Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Cancer Res ; 81(23): 5849-5861, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34649947

ABSTRACT

Multiple noncoding natural antisense transcripts (ncNAT) are known to modulate key biological events such as cell growth or differentiation. However, the actual impact of ncNATs on cancer progression remains largely unknown. In this study, we identified a complete list of differentially expressed ncNATs in hepatocellular carcinoma. Among them, a previously undescribed ncNAT HNF4A-AS1L suppressed cancer cell growth by regulating its sense gene HNF4A, a well-known cancer driver, through a promoter-specific mechanism. HNF4A-AS1L selectively activated the HNF4A P1 promoter via HNF1A, which upregulated expression of tumor suppressor P1-driven isoforms, while having no effect on the oncogenic P2 promoter. RNA-seq data from 23 tissue and cancer types identified approximately 100 ncNATs whose expression correlated specifically with the activity of one promoter of their associated sense gene. Silencing of two of these ncNATs ENSG00000259357 and ENSG00000255031 (antisense to CERS2 and CHKA, respectively) altered the promoter usage of CERS2 and CHKA. Altogether, these results demonstrate that promoter-specific regulation is a mechanism used by ncNATs for context-specific control of alternative isoform expression of their counterpart sense genes. SIGNIFICANCE: This study characterizes a previously unexplored role of ncNATs in regulation of isoform expression of associated sense genes, highlighting a mechanism of alternative promoter usage in cancer.


Subject(s)
Carcinoma, Hepatocellular/pathology , Choline Kinase/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Liver Neoplasms/pathology , Membrane Proteins/metabolism , Promoter Regions, Genetic , RNA, Antisense/genetics , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Choline Kinase/antagonists & inhibitors , Choline Kinase/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, SCID , Prognosis , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
3.
FASEB J ; 35(2): e21287, 2021 02.
Article in English | MEDLINE | ID: mdl-33423335

ABSTRACT

Regulation of sphingolipid metabolism plays a role in cellular homeostasis, and dysregulation of these pathways is involved in cancer progression. Previously, our reports identified ceramide as an anti-metastatic lipid. In the present study, we investigated the biochemical alterations in ceramide-centered metabolism of sphingolipids that were associated with metastatic potential. We established metastasis-prone sublines of SKOV3 ovarian cancer cells using an in vivo selection method. These cells showed decreases in ceramide levels and ceramide synthase (CerS) 2 expression. Moreover, CerS2 downregulation in ovarian cancer cells promoted metastasis in vivo and potentiated cell motility and invasiveness. Moreover, CerS2 knock-in suppressed the formation of lamellipodia required for cell motility in this cell line. In order to define specific roles of ceramide species in cell motility controlled by CerS2, the effect of exogenous long- and very long-chain ceramide species on the formation of lamellipodia was evaluated. Treatment with distinct ceramides increased cellular ceramides and had inhibitory effects on the formation of lamellipodia. Interestingly, blocking the recycling pathway of ceramides by a CerS inhibitor was ineffective in the suppression of exogenous C24:1 -ceramide for the formation of lamellipodia. These results suggested that C24:1 -ceramide, a CerS2 metabolite, predominantly suppresses the formation of lamellipodia without the requirement for deacylation/reacylation. Moreover, knockdown of neutral ceramidase suppressed the formation of lamellipodia concomitant with upregulation of C24:1 -ceramide. Collectively, the CerS2-C24:1 -ceramide axis, which may be countered by neutral ceramidase, is suggested to limit cell motility and metastatic potential. These findings may provide insights that lead to further development of ceramide-based therapy and biomarkers for metastatic ovarian cancer.


Subject(s)
Cell Movement , Ceramides/metabolism , Membrane Proteins/metabolism , Ovarian Neoplasms/metabolism , Pseudopodia/metabolism , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Ceramides/pharmacology , Enzyme Inhibitors/pharmacology , Female , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Ovarian Neoplasms/pathology , Pseudopodia/drug effects , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
4.
Mol Carcinog ; 59(10): 1199-1208, 2020 10.
Article in English | MEDLINE | ID: mdl-32808708

ABSTRACT

Long noncoding RNAs (lncRNAs) can act as oncogene and tumor suppressor genes in many types of cancers including breast cancer (BC). Our previous study has indicated microRNA (miR)-125a-5p was downregulated and function as a tumor suppressor in BC. However, its upstream regulation mechanism is still unclear. In this study, we used bioinformatics algorithms, RNA pulldown assay, and dual-luciferase reports assay to predict and confirm lncRNA CERS6-AS1 interacted with miR-125a-5p. Then we found CERS6-AS1 was upregulated in BC tissues. Experimental results of tumor growth in nude mice show that CERS6-AS1 promotes tumor growth. Furthermore, CERS6-AS1 regulated BC susceptibility gene 1-associated protein 1 (BAP1) expression via sponging miR-125a-5p via Western blot analysis and quantitative polymerase chain reaction arrays. Finally, we showed that miR-125a-5p had opposing effects to those of CERS6-AS1 on BC cells, demonstrating that CERS6-AS1 may promote cell proliferation and inhibit cell apoptosis via sponging miR-125a-5p. Our results indicated CERS6-AS1 promote BC cell proliferation and inhibit cell apoptosis via sponging miR-125a-5p to upregulate BAP1 expression.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , Membrane Proteins/antagonists & inhibitors , MicroRNAs/genetics , Oligonucleotides, Antisense/genetics , RNA, Long Noncoding/genetics , Sphingosine N-Acyltransferase/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/genetics , Mice , Mice, Nude , Sphingosine N-Acyltransferase/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Xenograft Model Antitumor Assays
5.
Toxins (Basel) ; 12(5)2020 05 12.
Article in English | MEDLINE | ID: mdl-32408599

ABSTRACT

Weaned piglets (n = 3 × 6) were fed 0, 15 and 30 mg/kg diet fumonisin (FB1, FB2 and FB3, i.e., FBs, a sphinganine analogue mycotoxin), from the age of 35 days for 21 days, to assess mycotoxin induced, dose-dependent changes in the red cells' membrane. Ouabain sensitive Na+/K+ ATPase activity was determined from lysed red cell membranes, membrane fatty acid (FA) profile was analysed, as well as antioxidant and lipid peroxidation endpoints. Final body weight was higher in the 30 mg/kg group (vs. control), even besides identical cumulative feed intake. After 3 weeks, there was a difference between control and the 30 mg/kg group in red cell membrane sodium pump activity; this change was dose-dependent (sig.: 0.036; R2 = 0.58). Membrane FA profile was strongly saturated with non-systematic inter-group differences; pooled data provided negative correlation with sodium pump activity (all individual membrane n6 FAs). Intracellular antioxidants (reduced glutathione and glutathione peroxidase) and lipid peroxidation indicators (conj. dienes, trienes and malondialdehyde) were non-responsive. We suppose a ceramide synthesis inhibitor (FB1) effect exerted onto the cell membrane, proven to be toxin dose-dependent and increasing sodium pump activity, with only indirect FA compositional correlations and lack of lipid peroxidation.


Subject(s)
Erythrocyte Membrane/drug effects , Fumonisins/toxicity , Oxidative Stress/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Administration, Oral , Animals , Antioxidants/metabolism , Dose-Response Relationship, Drug , Erythrocyte Membrane/metabolism , Fatty Acids/metabolism , Fumonisins/administration & dosage , Lipid Peroxidation/drug effects , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/metabolism , Sus scrofa , Up-Regulation
6.
Mol Metab ; 21: 36-50, 2019 03.
Article in English | MEDLINE | ID: mdl-30655217

ABSTRACT

OBJECTIVE: Ectopic fat deposition is associated with increased tissue production of ceramides. Recent genetic mouse studies suggest that specific sphingolipid C16:0 ceramide produced by ceramide synthase 6 (CerS6) plays an important role in the development of insulin resistance. However, the therapeutic potential of CerS6 inhibition not been demonstrated. Therefore, we pharmacologically investigated the selective ablation of CerS6 using antisense oligonucleotides (ASO) in obese insulin resistance animal models. METHODS: We utilized ASO as therapeutic modality, CerS6 ASO molecules designed and synthesized were initially screened for in-vitro knock-down (KD) potency and cytotoxicity. ASOs with >85% inhibition of CerS6 mRNA were selected for further investigations. Most promising ASOs verified for in-vivo KD efficacy in healthy mice. CerS6 ASO (AAGATGAGCCGCACC) was found most active with hepatic reduction of CerS6 mRNA expression. Prior to longitudinal metabolic studies, we performed a dose titration target engagement analysis with CerS6 ASO in healthy mice to select the optimal dose. Next, we utilized leptin deficiency ob/ob and high fat diet (HFD) induced obese mouse models for pharmacological efficacy study. RESULTS: CerS6 expression were significantly elevated in the liver and brown adipose, this was correlated with significantly elevated C16:0 ceramide concentrations in plasma and liver. Treatment with CerS6 ASO selectively reduced CerS6 expression by ∼90% predominantly in the liver and this CerS6 KD resulted in a significant reduction of C16:0 ceramide by about 50% in both liver and plasma. CerS6 KD resulted in lower body weight gain and accompanied by a significant reduction in whole body fat and fed/fasted blood glucose levels (1% reduction in HbA1c). Moreover, ASO-mediated CerS6 KD significantly improved oral glucose tolerance (during oGTT) and mice displayed improved insulin sensitivity. Thus, CerS6 appear to play an important role in the development of obesity and insulin resistance. CONCLUSIONS: Our investigations identified specific and selective therapeutic valid ASO for CerS6 ablation in in-vivo. CerS6 should specifically be targeted for the reduction of C16:0 ceramides, that results in amelioration of insulin resistance, hyperglycemia and obesity. CerS6 mediated C16:0 ceramide reduction could be a potentially attractive target for the treatment of insulin resistance, obesity and type 2 diabetes.


Subject(s)
Ceramides/metabolism , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Oligonucleotides, Antisense/metabolism , Sphingosine N-Acyltransferase/metabolism , Adipose Tissue, Brown/metabolism , Animals , Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Knockdown Techniques , Hep G2 Cells , Humans , Insulin Resistance , Leptin/deficiency , Liver/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Oligonucleotides, Antisense/pharmacology , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/genetics , Thionucleotides , Weight Gain
7.
Biochimie ; 149: 41-50, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29625159

ABSTRACT

Fumonisin B1 toxin (FB1) is a well-known competitive inhibitor of ceramide synthase (CS) in yeast. However, FB1 is unable to obstruct CS from Trichoderma spp., which are well-known biocontrol agents. To explore the contrasting binding modes, a comparative structural analysis of complexes of FB1 with these two CS proteins was carried out. Formation of activation loop on the binding of substrates with the CS from yeast was observed but when inhibitor interacted with the activation loop, it transformed into helix leading to the potentially inactivated state of the enzyme. In yeast homologue of the enzyme, the inhibitor and substrates compete for the same binding site. Whereas, in the CS protein from Trichoderma guizhouense, no such competition for substrate binding site was observed and the binding pocket of the enzyme could easily accommodate FB1 molecule along with the two interacting native substrates, which may lead to the successful catalysis.


Subject(s)
Fumonisins/chemistry , Sphingolipids/chemistry , Sphingosine N-Acyltransferase/chemistry , Trichoderma/enzymology , Fumonisins/pharmacology , Protein Binding , Protein Conformation , Signal Transduction/drug effects , Sphingosine N-Acyltransferase/antagonists & inhibitors , Substrate Specificity , Trichoderma/chemistry , Yeasts/chemistry , Yeasts/enzymology
8.
FASEB J ; 32(1): 130-142, 2018 01.
Article in English | MEDLINE | ID: mdl-28864659

ABSTRACT

Perilipin 2 (PLIN2) is a lipid-droplet protein that is up-regulated in alcoholic steatosis and associated with hepatic accumulation of ceramides, bioactive lipids implicated in alcoholic liver disease pathogenesis. The specific role of ceramide synthetic enzymes in the regulation of PLIN2 and promotion of hepatocellular lipid accumulation is not well understood. We examined the effects of pharmacologic ceramide synthesis inhibition on hepatic PLIN2 expression, steatosis, and glucose and lipid homeostasis in mice with alcoholic steatosis and in ethanol-incubated human hepatoma VL17A cells. In cells, pharmacologic inhibition of ceramide synthase reduced lipid accumulation by reducing PLIN2 RNA stability. The subtype ceramide synthase (CerS)6 was specifically up-regulated in experimental alcoholic steatosis in vivo and in vitro and was up-regulated in zone 3 hepatocytes in human alcoholic steatosis. In vivo ceramide reduction by inhibition of de novo ceramide synthesis reduced PLIN2 and hepatic steatosis in alcohol-fed mice, but only de novo synthesis inhibition, not sphingomyelin hydrolysis, improved glucose tolerance and dyslipidemia. These findings implicate CerS6 as a novel regulator of PLIN2 and suggest that ceramide synthetic enzymes may promote the earliest stage of alcoholic liver disease, alcoholic steatosis.-Williams, B., Correnti, J., Oranu, A., Lin, A., Scott, V., Annoh, M., Beck, J., Furth, E., Mitchell, V., Senkal, C. E., Obeid, L., Carr, R. M. A novel role for ceramide synthase 6 in mouse and human alcoholic steatosis.


Subject(s)
Fatty Liver, Alcoholic/enzymology , Membrane Proteins/metabolism , Sphingosine N-Acyltransferase/metabolism , Animals , Biosynthetic Pathways , Cell Line , Ceramides/biosynthesis , Disease Models, Animal , Ethanol , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/genetics , Glucose/metabolism , Humans , Lipid Metabolism , Liver/drug effects , Liver/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Perilipin-2/genetics , Perilipin-2/metabolism , RNA Stability , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/genetics , Up-Regulation/drug effects
9.
J Mol Med (Berl) ; 95(10): 1053-1064, 2017 10.
Article in English | MEDLINE | ID: mdl-28695226

ABSTRACT

Cystic fibrosis is the most common genetic disease, in which symptoms may be alleviated but not fully eliminated. Ceramides have long been implicated in the inflammatory etiology of cystic fibrosis, with contradicting reports with regards to their role. Recently, significant biological and biophysical differences have been observed between long- and very long-chain ceramides. This work reveals that long-chain ceramides are upregulated whereas very long-chain ceramides are downregulated in cell lines, mouse animal model, and patients with cystic fibrosis, compared with their controls. Treatment with fenretinide decreases the levels of long-chain ceramides and increases the levels of very long-chain ceramides. Our results show that restoration of cystic fibrosis conductance regulator (CFTR) expression is associated with normalization of aberrant levels of specific ceramides. This demonstrates for the first time a correlation between CFTR protein expression and regulation of specific ceramide levels. Furthermore, using cystic fibrosis lung epithelial cell lines, we demonstrate that this effect can be attributed to the transcriptional downregulation of ceramide synthase 5 (Cers5) enzyme. We also discovered a partial synergism between fenretinide and zinc (Zn2+), which deficiency has been reported in patients with cystic fibrosis. Overall, in addition to having direct translational application, we believe that our findings contribute to the understanding of ceramide metabolism in cystic fibrosis, as well as other inflammatory diseases where imbalances of ceramides have also been observed. KEY MESSAGES: Long- and very long-chain ceramides (LCCs and VLCCs) are biochemically distinct. LCCs are upregulated whereas VLCCs are downregulated in cystic fibrosis. Fenretinide downregulates the levels of LCCs and upregulates the levels of VLCCs. Fenretinide changes the balance of LCCs and VLCCs by downregulating Cers5 enzyme. Fenretinide and zinc ions cooperate in the modulation of ceramide levels.


Subject(s)
Ceramides/metabolism , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Down-Regulation/drug effects , Fenretinide/therapeutic use , Sphingosine N-Acyltransferase/metabolism , Adolescent , Adult , Animals , Cell Line , Ceramides/analysis , Ceramides/blood , Cystic Fibrosis/blood , Disease Models, Animal , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , PPAR gamma/agonists , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/genetics , Transcriptional Activation/drug effects , Young Adult
10.
FEBS Lett ; 590(7): 971-81, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26950647

ABSTRACT

Drosophila Ceramide Synthase (CerS) Schlank regulates both ceramide synthesis and fat metabolism. Schlank contains a catalytic lag1p motif and, like many CerS in other species, a homeodomain of unknown function. Here, we show that the Drosophila CerS Schlank is imported into the nucleus and requires two nuclear localization signals (NLSs) within its homeodomain and functional Importin-ß import machinery. Expression of Schlank variants containing the homeodomain without functional lag1p motif rescued the fat metabolism phenotype of schlank mutants whereas a variant with a mutated NLS site did not rescue. Thus, the homeodomain of Schlank is involved in the regulation of lipid metabolism independent of the catalytic lag1p motif.


Subject(s)
Cell Nucleus/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Fat Body/metabolism , Lipid Metabolism , Nuclear Localization Signals/metabolism , Sphingosine N-Acyltransferase/metabolism , Active Transport, Cell Nucleus , Amino Acid Motifs , Amino Acid Substitution , Animals , Animals, Genetically Modified , Catalytic Domain , Cell Line , Cell Nucleus/enzymology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Fat Body/cytology , Fat Body/enzymology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mutation , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/genetics , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/chemistry , Sphingosine N-Acyltransferase/genetics , beta Karyopherins/antagonists & inhibitors , beta Karyopherins/genetics , beta Karyopherins/metabolism
11.
PLoS One ; 11(1): e0146618, 2016.
Article in English | MEDLINE | ID: mdl-26783755

ABSTRACT

We previously reported that ceramide synthase 6 (CerS6) is elevated in response to folate stress in cancer cells, leading to enhanced production of C16-ceramide and apoptosis. Antifolate methotrexate (MTX), a drug commonly used in chemotherapy of several types of cancer, is a strong inhibitor of folate metabolism. Here we investigated whether this drug targets CerS6. We observed that CerS6 protein was markedly elevated in several cancer cell lines treated with MTX. In agreement with the enzyme elevation, its product C16-ceramide was also strongly elevated, so as several other ceramide species. The increase in C16-ceramide, however, was eliminated in MTX-treated cells lacking CerS6 through siRNA silencing, while the increase in other ceramides sustained. Furthermore, the siRNA silencing of CerS6 robustly protected A549 lung adenocarcinoma cells from MTX toxicity, while the silencing of another ceramide synthase, CerS4, which was also responsive to folate stress in our previous study, did not interfere with the MTX effect. The rescue effect of CerS6 silencing upon MTX treatment was further confirmed in HCT116 and HepG2 cell lines. Interestingly, CerS6 itself, but not CerS4, induced strong antiproliferative effect in several cancer cell lines if elevated by transient transfection. The effect of MTX on CerS6 elevation was likely p53 dependent, which is in agreement with the hypothesis that the protein is a transcriptional target of p53. In line with this notion, lometrexol, the antifolate inducing cytotoxicity through the p53-independent mechanism, did not affect CerS6 levels. We have also found that MTX induces the formation of ER aggregates, enriched with CerS6 protein. We further demonstrated that such aggregation requires CerS6 and suggests that it is an indication of ER stress. Overall, our study identified CerS6 and ceramide pathways as a novel MTX target.


Subject(s)
Cell Proliferation/drug effects , Membrane Proteins/antagonists & inhibitors , Methotrexate/pharmacology , Sphingosine N-Acyltransferase/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Ceramides/metabolism , HCT116 Cells , HeLa Cells , Hep G2 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Targeted Therapy , RNA, Small Interfering/pharmacology , Sphingosine N-Acyltransferase/genetics , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Protein p53/genetics
12.
Biochem J ; 473(5): 593-603, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26635357

ABSTRACT

Ceramide makes up the acyl-backbone of sphingolipids and plays a central role in determining the function of these essential membrane lipids. In Arabidopsis, the varied chemical composition of ceramide is determined by the specificity of three different isoforms of ceramide synthase, denoted LAG one homologue 1, -2 and -3 (LOH1, LOH2 and LOH3), for a range of long-chain base (LCB) and acyl-CoA substrates. The contribution of each of these isoforms to the synthesis of ceramide was investigated by in vitro ceramide synthase assays. The plant LCB phytosphingosine was efficiently used by the LOH1 and LOH3 isoforms, with LOH1 having the lowest Km for the LCB substrate of the three isoforms. In contrast, sphinganine was used efficiently only by the LOH2 isoform. Acyl-CoA specificity was also distinguished between the three isoforms with LOH2 almost completely specific for palmitoyl-CoA whereas the LOH1 isoform showed greatest activity with lignoceroyl- and hexacosanoyl-CoAs. Interestingly, unsaturated acyl-CoAs were not used efficiently by any isoform whereas unsaturated LCB substrates were preferred by LOH2 and 3. Fumonisin B1 (FB1) is a general inhibitor of ceramide synthases but LOH1 was found to have a much lower Ki than the other isoforms pointing towards the origin of FB1 sensitivity in plants. Overall, the data suggest distinct roles and modes of regulation for each of the ceramide synthases in Arabidopsis sphingolipid metabolism.


Subject(s)
Arabidopsis Proteins/antagonists & inhibitors , Fumonisins/chemistry , Sphingosine N-Acyltransferase/antagonists & inhibitors , Acyl Coenzyme A/metabolism , Arabidopsis/enzymology , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Enzyme Assays , Hydroxylation , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Kinetics , Microsomes/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine N-Acyltransferase/chemistry , Sphingosine N-Acyltransferase/genetics , Substrate Specificity
13.
J Clin Invest ; 126(1): 254-65, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26650179

ABSTRACT

Sphingolipids make up a family of molecules associated with an array of biological functions, including cell death and migration. Sphingolipids are often altered in cancer, though how these alterations lead to tumor formation and progression is largely unknown. Here, we analyzed non-small-cell lung cancer (NSCLC) specimens and cell lines and determined that ceramide synthase 6 (CERS6) is markedly overexpressed compared with controls. Elevated CERS6 expression was due in part to reduction of microRNA-101 (miR-101) and was associated with increased invasion and poor prognosis. CERS6 knockdown in NSCLC cells altered the ceramide profile, resulting in decreased cell migration and invasion in vitro, and decreased the frequency of RAC1-positive lamellipodia formation while CERS6 overexpression promoted it. In murine models, CERS6 knockdown in transplanted NSCLC cells attenuated lung metastasis. Furthermore, combined treatment with l-α-dimyristoylphosphatidylcholine liposome and the glucosylceramide synthase inhibitor D-PDMP induced cell death in association with ceramide accumulation and promoted cancer cell apoptosis and tumor regression in murine models. Together, these results indicate that CERS6-dependent ceramide synthesis and maintenance of ceramide in the cellular membrane are essential for lamellipodia formation and metastasis. Moreover, these results suggest that targeting this homeostasis has potential as a therapeutic strategy for CERS6-overexpressing NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Membrane Proteins/physiology , Sphingosine N-Acyltransferase/physiology , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Ceramides/metabolism , Dimyristoylphosphatidylcholine/pharmacology , Humans , Lung Neoplasms/drug therapy , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , MicroRNAs/physiology , Neoplasm Metastasis , Phenotype , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/genetics
14.
Tumour Biol ; 36(8): 5763-71, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25724183

ABSTRACT

Hepatocellular carcinoma (HCC) is a highly aggressive and lethal neoplasm with poor prognosis. The aim of this study is to investigate the anticancer activity of cinobufotalin, a bufadienolide isolated from toad venom, in cultured HCC cells, and to study the underlying mechanisms. We found that cinobufotalin (at nmol/L) significantly inhibited HCC cell growth and survival while inducing considerable cell apoptosis. Further, cinobufotalin inhibited sphingosine kinase 1 (SphK1) activity and induced pro-apoptotic ceramide production. Ceramide synthase-1 small hairpin RNA (shRNA)-depletion inhibited cinobufotalin-induced ceramide production and HCC cell apoptosis. On the other hand, the glucosylceramide synthase (GCS) inhibitor 1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP) facilitated cinobufotalin-induced ceramide production and cell apoptosis. SphK1 inhibitor II (SKI-II), similar to cinobufotalin, increased cellular ceramide level and promoted HCC cell apoptosis. Finally, we observed that cinobufotalin inactivated Akt-S6K1 signaling in HepG2 cells, which was again inhibited by ceramide synthase-1 shRNA-depletion. In conclusion, the results of this study suggest that cinobufotalin induces growth inhibition and apoptosis in cultured HCC cells through ceramide production. Cinobufotalin may be investigated as a novel anti-HCC agent.


Subject(s)
Bufanolides/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Ceramides/biosynthesis , Liver Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucosyltransferases/antagonists & inhibitors , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Morpholines/administration & dosage , Sphingosine N-Acyltransferase/antagonists & inhibitors
15.
Cell Death Dis ; 6: e1691, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25766330

ABSTRACT

Although numerous pathogenic changes within the mitochondrial respiratory chain (RC) have been associated with an elevated occurrence of apoptosis within the affected tissues, the mechanistic insight into how mitochondrial dysfunction initiates apoptotic cell death is still unknown. In this study, we show that the specific alteration of the cytochrome c oxidase (COX), representing a common defect found in mitochondrial diseases, facilitates mitochondrial apoptosis in response to oxidative stress. Our data identified an increased ceramide synthase 6 (CerS6) activity as an important pro-apoptotic response to COX dysfunction induced either by chemical or genetic approaches. The elevated CerS6 activity resulted in accumulation of the pro-apoptotic C16 : 0 ceramide, which facilitates the mitochondrial apoptosis in response to oxidative stress. Accordingly, inhibition of CerS6 or its specific knockdown diminished the increased susceptibility of COX-deficient cells to oxidative stress. Our results provide new insights into how mitochondrial RC dysfunction mechanistically interferes with the apoptotic machinery. On the basis of its pivotal role in regulating cell death upon COX dysfunction, CerS6 might potentially represent a novel target for therapeutic intervention in mitochondrial diseases caused by COX dysfunction.


Subject(s)
Apoptosis/genetics , Cytochrome-c Oxidase Deficiency/metabolism , Electron Transport Complex IV/biosynthesis , Membrane Proteins/biosynthesis , Sphingosine N-Acyltransferase/biosynthesis , Animals , Cytochrome-c Oxidase Deficiency/genetics , Cytochrome-c Oxidase Deficiency/pathology , Electron Transport Complex IV/genetics , HeLa Cells , Humans , Membrane Proteins/antagonists & inhibitors , Mice , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress , Oxygen Consumption , Sphingosine N-Acyltransferase/antagonists & inhibitors
16.
Arch Toxicol ; 89(9): 1619-29, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25155190

ABSTRACT

Fumonisin B1 (FB1) is a well-known inhibitor of de novo sphingolipid biosynthesis, due to its ability to inhibit ceramide synthases (CerS) activity. In mammals, this toxin triggers broad clinical symptoms with multi-organ dysfunction such as hepatotoxicity or pulmonary edema. The molecular mechanism of CerS inhibition by FB1 remains unknown. Due to the existence of six mammalian CerS isoforms with a tissue-specific expression pattern, we postulated that the organ-specific adverse effects of FB1 might be due to different CerS isoforms. The sphingolipid contents of lung and liver were compared in normal and FB1-exposed piglets (gavage with 1.5 mg FB1/kg body weight daily for 9 days). The effect of the toxin on each CerS was deduced from the analysis of its effects on individual ceramide (Cer) and sphingomyelin (SM) species. As expected, the total Cer content decreased by half in the lungs of FB1-exposed piglets, while in contrast, total Cer increased 3.5-fold in the livers of FB1-exposed animals. Our data also indicated that FB1 is more prone to bind to CerS4 and CerS2 to deplete lung and to enrich liver in d18:1/C20:0 and d18:1/C22:0 ceramides. It also interact with CerS1 to enrich liver in d18:1/C18:0 ceramides. Cer levels were counterbalanced by those of SM. In conclusion, these results demonstrate that the specificity of the effects of FB1 on tissues and organs is due to the effects of the toxin on CerS4, CerS2, and CerS1.


Subject(s)
Fumonisins/toxicity , Liver/drug effects , Lung/drug effects , Sphingosine N-Acyltransferase/antagonists & inhibitors , Animals , Enzyme Inhibitors/toxicity , Liver/metabolism , Lung/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Sphingolipids/biosynthesis , Sphingosine N-Acyltransferase/metabolism , Swine , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism
17.
Oxid Med Cell Longev ; 2014: 823071, 2014.
Article in English | MEDLINE | ID: mdl-24817993

ABSTRACT

Oxidized low density of lipoprotein (oxLDL) is the major lipid found in atherosclerotic lesion and elevated plasma oxLDL is recognized to be a risk factor of atherosclerosis. Whether plasma oxLDL could be transported across endothelial cells and initiate atherosclerotic changes remains unknown. In an established in vitro cellular transcytosis model, the present study found that oxLDL could traffic across vascular endothelial cells and further that the regulation of endogenous ceramide production by ceramide metabolizing enzyme inhibitors significantly altered the transcytosis of oxLDL across endothelial cells. It was found that acid sphingomyelinase inhibitor, desipramine (DES), and de novo ceramide synthesis inhibitor, myriocin (MYR), both decreasing the endogenous ceramide production, significantly inhibited the transcytosis of oxLDL. Ceramidase inhibitor, N-oleoylethanolamine (NOE), and sphingomyelin synthase inhibitor, O-Tricyclo[5.2.1.02,6]dec-9-yl dithiocarbonate potassium salt (D609), both increasing the endogenous ceramide production, significantly upregulated the transcytosis of oxLDL. In vivo, injection of fluorescence labeled oxLDL into mice body also predisposed to the subendothelial retention of these oxidized lipids. The observations provided in the present study demonstrate that endogenous ceramide contributes to the transcytosis of oxLDL across endothelial cells and promotes the initiating step of atherosclerosis-the subendothelial retention of lipids in vascular wall.


Subject(s)
Ceramides/metabolism , Lipoproteins, LDL/metabolism , Animals , Bridged-Ring Compounds/pharmacology , Ceramidases/antagonists & inhibitors , Ceramidases/metabolism , Desipramine/pharmacology , Endocannabinoids/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Ethanolamines/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Norbornanes , Oleic Acids/pharmacology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/metabolism , Thiocarbamates , Thiones/pharmacology , Transcytosis/drug effects , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Transferases (Other Substituted Phosphate Groups)/metabolism , Up-Regulation/drug effects
18.
Biochem Pharmacol ; 80(11): 1632-40, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20735991

ABSTRACT

Ceramides serve as bioactive molecules with important roles in cell proliferation and apoptosis. Ceramides (Cer) with different N-acyl side chains (C(14:0)-Cer-C(26:0)-Cer) possess distinctive roles in cell signaling and are differentially expressed in HCT-116 colon cancer cells. Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, exhibiting antiproliferative effects, activates the sphingolipid pathway. To elucidate the mechanism, HCT-116 cells were treated with 50µM celecoxib leading to a significant increase of C(16:0)-Cer. Interestingly, 50µM celecoxib resulted in a 2.8-fold increase of ceramide synthase (CerS) activity as measured by a cell-based activity assay. siRNA against several CerSs revealed that CerS6 was predominantly responsible for the increase of C(16:0)-Cer in HCT-116 cells. Moreover, the silencing of CerS6 partially protected HCT-116 cells from the toxic effects induced by celecoxib. Treatment of cells with celecoxib and fumonisin B1 (inhibitor of CerSs) or myriocin (inhibitor of l-serine palmitoyl transferase) or desipramine (inhibitor of acid sphingomyelinase and acid ceramidase) revealed that the increase of C(16:0)-Cer results predominantly from activation of the salvage pathway. Using the nude mouse model we demonstrated that celecoxib induces also in vivo a significant increase of C(16:0)-Cer in stomach, small intestine and tumor tissue. In conclusion, celecoxib causes a specific increase of C(16:0)-Cer by activating CerS6 and the salvage pathway, which contribute to the toxic effects of celecoxib.


Subject(s)
Ceramides/biosynthesis , Membrane Proteins/metabolism , Pyrazoles/pharmacology , Sphingosine N-Acyltransferase/metabolism , Sulfonamides/pharmacology , Animals , Celecoxib , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , HCT116 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mice , Mice, Nude , Pyrazoles/toxicity , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/physiology , Sulfonamides/toxicity , Tissue Distribution/drug effects , Tissue Distribution/physiology
19.
J Pharmacol Exp Ther ; 324(2): 548-57, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18042827

ABSTRACT

Although mechanisms involved in the pathogenesis of asthma remain unclear, roles for oxidative/nitrosative stress, epithelial cell apoptosis, and airway inflammation have been documented. Ceramide is a sphingolipid with potent proinflammatory and proapoptotic properties. This study aimed at determining whether increased formation of ceramide contributes to the development of airway inflammation and hyper-responsiveness, using a well characterized in vivo model of allergic asthmatic response and airway inflammation in ovalbumin-sensitized guinea pigs. Aerosol administration of ovalbumin increased ceramide levels and ceramide synthase activity in the airway epithelium associated with respiratory abnormalities, such as cough, dyspnea, and severe bronchoconstriction. These abnormalities correlated with nitrotyrosine formation in the airway epithelium and oxidative/nitrosative stress, epithelial cell apoptosis, and airway inflammation evident by the infiltration of neutrophils and eosinophils in lung tissues, mast cell degranulation, and release of prostaglandin D(2) and proinflammatory cytokines. Inhibition of de novo ceramide synthesis with the competitive and reversible inhibitor of ceramide synthase fumonisin B1 (0.25, 0.5 and 1 mg/kg b.wt.), given i.p. daily for 4 days before allergen challenge, attenuated nitrotyrosine formation and oxidative/nitrosative stress, epithelial cell apoptosis, and airway inflammation while improving the respiratory and histopathological abnormalities. These results implicate ceramide in the development of allergic asthmatic response and airway inflammation. Strategies aimed at reducing the levels of ceramide and downstream events should yield promising novel anti-asthmatic agents.


Subject(s)
Asthma/enzymology , Disease Models, Animal , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/biosynthesis , Allergens/toxicity , Animals , Asthma/chemically induced , Asthma/drug therapy , Bronchoconstriction/drug effects , Bronchoconstriction/physiology , Fumonisins/pharmacology , Fumonisins/therapeutic use , Guinea Pigs , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/enzymology , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...