[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

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (817)

Search Parameters:
Keywords = immunological tests

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
48 pages, 2851 KiB  
Review
Pollen–Food Allergy Syndrome: Allergens, Clinical Insights, Diagnostic and Therapeutic Challenges
by Laura Haidar, Camelia Felicia Bănărescu, Cristina Uța, Sandra Iulia Moldovan, Elena-Larisa Zimbru, Răzvan-Ionuț Zimbru, Elena Ciurariu, Marius Georgescu and Carmen Panaitescu
Appl. Sci. 2025, 15(1), 66; https://doi.org/10.3390/app15010066 - 25 Dec 2024
Abstract
Pollen–food allergy syndrome (PFAS), also known as oral allergy syndrome, is a common condition affecting individuals sensitized to pollens such as birch, ragweed, and grass. This syndrome arises from immunological cross-reactivity between pollen allergens and structurally similar proteins found in various fruits, vegetables, [...] Read more.
Pollen–food allergy syndrome (PFAS), also known as oral allergy syndrome, is a common condition affecting individuals sensitized to pollens such as birch, ragweed, and grass. This syndrome arises from immunological cross-reactivity between pollen allergens and structurally similar proteins found in various fruits, vegetables, and nuts. Although typically presenting with mild oral and pharyngeal symptoms, PFAS can occasionally result in severe allergic reactions, underscoring its clinical significance. This review explores the pathophysiology of PFAS, highlighting the molecular mechanisms underlying cross-reactivity and examining the main protein families involved, including those contributing to variations in symptom severity. Current diagnostic approaches, including skin prick testing, specific immunoglobulin E measurements, and component-resolved diagnostics, are discussed. Emerging diagnostic tools and biomarkers with potential to enhance accuracy are also examined. Therapeutic strategies for PFAS primarily focus on symptom management and avoidance of trigger foods. However, novel approaches such as allergen immunotherapy and biologics targeting key immune pathways are gaining traction as potential interventions for more severe or refractory cases. By addressing the diagnostic and therapeutic challenges of PFAS, this paper aims to provide clinicians and researchers with a comprehensive understanding of this condition, fostering improved patient care and the development of innovative treatment strategies. Full article
(This article belongs to the Special Issue New Diagnostic and Therapeutic Approaches in Food Allergy)
Show Figures

Figure 1

Figure 1
<p>The figure illustrates the PFAS mechanism, highlighting its two main phases: the sensitization phase and the clinical phase. During the sensitization phase, inhaled allergens, such as pollen, are presented by antigen-presenting cells to T helper 2 cells. These cells release cytokines like IL-4 and IL-13, which stimulate B cells to produce allergen-specific IgE antibodies. The IgE antibodies bind to the surface of mast cells, effectively priming them for future allergen exposure. During the clinical phase, ingestion of a cross-reactive allergen (e.g., fruits or vegetables) leads to the binding of the allergen to IgE on the surface of mast cells. This triggers mast cell degranulation, releasing inflammatory mediators such as histamine, leukotrienes, and prostaglandins. These mediators cause the characteristic symptoms of PFAS, including itching, swelling, or discomfort in the oral cavity, and also systemic symptoms. The cross-reactivity between pollen allergens and structurally similar food allergens is central to the development of PFAS symptoms.</p>
Full article ">Figure 2
<p>Stability and potential for severe systemic reactions of allergenic proteins [<a href="#B33-applsci-15-00066" class="html-bibr">33</a>,<a href="#B44-applsci-15-00066" class="html-bibr">44</a>,<a href="#B119-applsci-15-00066" class="html-bibr">119</a>,<a href="#B120-applsci-15-00066" class="html-bibr">120</a>]. The chart illustrates the relative stability (resistance to heat and digestion) and associated risk of severe systemic allergic reactions among different allergenic protein families. Proteins such as PR-10 and profilins are shown with lower stability and reduced potential for systemic reactions, while LTPs exhibit high stability and significant potential for systemic allergic responses. The arrow gradient represents the continuum from low to high stability and systemic reaction potential. GRPs: Gibberellin-Regulated Proteins; IFRs: Isoflavonoid reductases; LTPs: Lipid Transfer Proteins; PR-10: pathogenesis-related protein group 10; TLPs: Thaumatin-like proteins.</p>
Full article ">Figure 3
<p>Global distribution of key plants associated with PFAS, highlighting the most relevant species and their geographical ranges [<a href="#B121-applsci-15-00066" class="html-bibr">121</a>,<a href="#B122-applsci-15-00066" class="html-bibr">122</a>]. Created with ChartMap.net.</p>
Full article ">Figure 4
<p>PFAS diagnosis flowchart. The flowchart illustrates a structured approach for clinicians to evaluate and confirm the diagnosis of pollen–food allergy syndrome (PFAS).</p>
Full article ">
21 pages, 4110 KiB  
Article
Succinate Regulates Endothelial Mitochondrial Function and Barrier Integrity
by Reham Atallah, Juergen Gindlhuber, Wolfgang Platzer, Rishi Rajesh and Akos Heinemann
Antioxidants 2024, 13(12), 1579; https://doi.org/10.3390/antiox13121579 - 21 Dec 2024
Viewed by 317
Abstract
Endothelial dysfunction is a hallmark of several pathological conditions, including cancer, cardiovascular disease and inflammatory disorders. In these conditions, perturbed TCA cycle and subsequent succinate accumulation have been reported. The role of succinate as a regulator of immunological responses and inflammation is increasingly [...] Read more.
Endothelial dysfunction is a hallmark of several pathological conditions, including cancer, cardiovascular disease and inflammatory disorders. In these conditions, perturbed TCA cycle and subsequent succinate accumulation have been reported. The role of succinate as a regulator of immunological responses and inflammation is increasingly being recognized. Nevertheless, how endothelial cell function and phenotype are altered by elevated intracellular succinate has not been addressed yet. Thus, we employed numerous in vitro functional assays using primary HUVECs and diethyl succinate (DES), a cell membrane-permeable succinate analogue. An MTS assay 1 h post stimulation with DES suggested reduced metabolic activity in HUVECs. Concurrently, elevated production of ROS, including mitochondrial superoxide, and a reduction in mitochondrial membrane potential were observed. These findings were corroborated by Seahorse mito-stress testing, which revealed that DES acutely lowered the OCR, maximal respiration and ATP production. Given the link between mitochondrial stress and apoptosis, we examined important survival signalling pathways. DES transiently reduced ERK1/2 phosphorylation, a response that was followed by a skewed pro-apoptotic shift in the BAX to BCL2L1 gene expression ratio, which coincided with upregulating VEGF gene expression. This indicated an induction of mixed pro-apoptotic and pro-survival signals in the cell. However, the BAX/BCL-XL protein ratio was unchanged, suggesting that the cells did not commit themselves to apoptosis. An MTS assay, caspase 3/7 activity assay and annexin V/propidium iodide staining confirmed this finding. By contrast, stimulation with DES induced acute endothelial barrier permeability, forming intercellular gaps, altering cell size and associated actin filaments without affecting cell count. Notably, during overnight DES exposure gradual recovery of the endothelial barrier and cell sprouting was observed, alongside mitochondrial membrane potential restoration, albeit with sustained ROS production. COX-2 inhibition and EP4 receptor blockade hindered barrier restoration, implicating a role of COX-2/PGE2/EP4 signalling in this process. Interestingly, ascorbic acid pre-treatment prevented DES-induced acute barrier disruption independently from ROS modulation. In conclusion, succinate acts as a significant regulator of endothelial mitochondrial function and barrier integrity, a response that is counterbalanced by upregulated VEGF and prostaglandin production by the endothelial cells. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of DES on metabolic and mitochondrial function in HUVECs. (<b>A</b>) MTS assay of HUVECs stimulated with DES at indicated concentrations for 1 h (n = 3). (<b>B</b>) DHR 123 geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for selected time points (n = 5). (<b>C</b>) MitoSOX geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for selected time points (n = 3). (<b>D</b>) TMRE geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for selected time points (n = 4). For (<b>B</b>–<b>D</b>), the dotted line refers to vehicle-treated cells. (<b>E</b>) Representative microscopic image for 3 independent experiments with TMRE in HUVECs stimulated with 10 mM DES for indicated times. (<b>F</b>) Mito-stress assay of HUVECs demonstrating OCR with sequential addition of treatments. Acute response to DES, maximal respiration and ATP production were calculated (n = 5). For (<b>A</b>–<b>D</b>,<b>F</b>), data are presented as mean and SEM, with statistical significance determined using two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>B</b>–<b>D</b>) or one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>,<b>F</b>). For (<b>C</b>,<b>D</b>), * refers to comparison between vehicle and 10 mM DES, while # refers to comparison between vehicle and 5 mM DES. *, # <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 2
<p>Effect of DES on HUVECs viability. (<b>A</b>) Western blot of phosphorylated ERK1/2 and ratio of pERK/tERK/β-actin in HUVECs stimulated with 10 mM DES for indicated time points (n = 4). (<b>B</b>) RT-PCR of BAX, BCL2L1 and VEGF mRNA expression in HUVECs stimulated with 10 mM DES for 4 h (n = 5). (<b>C</b>) Western blot of BAX and BCL-XL in HUVECs stimulated with 10 mM DES for 6 h (n = 5). (<b>D</b>) MTS assay of HUVECs stimulated with indicated DES concentrations for 6 h (n = 3). (<b>E</b>) Caspase 3/7 activity of HUVECs post treatment with indicated DES concentrations for 16 h (n = 3). (<b>F</b>) Representative dot plot of annexin V/propidium iodide staining of HUVECs treated with indicated DES concentrations for 16 h. Live and apoptotic cells were quantified (n = 9). Data are presented as mean and SEM, with statistical significance determined using one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>,<b>D</b>–<b>F</b>) or paired <span class="html-italic">t</span>-test (<b>B</b>,<b>C</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; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Acute effect of DES on HUVECs barrier integrity. (<b>A</b>) Resistance of HUVECs monolayer stimulated with DES at indicated concentrations (n = 4). (<b>B</b>) Immunofluorescence staining of VE-cadherin and F-actin in HUVECs stimulated with 10 mM DES at selected time points. The image is representative of 3 independent experiments. Zoomed-in images are shown on the upper right side for each condition, scale bar = 50 µm. (<b>C</b>) Image analysis demonstrating number of nuclei, gap size, average cell size and median fluorescence intensity (MFI) of phalloidin-stained stress fibres. Data are presented as mean and SEM, with statistical significance determined using two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>) or one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>C</b>). For (<b>A</b>), * refers to comparison between vehicle and 10 mM DES, while # refers to comparison between vehicle and 5 mM DES. * <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 4
<p>Restoration of HUVECs barrier and sprouting during overnight stimulation with DES. (<b>A</b>) Resistance of HUVECs monolayer with overnight stimulation with DES at indicated concentrations (n = 6). (<b>B</b>) Spheroid sprouting assay of HUVECs stimulated with 10 mM DES for 16 h. Number of sprouts and total sprout length were calculated. The image is representative of three independent experiments. (<b>C</b>) Western blot of HIF-1α in HUVECs stimulated with DES at indicated concentrations for 16 h (n = 8). (<b>D</b>) Western blot of phosphorylated ERK1/2 and ratio of pERK/tERK/β-actin in HUVECs stimulated with indicated DES concentrations for 16 h (n = 4). (<b>E</b>) MTS assay of HUVECs stimulated with DES at indicated concentrations for 16 h (n = 3). (<b>F</b>) DHR 123 geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 16 h (n = 6). (<b>G</b>) MitoSOX geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 16 h (n = 5). (<b>H</b>) TMRE geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 16 h (n = 4). Data are presented as mean and SEM, with statistical significance determined using two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>), paired <span class="html-italic">t</span>-test (<b>B</b>) or one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>C</b>–<b>H</b>). ns refers to no statistical difference between vehicle and either 5 mM DES or 10 mM DES. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>COX-2/PGE<sub>2</sub>/EP4 contribution to barrier recovery in HUVECs. (<b>A</b>) Western blot of COX-2 in HUVECs after 16 h of stimulation with DES at indicated concentrations (n = 6). (<b>B</b>) Radioimmunoassay for quantification of PGE<sub>2</sub> in supernatants of HUVECs stimulated with DES for 16 h (n = 3). (<b>C</b>) Resistance of HUVECs monolayer treated with diclofenac ~1 h prior to stimulation with 10 mM DES (n = 3). (<b>D</b>) Resistance of HUVECs monolayer stimulated with PGE<sub>2</sub> (n = 3). (<b>E</b>) Resistance of HUVECs monolayer treated with EP2 antagonist ~2 h post treatment with 10 mM DES (n = 3). (<b>F</b>) Resistance of HUVECs monolayer treated with EP4 antagonist ~2 h post treatment with 10 mM DES (n = 3). Data are presented as mean and SEM, with statistical significance determined using one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>,<b>B</b>) or two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>C</b>–<b>F</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.</p>
Full article ">Figure 6
<p>Ascorbic acid prevention of initial barrier drop induced by DES in HUVECs. (<b>A</b>) Resistance of HUVECs monolayer treated with ascorbic acid at demonstrated concentrations 15 min prior to stimulation with DES at indicated concentrations (n = 4). (<b>B</b>) DHR 123 geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 1 h with/without 15 min of ascorbic acid pre-treatment (n = 3). (<b>C</b>) MitoSOX geometric mean of fluorescence intensity in HUVECs stimulated with DES at indicated concentrations for 1 h with/without 15 min of ascorbic acid pre-treatment (n = 3). Data are presented as mean and SEM, with statistical significance determined using two-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>A</b>) or one-way ANOVA for repeated measures followed by Tukey’s post hoc test (<b>B</b>,<b>C</b>). For (<b>A</b>), * refers to comparison between 10 mM DES and AA + 10 mM DES, while # refers to comparison between 5 mM DES and AA + 5 mM DES. *, # <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. AA denotes ascorbic acid.</p>
Full article ">
11 pages, 1786 KiB  
Article
Molecular Profiles of Sensitization to Non-Specific Lipid Transfer Proteins in Lithuania: Single Center Experience
by Sandra Sakalauskaite, Ligita Pilkyte, Edita Gasiuniene and Brigita Gradauskiene
Int. J. Mol. Sci. 2024, 25(24), 13535; https://doi.org/10.3390/ijms252413535 - 18 Dec 2024
Viewed by 379
Abstract
Non-specific Lipid Transfer proteins (nsLTPs) are relevant allergens of several pollens and plant foods. Sensitization to nsLTPs is not typical in our region. Still, it has become an increasingly common cause of IgE-mediated food allergies and food-induced anaphylaxis in Northern Europe in recent [...] Read more.
Non-specific Lipid Transfer proteins (nsLTPs) are relevant allergens of several pollens and plant foods. Sensitization to nsLTPs is not typical in our region. Still, it has become an increasingly common cause of IgE-mediated food allergies and food-induced anaphylaxis in Northern Europe in recent decades. No in-depth studies describe the prevalence of sensitization of molecular components to nsLTPs in Lithuania. This study aimed to determine the sensitization profile of atopic patients at the Immunology and Allergy Department of Kauno Klinikos to the components of nsLTPs, using molecular allergen component analysis. Sixty Lithuanian adults with symptoms of allergic rhinitis and/or allergic asthma and/or food allergies were included into the study. Specific immunoglobulin E (IgE) levels were measured using two in vitro techniques: allergen extract and molecular component analysis. Results showed that 25% of subjects were sensitized to nsLTP-containing allergen sources, mostly to Zea m 14, Mal d 3, Vit v 1, and Art v 3. The median amount of total IgE was higher in nsLTP-sensitized patients than in nsLTP-nonsensitized patients. Based on Cohen’s Kappa and McNemar tests, the results of allergen extract and component analysis tests do not always agree, especially when we determine the sensitization to allergen sources containing nsLTPs. Molecular allergen component analysis could be the first choice in determining detailed sensitization to nsLTPs in patients who experienced anaphylaxis of unknown origin. Full article
(This article belongs to the Special Issue Molecular Therapeutic Strategies in Allergic Diseases)
Show Figures

Figure 1

Figure 1
<p>The prevalence of sensitization of the whole allergen source (<b>a</b>) and their separate molecular components (<b>b</b>) in studied group. Cases of sensitization indicated in both parts—%.</p>
Full article ">Figure 2
<p>The prevalence of sensitization to nsLTP-containing allergen sources (<b>a</b>) and to different nsLTPs (<b>b</b>). Data presented as percentage of the whole group (<b>a</b>) and percentage of nsLTP-sensitized subjects (<b>b</b>).</p>
Full article ">Figure 3
<p>The amount of total IgE in nsLTP-sensitized subjects or without sensitization to nsLTPs (minimum data value, lower quartile value, median value, upper quartile value, maximum data value, outliers).</p>
Full article ">Figure 4
<p>The sensitization profiles to nsLTP components. Different patients indicated as (<b>A</b>–<b>O</b>). The concentration of sIgE up to 5 kUA/L (<b>A</b>–<b>I</b>), up to 15 kUA/L (<b>J</b>–<b>L</b>), and up to 50 (<b>M</b>–<b>O</b>).</p>
Full article ">
17 pages, 4126 KiB  
Article
In Vivo Induction of Leukemia-Specific Adaptive and Innate Immune Cells by Treatment of AML-Diseased Rats and Therapy-Refractory AML Patients with Blast Modulating Response Modifiers
by Michael Atzler, Tobias Baudrexler, Daniel Christoph Amberger, Nicole Rogers, Alexander Rabe, Joerg Schmohl, Ruixiao Wang, Andreas Rank, Olga Schutti, Klaus Hirschbühl, Marit Inngjerdingen, Diana Deen, Britta Eiz-Vesper, Christoph Schmid and Helga Maria Schmetzer
Int. J. Mol. Sci. 2024, 25(24), 13469; https://doi.org/10.3390/ijms252413469 - 16 Dec 2024
Viewed by 344
Abstract
There is a high medical need to develop new strategies for the treatment of patients with acute myeloid leukemia (AML) refractory to conventional therapy. In vitro, the combinations of the blast-modulatory response modifiers GM-CSF + Prostaglandin E1, (summarized as Kit M) have been [...] Read more.
There is a high medical need to develop new strategies for the treatment of patients with acute myeloid leukemia (AML) refractory to conventional therapy. In vitro, the combinations of the blast-modulatory response modifiers GM-CSF + Prostaglandin E1, (summarized as Kit M) have been shown to convert myeloid leukemic blasts into antigen-presenting dendritic cells of leukemic origin (DCleu) that were able to (re-)activate the innate and adaptive immune system, direct it specifically against leukemic blasts, and induce memory cells. This study aimed to investigate the immune modulatory capacity and antileukemic efficacy of Kit M in vivo. Brown Norway rats suffering from AML were treated with Kit M (twofold application). Blasts and immune cells were monitored in peripheral blood (PB) and spleen. Upon the observation of promising immune modulatory effects in the treated animals, two patients with AML refractory to multiple lines of therapy were offered treatment with Kit M on an individualized basis. Safety, as well as immunological and clinical effects, were monitored. Samples obtained from a third patient in similar clinical conditions not receiving Kit M were used as controls for immune monitoring tests. Animal experiments: Drugs were well tolerated by the treated animals. After 9 days of treatment, DCleu and memory-like T cells increased in the peripheral blood, whereas regulatory T cells, especially blasts, decreased in treated as compared to untreated control animals. Clinical courses: No severe side effects were observed. In patient 1482, PB blasts remained under the detection threshold during 27 days of treatment, thrombocytes were normalized, and (leukemia specific) immune effector cells of the adaptive and innate immune system increased up to 800-fold compared to the start of treatment. Patient 1601 responded with a 12% reduction in blasts in PB immediately after Kit M treatment. Several subtypes of (leukemia-specific) immune effector cells in PB increased up to four-fold during the 19 days of treatment. In contrast, immune-reactive cells decreased under mild chemotherapy in the PB of control patient 1511 with comparably refractory AML. Within the limitation of low numbers in both animal experiments and clinical applications, our data suggest that Kit M treatment of AML-diseased rats and patients is feasible and may induce leukemia-specific immune reactions and clinical improvement. A larger series and a prospective clinical trial will be required to confirm our observations. Beyond optimized doses and schedules of the applied compounds, the combination with other antileukemic strategies or the application of Kit M in less proliferative stages of the myeloid diseases need to be discussed. If effects are confirmed, the concept may add to the armamentarium of treatments for highly aggressive blood cancer. Full article
(This article belongs to the Special Issue Drug-Induced Modulation and Immunotherapy of Leukemia)
Show Figures

Figure 1

Figure 1
<p>Treatment of leukemia-diseased rats with Kit M induced DC/DC<sub>leu</sub> and DC/DC<sub>leu</sub>-activated immune-reactive and memory-like T cells and reduced regulatory T cells and blasts, preferentially in PB/spleen. Cell subtypes are given in <a href="#app1-ijms-25-13469" class="html-app">Supplementary Table S1</a>.</p>
Full article ">Figure 2
<p><b>Clinical course of disease of P1482 following low-dose chemotherapy and Kit M treatment.</b> Chemotherapy (hydroxycarbamide, cytarabine) was given from the start of observation to day 11. Kit M treatment was between day 11 and 38; no treatment was from day 38 to the end of observation. Blood cells (thrombocytes, hemoglobin, neutrophils, blasts) in peripheral blood (PB) and frequencies of BM blasts and leukocytes/white blood cells (WBCs) are given. ↑ Timepoints of erythrocyte transfusions; ↓ timepoints of thrombocyte transfusions. Immune monitoring at defined timepoints showed, in contrast to the samples obtained before treatment and from the patient without Kit M treatment, a continuous increase in DCs and proliferating CD8<sup>+</sup> T cells, of T<sub>non-naive</sub> (and a decrease in T<sub>naive</sub>) of the CD8 and CD4-lines, of Th<sub>1</sub>+ and Th<sub>17</sub>+, CD4<sup>+</sup> T cells, and of Bcell<sub>memory</sub> over the 4-week Kit M treatment. The same was true for T<sub>cm</sub> and T<sub>em</sub> of the CD8 and CD4-lines and for frequencies of NK cells (either CD161<sup>+</sup> or CD56<sup>+</sup>), of CIK cells (either CD161<sup>+</sup> or CD56<sup>+</sup>), Can be removed and iNKT cells (of the NK- as well as the CD3-type) (<a href="#ijms-25-13469-f005" class="html-fig">Figure 5</a>A, P1482).</p>
Full article ">Figure 3
<p><b>Clinical course of disease of P1601 during Kit M treatment ± chemotherapy.</b> No treatment was given between day 1 and 9, Kit M treatment was from day 9 to 26, and chemotherapeutical treatment (hydroxycarbamide) was from day 19 to 29. Patients’ pneumonia was additionally treated daily by prednisolone on day 12–14. Blood cells (thrombocytes, hemoglobin, neutrophils, blasts) in peripheral blood (PB) and leukocytes/white blood cells (WBCs) are given. ↑ Timepoints of erythrocyte transfusions; ↓ timepoints of thrombocyte-transfusions; <span class="html-fig-inline" id="ijms-25-13469-i001"><img alt="Ijms 25 13469 i001" src="/ijms/ijms-25-13469/article_deploy/html/images/ijms-25-13469-i001.png"/></span> pleural punctures.</p>
Full article ">Figure 4
<p><b>Clinical course of disease of P1511 during chemotherapy (control without Kit M).</b> Chemotherapy (cytarabine, midostaurin) was given from the start to the end of observation. Blood cells (thrombocytes, hemoglobin, blasts) in peripheral blood (PB) and leukocytes/white blood cells (WBCs) are given. ↑ Timepoints of erythrocyte transfusion; ↓ timepoints of thrombocyte transfusion.</p>
Full article ">Figure 5
<p><b>Immune monitoring under the treatment of therapy refractory patients (P1482, P1601) with Kit M compared to a control patient (P1511).</b> All values in the course of the disease are given as ‘fold change’ values, referring to the value at the beginning of observation. P1511: chemotherapy during the whole observation time. P1482: low-dose chemotherapy from day 1 to 11, Kit M treatment between day 11 and 38, no treatment from day 38 to the end of observation. P1601: no treatment between day 1 and 9, Kit M treatment from day 9 to 26, chemotherapy from day 19 to 29. * Application of prednisolone from day 12 to 14. Details and abbreviations on cellular subtypes are given in <a href="#app1-ijms-25-13469" class="html-app">Supplementary Table S2</a>. Details on individual treatments are provided in <a href="#app1-ijms-25-13469" class="html-app">Supplementary Table S3</a>. (<b>A</b>) Effects of Kit M treatment on patients’ DCs, cells of adaptive immunity, memory cells, and cells of innate immunity in the course of the disease. (<b>B</b>) Effects of Kit M treatment (P1482, P1601) (vs. no Kit M treatment (P1511)) on the provision of leukemia-specific cells.</p>
Full article ">Figure 5 Cont.
<p><b>Immune monitoring under the treatment of therapy refractory patients (P1482, P1601) with Kit M compared to a control patient (P1511).</b> All values in the course of the disease are given as ‘fold change’ values, referring to the value at the beginning of observation. P1511: chemotherapy during the whole observation time. P1482: low-dose chemotherapy from day 1 to 11, Kit M treatment between day 11 and 38, no treatment from day 38 to the end of observation. P1601: no treatment between day 1 and 9, Kit M treatment from day 9 to 26, chemotherapy from day 19 to 29. * Application of prednisolone from day 12 to 14. Details and abbreviations on cellular subtypes are given in <a href="#app1-ijms-25-13469" class="html-app">Supplementary Table S2</a>. Details on individual treatments are provided in <a href="#app1-ijms-25-13469" class="html-app">Supplementary Table S3</a>. (<b>A</b>) Effects of Kit M treatment on patients’ DCs, cells of adaptive immunity, memory cells, and cells of innate immunity in the course of the disease. (<b>B</b>) Effects of Kit M treatment (P1482, P1601) (vs. no Kit M treatment (P1511)) on the provision of leukemia-specific cells.</p>
Full article ">
11 pages, 2821 KiB  
Article
Overweight and Obesity Contribute to Inflammation and Reduction in Mean Corpuscular Volume and Mean Corpuscular Hemoglobin in Schoolchildren
by Bárbara Leles Fernandes, Alexandre Wallace Dias Cozer, Filipe Caldeira Vasconcelos Souza, Luana Dias Santiago, Marlucy Rodrigues Lima, Pauline Martins Leite, Alda Maria Soares Silveira, Barbara Nery Enes, Marcelo Henrique Fernandes Ottoni, Rafael Silva Gama and Thalisson Artur Ribeiro Gomides
Obesities 2024, 4(4), 524-534; https://doi.org/10.3390/obesities4040041 - 13 Dec 2024
Viewed by 398
Abstract
Background: The inflammation associated with overweight and obesity seems to alter iron metabolism, but there are few studies evaluating those conditions in children. Thus, we aimed to evaluate the leukometric, immunological, and hematimetric parameters of overweight and obese schoolchildren. Methods: This is a [...] Read more.
Background: The inflammation associated with overweight and obesity seems to alter iron metabolism, but there are few studies evaluating those conditions in children. Thus, we aimed to evaluate the leukometric, immunological, and hematimetric parameters of overweight and obese schoolchildren. Methods: This is a cross-sectional study in which 39 children living in Chonim de Cima (Brazil) underwent anthropometric, hematological, and immunological assessments. The evaluated parameters were compared between the study group (overweight/obesity, n = 15) and the control group (n = 24). Unpaired t-test, Mann–Whitney test, and linear regression were used for statistical tests, and the panoramic profile was used to illustrate differences between groups. Results: The study group had lower mean corpuscular volume (MCV) and mean corpuscular hemoglobin (MCH) and higher TNF levels compared to the control group. Positive correlations were observed between BMI-for-age percentile and total leukocytes (r = 0.1493; p = 0.0151) or neutrophils (r = 0.1395; p = 0.0192). Negative correlations between the BMI-for-age percentile and MCV (r = 0.1464; p = 0.0162) and MCH (r = 0.1460; p = 0.0164) were found. Furthermore, through the panoramic profile, it was noted that the study group had a higher frequency of individuals with high levels of TNF and lower frequencies of individuals with increased hemoglobin and serum iron. Conclusions: Our data suggest that overweight and obesity contribute to a pro-inflammatory context (leukocytes, neutrophils, and TNF) and MCV and MCH reduction in schoolchildren. Full article
Show Figures

Figure 1

Figure 1
<p>The study group presents differences in hematimetric indices compared to the control group. The study group (overweight and obese) has lower Mean Corpuscular Volume (<b>A</b>) and lower Mean Corpuscular Hemoglobin (<b>B</b>) compared to the control group. The differences between the groups (<b>A</b>,<b>B</b>) were considered statistically significant if <span class="html-italic">p</span> &lt; 0.05 (Unpaired <span class="html-italic">t</span>-test). * Statistically significant differences.</p>
Full article ">Figure 2
<p>Comparison of serum levels of TNF, IFN-γ, and IL-10 between the control group and the study group (overweight and obese). Serum TNF levels are higher in schoolchildren in the study group (overweight and obese) compared to the control group (<b>A</b>). There were no statistically significant differences between serum levels of IFN-γ and IL-10 between the groups evaluated (<b>B</b>,<b>C</b>). The Mann–Whitney test was used for the statistical evaluation. * Values of <span class="html-italic">p</span> &lt; 0.05 were considered significant.</p>
Full article ">Figure 3
<p>Positive correlation between BMI-for-age percentile and leukocytes in schoolchildren. (<b>A</b>) Positive correlation between BMI-for-age percentile and total leukocytes in schoolchildren. (<b>B</b>) Positive correlation between BMI-for-age percentile and neutrophils in schoolchildren. White circles represent individuals in the control group, and red circles represent individuals in the study group (overweight and obese). <span class="html-italic">p</span>-values &lt; 0.05 indicate a statistically significant correlation (linear regression).</p>
Full article ">Figure 4
<p>Negative correlation between BMI-for-age and hematimetric parameters. Negative correlation between BMI-for-age percentile and Mean Corpuscular Volume (<b>A</b>) and Mean Corpuscular Hemoglobin (<b>B</b>) in schoolchildren. White circles represent individuals in the control group, and red circles represent individuals in the study group (overweight and obese). The correlations (<b>A</b>,<b>B</b>) were statistically significant if <span class="html-italic">p</span> &lt; 0.05 (linear regression).</p>
Full article ">Figure 5
<p>Analysis of the correlation between serum iron and mean corpuscular volume (MCV). Peripheral blood was collected from all study participants for analysis. Eutrophic (control) (white circles) and overweight and obese (study group) (red circles) schoolchildren were analyzed. Serum iron and mean corpuscular volume were counted using routine biochemical reactions and hematologic cell counters, respectively. The correlation between serum iron and MCV was then calculated using linear regression. (<b>A</b>) No correlation between MCV and Serum Iron among all individuals evaluated. (<b>B</b>) Positive Correlation between MCV and Serum Iron in the study group (overweight and obese). A significant correlation was considered when <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Panoramic profile of immune and hematologic characteristics of the control group and the study group. (<b>A</b>) Matrix for individuals with low levels—LL (white boxes) and high levels—HL (black boxes) of the evaluated parameters (TNF, Fe, IL-10, IFN-γ, and Hb). Below each matrix (control group and study group), the number of HL (Nº HL) and frequency of HL (% HL) individuals are observed (black boxes). (<b>B</b>) On the side, you can see a table of arrows for comparing the profiles presented by the groups, where the upward arrow (↑) indicates that the frequency of HL is greater than 50%, and the downward arrow (↓) indicates that the frequency of HL is less than 50%.</p>
Full article ">
15 pages, 2551 KiB  
Article
Oral Vaccine Formulation for Immunocastration Using a Live-Attenuated Salmonella ΔSPI2 Strain as an Antigenic Vector
by Sergio A. Bucarey, Lucy D. Maldonado, Francisco Duarte, Alejandro A. Hidalgo and Leonardo Sáenz
Vaccines 2024, 12(12), 1400; https://doi.org/10.3390/vaccines12121400 - 12 Dec 2024
Viewed by 405
Abstract
Immunization against Gonadotropin-Releasing Hormone (GnRH) has been successfully explored and developed for the parenteral inoculation of animals, aimed at controlling fertility, reducing male aggressiveness, and preventing boar taint. Although effective, these vaccines may cause adverse reactions at the injection site, including immunosuppression and [...] Read more.
Immunization against Gonadotropin-Releasing Hormone (GnRH) has been successfully explored and developed for the parenteral inoculation of animals, aimed at controlling fertility, reducing male aggressiveness, and preventing boar taint. Although effective, these vaccines may cause adverse reactions at the injection site, including immunosuppression and inflammation, as well as the involvement of laborious and time-consuming procedures. Oral vaccines represent an advancement in antigen delivery technology in the vaccine industry. In this study, a Salmonella enterica serovar Typhimurium (S. Typhimurium) mutant lacking the pathogenicity island 2 (S. Typhimurium ΔSPI2) was used as a vehicle and mucosal adjuvant to deliver two genetic constructs in an attempt to develop an oral immunological preparation against gonadotropin hormone-releasing hormone (GnRH). S. Typhimurium ΔSPI2 was transformed to carry two plasmids containing a modified GnRH gene repeated in tandem (GnRXG/Q), one under eukaryotic expression control (pDNA::GnRXG/Q) and another under prokaryotic expression control (pJexpress::GnRXG/Q). A group of three male BALB/c mice were orally immunized and vaccination-boosted 30 days later. The oral administration of S. Typhimurium ΔSPI2 transformed with both plasmids was effective in producing antibodies against GnRXG/Q, leading to a decrease in serum testosterone levels and testicular tissue atrophy, evidenced by a reduction in the transverse tubular diameter of the seminiferous tubules and a decrease in the number of layers of the seminiferous epithelium in the testes of the inoculated mice. These results suggest that S. Typhimurium ΔSPI2 can be used as a safe and simple system to produce an oral formulation against GnRH and that Salmonella-mediated oral antigen delivery is a novel, yet effective, alternative to induce an immune response against GnRH in a murine model, warranting further research in other animal species. Full article
(This article belongs to the Special Issue Research on Immune Response and Vaccines: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Survival percentage of <span class="html-italic">S.</span> Typhimurium 14028s and <span class="html-italic">S.</span> Typhimurium 14028s ∆SPI2 in murine RAW264.7 macrophages. (<b>A</b>) Survival at 24 h post internalization. (<b>B</b>) Survival at 48 h post internalization. An intracellular survival assay was performed, and intracellular bacteria were recovered at the indicated times. Survival percentages were calculated from the bacteria recovered at t<sub>0</sub>. The bars represent the standard deviation (SD). The assays were performed in triplicate. (*) Indicates a significant difference with <span class="html-italic">p</span> &lt; 0.05 for the attenuated 14028s ∆SPI2 strain compared to the wild-type 14028s strain.</p>
Full article ">Figure 2
<p>Detection of GnRXG/Q by Western blot. An isolated colony of the recombinant strain <span class="html-italic">S.</span> Typhimurium 14028s ∆SPI2/pJEX was cultured in LB broth and induced with IPTG. The purified GnRXG/Q antigen (~30 kDa) was used as a positive control. The extracts were analyzed by Western blot. The band was photographed under a transilluminator. Lane 1 corresponds to the positive control (GnRXG/Q purified protein), and Lane 2 corresponds to the protein expressed in the recombinant strain <span class="html-italic">S.</span> Typhimurium 14028s ∆SPI2/pJEX.</p>
Full article ">Figure 3
<p>IgG anti-GnRXG/Q antibody levels expressed as arithmetic means of absorbance values measured at 450 nm on days 0, 15, 30, 45, and 60 post immunization. Male BALB/c mice (<span class="html-italic">n</span> = 3) were orally immunized on days 0 and 30 with 10<sup>9</sup> CFU of <span class="html-italic">S.</span> Typhimurium 14028s ΔSPI2/pDNAX3::GnRXG/Q (pDNAX3) with 10<sup>9</sup> CFU of <span class="html-italic">S.</span> Typhimurium 14028s ΔSPI2/pJEX::GnRXG/Q (pJEX), subcutaneously with 100 µg of GnRXG/Q purified by affinity chromatography using aluminum as an adjuvant (Control +), and orally with 200 µL of PBS (Control −). IgG anti-GnRXG/Q levels were measured by ELISA on days 0, 15, 30, 45, and 60 post immunization. (*) Indicates a significant difference with <span class="html-italic">p</span> &lt; 0.05 for any of the treated groups compared with the negative control.</p>
Full article ">Figure 4
<p>Serum testosterone concentration of the study animals, based on a standard curve. (<b>A</b>) Day 15 post immunization. (<b>B</b>) Day 30 post immunization. (<b>C</b>) Day 45 post immunization. Male BALB/c mice (<span class="html-italic">n</span> = 3) were orally immunized on days 0 and 30 with 10<sup>9</sup> CFU of <span class="html-italic">S.</span> Typhimurium 14028s ΔSPI2/pDNAX3::GnRXG/Q (pDNAX3) with 10<sup>9</sup> CFU of <span class="html-italic">S.</span> Typhimurium 14028s ΔSPI2/pJEX::GnRXG/Q (pJEX), subcutaneously with 100 µg of GnRXG/Q purified by affinity chromatography using aluminum as an adjuvant (Control +), and orally with 200 µL of PBS (Control −). Serum testosterone levels were measured by commercial ELISA (IBL-AMERICA kit) on days 15, 30, and 45 post immunization. (***) Indicates a difference of <span class="html-italic">p</span> &lt; 0.001 compared to the negative control. (**) Indicates a difference of <span class="html-italic">p</span> &lt; 0.01 compared to the negative control. (*) Indicates a difference of <span class="html-italic">p</span> &lt; 0.05 compared to the negative control.</p>
Full article ">Figure 5
<p>Histological sections of seminiferous tubules, stained with hematoxylin-eosin, observed at 40×. The images correspond to the negative control group immunized with 200 μL of oral PBS (<b>A</b>), the positive control group immunized subcutaneously with 100 μg of GnRXG/Q purified by aluminum affinity chromatography as an adjuvant (<b>B</b>), the experimental group orally immunized with strain <span class="html-italic">S.</span> Typhimurium 14028s ∆SPI2/pDNAX3, (<b>C</b>) and the experimental group immunized orally with strain <span class="html-italic">S.</span> Typhimurium 14028s ∆SPI2/pJEX (<b>D</b>). The black lines show the transverse tubular diameter, represented by the means of two diametrically opposed measurements. The black solid bar at the lower right corner of each figure is 100 µm long.</p>
Full article ">
8 pages, 479 KiB  
Article
Direct Immunofluorescence in Oral Lichen Planus and Related Lesions: Sensitivity, Specificity, and Diagnostic Accuracy in a Single Diagnostic Center in Poland
by Katarzyna Osipowicz, Konrad Szymański, Ewelina Pietrzyk, Emilia Milczarek, Cezary Kowalewski, Renata Górska and Katarzyna Woźniak
Dent. J. 2024, 12(12), 396; https://doi.org/10.3390/dj12120396 - 6 Dec 2024
Viewed by 434
Abstract
Objectives: Our study aimed to establish the basic reliability parameters of direct immunofluorescence test results in patients with oral lichen planus. Methods: We conducted an evaluation of individual antibody classes in the DIF and ELISA (BP180 antigen), comparing these results with the classical [...] Read more.
Objectives: Our study aimed to establish the basic reliability parameters of direct immunofluorescence test results in patients with oral lichen planus. Methods: We conducted an evaluation of individual antibody classes in the DIF and ELISA (BP180 antigen), comparing these results with the classical histopathological (HP) examination in a group of patients treated within the standard healthcare in our clinic. Results: Among 66 participants with oral changes indicative of LP, only 50% received histopathological confirmation of the LP diagnosis. Among those with a DIF profile entirely typical for LP (C3+, F+), 57.1% had a positive HP result. Fibrinogen deposits were identified in 42.4% and 36.4% of individuals with positive HP results for F1 and F2, respectively; 78.8% of patients with negative HP and 57.6% with positive HP exhibited no fibrinogen deposits. Simultaneous positivity for F1 and F2 occurred in all cases where F1 was positive. HP confirmed positive DIF for C3 in 50% of cases. Fibrinogen deposits demonstrated the highest diagnostic accuracy (61%). Sensitivity and specificity for fibrinogen deposits were 36% and 42% for F1 and 79% and 82% for F2. The positive predictive values were 67% for F1 and 67% for F2, while the negative predictive values were 58% for F1 and 56% for F2. Overall diagnostic accuracy was reported at 61% for F1 and 59% for F2. Conclusions: Our data indicate the complementarity of HP and immunological test results and the necessity of using both methods together in cases of doubt. Full article
(This article belongs to the Section Dental Education)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>–<b>h</b>). Upper row—LP confirmed in histopathology: (<b>a</b>) strong deposition of F1 in a linear pattern along the BMZ (DIF, ×200), (<b>b</b>) strong deposition of F2 in a linear pattern along the BMZ (DIF, ×200), (<b>c</b>) deposition of complement C3 in a granular pattern along the BMZ (DIF, ×400). Middle row—LP-like lesions without LP histopathology: (<b>d</b>) strong deposition of F1 in a linear pattern along the BMZ (DIF, ×200), (<b>e</b>) strong deposition of F2 in a linear pattern along the BMZ (DIF, ×200), (<b>f</b>) deposition of complement C3 in a granular pattern along the BMZ (DIF, ×400). Lower row—MMP patient with LP-like lesions: (<b>g</b>) strong deposition of IgG in a linear pattern along the BMZ (DIF, ×200), (<b>h</b>) deposition of linear complement C3 along the BMZ (DIF, ×200). LP-lichen planus, DIF—direct immunofluorescence, F1-fibrinogen 1, F2—fibrinogen 2, BMZ—basement membrane zone, MMP—mucous membrane pemphigoid.</p>
Full article ">
21 pages, 11820 KiB  
Article
The Impact of Resident Adipose Tissue Macrophages on Adipocyte Homeostasis and Dedifferentiation
by Julia Neugebauer, Nora Raulien, Lilli Arndt, Dagmar Akkermann, Constance Hobusch, Andreas Lindhorst, Janine Fröba and Martin Gericke
Int. J. Mol. Sci. 2024, 25(23), 13019; https://doi.org/10.3390/ijms252313019 - 4 Dec 2024
Viewed by 464
Abstract
Obesity is concurrent with immunological dysregulation, resulting in chronic low-grade inflammation and cellular dysfunction. In pancreatic islets, this loss of function has been correlated with mature β-cells dedifferentiating into a precursor-like state through constant exposure to inflammatory stressors. As mature adipocytes likewise have [...] Read more.
Obesity is concurrent with immunological dysregulation, resulting in chronic low-grade inflammation and cellular dysfunction. In pancreatic islets, this loss of function has been correlated with mature β-cells dedifferentiating into a precursor-like state through constant exposure to inflammatory stressors. As mature adipocytes likewise have the capability to dedifferentiate in vitro and in vivo, we wanted to analyze this cellular change in relation to adipose tissue (AT) inflammation and adipose tissue macrophage (ATM) activity. Using our organotypic AT explant culture method combined with a double-reporter mouse model for labeling ATMs and mature adipocytes, we were able to visualize and quantify dedifferentiated fat (DFAT) cells in AT explants. Preliminary testing showed increased dedifferentiation after tamoxifen (TAM) stimulation, making TAM-dependent lineage-tracing models unsuitable for quantification of naturally occurring DFAT cells. The regulatory role of ATMs in adipocyte dedifferentiation was shown through macrophage depletion using Plexxicon 5622 or clodronate liposomes, which significantly increased DFAT cell levels. Subsequent bulk RNA sequencing of macrophage-depleted explants revealed enrichment of the tumor necrosis factor α (TNFα) signaling pathway as well as downregulation of associated genes. Direct stimulation with TNFα decreased adipocyte dedifferentiation, while application of a TNFα-neutralizing antibody did not significantly alter DFAT cell levels. Our findings suggest a regulatory role of resident ATMs in maintaining the mature adipocyte phenotype and preventing excessive adipocyte dedifferentiation. The specific regulatory pathways as well as the impact that DFAT cells might have on ATMs, and vice versa, are subject to further investigation. Full article
(This article belongs to the Special Issue Advance of Cell Metabolism in Endocrine Diseases)
Show Figures

Figure 1

Figure 1
<p>Adipocyte dedifferentiation in AT explant culture. Epididymal white adipose tissue (WAT) explants from homozygous MacFat mice (n = 11–14, N = 5–6) were cultivated under standard cultivation conditions for 10 days. (<b>A</b>) Microscopy analysis of adipose tissue (AT) explants from MacFat mice (red: adipocytes; green: adipose tissue macrophages (ATMs); arrow: DFAT cell) in explant culture at different timepoints. (<b>B</b>) Representative flow cytometry dot plots showing increase in dedifferentiated fat (DFAT) cells (GFP<sup>−</sup>; tdTo<sup>+</sup>) (upper row) and a shift from anti- (CD11c<sup>−</sup>; CD301<sup>+</sup>) to pro-inflammatory (CD11c<sup>+</sup>; CD301<sup>−</sup>) ATM phenotype (lower row). (<b>C</b>) Proportion of live cells (4′,6-diamidino-2-phenylindole (DAPI)-negative, n = 14, N = 6) from single cells. (<b>D</b>,<b>E</b>) Proportion of (<b>D</b>) ATMs (GFP<sup>+</sup>; tdTo<sup>−</sup>, n = 14, N = 6) and (<b>E</b>) DFAT cells (GFP<sup>−</sup>; tdTo<sup>+</sup>, n = 14, N = 6) from the stromal vascular fraction (SVF). (<b>F</b>) Ratio of pro- (M1) to anti-inflammatory (M2) macrophages (n = 11, N = 5). Red line marks a balanced 1:1 ratio. (<b>G</b>,<b>H</b>) Proportion of (<b>G</b>) pro- (M1, CD11c<sup>+</sup>; CD301<sup>−</sup>, n = 11, N = 5) and (<b>H</b>) anti-inflammatory (M2, CD11c<sup>−</sup>; CD301<sup>+</sup>, n = 11, N = 5) macrophages from ATMs. Data shown as means ± SEM. Scale bars = 100 µm. ** <span class="html-italic">p</span>-value &lt; 0.01; *** <span class="html-italic">p</span>-value &lt; 0.001; **** <span class="html-italic">p</span>-value &lt; 0.0001, ns means not significantly.</p>
Full article ">Figure 2
<p>Effects of tamoxifen (TAM) on adipocyte dedifferentiation and ATM phenotype, comparing homo- and heterozygous MacFat mice. Epididymal WAT explants from homo- and heterozygous MacFat mice (n = 4–6, N = 3 per genotype) were stimulated with 1 or 10 µM TAM or 10 µL ethanol (EtOH, control) for 10 days. (<b>A</b>,<b>B</b>) Microscopic imaging of TAM-stimulated (10 µM TAM) and non-stimulated (EtOH) AT explants from (<b>A</b>) hetero- and (<b>B</b>) homozygous MacFat mice (red: adipocytes; green: ATMs; arrow_ DFAT cells) on day 10 in explant culture. (<b>C</b>,<b>D</b>) Representative flow cytometry dot plots of (<b>C</b>) hetero- and (<b>D</b>) homozygous AT explants showing DFAT cell (GFP<sup>−</sup>; tdTo<sup>+</sup>) increase (upper rows) and a shift from anti- (CD11c<sup>−</sup>; CD301<sup>+</sup>) to pro-inflammatory (CD11c<sup>+</sup>; CD301<sup>−</sup>) ATM phenotype (lower rows) under TAM stimulation on day 10 in explant culture. (<b>E</b>,<b>F</b>) Proportion of DFAT cells (GFP<sup>−</sup>; tdTo<sup>+</sup>) from SVF at different timepoints during cultivation of (<b>E</b>) hetero- (n = 5–6) and (<b>F</b>) homozygous (n = 4–6) AT explants under TAM stimulation. (<b>G</b>,<b>H</b>) Proportion of ATMs (GFP<sup>+</sup>; tdTo<sup>−</sup>) from SVF on different timepoints during cultivation of (<b>G</b>) hetero- (n = 5–6) and (<b>H</b>) homozygous (n = 4–6) AT explants under TAM stimulation. (<b>I</b>,<b>J</b>) Ratio of pro- (M1) to anti-inflammatory (M2) macrophages at different timepoints during cultivation of (<b>I</b>) hetero- (n = 5–6) and (<b>J</b>) homozygous (n = 4–6) AT explants under TAM stimulation. Red line marks a balanced 1:1 ratio. Data represented as means ± SEM. Scale bars = 100 µm. * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01; *** <span class="html-italic">p</span>-value &lt; 0.001, ns means not significantly.</p>
Full article ">Figure 3
<p>ATM depletion through Plexxicon 5622 (PLX) or clodronate liposomes amplifies DFAT cell levels. Epididymal WAT explants from homozygous MacFat mice (n = 6, N = 3 per condition) were cultivated under ATM depleting conditions with PLX or clodronate liposomes for 10 days. (<b>A</b>,<b>B</b>) Microscopic imaging on day 10 of cultivation of AT explants (red: adipocytes; green: ATMs; arrow: DFAT cells) stimulated with (<b>A</b>) dimethyl sulfoxide (DMSO, ctrl)/40 µM PLX or (<b>B</b>) PBS liposomes (ctrl)/clodronate liposomes. (<b>C</b>,<b>D</b>) Representative flow cytometry dot plots showing DFAT cell (GFP<sup>−</sup>; tdTo<sup>+</sup>) increase on day 10 after (<b>C</b>) PLX or (<b>D</b>) clodronate liposome stimulation. (<b>E</b>,<b>F</b>) Decreasing proportion of ATMs (GFP<sup>+</sup>; tdTo<sup>−</sup>) from SVF with (<b>E</b>) PLX or (<b>F</b>) clodronate liposomes. (<b>G</b>,<b>H</b>) Increasing numbers of DFAT cells (GFP<sup>−</sup>; tdTo<sup>+</sup>) coinciding with (<b>G</b>) PLX or (<b>H</b>) clodronate liposome stimulation. Data shown as means ±SEM. Scale bars = 100 µm. * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01; *** <span class="html-italic">p</span>-value &lt; 0.001; ns means not significantly.</p>
Full article ">Figure 4
<p>Bulk RNA sequencing of macrophage-depleted AT explants. Epididymal WAT explants from homozygous MacFat mice (n = 4, N = 2) were cultivated under ATM-depleting conditions with clodronate liposomes for 10 days and analyzed via bulk RNA sequencing. (<b>A</b>) Pathway analysis of the gene expression profile in macrophage-depleted AT explants. (<b>B</b>) Heat map of genes coding for mature adipocytes. (<b>C</b>) Heat map of genes coding for inflammatory cytokines, chemokines, and enzymes regulated through the TNFα signaling pathway.</p>
Full article ">Figure 5
<p>TNFα signaling affects adipocyte dedifferentiation in explant culture. Epididymal WAT explants from homozygous MacFat mice (n = 8, N = 4) were stimulated with 10 or 50 ng/mL TNFα and cultivated for 10 days. (<b>A</b>) Microscopy imaging of TNFα-stimulated and non-stimulated AT explants (red: adipocytes; green: ATMs; arrow: DFAT cells) on day 10 of cultivation. (<b>B</b>) Representative flow cytometry dot plots showing DFAT cells (GFP<sup>−</sup>; tdTo<sup>+</sup>) and ATMs (GFP<sup>+</sup>; tdTo<sup>−</sup>) (upper row) as well as the ratio of pro- (CD11c<sup>+</sup>; CD301<sup>−</sup>) to anti-inflammatory (CD11c<sup>−</sup>; CD301<sup>+</sup>) macrophages (lower row). (<b>C</b>) Proportion of DFAT cells (GFP<sup>−</sup>; tdTo<sup>+</sup>) from SVF at different timepoints during cultivation after TNFα stimulation. (<b>D</b>) Proportion of ATMs (GFP<sup>+</sup>; tdTo<sup>−</sup>) from SVF at different timepoints during cultivation after TNFα stimulation. (<b>E</b>) Ratio of pro- (M1) to anti-inflammatory (M2) macrophages at different timepoints during cultivation after TNFα stimulation. Red line indicates a balanced 1:1 ratio. Data shown as means ± SEM. Scale bars = 100 µm. * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01, ns means not significant.</p>
Full article ">
14 pages, 787 KiB  
Article
Soluble P-Selectin as an Indicator of Cutaneous Microangiopathy in Uncomplicated Young Patients with Type 1 Diabetes
by Jolanta Neubauer-Geryk, Małgorzata Myśliwiec, Katarzyna Zorena and Leszek Bieniaszewski
Life 2024, 14(12), 1587; https://doi.org/10.3390/life14121587 - 2 Dec 2024
Viewed by 467
Abstract
This study aimed to analyze the relationship between cutaneous microcirculation reactivity, retinal circulation, macrocirculation function, and specific adhesion molecules in young patients with uncomplicated type 1 diabetes. Fifty-five patients with type 1 diabetes mellitus (T1DM), aged 8 to 18 years, were divided into [...] Read more.
This study aimed to analyze the relationship between cutaneous microcirculation reactivity, retinal circulation, macrocirculation function, and specific adhesion molecules in young patients with uncomplicated type 1 diabetes. Fifty-five patients with type 1 diabetes mellitus (T1DM), aged 8 to 18 years, were divided into subgroups based on skin microcirculation reactivity. The cutaneous microcirculatory vessels were considered reactive if post-test PORH coverage increased compared to pre-test coverage. Optical coherence tomography (OCT) was conducted to detect early retinopathic changes. Macrocirculation was described using pulsatility indices (PIs) determined for common carotid (CCA) and peripheral arteries of the upper and lower limbs. The ankle–brachial index was also assessed. There were no significant differences in retinal circulation and macrocirculation between the studied subgroups. However, there were significant differences between the various subgroups concerning the age at onset of diabetes and the sP-selectin levels but not ICAM-1 and sVCAM-1. The sP-selectin differences remained true after adjusting for age at onset. The sP-selectin level was significantly higher in the subgroup of patients with non-reactive cutaneous microcirculation. The results of our study indicate that sP-selectin may be considered as an immunological marker for cutaneous abnormalities, which serve as an early indicator of endothelial dysfunction in young patients with type 1 diabetes in the absence of classical complication. Full article
(This article belongs to the Special Issue Management of Patients with Diabetes)
Show Figures

Figure 1

Figure 1
<p>The result of the optical coherence tomography (OCT) examination of a subject with T1DM who participated in the present study. Macula thickness OU—the abbreviation <span class="html-italic">OU</span> is derived from the Latin term oculus uterque, which translates to “both eyes”. Retinal thickness is measured as the distance between the internal limiting membrane (ILM) and the retinal pigment epithelium (RPE)- white arrows. OD—right eye; OS—left eye.</p>
Full article ">
27 pages, 734 KiB  
Review
Hypersensitivity to Lanolin: An Old–New Problem
by Kinga Lis
Life 2024, 14(12), 1553; https://doi.org/10.3390/life14121553 - 26 Nov 2024
Viewed by 421
Abstract
Lanolin is a fatty substance derived from sheep’s fleece. The ancient Greeks used the moisturizing and skin-protective properties of this substance. The technique of industrial production of lanolin was developed in Germany in the 19th century. Since then, this natural wax has become [...] Read more.
Lanolin is a fatty substance derived from sheep’s fleece. The ancient Greeks used the moisturizing and skin-protective properties of this substance. The technique of industrial production of lanolin was developed in Germany in the 19th century. Since then, this natural wax has become an extremely popular base for many different cosmetic and pharmaceutical preparations intended for the treatment and care of the skin. In addition to its medicinal and cosmetic applications, lanolin is also widely used for industrial purposes. Hypersensitivity to lanolin has raised many questions and controversies for almost 100 years. Although lanolin has significant dermoprotective properties and when applied to intact skin without inflammatory changes, it lubricates it, improves its lipid barrier, and maintains proper moisture, it can also cause contact hypersensitivity when in contact with pathologically changed or damaged skin. It can, in the same person, both protect and damage the skin, depending on the condition of the skin to which the cosmetic or medicine containing lanolin is applied. The nature of the observed reactions and the circumstances of their occurrence, as well as the lack of a clear answer to the question of whether this wax causes allergies or not, make this phenomenon one of the so-called dermatological paradoxes. Although unusual reactions to lanolin have been the subject of research for many years, they still raise many questions to which there is still no clear answer. This is mainly due to the imperfection and incompleteness of the available publications. Although many different studies have been published on hypersensitivity to lanolin, most of them are retrospective analyses of the results of routinely performed epidermal patch tests or descriptions of clinical cases. Such reports and analyses, although undoubtedly very important, are a poor tool for assessing the sensitizing potential of lanolin and/or its derivatives. It is difficult to determine the causative factors, to define lanolin allergens, to investigate immunological mechanisms, or to assess the clinical significance of this phenomenon. There is a definite lack of standardized studies on the nature of lanolin hypersensitivity involving well-selected groups of patients and healthy volunteers, which would be conducted in a reproducible manner under laboratory and/or clinical conditions. As of today, lanolin hypersensitivity seems to be both an old and new problem that still remains unresolved. Full article
Show Figures

Figure 1

Figure 1
<p>Commonly used types of products containing lanolin [<a href="#B4-life-14-01553" class="html-bibr">4</a>].</p>
Full article ">
18 pages, 2835 KiB  
Article
Application of the Sponge Model Implants in the Study of Vaccine Memory in Mice Previously Immunized with LBSap
by Mariana Ferreira Lanna, Lucilene Aparecida Resende, Paula Mello De Luca, Wanessa Moreira Goes, Maykelin Fuentes Zaldívar, André Tetzl Costa, Walderez Ornelas Dutra, Alexandre Barbosa Reis, Olindo Assis Martins-Filho, Kenneth Jhon Gollob, Sandra Aparecida Lima de Moura, Edelberto Santos Dias, Érika Michalsky Monteiro, Denise Silveira-Lemos and Rodolfo Cordeiro Giunchetti
Vaccines 2024, 12(12), 1322; https://doi.org/10.3390/vaccines12121322 - 26 Nov 2024
Viewed by 515
Abstract
Background/Objectives: Considering the large number of candidates in vaccine-testing studies against different pathogens and the amount of time spent in the preclinical and clinical trials, there is a pressing need to develop an improved in vivo system to quickly screen vaccine candidates. The [...] Read more.
Background/Objectives: Considering the large number of candidates in vaccine-testing studies against different pathogens and the amount of time spent in the preclinical and clinical trials, there is a pressing need to develop an improved in vivo system to quickly screen vaccine candidates. The model of a polyester–polyurethane sponge implant provides a rapid analysis of the specific stimulus–response, allowing the study of a compartmentalized microenvironment. The sponge implant’s defined measurements were standardized as a compartment to assess the immune response triggered by the vaccinal antigen. The LBSap vaccine (composed of Leishmania braziliensis antigens associated with saponin adjuvant) was used in the sponge model to assess the antigen-specific immunological biomarker, including memory generation after initial contact with the antigen. Methods: Mice strains (Swiss, BALB/c, and C57BL/6) were previously immunized using LBSap vaccine, followed by an antigenic booster performed inside the sponge implant. The sponge implants were assessed after 72 h, and the immune response pattern was analyzed according to leukocyte immunophenotyping and cytokine production. Results: After LBSap vaccination, the innate immune response of the antigenic booster in the sponge implants demonstrated higher levels in the Ly+ neutrophils and CD11c+ dendritic cells with reduced numbers of F4/80+ macrophages. Moreover, the adaptive immune response in Swiss mice demonstrated a high CD3+CD4+ T-cell frequency, consisting of an effector memory component, in addition to a cytoxicity response (CD3+CD8+ T cells), displaying the central memory biomarker. The major cell surface biomarker in the BALB/c mice strain was related to CD3+CD4+ effector memory, while the increased CD3+CD8+ effector memory was highlighted in C57/BL6. The cytokine profile was more inflammatory in Swiss mice, with the highest levels of IL-6, TNF, IFN-g, and IL-17, while the same cytokine was observed in in C57BL/6 yet modulated by enhanced IL-10 levels. Similar to Swiss mice, BALB/c mice triggered an inflammatory environment after the antigenic booster in the sponge implant with the increased levels in the ILL-6, TNF, and IFN-g. Conclusions: The findings emphasized the impact of genetic background on the populations engaged in immune responses, suggesting that this model can be utilized to enhance and track both innate and adaptive immune responses in vaccine candidates. Consequently, these results may inform the selection of the most suitable experimental model for biomolecule testing, taking into account how the unique characteristics of each mouse strain affect the immune response dynamics. Full article
(This article belongs to the Special Issue Research on Immune Response and Vaccines: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Experimental design and study groups. The animals were divided into three control groups and one experimental group, with a total of 5 mice per group. Statistical analyses were performed using ANOVA, followed by Tukey’s test. The implanted sponge discs were removed and sent for immunophenotyping analysis by flow cytometry and for investigation of soluble cytokines.</p>
Full article ">Figure 2
<p>Immunophenotypic profile of leukocytes in sponge implants. The cellular infiltrate removed from the implants was labeled with a monoclonal antibody for quantification by flow cytometry of leukocyte subpopulations (CD45+) of innate immunity cells: neutrophils (LY+), macrophages (F4/80+), and dendritic cells (CD11c+). Data are reported as mean percentage ± standard deviation (<span class="html-italic">n</span> = 5 mice per group). Intragroup and intergroup comparisons were evaluated using ANOVA, followed by Tukey’s test, and significant differences (<span class="html-italic">p</span> &lt; 0.05) are highlighted by connecting lines within the lineage and by the letters “S”, “B”, and “C” for comparisons between Swiss, BALB/c, and C57BL/6 mice, respectively.</p>
Full article ">Figure 3
<p>Immunophenotypic profile of lymphocytes in sponge implants. The cellular infiltrate was quantified by flow cytometry for the TCD3+ CD4+ and TCD3+ CD8+ lymphocyte subpopulations, in addition to the EM effector memory (CD62L+CCR7+) and CM central (CD62L+CCR7+) subpopulations. Data are reported as mean percentage ± standard deviation (<span class="html-italic">n</span> = 5 mice per group). Intragroup and intergroup comparisons were evaluated using ANOVA, followed by Tukey’s test, and significant differences (<span class="html-italic">p</span> &lt; 0.05) are highlighted by connecting lines within the lineage and by the letters “S”, “B”, and “C” for comparisons between Swiss, BALB/c, and C57BL/6 mice, respectively.</p>
Full article ">Figure 4
<p>Soluble cytokines in the microenvironment of sponge implants. Data are reported as mean fluorescence intensity (MFI) ± standard deviation for each cytokine (IL-6, TNF, IFN-γ, IL-10, and IL-17) (<span class="html-italic">n</span> = 5 mice per group). Significant differences (<span class="html-italic">p</span> &lt; 0.05) were evaluated using ANOVA, followed by Tukey’s test, and are represented by connecting lines above the graphs (differences considering the same lineage but different groups) and by the letters “S”, “B”, and “C” for comparisons among Swiss, BALB/c, and C57BL/6 mice, respectively.</p>
Full article ">Figure 5
<p>Ascending biomarker signature in sponge implants. This figure illustrates the frequency of biomarker levels above the global median cutoff for each cell subset (CD45+, Ly+, F4/80+, CD11c+, CD3+CD4+, CD3+CD4+ EM, CD3+ CD8+, CD3+CD8+ CM, and CD3+CD8+ EM) and soluble cytokines (IL-6, TNF, IFN-γ, IL-10, and IL-17) across different experimental groups: S/S (blue), S/I (red), V/S (green), and V/I (purple) for Swiss, BALB/c, and C57BL/6 mice (<span class="html-italic">n</span> = 5 mice per group). Significant differences (<span class="html-italic">p</span> &lt; 0.05) were evaluated using ANOVA, followed by Tukey’s test. Biomarkers with frequencies equal to or above 75% were considered relevant and are highlighted in underlined bold format for emphasis.</p>
Full article ">
19 pages, 1316 KiB  
Review
Synovial Fluid Markers and Extracellular Vesicles in Rheumatoid Arthritis
by Veronika Smolinska, Daniela Klimova, Lubos Danisovic and Stefan Harsanyi
Medicina 2024, 60(12), 1945; https://doi.org/10.3390/medicina60121945 - 26 Nov 2024
Viewed by 599
Abstract
In recent years, numerous potential prognostic biomarkers for rheumatoid arthritis (RA) have been investigated. Despite these advancements, clinical practice primarily relies on autoantibody tests—for rheumatoid factor (RF) and anti-citrullinated protein antibody (anti-CCP)—alongside inflammatory markers, such as the erythrocyte sedimentation rate (ESR) and C-reactive [...] Read more.
In recent years, numerous potential prognostic biomarkers for rheumatoid arthritis (RA) have been investigated. Despite these advancements, clinical practice primarily relies on autoantibody tests—for rheumatoid factor (RF) and anti-citrullinated protein antibody (anti-CCP)—alongside inflammatory markers, such as the erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP). Expanding the repertoire of diagnostic and therapeutic biomarkers is critical for improving clinical outcomes in RA. Emerging evidence highlights the significance of synovial fluid biomarkers, including aggrecan, matrix metalloproteinases, glucosyl-galactosyl-pyridinoline, hyaluronic acid, S100 proteins, calprotectin, and various cytokines, as well as immunological markers. Additionally, specific components of extracellular vesicles, such as non-coding RNAs, heat shock proteins, and lipids, are gaining attention. This review focuses on molecular markers found in synovial fluid and extracellular vesicles, excluding clinical and imaging biomarkers, and explores their potential applications in the diagnosis and management of RA. Full article
(This article belongs to the Section Hematology and Immunology)
Show Figures

Figure 1

Figure 1
<p>Synovial fluid collection, laboratory analysis, and biomarker detection.</p>
Full article ">Figure 2
<p>Healthy joint vs. joint affected by RA.</p>
Full article ">
18 pages, 2149 KiB  
Article
An Analysis of Longan Honey from Taiwan and Thailand Using Flow Cytometry and Physicochemical Analysis
by Lekhnath Kafle and Tandzisile Zine Mabuza
Foods 2024, 13(23), 3772; https://doi.org/10.3390/foods13233772 - 25 Nov 2024
Viewed by 1051
Abstract
The increase in honey fraud in the global market has highlighted the importance of pollen analysis in determining or confirming the botanical and geographical origins of honey. Numerous studies are currently underway to develop efficient and rapid methods for the determination of the [...] Read more.
The increase in honey fraud in the global market has highlighted the importance of pollen analysis in determining or confirming the botanical and geographical origins of honey. Numerous studies are currently underway to develop efficient and rapid methods for the determination of the quality, botanical, and geographical origin of honey. Typically, the physicochemical analysis of honey is used to evaluate its quality and geographical source. In this study, flow cytometry, a technique extensively employed in immunology and hematology, was first applied to analyze and characterize pollen from longan honeys from Taiwan and Thailand. The flow cytometry was employed for forward scatter (FSC), side scatter (SSC), Y610-A, and NUV450 to analyze longan honey samples from Taiwan and Thailand. Taiwan’s longan honeys were rich in pollens; however, based upon the FSC and SSC analyses, the pollens from Thai longan honeys were larger and more granular. The Y610/20 emission area was greatest in Thai pollens. The NUV450 measured in the near UV laser was also greater in Thai pollen. Additionally, honey samples were also analysed for physiochemical properties including moisture content, pH, ash content, viscosity, and hydroxymethylfurfural (HMF) for physiochemical properties of longan honey samples from both countries. The moisture content of honey from Taiwan varied between 20.90% and 23.40%, whereas honey from Thailand ranged from 19.50% to 23.50%. A total of 60% of Taiwan’s longan honey was found to have a dark amber color, and only 20% of Thai longan honey exhibited a dark amber color. Furthermore, the pH range of honey from Taiwan was found to be between 4.00 and 4.16, and the pH of Thai honey ranged from 4.01 to 4.12. The ash content of honey samples from Taiwan ranged from 0.05% to 0.23%, and Thai honey had a range of 0.01% to 0.9%. All samples were negative for the Fiehe’s test, indicating the absence of HMF. This analysis lays the groundwork for rapid identification the origins of the honey, applying flow cytometry in conjunction with physicochemical analysis to assess its quality. Full article
(This article belongs to the Section Food Analytical Methods)
Show Figures

Figure 1

Figure 1
<p>Scatter plots of pollen from Taiwanese longan honey samples. The fluorescent stain 4′,6-diamidino-2-phenylindole (DAPI) and chlorophyll protein (Chlorophyll a BL690-A) were used to create subpopulations. P1, P2, P3, and P4 indicated four randomly selected subpopulations of pollen. Chla BL690-A = Chlorophyll a BL690-A. DAPI NUV450-A = DAPI NUV450-A Nucleic Acid Stain.</p>
Full article ">Figure 2
<p>Pollen subpopulations of Thai longan honey samples analyzed by scatter plots. The fluorescent stain 4′,6-diamidino-2-phenylindole (DAPI) and chlorophyll protein (Chlorophyll a BL690-A) were used to create subpopulations. P1, P2, P3, and P4 indicated four randomly selected subpopulations of pollen. Chla BL690-A = Chlorophyll a BL690-A. DAPI NUV450-A = DAPI NUV450-A Nucleic Acid Stain.</p>
Full article ">Figure 3
<p>Dot plot for showing pollen distribution of Taiwan, Thai longan honeys, and artificial honey. The pollen is as measured in the forward (FSC) and side scatter (SSC); the area marked 10 μm excludes particles or debris.</p>
Full article ">
19 pages, 7043 KiB  
Article
Flow Cytometry Analyses of Meningioma Immune Cell Composition Using a Short, Optimized Digestion Protocol
by Gillian Dao Nyesiga, Jeppe Lohfert Haslund-Vinding, Josephine Budde, Josefine Føns Lange, Nadja Blum, Kotryna Dukstaite, Lars Ohlsson, Tiit Mathiesen, Anders Woetmann and Frederik Vilhardt
Cancers 2024, 16(23), 3942; https://doi.org/10.3390/cancers16233942 - 25 Nov 2024
Viewed by 612
Abstract
Background: Current challenges in meningioma treatment, including post-surgical complications and cognitive impairments, highlight the need for new treatment alternatives. Immunological interventions have shown promise. However, there is a knowledge gap in characterizing infiltrating immune cells in meningioma and their interplay. Further studies on [...] Read more.
Background: Current challenges in meningioma treatment, including post-surgical complications and cognitive impairments, highlight the need for new treatment alternatives. Immunological interventions have shown promise. However, there is a knowledge gap in characterizing infiltrating immune cells in meningioma and their interplay. Further studies on immune cells in single-cell suspensions from digested meningioma tissues could identify targetable mechanisms for non-surgical treatment options with fewer side effects. This study aimed to optimize a protocol for faster digestion of meningioma tissues into viable single-cell suspensions and to identify infiltrating immune cell populations. Methods: We modified a commercial kit intended for whole skin dissociation to digest resected meningioma tissues into viable single-cell suspensions. Tumor-infiltrating immune cell populations were characterized using flow cytometry. Results: Flow cytometry analyses revealed that the digested tissue was composed of viable immune cells, including predominantly CD14+ macrophages and CD3+ T cells, with minor populations of CD56+ NK cells and CD19+ B cells. In both of the two patient samples tested, half of the tumor-associated macrophages were TIM-3+, with a small proportion co-expressing CD83. Women were more likely to have a lower proportion of immune cells, B cells, and NK cells. Female patients with a high proportion of immune cells had a higher proportion of macrophages. Conclusion: We successfully optimized a protocol for generating single-cell suspensions with viable immune cells from meningioma tissues, revealing infiltrating antigen-presenting cells with an immunosuppressive phenotype, and lymphocytes. This short protocol allows advanced analyses of tumor-infiltrating cells using techniques such as single-cell RNA sequencing and flow cytometry, which require live, dissociated cells. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>An overview of the optimized protocol for digestion of meningioma for viable single-cell suspensions comprising immune cells. The illustration was created using BioRender.</p>
Full article ">Figure 2
<p>Assessment of cell suspension after digesting meningioma using flow cytometry. (<b>A</b>) Gating strategy for the samples from flow cytometry analyses. (<b>B</b>) Viability of single cells without debris clouds and (<b>C</b>) proportion of CD45<sup>+</sup> immune cells after meningioma tumors were incubated in digestion reagents for 10, 20, 30, and 40 min. See <a href="#app1-cancers-16-03942" class="html-app">Supplementary Figure S1</a> for more data. (<b>D</b>) The viability of single cell without debris clouds, and the proportion of CD45<sup>+</sup> immune cells of digested meningioma samples. (<b>E</b>,<b>F</b>) Samples digested the same day (fresh) compared to samples digested the next day (O/N) assessed (<b>E</b>) via the viability of single cells and (<b>F</b>) via the proportion of CD45<sup>+</sup> immune cells. Data is presented as individual values ((<b>B</b>,<b>C</b>) with <span class="html-italic">n</span> = 1 from 1 experiment) or as mean with SEM ((<b>D</b>) with <span class="html-italic">n</span> = 20 from 15 experiments, and (<b>E</b>,<b>F</b>) with <span class="html-italic">n</span> = 7 from 7 experiments) for Fresh and <span class="html-italic">n</span> = 13 with 9 experiments for O/N. An unpaired <span class="html-italic">t</span>-test was used for statistical comparisons. Statistical significances are presented as ns = non-significant.</p>
Full article ">Figure 3
<p>An overview of the myeloid compartment of the digested meningioma tissue using flow cytometry. Debris, doublets, and dead cells were excluded before the CD45<sup>+</sup> gate (as shown in <a href="#cancers-16-03942-f002" class="html-fig">Figure 2</a>A). (<b>A</b>) The gating strategy for <a href="#cancers-16-03942-f003" class="html-fig">Figure 3</a>D. (<b>B</b>) The gating strategy for <a href="#cancers-16-03942-f003" class="html-fig">Figure 3</a>E. (<b>C</b>) The gating strategy for <a href="#cancers-16-03942-f003" class="html-fig">Figure 3</a>F. (<b>D</b>) The proportion of CD11c<sup>+</sup> cells, CD14<sup>+</sup> cells, and CD64<sup>+</sup> cells of viable CD45<sup>+</sup> immune cells. (<b>E</b>) Proportions of CD11c<sup>+</sup>CD14<sup>+</sup>, CD14<sup>+</sup>HLA-DR<sup>+</sup>, and CD11c<sup>+</sup>HLA-DR<sup>+</sup> cells. (<b>F</b>) HLA-DR<sup>+</sup> and CD64<sup>+</sup> cells of CD11c<sup>+</sup>CD14<sup>+</sup> cells. Data is presented as mean with SEM ((<b>D</b>) with <span class="html-italic">n</span> = 17 from 10 experiments for CD11c, <span class="html-italic">n</span> = 19 from 12 experiments for CD14, <span class="html-italic">n</span> = 9 from 6 experiments for HLA-DR, and <span class="html-italic">n</span> = 4 from 2 experiments for CD64; (<b>E</b>) with <span class="html-italic">n</span> = 15 from 9 experiments for CD11c<sup>+</sup>CD14<sup>+</sup>, <span class="html-italic">n</span> = 9 from 6 experiments for CD14<sup>+</sup>HLA-DR<sup>+</sup>, and <span class="html-italic">n</span> = 9 from 6 experiments; and (<b>F</b>) with <span class="html-italic">n</span> = 9 from 6 experiments for HLA-DR and <span class="html-italic">n</span> = 4 from 2 experiments. Either a <span class="html-italic">t</span>-test or one-way ANOVA with post-hoc testing was used for statistical comparisons. Statistical significances are presented as ns = non-significant.</p>
Full article ">Figure 4
<p>The expression of TIM-3 and CD83 in CD45<sup>+</sup> immune cells was assessed using flow cytometry. (<b>A</b>) CD14/TIM-3 of viable CD45<sup>+</sup> cells. (<b>B</b>) CD14/CD83 of viable CD45<sup>+</sup> immune cells. (<b>C</b>) CD14 of viable CD45<sup>+</sup> cells. (<b>D</b>) CD4 of viable CD45<sup>+</sup> cells. (<b>E</b>) CD14/CD4 of viable CD45<sup>+</sup> cells. (<b>F</b>) CD83/TIM-3 of CD14<sup>+</sup>CD4<sup>+</sup> cells. Representative data for 2 patients with WHO grade 1 meningioma from 2 experiments.</p>
Full article ">Figure 5
<p>Overview of the lymphoid compartment. (<b>A</b>) The gating strategy for the flow cytometric analyses. (<b>B</b>) T cell proportion of CD45<sup>+</sup> immune cells. (<b>C</b>) The proportion of CD4<sup>+</sup> helper T cells and CD8<sup>+</sup> cytotoxic T cells. (<b>D</b>) The proportion of CD19<sup>+</sup> B cells. (<b>E</b>) The proportion of CD56<sup>+</sup> NK cells. Data is presented as mean with SEM ((<b>B</b>) with <span class="html-italic">n</span> = 5 from 4 experiments, (<b>C</b>) with <span class="html-italic">n</span> = 3 from 3 experiments, (<b>D</b>) with <span class="html-italic">n</span> = 22 from 14 experiments, and (<b>E</b>) with <span class="html-italic">n</span> = 12 from 7 experiments). A <span class="html-italic">t</span>-test was used for statistical comparison. Statistical significances are presented as ** = <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Assessment of CD45RA and CD45RO expression on immune cells within meningioma. (<b>A</b>) The gating strategy for the flow cytometry analyses. The proportion of CD45RA and CD45RO of the following populations: (<b>B</b>) CD45<sup>+</sup> cells, (<b>C</b>) CD4<sup>+</sup> cells, (<b>D</b>) CD8<sup>+</sup> cells, and (<b>E</b>) CD4<sup>−</sup>CD8<sup>−</sup> cells. Data is presented as mean with SEM ((<b>B</b>–<b>E</b>) with <span class="html-italic">n</span> = 15 from 13 experiments), and a <span class="html-italic">t</span>-test was used for statistical comparisons. Statistical significances are presented as * = <span class="html-italic">p</span> &lt; 0.05 and **** = <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
18 pages, 939 KiB  
Review
Periodontal Molecular Diagnostics: State of Knowledge and Future Prospects for Clinical Application
by Ewa Dolińska, Patryk Wiśniewski and Małgorzata Pietruska
Int. J. Mol. Sci. 2024, 25(23), 12624; https://doi.org/10.3390/ijms252312624 - 25 Nov 2024
Viewed by 655
Abstract
Periodontitis leads to immunologically mediated loss of periodontium and, if untreated, can result in tooth loss. Periodontal diseases are the most prevalent in the world and have a very strong impact on patients’ well-being and general health. Their treatment generates enormous costs. Given [...] Read more.
Periodontitis leads to immunologically mediated loss of periodontium and, if untreated, can result in tooth loss. Periodontal diseases are the most prevalent in the world and have a very strong impact on patients’ well-being and general health. Their treatment generates enormous costs. Given the above, precise, prompt, and predictive diagnosis of periodontal disease is of paramount importance for clinicians. The aim of the study was to summarize the state-of-the-art knowledge of molecular periodontal diagnostics and the utility of its clinical application. There is a great need to have diagnostic tests that not only describe the periodontal destruction that has occurred in the tissues but also allow clinicians to detect disease at a subclinical level before the changes occur. A test that would enable clinicians to follow the course of the disease and detect areas prone to exacerbation could be used to evaluate the effectiveness of ongoing periodontal therapies. Unfortunately, there is no such diagnostic method yet. A hopeful prospect is molecular diagnostics. There are numerous studies on biomarkers of periodontal disease. Point-of-care tests are also emerging. There are possibilities for processing large biological datasets (omics data). However, all of the above have a minor role in the overall single-patient diagnostics process. Despite advances in microbiological, molecular, and genetic research, the basis of periodontal diagnosis is still clinical examination enriched by the evaluation of radiological images. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the destruction of the alveolar bone and connective tissue in the course of periodontitis. Lipopolysaccharide (LPS), a major virulence factor for Gram-negative bacteria, binds to the lipid-binding protein (LBP). This complex is recognized by the CD14 receptor on monocytes/macrophages. Association of the LPS–LBP complex with the CD14 receptor initiates trans-membrane signaling that activates the cell to synthesize and secrete prostaglandin 2 (PGE2), the cytokines TNFα and IL-1β, and extracellular matrix metalloproteinases (MMPs). TNFα and IL-1β bind to receptors on fibroblasts and initiate signaling for the synthesis and secretion of MMPs and PGE2. MMPs mediate the loss of gingival extracellular matrix and periodontal ligament, while PGE2 leads to bone destruction. TNFα and IL-1β can also directly influence minor bone loss.</p>
Full article ">Figure 2
<p>Current and potential diagnostic possibilities on a simplified model of periodontal disease pathogenesis.</p>
Full article ">Figure 3
<p>Hypotheses on the role of bacteria in the etiopathogenesis of periodontal disease and milestones in the study of the oral microbiome on a timeline. (HOMIM—Human Oral Microbe Identification Microarray, HOMD—the Human Oral Microbiome Database, IMPEDE—Inflammation-Mediated Polymicrobial Emergence and Dysbiotic Exacerbation).</p>
Full article ">
Back to TopTop