[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

Journals

Article Types

Countries / Regions

Search Results (8)

Search Parameters:
Keywords = UGCG

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 1600 KiB  
Article
Peripheral Upregulation of Parkinson’s Disease-Associated Genes Encoding α-Synuclein, β-Glucocerebrosidase, and Ceramide Glucosyltransferase in Major Depression
by Razvan-Marius Brazdis, Claudia von Zimmermann, Bernd Lenz, Johannes Kornhuber and Christiane Mühle
Int. J. Mol. Sci. 2024, 25(6), 3219; https://doi.org/10.3390/ijms25063219 - 12 Mar 2024
Viewed by 1158
Abstract
Due to the high comorbidity of Parkinson’s disease (PD) with major depressive disorder (MDD) and the involvement of sphingolipids in both conditions, we investigated the peripheral expression levels of three primarily PD-associated genes: α-synuclein (SNCA), lysosomal enzyme β-glucocerebrosidase (GBA1), [...] Read more.
Due to the high comorbidity of Parkinson’s disease (PD) with major depressive disorder (MDD) and the involvement of sphingolipids in both conditions, we investigated the peripheral expression levels of three primarily PD-associated genes: α-synuclein (SNCA), lysosomal enzyme β-glucocerebrosidase (GBA1), and UDP-glucose ceramide glucosyltransferase (UGCG) in a sex-balanced MDD cohort. Normalized gene expression was determined by quantitative PCR in patients suffering from MDD (unmedicated n = 63, medicated n = 66) and controls (remitted MDD n = 39, healthy subjects n = 61). We observed that expression levels of SNCA (p = 0.036), GBA1 (p = 0.014), and UGCG (p = 0.0002) were higher in currently depressed patients compared to controls and remitted patients, and expression of GBA1 and UGCG decreased in medicated patients during three weeks of therapy. Additionally, in subgroups, expression was positively correlated with the severity of depression and anxiety. Furthermore, we identified correlations between the gene expression levels and PD-related laboratory parameters. Our findings suggest that SNCA, GBA1, and UGCG analysis could be instrumental in the search for biomarkers of MDD and in understanding the overlapping pathological mechanisms underlying neuro-psychiatric diseases. Full article
(This article belongs to the Special Issue Molecular Research on Depression)
Show Figures

Figure 1

Figure 1
<p>Peripheral gene expressions of <span class="html-italic">SNCA</span> (<b>a</b>), <span class="html-italic">GBA</span> (<b>b</b>), and <span class="html-italic">UGCG</span> (<b>c</b>) were significantly higher in patients with current MDE (combined unmedicated patients (PU) and medicated patients (PM)) at inclusion compared to unaffected individuals (combined remitted patients (PR) and healthy subjects (HC)). These levels remained for <span class="html-italic">SNCA</span> (<b>d</b>) but decreased between inclusion (T1) and follow-up (T2) after on average three weeks of treatment as usual for <span class="html-italic">GBA1</span> (<b>e</b>) and <span class="html-italic">UGCG</span> (<b>f</b>) in the group of initially medicated patients. Normalized gene expression relative to reference genes is shown on a logarithmic <span class="html-italic">y</span>-axis. The numbers of individuals are provided below the <span class="html-italic">x</span>-axis. <span class="html-italic">p</span>-values from Mann–Whitney U test (<b>a</b>–<b>c</b>) and Wilcoxon test for paired values (<b>d</b>–<b>f</b>). Box plots with median and interquartile range.</p>
Full article ">Figure 2
<p>Positive correlations between depression severity assessed by HAM-D (<b>a</b>–<b>c</b>), MADRS (<b>d</b>–<b>f</b>), and STAI trait (<b>g</b>–<b>i</b>) with peripheral gene expressions of <span class="html-italic">SNCA</span>, <span class="html-italic">GBA</span>, and <span class="html-italic">UGCG</span> in patients with remitted major depressive disorder (PR) separated in female (red dots) and male (blue dots) subgroups at inclusion. Linear regression line for the combined group with 95% confidence interval and statistics (Spearman correlation, in bold for <span class="html-italic">p</span> &lt; 0.05). Sex-stratified statistical data are in <a href="#ijms-25-03219-t003" class="html-table">Table 3</a>.</p>
Full article ">
14 pages, 1977 KiB  
Article
Interaction of Fabry Disease and Diabetes Mellitus: Suboptimal Recruitment of Kidney Protective Factors
by Maria D. Sanchez-Niño, Maria I. Ceballos, Sol Carriazo, Aranzazu Pintor-Chocano, Ana B. Sanz, Moin A. Saleem and Alberto Ortiz
Int. J. Mol. Sci. 2023, 24(21), 15853; https://doi.org/10.3390/ijms242115853 - 1 Nov 2023
Viewed by 1661
Abstract
Fabry disease is a lysosomal disease characterized by globotriaosylceramide (Gb3) accumulation. It may coexist with diabetes mellitus and both cause potentially lethal kidney end-organ damage. However, there is little information on their interaction with kidney disease. We have addressed the interaction between Fabry [...] Read more.
Fabry disease is a lysosomal disease characterized by globotriaosylceramide (Gb3) accumulation. It may coexist with diabetes mellitus and both cause potentially lethal kidney end-organ damage. However, there is little information on their interaction with kidney disease. We have addressed the interaction between Fabry disease and diabetes in data mining of human kidney transcriptomics databases and in Fabry (Gla-/-) and wild type mice with or without streptozotocin-induced diabetes. Data mining was consistent with differential expression of genes encoding enzymes from the Gb3 metabolic pathway in human diabetic kidney disease, including upregulation of UGCG, the gene encoding the upstream and rate-limiting enzyme glucosyl ceramide synthase. Diabetic Fabry mice displayed the most severe kidney infiltration by F4/80+ macrophages, and a lower kidney expression of kidney protective genes (Pgc1α and Tfeb) than diabetic wild type mice, without a further increase in kidney fibrosis. Moreover, only diabetic Fabry mice developed kidney insufficiency and these mice with kidney insufficiency had a high expression of Ugcg. In conclusion, we found evidence of interaction between diabetes and Fabry disease that may increase the severity of the kidney phenotype through modulation of the Gb3 synthesis pathway and downregulation of kidney protective genes. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

Figure 1
<p>Impact of diabetes on genes encoding enzymes in the Gb3 pathway and on kidney function in wild type and Fabry mice. (<b>a</b>) Scheme of Gb3 synthesis and metabolism: glucosylceramide synthase encoded by <span class="html-italic">Ugcg</span>, Gb3 synthase encoded by <span class="html-italic">A4galt</span>, acid ceramidase encoded by <span class="html-italic">Asah1</span>, and alpha-galactosidase A encoded by <span class="html-italic">Gla</span>. Arrows present a summary of data shown in panels (<b>b</b>–<b>d</b>). (<b>b</b>–<b>d</b>) Kidney mRNA was measured by real time RT-PCR in diabetic wild-type or Fabry mice or vehicle controls. (<b>b</b>) <span class="html-italic">Ugcg</span>, (<b>c</b>) <span class="html-italic">A4galt</span>, (<b>d</b>) <span class="html-italic">Asah1</span>. (<b>e</b>) Plasma creatinine and (<b>f</b>) plasma urea. Data expressed as mean ± SEM of 5–15 animals per group. * <span class="html-italic">p</span> &lt; 0.05 vs. respective vehicle group. Significance (<span class="html-italic">p</span> &lt; 0.05) was assessed by Student’s <span class="html-italic">t-</span>test for two groups of data and ANOVA for three or more groups with Bonferroni post-hoc correction.</p>
Full article ">Figure 2
<p>Impact of diabetes on kidney inflammation in Fabry mice. (<b>a</b>) <span class="html-italic">Mcp1</span>, (<b>b</b>) <span class="html-italic">Rantes</span> and (<b>c</b>) <span class="html-italic">Fn14</span> mRNA expression in kidneys was measured by real time RT-PCR. (<b>d</b>) Correlation between serum urea and kidney <span class="html-italic">Fn14</span> mRNA expression as assessed by linear regression. (<b>e</b>) Immunohistochemistry assessment of F4/80+ macrophages: representative images and quantitation. * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle WT. ** <span class="html-italic">p</span> &lt; 0.05 vs. vehicle Fabry.Original magnification ×20. Data expressed as mean ± SEM of 5–15 animals per group. Significance (<span class="html-italic">p</span> &lt; 0.05) was assessed by Student’s <span class="html-italic">t-</span>test for two groups of data and ANOVA for three or more groups with Bonferroni post-hoc correction.</p>
Full article ">Figure 3
<p>Impact of diabetes on kidney fibrosis in Fabry mice. (<b>a</b>) Kidney <span class="html-italic">Col1a2</span> and (<b>b</b>) <span class="html-italic">Fn1</span> mRNA expression was measured by real time RT-PCR. (<b>c</b>) Sirius red staining for collagen deposition: representative images and quantitation. (<b>d</b>) Immunohistochemistry disclosed milder fibronectin deposition in diabetic Fabry mice than in diabetic WT mice. Original magnification ×20. * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle WT. ** <span class="html-italic">p</span> &lt; 0.01 vs. WT diabetic, *** <span class="html-italic">p</span> &lt; 0.0005 vs. WT diabetic. Data expressed as mean ± SEM of 5–15 animals per group. Significance (<span class="html-italic">p</span> &lt; 0.05) was assessed by Student’s <span class="html-italic">t-</span>test for two groups of data and ANOVA for three or more groups with Bonferroni post-hoc correction.</p>
Full article ">Figure 4
<p>Impact of diabetes on the expression of kidney nephroprotective genes in Fabry mice. (<b>a</b>) Kidney <span class="html-italic">Klotho</span> mRNA, (<b>b</b>) <span class="html-italic">Pgc1α</span> mRNA, (<b>c</b>) <span class="html-italic">Tfeb</span> mRNA. * <span class="html-italic">p</span> &lt; 0.05 vs. WT vehicle, ** <span class="html-italic">p</span> &lt; 0.05 vs. WT diabetic. Data expressed as mean ± SEM of 5–15 animals per group. Significance (<span class="html-italic">p</span> &lt; 0.05) was assessed by Student’s <span class="html-italic">t</span>-test for two groups of data and ANOVA for three or more groups with Bonferroni post-hoc correction.</p>
Full article ">
13 pages, 865 KiB  
Review
Update on Glycosphingolipids Abundance in Hepatocellular Carcinoma
by Frances L. Byrne, Ellen M. Olzomer, Nina Lolies, Kyle L. Hoehn and Marthe-Susanna Wegner
Int. J. Mol. Sci. 2022, 23(9), 4477; https://doi.org/10.3390/ijms23094477 - 19 Apr 2022
Cited by 5 | Viewed by 2638
Abstract
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer. Low numbers of HCC patients being suitable for liver resection or transplantation and multidrug resistance development during pharmacotherapy leads to high death rates for HCC patients. Understanding the molecular mechanisms of [...] Read more.
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer. Low numbers of HCC patients being suitable for liver resection or transplantation and multidrug resistance development during pharmacotherapy leads to high death rates for HCC patients. Understanding the molecular mechanisms of HCC etiology may contribute to the development of novel therapeutic strategies for prevention and treatment of HCC. UDP-glucose ceramide glycosyltransferase (UGCG), a key enzyme in glycosphingolipid metabolism, generates glucosylceramide (GlcCer), which is the precursor for all glycosphingolipids (GSLs). Since UGCG gene expression is altered in 0.8% of HCC tumors, GSLs may play a role in cellular processes in liver cancer cells. Here, we discuss the current literature about GSLs and their abundance in normal liver cells, Gaucher disease and HCC. Furthermore, we review the involvement of UGCG/GlcCer in multidrug resistance development, globosides as a potential prognostic marker for HCC, gangliosides as a potential liver cancer stem cell marker, and the role of sulfatides in tumor metastasis. Only a limited number of molecular mechanisms executed by GSLs in HCC are known, which we summarize here briefly. Overall, the role GSLs play in HCC progression and their ability to serve as biomarkers or prognostic indicators for HCC, requires further investigation. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

Figure 1
<p>GSLs and HCC related publications and ceramide concentrations in glycosphingolipid-enriched microdomains (GEMs) in NMuLi/UGCG OE and control cells. (<b>A</b>) Publications identified by the key word combinations <span class="html-italic">HCC glucosylceramide</span>, <span class="html-italic">HCC glycosphingolipids</span>, <span class="html-italic">Gaucher HCC</span>, <span class="html-italic">Gaucher liver cancer</span>, <span class="html-italic">liver cancer glycosphingolipids</span>, <span class="html-italic">hepatocellular carcinoma gangliosides</span>, <span class="html-italic">hepatocellular carcinoma globosides</span>, <span class="html-italic">hepatocellular carcinoma hexosylceramide</span>, and <span class="html-italic">hepatocellular carcinoma</span> lactosylceramide between 1967 and 2021 [<a href="#B17-ijms-23-04477" class="html-bibr">17</a>]. Notably, the majority of these studies were identified by the keywords <span class="html-italic">liver, cancer,</span> and <span class="html-italic">glycosphingolipids</span>. Blue bars represent years 2010 to 2021 (increase in published studies). (<b>B</b>) GEMs were isolated by sucrose density centrifugation and GEMs verified by cholesterol and GlcCer concentration determination. NMuLi/EV-2 = empty vector control; NMuLi/UGCG OE = UGCG overexpressing cells. The GEM verification for these samples is published in [<a href="#B18-ijms-23-04477" class="html-bibr">18</a>]. Data are represented as a mean of n = 3 ± SEM. Unpaired t test with Welch’s correction. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Schematic overview of ganglioside-mediated effects in HCC. (<b>A</b>) Ganglioside-mediated effects on cell migration. (<b>B</b>) Ganglioside-mediated effects on cancer stem cells and immune response.</p>
Full article ">
16 pages, 3066 KiB  
Article
Inhibition of Ceramide Glycosylation Enhances Cisplatin Sensitivity in Cholangiocarcinoma by Limiting the Activation of the ERK Signaling Pathway
by Piyasiri Chueakwon, Peeranat Jatooratthawichot, Krajang Talabnin, James R. Ketudat Cairns and Chutima Talabnin
Life 2022, 12(3), 351; https://doi.org/10.3390/life12030351 - 28 Feb 2022
Cited by 5 | Viewed by 2825
Abstract
Cholangiocarcinoma (CCA) is an aggressive tumor of the biliary epithelium with poor survival that shows limited response to conventional chemotherapy. Increased expression of glucosylceramide synthase (GCS) contributes to drug resistance and the progression of various cancers; the expression profiles of GCS (UGCG) and [...] Read more.
Cholangiocarcinoma (CCA) is an aggressive tumor of the biliary epithelium with poor survival that shows limited response to conventional chemotherapy. Increased expression of glucosylceramide synthase (GCS) contributes to drug resistance and the progression of various cancers; the expression profiles of GCS (UGCG) and the genes for glucocerebrosidases 1, 2, and 3 (GBA1, GBA2, and GBA3) were therefore studied in CCA. The biological functions of GCS for cell proliferation and cisplatin sensitivity in CCA were explored. GCS expression was higher in CCA tumor tissues than that of GBA1, GBA2, and GBA3. Verification of GCS expression in 29 paired frozen CCA tissues showed that 8 of 29 cases (27.6%) had high GCS expression. The expression of GCS and GBA2 was induced in CCA cell lines following low-dose cisplatin treatment. Suppression of GCS by either palmitoylamino-3-morpholino-1-propanol (PPMP), GCS knockdown or a combination of the two resulted in reduced cell proliferation. These treatments enhanced the effect of cisplatin-induced CCA cell death, increased the expression of apoptotic proteins and reduced phosphorylation of ERK upon cisplatin treatment. Taken together, inhibition of the GCS increased cisplatin-induced CCA apoptosis via the inhibition of the ERK signaling pathway. Thus, targeting GCS might be a strategy for CCA treatment. Full article
Show Figures

Figure 1

Figure 1
<p>Expression of ceramide-metabolizing enzymes in paired CCA tissues. (<b>A</b>) Diagram of the sphingolipids biosynthesis pathways. Expression data were obtained from the GEO database (GSE76297 dataset) for a comparison of: (<b>B</b>) <span class="html-italic">GCS</span> mRNA expression, (<b>C</b>) <span class="html-italic">GBA1</span> mRNA expression, (<b>D</b><span class="html-italic">)</span> <span class="html-italic">GBA2</span> mRNA expression and (<b>E</b>) <span class="html-italic">GBA3</span> mRNA expression. The Mann-Whitney was used to compare the expression between tumor (n = 92) and non-tumor tissues (n = 91) as well as paired tissues. ***, <span class="html-italic">p</span> &lt; 0.001 versus non-tumor tissues.</p>
Full article ">Figure 2
<p><span class="html-italic">GCS</span> expression in 29 paired frozen CCA tissues. (<b>A</b>) <span class="html-italic">GCS</span> mRNA expression was determined by qPCR. (<b>B</b>) Cut-off at 1.5-fold change (tumor/adjacent normal) was used for dichotomized <span class="html-italic">GCS</span> mRNA expression in 2 groups. Cut-off value ≤ 1.5-fold change and &gt; 1.5-fold change denoted as low and high <span class="html-italic">GCS</span> expression, respectively. (<b>C</b>) Kaplan-Meier analysis of overall survival in low and high <span class="html-italic">GCS</span> cases. Log Rank test was used for survival analysis. *, <span class="html-italic">p</span> &lt; 0.05 versus adjacent normal tissues.</p>
Full article ">Figure 3
<p>Changes of ceramide-metabolizing enzymes upon cisplatin treatment. (<b>A</b>) Respective basal mRNA expression of GCS, GBA1, and GBA2 were determined in two CCA cell lines (KKU-100 and KKU-213A). (<b>B</b>) KKU-100 and KKU-213A were treated with cisplatin at 0, 10, 20, 40, and 60 µM for 24 and 48 h. (<b>C</b>) Respective mRNA expression of GCS, GBA2, and GCS/GBA2 ratio was determined in KKU-100 and KKU-213A after 24 h of cisplatin treatment. Values are expressed as the mean ± SEM of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 versus cisplatin at 0 µM. ND: not detected.</p>
Full article ">Figure 4
<p>Suppression of GCS reduces CCA cell growth. (<b>A</b>) mRNA and protein levels of GCS after suppression by siRNAs for 24 and 48 h. (<b>B</b>) Cell proliferation after GCS suppression by siRNAs for 0–48 h, (<b>C</b>) PPMP (GCS inhibitor), and (<b>D</b>) co-treatment by siRNAs and PPMP. (<b>E</b>,<b>F</b>) The relative mRNA expression of growth-related genes in siGCS-treated KKU-213A or PPMP-treated KKU-213A were determined using qPCR. Values are expressed as the mean ± SEM of three independent experiments. * <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 versus siCTRL. The original unedited blot was presented in <a href="#app1-life-12-00351" class="html-app">Figure S1</a>.</p>
Full article ">Figure 5
<p>Suppression of GCS enhances cisplatin sensitivity. (<b>A</b>) Inducible mRNA expression of GCS following low-dose cisplatin (0, 10, 20, and 40 μM). Cell viability of KKU-213A was determined after (<b>B</b>) GCS inhibition by siRNAs for 24 h and treated with cisplatin for 48 h, (<b>C</b>) GCS inhibition by PPMP for 24 h and in combination with cisplatin for 48 h, and (<b>D</b>) co-treatment of GCS with siRNAs and PPMP for 24 h plus cisplatin for 48 h. Values are expressed as the mean ± SEM of three independent experiments. * <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 versus cisplatin at 0 µM.</p>
Full article ">Figure 6
<p>Suppression of GCS induces CCA cell apoptosis via the attenuation of ERK activation. KKU-213A was exposed by PPMP alone (10 µM), cisplatin alone (10 or 20 µM), or the combination of both for 24 h. Protein expression potentially involved (<b>A</b>) apoptosis and (<b>B</b>) survival signaling pathway observed by Western blot assay. Relative protein expression was measured by Image J software (version 1.53a). Values are expressed as the mean ± SEM of three independent experiments. * <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 versus cisplatin at 0 µM. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 versus cisplatin at 10 or 20 µM. The original unedited blot was presented in <a href="#app1-life-12-00351" class="html-app">Figure S2</a>.</p>
Full article ">
19 pages, 2832 KiB  
Article
The Sphingolipid Asset Is Altered in the Nigrostriatal System of Mice Models of Parkinson’s Disease
by Victor Blokhin, Maria Shupik, Ulyana Gutner, Ekaterina Pavlova, Albert T. Lebedev, Olga Maloshitskaya, Vsevolod Bogdanov, Sergey Sokolov, Alice Alessenko and Michael Ugrumov
Biomolecules 2022, 12(1), 93; https://doi.org/10.3390/biom12010093 - 6 Jan 2022
Cited by 4 | Viewed by 2371
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease incurable due to late diagnosis and treatment. Therefore, one of the priorities of neurology is to study the mechanisms of PD pathogenesis at the preclinical and early clinical stages. Given the important role of sphingolipids in [...] Read more.
Parkinson’s disease (PD) is a neurodegenerative disease incurable due to late diagnosis and treatment. Therefore, one of the priorities of neurology is to study the mechanisms of PD pathogenesis at the preclinical and early clinical stages. Given the important role of sphingolipids in the pathogenesis of neurodegenerative diseases, we aimed to analyze the gene expression of key sphingolipid metabolism enzymes (ASAH1, ASAH2, CERS1, CERS3, CERS5, GBA1, SMPD1, SMPD2, UGCG) and the content of 32 sphingolipids (subspecies of ceramides, sphingomyelins, monohexosylceramides and sphinganine, sphingosine, and sphingosine-1-phosphate) in the nigrostriatal system in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse models of the preclinical and clinical stages of PD. It has been shown that in PD models, the expression of five of the nine studied genes (CERS1, CERS5, ASAH1, ASAH2, and GBA1) increases but only in the substantia nigra (SN) containing dopaminergic cell bodies. Changes in the expression of enzyme genes were accompanied by an increase in the content of 7 of the 32 studied sphingolipids. Such findings suggest these genes as attractive candidates for diagnostic purposes for preclinical and clinical stages of PD. Full article
(This article belongs to the Section Biomacromolecules: Lipids)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Design of experiments for modeling Parkinson’s disease at the preclinical and clinical stages in C57BL/6 mice by subcutaneous administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), once at a single dose of 18 mg/kg and three times at a single dose of 10 mg/kg, with a 2-h interval between injections, respectively. In the controls, saline was administered.</p>
Full article ">Figure 2
<p>Motor behavior: distance traveled in the open-field test in mice 2 weeks after administration of MPTP once at a dose of 18 mg/kg (1 × 18), three times at a single dose of 10 mg/kg (3 × 10), and in the controls (NaCl). * <span class="html-italic">p</span> = 0.05, significant difference compared to the controls; “+”: average; “−”: median.</p>
Full article ">Figure 3
<p>Changes in the dopamine (DA) concentration in the striatum (<b>A</b>) and changes in the dopamine content in the substantia nigra (<b>B</b>) in mice 2 weeks after subcutaneous administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): once at a single dose of 18 mg/kg (1 × 18) or three times at a single dose of 10 mg/kg with a 2-h interval between injections (3 × 10), compared to the controls (saline), taken as 100%. * <span class="html-italic">p</span> &lt; 0.05, significant difference compared to the controls, taken as 100%; # <span class="html-italic">p</span> &lt; 0.05, differences between the groups 1 × 18 and 3 × 10.</p>
Full article ">Figure 4
<p>Total (cumulative) concentration of all measured sphingomyelins (14-0, 16-1, 16-0, 18-1, 18-0, 20-1, 20-0, 22-1, 22-0, 24-1, 24-0, and 26-1) in the striatum and substantia nigra (<b>A</b>) in mice 2 weeks after the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): once at a single dose of 18 mg/kg (1 × 18) and three times at a single dose of 10 mg/kg (3 × 10), as well as the concentration of individual sphingomyelins in the striatum (<b>B</b>) and in the substantia nigra (<b>C</b>) in the same experiments and in the controls (saline). * <span class="html-italic">p</span> &lt; 0.05, significant differences compared to the controls; # <span class="html-italic">p</span> &lt; 0.05, significant differences between the groups 1 × 18 and 3 × 10.</p>
Full article ">Figure 5
<p>Concentration of ceramide 16-0 in the substantia nigra of mice 2 weeks after the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, once at a single dose of 18 mg/kg or three times at a single dose of 10 mg/kg. * <span class="html-italic">p</span> &lt; 0.05, significant differences compared to the controls.</p>
Full article ">Figure 6
<p>Concentration of sphingosine (SS), sphinganine (SG), and sphingosine-1-phosphate (S-1-P) in the striatum (<b>A</b>) and in the substantia nigra (<b>B</b>) in mice 2 weeks after the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, once at a single dose of 18 mg/kg or three times at a single dose of 10 mg/kg. * <span class="html-italic">p</span> &lt; 0.05, significant differences compared to the controls.</p>
Full article ">Figure 7
<p>Total (cumulative) concentration of all measured hexosylceramides, 18-0, 20-0, 22-0, 24-0, and 24-1, in the striatum and in the substantia nigra (<b>A</b>) in mice 2 weeks after the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine once at a single dose of 18 mg/kg (1 × 18) or three times at a single dose of 10 mg/kg (3 × 10), as well as the concentration of individual hexosylceramides, 20-0, 24-0, and 24-1, in the striatum (<b>B</b>) and in the substantia nigra (<b>C</b>) in the same experiments and in the controls (saline). * <span class="html-italic">p</span> &lt; 0.05, significant differences compared to the controls.</p>
Full article ">Figure 8
<p>Relative level of gene expression in the substantia nigra (<b>A</b>) and in the striatum (<b>B</b>) in mice 2 weeks after the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): once at a single dose of 18 mg/kg (1 × 18) or three times at a single dose of 10 mg/kg (3 × 10), with a 2-h interval between injections, as well as after saline administration in the controls. ASAH1, acid ceramidase; ASAH2, neutral ceramidase; CERS1, CERS3, CERS5, ceramide synthases 1, 3, and 5, respectively; GBA1, glucocerebrosidase; SMPD1, sphingomyelin phosphodiesterase 1 (acid sphingomyelinase); SMPD2, sphingomyelin phosphodiesterase 2 (neutral sphingomyelinase); UGCG, ceramide glucosyltransferase. * <span class="html-italic">p</span> &lt; 0.05, significant differences between the experiment and the controls.</p>
Full article ">Figure 9
<p>Scheme for the synthesis of sphingolipids. Lipids and enzymes that were examined in this study are marked in red. UGCG, ceramide glucosyltransferase; CERS1, CERS3, CERS5, ceramide synthases 1, 3, and 5, respectively; ASAH1, acid ceramidase; ASAH2, neutral ceramidase; GBA1, glucocerebrosidase; SMPD1, sphingomyelin phosphodiesterase 1 (acid sphingomyelinase); SMPD2, sphingomyelin phosphodiesterase 2 (neutral sphingomyelinase); SPT, serine-palmitoyl transferase; 3-KSR, 3-ketosphinganine reductase; DES, dihydroceramide desaturase; GALC, galactosyceramidase; UTG8A, 1-beta-galactosyltransferase; SPHK1, sphingosine kinase 1; SPHK2, sphingosine kinase 1; SGPP, sphingosine-1-phosphate phosphatase; SMS1, sphingomyelin synthase 1; SMS2, sphingomyelin synthase 2.</p>
Full article ">
22 pages, 10787 KiB  
Article
Blockade of Glycosphingolipid Synthesis Inhibits Cell Cycle and Spheroid Growth of Colon Cancer Cells In Vitro and Experimental Colon Cancer Incidence In Vivo
by Richard Jennemann, Martina Volz, Felix Bestvater, Claudia Schmidt, Karsten Richter, Sylvia Kaden, Johannes Müthing, Hermann-Josef Gröne and Roger Sandhoff
Int. J. Mol. Sci. 2021, 22(19), 10539; https://doi.org/10.3390/ijms221910539 - 29 Sep 2021
Cited by 11 | Viewed by 3075
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed cancers in humans. At early stages CRC is treated by surgery and at advanced stages combined with chemotherapy. We examined here the potential effect of glucosylceramide synthase (GCS)-inhibition on CRC biology. GCS is [...] Read more.
Colorectal cancer (CRC) is one of the most frequently diagnosed cancers in humans. At early stages CRC is treated by surgery and at advanced stages combined with chemotherapy. We examined here the potential effect of glucosylceramide synthase (GCS)-inhibition on CRC biology. GCS is the rate-limiting enzyme in the glycosphingolipid (GSL)-biosynthesis pathway and overexpressed in many human tumors. We suppressed GSL-biosynthesis using the GCS inhibitor Genz-123346 (Genz), NB-DNJ (Miglustat) or by genetic targeting of the GCS-encoding gene UDP-glucose-ceramide-glucosyltransferase- (UGCG). GCS-inhibition or GSL-depletion led to a marked arrest of the cell cycle in Lovo cells. UGCG silencing strongly also inhibited tumor spheroid growth in Lovo cells and moderately in HCT116 cells. MS/MS analysis demonstrated markedly elevated levels of sphingomyelin (SM) and phosphatidylcholine (PC) that occurred in a Genz-concentration dependent manner. Ultrastructural analysis of Genz-treated cells indicated multi-lamellar lipid storage in vesicular compartments. In mice, Genz lowered the incidence of experimentally induced colorectal tumors and in particular the growth of colorectal adenomas. These results highlight the potential for GCS-based inhibition in the treatment of CRC. Full article
(This article belongs to the Special Issue Activity, Function and Druggability of Cancer-Related Enzymes)
Show Figures

Figure 1

Figure 1
<p>UGCG expression in human colorectal cancer (CRC) with relation to survival. (<b>A</b>) Sphingolipid synthesis pathway; the enzyme glucosylceramide synthase (GCS) encoded by the gene <span class="html-italic">UGCG</span> is the initial enzyme in the glycosphingolipid (GSL)-biosynthesis pathway. Targeting GCS using genetic approaches or with specific inhibitors such as Genz-123346 disrupts the synthesis of glucosylceramide (GlcCer) and its downstream GSL-products and may lead to accumulation of precursors. (GSLs are abbreviated according to the joint commission on biochemical nomenclature of glycolipids [<a href="#B23-ijms-22-10539" class="html-bibr">23</a>]). (<b>B</b>) The expression of <span class="html-italic">UGCG</span> mRNA was lower in human CRC as compared to normal colon. (<b>C</b>) However, patients with high <span class="html-italic">UGCG</span> mRNA expression in colorectal tumors had a reduced overall survival time than patients with lower <span class="html-italic">UGCG</span> expression.</p>
Full article ">Figure 2
<p>Treatment of Lovo and HCT116 cells with Genz lead to reduced GSL expression and cell cycle arrest. (<b>A</b>) Formula of the GCS inhibitor Genz-123346 (<span class="html-italic">N</span>-[(1<span class="html-italic">R</span>,2<span class="html-italic">R</span>)-1-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-hydroxy-3-pyrrolidin-1-ylpropan-2-yl]nonanamide). (<b>B</b>,<b>C</b>) As shown by thin layer chromatography (TLC), GSL-synthesis was almost completely disrupted by treatment of Lovo (<b>B</b>) and HCT116 cells (<b>C</b>) with 1 µM Genz in the culture medium after 4 and 6 days; x, no GSL-positive band; running solvent for neutral GSLs was CHCl<sub>3</sub>/CH<sub>3</sub>OH, H<sub>2</sub>O, 62.5:30:6 (<span class="html-italic">v</span>/<span class="html-italic">v</span>) and for acidic GSLs CHCl<sub>3</sub>/CH<sub>3</sub>OH, 0.2% CaCl<sub>2</sub>, 60:35:8 (<span class="html-italic">v</span>/<span class="html-italic">v</span>). (<b>D</b>) Anti-Gb<sub>3</sub>Cer immune overlay of the neutral GSL-fraction from Lovo and HCT116 cells. Chemical staining with orcinol reagent (left) and immune overlay with anti-Gb<sub>3</sub>Cer antibodies. HCT116 cells expressed globosides in addition to hexosylceramides and lactosylceramides, which were not detectable in Lovo cells. (<b>E</b>,<b>F</b>, quantification) Cell cycle arrest of the Lovo cells in S and G2/M phases occurred after treatment with 1 µM Genz (<b>G</b>,<b>H</b>, quantification). A significant influence on the cell cycle of HCT116 cells as shown by FACS analysis occurred at 5 µM Genz and higher concentrations. (<b>I</b>–<b>L</b>) One micrometer of Genz inhibited growth of tumor spheroids from Lovo (<b>I</b>,<b>J</b>, quantification) and HCT116 cells (<b>K</b>,<b>L</b>, quantification). Note: HCT116 microspheres broke apart with elevated Genz concentrations after longer incubation period (<b>K</b>) and could therefore not be included in the calculations (<b>L</b>, nd, not done); each data point on the graphs represents one biological replicate; scale bars, 100 µM; significances are *, <span class="html-italic">p</span> ≤ 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>Miglustat treatment leads to cell cycle arrest in Lovo cells and inhibits growth of Lovo and HCT116 tumor spheroids. (<b>A</b>) Formula of Miglustat. (<b>B</b>,<b>C</b>) n-butyl-deoxynojirimycin (NB-DNJ, Miglustat) inhibits GSL-synthesis of Lovo (<b>B</b>) and HCT116 cells (<b>C</b>) almost completely after four days; x, no GSL-positive band; running solvent for neutral- and acidic GSLs was CHCl<sub>3</sub>/CH<sub>3</sub>OH, 0.2% CaCl<sub>2</sub>, 60:35:8 (<span class="html-italic">v</span>/<span class="html-italic">v</span>). (<b>D</b>,<b>E</b>, quantification) Miglustat treatment led to a visible arrest of the cell cycle in Lovo cells but not in HCT116 cells (<b>F</b>,<b>G</b>, quantification). (<b>H</b>–<b>K</b>) Miglustat inhibited tumor spheroid growth of Lovo- (<b>H</b>,<b>I</b>, quantification) and HCT116 cells (<b>J</b>,<b>K</b>, quantification); each data point on the graphs represents one biological replicate; scale bars, 100 µM; significances are *, <span class="html-italic">p</span> ≤ 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
<p>TLC and cell cycle determination of <span class="html-italic">UGCG</span>-gRNA treated HCT116- and Lovo cells. (<b>A</b>,<b>D</b>) An almost complete depletion of neutral and acidic GSLs was achieved in HCT116- (<b>A</b>) and Lovo cells (<b>D</b>) by <span class="html-italic">UGCG</span>-guide-RNA (gRNA); x, no GSL-positive band; running solvent for neutral- and acidic GSLs was CHCl<sub>3</sub>/CH<sub>3</sub>OH, 0.2% CaCl<sub>2</sub>, 60:35:8 (<span class="html-italic">v</span>/<span class="html-italic">v</span>). (<b>B</b>,<b>C</b>, quantification) <span class="html-italic">UGCG</span>-gRNA treated HCT116 cells did not show an arrest of the cell cycle, similar as HCT116 cells treated with low doses of Genz (<a href="#ijms-22-10539-f002" class="html-fig">Figure 2</a>). (<b>E</b>–<b>G</b>, quantification) Genetic depletion using <span class="html-italic">UGCG</span>-gRNA-application (<b>E</b>) and treatment of Lovo cells with 5 µM Genz (<b>F</b>) resulted in an arrest of the cell cycle, whereas the response in Genz-treated cells was slightly more pronounced; significances are *, <span class="html-italic">p</span> ≤ 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>Lovo and HCT116 cells present an elevated number of multivesicular bodies (MVBs) which contain conspicuous structured vesicles upon Genz treatment. (<b>A</b>) The number of multivesicular bodies with vesicles displaying multilamellar structures was elevated in Genz-treated colon carcinoma cells. MVB-number increased markedly with 1 µM Genz in Lovo cells. Only low numbers of MVBs were seen in HCT116 cells upon treatment with 1 µM Genz. The numbers of MVBs rose markedly with higher Genz concentrations; scale bars, 1 µm. (<b>B</b>) Vesicles inside MVBs of Lovo cells were canonically spheroid, with potential to form onion-like superstructures and myelin-like compaction of stacked membranes (<b>B</b>, insert). (<b>C</b>) MVBs of HCT116 cells displayed angular overall shapes (here triangular), a combination of apparently stiff sheets with sharp cornered rims. The insert demonstrates a widening of the expectedly lipophilic middle-leaflet of the canonical bi-laminar unit-membrane (arrowhead), scale bars in (<b>B</b>,<b>D</b>), 500 nm; in insets 100 nm. (<b>D</b>) The number of MVBs increased with elevated Genz-concentrations; shown are mean numbers of MVBs per µm<sup>2</sup> cytosol, each from 10 cells. (<b>E</b>,<b>F</b>) The expression levels of the autophagy marker Lc3-II rose markedly in both Lovo- (<b>E</b>) and HCT116 cells (<b>F</b>) upon increased Genz concentrations; significances are *, <span class="html-italic">p</span> ≤ 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>Genz treatment of Lovo and HCT116 cells lead to a concentration-dependent increase of sphingomyelin (SM). Lipids were quantified by C18-reversed phase UPLC/MS<sup>2</sup> in MRM mode using internal standards for each lipid class. Major sphingolipids containing non-hydroxy fatty acids (N) and (C18)-sphingosine (S) were recorded; Cer, ceramide, HexCer: hexosylceramide, SM, sphingomyelin, and PC: phosphatidylcholine. (<b>A</b>,<b>B</b>) HexCer was reduced to a similar extent in Genz- or <span class="html-italic">UGCG</span>-gRNA-treated Lovo (<b>A</b>) or HCT116 cells (<b>B</b>). The SM content in Lovo (<b>A</b>) and HCT116 (<b>B</b>) cells was remarkably elevated upon Genz treatment and much higher as in cells treated with <span class="html-italic">UGCG</span>-gRNA in which the SM-increase correlated vice versa with the reduction of GSLs. The ceramide levels in Lovo and HCT116 cells upon Genz treatment were similar as in controls. (<b>C</b>,<b>D</b>) SM levels in Miglustat-treated Lovo (<b>C</b>) and HCT116 cells (<b>D</b>) were also elevated as compared to controls. Ceramides appeared unaltered upon Miglustat treatment; each data point on the graphs represents one biological replicate; significances are *, <span class="html-italic">p</span> ≤ 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>Genz treatment lowered the incidence of experimental CRC in vivo. (<b>A</b>) Scheme of the treatment course. CRC was induced in C57Bl6 mice with azoxymethane (AOM) at the age of 8 weeks, followed by a tumor promoting 5-day cycle of dextrane sulfate (DSS) applied by the drinking water starting one week after the AOM-treatment. Mice, <span class="html-italic">n</span> = 24, received chow diet until five weeks after tumor induction. Then one cohort of animals, <span class="html-italic">n</span> = 10, received 0.225% Genz in the food. All animals were sacrificed 15 weeks after tumor induction according to the termination criteria from the animal application. (<b>B</b>) Scheme of the human and murine intestinal tract. Colorectal tumors in humans develop preferentially in the areas of the sigmoid colon and the rectum but also in other areas of the large intestine. In this mouse model, we observed tumors predominantly restricted to the rectal area of the colon with a critical load at fifteen weeks after tumor induction. (<b>C</b>) Treatment of mice with Genz resulted in a markedly lower number of colorectal tumors in the distal part of the colon. (<b>D</b>) The size of tumors detected in the colons of Genz-treated mice was also notably smaller as compared to chow fed mice; each data point represents the mean size of all tumors detected in one mouse; significances are *, <span class="html-italic">p</span> ≤ 0.05; **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 8
<p>The number of colorectal adenomas were reduced in Genz-treated mice. (<b>A</b>) Ki67 staining indicated proliferative cells in colons of mice. Both Genz-treated mice and controls developed predominantly adenomas with its typical polypoid-like shape (<b>A</b>,<b>B</b>, middle images) and only low numbers of adenocarcinomas (<b>A</b>,<b>B,</b> lower images). However, Genz-feeding markedly reduced the number of adenomas as compared to chow-fed controls (<b>B</b>). (<b>C</b>) The number of Ki67 positive cells in the colorectal parts of the colons was lower in crypts of Genz-treated mice as compared to controls; each data point represents mean numbers of Ki67 positive cells of approximately 50 crypts per mouse. Counted were positive cells only from completely vertically cut villi apart from the tumor areas; significances are *, <span class="html-italic">p</span> ≤ 0.05; ***, <span class="html-italic">p</span> &lt; 0.001. (<b>D</b>, F4/80) Both Genz-fed mice and controls lacked macrophage infiltrations in the tumor but showed an enrichment in peritumoral areas. (<b>D</b>, CD3) Only a few cells stained positive with the T cell marker CD3; T, tumor area. Genz treatment did not lead to increased apoptosis (<b>D</b>, TUNEL); a few cells with smaller nuclei than epithelial cells stained positive in tumors of treated mice and controls; scale bars, 100 µM.</p>
Full article ">Figure 9
<p>Genz treatment alters the sphingolipid composition in the intestine. (<b>A</b>) TLC-analysis of neutral and acidic GSLs from the intestine of Genz-treated mice. The intestine of adult mice contained predominantly the neutral GSLs HexCer and GA1. Lipids were quantified by C18-reversed phase UPLC/MS<sup>2</sup> in MRM mode using internal standards for each lipid class except for GA1, which was normalized to internal HexCer standards. Sphingolipids containing either non-hydroxy fatty acids (N) or alpha-hydroxy fatty acids (<b>A</b>) in combination with (C18)-sphingosine (S) or with (C18)-phytosphingosine (P) were recorded; Cer: ceramide, HexCer, hexosylceramide, GA1: asialo-ganglioside 1 or gangliotetraosylceramide (Gg<sub>4</sub>Cer, see <a href="#ijms-22-10539-f001" class="html-fig">Figure 1</a>A), and SM: sphingomyelin. (<b>B</b>,<b>C</b>) MS<sup>2</sup>-analysis of HexCer (<b>B</b>) and GA1 (<b>C</b>). AS- as well as NS-species markedly decreased in the intestine of Genz-treated mice. AP-GSLs were less effectively downregulated. Genz-application did not lead to a reduction of NP-GSLs; for designation of ceramide anchors, see abbreviations. (<b>D</b>) NP-Cers increased in intestines of Genz-treated mice as compared to chow-fed controls. (<b>E</b>) The SM content was markedly elevated and PC remained essentially unaltered upon Genz treatment; significances are *, <span class="html-italic">p</span> ≤ 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
17 pages, 1155 KiB  
Review
Roles of Gangliosides in Hypothalamic Control of Energy Balance: New Insights
by Kei-ichiro Inamori and Jin-ichi Inokuchi
Int. J. Mol. Sci. 2020, 21(15), 5349; https://doi.org/10.3390/ijms21155349 - 28 Jul 2020
Cited by 10 | Viewed by 3134
Abstract
Gangliosides are essential components of cell membranes and are involved in a variety of physiological processes, including cell growth, differentiation, and receptor-mediated signal transduction. They regulate functions of proteins in membrane microdomains, notably receptor tyrosine kinases such as insulin receptor (InsR) and epidermal [...] Read more.
Gangliosides are essential components of cell membranes and are involved in a variety of physiological processes, including cell growth, differentiation, and receptor-mediated signal transduction. They regulate functions of proteins in membrane microdomains, notably receptor tyrosine kinases such as insulin receptor (InsR) and epidermal growth factor receptor (EGFR), through lateral association. Studies during the past two decades using knockout (KO) or pharmacologically inhibited cells, or KO mouse models for glucosylceramide synthase (GCS; Ugcg), GM3 synthase (GM3S; St3gal5), and GD3 synthase (GD3S; St8sia1) have revealed essential roles of gangliosides in hypothalamic control of energy balance. The a-series gangliosides GM1 and GD1a interact with leptin receptor (LepR) and promote LepR signaling through activation of the JAK2/STAT3 pathway. Studies of GM3S KO cells have shown that the extracellular signal-regulated kinase (ERK) pathway, downstream of the LepR signaling pathway, is also modulated by gangliosides. Recent studies have revealed crosstalk between the LepR signaling pathway and other receptor signaling pathways (e.g., InsR and EGFR pathways). Gangliosides thus have the ability to modulate the effects of leptin by regulating functions of such receptors, and by direct interaction with LepR to control signaling. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

Figure 1
<p>Biosynthetic pathway of ganglio-series gangliosides. GCS (<span class="html-italic">Ugcg</span>), a glucosyltransferase, catalyzes the first step in synthesis of ganglio-series gangliosides. Subsequently, LacCerS (<span class="html-italic">B4galt5/6</span>) adds a galactose residue onto GlcCer to form LacCer. GM3S (<span class="html-italic">St3gal5</span>) is a sialyltransferase required for initiation of synthesis of a- and b-series gangliosides. GD3S (<span class="html-italic">St8sia1</span>) is a sialyltransferase required for synthesis of b-series gangliosides. Four species (GM1, GD1a, GD1b, GT1b) comprise the majority of total brain gangliosides in mammals.</p>
Full article ">Figure 2
<p>Leptin receptor signaling pathway, and alterations in signaling that characterize various ganglioside-deficient KO mouse models. Up and down arrows indicate increased or decreased activation of STAT3 or ERK in the pathways for the models. GCS icKO: tamoxifen-inducible, neuron-specific, conditional GCS KO.</p>
Full article ">
3071 KiB  
Article
Role of Intracellular Lipid Logistics in the Preferential Usage of Very Long Chain-Ceramides in Glucosylceramide
by Toshiyuki Yamaji, Aya Horie, Yuriko Tachida, Chisato Sakuma, Yusuke Suzuki, Yasunori Kushi and Kentaro Hanada
Int. J. Mol. Sci. 2016, 17(10), 1761; https://doi.org/10.3390/ijms17101761 - 21 Oct 2016
Cited by 19 | Viewed by 5313
Abstract
Ceramide is a common precursor of sphingomyelin (SM) and glycosphingolipids (GSLs) in mammalian cells. Ceramide synthase 2 (CERS2), one of the six ceramide synthase isoforms, is responsible for the synthesis of very long chain fatty acid (C20–26 fatty acids) (VLC)-containing ceramides (VLC-Cer). It [...] Read more.
Ceramide is a common precursor of sphingomyelin (SM) and glycosphingolipids (GSLs) in mammalian cells. Ceramide synthase 2 (CERS2), one of the six ceramide synthase isoforms, is responsible for the synthesis of very long chain fatty acid (C20–26 fatty acids) (VLC)-containing ceramides (VLC-Cer). It is known that the proportion of VLC species in GSLs is higher than that in SM. To address the mechanism of the VLC-preference of GSLs, we used genome editing to establish three HeLa cell mutants that expressed different amounts of CERS2 and compared the acyl chain lengths of SM and GSLs by metabolic labeling experiments. VLC-sphingolipid expression was increased along with that of CERS2, and the proportion of VLC species in glucosylceramide (GlcCer) was higher than that in SM for all expression levels of CERS2. This higher proportion was still maintained even when the proportion of C16-Cer to the total ceramides was increased by disrupting the ceramide transport protein (CERT)-dependent C16-Cer delivery pathway for SM synthesis. On the other hand, merging the Golgi apparatus and the endoplasmic reticulum (ER) by Brefeldin A decreased the proportion of VLC species in GlcCer probably due to higher accessibility of UDP-glucose ceramide glucosyltransferase (UGCG) to C16-rich ceramides. These results suggest the existence of a yet-to-be-identified mechanism rendering VLC-Cer more accessible than C16-Cer to UGCG, which is independent of CERT. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Isolation of <span class="html-italic">CERS2</span>-deficient clones. (<b>A</b>) The target site of the TALEN-CERS2 pair in the human <span class="html-italic">CERS2</span> gene and indel analysis in TALEN-CERS2–treated (TAL-CERS2) clones. The target sequences (CERS2 Fw and Rv) are located in intron 1 and exon 2, respectively. The sequence between the TALEN target sequences contains the start codon (yellow box) and the splicing acceptor (SA) at the end of intron 1 (blue lined box). Green boxes highlight the portions corresponding to the start codon and the SA. Deletions are shown in red and their lengths are specified on the right of the sequences; (<b>B</b>) Western blot analysis of CERS2 in the TAL-CERS2 clones. Cell lysates from the parent cells and CERS2-deficient mutants were immunoblotted with anti-CERS2 antibody (<b>top</b>) and anti-α-tubulin antibody (<b>middle</b>). Part of the enlarged view of the top image is shown at the <b>bottom</b>. The arrow and arrowhead indicate CERS2; P, parent cell line; 16, TAL-CERS2#16 clone; 18, TAL-CERS2#18 clone; 13, TAL-CERS2#13 clone; (-), no transfection; M, mock; and C2, CERS2 cDNA restoration. Note that overexpression of CERS2 resulted in formation of high molecular weight aggregates.</p>
Full article ">Figure 2
<p>Metabolic labeling of lipids in TAL-CERS2 clones. (<b>A</b>) Metabolic labeling of sphingolipids with radioactive serine in the parent cells and TAL-CERS2 clones shown in <a href="#ijms-17-01761-f001" class="html-fig">Figure 1</a>B. Cells were cultured with [<sup>14</sup>C]serine for 16 h, and lipids extracted from the cells were separated by normal-phase thin-layer chromatography (TLC) (Method 1). Radioactive image of an analyzed TLC plate is shown. Cer, ceramide; GalCer, galactosylceramide; PE, phosphatidylethanolamine; PS, phosphatidylserine; PC, phosphatidylcholine. P, parent cell line; 16, TAL-CERS2#16 clone; 18, TAL-CERS2#18 clone; 13, TAL-CERS2#13 clone; (-), no transfection; M, mock; C2, CERS2 cDNA restoration. The darker image is shown in <a href="#app1-ijms-17-01761" class="html-app">Figure S3A</a>; (<b>B</b>) comparison of the proportion of the upper band intensities (indicating the proportion of very long chain (VLC)) of GlcCer and sphingomyelin (SM). The enlarged and darker images of [<sup>14</sup>C]serine-labeled GlcCer and SM shown in (<b>A</b>) are aligned. The proportions of VLC-containing lipids are expressed as the upper (VLC-containing) band intensity as a percentage of the sum of the upper (VLC) and lower (C16 long chain fatty acid (FA) (LC)-containing) band intensities of GlcCer and SM for each cell line shown in <a href="#ijms-17-01761-f001" class="html-fig">Figure 1</a>B: mean percentage ± standard deviation (SD) obtained from five independent experiments. The Student’s <span class="html-italic">t</span>-test was used. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Different fatty acid preferences of GlcCer and SM. (<b>A</b>,<b>B</b>) Metabolic labeling of sphingolipids with radioactive serine for a short time. Cells were cultured with [<sup>14</sup>C]serine for 2 h, and lipids extracted from the cells were separated by TLC. (<b>A</b>) Separation between upper (VLC-containing) and lower (LC-containing) bands of ceramide by normal-phase TLC (Method 3 described in the “Experimental Section”). Non-radiolabeled ceramide standards (C16:0, C24:1, and C24:0) were developed simultaneously and visualized by iodine vapor. The positions of the spot origin (ori) and the solvent front (top) in two images were aligned; (<b>B</b>) separation of upper and lower bands of GlcCer and SM by normal-phase TLC according to Method 1. Radioactive images of analyzed TLC plates are shown. The darker image is shown in <a href="#app1-ijms-17-01761" class="html-app">Figure S6B</a>; (<b>C</b>) comparison of the proportion of upper band intensities (indicating the proportion of VLC) of ceramide, GlcCer, and SM shown in (<b>A</b>,<b>B</b>). The enlarged and darker images of [<sup>14</sup>C]serine-labeled ceramide, GlcCer, and SM shown in (<b>A</b>,<b>B</b>) are aligned. The proportions of VLC-containing lipids are expressed as described in <a href="#ijms-17-01761-f002" class="html-fig">Figure 2</a>B: mean percentage ± SD obtained from three independent experiments. The Bonferroni corrected Student’s <span class="html-italic">t</span>-test was used for multiple comparisons. * <span class="html-italic">p</span> &lt; 0.017.</p>
Full article ">Figure 4
<p><span class="html-italic">CERT</span> gene disruption by the TALEN–CERT pair in the TAL-CERS2#18 clone. (<b>A</b>) The target site of the TALEN–CERT pair in the human <span class="html-italic">CERT</span> gene and indel analysis of the TAL-CERS2#18-CERT#10 (#18-10) clones. The target sequences (CERT Fw and Rv) are located in exon 2, which codes for part of the pleckstrin homology (PH) domain. Deletions and replacements are shown in red and their lengths are specified on the right of the sequences. WT, wild type; (<b>B</b>) Western blot analysis of CERT in the TAL-CERS2#18 clones. Cell lysates from the parent cells and the indicated mutants were immunoblotted with anti-CERT antibody (<b>top</b>) and anti-α-tubulin antibody (<b>bottom</b>). The arrow indicates CERT at 68 kD. P, parent cell line; 18, TAL-CERS2#18 clone; 18-10, TAL-CERS2#18-CERT#10 clone.</p>
Full article ">Figure 5
<p>Effects of CERT disruption and Brefeldin A (BFA) treatment on the proportion of VLC species in sphingolipids. (<b>A</b>,<b>B</b>) Metabolic labeling of sphingolipids with radioactive serine in the parent cells (P), TAL-CERS2#18 clone (#18), and TAL-CERS2#18-CERT#10 clone (#18-10). Cells were cultured with [<sup>14</sup>C]serine in the presence (+) or absence (-) of BFA for 2 h, and lipids extracted from the cells were separated by Method 1 for GlcCer and SM in (<b>A</b>) and Method 3 for ceramide analysis in (<b>B</b>). The darker images are shown in <a href="#app1-ijms-17-01761" class="html-app">Figures S7A and S7C</a>; (<b>C</b>) comparison of the proportion of VLC of ceramide, GlcCer, and SM shown in (<b>A</b>,<b>B</b>). The method used is as described in <a href="#ijms-17-01761-f002" class="html-fig">Figure 2</a>B. The enlarged and darker images of [<sup>14</sup>C]serine-labeled ceramide, GlcCer, and SM shown in (<b>A</b>,<b>B</b>) are aligned. The proportions of VLC-containing lipids are expressed as described in <a href="#ijms-17-01761-f002" class="html-fig">Figure 2</a>B: mean percentage ± SD obtained from three independent experiments. The mean percentage in the TAL-CERS2#18 clone was compared with that in the TAL-CERS2#18-CERT#10 clone (#18-10) and that in the BFA-treated TAL-CERS2#18 clone, and the Bonferroni corrected Student’s <span class="html-italic">t</span>-test was used for the multiple comparisons. * <span class="html-italic">p</span> &lt; 0.025.</p>
Full article ">Figure 6
<p>Effect of UGCG overexpression on the proportion of VLC in GlcCer. (<b>A</b>) Western blot analysis of UGCG in UGCG-overexpressed TAL-CERS2#18 cells. Cell lysates from TAL-CERS2#18 cells and UGCG-overexpressed TAL-CERS2#18 cells were immunoblotted with anti-HA antibody (HA), anti-UGCG antibody (<b>top</b>), and anti-α-tubulin antibody (<b>bottom</b>). 18, TAL-CERS2#18 clone; UGCG, UGCG overexpression. The whole images are shown in <a href="#app1-ijms-17-01761" class="html-app">Figure S8</a>. (-), no transfection; UGCG, UGCG cDNA overexpression; (<b>B</b>) metabolic labeling of sphingolipids with radioactive serine in TAL-CERS2#18 and UGCG-overexpressed TAL-CERS2#18 cells. Cells were cultured with [<sup>14</sup>C]serine for 2 h, and lipids extracted from the cells were separated by Method 1 for GlcCer; (<b>C</b>) comparison of the proportions of VLC-GlcCer between endogenous GlcCer in TAL-CERS2#18 cells and overexpressed GlcCer in UGCG-overexpressed TAL-CERS2#18 cells. The proportions of VLC-containing GlcCer are expressed as described in <a href="#ijms-17-01761-f002" class="html-fig">Figure 2</a>B: mean percentage ± SD obtained from three independent experiments. The Student’s <span class="html-italic">t</span>-test was used. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
Back to TopTop