Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
1.
J Biol Chem ; 300(3): 105728, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38325740

ABSTRACT

Serine palmitoyltransferase (SPT) catalyzes the pyridoxal-5'-phosphate (PLP)-dependent decarboxylative condensation of l-serine and palmitoyl-CoA to form 3-ketodihydrosphingosine (KDS). Although SPT was shown to synthesize corresponding products from amino acids other than l-serine, it is still arguable whether SPT catalyzes the reaction with d-serine, which is a question of biological importance. Using high substrate and enzyme concentrations, KDS was detected after the incubation of SPT from Sphingobacterium multivorum with d-serine and palmitoyl-CoA. Furthermore, the KDS comprised equal amounts of 2S and 2R isomers. 1H-NMR study showed a slow hydrogen-deuterium exchange at Cα of serine mediated by SPT. We further confirmed that SPT catalyzed the racemization of serine. The rate of the KDS formation from d-serine was comparable to those for the α-hydrogen exchange and the racemization reaction. The structure of the d-serine-soaked crystal (1.65 Å resolution) showed a distinct electron density of the PLP-l-serine aldimine, interpreted as the racemized product trapped in the active site. The structure of the α-methyl-d-serine-soaked crystal (1.70 Å resolution) showed the PLP-α-methyl-d-serine aldimine, mimicking the d-serine-SPT complex prior to racemization. Based on these enzymological and structural analyses, the synthesis of KDS from d-serine was explained as the result of the slow racemization to l-serine, followed by the reaction with palmitoyl-CoA, and SPT would not catalyze the direct condensation between d-serine and palmitoyl-CoA. It was also shown that the S. multivorum SPT catalyzed the racemization of the product KDS, which would explain the presence of (2R)-KDS in the reaction products.


Subject(s)
Serine C-Palmitoyltransferase , Serine , Sphingobacterium , Catalytic Domain , Crystallization , Deuterium Exchange Measurement , Electrons , Hydrogen/metabolism , Palmitoyl Coenzyme A/metabolism , Serine/analogs & derivatives , Serine/metabolism , Serine C-Palmitoyltransferase/chemistry , Serine C-Palmitoyltransferase/metabolism , Sphingobacterium/enzymology , Sphingobacterium/metabolism , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis , Sphingosine/metabolism , Stereoisomerism , Substrate Specificity
2.
J Biol Chem ; 299(9): 105126, 2023 09.
Article in English | MEDLINE | ID: mdl-37543362

ABSTRACT

Oxidative stress triggered by aging, radiation, or inflammation impairs ovarian function by inducing granulosa cell (GC) apoptosis. However, the mechanism inducing GC apoptosis has not been characterized. Here, we found that ovarian GCs from aging patients showed increased oxidative stress, enhanced reactive oxygen species activity, and significantly decreased expression of the known antiapoptotic factor sphingosine-1-phosphate/sphingosine kinase 1 (SPHK1) in GCs. Interestingly, the expression of Krüppel-like factor 12 (KLF12) was significantly increased in the ovarian GCs of aging patients. Furthermore, we determined that KLF12 was significantly upregulated in hydrogen peroxide-treated GCs and a 3-nitropropionic acid-induced in vivo model of ovarian oxidative stress. This phenotype was further confirmed to result from inhibition of SPHK1 by KLF12. Interestingly, when endogenous KLF12 was knocked down, it rescued oxidative stress-induced apoptosis. Meanwhile, supplementation with SPHK1 partially reversed oxidative stress-induced apoptosis. However, this function was lost in SPHK1 with deletion of the binding region to the KLF12 promoter. SPHK1 reversed apoptosis caused by hydrogen peroxide-KLF12 overexpression, a result further confirmed in an in vitro ovarian culture model and an in vivo 3-nitropropionic acid-induced ovarian oxidative stress model. Overall, our study reveals that KLF12 is involved in regulating apoptosis induced by oxidative stress in aging ovarian GCs and that sphingosine-1-phosphate/SPHK1 can rescue GC apoptosis by interacting with KLF12 in negative feedback.


Subject(s)
Aging , Apoptosis , Granulosa Cells , Hydrogen Peroxide , Kruppel-Like Transcription Factors , Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor) , Sphingosine , Female , Humans , Aging/metabolism , Feedback, Physiological , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Hydrogen Peroxide/pharmacology , In Vitro Techniques , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Lysophospholipids/biosynthesis , Lysophospholipids/metabolism , Organ Culture Techniques , Oxidative Stress/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Promoter Regions, Genetic , Sphingosine/biosynthesis , Sphingosine/metabolism , Reactive Oxygen Species/metabolism
3.
J Appl Microbiol ; 130(6): 1981-1992, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33190388

ABSTRACT

AIMS: To genetically engineer the oleaginous yeast Yarrowia lipolytica for de novo production of tetraacetylphytosphingosine (TAPS), a precursor of phytosphingosine, and optimization of fermentation conditions for high yield. METHODS AND RESULTS: We successfully constructed a TAPS-producing Y. lipolytica CE3 strain by co-expression of Wickerhamomyces ciferrii-derived acetyl transferases, Sli1p and Atf2p. Next, we optimized several environmental factors including temperature, initial pH and C/N ratio for TAPS production in a shake culture. Deletion of LCB4 in CE3 strain increased the volumetric TAPS titre and cell-specific yield to 142·1 ± 10·7 mgTAPS  l-1 and 3·08 ± 0·11 mgTAPS  gDCW -1 , respectively, in a shake flask culture incubated for 120 h at 28°C with glycerol as the carbon source. Finally, we developed a 5-l fed-batch process with NaOH-mediated pH control and olive oil as a carbon source, exhibiting 650 ± 24 mgTAPS  l-1 of TAPS production within 56 h of the fermentation. CONCLUSIONS: The introduction of codon-optimized Sli1p and Atf2p, deletion of LCB4 gene and sexual hybridization, accompanied by specific fermentation conditions, enhanced TAPS yield in Y. lipolytica. SIGNIFICANCE AND IMPACT OF THE STUDY: Our results highlight Y. lipolytica as a promising candidate for the industrial production of TAPS, an important component of cosmetic formulations.


Subject(s)
Sphingosine/analogs & derivatives , Yarrowia/genetics , Yarrowia/metabolism , Batch Cell Culture Techniques , Fermentation , Fungal Proteins/genetics , Fungal Proteins/metabolism , Industrial Microbiology , Metabolic Engineering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomycetales/enzymology , Saccharomycetales/genetics , Sphingosine/analysis , Sphingosine/biosynthesis
4.
J Cell Mol Med ; 24(18): 10290-10301, 2020 09.
Article in English | MEDLINE | ID: mdl-32803879

ABSTRACT

There are many different types of cardiovascular diseases, which impose a huge economic burden due to their extremely high mortality rates, so it is necessary to explore the underlying mechanisms to achieve better supportive and curative care outcomes. Sphingosine 1-phosphate (S1P) is a bioactive lipid mediator with paracrine and autocrine activities that acts through its cell surface S1P receptors (S1PRs) and intracellular signals. In the circulatory system, S1P is indispensable for both normal and disease conditions; however, there are very different views on its diverse roles, and its specific relevance to cardiovascular pathogenesis remains elusive. Here, we review the synthesis, release and functions of S1P, specifically detail the roles of S1P and S1PRs in some common cardiovascular diseases, and then address several controversial points, finally, we focus on the development of S1P-based therapeutic approaches in cardiovascular diseases, such as the selective S1PR1 modulator amiselimod (MT-1303) and the non-selective S1PR1 and S1PR3 agonist fingolimod, which may provide valuable insights into potential therapeutic strategies for cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Lysophospholipids/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine/analogs & derivatives , Animals , Humans , Lysophospholipids/biosynthesis , Models, Biological , Signal Transduction , Sphingosine/biosynthesis , Sphingosine/metabolism
5.
Handb Exp Pharmacol ; 259: 49-76, 2020.
Article in English | MEDLINE | ID: mdl-29460151

ABSTRACT

There is substantial evidence that the enzymes, sphingosine kinase 1 and 2, which catalyse the formation of the bioactive lipid sphingosine 1-phosphate, are involved in pathophysiological processes. In this chapter, we appraise the evidence that both enzymes are druggable and describe how isoform-specific inhibitors can be developed based on the plasticity of the sphingosine-binding site. This is contextualised with the effect of sphingosine kinase inhibitors in cancer, pulmonary hypertension, neurodegeneration, inflammation and sickling.


Subject(s)
Enzyme Inhibitors/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Anemia, Sickle Cell , Binding Sites , Humans , Hypertension, Pulmonary , Inflammation , Lysophospholipids/biosynthesis , Neoplasms , Neurodegenerative Diseases , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis
6.
Biochem Biophys Res Commun ; 516(3): 934-940, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31277939

ABSTRACT

This study shows the effects of tamoxifen, a known estrogen receptor antagonist used in the treatment of breast cancer, on the sphingolipid pathway of Trypanosoma cruzi, searching for potential chemotherapeutic targets. A dose-dependent epimastigote growth inhibition at increasing concentration of tamoxifen was determined. In blood trypomastigotes, treatment with 10 µM showed 90% lysis, while 86% inhibition of intracellular amastigote development was obtained using 50 µM. Lipid extracts from treated and non-treated metabolically labelled epimastigotes evidenced by thin layer chromatography different levels of sphingolipids and MALDI-TOF mass spectrometry analysis assured the identity of the labelled species. Comparison by HPLC-ESI mass spectrometry of lipids, notably exhibited a dramatic increase in the level of ceramide in tamoxifen-treated parasites and a restrained increase of ceramide-1P and sphingosine, indicating that the drug is acting on the enzymes involved in the final breakdown of ceramide. The ultrastructural analysis of treated parasites revealed characteristic morphology of cells undergoing an apoptotic-like death process. Flow cytometry confirmed cell death by an apoptotic-like machinery indicating that tamoxifen triggers this process by acting on the parasitic sphingolipid pathway.


Subject(s)
Antiprotozoal Agents/pharmacology , Life Cycle Stages/drug effects , Lipid Metabolism/drug effects , Sphingolipids/antagonists & inhibitors , Tamoxifen/pharmacology , Trypanosoma cruzi/drug effects , Animals , Apoptosis/drug effects , Ceramides/antagonists & inhibitors , Ceramides/biosynthesis , Chagas Disease/drug therapy , Chagas Disease/parasitology , Disease Models, Animal , Drug Repositioning , Estrogen Antagonists/pharmacology , Mice , Mice, Inbred BALB C , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Sphingolipids/biosynthesis , Sphingosine/antagonists & inhibitors , Sphingosine/biosynthesis , Trypanosoma cruzi/growth & development , Trypanosoma cruzi/metabolism
7.
IUBMB Life ; 71(9): 1284-1292, 2019 09.
Article in English | MEDLINE | ID: mdl-30889317

ABSTRACT

This study was designed to explore whether exosomal sphingosine 1-phosphate (S1P) from mesenchymal stem cells (MSCs) regulate the Treg/Th17 balance in aplastic anemia (AA) patients and to validate the underlying mechanism. To address this, exosomes from human bone marrow MSCs (MSCs-Exos) were co-cultured with CD4+ T cells from AA patients (AA CD4+ T cells), which were transfected with si-S1PR1, si-S1PR3, or not. The proportion of Th17 and Treg was evaluated by flow cytometry. The levels of Th17-associated interleukin-17 (IL-17), Treg-associated IL-10, and transforming growth factor-ß were determined by ELISA. S1P content in MSCs-Exos isolated from control, si-SphK1, or si-SphK2 transfected MSCs was examined by LC-MS/MS. Hematoxylin and eosin staining of bone marrow tissues was performed to evaluate the effect of MSCs-Exos in AA mice. Our results showed that MSCs-Exos reversed the increased Th17/Treg in AA through SphK1-mediated exosomal S1P enrichment. Furthermore, the promotion of Treg differentiation by exosomal S1P from MSCs was mediated through the receptor S1PR1 expressed on CD4+ T cells. Further in vivo experiments showed that MSCs-Exos reversed the increased Th17/Treg and alleviated AA progression in AA mice. In summary, SphK1-mediated enrichment of exosomal S1P secreted by MSCs reversed the increased Treg/Th17 ratio via the receptor S1PR1 in AA patients. © 2019 IUBMB Life, 71(9):1284-1292, 2019.


Subject(s)
Anemia, Aplastic/immunology , Lysophospholipids/immunology , Sphingosine/analogs & derivatives , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Anemia, Aplastic/genetics , Anemia, Aplastic/pathology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Chromatography, Liquid , Coculture Techniques , Exosomes/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lysophospholipids/biosynthesis , Lysophospholipids/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Sphingosine/biosynthesis , Sphingosine/immunology , Sphingosine/metabolism , Tandem Mass Spectrometry , Transforming Growth Factor beta/genetics
8.
Acta Derm Venereol ; 99(6): 594-601, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30834454

ABSTRACT

Sphingosine-1-phosphate (S1P) is a signalling sphingolipid metabolite that regulates important cell processes, including cell proliferation and apoptosis. Circulating S1P levels have been reported to be increased in patients with psoriasis relative to healthy patients. The aim of this study was to examine the potency of S1P inhibition using an imiquimod-induced psoriasis mouse model. Both topical ceramidase and sphingosine kinase 1/2 inhibition, which blocks S1P generation, alleviated imiquimod-induced skin lesions and reduced the serum interleukin 17-A levels induced by application of imiquimod. These treatments also normalized skin mRNA levels of genes associated with inflammation and keratinocyte differentiation. Inhibition of sphingosine kinase 2, but not sphingosine kinase 1, diminished levels of suppressor of cytokine signalling 1 and blocked T helper type 17 differentiation of naïve CD4+ T cells; imiquimod-induced psoriasis-like skin symptoms were also ameliorated. These results indicate the distinct effects of sphingosine kinase 1 and sphingosine kinase 2 inhibition on T helper type 17 generation and suggest molecules that inhibit S1P formation, including ceramidase and sphingosine kinase 2 inhibitors, as novel therapeutic candidates for psoriasis.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Enzyme Inhibitors/pharmacology , Lysophospholipids/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Psoriasis/drug therapy , Sphingosine/analogs & derivatives , Administration, Topical , Animals , Cell Differentiation/drug effects , Ceramidases/antagonists & inhibitors , Disease Models, Animal , Gene Expression/drug effects , Imiquimod , Immunity/drug effects , Inflammation/genetics , Interleukin-17/blood , Male , Mice , Psoriasis/chemically induced , Psoriasis/pathology , Quinolones/pharmacology , RNA, Messenger/metabolism , Sphingosine/biosynthesis , Suppressor of Cytokine Signaling 1 Protein , Th17 Cells
9.
Int J Mol Sci ; 20(20)2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31614447

ABSTRACT

Ceramide and sphingosine display a unique profile during brain development, indicating their critical role in myelinogenesis. Employing advanced technology such as gas chromatography-mass spectrometry, high performance liquid chromatography, and immunocytochemistry, along with cell culture and molecular biology, we have found an accumulation of sphingosine in brain tissues of patients with multiple sclerosis (MS) and in the spinal cord of rats induced with experimental autoimmune encephalomyelitis. The elevated sphingosine leads to oligodendrocyte death and fosters demyelination. Ceramide elevation by serine palmitoyltransferse (SPT) activation was the primary source of the sphingosine elevation as myriocin, an inhibitor of SPT, prevented sphingosine elevation and protected oligodendrocytes. Supporting this view, fingolimod, a drug used for MS therapy, reduced ceramide generation, thus offering partial protection to oligodendrocytes. Sphingolipid synthesis and degradation in normal development is regulated by a series of microRNAs (miRNAs), and hence, accumulation of sphingosine in MS may be prevented by employing miRNA technology. This review will discuss the current knowledge of ceramide and sphingosine metabolism (synthesis and breakdown), and how their biosynthesis can be regulated by miRNA, which can be used as a therapeutic approach for MS.


Subject(s)
Ceramides/biosynthesis , MicroRNAs/genetics , Multiple Sclerosis/genetics , Serine C-Palmitoyltransferase/metabolism , Sphingosine/biosynthesis , Animals , Brain/metabolism , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Humans , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Rats , Serine C-Palmitoyltransferase/antagonists & inhibitors
10.
Int J Mol Sci ; 20(15)2019 Jul 26.
Article in English | MEDLINE | ID: mdl-31357484

ABSTRACT

Sphingosine 1-phosphate (S1P) is a potent lipid mediator that modulates inflammation and angiogenesis. In this study, we investigated the possible involvement of S1P in the pathology of light-induced retinal degeneration in vivo and in vitro. The intracellular S1P and sphingosine kinase (SphK) activity in a photoreceptor cell line (661W cells) was significantly increased by exposure to light. The enhancement of SphK1 expression was dependent on illumination, and all-trans-retinal significantly promoted SphK1 expression. S1P treatment reduced protein kinase B (Akt) phosphorylation and increased the protein expression of cleaved caspase-3, and induced photoreceptor cell apoptosis. In vivo, light exposure enhanced the expression of SphK1 in the outer segments of photoreceptors. Intravitreal injection of a SphK inhibitor significantly suppressed the thinning of the outer nuclear layer and ameliorated the attenuation of the amplitudes of a-waves and b-waves of electroretinograms during light-induced retinal degeneration. These findings imply that light exposure induces the synthesis of S1P in photoreceptors by upregulating SphK1, which is facilitated by all-trans-retinal, causing retinal degeneration. Inhibition of this enhancement may be a therapeutic target of outer retinal degeneration, including age-related macular degeneration.


Subject(s)
Light , Lysophospholipids/biosynthesis , Photoreceptor Cells/metabolism , Photoreceptor Cells/radiation effects , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Sphingosine/analogs & derivatives , Stress, Physiological/radiation effects , Animals , Apoptosis , Cell Line , Disease Models, Animal , Disease Susceptibility , Electroretinography , Humans , Light/adverse effects , Macular Degeneration/etiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Photoreceptor Cells/pathology , Retina/metabolism , Retina/pathology , Retina/radiation effects , Retinal Degeneration/diagnostic imaging , Retinal Degeneration/pathology , Sphingosine/biosynthesis , Tomography, Optical Coherence
11.
J Lipid Res ; 59(3): 462-474, 2018 03.
Article in English | MEDLINE | ID: mdl-29326159

ABSTRACT

Sphingosine kinase 1 (SK1) is required for production of sphingosine-1-phosphate (S1P) and thereby regulates many cellular processes, including cellular growth, immune cell trafficking, and inflammation. To produce S1P, SK1 must access sphingosine directly from membranes. However, the molecular mechanisms underlying SK1's direct membrane interactions remain unclear. We used hydrogen/deuterium exchange MS to study interactions of SK1 with membrane vesicles. Using the CRISPR/Cas9 technique to generate HCT116 cells lacking SK1, we explored the effects of membrane interface disruption and the function of the SK1 interaction site. Disrupting the interface resulted in reduced membrane association and decreased cellular SK1 activity. Moreover, SK1-dependent signaling, including cell invasion and endocytosis, was abolished upon mutation of the membrane-binding interface. Of note, we identified a positively charged motif on SK1 that is responsible for electrostatic interactions with membranes. Furthermore, we demonstrated that SK1 uses a single contiguous interface, consisting of an electrostatic site and a hydrophobic site, to interact with membrane-associated anionic phospholipids. Altogether, these results define a composite domain in SK1 that regulates its intrinsic ability to bind membranes and indicate that this binding is critical for proper SK1 function. This work will allow for a new line of thinking for targeting SK1 in disease.


Subject(s)
Lipids/chemistry , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/analogs & derivatives , Binding Sites , Cell Membrane/metabolism , Deuterium Exchange Measurement , HCT116 Cells , Humans , Lysophospholipids/biosynthesis , Mass Spectrometry , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Signal Transduction , Sphingosine/biosynthesis , Sphingosine/metabolism
12.
J Lipid Res ; 58(4): 636-648, 2017 04.
Article in English | MEDLINE | ID: mdl-28188148

ABSTRACT

The pathophysiology of human keratoconus (KC), a bilateral progressive corneal disease leading to protrusion of the cornea, stromal thinning, and scarring, is not well-understood. In this study, we investigated a novel sphingolipid (SPL) signaling pathway through which KC may be regulated. Using human corneal fibroblasts (HCFs) and human KC cells (HKCs), we examined the SPL pathway modulation. Both cell types were stimulated by the three transforming growth factor (TGF)-ß isoforms: TGF-ß1 (T1), TGF-ß2 (T2), and TGF-ß3 (T3). All samples were analyzed using lipidomics and real-time PCR. Our data showed that HKCs have increased levels of signaling SPLs, ceramide (Cer), and sphingosine 1-phosphate (S1P). Treatment with T1 reversed the increase in Cer in HKCs and treatment with T3 reversed the increase in S1P. S1P3 receptor mRNA levels were also significantly upregulated in HKCs, but were reduced to normal levels following T3 treatment. Furthermore, stimulation with Cer and S1P led to significant upregulation of fibrotic markers in HCFs, but not in HKCs. Additionally, stimulation with a Cer synthesis inhibitor (FTY720) led to significant downregulation of specific fibrotic markers in HKCs (TGF-ß1, collagen type III, and α smooth muscle actin) without an effect on healthy HCFs, suggesting a causative role of Cer and S1P in fibrogenesis. Overall, this study suggests an association of the SPL signaling pathway in KC disease and its relation with the TGF-ß pathway.


Subject(s)
Ceramides/genetics , Keratoconus/genetics , Lysophospholipids/biosynthesis , Sphingolipids/genetics , Sphingosine/analogs & derivatives , Cell Line , Ceramides/administration & dosage , Cornea/metabolism , Cornea/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/physiology , Fingolimod Hydrochloride/administration & dosage , Humans , Keratoconus/pathology , Lysophospholipids/administration & dosage , RNA, Messenger/genetics , Signal Transduction , Sphingolipids/isolation & purification , Sphingolipids/metabolism , Sphingosine/administration & dosage , Sphingosine/biosynthesis , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta2/administration & dosage , Transforming Growth Factor beta3/administration & dosage
13.
Biochim Biophys Acta ; 1854(10 Pt B): 1718-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25936777

ABSTRACT

Fam20C is an atypical kinase implicated in bio-mineralization and phosphate homeostasis disorders, and has recently been shown to account for the activity of an orphan enzyme ("genuine casein kinase", G-CK) previously characterized for its ability to phosphorylate casein and a plethora of secreted proteins at serine residues specified by the S-x-E/pS motif. Fam20C/G-CK activity is only appreciable in the presence of high Mn2+ concentration (>1 mM), and is negligible if Mn2+ is replaced by physiological Mg2+ concentrations. Here we show that sphingosine (but not its biological precursor ceramide) not only stimulates several-fold Fam20C activity in the presence of Mn2+, but also confers a comparable activity to Fam20C assayed with Mg2+. Activation by sphingosine is evident using a variety of substrates, and is accounted for by both higher Vmax and decreased KmATP, as judged from kinetics run with the ß(28-40) substrate peptide and a physiological substrate, BMP-15. Sphingosine also protects Fam20C from thermal inactivation. Consistent with the in vitro results, by treating Fam20C expressing HEK293T cells with myriocin, a potent inhibitor of the sphingosine biosynthetic pathway, the activity of Fam20C released into the conditioned medium is substantially decreased corroborating the concept that sphingosine (or related metabolite(s)) is a co-factor required by Fam20C to optimally display its biological functions. None of the small molecule kinase inhibitors tested so far were able to inhibit Fam20C. Interestingly however fingolimod, an immunosuppressive drug structurally related to sphingosine, used for the treatment of multiple sclerosis, is a powerful activator of Fam20C, both wild type and its pathogenic, loss of function, T268M mutant. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.


Subject(s)
Casein Kinase I/genetics , Extracellular Matrix Proteins/genetics , Multiple Sclerosis/genetics , Sphingosine/biosynthesis , Amino Acid Sequence , Casein Kinase I/chemistry , Extracellular Matrix Proteins/chemistry , Fatty Acids, Monounsaturated/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Multiple Sclerosis/enzymology , Phosphorylation , Sphingosine/antagonists & inhibitors , Sphingosine/metabolism , Transcriptional Activation/drug effects
14.
Development ; 140(1): 136-46, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23154413

ABSTRACT

Duchenne muscular dystrophy is a lethal genetic disease characterized by the loss of muscle integrity and function over time. Using Drosophila, we show that dystrophic muscle phenotypes can be significantly suppressed by a reduction of wunen, a homolog of lipid phosphate phosphatase 3, which in higher animals can dephosphorylate a range of phospholipids. Our suppression analyses include assessing the localization of Projectin protein, a titin homolog, in sarcomeres as well as muscle morphology and functional movement assays. We hypothesize that wunen-based suppression is through the elevation of the bioactive lipid Sphingosine 1-phosphate (S1P), which promotes cell proliferation and differentiation in many tissues, including muscle. We confirm the role of S1P in suppression by genetically altering S1P levels via reduction of S1P lyase (Sply) and by upregulating the serine palmitoyl-CoA transferase catalytic subunit gene lace, the first gene in the de novo sphingolipid biosynthetic pathway and find that these manipulations also reduce muscle degeneration. Furthermore, we show that reduction of spinster (which encodes a major facilitator family transporter, homologs of which in higher animals have been shown to transport S1P) can also suppress dystrophic muscle degeneration. Finally, administration to adult flies of pharmacological agents reported to elevate S1P signaling significantly suppresses dystrophic muscle phenotypes. Our data suggest that localized intracellular S1P elevation promotes the suppression of muscle wasting in flies.


Subject(s)
Down-Regulation/genetics , Drosophila melanogaster/genetics , Lysophospholipids/genetics , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/prevention & control , Phenotype , Sphingosine/analogs & derivatives , Up-Regulation/genetics , Animals , Lysophospholipids/biosynthesis , Muscular Dystrophy, Animal/diagnosis , Mutation , Myofibrils/genetics , Myofibrils/metabolism , Myofibrils/pathology , Signal Transduction/genetics , Sphingosine/biosynthesis , Sphingosine/genetics
15.
J Pharmacol Exp Ther ; 358(2): 359-70, 2016 08.
Article in English | MEDLINE | ID: mdl-27317800

ABSTRACT

Initially discovered as abundant components of eukaryotic cell membranes, sphingolipids are now recognized as important bioactive signaling molecules that modulate a variety of cellular functions, including those relevant to cancer and immunologic, inflammatory, and cardiovascular disorders. In this review, we discuss recent advances in our understanding of the role of sphingosine-1-phosphate (S1P) receptors in the regulation of vascular function, and focus on how de novo biosynthesized sphingolipids play a role in blood pressure homeostasis. The therapeutic potential of new drugs that target S1P signaling is also discussed.


Subject(s)
Blood Pressure , Homeostasis , Lysophospholipids/biosynthesis , Lysophospholipids/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Endothelium, Vascular/metabolism , Humans , Sphingosine/biosynthesis , Sphingosine/metabolism , Vasodilation
16.
FASEB J ; 29(7): 2803-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25805832

ABSTRACT

Sphingosine kinase 1 (SK1), the enzyme responsible for sphingosine 1-phosphate (S1P) production, is overexpressed in many human solid tumors. However, its role in clear cell renal cell carcinoma (ccRCC) has not been described previously. ccRCC cases are usually associated with mutations in von Hippel-Lindau (VHL) and subsequent normoxic stabilization of hypoxia-inducible factor (HIF). We previously showed that HIF-2α up-regulates SK1 expression during hypoxia in glioma cells. Therefore, we hypothesized that the stabilized HIF in ccRCC cells will be associated with increased SK1 expression. Here, we demonstrate that SK1 is overexpressed in 786-0 renal carcinoma cells lacking functional VHL, with concomitant high S1P levels that appear to be HIF-2α mediated. Moreover, examining the TCGA RNA seq database shows that SK1 expression was ∼2.7-fold higher in solid tumor tissue from ccRCC patients, and this was associated with less survival. Knockdown of SK1 in 786-0 ccRCC cells had no effect on cell proliferation. On the other hand, this knockdown resulted in an ∼3.5-fold decrease in invasion, less phosphorylation of focal adhesion kinase (FAK), and an ∼2-fold decrease in angiogenesis. Moreover, S1P treatment of SK1 knockdown cells resulted in phosphorylation of FAK and invasion, and this was mediated by S1P receptor 2. These results suggest that higher SK1 and S1P levels in VHL-defective ccRCC could induce invasion in an autocrine manner and angiogenesis in a paracrine manner. Accordingly, targeting SK1 could reduce both the invasion and angiogenesis of ccRCC and therefore improve the survival rate of patients.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Mutation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/etiology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Down-Regulation , Focal Adhesion Kinase 1/metabolism , Gene Knockdown Techniques , Humans , Kidney Neoplasms/etiology , Kidney Neoplasms/pathology , Lysophospholipids/biosynthesis , Neoplasm Invasiveness , Neovascularization, Pathologic , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis , Sphingosine-1-Phosphate Receptors , Up-Regulation
17.
J Immunol ; 193(6): 3165-74, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25127862

ABSTRACT

Chronic lymphocytic leukemia (CLL) is characterized by the progressive accumulation of clonal B lymphocytes. Proliferation occurs in lymphoid tissues upon interaction of leukemic cells with a supportive microenvironment. Therefore, the mobilization of tissue-resident CLL cells into the circulation is a useful therapeutic strategy to minimize the reservoir of tumor cells within survival niches. Because the exit of normal lymphocytes from lymphoid tissues depends on the presence of sphingosine-1 phosphate (S1P) and the regulated expression of S1P receptor-1 (S1PR1), we investigated whether the expression and function of S1PR1 can be modulated by key microenvironment signals. We found that activation of CLL cells with CXCL12, fibroblast CD40L(+), BCR cross-linking, or autologous nurse-like cells reduces their S1PR1 expression and the migratory response toward S1P. Moreover, we found that S1PR1 expression was reduced in the proliferative/activated subset of leukemic cells compared with the quiescent subset from the same patient. Similarly, bone marrow-resident CLL cells expressing high levels of the activation marker CD38 showed a lower expression of S1PR1 compared with CD38(low) counterparts. Finally, given that treatment with BCR-associated kinase inhibitors induces a transient redistribution of leukemic cells from lymphoid tissues to circulation, we studied the effect of the Syk inhibitors piceatannol and R406 on S1PR1 expression and function. We found that they enhance S1PR1 expression in CLL cells and their migratory response toward S1P. Based on our results, we suggest that the regulated expression of S1PR1 might modulate the egress of the leukemic clone from lymphoid tissues.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lysophospholipids/immunology , Oxazines/pharmacology , Pyridines/pharmacology , Receptors, Lysosphingolipid/immunology , Sphingosine/analogs & derivatives , Stilbenes/pharmacology , ADP-ribosyl Cyclase 1/biosynthesis , Adult , Aged , Aged, 80 and over , Animals , B-Lymphocytes , CD40 Ligand/biosynthesis , Cell Movement , Chemokine CXCL12/biosynthesis , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lysophospholipids/biosynthesis , Male , Membrane Glycoproteins/biosynthesis , Mice , Middle Aged , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcr/biosynthesis , Receptors, CXCR4 , Receptors, Lysosphingolipid/biosynthesis , Sphingosine/biosynthesis , Sphingosine/immunology , Sphingosine-1-Phosphate Receptors , Syk Kinase , Tumor Cells, Cultured , Tumor Microenvironment
18.
Nature ; 465(7301): 1084-8, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20577214

ABSTRACT

Tumour-necrosis factor (TNF) receptor-associated factor 2 (TRAF2) is a key component in NF-kappaB signalling triggered by TNF-alpha. Genetic evidence indicates that TRAF2 is necessary for the polyubiquitination of receptor interacting protein 1 (RIP1) that then serves as a platform for recruitment and stimulation of IkappaB kinase, leading to activation of the transcription factor NF-kappaB. Although TRAF2 is a RING domain ubiquitin ligase, direct evidence that TRAF2 catalyses the ubiquitination of RIP1 is lacking. TRAF2 binds to sphingosine kinase 1 (SphK1), one of the isoenzymes that generates the pro-survival lipid mediator sphingosine-1-phosphate (S1P) inside cells. Here we show that SphK1 and the production of S1P is necessary for lysine-63-linked polyubiquitination of RIP1, phosphorylation of IkappaB kinase and IkappaBalpha, and IkappaBalpha degradation, leading to NF-kappaB activation. These responses were mediated by intracellular S1P independently of its cell surface G-protein-coupled receptors. S1P specifically binds to TRAF2 at the amino-terminal RING domain and stimulates its E3 ligase activity. S1P, but not dihydro-S1P, markedly increased recombinant TRAF2-catalysed lysine-63-linked, but not lysine-48-linked, polyubiquitination of RIP1 in vitro in the presence of the ubiquitin conjugating enzymes (E2) UbcH13 or UbcH5a. Our data show that TRAF2 is a novel intracellular target of S1P, and that S1P is the missing cofactor for TRAF2 E3 ubiquitin ligase activity, indicating a new paradigm for the regulation of lysine-63-linked polyubiquitination. These results also highlight the key role of SphK1 and its product S1P in TNF-alpha signalling and the canonical NF-kappaB activation pathway important in inflammatory, antiapoptotic and immune processes.


Subject(s)
Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , TNF Receptor-Associated Factor 2/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Biocatalysis , Cell Line , Enzyme Activation , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Lysine/metabolism , Lysophospholipids/biosynthesis , Lysophospholipids/chemistry , Mice , Models, Molecular , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Protein Structure, Tertiary , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Sphingosine/biosynthesis , Sphingosine/chemistry , Sphingosine/metabolism , Substrate Specificity , TNF Receptor-Associated Factor 2/chemistry , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination/drug effects
19.
J Biol Chem ; 289(5): 2801-14, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24318881

ABSTRACT

Apolipoprotein M (apoM), a lipocalin family member, preferentially associates with plasma HDL and binds plasma sphingosine 1-phosphate (S1P), a signaling molecule active in immune homeostasis and endothelial barrier function. ApoM overexpression in ABCA1-expressing HEK293 cells stimulated larger nascent HDL formation, compared with cells that did not express apoM; however, the in vivo role of apoM in HDL metabolism remains poorly understood. To test whether hepatic apoM overexpression increases plasma HDL size, we generated hepatocyte-specific apoM transgenic (APOM Tg) mice, which had an ∼3-5-fold increase in plasma apoM levels compared with wild-type mice. Although HDL cholesterol concentrations were similar to wild-type mice, APOM Tg mice had larger plasma HDLs enriched in apoM, cholesteryl ester, lecithin:cholesterol acyltransferase, and S1P. Despite the presence of larger plasma HDLs in APOM Tg mice, in vivo macrophage reverse cholesterol transport capacity was similar to that in wild-type mice. APOM Tg mice had an ∼5-fold increase in plasma S1P, which was predominantly associated with larger plasma HDLs. Primary hepatocytes from APOM Tg mice generated larger nascent HDLs and displayed increased sphingolipid synthesis and S1P secretion. Inhibition of ceramide synthases in hepatocytes increased cellular S1P levels but not S1P secretion, suggesting that apoM is rate-limiting in the export of hepatocyte S1P. Our data indicate that hepatocyte-specific apoM overexpression generates larger nascent HDLs and larger plasma HDLs, which preferentially bind apoM and S1P, and stimulates S1P biosynthesis for secretion. The unique apoM/S1P-enriched plasma HDL may serve to deliver S1P to extrahepatic tissues for atheroprotection and may have other as yet unidentified functions.


Subject(s)
Apolipoproteins/genetics , Apolipoproteins/metabolism , Hepatocytes/metabolism , Lipocalins/genetics , Lipocalins/metabolism , Lipoproteins, HDL/metabolism , Liver/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Apolipoproteins E/blood , Apolipoproteins M , Female , Humans , Lipoproteins, HDL/blood , Lipoproteins, HDL/chemistry , Liver/cytology , Lysophospholipids/biosynthesis , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Particle Size , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Sphingosine/biosynthesis , Sphingosine/metabolism
20.
Stroke ; 46(8): 2260-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26138121

ABSTRACT

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) is a complex stroke subtype characterized by an initial brain injury, followed by delayed cerebrovascular constriction and ischemia. Current therapeutic strategies nonselectively curtail exacerbated cerebrovascular constriction, which necessarily disrupts the essential and protective process of cerebral blood flow autoregulation. This study identifies a smooth muscle cell autocrine/paracrine signaling network that augments myogenic tone in a murine model of experimental SAH: it links tumor necrosis factor-α (TNFα), the cystic fibrosis transmembrane conductance regulator, and sphingosine-1-phosphate signaling. METHODS: Mouse olfactory cerebral resistance arteries were isolated, cannulated, and pressurized for in vitro vascular reactivity assessments. Cerebral blood flow was measured by speckle flowmetry and magnetic resonance imaging. Standard Western blot, immunohistochemical techniques, and neurobehavioral assessments were also used. RESULTS: We demonstrate that targeting TNFα and sphingosine-1-phosphate signaling in vivo has potential therapeutic application in SAH. Both interventions (1) eliminate the SAH-induced myogenic tone enhancement, but otherwise leave vascular reactivity intact; (2) ameliorate SAH-induced neuronal degeneration and apoptosis; and (3) improve neurobehavioral performance in mice with SAH. Furthermore, TNFα sequestration with etanercept normalizes cerebral perfusion in SAH. CONCLUSIONS: Vascular smooth muscle cell TNFα and sphingosine-1-phosphate signaling significantly enhance cerebral artery tone in SAH; anti-TNFα and anti-sphingosine-1-phosphate treatment may significantly improve clinical outcome.


Subject(s)
Lysophospholipids/biosynthesis , Sphingosine/analogs & derivatives , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/physiopathology , Tumor Necrosis Factor-alpha/biosynthesis , Vasoconstriction/physiology , Animals , Cerebral Arteries/drug effects , Cerebral Arteries/physiology , Gene Targeting/methods , Lysophospholipids/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Organ Culture Techniques , Phenylephrine/administration & dosage , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/biosynthesis , Sphingosine/deficiency , Subarachnoid Hemorrhage/therapy , Tumor Necrosis Factor-alpha/deficiency , Vasoconstriction/drug effects , Vasomotor System/drug effects , Vasomotor System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL