[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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (937)

Search Parameters:
Keywords = ceramide

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 445 KiB  
Article
Investigating a New Way to Assess Metabolic Risk in Pregnant Females with Prior RYGB Surgery
by Teresa Gisinger, Birgit Reiter, Karin Preindl, Thomas Stimpfl, Liliana-Imi Gard, Sabina Baumgartner-Parzer, Alexandra Kautzky-Willer and Michael Leutner
Nutrients 2024, 16(16), 2704; https://doi.org/10.3390/nu16162704 (registering DOI) - 14 Aug 2024
Abstract
Background: Obesity in pregnancy is linked to adverse clinical outcomes such as gestational diabetes. Recently, a risk score calculated by different ceramide concentrations was recognized as a new way to investigate cardiovascular risk. The aim was to analyze if the ceramide risk score [...] Read more.
Background: Obesity in pregnancy is linked to adverse clinical outcomes such as gestational diabetes. Recently, a risk score calculated by different ceramide concentrations was recognized as a new way to investigate cardiovascular risk. The aim was to analyze if the ceramide risk score and cardiometabolic risk vary between normal-weight, obese, and females with prior Roux-en-Y bypass surgery (RYGB) during pregnancy. Methods: Three cohorts were investigated: first, 25 pregnant females with a history of RYGB; second, 19 with preconception BMI ≥ 35 kg/m2; and third, 19 normal-weight (preconception BMI < 25 kg/m2). Around the 24th to 28th weeks of gestation routine laboratory assessments, 3 h 75 g oral and intravenous glucose tolerance tests were carried out. The correlation of ceramide risk scores and ceramide ratios (Cer(d18:1/18:0)/Cer(d18:1/16:0)) with metabolic parameters was analyzed via Pearson correlation. The cohorts were compared via ANOVA and unpaired t-tests. Results: The RYGB cohort had lower ceramide risk scores and ratios compared to obese pregnant females (7.42 vs.9.34, p = 0.025; 0.33 vs.0.47, p < 0.001). Ceramide risk score and ratio were found to correlate negatively with insulin sensitivity (measured with the Matsuda (r = −0.376, p = 0.031; r = −0.455, p = 0.008) and calculated sensitivity index (r = −0.358, p = 0.044; r = −0.621, p < 0.001)) in females without RYGB. The ceramide risk score correlated positively with body fat in RYGB females (r = 0.650, p = 0.012). Conclusions: We found that females after RYGB have lower ceramide risk scores and ceramide ratios compared to obese pregnant females, possibly indicating lower metabolic risk. Full article
(This article belongs to the Special Issue Nutrition and Supplements during Pregnancy (2nd Edition))
21 pages, 10664 KiB  
Article
Overexpression of the β-Subunit of Acid Ceramidase in the Epidermis of Mice Provokes Atopic Dermatitis-like Skin Symptoms
by Miho Sashikawa-Kimura, Mariko Takada, Md Razib Hossain, Hidetoshi Tsuda, Xiaonan Xie, Mayumi Komine, Mamitaro Ohtsuki and Genji Imokawa
Int. J. Mol. Sci. 2024, 25(16), 8737; https://doi.org/10.3390/ijms25168737 - 10 Aug 2024
Viewed by 429
Abstract
We previously reported that a pathogenic abnormality in the barrier and water-holding functions of the stratum corneum (SC) in the skin of patients with atopic dermatitis (AD) is mainly attributable to significantly decreased levels of total ceramides in the SC. That decrease is [...] Read more.
We previously reported that a pathogenic abnormality in the barrier and water-holding functions of the stratum corneum (SC) in the skin of patients with atopic dermatitis (AD) is mainly attributable to significantly decreased levels of total ceramides in the SC. That decrease is mediated by the abnormal expression of a novel ceramide-reducing enzyme, sphingomyelin/glucosylceramide deacylase (SGDase), which is the β-subunit (ASAH1b) of acid ceramidase. In this study, we determined whether mice overexpressing ASAH1b in their epidermis develop AD-like skin symptoms. We generated transgenic (TG) mice overexpressing ASAH1b, regulated by the involucrin promoter, to localize its expression in the upper epidermis. After hair removal using a depilatory cream containing glycolic acid, the TG mice without any visible skin inflammation at 8 weeks of age had increased levels of ASAH1b and decreased levels of SC ceramide, with disrupted barrier functions measured by trans-epidermal water loss compared to the wild-type (WT) mice. Interestingly, enzymatic assays revealed that SGDase activity was not detectable in the skin of the TG mice compared to WT mice. Immunological staining revealed that there was an increased expression level of IL-33 in the epidermis and an accumulation of macrophages in the dermis of TG mice compared to WT mice, which are phenotypic characteristics of AD, that were exacerbated by tape-stripping of the skin. In the skin of the TG mice, the mRNA levels of IL-5, CCL11, IL-22, CXCL10, and IFNγ were significantly upregulated compared to the WT mice, and tape-stripping significantly increased the mRNA levels of IL-4, IL-33, CXCL1, CXCL12, TLR9, and CD163 compared to WT mice. These findings strongly indicate that the skin of the depilatory cream-treated TG mice exists in an atopic dry skin condition that is highly sensitive to various environmental stimuli. The sum of our results suggests that ASAH1b itself, even in the absence of its enzymatic activity, is a major etiologic factor for atopic dry skin symptoms via an unknown mechanism. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>Appearance of the ear skin, H&amp;E staining, skin score and thickness of the epidermis before and after repeated tape-stripping. (<b>A</b>) (<b>a</b>) WT ears at day 0 before tape-stripping, (<b>b</b>) WT ears at day 2 after tape-stripping of the right ear (red arrow); the left ear (blue arrow) was not tape-stripped as a control, (<b>c</b>) H&amp;E staining of the left WT ear at day 2, (<b>d</b>) H&amp;E staining of right WT ear at day 2 after tape-stripping, (<b>e</b>) TG ears at day 0 before tape-stripping, (<b>f</b>) TG ears at day 2 after tape-stripping of the right ear (red arrow); the left ear (blue arrow) was not tape-stripped as a control, (<b>g</b>) H&amp;E staining of the left TG ear at day 2, (<b>h</b>) H&amp;E staining of the right TG ear at day 2 after tape-stripping. (<b>B</b>) (<b>a</b>) Total visible score on the outside ear skin of TG and WT mice at the age of 8 weeks before and day 2 after tape-stripping. n = 7 for WT, n = 5 for TG, *: <span class="html-italic">p</span> &lt; 0.05 by Mann–Whitney comparisons test. (<b>b</b>) Thickness of the epidermis of the outside of the ear measured using Image J software (imageJ 1.54g). n = 5~7, ***: <span class="html-italic">p</span> &lt; 0.001, ****: <span class="html-italic">p</span> &lt; 0.0001 by Tukey’s multiple comparisons test. nd: not significant difference.</p>
Full article ">Figure 2
<p>Surface appearance of the dorsal skin of TG and WT mice at the age of 8 weeks after hair removal using electric hair clippers and barrier function values three days after hair removal. (<b>A</b>) Skin appearance three days after hair removal. (<b>B</b>) Barrier function measured by TEWL three days after hair removal, TG; n = 6, WT; n = 4, ns: not significant, by Mann–Whitney test.</p>
Full article ">Figure 3
<p>Surface appearance of the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks and barrier function before and at day 2 after tape-stripping. (<b>A</b>) Skin appearance at Days 0, 1 and 2, <b>right</b> and <b>left</b> sides (separated by red line) of the dorsal skin show tape-stripped and non-tape-stripped areas, respectively. (<b>B</b>) Barrier function measured by TEWL before and at day 2 after tape-stripping, n = 5, ****: <span class="html-italic">p</span> &lt; 0.0001, ***: <span class="html-italic">p</span> &lt; 0.001 by Tukey’s multiple comparisons test.</p>
Full article ">Figure 4
<p>H&amp;E staining of the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and after tape-stripping. (<b>A</b>) H&amp;E staining, (<b>a</b>) WT dorsal skin before tape-stripping, (<b>b</b>) WT dorsal skin at day 2 after tape-stripping, (<b>c</b>) TG dorsal skin before tape-stripping, (<b>d</b>) TG dorsal skin at day 2 after tape-stripping, bars = 100 μm, (<b>B</b>) Epidermal thickness, analysis by measurement using Image J software (imageJ 1.54g). n = 6, ****: <span class="html-italic">p</span> &lt; 0.0001 by Tukey’s multiple comparisons test.</p>
Full article ">Figure 5
<p>Immunostaining of ASAH1 with an anti-ASAH1b antibody in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. (<b>A</b>) Immunostaining, bars = 100 μm, (<b>B</b>) Stained area, analysis by measurement with a Keyence Image Analyzer. n = 6, ****: <span class="html-italic">p</span> &lt; 0.0001, **: <span class="html-italic">p</span> &lt; 0.01 by Tukey’s multiple comparisons test. TS = tape-stripped.</p>
Full article ">Figure 6
<p>Immunostaining of ceramide with an anti-ceramide antibody in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. (<b>A</b>) Immunostaining, bars = 100 μm, (<b>a</b>) WT/TS (−), (<b>b</b>) WT/TS (+), (<b>c</b>) TG/TS (−), (<b>d</b>) TG/TS (+), (<b>B</b>) Stained area, analysis by measurement with a Keyence Image Analyzer, n = 6, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test. TS: tape-stripped.</p>
Full article ">Figure 7
<p>Western blotting of ASAH1b protein in epidermal homogenates of the depilatory cream-treated dorsal skin and abdominal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. ME = treated with 2-mercaptoethanol. The epidermal homogenates were obtained at 8 weeks of age or at day 2 after tape-stripping.</p>
Full article ">Figure 8
<p>Immunostaining of macrophages using various macrophage markers in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. (<b>A</b>) Immunostaining of F4/80, bars = 100 μm, analysis by measurement with a Keyence Image Analyzer, n = 5, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test. (<b>B</b>) Immunostaining of CD80, bars = 100 μm, analysis by measurement with a Keyence Image Analyzer, n = 3, ***: <span class="html-italic">p</span> &lt; 0.001, **: <span class="html-italic">p</span> &lt; 0.01 by Tukey’s multiple comparisons test. (<b>C</b>) Immunostaining of CD163, bars = 100 μm, analysis by measurement with a Keyence Image Analyzer, n = 5, *: <span class="html-italic">p</span> &lt; 0.05, by Tukey’s multiple comparisons test. (<b>a</b>) WT/TS (−), (<b>b</b>) WT/TS (+), (<b>c</b>) TG/TS (−), (<b>d</b>) TG/TS (+), TS: Tape-stripped. ns: not significant.</p>
Full article ">Figure 9
<p>Toluidine blue staining of mast cells (<b>A</b>) and immunostaining with an anti-IL-33 antibody (<b>B</b>) in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. bars = 100 μm, analysis by measurement with a Keyence Image Analyzer. n = 6, ***: <span class="html-italic">p</span> &lt; 0.001, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test. (<b>a</b>) WT/TS (−), (<b>b</b>) WT/TS (+), (<b>c</b>) TG/TS (−), (<b>d</b>) TG/TS (+), TS: Tape-stripping.</p>
Full article ">Figure 10
<p>Effects of the overexpression of ASAH1b on the activity of SGDase in the epidermis. (<b>A</b>): LC-MS-MS spectrum for released SPC by sphingolipid ceramide N-deacylase from <span class="html-italic">Pseudomonas</span> sp. as a positive control. (<b>B</b>): LC-MS-MS spectrum for released SPC by SGDase in the epidermal homogenate from TG/TS (+). (<b>C</b>): Time course of the released SPC after enzymatic reaction with epidermal homogenate or sphingolipid ceramide N-deacylase from <span class="html-italic">Pseudomonas</span> sp. as positive control.An epidermal homogenate was incubated for 5 h at 37 °C with varying amounts of SM. The enzymatic activities expressed as released SPC for WT/TS(-), WT/TS(+), TG/TS(-), and TG/TS(+) were located at O level. The final reaction mixtures contained 50 mM potassium acetate buffer (pH 4.7), the enzyme source, the substrate, 0.1% Triton X-100 and 20 mM CaCl<sub>2</sub>. The rate of SPC generation was measured as a function of SGDase by LC-MS-MS analysis. PC: positive control using sphingolipid ceramide N-deacylase from <span class="html-italic">Pseudomonas</span> sp. as SGDase. TS: Tape-stripping.</p>
Full article ">Figure 11
<p>Effects of the overexpression of ASAH1b on the activity of ASAH1 in the epidermis. (<b>A</b>): LC-MS-MS spectrum of released sphingosine (SPH) after enzymatic reaction with epidermal homogenate. (<b>B</b>): Enzymatic activity of ASAH1 expressed as released SPH in epidermal homogenates of WT and TG mice. An epidermal homogenate was incubated for 12 h at 37 °C with varying amounts of ceramide substrates. The final reaction mixtures contained 50 mM potassium acetate buffer (pH 4.7), the enzyme source, the substrate, 0.1% Triton X-100 and 20 mM CaCl<sub>2</sub>. Blue rectangles indicate TG mice. The rate of sphingosine (SPH) generation was measured as a function of ASAH1 by LC-MS-MS analysis. n = 6, ****: <span class="html-italic">p</span> &lt; 0.0001, **: <span class="html-italic">p</span> &lt; 0.01, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test.</p>
Full article ">Figure 12
<p>mRNA expression levels of inflammatory cytokines and chemokines in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. n = 3~9, ****: <span class="html-italic">p</span> &lt; 0.0001, ***: <span class="html-italic">p</span> &lt; 0.001, **: <span class="html-italic">p</span> &lt; 0.01, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test.</p>
Full article ">
16 pages, 5630 KiB  
Article
Angiotensin II Alters Mitochondrial Membrane Potential and Lipid Metabolism in Rat Colonic Epithelial Cells
by Darby D. Toth, Christopher L. Souder, Sarah Patuel, Cole D. English, Isaac Konig, Emma Ivantsova, Wendi Malphurs, Jacqueline Watkins, Kaylie Anne Costa, John A. Bowden, Jasenka Zubcevic and Christopher J. Martyniuk
Biomolecules 2024, 14(8), 974; https://doi.org/10.3390/biom14080974 - 9 Aug 2024
Viewed by 336
Abstract
An over-active renin-angiotensin system (RAS) is characterized by elevated angiotensin II (Ang II). While Ang II can promote metabolic and mitochondrial dysfunction in tissues, little is known about its role in the gastrointestinal system (GI). Here, we treated rat primary colonic epithelial cells [...] Read more.
An over-active renin-angiotensin system (RAS) is characterized by elevated angiotensin II (Ang II). While Ang II can promote metabolic and mitochondrial dysfunction in tissues, little is known about its role in the gastrointestinal system (GI). Here, we treated rat primary colonic epithelial cells with Ang II (1–5000 nM) to better define their role in the GI. We hypothesized that Ang II would negatively affect mitochondrial bioenergetics as these organelles express Ang II receptors. Ang II increased cellular ATP production but reduced the mitochondrial membrane potential (MMP) of colonocytes. However, cells maintained mitochondrial oxidative phosphorylation and glycolysis with treatment, reflecting metabolic compensation with impaired MMP. To determine whether lipid dysregulation was evident, untargeted lipidomics were conducted. A total of 1949 lipids were detected in colonocytes spanning 55 distinct (sub)classes. Ang II (1 nM) altered the abundance of some sphingosines [So(d16:1)], ceramides [Cer-AP(t18:0/24:0)], and phosphatidylcholines [OxPC(16:0_20:5(2O)], while 100 nM Ang II altered some triglycerides and phosphatidylserines [PS(19:0_22:1). Ang II did not alter the relative expression of several enzymes in lipid metabolism; however, the expression of pyruvate dehydrogenase kinase 2 (PDK2) was increased, and PDK2 can be protective against dyslipidemia. This study is the first to investigate the role of Ang II in colonic epithelial cell metabolism. Full article
Show Figures

Figure 1

Figure 1
<p>Cytotoxicity of Ang II to colonocytes at 72 h. (<b>A</b>) Cytotoxicity, (<b>B</b>) Cell viability. The lysis control was used as a positive control for the assay (induces cell death of colonocytes). The columns represent the mean relative fluorescence ± standard deviation. Different letters denote significant differences from the media-only control (One-way ANOVA, Dunnett multiple comparison test, n = 4/experiment, significance determined at <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>ATP levels after exposure to Ang II at 72 h. Carbonyl cyanide-4-phenylhydrazone (FCCP) was used as a positive control. Relative luminescence is graphed for each experimental group (horizontal bar represents mean relative luminescence ± standard deviation). Asterisks (****) denotes significant differences from the media-only control (One-way ANOVA followed by a Dunnett multiple comparison test, n = 4/experiment, significance determined at <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 3
<p>Mitochondrial membrane potential (MMP) after exposure to Ang II at 72 h. Carbonyl cyanide-4-phenylhydrazone (FCCP) was used as a positive control as it acts as an uncoupling agent for mitochondrial membranes. Relative fluorescence is based on the red/green signal intensity, and all data are normalized to the media-only control (mean relative fluorescence ± standard deviation). Asterisk denotes significant differences compared to the media-only control (one-way ANOVA followed by a Dunnett multiple comparison test, n = 4/experiment, significance determined at * <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>Normalized oxygen consumption rate and extracellular acidification rate for rat epithelial colonocytes after a 24 h exposure to Ang II. (<b>A</b>) Oxygen consumption rates over time (<b>B</b>) Acidification rates over time. Data are represented as mean ± standard deviation (one-way ANOVA followed by a Dunnett multiple comparison test, n = 4 replicates/groups, significance determined at <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Lipid abundance and categorical classification of lipids in rat epithelial colonocytes (all lipids detected in all treatments). Abbreviations: triglycerides (TG), plasmanyl-TG (plasmanyl-triglycerides), phosphatidylcholine (PC), phosphatidylethanolamines (PE), ceramide (Cer), diacylglycerol (DG), plasmanyl-PC (plasmanyl-phosphatidylcholine), plasmenyl-PE (plasmenyl- phosphatidylethanolamines), phosphatidylserines (PS), plasmenyl-PS (plasmenyl-phosphatidylethanolamines), oxidized phosphatidylcholines (OxPC), phosphatidylglycerol (PG), phosphoinositide (PI), oxidized phosphatidylethanolamines (PE), dimethyl-phosphatidylethanolamine (DMPE), hemibismonoacylglycerophosphate (HBMP), plasmenyl-PC (plasmenyl-phosphatidylcholine), polyethylene glycol (PEG), oxidized lysophosphatidylcholines (OxLPC), lysophosphatidylcholines (LPC), oxidized triglycerides (OxTG), cardiolipins (CL), monomethyl-phosphatidylethanolamine (MMPE), lysophosphatidylethanolamine (LPE), and glucosylceramide non-hydroxyfatty acid-sphingosine (HexCer-NS).</p>
Full article ">Figure 6
<p>(<b>A</b>-Top graph) Principal component analysis scores plot for rat colonocyte lipids with each point representing the lipids in a single sample, the ellipses representing the 95% confidence interval, and the colored groups representing the three different treatments (blue = control, red = low Ang II, and green = high Ang II). (<b>B</b>-bottom graph) Heatmap showing significant changes in the levels of lipids following exposure to Ang II. Data were subjected to ANOVA followed by Fisher’s least significant difference method (Fisher’s LSD), and significant changes were set at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>Volcano plots of (<b>A</b>) 1 nM of Ang II and (<b>B</b>) 100 nM of Ang II and the differentially abundant lipids (<span class="html-italic">p</span> &lt; 0.05) outlined in red and blue.</p>
Full article ">Figure 8
<p>Relative concentrations of lipid abundance by sample weight in rat epithelial colonocytes exposed to 1 nM of Ang II (<b>left panel</b>). The most abundant lipids measured include So(d16:1) and Cer-AP(t18:0/24:0). Abbreviations: sphingosine (So), alpha-hydroxy-fatty acid phytosphingosine ceramide (Cer-AP), oxidized lysophosphatidylcholines (OxLPC), and phosphatidylethanolamines (PE). Relative concentrations of lipid abundance by sample weight in rat epithelial colonocytes exposed to 100 nM of Ang II (<b>right panel</b>). The most abundant lipid measured was So(d16:1). Abbreviations: sphingosine (So), oxidized phosphatidylcholines (OxPC), phosphatidylethanolamines (PE), and phosphatidylserines (PS). The black dots represent the metabolite levels in all samples, and the yellow diamond represents the average value.</p>
Full article ">Figure 9
<p>Relative gene expression for rat colonocytes after exposure to Ang II. (<b>a</b>) <span class="html-italic">PDK1,</span> (<b>b</b>) <span class="html-italic">PDK2</span>, (<b>c</b>) <span class="html-italic">PDK4</span>. Data are represented as mean ± standard deviation. Asterisks (**) denote significant differences from the media-only control (data were evaluated using a Mann–Whitney U test, n = 4/experiment, significance determined at <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
14 pages, 2161 KiB  
Article
Brain Region-Specific Expression Levels of Synuclein Genes in an Acid Sphingomyelinase Knockout Mouse Model: Correlation with Depression-/Anxiety-Like Behavior and Locomotor Activity in the Absence of Genotypic Variation
by Razvan-Marius Brazdis, Iulia Zoicas, Johannes Kornhuber and Christiane Mühle
Int. J. Mol. Sci. 2024, 25(16), 8685; https://doi.org/10.3390/ijms25168685 - 9 Aug 2024
Viewed by 276
Abstract
Accumulating evidence suggests an involvement of sphingolipids, vital components of cell membranes and regulators of cellular processes, in the pathophysiology of both Parkinson’s disease and major depressive disorder, indicating a potential common pathway in these neuropsychiatric conditions. Based on this interaction of sphingolipids [...] Read more.
Accumulating evidence suggests an involvement of sphingolipids, vital components of cell membranes and regulators of cellular processes, in the pathophysiology of both Parkinson’s disease and major depressive disorder, indicating a potential common pathway in these neuropsychiatric conditions. Based on this interaction of sphingolipids and synuclein proteins, we explored the gene expression patterns of α-, β-, and γ-synuclein in a knockout mouse model deficient for acid sphingomyelinase (ASM), an enzyme catalyzing the hydrolysis of sphingomyelin to ceramide, and studied associations with behavioral parameters. Normalized Snca, Sncb, and Sncg gene expression was determined by quantitative PCR in twelve brain regions of sex-mixed homozygous (ASM−/−, n = 7) and heterozygous (ASM+/−, n = 7) ASM-deficient mice, along with wild-type controls (ASM+/+, n = 5). The expression of all three synuclein genes was brain region-specific but independent of ASM genotype, with β-synuclein showing overall higher levels and the least variation. Moreover, we discovered correlations of gene expression levels between brain regions and depression- and anxiety-like behavior and locomotor activity, such as a positive association between Snca mRNA levels and locomotion. Our results suggest that the analysis of synuclein genes could be valuable in identifying biomarkers and comprehending the common pathological mechanisms underlying various neuropsychiatric disorders. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Brain-specific variation of α, β-, and γ-synuclein gene expression in twelve regions with uniformity across the three acid sphingomyelinase (ASM) genotypes. (<b>a</b>) <span class="html-italic">Snca</span>, (<b>b</b>) <span class="html-italic">Sncb</span>, and (<b>c</b>) <span class="html-italic">Sncg</span> were expressed differently in twelve brain regions: frontal cortex (FC), dorsal striatum (DS), lateral septum (LS), ventral striatum (VS), amygdala (AM), dorsal hippocampus (DH), thalamus (TH), hypothalamus (HY), ventral hippocampus (VH), dorsal mesencephalon (DM), ventral mesencephalon (VM), and cerebellum (CE). No statistically significant differences were observed between homozygous ASM-deficient (ASM−/−, <span class="html-italic">n</span> = 7), heterozygous ASM-deficient (ASM+/−, <span class="html-italic">n</span> = 7), and wild-type (ASM+/+, <span class="html-italic">n</span> = 5) mice. Data represent individual data points with means as bars.</p>
Full article ">Figure 2
<p>Heat maps of Spearman correlation coefficient (ρ) between (<b>a</b>) <span class="html-italic">Snca</span>, (<b>b</b>) <span class="html-italic">Sncb</span>, and (<b>c</b>) <span class="html-italic">Sncg</span> expression in twelve brain regions, frontal cortex (FC), dorsal striatum (DS), lateral septum (LS), ventral striatum (VS), amygdala (AM), dorsal hippocampus (DH), thalamus (TH), hypothalamus (HY), ventral hippocampus (VH), dorsal mesencephalon (DM), ventral mesencephalon (VM), and cerebellum (CE), for the entire group of mice (total, <span class="html-italic">n</span> = 19). ρ index ranges from −1 to +1; blue indicates a positive correlation, and red a negative correlation (darker color indicates a stronger correlation); white (ρ = 0) represents no correlation. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 for the significance level of the correlation.</p>
Full article ">Figure 3
<p>The behavioral phenotype of homozygous ASM knockout (ASM−/−, <span class="html-italic">n</span> = 7), heterozygous ASM-deficient (ASM+/−, <span class="html-italic">n</span> = 7), and wild-type (ASM+/+, <span class="html-italic">n</span> = 5) mice. (<b>a</b>) Percentage of immobility time, as an indicator of depression-like behavior, was assessed in the forced swim test; (<b>b</b>) Percentage of time spent in the open arms of the elevated plus-maze is an indicator of anxiety-like behavior; (<b>c</b>) The number of entries into the closed arm of the elevated plus-maze is an indicator of locomotor activity; (<b>a</b>,<b>b</b>) ASM−/− mice showed a reduced depression-like phenotype, but increased anxiety-like behavior compared with ASM+/+ mice. Locomotor activity was reduced in ASM−/− mice compared with ASM+/− mice. Data represent the means + SEM. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Heat maps of Spearman correlation coefficient (ρ) between (<b>a</b>) <span class="html-italic">Snca</span>, (<b>b</b>) <span class="html-italic">Sncb</span>, and (<b>c</b>) <span class="html-italic">Sncg</span> expression and depression-like behavior (D) expressed as percentage immobility in the forced swim test, anxiety-like behavior (A) indicated by the percentage of time spent in the open arms of the elevated plus-maze, and locomotor activity (L) assessed by the number of closed arm entries in the elevated plus-maze in twelve brain regions: frontal cortex (FC), dorsal striatum (DS), lateral septum (LS), ventral striatum (VS), amygdala (AM), dorsal hippocampus (DH), thalamus (TH), hypothalamus (HY), ventral hippocampus (VH), dorsal mesencephalon (DM), ventral mesencephalon (VM), and cerebellum (CE), for the entire group of mice [<span class="html-italic">n</span> = 19, male <span class="html-italic">n</span> = 8, female <span class="html-italic">n</span> = 11; wild-type (ASM+/+) <span class="html-italic">n</span> = 5, homozygous ASM-deficient (ASM−/−) <span class="html-italic">n</span> = 7 and heterozygous ASM-deficient (ASM+/−) <span class="html-italic">n</span> = 7]. ρ index ranges from −1 to +1; blue indicates a positive correlation, and red a negative correlation (darker color indicates a stronger correlation); white (ρ = 0) represents no correlation. * <span class="html-italic">p</span> &lt; 0.05 for the significance level of the correlation.</p>
Full article ">Figure 5
<p>Associations of synuclein expression data with behavioral measures: (<b>a</b>) Negative correlation of <span class="html-italic">Sncb</span> expression with depression-like behavior, expressed as percentage immobility in the forced swim test, in the ventral striatum (VS) of female (red, <span class="html-italic">n</span> = 8) and male (blue, <span class="html-italic">n</span> = 11) mice; (<b>b</b>) Positive correlation of <span class="html-italic">Snca</span> expression with percentage of time spent in the open arms of the elevated plus-maze, as an inverse indicator of anxiety-like behavior, in the amygdala (AM) of female (red) and male (blue) heterozygous ASM-deficient (ASM+/−, <span class="html-italic">n</span> = 7) mice; (<b>c</b>) Positive correlation of <span class="html-italic">Snca</span> expression with number of closed arm entries in the elevated plus-maze, as an indicator of locomotor activity, in the cerebellum (CE) of combined female homozygous ASM-deficient (ASM−/−, <span class="html-italic">n</span> = 1), heterozygous ASM-deficient (ASM+/−, <span class="html-italic">n</span> = 4), and wild-type (ASM+/+, <span class="html-italic">n</span> = 3) mice, (<b>d</b>) as well as in female ASM+/+ (<span class="html-italic">n</span> = 3) and male ASM+/+ (<span class="html-italic">n</span> = 2) mice. Linear regression line for the combined group with 95% confidence interval and statistics (Spearman correlation, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
25 pages, 6331 KiB  
Article
Modulation of Ceramide-Induced Apoptosis in Enteric Neurons by Aryl Hydrocarbon Receptor Signaling: Unveiling a New Pathway beyond ER Stress
by Mallappa Anitha, Supriya M. Kumar, Imhoi Koo, Gary H. Perdew, Shanthi Srinivasan and Andrew D. Patterson
Int. J. Mol. Sci. 2024, 25(16), 8581; https://doi.org/10.3390/ijms25168581 - 6 Aug 2024
Viewed by 472
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a persistent organic pollutant and a potent aryl hydrocarbon receptor (AHR) ligand, causes delayed intestinal motility and affects the survival of enteric neurons. In this study, we investigated the specific signaling pathways and molecular targets involved in TCDD-induced enteric [...] Read more.
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a persistent organic pollutant and a potent aryl hydrocarbon receptor (AHR) ligand, causes delayed intestinal motility and affects the survival of enteric neurons. In this study, we investigated the specific signaling pathways and molecular targets involved in TCDD-induced enteric neurotoxicity. Immortalized fetal enteric neuronal (IM-FEN) cells treated with 10 nM TCDD exhibited cytotoxicity and caspase 3/7 activation, indicating apoptosis. Increased cleaved caspase-3 expression with TCDD treatment, as assessed by immunostaining in enteric neuronal cells isolated from WT mice but not in neural crest cell-specific Ahr deletion mutant mice (Wnt1Cre+/−/Ahrb(fl/fl)), emphasized the pivotal role of AHR in this process. Importantly, the apoptosis in IM-FEN cells treated with TCDD was mediated through a ceramide-dependent pathway, independent of endoplasmic reticulum stress, as evidenced by increased ceramide synthesis and the reversal of cytotoxic effects with myriocin, a potent inhibitor of ceramide biosynthesis. We identified Sptlc2 and Smpd2 as potential gene targets of AHR in ceramide regulation by a chromatin immunoprecipitation (ChIP) assay in IM-FEN cells. Additionally, TCDD downregulated phosphorylated Akt and phosphorylated Ser9-GSK-3β levels, implicating the PI3 kinase/AKT pathway in TCDD-induced neurotoxicity. Overall, this study provides important insights into the mechanisms underlying TCDD-induced enteric neurotoxicity and identifies potential targets for the development of therapeutic interventions. Full article
(This article belongs to the Section Molecular Toxicology)
Show Figures

Figure 1

Figure 1
<p>TCDD induces apoptosis in IM-FEN cells. Cells were treated with vehicle or various doses of TCDD (0.1, 1, and 10 nM) for 24 h. Representative images of cleaved caspase-3 and cleaved PARP Western blots are included (<b>A</b>,<b>B</b>). Results from the protein bands’ density normalized to GAPDH have been included in the correspondent graphs. (<b>C</b>) Representative images of neuronal marker TUJ1 (red), TUNEL (green), and DAPI (blue) immunostaining in IM-FEN cells treated with vehicle and 10 nM TCDD for 24 h. Arrows point to the apoptotic cells (yellow) with condensed nuclei. Magnified image of the neuron shows the DNA fragmentation seen during apoptosis. Scale bar, 90 μm. The data represent three independent experiments. Results are mean ± SEM, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 1 Cont.
<p>TCDD induces apoptosis in IM-FEN cells. Cells were treated with vehicle or various doses of TCDD (0.1, 1, and 10 nM) for 24 h. Representative images of cleaved caspase-3 and cleaved PARP Western blots are included (<b>A</b>,<b>B</b>). Results from the protein bands’ density normalized to GAPDH have been included in the correspondent graphs. (<b>C</b>) Representative images of neuronal marker TUJ1 (red), TUNEL (green), and DAPI (blue) immunostaining in IM-FEN cells treated with vehicle and 10 nM TCDD for 24 h. Arrows point to the apoptotic cells (yellow) with condensed nuclei. Magnified image of the neuron shows the DNA fragmentation seen during apoptosis. Scale bar, 90 μm. The data represent three independent experiments. Results are mean ± SEM, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Cytotoxicity and apoptosis induced by TCDD are AHR-dependent. IM-FEN cells were pretreated with an AHR antagonist, CH-223191 (10 µM), for 1 h and then treated with 10 nM TCDD at different time points (3, 6, 12, and 24 h). (<b>A</b>) Cytotoxicity was assessed by an LDH release assay. The percentage of cytotoxicity was calculated relative to the maximum LDH release control (10% Triton<sup>®</sup> X-100). (<b>B</b>) Cell death by apoptosis was assessed by measuring Caspase-3/7 activity 1 h after adding the Caspase−Glo-3/7 reagent. Statistical analysis of LDH cytotoxicity assay data shows significant differences between vehicle and 10 nM TCDD after 6, 12, and 24 h of treatment. Not significant (ns) differences were observed between the vehicle and 10 nM TCDD + CH-223191 experimental groups. Results from the Caspase 3/7 assay show significant differences between vehicle and 10 nM TCDD treatment at all timepoints studied, whereas n.s. differences were observed when comparing the vehicle and 10 nM TCDD + CH-223191 experimental groups. Apoptosis was assessed by TUJ1/cleaved caspase-3 immunostaining of myenteric neurons isolated from (<b>C</b>) <span class="html-italic">Wnt1Cre<sup>−/−</sup>/Ahr<sup>b(fl/fl)</sup></span> (control) mice and (<b>D</b>) <span class="html-italic">Wnt1Cre<sup>+/−</sup>/Ahr<sup>b(fl/fl)</sup></span> (neural crest-specific <span class="html-italic">Ahr<sup>−/−</sup></span>) mice treated with vehicle and 10 nM TCDD for 24 h. Representative images show TUJ1 (red) and cleaved caspase-3 (green). The white arrow points to Tuj1<sup>+</sup> cell bodies, and the white arrowhead points to the axons. The yellow arrowhead points to cleaved caspase-3-positive neurons. Scale bar, 90 μm. Results are mean ± SEM; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2 Cont.
<p>Cytotoxicity and apoptosis induced by TCDD are AHR-dependent. IM-FEN cells were pretreated with an AHR antagonist, CH-223191 (10 µM), for 1 h and then treated with 10 nM TCDD at different time points (3, 6, 12, and 24 h). (<b>A</b>) Cytotoxicity was assessed by an LDH release assay. The percentage of cytotoxicity was calculated relative to the maximum LDH release control (10% Triton<sup>®</sup> X-100). (<b>B</b>) Cell death by apoptosis was assessed by measuring Caspase-3/7 activity 1 h after adding the Caspase−Glo-3/7 reagent. Statistical analysis of LDH cytotoxicity assay data shows significant differences between vehicle and 10 nM TCDD after 6, 12, and 24 h of treatment. Not significant (ns) differences were observed between the vehicle and 10 nM TCDD + CH-223191 experimental groups. Results from the Caspase 3/7 assay show significant differences between vehicle and 10 nM TCDD treatment at all timepoints studied, whereas n.s. differences were observed when comparing the vehicle and 10 nM TCDD + CH-223191 experimental groups. Apoptosis was assessed by TUJ1/cleaved caspase-3 immunostaining of myenteric neurons isolated from (<b>C</b>) <span class="html-italic">Wnt1Cre<sup>−/−</sup>/Ahr<sup>b(fl/fl)</sup></span> (control) mice and (<b>D</b>) <span class="html-italic">Wnt1Cre<sup>+/−</sup>/Ahr<sup>b(fl/fl)</sup></span> (neural crest-specific <span class="html-italic">Ahr<sup>−/−</sup></span>) mice treated with vehicle and 10 nM TCDD for 24 h. Representative images show TUJ1 (red) and cleaved caspase-3 (green). The white arrow points to Tuj1<sup>+</sup> cell bodies, and the white arrowhead points to the axons. The yellow arrowhead points to cleaved caspase-3-positive neurons. Scale bar, 90 μm. Results are mean ± SEM; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>TCDD-induced apoptosis in IM-FEN cells is independent of ER stress. Cells were treated with Veh and 10 nM TCDD for 24 h. Some cells received pre-treatment with the AHR antagonist CH-223191 (10 µM) for one hour before a 24 h exposure to 10 nM TCDD. Thapsigargin (300 nM, 5 h treatment) was used as a positive control for ER stress. Representative images of GRP78, IRE1α, and CHOP Western blots are included. Results from the protein bands’ density normalized to GAPDH have been included in the correspondent graphs. Results are mean ± SEM. Not significant (ns) differences were observed between vehicle and 10 nM TCDD with or without CH-223191 experimental groups.</p>
Full article ">Figure 4
<p>Regulation of ceramide biosynthesis and sphingolipid metabolism by TCDD in IM-FEN cells. Quantitative PCR (qPCR), neutral sphingomyelinase (N-SMase) activity assays, lipidomics, and chromatin immunoprecipitation (ChIP) assays to elucidate the effects of TCDD treatment on ceramide metabolism and gene regulation. (<b>A</b>) Heat map visualization shows the normalized expression levels of key genes involved in ceramide biosynthesis (<span class="html-italic">Sptlc1</span>, <span class="html-italic">Sptlc2</span>, <span class="html-italic">Cers2</span>, <span class="html-italic">Cser5</span>, <span class="html-italic">Cers6</span>, <span class="html-italic">Degs1</span>, <span class="html-italic">Smpd1</span>, <span class="html-italic">Smpd2</span>, <span class="html-italic">Smpd3</span>, and <span class="html-italic">Smpd4</span>) in IM-FEN cells treated with vehicle or 10 nM TCDD over various time points (30 min, 1, 3, 6, 12, and 24 h), with upregulated genes in red and downregulated genes in blue. Columns represent the timepoint, and rows represent individual genes. (<b>B</b>) N-SMase activity of vehicle and 10 nM TCDD-treated IM-FEN cells for 24 h. (<b>C</b>) Lipidomics analysis presented through a heatmap, illustrating the log2 mean-centered normalized data of sphingolipids in vehicle and 10 nM TCDD-treated IM-FEN cells, with tiles colored red for high abundance and blue for low abundance. (<b>D</b>) Statistically significant changes in sphingolipids, sorted into categories, are highlighted to show the alterations in ceramide, sphingomyelin, and hexosylceramide levels. log2 mean-centered data were imported into R, and the Complex Heatmap package was used to create the heatmap. (<b>E</b>,<b>F</b>) IM-FEN cells were treated with vehicle and 10 nM TCDD for 1 h. (<b>E</b>) ChIP-qPCR result for <span class="html-italic">Cyp1a1</span> gene promoter. (<b>F</b>) ChIP-qPCR results for <span class="html-italic">Sptlc2</span> and <span class="html-italic">Smpd2</span> gene promoters that were quantified by normalization with the corresponding input signal. Results are mean ± SEM, * <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 4 Cont.
<p>Regulation of ceramide biosynthesis and sphingolipid metabolism by TCDD in IM-FEN cells. Quantitative PCR (qPCR), neutral sphingomyelinase (N-SMase) activity assays, lipidomics, and chromatin immunoprecipitation (ChIP) assays to elucidate the effects of TCDD treatment on ceramide metabolism and gene regulation. (<b>A</b>) Heat map visualization shows the normalized expression levels of key genes involved in ceramide biosynthesis (<span class="html-italic">Sptlc1</span>, <span class="html-italic">Sptlc2</span>, <span class="html-italic">Cers2</span>, <span class="html-italic">Cser5</span>, <span class="html-italic">Cers6</span>, <span class="html-italic">Degs1</span>, <span class="html-italic">Smpd1</span>, <span class="html-italic">Smpd2</span>, <span class="html-italic">Smpd3</span>, and <span class="html-italic">Smpd4</span>) in IM-FEN cells treated with vehicle or 10 nM TCDD over various time points (30 min, 1, 3, 6, 12, and 24 h), with upregulated genes in red and downregulated genes in blue. Columns represent the timepoint, and rows represent individual genes. (<b>B</b>) N-SMase activity of vehicle and 10 nM TCDD-treated IM-FEN cells for 24 h. (<b>C</b>) Lipidomics analysis presented through a heatmap, illustrating the log2 mean-centered normalized data of sphingolipids in vehicle and 10 nM TCDD-treated IM-FEN cells, with tiles colored red for high abundance and blue for low abundance. (<b>D</b>) Statistically significant changes in sphingolipids, sorted into categories, are highlighted to show the alterations in ceramide, sphingomyelin, and hexosylceramide levels. log2 mean-centered data were imported into R, and the Complex Heatmap package was used to create the heatmap. (<b>E</b>,<b>F</b>) IM-FEN cells were treated with vehicle and 10 nM TCDD for 1 h. (<b>E</b>) ChIP-qPCR result for <span class="html-italic">Cyp1a1</span> gene promoter. (<b>F</b>) ChIP-qPCR results for <span class="html-italic">Sptlc2</span> and <span class="html-italic">Smpd2</span> gene promoters that were quantified by normalization with the corresponding input signal. Results are mean ± SEM, * <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 4 Cont.
<p>Regulation of ceramide biosynthesis and sphingolipid metabolism by TCDD in IM-FEN cells. Quantitative PCR (qPCR), neutral sphingomyelinase (N-SMase) activity assays, lipidomics, and chromatin immunoprecipitation (ChIP) assays to elucidate the effects of TCDD treatment on ceramide metabolism and gene regulation. (<b>A</b>) Heat map visualization shows the normalized expression levels of key genes involved in ceramide biosynthesis (<span class="html-italic">Sptlc1</span>, <span class="html-italic">Sptlc2</span>, <span class="html-italic">Cers2</span>, <span class="html-italic">Cser5</span>, <span class="html-italic">Cers6</span>, <span class="html-italic">Degs1</span>, <span class="html-italic">Smpd1</span>, <span class="html-italic">Smpd2</span>, <span class="html-italic">Smpd3</span>, and <span class="html-italic">Smpd4</span>) in IM-FEN cells treated with vehicle or 10 nM TCDD over various time points (30 min, 1, 3, 6, 12, and 24 h), with upregulated genes in red and downregulated genes in blue. Columns represent the timepoint, and rows represent individual genes. (<b>B</b>) N-SMase activity of vehicle and 10 nM TCDD-treated IM-FEN cells for 24 h. (<b>C</b>) Lipidomics analysis presented through a heatmap, illustrating the log2 mean-centered normalized data of sphingolipids in vehicle and 10 nM TCDD-treated IM-FEN cells, with tiles colored red for high abundance and blue for low abundance. (<b>D</b>) Statistically significant changes in sphingolipids, sorted into categories, are highlighted to show the alterations in ceramide, sphingomyelin, and hexosylceramide levels. log2 mean-centered data were imported into R, and the Complex Heatmap package was used to create the heatmap. (<b>E</b>,<b>F</b>) IM-FEN cells were treated with vehicle and 10 nM TCDD for 1 h. (<b>E</b>) ChIP-qPCR result for <span class="html-italic">Cyp1a1</span> gene promoter. (<b>F</b>) ChIP-qPCR results for <span class="html-italic">Sptlc2</span> and <span class="html-italic">Smpd2</span> gene promoters that were quantified by normalization with the corresponding input signal. Results are mean ± SEM, * <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>Short-Chain Ceramides induce cytotoxicity and apoptosis in IM-FEN cells. IM-FEN cells were treated with vehicle, 25 µM C<sub>2</sub>-ceramide, C<sub>6</sub>-ceramide, C<sub>2</sub>-DHC, and C<sub>6</sub>-DHC for 30 min, 1-, 3-, 6-, 12-, and 24-h. (<b>A</b>) Cytotoxicity was assessed by the LDH release assay. The percentage of cytotoxicity was calculated relative to the maximum LDH release control (10% Triton<sup>®</sup> X-100). (<b>B</b>) Apoptosis was assessed by measuring Caspase 3/7 activity. The statistical significance of the C<sub>2</sub>-ceramide and C<sub>6</sub>-ceramide treatment groups is shown with respect to vehicle. Not significant (ns) differences were observed between the vehicle and C<sub>2</sub>- and C<sub>6</sub>-DHC experimental groups. Results are mean ± SEM, * <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 Cont.
<p>Short-Chain Ceramides induce cytotoxicity and apoptosis in IM-FEN cells. IM-FEN cells were treated with vehicle, 25 µM C<sub>2</sub>-ceramide, C<sub>6</sub>-ceramide, C<sub>2</sub>-DHC, and C<sub>6</sub>-DHC for 30 min, 1-, 3-, 6-, 12-, and 24-h. (<b>A</b>) Cytotoxicity was assessed by the LDH release assay. The percentage of cytotoxicity was calculated relative to the maximum LDH release control (10% Triton<sup>®</sup> X-100). (<b>B</b>) Apoptosis was assessed by measuring Caspase 3/7 activity. The statistical significance of the C<sub>2</sub>-ceramide and C<sub>6</sub>-ceramide treatment groups is shown with respect to vehicle. Not significant (ns) differences were observed between the vehicle and C<sub>2</sub>- and C<sub>6</sub>-DHC experimental groups. Results are mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Ceramide involvement in TCDD-induced cytotoxicity in IM-FEN cells. Cells were pretreated with 10 µM myriocin, an inhibitor of ceramide synthesis, for an hour and then treated with 10 nM TCDD at different time points (3, 6, 12, and 24 h). Cytotoxicity was assessed by the LDH release assay. The percentage of cytotoxicity was calculated relative to the maximum LDH release control (10% Triton<sup>®</sup> X-100). The statistical significance of 10 nM TCDD with or without myriocin is shown with respect to vehicle. Results are mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001. ns: Not significant.</p>
Full article ">Figure 7
<p>Impact of TCDD and ceramide on neuronal survival pathways via Akt and GSK-3β modulation. IM-FEN cells were treated with vehicle and various doses of TCDD (0.1, 1, and 10 nM) and C<sub>2</sub> ceramide (25 µM) for 24 h. Representative images of Phospho-Akt (<b>A</b>,<b>C</b>) and Phospho-GSK-3β (<b>B</b>,<b>D</b>) Western blots are included. Results from the protein bands’ density normalized to corresponding Akt and GSK-3β have been included in the correspondent graphs. Results are mean ± SEM, *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 8
<p>A proposed model showing enhanced ceramide synthesis leading to apoptosis in IM-FEN cells. Key components showing activators in blue arrows and inhibitory associations in red lines.</p>
Full article ">
13 pages, 1566 KiB  
Article
Determination of Gb3 and Lyso-Gb3 in Fabry Disease-Affected Patients by LC-MRM/MS
by Gennaro Battaglia, Gabriella Pinto, Carolina Fontanarosa, Michele Spinelli, Anna Illiano, Stefania Serpico, Lorenzo Chiariotti, Roberta Risoluti, Stefano Materazzi and Angela Amoresano
Separations 2024, 11(8), 239; https://doi.org/10.3390/separations11080239 - 6 Aug 2024
Viewed by 501
Abstract
Limited or absent activity of the enzyme α-galactosidase A (α-Gal A), due to mutation in the related gene on the X chromosome, leads to the development of a rare hereditary and genetic disease known as Fabry disease (FD). This pathology involves a progressive [...] Read more.
Limited or absent activity of the enzyme α-galactosidase A (α-Gal A), due to mutation in the related gene on the X chromosome, leads to the development of a rare hereditary and genetic disease known as Fabry disease (FD). This pathology involves a progressive accumulation in various organs of the substrates of the enzyme e.g., globotriaosylceramide (Gb3) and its deacylated form, globotriaosylsphingosine (Lyso-Gb3), suggesting these molecules as biomarkers of Fabry disease. The present paper describes the development of an analytical strategy for the identification and quantification of Gb3 and Lyso-Gb3, in serum and blood samples by using liquid chromatography (LC) coupled to mass spectrometry in multiple reaction monitoring (MRM/MS) ion mode. The best experimental conditions were obtained by extracting the glycolipids with chloroform/methanol/H2O (2/1/0.3) and by separating them on a C4 column with a linear gradient (A: H2O with 2 mM ammonium formate. B: methanol with 1 mM ammonium formate, both acidified with 0.2% formic acid). The best transitions (a combination of precursor and fragment ions—m/z) were 786.8 m/z > 268.3 m/z for Lyso-GB3, 1137.3 m/z > 264.3 m/z for Gb3, 1039.3 m/z > 264.4 m/z for N-heptadecanoyl-ceramide trihexoside, and 843.5 m/z > 264.3 m/z for N-glycinated lyso-ceramide trihexoside, the latter being used as an internal standard. The developed method provided a reliable, fast, and effective procedure for direct measurements of GB3 and Lyso-GB3 in serum and blood for diagnosis of Fabry disease, suggesting this method as a complementary assay to the current enzymatic test. Therefore, this approach could open new insights into the clinical diagnostics of lysosomal storage disorders. Full article
(This article belongs to the Section Bioanalysis/Clinical Analysis)
Show Figures

Figure 1

Figure 1
<p>Total ion current chromatogram for the analysis of standard mixture solution at a concentration of 400 µg/L (<b>A</b>). MRM transition for the analysis of standard mixture solution at a concentration of 400 µg/L (<b>B</b>).</p>
Full article ">Figure 2
<p>Extraction tests of N-Glycinated lyso-ceramide trihexoside by using several solvents.</p>
Full article ">Figure 3
<p>TIC serum and blood of a representative control sample.</p>
Full article ">Figure 4
<p>Heatmap hierarchical clustering of GB3 and lyso-Gb3 in blood from Fabry and control subjects.</p>
Full article ">
17 pages, 3189 KiB  
Article
Functional Insights into the Sphingolipids C1P, S1P, and SPC in Human Fibroblast-like Synoviocytes by Proteomic Analysis
by Thomas Timm, Christiane Hild, Gerhard Liebisch, Markus Rickert, Guenter Lochnit and Juergen Steinmeyer
Int. J. Mol. Sci. 2024, 25(15), 8363; https://doi.org/10.3390/ijms25158363 - 31 Jul 2024
Viewed by 365
Abstract
The (patho)physiological function of the sphingolipids ceramide-1-phosphate (C1P), sphingosine-1-phosphate (S1P), and sphingosylphosphorylcholine (SPC) in articular joints during osteoarthritis (OA) is largely unknown. Therefore, we investigated the influence of these lipids on protein expression by fibroblast-like synoviocytes (FLSs) from OA knees. Cultured human FLSs [...] Read more.
The (patho)physiological function of the sphingolipids ceramide-1-phosphate (C1P), sphingosine-1-phosphate (S1P), and sphingosylphosphorylcholine (SPC) in articular joints during osteoarthritis (OA) is largely unknown. Therefore, we investigated the influence of these lipids on protein expression by fibroblast-like synoviocytes (FLSs) from OA knees. Cultured human FLSs (n = 7) were treated with 1 of 3 lipid species—C1P, S1P, or SPC—IL-1β, or with vehicle. The expression of individual proteins was determined by tandem mass tag peptide labeling followed by high-resolution electrospray ionization (ESI) mass spectrometry after liquid chromatographic separation (LC-MS/MS/MS). The mRNA levels of selected proteins were analyzed using RT-PCR. The 3sphingolipids were quantified in the SF of 18 OA patients using LC-MS/MS. A total of 4930 proteins were determined using multiplex MS, of which 136, 9, 1, and 0 were regulated both reproducibly and significantly by IL-1β, C1P, S1P, and SPC, respectively. In the presence of IL-1ß, all 3 sphingolipids exerted ancillary effects. Only low SF levels of C1P and SPC were found. In conclusion, the 3 lipid species regulated proteins that have not been described in OA. Our results indicate that charged multivesicular body protein 1b, metal cation symporter ZIP14, glutamine-fructose-6-P transaminase, metallothionein-1F and -2A, ferritin, and prosaposin are particularly interesting proteins due to their potential to affect inflammatory, anabolic, catabolic, and apoptotic mechanisms. Full article
(This article belongs to the Special Issue Proteomics and Its Applications in Disease 3.0)
Show Figures

Figure 1

Figure 1
<p>Venn diagram of proteins that are reproducibly regulated by (<b>A</b>) C1P (red), C1P in the presence of IL-1ß (yellow), and IL-1ß alone (green); (<b>B</b>) S1P (blue), S1P in the presence of IL-1ß (orange), and IL-1ß alone (green); (<b>C</b>) SPC in the presence of IL-1ß (violet) and IL-1ß alone (green); and (<b>D</b>) IL-1ß in the presence of C1P (yellow), S1P (orange), or SPC (violet). The number of proteins that can be reproducibly regulated is shown. In <a href="#app1-ijms-25-08363" class="html-app">Tables S1 and S2</a>, the AR of each protein is listed together with the results of the statistical analysis.</p>
Full article ">Figure 2
<p>C1P and S1P reproducibly and significantly regulate the level of proteins in FLSs. Protein levels of (<b>A</b>) stromelysin, charged multivesicular body protein 1b, metal cation symporter ZIP14; (<b>B</b>) cPLA2, cytochrome c oxidase subunit 1, SOD; and (<b>C</b>) long-chain-fatty-acid–CoA ligase 4, ICAM-1, and glutamine fructose-6-Ptransaminase were quantified by MS in duplicate in the 7 biological replicates. The dot plots represent the data obtained from the resulting 14 replicates and illustrate the x-fold abundance of the proteins in the treated FLS cells in comparison to that of only vehicle-treated controls (which are normalized to 1 and shown as a dotted line). The mean value ± SD is represented by lines within each figure (<span class="html-italic">n</span> = 7). * 0.05 ≥ <span class="html-italic">p</span> &gt; 0.01; ** 0.01 ≥ <span class="html-italic">p</span> &gt; 0.001; *** <span class="html-italic">p</span> ≥ 0.001.</p>
Full article ">Figure 3
<p>The biological processes of FLS being altered by C1P. The 9 proteins were reproducibly upregulated by more than 1.2-fold by C1P in FLS during 48 h of treatment, and <a href="#app1-ijms-25-08363" class="html-app">Table S1</a> provides further data on these proteins. The Go Slim categories for proteins were generated by Proteome Discoverer 2.5 software using the Gene Ontology (GO) database.</p>
Full article ">Figure 4
<p>The molecular functions of FLS being altered by C1P. The 9 proteins were reproducibly upregulated by more than 1.2-fold by C1P in FLS during 48 h of treatment, and <a href="#app1-ijms-25-08363" class="html-app">Table S1</a> provides further data on these proteins. The Go Slim categories for proteins were generated by Proteome Discoverer 2.5 software using the Gene Ontology (GO) database.</p>
Full article ">Figure 5
<p>The cellular localization of 9 proteins being regulated by C1P. The 9 proteins were reproducibly upregulated by more than 1.2-fold by C1P in FLS during 48 h of treatment, and <a href="#app1-ijms-25-08363" class="html-app">Table S1</a> provides further data on these proteins. The Go Slim categories for proteins were generated by Proteome Discoverer 2.5 software using the Gene Ontology (GO) database.</p>
Full article ">Figure 6
<p>Levels of C1P 16:0 ad SPC 18:1;O2 in knee SF of patients with early-stage or late-stage knee OA. Sphingolipids were quantified by LC-MS/MS in extracts of SF obtained from 8 patients with eOA (black circle) and 10 patients with lOA (blue open circle). Data presented indicate mean ± SD of lipid concentration in SF (N = 8 or 10).</p>
Full article ">
15 pages, 4065 KiB  
Article
Correlations between Skin Condition Parameters and Ceramide Profiles in the Stratum Corneum of Healthy Individuals
by Fuminari Akiyama, Natsumi Takahashi, Yuto Ueda, Shizuno Tada, Nobuyuki Takeuchi, Yusuke Ohno and Akio Kihara
Int. J. Mol. Sci. 2024, 25(15), 8291; https://doi.org/10.3390/ijms25158291 - 29 Jul 2024
Viewed by 398
Abstract
Ceramides are essential lipids for skin barrier function, and various classes and species exist in the human stratum corneum (SC). To date, the relationship between skin conditions and ceramide composition in healthy individuals has remained largely unclear. In the present study, we measured [...] Read more.
Ceramides are essential lipids for skin barrier function, and various classes and species exist in the human stratum corneum (SC). To date, the relationship between skin conditions and ceramide composition in healthy individuals has remained largely unclear. In the present study, we measured six skin condition parameters (capacitance, transepidermal water loss, scaliness, roughness, multilayer exfoliation, and corneocyte cell size) for the SC of the cheeks and upper arms of 26 healthy individuals and performed correlation analyses with their SC ceramide profiles, which we measured via liquid chromatography–tandem mass spectrometry. In the cheeks, high levels and/or ratios of two free ceramide classes containing an extra hydroxyl group in the long-chain moiety and a protein-bound ceramide class containing 6-hydroxysphingosine correlated with healthy skin conditions. In contrast, the ratios of two other free ceramide classes, both containing sphingosine, and a protein-bound ceramide class containing 4,14-sphingadiene correlated with unhealthy skin conditions, as did shortening of the carbon chain of the fatty acid portion of two ceramide classes containing non-hydroxy fatty acids. Thus, our findings help to elucidate the relationship between skin conditions and ceramide composition. Full article
Show Figures

Figure 1

Figure 1
<p>Skin condition parameters. The parameters ((<b>A</b>), capacitance; (<b>B</b>), transepidermal water loss [TEWL]; (<b>C</b>), multilayer exfoliation; (<b>D</b>), corneocyte cell size; (<b>E</b>), roughness; (<b>F</b>), scaliness) were measured for the cheeks and upper arms of healthy women (31–49 years old; n = 26) in winter. Boxes indicate the interquartile range (IQR), and lines in boxes represent the median. Whiskers indicate minimum and maximum values within 1.5 times the IQR. Circles and crosses represent outliers and means, respectively (** <span class="html-italic">p</span> &lt; 0.01; Welch’s <span class="html-italic">t</span>-test). AU, arbitrary unit.</p>
Full article ">Figure 2
<p>Skin condition parameters for the cheek in the dry and non-dry groups. The parameters ((<b>A</b>), capacitance; (<b>B</b>), transepidermal water loss [TEWL]; (<b>D</b>), multilayer exfoliation; (<b>E</b>), corneocyte cell size; (<b>G</b>), roughness; (<b>H</b>), scaliness) were measured in winter and were compared between the dry and non-dry groups (n = 13 each). Bars and whiskers indicate means and standard deviations (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; Welch’s <span class="html-italic">t</span>-test). AU, arbitrary unit. (<b>C</b>) After collecting the stratum corneum by tape stripping, samples were subjected to brilliant green/gentian violet staining. The images are typical examples from each of the groups. Scale bars, 200 μm. (<b>F</b>) Images of the skin surface were obtained using Visioscan VC98. The images are typical examples from each of the groups. Scale bars, 1 mm.</p>
Full article ">Figure 3
<p>Ceramide profiles. Stratum corneum samples were collected from the cheeks and upper arms of healthy women (31–49 years old; n = 26) by tape stripping in winter, and ceramides were quantified via liquid chromatography coupled with tandem mass spectrometry. Quantities of free ceramides (<b>A</b>), protein-bound ceramides (<b>B</b>), and total ceramides (<b>C</b>) are shown in box-and-whisker plots. Boxes indicate the interquartile range (IQR), and lines in boxes represent the median. Whiskers indicate minimum and maximum values within 1.5 times the IQR. Circles and crosses represent outliers and means, respectively (** <span class="html-italic">p</span> &lt; 0.01; Welch’s <span class="html-italic">t</span>-test). AU, arbitrary unit. The proportions of free ceramides (<b>D</b>) and protein-bound ceramides (<b>E</b>) comprised by each ceramide class are shown in pie charts.</p>
Full article ">Figure 4
<p>Ceramide class composition of cheek samples from the dry and non-dry groups. Quantities of ceramides ((<b>A</b>), free ceramide classes; (<b>B</b>), protein-bound ceramide classes; (<b>C</b>), total ceramides) and ratios of ceramide classes to total ceramides ((<b>D</b>), free ceramide classes; (<b>E</b>), protein-bound ceramide classes) in the cheeks in winter were compared between the dry and non-dry groups (n = 13 each). Bars and whiskers represent means and standard deviations (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; Welch’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 5
<p>Correlations between ceramide class ratios and skin condition parameters. Correlations were analyzed between seven ceramide classes (NS, NP, AS, EOS, EOH, PB-H, and PB-SD) and six skin condition parameters (capacitance, transepidermal water loss [TEWL], scaliness, roughness, multilayer exfoliation, and corneocyte cell size) for samples taken from the cheeks in winter. (<b>A</b>) The strength of each correlation is indicated in the heatmap. Red and blue indicate correlations with healthy and unhealthy skin conditions, respectively. The numbers are the correlation coefficients (<span class="html-italic">R</span>). (<b>B</b>) Representative scatter plots of the ceramide class ratios and skin condition parameters (magenta, dry group; light blue, non-dry group).</p>
Full article ">Figure 6
<p>Ceramide chain lengths for the cheek in the dry and non-dry groups. Weighted averages of the chain lengths of free ceramide classes (<b>A</b>) and fatty acid composition of NS (<b>B</b>) and NH (<b>C</b>) for the cheeks in winter were compared between the dry and non-dry groups (n = 13 each). Bars and whiskers represent means and standard deviations (* <span class="html-italic">p</span> &lt; 0.05; Welch’s <span class="html-italic">t</span>-test). (<b>D</b>) Analysis of correlations in the NS and NH classes between FA chain length and skin condition parameters (capacitance, transepidermal water loss [TEWL], scaliness, roughness, multilayer exfoliation, and corneocyte cell size) for the cheeks in winter. The strength of each correlation is indicated in the heatmap. Red and blue indicate correlation with healthy and unhealthy skin conditions, respectively. The numbers are the correlation coefficients (<span class="html-italic">R</span>). (<b>E</b>) Representative scatter plots of the fatty acid chain lengths of NS and NH and skin condition parameters (magenta, dry group; light blue, non-dry group).</p>
Full article ">Figure 7
<p>Differences in ceramide levels and class composition in the cheeks between seasons. Stratum corneum samples were collected from the cheeks of healthy women (31–49 years old; n = 26) by tape stripping in summer and winter, and ceramides were quantified via liquid chromatography coupled with tandem mass spectrometry. (<b>A</b>) Bars and whiskers represent means and standard deviations of total ceramide quantities (** <span class="html-italic">p</span> &lt; 0.01; paired Student’s <span class="html-italic">t</span>-test). (<b>B</b>) The ratios of the ceramide classes are shown in pie charts.</p>
Full article ">
17 pages, 2028 KiB  
Article
Sphingosine 1-Phosphate Stimulates ER to Golgi Ceramide Traffic to Promote Survival in T98G Glioma Cells
by Paola Giussani, Loredana Brioschi, Enida Gjoni, Elena Riccitelli and Paola Viani
Int. J. Mol. Sci. 2024, 25(15), 8270; https://doi.org/10.3390/ijms25158270 - 29 Jul 2024
Viewed by 347
Abstract
Glioblastoma multiforme is the most common and fatal brain tumor among human cancers. Ceramide (Cer) and Sphingosine 1-phosphate (S1P) have emerged as bioeffector molecules that control several biological processes involved in both cancer development and resistance. Cer acts as a tumor suppressor, inhibiting [...] Read more.
Glioblastoma multiforme is the most common and fatal brain tumor among human cancers. Ceramide (Cer) and Sphingosine 1-phosphate (S1P) have emerged as bioeffector molecules that control several biological processes involved in both cancer development and resistance. Cer acts as a tumor suppressor, inhibiting cancer progression, promoting apoptosis, enhancing immunotherapy and sensitizing cells to chemotherapy. In contrast, S1P functions as an onco-promoter molecule, increasing proliferation, survival, invasiveness, and resistance to drug-induced apoptosis. The pro-survival PI3K/Akt pathway is a recognized downstream target of S1P, and we have previously demonstrated that in glioma cells it also improves Cer transport and metabolism towards complex sphingolipids in glioma cells. Here, we first examined the possibility that, in T98G glioma cells, S1P may regulate Cer metabolism through PI3K/Akt signaling. Our research showed that exogenous S1P increases the rate of vesicular trafficking of Cer from the endoplasmic reticulum (ER) to the Golgi apparatus through S1P receptor-mediated activation of the PI3K/Akt pathway. Interestingly, the effect of S1P results in cell protection against toxicity arising from Cer accumulation in the ER, highlighting the role of S1P as a survival factor to escape from the Cer-generating cell death response. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of S1P and PTX on AKT phosphorylation in T98G glioma cells. T98G cells were serum-starved for 24 h and then: (<b>panel A</b>) incubated for the indicated times in serum-free medium at 37 °C in the presence or absence (control) of 200 nM S1P; ** <span class="html-italic">p</span> &lt; 0.01 versus time 0 (one-way ANOVA followed by Tukey’s post hoc test); (<b>panel B</b>) pre-treated in serum-free medium for 2 h with 100 ng/mL (100 μM) PTX. Then, 200 nM S1P was added for 30 min. Cells were harvested for immunoblot analysis of phospho-AKT and AKT levels. All values are the mean ± S.D. of at least three individual experiments. *** <span class="html-italic">p</span> &lt; 0.001 versus CT (one-way ANOVA followed by Tukey’s post hoc test).</p>
Full article ">Figure 2
<p>Effect of S1P on [<sup>3</sup>H]Sph inT98G glioma cells treated with or without PTX and BFA. T98G cells were serum-starved for 24 h and then: (<b>panel A</b>) pulsed with 0.15 μCi/mL [C3-<sup>3</sup>H]Sph for 1 h; (<b>panel B</b>) incubated in serum-free medium in the absence or presence of 100 ng/mL PTX. After 2 h, cells were pulsed with 0.15 μCi/mL [C3-<sup>3</sup>H]Sph for 30 min (<b>panel C</b>) and then pulsed with 0.15 μCi/mL [C3-<sup>3</sup>H]Sph for 30 min at 37 °C in the absence or presence of 1 μg/mL BFA. Then the cells were chased for (<b>panel A</b>) the indicated times (<b>panel B</b>, <b>C</b>) for 1 h, at 37 °C in the presence or absence of 200 nM S1P in the absence or presence of (<b>panel B</b>) 100 ng/mL PTX; (<b>panel C</b>) 1 μg/mL BFA. Radioactivity associated with each lipid was determined. (<b>panel A</b>) HPTLC plate. Values are expressed as nCi/mg protein (<b>panel A</b>–<b>C</b>). All values are the mean ± S.D. of at least three individual experiments performed in triplicate. * <span class="html-italic">p</span> &lt; 0.05 versus Ctrl; ** <span class="html-italic">p</span> &lt; 0.01 versus Ctrl (one-way ANOVA followed by Tukey’s post hoc test).</p>
Full article ">Figure 2 Cont.
<p>Effect of S1P on [<sup>3</sup>H]Sph inT98G glioma cells treated with or without PTX and BFA. T98G cells were serum-starved for 24 h and then: (<b>panel A</b>) pulsed with 0.15 μCi/mL [C3-<sup>3</sup>H]Sph for 1 h; (<b>panel B</b>) incubated in serum-free medium in the absence or presence of 100 ng/mL PTX. After 2 h, cells were pulsed with 0.15 μCi/mL [C3-<sup>3</sup>H]Sph for 30 min (<b>panel C</b>) and then pulsed with 0.15 μCi/mL [C3-<sup>3</sup>H]Sph for 30 min at 37 °C in the absence or presence of 1 μg/mL BFA. Then the cells were chased for (<b>panel A</b>) the indicated times (<b>panel B</b>, <b>C</b>) for 1 h, at 37 °C in the presence or absence of 200 nM S1P in the absence or presence of (<b>panel B</b>) 100 ng/mL PTX; (<b>panel C</b>) 1 μg/mL BFA. Radioactivity associated with each lipid was determined. (<b>panel A</b>) HPTLC plate. Values are expressed as nCi/mg protein (<b>panel A</b>–<b>C</b>). All values are the mean ± S.D. of at least three individual experiments performed in triplicate. * <span class="html-italic">p</span> &lt; 0.05 versus Ctrl; ** <span class="html-italic">p</span> &lt; 0.01 versus Ctrl (one-way ANOVA followed by Tukey’s post hoc test).</p>
Full article ">Figure 3
<p>Effect of CERT downregulation on the intracellular distribution of BODIPY-C<sub>5</sub>Cer in T98G glioma cells. Cells grown in DMEM supplemented with 10% FCS were transfected with a mix of H87 and H424 siRNA for CERT (siCERT) and the corresponding non-targeting NT87 and H424 as control (siNT) as described in Mat. and Meth. (<b>Panel A</b>) the cells were analyzed for CERT levels. 72 h after transfection, cells were washed twice with PBS and harvested. Cell lysates (20 μg of protein) from two different preparations of siNT and siCERT transfected cells were analyzed by immunoblotting with a polyclonal anti-CERT antibody and polyclonal anti-GAPDH antibody. *** <span class="html-italic">p</span> &lt; 0.001 versus siCT cells (one-way ANOVA followed by Tukey’s post hoc test). (<b>Panel B</b>) the cells were analyzed for fluorescence distribution. T98G glioma cells grown on a coverslip were transfected for silencing as previously described and 48 h after transfection, cells were pre-treated in serum-free medium at 37 °C for 30 min with or without 200 nM S1P and/or 10 μM LY294002 and then, cells were incubated with 2.5 μM BODIPY-C<sub>5</sub>Cer for 30 min at 4 °C. Labeled cells were further incubated at 37 °C for the indicated times with or without 200 nM S1P in the presence or absence of 10 μM LY294002 in order to allow the intracellular redistribution of fluorescent ceramides. Images are representative of at least five different experiments and were identically processed and printed.</p>
Full article ">Figure 4
<p>Effect of etoposide on ceramide biosynthesis and viability in T98G glioma cells and rescue activity of myriocin, fumonisin B1, and S1P on etoposide-induced cell toxicity. T98G cells were serum-starved for 24 h and then incubated in serum-free medium in the absence or presence of increasing doses of etoposide, and cell viability was assessed after 72 h of incubation (<b>panel A</b>), * <span class="html-italic">p</span> &lt; 0.05 versus “0” (one-way ANOVA followed by Tukey’s post hoc test); ** <span class="html-italic">p</span> &lt; 0.01 versus “0” (one-way ANOVA followed by Tukey’s post hoc test); serum-starved cells were pre-treated or not for 1 h with 400 µM etoposide (ETO), then pulsed with 3 μCi/mL [<sup>14</sup>C]Palmitate for 6 h with or without 400 μM etoposide, after that, radioactivity associated with each lipid was determined. Values are expressed as nCi/mg protein. All values are the mean ± S.D. of at least three individual experiments performed in triplicate (<b>panel B</b>), ** <span class="html-italic">p</span> &lt; 0.01 versus CT (one-way ANOVA followed by Tukey’s post hoc test); serum-starved cells were incubated with 10 μM myriocin (MYR) or fumonisin B1 (Fb1) in the presence or absence of 400 μM ETO and cell viability was assessed 24 h later (<b>panel C</b>); starved cells were treated for 24 h with 400 μM etoposide alone, or together with 200 nM S1P, 200 nM S1P and 10 μM LY294002 (LY29) or 10 μM LY29 alone. (<b>panel D</b>) Cell viability was evaluated by the MTT assay. The viability of untreated cells was regarded as 100%. Mean ± S.D. is shown. (<b>Panel C</b>,<b>D</b>): One-way ANOVA with Tukey post hoc test. Data are presented as mean ± standard deviation ** <span class="html-italic">p</span> &lt; 0.01; * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
19 pages, 1952 KiB  
Article
Diagnostic Value of Menstrual Blood Lipidomics in Endometriosis: A Pilot Study
by Natalia Starodubtseva, Vitaliy Chagovets, Alisa Tokareva, Madina Dumanovskaya, Eugenii Kukaev, Anastasia Novoselova, Vladimir Frankevich, Stanislav V. Pavlovich and Gennady Sukhikh
Biomolecules 2024, 14(8), 899; https://doi.org/10.3390/biom14080899 - 24 Jul 2024
Viewed by 394
Abstract
Endometriosis is a prevalent chronic inflammatory disease characterized by a considerable delay between initial symptoms and diagnosis through surgery. The pressing need for a timely, non-invasive diagnostic solution underscores the focus of current research efforts. This study examines the diagnostic potential of the [...] Read more.
Endometriosis is a prevalent chronic inflammatory disease characterized by a considerable delay between initial symptoms and diagnosis through surgery. The pressing need for a timely, non-invasive diagnostic solution underscores the focus of current research efforts. This study examines the diagnostic potential of the menstrual blood lipidome. The lipid profile of 39 samples (23 women with endometriosis and 16 patients in a control group) was acquired using reverse-phase high-performance liquid chromatography–mass spectrometry with LipidMatch processing and identification. Profiles were normalized based on total ion counts. Significant differences in lipids were determined using the Mann–Whitney test. Lipids for the diagnostic model, based on logistic regression, were selected using a combination of variance importance projection filters and Akaike information criteria. Levels of ceramides, sphingomyelins, cardiolipins, triacylglycerols, acyl- and alkenyl-phosphatidylethanolamines, and alkenyl-phosphatidylcholines increased, while acyl- and alkyl-phosphatidylcholines decreased in cases of endometriosis. Plasmenylphosphatidylethanolamine PE P-16:0/18:1 and cardiolipin CL 16:0_18:0_22:5_22:6 serve as marker lipids in the diagnostic model, exhibiting a sensitivity of 81% and specificity of 85%. The diagnostic approach based on dried spots of menstrual blood holds promise as an alternative to traditional non-invasive methods for endometriosis screening. Full article
Show Figures

Figure 1

Figure 1
<p>The association of the dried menstrual blood spot lipids with endometriosis. (<b>A</b>) A volcano plot of lipids detected in the dried menstrual blood spots. Orange color marks lipids with statistically significant differences between groups studied (<span class="html-italic">p</span>-value &lt; 0.05, Mann–Whitney test) and a median fold change greater than 1.5 is labeled. (<b>B</b>) Correlation diagram of the lipids with the severity of endometriosis (stage) (<span class="html-italic">p</span> &lt; 0.05). X represents the absence of a significant association. Cer-NS—ceramide, Cer-NP—physphingosine ceramide, CL—cardiolipine, oxCL—oxidized cardiolipins, PC—phosphatidylcholine, PC O—plasmanylphosphatidylcholines, PC P—plasmenylphosphatidylcholines, oxPC—oxidized phosphatidylcholines, PE P—plasmenylphosphatidylethanolamine, LPE P —plasmenyllysophosphatidylethanolamine, PG — phosphatidylglycerol, TG —triacylglycerol.</p>
Full article ">Figure 2
<p>Base peak chromatogram (black), extracted ion chromatogram of PE P-16:0/18:1 (red) and extracted ion chromatogram of CL 16:0_18:0_22:5_22:6 (blue) with tandem mass spectra. Extracted ion chromatogram of CL 16:0_18:0_22:5_22:6 is scaled five times. Negative ion mode.</p>
Full article ">Figure 3
<p>Receiver operating characteristic curve obtained after cross-validation for logistic regression model developed (1). Coefficients in endometriosis diagnostic model, based on logic regression, are presented underneath.</p>
Full article ">Figure 4
<p>Diagnostic potential of plasmenylphosphatidylethanolamine PE P-16:0/18:1 and cardiolipin CL 16:0_18:0_22:5_22:6: (<b>A</b>) Boxplots illustrating the levels of these markers in menstrual blood; (<b>B</b>) dried menstrual blood spots plotted in the space of the relative proportion of plasmenyl-phosphatidylethanolamine PE P-16:0/18:1 and cardiolipin CL 16:0_18:0_22:5_22:6. The green points represent samples from the control group patients, while the ocher points represent samples from the endometriosis group.</p>
Full article ">
15 pages, 15132 KiB  
Article
Ceramide Ehux-C22 Targets the miR-199a-3p/mTOR Signaling Pathway to Regulate Melanosomal Autophagy in Mouse B16 Cells
by Jiyue Wan, Shumiao Zhang, Guiling Li, Shiying Huang, Jian Li, Zhengxiao Zhang and Jingwen Liu
Int. J. Mol. Sci. 2024, 25(15), 8061; https://doi.org/10.3390/ijms25158061 - 24 Jul 2024
Viewed by 423
Abstract
Melanosomes are specialized membrane-bound organelles where melanin is synthesized and stored. The levels of melanin can be effectively reduced by inhibiting melanin synthesis or promoting melanosome degradation via autophagy. Ceramide, a key component in the metabolism of sphingolipids, is crucial for preserving the [...] Read more.
Melanosomes are specialized membrane-bound organelles where melanin is synthesized and stored. The levels of melanin can be effectively reduced by inhibiting melanin synthesis or promoting melanosome degradation via autophagy. Ceramide, a key component in the metabolism of sphingolipids, is crucial for preserving the skin barrier, keeping it hydrated, and warding off the signs of aging. Our preliminary study indicated that a long-chain C22-ceramide compound (Ehux-C22) isolated from the marine microalga Emiliania huxleyi, reduced melanin levels via melanosomal autophagy in B16 cells. Recently, microRNAs (miRNAs) were shown to act as melanogenesis-regulating molecules in melanocytes. However, whether the ceramide Ehux-C22 can induce melanosome autophagy at the post-transcriptional level, and which potential autophagy-dependent mechanisms are involved, remains unknown. Here, miR-199a-3p was screened and identified as a novel upregulated miRNA in Ehux-C22-treated B16 cells. An in vitro high melanin expression model in cultured mouse melanoma cells (B16 cells) was established by using 0.2 μM alpha-melanocyte-stimulating hormone(α-MSH) and used for subsequent analyses. miR-199a-3p overexpression significantly enhanced melanin degradation, as indicated by a reduction in the melanin level and an increase in melanosome autophagy. Further investigation demonstrated that in B16 cells, Ehux-C22 activated miR-199a-3p and inhibited mammalian target of rapamycin(mTOR) level, thus activating the mTOR-ULK1 signaling pathway by promoting the expression of unc-51-like autophagy activating kinase 1 (ULK1), B-cell lymphoma-2 (Bcl-2), Beclin-1, autophagy-related gene 5 (ATG5), and microtubule-associated protein light chain 3 (LC3-II) and degrading p62. Therefore, the roles of Ehux-C22-regulated miR-199a-3p and the mTOR pathway in melanosomal autophagy were elucidated. This research may provide novel perspectives on the post-translational regulation of melanin metabolism, which involves the coordinated control of melanosomes. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Impact of Ehux-C22 on the regulation of autophagy-associated miRNAs and their respective target genes in B16 cells: (<b>A</b>) Analysis of miRNA expression levels; (<b>B</b>) Assessment of target gene mRNA levels. Error bars indicate means ± SD of three biological replicates. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>The targeting relationship between miR-199a-3p and the target gene mTOR: (<b>A</b>) The miR-199a-3p target locus was within the 3′UTR of mTOR. Red represented the seed sequence, and green represented the mutant region. (<b>B</b>) The dual-luciferase reporter assay was employed to measure the Luciferase relative activity. Error bars indicate means ± SD of three biological replicates. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>The expression level of miR-199a-3p in B16 cells. After transfection with miR-199a-3p or miR-199a-3p inhibitor for 6 h, B16 cells were incubated in DMEM with 0.2 µM α-MSH, and Ehux-C22 was added according to the group, followed by incubation for 48 h. Error bars indicate means ± SD of three biological replicates. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Effect of miR-199a-3p on melanin levels in B16 cells: (<b>A</b>–<b>D</b>) Images were observed by optical microscopy at 100× magnification. (<b>A</b>) Blank control: 0.2 µM α-MSH; (<b>B</b>) 0.2 µM α-MSH+2 µM Ehux-C22; (<b>C</b>) 0.2 µM α-MSH+2 µM Ehux-C22 + miR-199a-3p inhibitor; (<b>D</b>) 0.2 µM α-MSH+miR-199a-3p. (<b>E</b>) The absorbance (490 nm) was measured to calculate the melanin level. Error bars indicate means ± SD of three biological replicates. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Electron micrograph of melanosomes structure in B16 cells: (<b>A</b>) Blank control: 0.2 µM α-MSH; (<b>B</b>) 0.2 µM α-MSH+2 µM Ehux-C22; (<b>C</b>) 0.2 µM α-MSH + 2 µM Ehux-C22 + miR-199a-3p inhibitor; (<b>D</b>) 0.2 µM α-MSH+miR-199a-3p. The scale bars represent 5.0 µm (<b>A-1</b>–<b>D-1</b>), 1.0 µm (<b>A-2</b>–<b>D-2</b>) and 0.5 µm (<b>A-3</b>–<b>D-3</b>). M-I/II/III/IV: I/II/III/IV stage melanosome; V: vacuole; AS: autolysosome; MC: melanosome complex; N: nucleus.</p>
Full article ">Figure 6
<p>Impact of miR-199a-3p on autophagic flux in B16 cells: (<b>A</b>) Two hours after transfection with mRFP-GFP-LC3 adenovirus, B16 cells were treated with control: 0.2 µM α-MSH, 0.2 µM α-MSH+2 µM Ehux-C22, 0.2 µM α-MSH+miR-199a-3p or 0.2 µM α-MSH+Ehux-C22+miR-199a-3p inhibitor. After 36 h, fluorescent LC3-positive puncta were observed by confocal microscopy. (<b>B</b>) Autolysosomes (red puncta) were quantified as the number of puncta (red puncta/total puncta) in the merged images. (<b>C</b>) Autophagosomes (yellow puncta) were quantified as the number of puncta (yellow puncta/total puncta) in the merged images. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>Impact of miR-199a-3p on the expression of mTOR in B16 cells: (<b>A</b>) Representative band from a Western blot for mTOR; (<b>B</b>) quantitative analysis by ImageJ 1.54d 30. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Protein expression levels of key autophagy-related components of the mTOR-ULK1 signaling pathway: (<b>A</b>) Representative Western blot bands for unc-51-like autophagy activating kinase 1 (ULK1), Beclin-1, p-Beclin-1 (Ser15), p-Bcl-2 (Ser70), autophagy-related gene 5 (ATG5), p62, and microtubule-associated protein light chain 3 (LC3-II/I). (<b>B</b>) Relative quantitative analysis of proteins by ImageJ 1.54d 30. * <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>Signaling pathway through which Ehux-C22 regulates miR-199a-3p targeting mTOR to induce melanosome autophagy in B16 cells.</p>
Full article ">
25 pages, 5039 KiB  
Article
Functional Nanostructured Lipid Carrier-Enriched Hydrogels Tailored to Repair Damaged Epidermal Barrier
by Radwan Joukhadar, Laura Nižić Nodilo, Jasmina Lovrić, Anita Hafner, Ivan Pepić and Mario Jug
Gels 2024, 10(7), 466; https://doi.org/10.3390/gels10070466 - 16 Jul 2024
Viewed by 711
Abstract
In this study, a functional nanostructured lipid carriers (NLCs)-based hydrogel was developed to repair the damaged epidermal skin barrier. NLCs were prepared via a high-energy approach, using argan oil and beeswax as liquid and solid lipids, respectively, and were loaded with ceramides and [...] Read more.
In this study, a functional nanostructured lipid carriers (NLCs)-based hydrogel was developed to repair the damaged epidermal skin barrier. NLCs were prepared via a high-energy approach, using argan oil and beeswax as liquid and solid lipids, respectively, and were loaded with ceramides and cholesterol at a physiologically relevant ratio, acting as structural and functional compounds. Employing a series of surfactants and optimizing the preparation conditions, NLCs of 215.5 ± 0.9 nm in size and a negative zeta potential of −42.7 ± 0.9 were obtained, showing acceptable physical and microbial stability. Solid state characterization by differential scanning calorimetry and X-ray powder diffraction revealed the formation of imperfect crystal NLC-type. The optimized NLC dispersion was loaded into the gel based on sodium hyaluronate and xanthan gum. The gels obtained presented a shear thinning and thixotropic behavior, which is suitable for dermal application. Incorporating NLCs enhanced the rheological, viscoelastic, and textural properties of the gel formed while retaining the suitable spreadability required for comfortable application and patient compliance. The NLC-loaded gel presented a noticeable occlusion effect in vitro. It provided 2.8-fold higher skin hydration levels on the ex vivo porcine ear model than the NLC-free gel, showing a potential to repair the damaged epidermal barrier and nourish the skin actively. Full article
(This article belongs to the Special Issue Multifunctional Hydrogel for Wound Healing and Tissue Repair)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The effect of the selected emulsifier/GMS blends on the properties of NLCs formed: mean particle size (<span class="html-italic">MPS</span>, <b>A</b>), polydispersity index (<span class="html-italic">PDI</span>, <b>B</b>), particle size distribution (<b>C</b>), zeta potential (ζ, <b>D</b>), and concentration of the particles formed (<b>E</b>). One asterisk (*) denotes a statistically significant difference compared to PS80 (<span class="html-italic">p</span> &lt; 0.0001), while two asterisks (**) denote a statistically insignificant difference compared to PS80 (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 2
<p>The effect of the homogenization rate on the mean particle size (MPS, <b>A</b>), polydispersity index (PDI, <b>A</b>), zeta potential (ζ, <b>B</b>), and particle concentration (<b>C</b>) of NLC_PS80 dispersion. One asterisk (*) denotes a statistically significant difference compared to the sample obtained at 12,000 rpm (<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 3
<p>Solid-state analysis of lipid components and freeze-dried NLC_PS80 formulation performed by DSC (<b>A</b>,<b>B</b>) and XRPD (<b>C</b>).</p>
Full article ">Figure 4
<p>Shear viscosity profiles of blank (<b>left</b>) and NLC-loaded HaXa hydrogels (<b>right</b>) containing 1% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) of HA and variable amounts of XA (0.5, 1.0, 1.5, and 2.0% (<span class="html-italic">w</span>/<span class="html-italic">w</span>), respectively). The shear viscosity curve of the optimized NLC dispersion (NLC_PS80) was added for comparison.</p>
Full article ">Figure 5
<p>Dependence of storage (<span class="html-italic">G</span>′) and loss (<span class="html-italic">G</span>″) moduli on shear strain for blank (<b>left</b>) and NLC-loaded HaXa hydrogels (<b>right</b>) containing 1% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) of HA and variable amounts of XA (0.5, 1.0, 1.5, and 2.0% (<span class="html-italic">w</span>/<span class="html-italic">w</span>), respectively).</p>
Full article ">Figure 6
<p>Storage (<span class="html-italic">G</span>′) and loss (<span class="html-italic">G</span>″) moduli as a function of angular frequency (ω) for blank and NLC-loaded HaXa hydrogel containing 1% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) of HA and 0.5% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) of XA.</p>
Full article ">Figure 7
<p>Textural properties of blank and NLC-loaded HaXa hydrogels containing 1% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) of HA and variable amounts of XA (0.5, 1.0, 1.5, and 2.0% (<span class="html-italic">w</span>/<span class="html-italic">w</span>), respectively) obtained by the backward extrusion test. Asterisks (*) denote a statistically significant difference compared to the corresponding blank HaXa gel (<span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 8
<p>Spreadability parameters (strength and work of shear) of blank and NLC-loaded HaXa hydrogels containing 1% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) of HA and variable amounts of XA (0.5, 1.0, 1.5, and 2.0% (<span class="html-italic">w</span>/<span class="html-italic">w</span>), respectively) obtained by the TTC spreadability test. Asterisks (*) denote a statistically significant difference compared to the corresponding blank HaXa hydrogel (<span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 9
<p>Biocompatibility of NLC_PS80 dispersion and NLC_HaXa_0.5 hydrogel on HaCaT cell line tested by MTT reduction assay. One asterisk (*) denotes a statistically significant difference compared to the negative control (<span class="html-italic">p</span> &lt; 0.001), while two asterisks (**) denote a statistically significant difference compared to the NLC_PS80 dispersion at the same concentration (<span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 10
<p>In vitro occlusion factor (OF) as a function of time for selected blank and NLC-loaded HaXa hydrogels containing 1% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) of HA and 0.5% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) of XA. Vaseline was used as a positive control. Asterisks (*) denote a statistically significant difference compared to the corresponding blank HaXa gel (<span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 11
<p>The relative changes in the skin hydration level (Δ<span class="html-italic">SHL</span>) observed after applying the blank and NLC-loaded HaXa_0.5 hydrogels on the ex vivo porcine ear model with disrupted epidermal barrier by SLS treatment. Untreated skin served as a control. One asterisk (*) denotes a statistically significant difference compared to the control (<span class="html-italic">p</span> &lt; 0.001), while two asterisks (**) denote a statistically significant difference compared to the skin treated with blank HaXa_0.5 gel (<span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 12
<p>The preparation protocol of NLC dispersions.</p>
Full article ">
18 pages, 757 KiB  
Review
The Utility of Lipidomic Analysis in Colorectal Cancer Diagnosis and Prognosis—A Systematic Review of Recent Literature
by Jakub Klekowski, Mariusz Chabowski, Małgorzata Krzystek-Korpacka and Mariusz Fleszar
Int. J. Mol. Sci. 2024, 25(14), 7722; https://doi.org/10.3390/ijms25147722 - 14 Jul 2024
Viewed by 807
Abstract
Colorectal cancer (CRC) is among the most prevalent and lethal malignancies. Lipidomic investigations have revealed numerous disruptions in lipid profiles across various cancers. Studies on CRC exhibit potential for identifying novel diagnostic or prognostic indicators through lipidomic signatures. This review examines recent literature [...] Read more.
Colorectal cancer (CRC) is among the most prevalent and lethal malignancies. Lipidomic investigations have revealed numerous disruptions in lipid profiles across various cancers. Studies on CRC exhibit potential for identifying novel diagnostic or prognostic indicators through lipidomic signatures. This review examines recent literature regarding lipidomic markers for CRC. PubMed database was searched for eligible articles concerning lipidomic biomarkers of CRC. After selection, 36 articles were included in the review. Several studies endeavor to establish sets of lipid biomarkers that demonstrate promising potential to diagnose CRC based on blood samples. Phosphatidylcholine, phosphatidylethanolamine, ceramides, and triacylglycerols (TAGs) appear to offer the highest diagnostic accuracy. In tissues, lysophospholipids, ceramides, and TAGs were among the most altered lipids, while unsaturated fatty acids also emerged as potential biomarkers. In-depth analysis requires both cell culture and animal studies. CRC involves multiple lipid metabolism alterations. Although numerous lipid species have been suggested as potential diagnostic markers, the establishment of standardized methods and the conduct of large-scale studies are necessary to facilitate their clinical application. Full article
Show Figures

Figure 1

Figure 1
<p>PRISMA flowchart of the literature review process.</p>
Full article ">
22 pages, 3203 KiB  
Article
Deletion of Sphingosine Kinase 2 Attenuates Acute Kidney Injury in Mice with Hemolytic-Uremic Syndrome
by Tina Müller, Nadine Krieg, Antonia I. Lange-Polovinkin, Bianka Wissuwa, Markus H. Gräler, Sophie Dennhardt and Sina M. Coldewey
Int. J. Mol. Sci. 2024, 25(14), 7683; https://doi.org/10.3390/ijms25147683 - 12 Jul 2024
Viewed by 456
Abstract
Typical hemolytic uremic syndrome (HUS) can occur as a severe systemic complication of infections with Shiga toxin (Stx)-producing Escherichia coli. Its pathology can be induced by Stx types, resulting in toxin-mediated damage to renal barriers, inflammation, and the development of acute kidney [...] Read more.
Typical hemolytic uremic syndrome (HUS) can occur as a severe systemic complication of infections with Shiga toxin (Stx)-producing Escherichia coli. Its pathology can be induced by Stx types, resulting in toxin-mediated damage to renal barriers, inflammation, and the development of acute kidney injury (AKI). Two sphingosine kinase (SphK) isozymes, SphK1 and SphK2, have been shown to be involved in barrier maintenance and renal inflammatory diseases. Therefore, we sought to determine their role in the pathogenesis of HUS. Experimental HUS was induced by the repeated administration of Stx2 in wild-type (WT) and SphK1 (SphK1−/−) or SphK2 (SphK2−/−) null mutant mice. Disease severity was evaluated by assessing clinical symptoms, renal injury and dysfunction, inflammatory status and sphingolipid levels on day 5 of HUS development. Renal inflammation and injury were found to be attenuated in the SphK2−/− mice, but exacerbated in the SphK1−/− mice compared to the WT mice. The divergent outcome appeared to be associated with oppositely altered sphingolipid levels. This study represents the first description of the distinct roles of SphK1−/− and SphK2−/− in the pathogenesis of HUS. The identification of sphingolipid metabolism as a potential target for HUS therapy represents a significant advance in the field of HUS research. Full article
(This article belongs to the Special Issue Sphingolipid Metabolism and Signaling: Role in Health and Diseases)
Show Figures

Figure 1

Figure 1
<p>Weight loss and HUS score of WT, SphK1<sup>−/−</sup>, and SphK2<sup>−/−</sup> mice with experimental hemolytic-uremic syndrome. Mice received vehicle (sham) or were challenged with Stx2 to induce experimental HUS and were followed up for 5 days (WT sham: <span class="html-italic">n</span> = 24, WT Stx: <span class="html-italic">n</span> = 24, SphK1<sup>−/−</sup> sham: <span class="html-italic">n</span> = 14, SphK1<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 14, SphK2<sup>−/−</sup> sham: <span class="html-italic">n</span> = 12, SphK2<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 12). (<b>A</b>) Progression of weight loss from day 1 to 5. (<b>B</b>) Overall weight loss on day 5. (<b>C</b>) HUS score from day 1 to 5. HUS score ranges from 1 = no signs of illness to 5 = dead. (<b>D</b>) HUS score on day 5. Data are expressed as (<b>A</b>,<b>C</b>) interleaved scatter plot, (<b>B</b>) scatter dot plot with median (interquartile range). and (<b>D</b>) bar plot with median (interquartile range) for <span class="html-italic">n</span> observations. Kruskal–Wallis test + Dunn’s multiple comparison test: * <span class="html-italic">p</span> &lt; 0.05: Stx group vs. the corresponding sham group, § <span class="html-italic">p</span> &lt; 0.05: Stx groups comparison. HUS, hemolytic-uremic syndrome; SphK, sphingosine kinase; Stx, Shiga toxin; WT, wild type.</p>
Full article ">Figure 2
<p>Hemogram analysis in WT, SphK1<sup>−/−</sup>, and SphK2<sup>−/−</sup> mice with experimental hemolytic-uremic syndrome. Mice received vehicle (sham) or were challenged with Stx2 to induce experimental HUS. Whole-blood parameters were assessed by determining (<b>A</b>) hematocrit, (<b>B</b>) erythrocytes, (<b>C</b>) hemoglobin, (<b>D</b>) thrombocyte count, (<b>E</b>) leukocyte count, (<b>F</b>) mean corpuscular volume, (<b>G</b>) mean corpuscular hemoglobin, and (<b>H</b>) mean corpuscular hemoglobin concentration in whole blood drawn on day 5 (WT sham: <span class="html-italic">n</span> = 24, WT Stx: <span class="html-italic">n</span> = 24, SphK1<sup>−/−</sup> sham: <span class="html-italic">n</span> = 14, SphK1<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 14, SphK2<sup>−/−</sup> sham: <span class="html-italic">n</span> = 12, SphK2<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 12). Data are expressed as (<b>A</b>–<b>H</b>) scatter dot plot with median (interquartile range) for <span class="html-italic">n</span> observations. Kruskal–Wallis test + Dunn’s multiple comparison test: * <span class="html-italic">p</span> &lt; 0.05: Stx group vs. the corresponding sham group, # <span class="html-italic">p</span> &lt; 0.05: sham groups comparison, § <span class="html-italic">p</span> &lt; 0.05: Stx groups comparison. SphK, sphingosine kinase; Stx, Shiga toxin; WT, wild type.</p>
Full article ">Figure 3
<p>Kidney injury and dysfunction in WT, SphK1<sup>−/−</sup>, and SphK2<sup>−/−</sup> mice with experimental hemolytic-uremic syndrome. Mice received vehicle (sham) or were challenged with Stx2 to induce experimental HUS. Kidney injury and dysfunction were assessed by determining plasma (<b>A</b>) NGAL (WT sham: <span class="html-italic">n</span> = 24, WT Stx: <span class="html-italic">n</span> = 24, SphK1<sup>−/−</sup> sham: <span class="html-italic">n</span> = 14, SphK1<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 14, SphK2<sup>−/−</sup> sham: <span class="html-italic">n</span> = 12, SphK2<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 12) and (<b>B</b>) urea (WT sham: <span class="html-italic">n</span> = 22, WT Stx: <span class="html-italic">n</span> = 23, SphK1<sup>−/−</sup> sham: <span class="html-italic">n</span> = 13, SphK1<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 14, and SphK2<sup>−/−</sup>: <span class="html-italic">n</span> = 12) levels, respectively, on day 5. Kidney injury was further investigated by scoring of (<b>C</b>) PAS staining and immunohistological (<b>D</b>) KIM-1 and (<b>E</b>) CD31 staining of renal sections (<span class="html-italic">n</span> = 8 per group) on day 5. Bar = 50 µm (400× magnification). Data are expressed as (<b>A</b>–<b>E</b>) scatter dot plot with median (interquartile range) for <span class="html-italic">n</span> observations. Images (<b>C</b>–<b>E</b>) are representative for renal sections of 8 mice per group. Kruskal–Wallis test + Dunn’s multiple comparison test: * <span class="html-italic">p</span> &lt; 0.05: Stx group vs. the corresponding sham group. CD31, cluster of differentiation 31; KIM-1, kidney injury molecule-1; NGAL, neutrophil gelatinase-associated lipocalin; PAS, periodic acid Schiff; SphK, sphingosine kinase; Stx, Shiga toxin; WT, wild type.</p>
Full article ">Figure 4
<p>Macrophage invasion and cytokine release in WT, SphK1<sup>−/−</sup>, and SphK2<sup>−/−</sup> mice with experimental hemolytic-uremic syndrome. Mice received vehicle (sham) or were challenged with Stx2 to induce experimental HUS. Macrophage invasion was assessed by determining the relative levels of (<b>A</b>) F4-80 in renal sections on day 5. Images are representative for renal sections of 8 mice per group. Bar = 50 µm (400× magnification). Cytokine levels of (<b>B</b>) RANTES, (<b>C</b>) MCP-5, (<b>D</b>) MDC, (<b>E</b>) IL-6, (<b>F</b>) KC, (<b>G</b>) GM-CSF, (<b>H</b>) IP-10, (<b>I</b>) MIP-3β, and (<b>J</b>) MIP-1α were measured on day 5 (WT sham: <span class="html-italic">n</span> = 4, WT Stx: <span class="html-italic">n</span> = 5, SphK1<sup>−/−</sup> sham: <span class="html-italic">n</span> = 5, SphK1<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 6, SphK2<sup>−/−</sup> sham: <span class="html-italic">n</span> = 5, SphK2<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 6). Data are expressed as (<b>A</b>–<b>J</b>) scatter dot plot with median (interquartile range) for <span class="html-italic">n</span> observations. Kruskal–Wallis test + Dunn’s multiple comparison test: * <span class="html-italic">p</span> &lt; 0.05: Stx group vs. the corresponding sham group, # <span class="html-italic">p</span> &lt; 0.05: sham groups comparison, § <span class="html-italic">p</span> &lt; 0.05: Stx groups comparison. GM-CSF, granulocyte-macrophage colony-stimulating factor; IL-6, interleukin-6; KC, keratinocyte-derived chemokine; MCP-5, monocyte chemoattractant protein-5; MDC, macrophage-derived chemokine; RANTES, Regulated And Normal T cell Expressed and Secreted; IP-10, interferon gamma-induced protein 10; MIP-3β, macrophage inflammatory protein-3-beta; MIP1α, macrophage inflammatory protein-3-alpha; SphK, sphingosine kinase; Stx, Shiga toxin; WT, wild type.</p>
Full article ">Figure 5
<p>Sphingolipid levels in plasma from WT, SphK1<sup>−/−</sup>, and SphK2<sup>−/−</sup> mice with experimental hemolytic-uremic syndrome. Mice received vehicle (sham) or were challenged with Stx2 to induce experimental HUS. To assess changes in plasma sphingolipid levels from mice with experimental HUS, levels of (<b>A</b>) S1P, (<b>B</b>) Sph and the ceramide (Cer) species (<b>C</b>) C16:0 Cer, (<b>D</b>) C22:0 Cer, (<b>E</b>) C24:0 Cer, and (<b>F</b>) C24:1 Cer were determined by LC-MS/MS. Data are expressed as scatter dot plots with median (interquartile range) for <span class="html-italic">n</span> observations. (<b>A</b>,<b>D</b>–<b>F</b>): WT: <span class="html-italic">n</span> = 24, SphK1<sup>−/−</sup>: <span class="html-italic">n</span> = 14, SphK2<sup>−/−</sup>: <span class="html-italic">n</span> = 11, (<b>B</b>): WT sham: <span class="html-italic">n</span> = 24, WT Stx: <span class="html-italic">n</span> = 23, SphK1<sup>−/−</sup> sham: <span class="html-italic">n</span> = 13, SphK1<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 13, SphK2<sup>−/−</sup> sham: <span class="html-italic">n</span> = 10, SphK2<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 10, (<b>C</b>): WT sham: <span class="html-italic">n</span> = 14, WT Stx: <span class="html-italic">n</span> = 11, SphK1<sup>−/−</sup> sham: <span class="html-italic">n</span> = 13, SphK1<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 12, SphK2<sup>−/−</sup> sham: <span class="html-italic">n</span> = 10, SphK2<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 10. Kruskal–Wallis test + Dunn’s multiple comparison test: # <span class="html-italic">p</span> &lt; 0.05: sham groups comparison, § <span class="html-italic">p</span> &lt; 0.05: Stx groups comparison. Cer, ceramide; HUS, hemolytic-uremic syndrome; S1P, sphingosine-1-phosphate; Sph, sphingosine; SphK, sphingosine kinase; Stx, Shiga toxin; WT, wild type.</p>
Full article ">Figure 6
<p>Sphingolipid levels in renal tissue from WT, SphK1<sup>−/−</sup> and SphK2<sup>−/−</sup> mice with experimental hemolytic-uremic syndrome. Mice received vehicle (sham) or were challenged with Stx2 to induce experimental HUS. To assess changes in sphingolipid levels in renal tissue from mice with experimental HUS, levels of (<b>A</b>) S1P, (<b>B</b>) Sph and ceramide species (<b>C</b>) C16:0 Cer, (<b>D</b>) C18:0 Cer, (<b>E</b>) C20:0 Cer, (<b>F</b>) C22:0 Cer, (<b>G</b>) C24:0 Cer, and (<b>H</b>) C24:1 Cer were determined by LC-MS/MS. Data are expressed as scatter dot plots with median (interquartile range) for <span class="html-italic">n</span> observations. (<b>A</b>–<b>H</b>): WT sham: <span class="html-italic">n</span> = 24, WT Stx: <span class="html-italic">n</span> = 24, SphK1<sup>−/−</sup> sham: <span class="html-italic">n</span> = 13–14, SphK1<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 14, SphK2<sup>−/−</sup> sham: <span class="html-italic">n</span> = 12, SphK2<sup>−/−</sup> Stx: <span class="html-italic">n</span> = 12. Kruskal–Wallis test + Dunn’s multiple comparison test: * <span class="html-italic">p</span> &lt; 0.05: Stx group vs. the corresponding sham group, # <span class="html-italic">p</span> &lt; 0.05: sham groups comparison, § <span class="html-italic">p</span> &lt; 0.05: Stx groups comparison. Cer, ceramide; HUS, hemolytic-uremic syndrome; S1P, sphingosine-1-phosphate; Sph, sphingosine; Stx, Shiga toxin; SphK, sphingosine kinase; WT, wild type.</p>
Full article ">
19 pages, 5547 KiB  
Article
Small and Large Extracellular Vesicles of Porcine Seminal Plasma Differ in Lipid Profile
by Pablo Martínez-Díaz, Ana Parra, Christian M. Sanchez-López, Josefina Casas, Xiomara Lucas, Antonio Marcilla, Jordi Roca and Isabel Barranco
Int. J. Mol. Sci. 2024, 25(13), 7492; https://doi.org/10.3390/ijms25137492 - 8 Jul 2024
Viewed by 558
Abstract
Seminal plasma contains a heterogeneous population of extracellular vesicles (sEVs) that remains poorly characterized. This study aimed to characterize the lipidomic profile of two subsets of differently sized sEVs, small (S-) and large (L-), isolated from porcine seminal plasma by size-exclusion chromatography and [...] Read more.
Seminal plasma contains a heterogeneous population of extracellular vesicles (sEVs) that remains poorly characterized. This study aimed to characterize the lipidomic profile of two subsets of differently sized sEVs, small (S-) and large (L-), isolated from porcine seminal plasma by size-exclusion chromatography and characterized by an orthogonal approach. High-performance liquid chromatography–high-resolution mass spectrometry was used for lipidomic analysis. A total of 157 lipid species from 14 lipid classes of 4 major categories (sphingolipids, glycerophospholipids, glycerolipids, and sterols) were identified. Qualitative differences were limited to two cholesteryl ester species present only in S-sEVs. L-sEVs had higher levels of all quantified lipid classes due to their larger membrane surface area. The distribution pattern was different, especially for sphingomyelins (more in S-sEVs) and ceramides (more in L-sEVs). In conclusion, this study reveals differences in the lipidomic profile of two subsets of porcine sEVs, suggesting that they differ in biogenesis and functionality. Full article
(This article belongs to the Special Issue New Progress in Extracellular Vesicles)
Show Figures

Figure 1

Figure 1
<p>Phenotypic characterization of large (L) and small (S) porcine seminal extracellular vesicles (sEVs). Box plots showing (<b>A</b>) total protein concentration and (<b>B</b>) particle concentration measured by nanoparticle tracking analysis. (<b>C</b>) Particle size distribution measured by dynamic light scattering analysis (blue line indicates S-sEVs and red line indicates L-sEVs). (<b>D</b>) Representative transmission electron microscopy images showing the morphology of sEVs. **** <span class="html-italic">p</span> &lt; 0.0001. Box plots: Boxes enclose the 25th and 75th percentiles, whiskers extend to the 5th and 95th percentiles, and line represents median.</p>
Full article ">Figure 2
<p>Representative flow cytometry plots (violet side scatter [violet-SSC]/direct side scatter [FSC]) showing CFSE, CD63, HSP90β and albumin positive events in samples of small and large seminal extracellular vesicles (sEVs) isolated from porcine seminal plasma.</p>
Full article ">Figure 3
<p>Lipid species identified and quantified in porcine seminal extracellular vesicles distributed by lipid categories (each with a different box color) and lipid classes within each lipid category. The data show the number of lipid species that were identified and quantified. Cer: ceramides; DHCer: dihydroceramides; SM: sphingomyelin; DHSM: dihydrosphingomyelin; HexCer: hexosylceramides; CDH: ceramide dihexoside; PC: phosphatidylcholines; LPC: lyso-phosphatidylcholines; PE: phosphatidylethanolamines; PE O-: ether-linked phosphatidylethanolamines; DG: diacylglycerols; TG: triacylglycerols; FC: free cholesterol; CE: cholesteryl esters.</p>
Full article ">Figure 4
<p>Histograms showing the differences between large (L-) and small (S-) seminal extracellular vesicles (sEVs) in the relative abundance of sphingolipids (<b>A</b>), glycerophospholipids (<b>B</b>), glycerolipids (<b>C</b>), and sterol lipids (<b>D</b>). Data are expressed as pmol eq/mg protein and are the mean ± SD. **** <span class="html-italic">p</span> value &lt; 0.0001, ** <span class="html-italic">p</span> value &lt; 0.01.</p>
Full article ">Figure 5
<p>Histograms showing the differences between large (L-) and small (S-) porcine seminal extracellular vesicles (sEVs) in the relative abundance of identified and quantified lipid classes distributed by lipid categories: sphingolipids (<b>A</b>), glycerophospholipids (<b>B</b>), glycerolipids (<b>C</b>), and sterol lipids (<b>D</b>). Data are expressed as pmol eq/mg protein and are the mean ± SD. **** <span class="html-italic">p</span> value &lt; 0.0001, *** <span class="html-italic">p</span> value &lt; 0.001, ** <span class="html-italic">p</span> value &lt; 0.01, * <span class="html-italic">p</span> value &lt; 0.05.</p>
Full article ">Figure 6
<p>Pie charts and supplementary tables showing the distribution of different categories (<b>A</b>) and classes of lipids (<b>B</b>–<b>E</b>) between small (S-) and large (L-) porcine seminal extracellular vesicles (sEVs). Data in tables show the mean percentage (%) ± SD. **** <span class="html-italic">p</span> value &lt; 0.0001, *** <span class="html-italic">p</span> value &lt; 0.001, ** <span class="html-italic">p</span> value &lt; 0.01, * <span class="html-italic">p</span> value &lt; 0.05, ns <span class="html-italic">p</span> value &gt; 0.05. a, b, c, d, e indicates differences at <span class="html-italic">p</span> &lt; 0.05 between lipid classes within a lipid category. SP: sphingolipids; GP: glycerophospholipids; GL: glycerolipids; ST: sterol lipids; Cer: ceramides; DHCer: dihydroceramides; SM: sphingomyelin; DHSM: dihydrosphingomyelin; HexCer: hexosylceramide; CDH: ceramide dihexoside; PC: phosphatidylcholine; LPC: lyso-phosphatidylcholine; PE: phosphatidylethanolamine; PE O-: ether-linked phosphatidylethanolamine; DG: diacylglycerols; TG: triacylglycerols; FC: free cholesterol; CE: cholesteryl esters.</p>
Full article ">
Back to TopTop