Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Bioorg Med Chem ; 32: 116011, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33461145

ABSTRACT

B13 is an acid ceramidase (ACDase) inhibitor. The two chiral centers of this aromatic amido alcohol lead to four stereoisomers, yet we have little knowledge about its erythro- enantiomers, (1R, 2S) and (1S, 2R). In this paper, for the first time, the synthesis of two erythro- enantiomers is described, and the compounds are evaluated along with two threo- enantiomers, (1R, 2R) and (1S, 2S). The key metabolites and sphingolipid (SL) profile of the full set of B13 stereoisomers in MCF7 breast carcinoma cells are presented. The results demonstrated that the erythro- enantiomers were more effective than the threo- enantiomers on growth inhibition in MCF7 cells, although there were no statistically significant differences within the threo- and erythro- series. Measurement of intracellular levels of the compounds indicated that the erythro- seemed a little more cell permeable than the threo- enantiomers; also, the (1R, 2S) isomer with the same stereo structure as natural ceramide (Cer) could be hydrolyzed and phosphorylated in MCF7 cells. Furthermore, we also observed the formation of C16 homologs from the full set of B13 isomers within the cells, indicating the occurrence of de-acylation and re-acylation of the amino group of the aromatic alcohol. Moreover, the decrease in the Cer/Sph ratio suggests that the growth inhibition from (1R, 2S) isomer is not because of the inhibition of ceramidases. Taken together, (1R, 2S) could be developed as a substitute of natural Cer.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , Propanolamines/pharmacology , Sphingolipids/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Molecular Structure , Propanolamines/chemical synthesis , Propanolamines/chemistry , Sphingolipids/metabolism , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
2.
J Neurochem ; 154(6): 662-672, 2020 09.
Article in English | MEDLINE | ID: mdl-32058598

ABSTRACT

A major dose-limiting side effect of docetaxel chemotherapy is peripheral neuropathy. Patients' symptoms include pain, numbness, tingling and burning sensations, and motor weakness in the extremities. The molecular mechanism is currently not understood, and there are no treatments available. Previously, we have shown an association between neuropathy symptoms of patients treated with paclitaxel and the plasma levels of neurotoxic sphingolipids, the 1-deoxysphingolipids (1-deoxySL) (Kramer et al, FASEB J, 2015). 1-DeoxySL are produced when the first enzyme of the sphingolipid biosynthetic pathway, serine palmitoyltransferase (SPT), uses L-alanine as a substrate instead of its canonical amino acid substrate, L-serine. In the current investigation, we tested whether 1-deoxySL accumulate in the nervous system following systemic docetaxel treatment in mice. In dorsal root ganglia (DRG), we observed that docetaxel (45 mg/kg cumulative dose) significantly elevated the levels of 1-deoxySL and L-serine-derived ceramides, but not sphingosine-1-phosphate (S1P). S1P is a bioactive sphingolipid and a ligand for specific G-protein-coupled receptors. In the sciatic nerve, docetaxel decreased 1-deoxySL and ceramides. Moreover, we show that in primary DRG cultures, 1-deoxysphingosine produced neurite swellings that could be reversed with S1P. Our results demonstrate that docetaxel chemotherapy up-regulates sphingolipid metabolism in sensory neurons, leading to the accumulation of neurotoxic 1-deoxySL. We suggest that the neurotoxic effects of 1-deoxySL on axons can be reversed with S1P.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Docetaxel/toxicity , Neurotoxicity Syndromes/prevention & control , Serine C-Palmitoyltransferase/metabolism , Sphingolipids/metabolism , Sphingolipids/toxicity , Animals , Axons/drug effects , Axons/pathology , Ceramides/metabolism , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Lipids/pharmacology , Lysophospholipids/pharmacology , Mice , Mice, Inbred C57BL , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Serine/metabolism , Serine C-Palmitoyltransferase/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
3.
PLoS One ; 14(4): e0215770, 2019.
Article in English | MEDLINE | ID: mdl-31002723

ABSTRACT

The purpose of this study was to determine the profile of bioactive sphingolipids in xenograft mouse tissues of head and neck squamous cell carcinoma. We utilized UHPLC-MS/MS to determine the profile of full set of ceramides, sphingosine, and sphingosine 1-phosphate in this xenograft mouse model. The tissues isolated and investigated were from brain, lung, heart, liver, spleen, kidney, bladder, tumors and blood. With the exception of equal volume of blood plasma (100ul), all tissues were studied with the same amount of protein (800ug). Results demonstrated that brain contained the highest level of ceramide and kidney had the highest level of sphingosine, whereas sphingosine 1-phosphate and dihydrosphingosine 1-phosphate were heavily presented in the blood. Brain also comprised the highest level of phospholipids. As for the species, several ceramides, usually present in very low amounts in cultured tumor cells, showed relatively high levels in certain tissues. This study highlights levels of bioactive sphingolipids profiles in xenograft mouse model of head and neck squamous cell carcinoma, and provides resources to investigate potential therapeutic targets and biomarkers that target bioactive sphingolipids metabolism pathways.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Sphingolipids/metabolism , Animals , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Ceramides/metabolism , Chromatography, High Pressure Liquid/methods , Disease Models, Animal , Head and Neck Neoplasms/pathology , Humans , Lysophospholipids/metabolism , Mice, Nude , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Tandem Mass Spectrometry/methods , Transplantation, Heterologous
4.
Bioorg Med Chem ; 26(23-24): 6067-6075, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30448190

ABSTRACT

The function of acid ceramidase (ACDase), whose congenital deficiency leads to Farber disease, has been recognized to be vital to tumor cell biology, and inhibition of its activity may be beneficial in cancer therapy. Therefore, manipulation of the activity of this enzyme may have significant effect, especially on cancer cells. LCL521, Di-DMG-B13, is a lysosomotropic inhibitor of ACDase. Here we define complexities in the actions of LCL521 on ACDase. Systematic studies in MCF7 cells showed dose and time divergent action of LCL521 on ACDase protein expression and sphingolipid levels. Low dose of LCL521 (1 µM) effectively inhibited ACDase in cells, but the effects were transient. A higher dose of LCL521 (10 µM) caused a profound decrease of sphingosine and increase of ceramide, but additionally affected the processing and regeneration of the ACDase protein, with biphasic and reversible effects on the expression of ACDase, which paralleled the long term changes of cellular sphingosine and ceramide. Finally, the higher concentrations of LCL521 also inhibited Dihydroceramide desaturase (DES-1). In summary, LCL521 exhibits significant effects on ACDase in a dose and time dependent manner, but dose range and treatment time need to be paid attention to specify its future exploration on ACDase targeted cancer treatment.


Subject(s)
Acetates/pharmacology , Acid Ceramidase/antagonists & inhibitors , Amines/pharmacology , Enzyme Inhibitors/pharmacology , Sphingolipids/antagonists & inhibitors , Acid Ceramidase/metabolism , Dose-Response Relationship, Drug , Humans , MCF-7 Cells , Molecular Structure , Sphingolipids/metabolism , Structure-Activity Relationship , Time Factors , Tumor Cells, Cultured
5.
Glia ; 66(3): 654-669, 2018 03.
Article in English | MEDLINE | ID: mdl-29193293

ABSTRACT

Recent evidence suggests that the oral drug Fingolimod (FTY720) for relapsing-remitting multiple sclerosis (MS) may act directly on the central nervous system (CNS) and modulate disease pathogenesis and progression in experimental models of MS. However, the specific subtype of sphingosine-1-phosphate (S1P) receptors that mediates the effect of FTY720 on the CNS cells has not been fully elucidated. Here, we report that S1P receptor 1 (S1PR1) is elevated in reactive astrocytes in an autoimmunity independent mouse model of MS and that selective S1PR1 modulation is sufficient to ameliorate the loss of oligodendrocytes and demyelination. The non-selective S1PR modulator, FTY720, or a short-lived S1PR1-specific modulator, CYM5442, was administered daily to mice while on cuprizone diet. Both FTY720- and CYM5422-treated mice displayed a significant reduction in oligodendrocyte apoptosis and astrocyte and microglial activation in comparison to vehicle-treated groups, which was associated with decreased production of proinflammatory mediators and down-regulation of astrocytic S1PR1 protein. Interestingly, S1PR1 modulation during the early phase of cuprizone intoxication was required to suppress oligodendrocyte death and consequent demyelination as drug treatment from 10 days after the initiation of cuprizone feeding was no longer effective. CYM5442 treatment during the brief cuprizone exposure significantly prevented Il-1ß, Il-6, Cxcl10, and Cxcl3 induction, resulting in suppression of subsequent reactive gliosis and demyelination. Our study identifies functional antagonism of S1PR1 as a major mechanism for the protective effect of FTY720 in the cuprizone model and suggests pathogenic contributions of astrocyte S1PR1 signaling in primary demyelination and its potential as a therapeutic target for CNS inflammation.


Subject(s)
Demyelinating Diseases/drug therapy , Fingolimod Hydrochloride/pharmacology , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Receptors, Lysosphingolipid/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain/drug effects , Brain/metabolism , Brain/pathology , Chimera , Cuprizone , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Gene Expression/drug effects , Gliosis/drug therapy , Gliosis/metabolism , Gliosis/pathology , Indans/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/metabolism , Neuroglia/pathology , Oxadiazoles/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors , White Matter/drug effects , White Matter/metabolism , White Matter/pathology
6.
J Transl Med ; 15(1): 120, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28583134

ABSTRACT

BACKGROUND: Accumulating evidence suggests that sphingosine kinase 1 (SphK1)/sphingosine 1-phosphate pathway plays a pivotal role in colon carcinogenesis. METHODS: To further support the evidence, we investigated the effects of SphK1 using three separate animal models: SphK1 knockout mice, SphK1 overexpressing transgenic mice, and SphK1 overexpression in human colon cancer xenografts. Using azoxymethane (AOM, colon carcinogen), we analyzed colon tumor development in SphK1 KO and SphK1 overexpression in intestinal epithelial cells regulated by a tet-on system. Then, we analyzed subcutaneous tumor growth using xenografts of HT-29 human colon cancer cell. Finally, immunohistochemical analyses for SphK1 and COX-2 were performed on human colon cancer tissue microarray. RESULTS: SphK1 KO mice, compared to wild-type mice, demonstrated a significant inhibition in colon cancer development induced by AOM (58.6% vs. 96.4%, respectively, P < 0.005). Tumor multiplicity (1.00 vs. 1.64 per colon, respectively, P < 0.05) and tumor volume (14.82 mm3 vs. 29.10 mm3, P < 0.05) were both significantly reduced in SphK1 KO mice compared to wild-type mice. Next, SphK1 overexpression in HT-29 enhanced tumor growth as compared to GFP control in nude mice (229.5 mm3 vs. 90.9 mm3, respectively, P < 0.05). Furthermore, overexpression of SphK1 in intestinal epithelial cells significantly enhances AOM-induced colon tumor formation (P < 0.05). Lastly, SphK1 and COX-2 intensity tended to reduce overall survival of late stage colon cancer patients. CONCLUSIONS: SphK1 expression regulates the early stage of colon carcinogenesis and tumor growth, thus inhibition of SphK1 may be an effective strategy for colon cancer chemoprevention.


Subject(s)
Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Aged , Animals , Azoxymethane , Carcinogenesis/pathology , Cell Proliferation , Cyclooxygenase 2/metabolism , Enterocytes/metabolism , Enterocytes/pathology , Female , HT29 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasm Staging
7.
J Lipid Res ; 58(7): 1439-1452, 2017 07.
Article in English | MEDLINE | ID: mdl-28490444

ABSTRACT

Acute kidney injury (AKI), resulting from chemotherapeutic agents such as cisplatin, remains an obstacle in the treatment of cancer. Cisplatin-induced AKI involves apoptotic and necrotic cell death, pathways regulated by sphingolipids such as ceramide and glucosylceramide. Results from this study indicate that C57BL/6J mice treated with cisplatin had increased ceramide and hexosylceramide levels in the renal cortex 72 h following cisplatin treatment. Pretreatment of mice with inhibitors of acid sphingomyelinase and de novo ceramide synthesis (amitriptyline and myriocin, respectively) prevented accumulation of ceramides and hexosylceramide in the renal cortex and protected from cisplatin-induced AKI. To determine the role of ceramide metabolism to hexosylceramides in kidney injury, we treated mice with a potent and highly specific inhibitor of glucosylceramide synthase, the enzyme responsible for catalyzing the glycosylation of ceramides to form glucosylceramides. Inhibition of glucosylceramide synthase attenuated the accumulation of the hexosylceramides and exacerbated ceramide accumulation in the renal cortex following treatment of mice with cisplatin. Increasing ceramides and decreasing glucosylceramides in the renal cortex sensitized mice to cisplatin-induced AKI according to markers of kidney function, kidney injury, inflammation, cell stress, and apoptosis. Under conditions of high ceramide generation, data suggest that metabolism of ceramides to glucosylceramides buffers kidney ceramides and helps attenuate kidney injury.-Dupre, T. V., M. A. Doll, P. P. Shah, C. N. Sharp, D. Siow, J. Megyesi, J. Shayman, A. Bielawska, J. Bielawski, L. J. Beverly, M. Hernandez-Corbacho, C. J. Clarke, A. J. Snider, R. G. Schnellmann, L. M. Obeid, Y. A. Hannun, and L. J. Siskind. Inhibiting glucosylceramide synthase exacerbates cisplatin-induced acute kidney injury. J. Lipid Res 2017. 58: 1439-1452.


Subject(s)
Acute Kidney Injury/chemically induced , Cisplatin/adverse effects , Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Acute Kidney Injury/metabolism , Acute Kidney Injury/physiopathology , Animals , Ceramides/metabolism , Kidney Cortex/blood supply , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Male , Mice , Rats , Reperfusion Injury/metabolism
8.
Anticancer Res ; 37(3): 1213-1218, 2017 03.
Article in English | MEDLINE | ID: mdl-28314284

ABSTRACT

BACKGROUND/AIM: Because patients with cancer of apparently equivalent stage often have different outcomes, it is necessary to gather additional information to complement cancer staging. Dysregulated sphingolipid metabolism contributes to carcinogenesis. In this retrospective pilot study, we tested the hypothesis that changes in serum levels of sphingolipids are associated with stage IV colorectal cancer (CRC). PATIENTS AND METHODS: We used commercially available serum samples from healthy males and patients with CRC (adenocarcinoma of the large intestine, stage IV with metastases). Blood samples were obtained immediately prior to anesthesia/surgery. We measured sphingolipid levels in sera using mass spectrometry. RESULTS: In serum of patients with CRC, the levels of C16-, C18-, C18:1-, and C24:1-ceramide, as well as those of sphingosine, were significantly higher than those of controls. In contrast, the levels of C24-sphingomyelin were significantly lower than those of controls. A global test of association showed that ceramides and sphingomyelins but not hexosylceramides were significantly associated with stage IV CRC. CONCLUSION: Sphingolipids have a potential of serving as novel, non-invasive, inexpensive, and effective blood-based biomarkers to complement CRC staging for better prognosis and more personalized medicine.


Subject(s)
Ceramides/blood , Colorectal Neoplasms/blood , Sphingomyelins/blood , Sphingosine/blood , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism , Male , Middle Aged , Pilot Projects , Prognosis , Retrospective Studies
9.
Anticancer Res ; 37(2): 455-463, 2017 02.
Article in English | MEDLINE | ID: mdl-28179290

ABSTRACT

BACKGROUND/AIM: Combining an anticancer agent fenretinide (HPR) or C6-pyridinium ceramide (LCL29) with Foscan-mediated photodynamic therapy (FoscanPDT) is expected to augment anticancer benefits of each substance. We showed that treatment with FoscanPDT+HPR enhanced accumulation of C16-dihydroceramide, and that fumonisin B1 (FB), an inhibitor of ceramide synthase, counteracted caspase-3 activation and colony-forming ability of head and neck squamous cell carcinoma (HNSCC) cells. Because cancer cells appear to be more susceptible to increased levels of the endoplasmic reticulum (ER) stress than normal cells, herein we tested the hypothesis that FoscanPDT combined with HPR or LCL29 induces FB-sensitive ER stress-associated apoptosis that affects cell survival. MATERIALS AND METHODS: Using an HNSCC cell line, we determined: cell survival by clonogenic assay, caspase-3 activity by spectrofluorometry, the expression of the ER markers BiP and CHOP by quantitative real-time polymerase chain reaction and western immunoblotting, and sphingolipid levels by mass spectrometry. RESULTS: Similar to HPR+FoscanPDT, LCL29+FoscanPDT induced enhanced loss of clonogenicity and caspase-3 activation, that were both inhibited by FB. Our additional pharmacological evidence showed that the enhanced loss of clonogenicity after the combined treatments was singlet oxygen-, ER stress- and apoptosis-dependent. The combined treatments induced enhanced, FB-sensitive, up-regulation of BiP and CHOP, as well as enhanced accumulation of sphingolipids. CONCLUSION: Our data suggest that enhanced clonogenic cell killing after the combined treatments is dependent on oxidative- and ER-stress, apoptosis, and FB-sensitive sphingolipid production, and should help develop more effective mechanism-based therapeutic strategies.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Ceramides/pharmacology , Endoplasmic Reticulum Stress/drug effects , Fenretinide/pharmacology , Fumonisins/pharmacology , Head and Neck Neoplasms/drug therapy , Mesoporphyrins/pharmacology , Photochemotherapy/methods , Pyridinium Compounds/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Combined Modality Therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Radiation-Sensitizing Agents/pharmacology , Squamous Cell Carcinoma of Head and Neck
10.
Neuromolecular Med ; 19(1): 46-56, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27388466

ABSTRACT

Plasma deoxy-sphingoid bases are elevated in type 2 diabetes patients and correlate with the stage of diabetic distal sensorimotor polyneuropathy; however, associations between deoxy-sphingolipids (DSL) and neuropathy in type 1 diabetes have not been examined. The primary aim of this exploratory pilot study was to assess the associations between multiple sphingolipid species including DSL and free amino acids and the presence of symptomatic neuropathy in a DCCT/EDIC type 1 diabetes subcohort. Using mass spectroscopy, plasma levels of DSL and free amino acids in DCCT/EDIC type 1 diabetes participants (n = 80), with and without symptoms of neuropathy, were investigated. Patient-determined neuropathy was based on 15-item self-administered questionnaire (Michigan Neuropathy Screening Instrument) developed to assess distal symmetrical peripheral neuropathy in diabetes. Patients who scored ≥4, or reported inability to sense their feet during walking or to distinguish hot from cold water while bathing were considered neuropathic. Plasma levels of ceramide, sphingomyelin, hexosyl- and lactosylceramide species, and amino acids were measured and analyzed relative to neuropathy status in the patient. Deoxy-C24-ceramide, C24- and C26-ceramide were higher in patients with neuropathy than those without neuropathy. Cysteine was higher in patients with neuropathy. No differences in other sphingolipids or amino acids were detected. The covariate-adjusted Odds Ratios of positive patient-reported neuropathy was associated with increased levels of deoxy-C24-, and deoxy-C24:1-ceramide; C22-, C24-, and C26-ceramide; and cysteine. Plasma deoxy-ceramide and ceramide species may have potential diagnostic and prognostic significance in diabetic neuropathy.


Subject(s)
Ceramides/blood , Diabetes Mellitus, Type 1/blood , Diabetic Neuropathies/blood , Adult , Amino Acids/blood , Cross-Sectional Studies , Cysteine/blood , Diabetes Mellitus, Type 1/complications , Diabetic Neuropathies/epidemiology , Female , Follow-Up Studies , Glycosphingolipids , Humans , Male , Observational Studies as Topic , Odds Ratio , Pilot Projects , Sensation Disorders/blood , Sensation Disorders/epidemiology , Sensation Disorders/etiology , Sphingolipids/blood , Sphingomyelins/blood , Tandem Mass Spectrometry , Young Adult
11.
FASEB J ; 31(2): 771-780, 2017 02.
Article in English | MEDLINE | ID: mdl-27836988

ABSTRACT

The molecular mechanisms responsible for the development of proteinuria and glomerulosclerosis in radiation nephropathy remain largely unknown. Podocytes are increasingly recognized as key players in the pathogenesis of proteinuria in primary and secondary glomerular disorders. The lipid-modulating enzyme sphingomyelin phosphodiesterase acid-like 3B (SMPDL3b) is a key determinant of podocyte injury and a known off target of the anti-CD20 antibody rituximab (RTX). The current study investigates the role of sphingolipids in radiation-induced podocytopathy. After a single dose of radiation (8 Gy), several ceramide species were significantly elevated. In particular, C16:00, C24:00, and C24:1 ceramides were the most abundant ceramide species detected. These changes were paralleled by a time-dependent drop in SMPDL3b protein, sphingosine, and sphingosine-1-phosphate levels. Interestingly, SMPDL3b-overexpressing podocytes had higher basal levels of sphingosine-1-phosphate and maintained basal ceramide levels after irradiation. Morphologically, irradiated podocytes demonstrated loss of filopodia and remodeling of cortical actin. Furthermore, the actin binding protein ezrin relocated from the plasma membrane to the cytosol as early as 2 h after radiation. In contrast, SMPDL3b overexpressing podocytes were protected from radiation-induced cytoskeletal remodeling. Treatment with RTX before radiation exposure partially protected podocytes from SMPDL3b loss, cytoskeletal remodeling, and caspase 3 cleavage. Our results demonstrate that radiation injury induces early cytoskeletal remodeling, down-regulation of SMPDL3b, and elevation of cellular ceramide levels. Overexpression of SMPDL3b and pretreatment with RTX confer a radioprotective effect in cultured podocytes. These findings indicate a potential role for SMPDL3b and RTX in radiation-induced podocytopathy.-Ahmad, A., Mitrofanova, A., Bielawski, J., Yang, Y., Marples, B., Fornoni, A., Zeidan, Y. H. Sphingomyelinase-like phosphodiesterase 3b mediates radiation-induced damage of renal podocytes.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Podocytes/metabolism , Podocytes/radiation effects , Sphingomyelin Phosphodiesterase/metabolism , Animals , Ceramides/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Down-Regulation , Gene Expression Regulation, Enzymologic , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacology , Mice , Mice, Inbred C57BL , Rituximab/administration & dosage , Rituximab/pharmacology , Sphingomyelin Phosphodiesterase/genetics
12.
Oncotarget ; 7(51): 83907-83925, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27880732

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are immune suppressive cells that are hallmarks of human cancer. MDSCs inhibit cytotoxic T lymphocytes (CTLs) and NK cell functions to promote tumor immune escape and progression, and therefore are considered key targets in cancer immunotherapy. Recent studies determined a key role of the apoptosis pathways in tumor-induced MDSC homeostasis and it is known that ceramide plays a key role in regulation of mammalian cell apoptosis. In this study, we aimed to determine the efficacy and underlying molecular mechanism of ceramide in suppression of MDSCs. Treatment of tumor-bearing mice with LCL521, a lysosomotropic inhibitor of acid ceramidase, significantly decreased MDSC accumulation in vivo. Using a MDSC-like myeloid cell model, we determined that LCL521 targets lysosomes and increases total cellular C16 ceramide level. Although MDSC-like cells have functional apoptosis pathways, LCL521-induced MDSC death occurs in an apoptosis- and necroptosis-independent mechanism. LCL521 treatment resulted in an increase in the number of autophagic vesicles, heterolysosomes and swollen ERs. Finally, concomitant inhibition of cathepsin B and cathepsin D was required to significantly decrease LCL521-induced cell death. Our observations indicate that LCL521 targets lysosomes to activate cathepsin B and cathepsin D, resulting in interrupted autophagy and ER stress that culminates in MDSC death. Therefore, a ceramidase inhibitor is potentially an effective adjunct therapeutic agent for suppression of MDSCs to enhance the efficacy of CTL-based cancer immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cathepsin B/metabolism , Cathepsin D/metabolism , Ceramides/metabolism , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/pharmacology , Lysosomes/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Sarcoma/drug therapy , Signal Transduction/drug effects , Acid Ceramidase/antagonists & inhibitors , Acid Ceramidase/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation , Lysosomes/enzymology , Lysosomes/pathology , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/enzymology , Myeloid-Derived Suppressor Cells/pathology , Sarcoma/enzymology , Sarcoma/immunology , Sarcoma/pathology , Time Factors
13.
J Lipid Res ; 57(11): 2028-2039, 2016 11.
Article in English | MEDLINE | ID: mdl-27623848

ABSTRACT

Th1 pro-inflammatory cytokines, i.e., TNF-α and IFN-γ, in combination are known to induce cell death in several cell types, including oligodendrocytes, but the mechanism of their synergistic cytotoxicity is unclear. Although ceramide (Cer) has been implicated in cytokine- and stress-induced cell death, its intracellular levels alone cannot explain cytokine synergy. We considered the possibility that Cer released as part of extracellular vesicles may contribute to cytokine-induced synergistic cell death. Using a human oligodendroglioma (HOG) cell line as a model, here we show that exosomes derived from TNF-α-treated "donor" cells, while being mildly toxic to fresh cultures (similar to individual cytokines), induce enhanced cell death when added to IFN-γ-primed target cultures in a fashion resembling the effect of cytokine combination. Further, the sphingolipid profiles of secreted exosomes, as determined by HPLC-MS/MS, revealed that the treatment with the cytokines time-dependently induced the formation and exosomal release, in particular of C16-, C24-, and C24:1-Cer species; C16-, C24-, and C24:1-dihydroCer species; and C16-, C24-, and C24:1-SM species. Finally, exogenous C6-Cer or C16-Cer mimicked and enhanced the cytotoxic effects of the cytokines upon HOG cells, thereby supporting the cell death-signaling role of extracellular Cer.


Subject(s)
Ceramides/metabolism , Interferon-gamma/metabolism , Oligodendroglioma/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Death/genetics , Cell Line, Tumor , Ceramides/chemistry , Ceramides/genetics , Chromatography, High Pressure Liquid , Exosomes , Extracellular Vesicles/metabolism , Humans , Interferon-gamma/administration & dosage , Interferon-gamma/genetics , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligodendroglioma/pathology , Sphingolipids/chemistry , Sphingolipids/metabolism , Tandem Mass Spectrometry , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/genetics
14.
Proc Natl Acad Sci U S A ; 113(21): 5928-33, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27162368

ABSTRACT

Sphingolipids exhibit extreme functional and chemical diversity that is in part determined by their hydrophobic moiety, ceramide. In mammals, the fatty acyl chain length variation of ceramides is determined by six (dihydro)ceramide synthase (CerS) isoforms. Previously, we and others showed that mutations in the major neuron-specific CerS1, which synthesizes 18-carbon fatty acyl (C18) ceramide, cause elevation of long-chain base (LCB) substrates and decrease in C18 ceramide and derivatives in the brain, leading to neurodegeneration in mice and myoclonus epilepsy with dementia in humans. Whether LCB elevation or C18 ceramide reduction leads to neurodegeneration is unclear. Here, we ectopically expressed CerS2, a nonneuronal CerS producing C22-C24 ceramides, in neurons of Cers1-deficient mice. Surprisingly, the Cers1 mutant pathology was almost completely suppressed. Because CerS2 cannot replenish C18 ceramide, the rescue is likely a result of LCB reduction. Consistent with this hypothesis, we found that only LCBs, the substrates common for all of the CerS isoforms, but not ceramides and complex sphingolipids, were restored to the wild-type levels in the Cers2-rescued Cers1 mutant mouse brains. Furthermore, LCBs induced neurite fragmentation in cultured neurons at concentrations corresponding to the elevated levels in the CerS1-deficient brain. The strong association of LCB levels with neuronal survival both in vivo and in vitro suggests high-level accumulation of LCBs is a possible underlying cause of the CerS1 deficiency-induced neuronal death.


Subject(s)
Brain/metabolism , Ceramides , Gene Expression , Membrane Proteins/deficiency , Neurites , Neurodegenerative Diseases , Sphingosine N-Acyltransferase/biosynthesis , Sphingosine N-Acyltransferase/deficiency , Animals , Brain/pathology , Cell Survival , Ceramides/biosynthesis , Ceramides/genetics , Disease Models, Animal , Humans , Mice , Mice, Mutant Strains , Neurites/metabolism , Neurites/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Sphingolipids/biosynthesis , Sphingolipids/genetics , Sphingosine N-Acyltransferase/genetics
15.
JCI Insight ; 1(5)2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27158676

ABSTRACT

We recently discovered that endothelial Nogo-B, a membrane protein of the ER, regulates vascular function by inhibiting the rate-limiting enzyme, serine palmitoyltransferase (SPT), in de novo sphingolipid biosynthesis. Here, we show that endothelium-derived sphingolipids, particularly sphingosine-1-phosphate (S1P), protect the heart from inflammation, fibrosis, and dysfunction following pressure overload and that Nogo-B regulates this paracrine process. SPT activity is upregulated in banded hearts in vivo as well as in TNF-α-activated endothelium in vitro, and loss of Nogo removes the brake on SPT, increasing local S1P production. Hence, mice lacking Nogo-B, systemically or specifically in the endothelium, are resistant to the onset of pathological cardiac hypertrophy. Furthermore, pharmacological inhibition of SPT with myriocin restores permeability, inflammation, and heart dysfunction in Nogo-A/B-deficient mice to WT levels, whereas SEW2871, an S1P1 receptor agonist, prevents myocardial permeability, inflammation, and dysfunction in WT banded mice. Our study identifies a critical role of endothelial sphingolipid biosynthesis and its regulation by Nogo-B in the development of pathological cardiac hypertrophy and proposes a potential therapeutic target for the attenuation or reversal of this clinical condition.

16.
J Photochem Photobiol B ; 159: 191-5, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27085050

ABSTRACT

We and others have shown that stresses, including photodynamic therapy (PDT), can disrupt the de novo sphingolipid biosynthesis pathway, leading to changes in the levels of sphingolipids, and subsequently, modulation of cell death. The de novo sphingolipid biosynthesis pathway includes a ceramide synthase-dependent reaction, giving rise to dihydroceramide, which is then converted in a desaturase-dependent reaction to ceramide. In this study we tested the hypothesis that combining Foscan-mediated PDT with desaturase inhibitor fenretinide (HPR) enhances cancer cell killing. We discovered that by subjecting SCC19 cells, a human head and neck squamous cell carcinoma cell line, to PDT+HPR resulted in enhanced accumulation of C16-dihydroceramide, not ceramide. Concomitantly, mitochondrial depolarization was enhanced by the combined treatment. Enhanced activation of caspase-3 after PDT+HPR was inhibited by FB. Enhanced clonogenic cell death after the combination was sensitive to FB, as well as Bcl2- and caspase inhibitors. Treatment of mouse SCCVII squamous cell carcinoma tumors with PDT+HPR resulted in improved long-term tumor cures. Overall, our data showed that combining PDT with HPR enhanced apoptotic cancer cell killing and antitumor efficacy of PDT. The data suggest the involvement of the de novo sphingolipid biosynthesis pathway in enhanced apoptotic cell killing after PDT+HPR, and identify the combination as a novel more effective anticancer treatment than either treatment alone.


Subject(s)
Apoptosis , Fenretinide/therapeutic use , Mesoporphyrins/therapeutic use , Photochemotherapy , Photosensitizing Agents/therapeutic use , Sphingolipids/biosynthesis , Cell Line, Tumor , Drug Therapy, Combination , Fenretinide/administration & dosage , Humans , Mesoporphyrins/administration & dosage , Photosensitizing Agents/administration & dosage
17.
BMC Cancer ; 15: 762, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26493335

ABSTRACT

BACKGROUND: Resistance to chemotherapy is common in gastroesophageal cancer. Mechanisms of resistance are incompletely characterised and there are no predictive biomarkers in clinical practice for cytotoxic drugs. We used new cell line models to characterise novel chemotherapy resistance mechanisms and validated them in tumour specimens to identify new targets and biomarkers for gastroesophageal cancer. METHODS: Cell lines were selected for resistance to oxaliplatin, cisplatin and docetaxel and gene expression examined using Affymetrix Exon 1.0 ST arrays. Leads were validated by qRT-PCR and HPLC of tumour metabolites. Protein expression and pharmacological inhibition of lead target SPHK1 was evaluated in independent cell lines, and by immunohistochemistry in gastroesophageal cancer patients. RESULTS: Genes with differential expression in drug resistant cell lines compared to the parental cell line they were derived from, were identified for each drug resistant cell line. Biological pathway analysis of these gene lists, identified over-represented pathways, and only 3 pathways - lysosome, sphingolipid metabolism and p53 signalling- were identified as over-represented in these lists for all three cytotoxic drugs investigated. The majority of genes differentially expressed in chemoresistant cell lines from these pathways, were involved in metabolism of glycosphingolipids and sphingolipids in lysosomal compartments suggesting that sphingolipids might be important mediators of cytotoxic drug resistance in gastroeosphageal cancers . On further investigation, we found that drug resistance (IC50) was correlated with increased sphingosine kinase 1(SPHK1) mRNA and also with decreased sphingosine-1-phosphate lysase 1(SGPL1) mRNA. SPHK1 and SGPL1 gene expression were inversely correlated. SPHK1:SGPL1 ratio correlated with increased cellular sphingosine-1-phosphate (S1P), and S1P correlated with drug resistance (IC50). High SPHK1 protein correlated with resistance to cisplatin (IC50) in an independent gastric cancer cell line panel and with survival of patients treated with chemotherapy prior to surgery but not in patients treated with surgery alone. Safingol a SPHK1 inhibitor, was cytotoxic as a single agent and acted synergistically with cisplatin in gastric cancer cell lines. CONCLUSION: Agents that inhibit SPHK1 or S1P could overcome cytotoxic drug resistance in gastroesophageal cancer. There are several agents in early phase human trials including Safingol that could be combined with chemotherapy or used in patients progressing after chemotherapy.


Subject(s)
Aldehyde-Lyases/genetics , Drug Resistance, Neoplasm/genetics , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lysophospholipids/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sphingosine/analogs & derivatives , Stomach Neoplasms/genetics , Aldehyde-Lyases/biosynthesis , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Lysophospholipids/biosynthesis , Male , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , RNA, Neoplasm/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , Sphingosine/biosynthesis , Sphingosine/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism
18.
Mol Biol Cell ; 26(24): 4451-65, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26446842

ABSTRACT

Cilia are important organelles formed by cell membrane protrusions; however, little is known about their regulation by membrane lipids. We characterize a novel activation mechanism for glycogen synthase kinase-3 (GSK3) by the sphingolipids phytoceramide and ceramide that is critical for ciliogenesis in Chlamydomonas and murine ependymal cells, respectively. We show for the first time that Chlamydomonas expresses serine palmitoyl transferase (SPT), the first enzyme in (phyto)ceramide biosynthesis. Inhibition of SPT in Chlamydomonas by myriocin led to loss of flagella and reduced tubulin acetylation, which was prevented by supplementation with the precursor dihydrosphingosine. Immunocytochemistry showed that (phyto)ceramide was colocalized with phospho-Tyr-216-GSK3 (pYGSK3) at the base and tip of Chlamydomonas flagella and motile cilia in ependymal cells. The (phyto)ceramide distribution was consistent with that of a bifunctional ceramide analogue UV cross-linked and visualized by click-chemistry-mediated fluorescent labeling. Ceramide depletion, by myriocin or neutral sphingomyelinase deficiency (fro/fro mouse), led to GSK3 dephosphorylation and defective flagella and cilia. Motile cilia were rescued and pYGSK3 localization restored by incubation of fro/fro ependymal cells with exogenous C24:1 ceramide, which directly bound to pYGSK3. Our findings suggest that (phyto)ceramide-mediated translocation of pYGSK into flagella and cilia is an evolutionarily conserved mechanism fundamental to the regulation of ciliogenesis.


Subject(s)
Ceramides/metabolism , Chlamydomonas/metabolism , Cilia/metabolism , Flagella/metabolism , Glycogen Synthase Kinase 3/metabolism , Acetylation , Animals , Cells, Cultured , Chlamydomonas/enzymology , Cilia/enzymology , Ependyma/cytology , Ependyma/metabolism , Flagella/enzymology , Humans , Mice , Mice, Inbred C57BL , Serine C-Palmitoyltransferase/metabolism , Tubulin/metabolism
19.
Proc Natl Acad Sci U S A ; 112(42): 12962-7, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26438849

ABSTRACT

Sphingolipids typically have an 18-carbon (C18) sphingoid long chain base (LCB) backbone. Although sphingolipids with LCBs of other chain lengths have been identified, the functional significance of these low-abundance sphingolipids is unknown. The LCB chain length is determined by serine palmitoyltransferase (SPT) isoenzymes, which are trimeric proteins composed of two large subunits (SPTLC1 and SPTLC2 or SPTLC3) and a small subunit (SPTssa or SPTssb). Here we report the identification of an Sptssb mutation, Stellar (Stl), which increased the SPT affinity toward the C18 fatty acyl-CoA substrate by twofold and significantly elevated 20-carbon (C20) LCB production in the mutant mouse brain and eye, resulting in surprising neurodegenerative effects including aberrant membrane structures, accumulation of ubiquitinated proteins on membranes, and axon degeneration. Our work demonstrates that SPT small subunits play a major role in controlling SPT activity and substrate affinity, and in specifying sphingolipid LCB chain length in vivo. Moreover, our studies also suggest that excessive C20 LCBs or C20 LCB-containing sphingolipids impair protein homeostasis and neural functions.


Subject(s)
Carbon/chemistry , Mutation , Neurodegenerative Diseases/enzymology , Serine C-Palmitoyltransferase/chemistry , Amino Acid Sequence , Animals , Humans , Mice , Molecular Sequence Data , Neurodegenerative Diseases/genetics , Sequence Homology, Amino Acid , Serine C-Palmitoyltransferase/genetics , Ubiquitination
20.
Oncotarget ; 6(27): 24246-60, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26327294

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent for several human cancers including primary effusion lymphoma (PEL), a rapidly progressive malignancy arising preferentially in immunocompromised patients. With conventional chemotherapy, PEL continues to portend high mortality, dictating the development of novel therapeutic strategies. Sphingosine kinase 2 (SphK2) represents a key gatekeeper for sphingolipid metabolism, responsible for conversion of ceramides to sphingosine-1-phosphate (S1P). We have previously demonstrated that targeting SphK2 using a novel selective inhibitor, ABC294640, leads to intracellular accumulation of ceramides and induces apoptosis for KSHV-infected PEL cells, while suppressing tumor progression in vivo. In the current study, we sought to determine whether specific ceramide/dh-ceramide species and related ceramide synthases (CerS) impact viability for KSHV-infected PEL cells during targeting of SphK2. We found that several specific ceramide and dihydro(dh)-ceramide species and their associated CerS reduce PEL survival and tumor expansion in vitro and in vivo. Moreover, we found that dhC16-Cer induces PEL apoptosis in part through activation of KSHV lytic gene expression. These data further implicate bioactive sphingolipids in regulation of PEL survival, and provide justification for future studies evaluating clinically relevant ceramide analogs or mimetics for their potential as therapeutic agents for PEL.


Subject(s)
Apoptosis , Ceramides/metabolism , Gene Expression Regulation, Viral , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/virology , Oxidoreductases/metabolism , Adamantane/analogs & derivatives , Adamantane/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pyridines/chemistry , RNA Interference , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...