">p</span> &lt; 0.05 and ns ≥ 0.05, one-way ANOVA with Bonferroni’s multiple comparison test. Line graphs displaying the perfusion ratio (occ/sham) before femoral artery ligation (FAL, baseline), directly after FAL (aFAL), at day 3 and day 7 in wildtype (WT), C3 −/−, and C3 −/− mice after treatment with (<b>c</b>) C48/80 or (<b>d</b>) MCP-1 as measured by LDI. <span class="html-italic">n</span> = 4 mice per group. <sup>Φ</sup> <span class="html-italic">p</span> &lt; 0.05 (WT compared with C3 −/−), <sup>Ω</sup> <span class="html-italic">p</span> &lt; 0.05 (WT compared with C3 −/− + C48/80), <sup>&amp;</sup> <span class="html-italic">p</span> &lt; 0.05 (C3 −/− compared with C3 −/− + C48/80), <sup>δ</sup> <span class="html-italic">p</span> &lt; 0.05 (WT compared with C3 −/− + MCP-1), <sup>ψ</sup> <span class="html-italic">p</span> &lt; 0.05 (C3 −/− compared to C3 −/− + MCP-1), two-way ANOVA with Bonferroni’s multiple comparison test. Data are means ± SEM.</p>
Full article ">
10 pages, 1191 KiB  
Review
Tryptase: The Silent Witness of Past and Ongoing Systemic Events
by Irena Oštrić Pavlović, Sara Radović, Danka Krtinić, Jelena Spirić, Nataša Kusić, Antonije Veličković and Vesna Tomić-Spirić
Medicina 2024, 60(9), 1380; https://doi.org/10.3390/medicina60091380 - 23 Aug 2024
Viewed by 399
Abstract
Introduction: Tryptase is an important biomarker widely used in the laboratory confirmation of severe hypersensitivity reactions, especially anaphylaxis. It also plays a crucial role in the diagnosis, risk stratification, management and prognostic evaluation of many other mast cell-related conditions. Aim: This paper aims [...] Read more.
Introduction: Tryptase is an important biomarker widely used in the laboratory confirmation of severe hypersensitivity reactions, especially anaphylaxis. It also plays a crucial role in the diagnosis, risk stratification, management and prognostic evaluation of many other mast cell-related conditions. Aim: This paper aims to highlight the role of serum tryptase, both in allergic disorders and other mast cell-related conditions. Two clinical cases regarding timely serum tryptase acquisition (in drug hypersensitivity reactions during the imaging procedure and perioperative anaphylaxis) are meant to emphasize the clinical potential of this protease. Method: We performed a comprehensive literature search of the PubMed/Medline and Scopus databases. From a total of 640 subject related publications, dating from 1940 to 2024, 45 articles written in English were selected. Literature search results: Total serum tryptase is a simple, cost-effective analysis with a normal baseline tryptase (sBT) level below 8.4 µg/L. Elevated sBT can indicate hereditary alpha-tryptasemia (HαT), mastocytosis and other non-allergic disorders. Patients with higher sBT levels, especially with insect venom allergy, have an increased risk of severe reactions and thereby require a prolonged treatment. All immediate systemic hypersensitivity reactions require a correlation between serum acute tryptase (sAT) and sBT. According to the guidelines, measuring sAT 30 min to 2 h after the symptom onset and sBT 24 h after the resolution, using the 20 + 2 rule and an sAT/sBT ratio of 1.685, improves the diagnostic accuracy in anaphylaxis. Conclusions: Tryptase levels should be acquired in all cases with clinical suspicion of MC degranulation. Given the increasing clinical relevance, elevated baseline serum tryptase levels require a multidisciplinary approach and further investigation. Full article
(This article belongs to the Special Issue Pitfalls and Challenges in Allergy Management)
Show Figures

Figure 1

Figure 1
<p>Clinical significance of serum tryptase measurement in acute reactions and chronic conditions.</p>
Full article ">
15 pages, 5830 KiB  
Article
Unexpected Expression and Function of FcεRI in Immortalized Breast Cancer Cells: A Cautionary Null Study
by Alexandria M. Ashbaugh, David O. Lyons, Carianna M. Keyser and Nicholas A. Pullen
Cells 2024, 13(16), 1399; https://doi.org/10.3390/cells13161399 - 22 Aug 2024
Viewed by 615
Abstract
The high-affinity IgE receptor, FcεRI, is typically associated with type 2 effectors such as mast cells (MC). The relatively unique expression profile of FcεRI and accumulating evidence from pre-clinical and clinical settings, such as MC interactions with tumors, have led us to study [...] Read more.
The high-affinity IgE receptor, FcεRI, is typically associated with type 2 effectors such as mast cells (MC). The relatively unique expression profile of FcεRI and accumulating evidence from pre-clinical and clinical settings, such as MC interactions with tumors, have led us to study MCs as a potential therapeutic target in breast cancer. Our work identified MCs interacting with tumor cells at primary sites using the 4T1 (BALB/c) adenocarcinoma model in vivo. However, this analysis was complicated by a surprising finding that the tumor cells intrinsically and strongly expressed FcεRI. We further studied the expression and function of FcεRI in breast cancer cells in vitro. The 4T1 cells expressed FcεRI to a level similar to mouse bone marrow-derived MC (BMMC). Additionally, two established breast cancer cultures derived from human T-47D cells, one estrogen-dependent (E3) and the other estrogen-withdrawn (EWD8), also expressed FcεRI with EWD8 cells showing the greatest abundance. Functional analyses indicated that IgE-mediated antigen stimulation did not elicit classic Ca2+ flux in breast cancer cells as seen in the respective species’ MCs; however, FcεRI crosslinking could stimulate IL-6 production from the T-47D derivatives. Preliminary analysis of primary breast cancer biopsy datasets using R2: Genomics Analysis and Visualization Platform was discordant with our in vivo model and in vitro observations. Indeed, FcεRI mRNA abundance declined in metastatic breast cancers compared to non-cancerous breast tissue. Altogether, we report a previously unidentified and immunologically substantive difference between breast cancer models and human primary tumors. Investigators pursuing FcεRI-relevant therapeutics in this context should be aware of this translational barrier. Full article
Show Figures

Figure 1

Figure 1
<p>Left: Representative images of CAE staining at 40X objective magnification. Mast cells circled in blue for context. Top: WT 4T1 tumors. Bottom: IL-6 tumors. Right: Quantification of the MC concentration between WT and KO tumors. KO tumors were more likely to have higher MC counts (** <span class="html-italic">p</span> &lt; 0.05) [<a href="#B13-cells-13-01399" class="html-bibr">13</a>].</p>
Full article ">Figure 2
<p>(<b>A</b>–<b>C</b>) Control “Wildtype” 4T1 tumors stained for FcεRI (green) and Hoechst (blue) at 20X objective magnification. (<b>D</b>–<b>F</b>) IL-6 knock-out 4T1 tumors stained for FceR-1 (green) and Hoechst (blue) 20X objective magnification. All images were taken with the Zeiss LSM-900 microscope.</p>
Full article ">Figure 3
<p>(<b>A</b>) Unstained negative control C57BL/6J BMMCs. (<b>B</b>) Stained BMMCs for anti-mouse FcεRI, &gt;90% positive, as expected. (<b>C</b>) Unstained negative control Jurkat. (<b>D</b>) Stained Jurkat for anti-mouse FcεRI. (<b>E</b>) Unstained negative control 4T1s. (<b>F</b>) Stained 4T1s for anti-mouse FcεRI &gt; 90%, an unexpected result.</p>
Full article ">Figure 4
<p>(<b>A</b>) Unstained negative control LAD2. (<b>B</b>) Stained LAD2 for anti-human FcεRI, ~80% positive as expected. (<b>C</b>) Unstained negative control EWD8. (<b>D</b>) Stained EWD8 for anti-human FcεRI, ~76% positive, an unexpected result. (<b>E</b>) Unstained negative control E3. (<b>F</b>) Stained E3 for anti-human FcεRI, ~36% positive, an unexpected result. (<b>G</b>) Unstained negative control MCF7. (<b>H</b>) Stained MCF7 for anti-human FcεRI, ~11% positive, an unexpected result.</p>
Full article ">Figure 5
<p>Mouse Cell Ca<sup>2+</sup> Assay. Left: NGS3 Ca<sup>2+</sup> (Fluo-4 AM fluorescence). After activation with TNP-KLH, NGS3 increased in Ca<sup>2+</sup>, as expected (red); compared to unstained (blue). Right: 4T1 Ca<sup>2+</sup> Assay. 4T1 did not have a difference in Ca<sup>2+</sup> release post-activation with TNP-KLH, however progressively increased in fluorescence as the assay proceeded in analysis when stained (blue); compared to unstained (orange).</p>
Full article ">Figure 6
<p>Human Cell Ca<sup>2+</sup> Assay. (<b>A</b>) LAD2 Ca<sup>2+</sup>. (<b>B</b>) E3 Ca<sup>2+</sup>. (<b>C</b>) EWD8 Ca<sup>2+</sup>. (<b>D</b>) MCF7 Ca<sup>2+</sup>. LAD2 showed increased Ca<sup>2+</sup> after the addition of antigen, as expected. E3, EWD8, and MCF7 showed no change in Ca<sup>2+</sup> post-antigen stimulation.</p>
Full article ">Figure 7
<p>FcεRI subunits expressed in mouse cells lines. (<b>A</b>) PCR gel from 4T1 and NGS3 cells (mouse lines). NGS3 expressed all three subunits (αβγ) as expected, and 4T1 expressed all three subunits (αβγ), which was an unexpected result. (<b>B</b>) Immunoblot confirming protein expression.</p>
Full article ">Figure 8
<p>Representative PCR gel from LAD2, E3, EWD8, and MCF7 (human lines). LAD2 expressed all three subunits as expected. E3 and EWD8 expressed all three subunits, an unexpected result. Repeated attempts to detect these subunits at the mRNA level in MCF7 were unsuccessful.</p>
Full article ">Figure 9
<p>IL-6 ELISA results. LAD2 and NGS3 produced IL-6 as expected when given IgE and antigen. E3 and EWD8 also produced IL-6 which was an unexpected result. 4T1 and MCF7 did not produce any IL-6, and thus are not depicted here.</p>
Full article ">Figure 10
<p>R2 Genomics search results. Top row: normal breast tissue. Bottom row: tumor breast tissue. GAPDH and ACTB from each dataset were included as internal reference standards (i.e., “loading controls”).</p>
Full article ">
Back to TopTop