Abstract
Sphingolipid metabolism plays a critical role in regulating processes that control cellular fate. This dynamic pathway can generate and degrade the central players: ceramide, sphingosine and sphingosine-1-phosphate in almost any membrane in the cell, adding an unexpected level of complexity in deciphering signaling events. While in vitro assays have been developed for most enzymes in SL metabolism, these assays are setup for optimal activity conditions and can fail to take into account regulatory components such as compartmentalization, substrate limitations, and binding partners that can affect cellular enzymatic activity. Therefore, many in-cell assays have been developed to derive results that are authentic to the cellular situation which may give context to alteration in SL mass. This review will discuss approaches for utilizing probes for mammalian in-cell assays to interrogate most enzymatic steps central to SL metabolism. The use of inhibitors in conjunction with these probes can verify the specificity of cellular assays as well as provide valuable insight into flux in the SL network. The use of inhibitors specific to each of the central sphingolipid enzymes are also discussed to assist researchers in further interrogation of these pathways.
INTRODUCTION.
Sphingolipids and their metabolizing enzymes comprise a dynamic network of critical players in cell signaling and biology. Three central sphingolipids have been extensively studied and implicated in signaling and biology in cells and in vivo. Ceramide (Cer), the central lipid in sphingolipid metabolism, has been implicated in cell death, cellular stress responses, senescence and cell cycle arrest (143, 226). Sphingosine (Sph), the immediate metabolite of ceramide breakdown, has been associated with growth arrest and cell death as well (90, 184). On the other hand, sphingosine-1-phosphate (S1P) has roles in cellular proliferation, migration, invasion, and inflammation, thereby often representing an opposing signal to Sph and Cer.
Eukaryotic cells regulate this network at both the enzyme and lipid levels throughout multiple sub cellular organelles. Consequently, regulation of the 40 plus enzymes central to metabolism of bioactive sphingolipids, along with hundreds (and possibly thousands) of distinct sphingolipid metabolic products, plays a large role in maintaining membrane composition as well as coordinating lipid signaling events (72). The amphipathic sphingolipids contain a hydrophobic fatty acyl chain attached to another hydrophobic sphingoid backbone, forming ceramide that can be derivatized with the addition of multiple polar head groups. Given the diversity of sphingolipids, both at the molecular and signaling level, there are numerous experimental approaches taking advantage of the various biochemical properties to determine regulation and signaling potential of these potent bioactive lipids.
Within the cell, the hydrophobic nature of sphingolipids, imparted by the two hydrocarbon chains, restricts the movement of Cer and complex sphingolipids to vesicular trafficking or protein-mediated transport, adding another potential level of regulation. On the other hand, the less abundant single chained sphingolipids, such as Sph and S1P, can be soluble in the cytosol and move much more freely between membranes, potentially confounding efforts to identify the compartment of origin of these potent signaling molecules (Fig. 1). To add even more intricacy, sphingolipid pathways are often subject to substrate availability, interconnecting metabolic pathways that utilize the same substrate. Consequently, when assessing sphingolipid metabolism, all of these considerations add levels of complexity to an already multifaceted system.
Figure 1.
Bioactive sphingolipids can be generated in multiple membranes throughout the cell. Generation of sphingolipids de novo begins with the condensation of serine and palmitoyl-CoA via the serine palmitoyltransferase complex (SPT). The 3-ketosphinganine (3-KSph) product is reduced by 3-ketosphinganine reductase (3-KR) to generate dihydrosphingosine (dHSph). Next, ceramide synthases (CerS) derivatize dHSph with one of various fatty acids to form dihydroceramides (dHCer) of various acyl-chain lengths. Dihydroceramide desaturase (DES) then reduces dHCer to form ceramide (Cer), which can be metabolized in the ER forming either ceramide phosphoethanolamine (CPE) or galactosylceramide (GalCer). Ceramide can also be transported to the Golgi via ceramide transport protein (CERT) or through vesicular trafficking to generate sphingomyelin (SM) or glycosphingolipids (GSL), respectively. These complex sphingolipids can then be transported to the plasma membrane (PM) where hydrolytic pathways can degrade SM (SMases) or GSL (Gcase) to produce Cer at the PM or in endolysosomes. The Cer that is produced can be further broken down by ceramidases (CDase) to form sphingosine (Sph). Free Sph can either serve as a substrate for CerS to generate ceramide in a processed termed “salvage/recycling” or begin clearance from the pathway by phosphorylation and subsequent degradation via sphingosine kinase (SK) and sphingosine lyase (SPL), respectively.
Thus, complementary approaches to measurements of sphingolipids have been designed and refined to gain the most accurate picture of how sphingolipid metabolism is regulated and which component is modulated under different experimental conditions and in response to various stimuli. For instance, the analysis of ceramide mass via the decades old diglyceride kinase detection assay is still an effective means for this purpose, although this has been largely superseded by LC/MS technology. Though, to further this method, the use of a tracer, such as 17 carbon dihydrosphingosine, can be utilized to distinguish between de novo and hydrolytically generated Cer. In this review, we provide an overview of commonly utilized techniques for the analysis of sphingolipid metabolism, along with parameters on the use of labels and inhibitors to probe sphingolipid homeostasis in mammalian cell lines.
1. Sphingolipid metabolism
1.1. Synthesis and Degradation
Sphingolipids represent the third most abundant lipid class found in mammalian cells, after glycerophospholipids and sterols, and are easily the most structurally diverse. They are typically distinguished by their 18-carbon amino alcohol backbone. This “sphingoid” backbone is synthesized in the ER through a condensation of serine and palmitoyl-CoA and eventually N-acylated with one of dozens of different fatty acids resulting in the formation of a family of distinct Cers (Fig. 1). This basic sphingolipid is modified at the 1-hydroxyl group to create a vast subset of sphingolipid subclasses. Thus, addition of phosphate, phosphocholine, or sugar groups creates ceramide 1-phosphate (C1P), sphingomyelin (SM), or glycosphingolipids, respectively. While synthesis and degradation of sphingolipids is almost always compartment-dependent, movement of lipids between membranes makes tracking of biologically relevant accumulations more difficult.
An important aspect of sphingolipid enzyme regulation is localization of the enzymes to different organelles in the cell, as presented in Figure 1. De novo synthesis occurs in the ER, where serine palmitoyltransferase (SPT), ceramide synthases (CerS), and ceramide desaturase (DeS) are localized. Transfer of Cer to the Golgi either through the ceramide transporter (CERT) or through vesicular pathways leads to the production of either SM or glucosylceramide (GluCer), respectively. Through vesicular transport, SM can travel to the plasma membrane (PM) whereas GluCer is utilized in the synthesis of more complex glycosphingolipids in additional Golgi sub-compartments prior to transport. At the PM, both SM and glycosphingolipids become part of a dynamic relationship between the catabolizing enzymes, sphingomyelinases (SMases), cerebrosidases, ceramidases (CDase), and others to influence membrane dynamics or be utilized in signaling events (87, 240). Endolysosomal action can begin the processes of clearance and recycling of sphingolipids. As the pH of lysosomes is acidic, another category of sphingolipid enzymes with acidic pH optima resides here. These enzymes help to coordinate the degradation of SM and hexosylCer (HexCer) (denoting GluCer and/or GalCer), and in turn generated from degradation of more complex glycosphingolipids) into Cer and eventually into Sph by the actions of acid CDase (aCDase), acid SMase (aSMase), and glucosyl-ceramidase (GBA). Sph, a soluble metabolite in cells, can be acted on by sphingosine kinases (SK1 or SK2) to generate S1P, which can be irreversibly degraded by S1P lyase (SPL), representing the only known clearance for metabolites from the sphingolipid network. Alternatively, Sph can be acylated to Cer, and S1P can be dephosphorylated by S1P phosphatases (SPP) and then re-acylated in the ER to re-enter the sphingolipid pathway.
1.2. Complexity of Sphingolipids
Sphingolipids can be metabolized and catabolized in specific cellular compartments, potentially resulting in different signaling and biologies. This complexity is underscored by the presence of at least 5 distinct ceramidases and 5 distinct sphingomyelinases (products of distinct genes) that show selective subcellular localization.
Cer can be generated de novo or via re-acylation in the ER, or by degradation of SM at the PM (38), in the nucleus (185), in lysosomes (88) or in mitochondria (165). Ceramide can then mediate specific cellular responses through activation of one or more serine/threonine phosphatases (35) as well as possibly other targets (cathepsins (79), PKCz(105))). Similarly, Cer can be degraded to Sph in almost any cellular membrane by the action of compartment-specific CDases (39). Sph can then be phosphorylated to S1P either in the nucleus (85, 176), at the PM (199), or possibly additional compartments by the actions of SKs. S1P can then be transported outside the cell to act on one of five G-protein coupled receptors (S1PRs1–5) generating pro-survival and inflammatory biologies (194, 207). S1P can also act on intracellular targets (200). The metabolic interconnections of bioactive sphingolipids also add to the complexity of their study. For example, actions attributed to acid sphingomyelinase cannot be a priori assigned to ceramide as the direct product. This was recently demonstrated in the case of the function of acid sphingomyelinase in regulating production of cytokines and chemokines where this was found to be dependent on further metabolism of ceramide to ceramide 1-phosphate (139). Given this interconnectivity of sphingolipid metabolism, methods for simultaneously monitoring the mass of multiple sphingolipid species in conjunction with their flux and intracellular transport/location are needed to allow more complete insight into the dynamic origins of sphingolipid signaling in cellular fate.
2. Determining Sphingolipid Mass
Detection of sphingolipid metabolism often begins with the analysis of cellular sphingolipid mass. This was classically accomplished by thin layer chromatography (TLC), including high performance TLC (HP-TLC) and the diacylglycerol kinase assay for ceramide (153), but in the last 15–20 years mass detection and quantitation have moved into the realm of mass spectrometry (MS). This field is now rapidly expanding with the development of new instrumentation and improved sensitivity. However, much of the current methods used to examine sphingolipids are still prone to the major drawback of analyzing total cellular sphingolipids as a snapshot of this dynamic network.
2.1. Mass Spectrometry
MS analysis is now recognized as the most sensitive and selective means of determining cellular concentrations of sphingolipids (63). As MS identifies molecules based on mass to charge ratio (m/z), initial studies were quick to identify the 246m/z “sphingoid backbone” fragment ion that is characteristic of collision-induced dissociation (CID) for most sphingolipid species (63). Though for SM, the 184.1m/z phosphocholine product ion was found to be the most intense product ion and thus is utilized for its identification (94). Interestingly, these initial studies were able to achieve sub-picomolar sensitivity (63), which, even with large technical advances, has only increased modestly over the past few years. The major advantage of MS over other techniques mentioned in the following sections is the added ability to quantify molecular species characterized by the presence of a specific fatty acid linked to a specific sphingoid backbone (ie. C16, C18, C24-Cer) for each sphingolipid class (i.e. Cer, SM, HexCer), allowing for a more precise and global approach for profiling the sphingolipid network. This aspect acquires particular relevance as it is becoming evident that individual species may exert specific biological functions (73).
High resolution instrumentation has led to an increase in selectivity that has allowed for the deconvolution of complex samples with multiple nearly isobaric (having identical mass) lipid species. Numerous robust and reliable methodologies exist (23, 186) that utilize HPLC MS/MS as a workhorse for the analysis of mammalian sphingolipids, including sphingoid bases, sphingoid base phosphates, Cer, HexCer, SM, C1P, and alpha-hydroxy ceramides (a-HO-Cer). In addition, methodologies for SM often require a hydrolysis step in order to remove contaminating isotopic peaks produced by the more abundant phosphatidylcholine. These contaminating peaks can have overlapping mass transitions generating the common 184.1m/z phosphocholine product ion. As demonstrated by Peng et al,(150) mass differences in lipids can be utilized with high mass accuracy MS to distinguish nearly isobaric sphingolipids. Indeed, these authors were able to separate an odd chain C23:0-Cer (635.6216 m/z) from the hydroxylated C22:1-HO-Cer (635.5852m/z) utilizing the less than 0.2 dalton mass difference generated via the extra oxygen (150). These developing methodologies also have the added benefit of increased functional sensitivity, owed to significantly reduced background when compared to standard low-resolution MS techniques (25, 150).
Though MS analysis of sphingolipids can be utilized without chromatographic separation, termed shotgun lipidomics (62, 67, 174), these methods are susceptive to isobaric interference, as well as loss in sensitivity due to ion suppression. Thus, the addition of the HPLC step, which takes advantage of the amphipathic nature of sphingolipids, represents an orthogonal separation to mass. Nevertheless, there are many considerations that must be taken into account when utilizing MS detections coupled with HPLC separation, especially for quantitative purposes. Primarily, the proper standards must be utilized for both method development and as internal standards to account for extraction efficiencies, ionization effects, and other differences that may arise from specific biochemical properties of the analyte. Ideally, internal standard mixes for simple sphingolipid subspecies would contain a stable isotope label for each molecular species that would generate odd massed parent and product ions. Accordingly, internal standard mass transitions would fall between the even numbered mass transitions that are produced by most endogenous sphingolipids, readily allowing for MS separation of internal standard masses from analytes of interest. Unfortunately, when considering the myriad of sphingolipid molecular species, only a small fraction is currently available as isotopically labeled standards. Therefore, many investigators utilize an array of representative odd chain or stable isotope sphingolipids as internal standards to account for the different chemical and biophysical properties of each sphingolipid subspecies (24, 127). The reverse phase chromatographic conditions typically used to separate sphingolipids prior to MS are also a major consideration. Elution of isobars or isomers, such as GluCer and galactosylceramide (GalCer), require separation prior to MS detection (127, 230). It should also be noted that if separation cannot be achieved under typical HPLC/MS conditions, the analytes are commonly reported as a grouped subspecies if detection is possible; for example, HexCer is used to denote any hexose-modified ceramide primarily including GluCer and GalCer. This is still useful in this case because most non-neuronal tissues contain much higher levels of GluCer than GalCer. In comparison to shotgun lipidomics, chromatographic separation substantially reduces ion suppression in the MS source by reducing the sample complexity at any given point during a gradient. Finally, extraction conditions should be considered carefully based on the sphingolipid species to be analyzed. Sphingolipids have been extracted from cell lysate utilizing a plethora of extraction techniques [reviewed in (36)], but major considerations include the disparity of hydrophobicity between sphingolipid molecular species and the sizable differences in polarity in sphingolipid sub species generated via the derivatized head group. When all of these considerations are accounted for, MS can be utilized as a powerful tool for the quantitative analysis of multiple classes of sphingolipids across many biological matrixes.
2.2. Fluorescence and Ultra Violet detection
Profiling sphingolipids has primarily been achieved via chromatography though the use of traditional HPLC detectors has been hampered as sphingolipids do not contain natural fluorophores or chromophores. Sample derivatization has played a key role in the early understanding of alterations in total cellular sphingolipid content. Iwamori et al. were among the first to derivatize Cer utilizing benzoyl chloride to generate UV absorbent Cer molecules which were detectable in the 230–280 nm range with a detection limit of 10 nmoles (86). Interestingly, these N-acyl benzoylated Cer derivatives were separated via normal phase chromatography on silica columns utilizing a mobile phase containing hexane/ethyl acetate. The major drawback with the derivatization technique is that it is highly sensitive to water, and the chemicals are highly toxic. By comparison, other derivatization techniques with fluorescent molecules yielded an order of magnitude more sensitivity and added selectivity as reverse phase HPLC began to be utilized for sphingolipids (161), thus allowing for the separation of Cer species based on their N-acyl chain length and degrees of desaturation. Indeed, Yano et al. utilized acetonitrile/methanol/ethyl acetate mobiles phases in conjunction with a C18 stationary phase and derivatization with the fluorescent reagent anthroyl-cyanide to separate and detect over 17 distinct Cer and dihydroceramide (dhCer) molecular species (242). Chromatographic separations such as these began to allow for clarification of the role and metabolism of sphingolipids as well as the molecular species contained in each group. The major drawback of these derivatization procedures comes from the inherent instability introduced with the derivatization products. Thus, samples typically cannot be stored for long periods of time and must be analyzed quickly following the reaction (202). Also, unlike MS analysis of the directly extracted lipids, derivatization reactions may introduce more variability, consequently reducing the recovery of sphingolipids or affecting the reproducibility of a sphingolipid analysis.
2.3. Thin Layer Chromatography Detection
Thin layer chromatography (TLC), is an indispensable tool for the analysis of both sphingolipid flux and mass. This relatively rapid method offers many advantages over other techniques noted in this section, as it is robust, simple, and economical. This decades-old system is still utilized by investigators desiring a broad overview of sphingolipid classes, typically by normal phase separation (49). The conditions commonly utilized are a solvent mixture of chloroform/methanol/aqueous solution on silica gel, alumina, or kieselguhr stationary phase (49, 108). After separation on the TLC plate, based on sphingolipid biophysical properties, analytes are visualized and quantified by densitometry after identification and comparison with standards. To increase the sensitivity of TLC analysis, cells are often labeled with radioactive or fluorescent probes and/or precursors, thereby greatly reducing background. Recently, high performance TLC has offered greater reproducibility and fidelity combined with a decrease in the necessary sample volume (49, 108, 205). These advances in chromatography have been utilized by Torretta et al. in conjunction with MALDI MS detection to analyze sphingolipid molecules that can be difficult to define by standard means (217). Thus, although TLC is an older method, it is still used extensively and effectively for measuring sphingolipids.
2.4. Diglyceride Kinase Assay
Another historically important method for the analysis of total cellular ceramide content is the diglyceride kinase (DGK) assay. The assay was developed to monitor diacylglycerol (DAG) content in cells through mass conversion to phosphatidic acid (160), but it was quickly realized that bacterial DGK will also phosphorylate the 1’ hydroxyl group on Cer to form C1P, thus allowing for radiometric quantitation of Cer in a given cell lysate as long as quantitative (full) conversion of ceramide is achieved (153). This is a critical consideration. Additional special consideration should be given to the form of the E. coli DGK enzyme, as it has been noted that lyophilized enzyme preparations may be prone to stimulation by endogenous lipids, potentially skewing Cer readings (153), a shortcoming that is obviated if DGK activity is in large excess and therefore not susceptible to activators or inhibitors. Labeled C1P product identification and quantitation is based on TLC plate mobility when compared to appropriate standards. This assay was also adapted for the analysis of free sphingoid bases by Van Veldhoven et al.; following chemical N-acylation with hexanoic anhydride resulting in the formation of C6-ceramide which was then used as a substrate for E. coli DGK and quantified (225). This method has been instrumental in the analysis of Cer and its roles in biology but has been mostly replaced by MS technology.
2.5. Antibodies
The subcellular site of sphingolipid generation is potentially of equal importance to changes in their mass since most of the bioactive sphingolipids demonstrate compartment specificity. Spatial context can be given to sphingolipid levels with the use of anti-sphingolipid antibodies (ASABs). Perhaps qualitative at best, ASABs also allow for the study of protein-lipid interactions through pulldown experiments. Therefore, while other methods in this section focus on evaluating sphingolipid mass, these antibodies begin to interrogate compartment-specific signaling events (92). Currently, ASABs are available for determining cellular pools of Cer, Sph, and S1P (172, 175). ASABs have been utilized with flow cytometry, immunohistochemistry and western blotting, as well as for lipid-protein immunoprecipitation. Moreover, ASABs have the potential to be utilized therapeutically. The Cer antibody has been utilized as a countermeasure against the harmful side effects of Cer-induced apoptosis in radiation (172). It should be noted, however, that the lipid specificity of many of these antibodies (especially anti-ceramide antibodies) has not been well established in vitro and even less so in cells (43, 92, 102).
3. Evaluating sphingolipid biosynthetic flux; use of probes and inhibitors
Levels of sphingolipid species in the cell can be regulated either by changes in the expression and/or activity of the enzymes that act on them, as well as by substrate availability/load. Additionally, localization of specific sphingolipid enzymes often dictates the metabolic fate of sphingolipids. This flux in the sphingolipid network both in localization and levels of metabolites has been shown to have biological implications. For example, roles have been established for mitochondrial (193) and nuclear Cer production in apoptosis (235), while Cer generated at the PM by nSMase2 is an essential part of breast cancer cells cytostatic responses to certain chemotherapeutics (185). Better understanding of the effects of Cer generation and metabolic flux will provide significant and more specific insight into Cer-mediated-biological processes and possibly chemotherapeutics.
Thus, analysis of flux in this dynamic network can provide important context to sphingolipid mass levels, be it enzymatic or spatial. Throughout the years, many of the aforementioned techniques for monitoring sphingolipid mass have been adapted into pulse/chase type experiments, often used in conjunction with inhibitors to validate flux at key points in sphingolipid metabolism (182). Many enzymes have a vast array of probes or inhibitors (Figures 2 and 3), and there are many reviews on this topic that comprehensively cover sphingolipid metabolism discussing the importance of functional groups and the classes of inhibitors available (44, 121, 126, 182). This section will highlight some of the more useful probes and the more potent or relevant inhibitors that lend themselves to the analysis of flux in situ for mammalian cells. Of note, many of the in situ labels can be used to monitor flux through multiple enzymes, briefly reviewed in the section on probing de novo flux.
Figure 2.
An overview of commonly used probes for sphingolipid metabolism. All labels represent non-radioactive label tracers that can be utilized to probe sphingolipid metabolism in cells. Typical concentrations and duration of labeling to accurately monitor flux are presented.
Figure 3.
An Overview of commonly used inhibitors of sphingolipid metabolism. Structures of commonly used inhibitors with concentration and duration for typical treatments for cellular use.
Many of the probes mentioned in the following sections share similar structures with endogenous sphingolipids, and therefore biochemical properties, leading to important considerations in their handling and use. Storing solutions of sphingolipid probes at −80°C can cause them to come out of solution, but most lipids are fairly stable and can be stored at −20°C.If a solution containing a probe appears to contain precipitant or is hazy, sonication and/or slight heat (<45°c) can be utilized to get the lipid back into solution. A caveat would be fluorescent lipid probes containing NBD or BODIPY fluorescent groups that can have limited photostability, though it should be noted that BODIPY is the more stable of the two probes.
Most sphingolipid probes are available in a variety of N-acyl chain lengths (from short to long) and are soluble in methanol or ethanol. Though most cells can tolerate small amounts of organic solvent in their media (for probe delivery), probes can also be complexed with bovine serum albumin allowing for delivery in aqueous solutions. Finally, it is important to note that sphingolipid probes or their metabolites have the potential to instigate cellular signaling, due to similarities to their endogenous counterparts, and therefore should be used at the lowest possible dose.
3.1. Serine palmitoyltransferase- probes and inhibitors
The first step in de novo sphingolipid metabolism occurs in the ER via SPT. The basic SPT heterodimer complex (SPTLC1 and SPTLC2 or SPTLC3) catalyzes a condensation reaction of serine and palmitoyl-CoA. In yeast, SPT is appreciated to be a multicomponent complex, including additional subunits. Both stable isotope and radioactive substrates have been utilized in the literature to probe this rate limiting step, though as noted by Merrill et al. special care must be exercised with the use of labeled palmitate, as even at low concentrations it can induce sphingolipid biologies (77). Isotopically labeled serine on the other hand was able to be utilized over a wide range of concentrations exhibiting only a slight ability to drive sphingolipid synthesis at mM concentrations (up to 30%) (130). Most methods utilized serine-depleted cell culture media that is reconstituted with isotopically labeled serine to increase the potential label uptake (69, 191). Concentrations of the isotopically labeled serine are typically in the mid μM range for standard cell culture media (Fig 2). It is important to note that serine is not an essential amino acid, and therefore cells can produce it. This becomes an important consideration for longer pulses, and therefore the levels of serine in the media and cells should be monitored at extended time points. Also of note, the signal to noise ratio is reduced for some stable isotopes (namely 13C serine) due to the relatively high natural abundance of 13C in the hydrocarbon chains of lipids producing large amounts of endogenous sphingolipids with identical masses to their labeled counterparts (93). Radioactive 14C serine has also been extensively utilized in both pulse/chase and steady state assays, and, while it is not subject to the same considerations regarding the background as 13C labeling, the method of detection does not lend itself to separation of sphingolipid molecular species. Typically, cells are labeled for minutes to hours in serine pulse/chase experiments as labeled serine reaches steady state in the cell within 10–20 min (130).
Due to utilization of palmitate by two major steps in ceramide synthesis (SPT and CerS), its incorporation can be more difficult to interpret than serine. One solution to evaluate de novo synthesis (which incorporates palmitate into both positions) and salvage (which incorporates palmitate only in the acyl position) can be achieved through the use and analysis of isotopologues (molecules with different isotopes at the same structural positions) and isotopomers (molecules with the same isotopes at different structural positions) generated via the incorporation of label into either the sphingoid backbone, N-acyl chain, or both. Indeed Sims et al. used 0.1mM [13C]palmitate to implicate de novo synthesis in the initial autophagic response of RAW264.7 cells to KLA (Kdo2-lipid A) treatment as most of the Cer mass generated in unlabeled cells could be accounted for in the labeled sphingoid backbone upon addition of the probe (190). As labeling to steady state tends to obfuscate flux results, it should be noted that, in the case of palmitate, labeling to steady state was relatively rapid. When HEK293 cells were labeled with 0.1mM of [13C] palmitate for 6 h, the palmitoyl-CoA pools utilized to produce 3-ketosphinganine nearly reached isotopic equilibrium (77). One major limitation in this approach is the biologic effects that higher concentrations of fatty acids (above 0.1mM) can have on ion transport, gene regulation, and even sphingolipid metabolism (77). A second limitation to both palmitate and serine labeling is the fact that both substrates for SPT are major components of cellular metabolism and therefore are subject to utilization throughout the cell producing numerous nonsphingolipid metabolites.
The discovery that other amino acids (besides serine) can be utilized for the generation of sphingolipids (151) presents an exciting opportunity to utilize other stable-isotope amino acids to probe generation of these non-canonical sphingolipid based (249). In this case, use of alanine or glycine by SPT generates deoxy-dihydrosphingosine and 1-deoxymethyl-dihydrosphingosine. Indeed, the use of isotopically labeled alanine and serine was utilized to verify that deoxysphingolipids, thought to initially be the result of action of mutations in the SPTLC1 gene (151), are regularly produced by mammalian cells (249).
The condensation reaction of SPT to form 3-ketosphinganine has multiple inhibitors including myriocin, lipoxamycin and fungins (44). The most potent of these, myriocin, also known as ISP-1, has an established Ki of 0.28 nM in vitro and an IC50 of 15 nM in CTLL-2 cells (133). It is commonly used between 0.1 and 5 μM for up to 24 h in mammalian cells (Fig 3) (5, 82, 104, 134), although the rationale for the higher concentrations is not known and currently should not be encouraged. Myriocin inhibition is likely driven through the similarities it has to a catalytic intermediate of the SPT reaction (68). Not surprisingly, structurally modified amino acids such as cycloserine and β-chloroalanine have also been utilized for the inhibition of SPT. Though as many enzymes utilize serine and alanine, these inhibitors are not very specific (124, 204). Indeed, Hanada et al. demonstrated that in SPT deficient cells, addition of Sph could rescue cell growth from myriocin treatment, but was ineffective against modified amino acids, indicating that the primary effects of cycloserine and β-chloroalanine on growth were not through SPT (70).
3.2. 3-Keto-dihydrosphingosine reductase - probes
The 3-keto-dihydrosphingosine reductase (KDSR) is one of the least understood of the enzymes central to sphingolipid metabolism. Only in vitro assays have been performed to directly measure KDSR activity. These assays utilize 10–100 μM of 3-[4,5- 3H]ketosphinganine for 10 min to 1 h (101, 129, 208) and with the addition of NADPH accounting for the reducing cofactor. The in vivo KDSR activity has not been well studied. No labeled 3KDS and DHS were detected in a flux assay where radio-labeled serine (such as [3H]serine) was added to the live cell culture (128, 133). This led to the notion that the 3KDS is a highly transient sphingolipid intermediate that is converted to downstream metabolites by KDSR at a fast rate, suggesting little/no regulation at the KDSR level. A recent study reported the detection of a stable pool of 3KDS in yeast S. cerevisiae at a level comparable to cellular DHS using a HPLC-ESI-MS/MS method (167). We believe the direct detection of both the substrate and product of KDSR makes it possible to develop labeling methods to follow the flux of serine into the sphingolipid pathway through KDSR and thus measure its activity and regulation in vivo. Therefore, as there are no known inhibitors for KDSR, methods for probing flux represent the first step to explore the physiological activity of KDSR, a long-ignored enzyme in the sphingolipid biosynthetic pathway.
3.3. Ceramide Synthases - probes and inhibitors
In mammals, CerSs constitute a family of 6 distinct enzymes, products of distinct genes (S. cerevisiae has two: Lag1 and Lac1). CerSs1–6 display distinct preference for the fatty acyl CoA substrates, thus generating ceramides, and subsequent complex sphingolipids, with distinct N-linked fatty acids. Probing flux through CerSs presents a challenge in that the CerSs are active in both de novo synthesis and salvage pathway; thus, the flow of these two pathways converge in the ER through CerS-mediated N-acylation of both dhSph and Sph, respectively. Regulation and function of these 6 enzymes, which have been implicated in cellular stress responses such as autophagy, apoptosis, and growth arrest, has been thoroughly reviewed by Wenger et al. (236). Thus, many probes have been developed to assess CerS activity in situ. The different isozymes of CerS have similar affinity towards the dhSph substrate (Km between 2 and 5 μM (103)), and consequently dhCer production appears to be driven by which CerS is present and/or active (135). Hence odd chain sphingoid bases such as d17dhSph and d17Sph are commonly utilized probes. These probes are also easily distinguishable from the 18 carbon backbones of most endogenous sphingolipids and readily allow for the HPLC/MS/MS quantitation of the different CerSs products (183, 196, 197). These d17-sphingoid bases are typically used between 0.5 and 2 μM from 5 min to 4 hours in situ (Fig 2)(134, 183). It is important to avoid higher concentrations of these probes as concentrations of sphingoid bases in the low μM range can exert significant biological effects (168). Also, when monitoring the product of CerS activity, consideration should be given to the substrate utilized, as dhSph requires a second step (via DeS activity) after CerS activity to generate labeled Cer.
Multiple fluorescent probes that do not require MS for detection have also been developed utilizing Sph analogs, including NBD sphingoid bases (215) and “clickable” derivatives (65). The utilization of a photoactivatable and clickable Sph (PACsph) showed that while the uptake of PACsph was very quick, reaching the ER 5 min after addition to the media, it required 4–6 h for PAC-metabolites to reach the PM (65). This study was not only able to visualize sphingolipid metabolism throughout the cell, but also utilized click chemistry to bind PAC-sphingolipid metabolites to interacting proteins.
[13C] palmitate is another tool used to probe flux through the CerS. As previously mentioned, there are considerations to the use of palmitate as a label (reviewed in (77)), but the incorporation of the [13C]palmitate into the sphingoid base, the N-acyl chain, or both presents an opportunity to deconvolute salvage from de novo lipid synthesis. Indeed, in a study of fatty acid effects on insulin resistance utilizing a 3h incubation with 13C palmitate, Hu et al. were able to implicate stimulation of the salvage pathway since most of the N-acyl labeled C16Cer pools (the predominant labeled position) did not contain a labeled sphingoid backbone (81). The 14C radioactive palmitate isotopologue has also been utilized. In a 2005 study, Becker et al. utilized labeled palmitate at 3.0 uCi/ml for 16–24h to validate that phorbol ester treatment was able to increase turnover through hydrolytic and CDase pathways to form free Sph which could be reacylated through salvage to generate bioactive Cer (16). Careful consideration must be given to probing CerS activity as the confluence of salvage and de novo pathways makes it one of the most complicated steps in sphingolipid metabolism to interpret.
The judicious use of inhibitors can also be employed to distinguish the salvage and de novo pathways. Utilizing inhibitors of SPT in conjunction with the potent and what appears to be highly specific CerS inhibitor fumonisin B1 (FB1), Becker et al. were able to demonstrate activation of the salvage pathway in response to sustained PKC activation. In this study, Cer generation upon PKC activation was sensitive to FB1, but not myriocin, thus metabolic flux though salvage was implicated (16). In mammalian cells, FB1 is typically used at a concentration of 5–100 μM (80, 82, 104, 134, 140, 170), and pretreatment of 1–6 h before further perturbation is common, likely to allow cellular uptake (Fig 3) (146, 239). In renal cells, FB1 has an IC50 of 35 μM for inhibition of Cer biosynthesis (141). FB1 contains anionic tricarballylic acids and aminpentol backbone that likely drive competition with the sphingoid and acyl-CoA substrates. The inhibitor itself has been linked to renal and liver toxicity, carcinogenesis, neurotoxicity, and pulmonary edema, which limit its use in vivo. Interestingly, these toxicities could all potentially be linked to the marked elevation in sphingolipid levels (46). The Sph analog FTY720 has also been utilized to decipher regulation amongst the various CerS isozymes, and while concentrations of FTY720 that could inhibit CerS cannot be reached in humans, it still remains a powerful tool for in situ studies. It has been shown to mainly affect long chain Cer synthesis as it predominantly inhibited CerS5 and/or CerS6 (18), though analogues of FTY720 have also been developed for the preferential inhibition of CerS2 and CerS4 (178). Recently, P053, was shown to specifically inhibit CerS1 activity (IC50 of 0.5 μM), demonstrating that very specific and potent CerS inhibitors can be developed (219).
3.4. Dihydroceramide desaturase- probes and inhibitors
As the last enzyme in the de novo generation of Cer, the DeS enzymes catalyze the formation of a double bond between the C4 and C5 position in the sphingoid backbone, although DeS2 also functions as a hydroxylase introducing an OH group on the C4 position of the sphingoid base (effectively making phytoCer). Only relatively recently has dhCer begun to be recognized as a bioactive molecule with implications in autophagy (37), mitochondrial ROS production (55), and growth arrest (123). Therefore, probing the conversion of dhCer to its delta-4 unsaturated metabolite has become an important endeavor. Initial analysis of DeS activity was performed utilizing the fluorescent C6-NBD-dhCer at 0.5 to 10 μM for up to 24 h in situ and in vitro (Fig 2)(37, 97, 136). Indeed, C6-NBD-dhCer was instrumental in the 1997 study by Kok et al., which led to the initial observations that desaturation of the sphingolipid backbone occurred during de novo synthesis and not later in metabolism via desaturation of complex dihydrosphingolipids (97). In a more selective and sensitive assay for DeS activity, D-erythro-2-N-[12′-(1″-Pyridinium)-dodecanoyl]-4,5-dihydrosphingosine bromide (C12dhCCPS) was utilized in cellulo at 0.5 μM and reached a pseudo steady state at 6 h (time course 15 min to 24 h) in two cancer cell lines (100). It should be noted that utilizing C12dhCCPS as substrate with MS as a detector, the transition of 577.4/79.8m/z/90CE (labeled PhytoCer derivative) can be utilized to also monitor the documented hydroxylase activity of DeS2 (Snider unpublished data), thus allowing for at least partial separation of the isoenzymes activities.
Interestingly, DeS regulation has been shown to occur via several mechanisms in the cell. DeS has been shown to be regulated via transcriptional mechanisms as well as by palmitoylation and translocation in response to fatty acid treatment (15). It has also been shown to be regulated through availability of the NADPH cofactor during oxidative stress conditions (84). Two commonly used inhibitors of DeS in the literature are the sphingolipid analog GT11 and the vitamin A analog Fenretinide. Fenretinide (4-hydroxyphenylretinamide, 4HPR) was reported as a competitive inhibitor with a Ki of 8.28 ±1.25 μM and an IC50 of 2.32 μM for enzyme activity in rat liver microsomes by Rahmaniyan in 2011 (164). 4HPR may also inhibit DeS through ROS generation (84), potentially reducing the cellular pools of the DeS cofactor, NADPH. In turn, these two different regulatory mechanisms of DeS activity may account for the different phenotypes seen in cancer cells at low (cytostatic (100)) and high (cytotoxic (233)) 4HPR doses. Building on previous work (232), Kraveka et al. demonstrated an optimal inhibition of DeS activity at 2 h with 1 μM 4HPR in human neuroblast cells, however 4HPR for DeS inhibition has been utilized in cells up to and above the cytotoxic dose of 5 μM for up to 48 h (6, 100, 180). On the other hand, the cyclopropane containing compound GT11 (C8-cyclopropenylceramide,C8CCP) is a sphingolipid analog and exhibited an IC50 of 23 nM for the inhibition of formation of Cer by DeS in primary cultured cerebellar neurons (218). GT11 has also been successfully utilized in vitro as a competitive inhibitor with a Ki of 13 μM. When considering the use of GT11, two important issues must be considered: 1) higher concentrations of GT11 (>5 μM) induce inhibition of SPT via accumulations of sphingoid base phosphates (218), and 2) the cylcopropene structure in GT11 is prone to oxidation via atmospheric oxygen, and stocks must be stored under nitrogen; thus, for extended use, the XM462 analog, which is more chemically stable, may be considered (136). Typically, GT11 concentrations below 2.5 μM are used in cells for 30 min to 2 h pretreatments with a total incubation of up to 24 h (164). Thus, there are multiple very potent and specific options for the inhibition of DeS that are helping to drive forward the understanding of this crucial step in sphingolipid metabolism.
3.5. Sphingomyelin synthase - probes and inhibitors
The generation of SM represents a metabolic fork in the road for ER pools of Cer. The transfer of Cer from the ER to the Golgi via CERT leads to the preferential generation of SM through sphingomyelin synthase 1 (SMS1) (212). SMS2 exhibits similar substrate specificity as SMS1, but can be localized at both the PM and Golgi (210). Non-sphingolipid metabolites may also need to be considered when probing SMS biological function since these enzymes also affect proliferative cellular signaling through the metabolism of phosphatidylcholine (the donor of the choline phosphate head group in the SMS reaction) to form the bioactive molecule DAG (71, 228). SMS activity has traditionally been assessed utilizing C6-NBD-Cer in cells at concentrations of 1–5 μM for 1–8 h (Fig 2) (7, 29, 96, 228). An in vitro study by the Ye group determined the Km for C6-NBD-Cer as a substrate for SMS at 7.5 μM. In a 2007 study evaluating the contribution of the two SMS isozymes to cellular SM, Tafesse et al. utilized C6-NBD-Cer in human cervical carcinoma HeLa cells to evaluate both flux and localization of SMNDB following knock down of either enzyme using RNA interference. While an HPLC analysis quantified the reduced incorporation of the label into SM in either SMS1 or SMS2 knockdowns, utilizing fluorescent microscopy indicated abolished Golgi staining in response to SMS1 knockdown. Therefore, the use of a single probe provided complementary information when different detection methods are employed (206).
The toxins lysenin and equinatoxin-II (EqtII) bind SM with high affinity and specificity, and both are effective probes for visualizing SM and for identifying relative levels of SM at the PM. Yachi et al. observed that, after permeabilization of cells, these stains localized to different organelles, with EqtII staining abolished upon knockdown of SMS1, indicating that these probes actually recognized different pools of SM (241). Finally, radioactive SMS substrate precursors, such as [3H] choline, are also utilized to monitor perturbations in SMS activity (47, 75, 119, 206) in cells, though newer stable isotope assays present less hazardous substrates. Methyl[14C]choline is typically utilized at 0.5–2μCi/ml for 4–5 hours for pulse conditions (206, 221) and takes between 24–60 hours to reach steady state labeling of SM (4, 119).
As there are differences in sub-cellular localization of SMS isoforms and distinct regulation of SM pools, inhibitors that could distinguish these activities would be highly desirable. The initial inhibitor discovered for SMS was the anti-viral reagent D609 (119), that exhibited an IC50 of 375 μM towards SMS1 and was less effective at inhibiting SMS2. It has been utilized in multiple cell types at a concentration of 20–25ug/ml for 2–6 h (Fig 3)(119, 125, 169). However, particular care must be taken with stocks as the xanthate group of D609 can be readily oxidized (t1/2 is less than 20 min in saline conditions at 24 °C (11)), causing a loss of biological activity. A more stable prodrug (methyleneoxybutyryl D609)) has been described (11). More potent and specific inhibitors of the isozymes activity have recently been developed, including D-Series inhibitors, SAPAs, and 2-Quinolone derivative (1, 45, 109). Li et al. reported on the 2-(4-(N-phenethylsulfamoyl)phenoxy)acetamides (SAPAs) molecules, one of which had a Km of 2.1 μM for SMS1 (109), a 100 fold more potent than D609. Also, greater specificity was achieved by the Kawamoto group who recently developed a 2-Quinolone derivative that has an IC50 in the nM range with over 100 fold selectivity for SMS2, potentially providing a much more effective tool for the interrogation of SMS activity (1). However these inhibitors are not suitable for cellular studies.
3.6. Glucosylceramide synthase - probes and inhibitors
While SM synthesis represents an “anabolic dead end”, the generation of GluCer, through glucosylceramide synthase (GCS) activity, is a stepping stone to the vastly complex family of glycosphingolipids. In this complicated arm of sphingolipid metabolism, utilizing molecular probes is the most effective way to track metabolic changes. Typical tracer experiments in cells utilize C6-NBD-Cer as a direct substrate for GCS at 1–100 μM for 30 min to 3 h and analyze the products using TLC/fluorimetric analysis (Fig 2) (29, 47, 96, 114, 122, 189, 231). Fluorescent molecules have been extensively utilized in the study of vesicular trafficking of newly synthesized complex sphingolipids. In an early study, Babia et al. demonstrated distinct NBD-GluCer or NBD-SM containing vesicles in HT-29 cells labeled with C6-NBD-Cer (9). An important consideration in the analysis of GCS flux is GalCer, an isomer of the GCS product, which can often be difficult to separate with HPLC and fluorescent detection techniques. As tissue distribution between these glycosphingolipid isomers is highly distinct, it is sometimes possible to deduce GCS activity in tissues with very low/absent formation of GalCer. Though to circumvent this issue, radioactive UDP-glucose and UDP-galactose have also been utilized in many studies. Brenkert et al. defined the effects of age on patterns of Cer glycosylation in rat brain lysate utilizing either glucose or galactose tracers to separate the metabolism of Cer into GluCer or GalCer isomers (33). It is also possible to separate these isomers utilizing borate impregnated HPTLC plates, as was demonstrated by Gupta et al. with a 2 h incubation of 500 μM C6-NBD-Cer in a breast cancer cell line (64).
Traditionally, inhibitors for glycosyltransferases are few, but GCS is an exception, with inhibitors generated from analogs of both substrates, UDPglucose and Cer. 1-phenyl-2-decanoylamino-3-morpholino-propanol (PDMP) is a ceramide analog which contains an aromatic ring and tetracyclic system in place of the alkyl chain and sphingoid base, respectively. PDMP is a mixed-type inhibitor that is typically used in situ at IC10 values (50 μM) (Fig 3) (14, 29, 56). Improved derivatives of PDMP include P4, a compound with an IC50 of 90 nM towards the inhibition of ceramide glycosylation by GCS in vitro (106). Recently C10, an analog of the potent drug eliglustat (used in treatment of Gaucher’s disease) has been utilized for the inhibition of GCS in situ (IC50 in low nanomolar range), and of particular importance, appears to have less off target effects then PDMP (unpublished data and (187, 188)). Subathra et al. utilized 0.15 μM C10 in mesangial cells for 48 h, decreasing GCS activity by 90% (201). As a counterpart to the ceramide analogs, immunosugar derivatives of UDPGlu have also been utilized to inhibit GCS, in particular deoxynojirimycin (DNJ) analogs, some of which have low μM IC50s (29, 41, 158, 159). There is the possibility of immunosugars having off-target effects throughout the cell and on other steps in glycosphingolipid metabolism, therefore when probing the initial step in this metabolic pathway, Cer analogs may present a better option.
3.6. Ceramide kinase - probes and inhibitors
Ceramide kinase (CERK) has been implicated in several cellular processes, including survival and proliferation (58) as well as apoptosis and inflammatory responses (60). C6-NBDCer has been employed as a substrate for probing CERK activity in vitro with a reported Km of 4.0 μM (224) when utilized with purified enzyme. Pettus et al. utilized 20μM C6-NBD-Cer for 2 h in A549 lung adenocarcinoma cells to demonstrate cytokine activation of CERK (154). More recently Matsuzaki et al. were able to take advantage of the increased sensitivity afforded by technical advances in chromatography and detectors to create a CERK activity assay utilizing 30 min treatment of 10 μM C6-NBD-Cer in HEPG2 cells (Fig 2) (122). Of note, due to the large range in NBD metabolite concentrations, absolute quantification was difficult, and results were expressed as a percentage of the total cellular concentration of the NBD metabolites. Matsuzaki also noted the additional polarity offered by the phosphorylation to the already less hydrophobic NBD labeled metabolite can affect partitioning during extraction; therefore, single phase extractions (48) or a solid phase extraction (224) should be considered to achieve optimal recovery. Fluorescent C5- BODIPY-Cer has also been utilized to study C1P trafficking, though at lower concentrations than its NBD counterpart (5 μM for 30–45min in COS-1 cells) (28). Radioactive phosphate tracers ([32P] orthophosphate) are also popular and typically used at 0.1–10 mCi/ml for 2–15 h in cells (60, 222–224).
There are few inhibitors for CERK activity. It has been noted that some SK inhibitors modestly inhibit CERK activity, though treatments with the SK inhibitors MP-A08 and F-12509A were in the 100’s of μM range to achieve only a 50% inhibition of CERK (155, 203). The inhibitor NVP-231 is highly specific and has an IC50 value of 12 nM for CERK activity, with an observed Ki of 7.4 nM (59). NVP-231 provided a 90% inhibition of CERK activity when utilized in cells at a concentration of 100 nM for 6 h (Fig 3), and increasing the concentration to 1 μM or increasing the time to 24 provided no significant improvement in its inhibitory effect (59). While there are few inhibitors for CERK, NVP-231 represents a potent option for the interrogation of CERK signaling potential.
3.7. Probing de novo flux
As mentioned above, many of the fluorescent products used to measure enzymatic activity in cells are recognized and further metabolized by sphingolipid enzymes. Therefore, if a comprehensive approach is taken to analyzing de novo sphingolipid synthesis, the metabolic fate of probes can provide insight into the activity of multiple enzymes. While exogenous treatments with C6-NBD-Cer have been extensively utilized to probe sphingolipid metabolism mostly at the Golgi (SMS, CERK, GCS) (112), there is still some contention as to the confounding effects that an omega positioned NBD fluorophore and short N-acyl chain length have on the recognition of this molecule versus endogenous sphingolipids by Golgi enzymes of sphingolipid metabolism. Thus, the utilization of a d17 sphingoid backbone or isotopically labeled precursor may produce the most faithful results to indicate regulation of endogenous sphingolipid metabolic flux. Indeed, a method was recently developed by Snider et at. utilizing mass spectrometry to monitor temporally distinct phases for the incorporation of a d17dhsph pulse into d17dhCer, d17Cer, d17SM, d17HexCer, d17Sph, and d17dhSph 1-phosphate, allowing for the dissection of enzymatic activities for CerS, DeS, SMS, GCS, CDases, and SK1, respectively (195). This method offers a much more comprehensive insight into de novo sphingolipid flux than those using fluorescent probes, as mass spectrometry can follow the incorporation of the d17dhsph probe into the myriad of sphingolipid molecular species that can be generated in the ER and Golgi. Additionally, the utilization of a dihydro sphingoid base as a probe allows for accurate monitoring of ceramidase activity, which occurs to a much greater extent on Cer molecules (as opposed to dhcer), thereby producing a different molecule (d17Sph) from the initial probe (d17dhSph) (195). In such a dynamic network, these methods for probing entire sections of sphingolipid metabolism will play a crucial role in moving forward the understanding of sphingolipid metabolism and transport.
4. Sphingolipid Turnover and Clearance
4.1. Sphingomyelinases - probes and inhibitors
SM is the most abundant sphingolipid at the PM, but it is also found in several other subcellular membranes. Therefore, hydrolysis of these SM pools provides an efficient means for cells to generate Cer quickly and in a compartment-specific manner. Consequently, the SMases play an important role in signaling for Cer-driven biologies, including apoptosis (3), growth arrest (185), and autophagy (152). As pH dependent activity has been observed, these enzymes are separated into 3 categories, acid (aSMase), neutral (nSMases), and alkaline. The radioactive substrate [choline-methyl-14C]-SM and fluorescent probes have been utilized for in vitro enzyme activity assays (116), and changing the pH from 4.0 to 7.4 (as well as the addition of Mg2+) will change the assay specificity from aSMase to nSMase activity. The C6-NBD-SM substrate is typically utilized in cells through direct addition to media at 2–4 μM for 30–60min to probe SMase activity (Fig 2) (96, 117). PM isolations (96) and back exchange conditions (0–4 degrees with BSA) (246) have been utilized to optimally assess SMase activity on fluorescent probes in a cellular context. Utilizing back exchange conditions, Hao et al. were able to establish two routes for C6-NBD-SM recycling at the PM, one with a label halftime of 1.5 min and one with a slower recycling halftime of 12 min (74). Further, utilizing a lipophilic dye (FM) that cannot flip-flop across the lipid bilayers, they interrogated endosomal fusion and were able to verify the hypothesis that both of the recycling pools were indeed internalized and included passage through early endosomes (74). Altering the temperature in the back exchange protocol was utilized to demonstrate defects in SM degradation in Niemann-pick type-A fibroblasts (NPA) that allowed SM to accumulate in the lysosomes. Liposomes containing 25 μM NBD-SM were initially loaded into the PM of cells at 7 degrees, with similar rates of internalization between normal and NP-A cells, but, as temperature and time were increased, normal cells demonstrated increased fluorescence in the Golgi apparatus while NBD-SM accumulated in the lysosomes in NP-A cells. Therefore, NBD-SM can be utilized to monitor flux through multiple components of SM degradation based on its transport between intercellular compartments (99).
For evaluation of cellular activity of SMases, steady state labeling of SM substrate can be reached within 16–48 h depending on the cell line and substrate utilized (e.g choline or palmitate)(2, 4, 50). Depletion of labeled SM then provides insight into SMase activity. To distinguish SM pools that where sensitive to hydrolysis in response to TNFa signaling, Levade’s group employed [methyl-3H]choline at 2μCi for 48h in conjunction with exogenous SMase treatments(2). Demonstrating that pools of SM generated after TNFa treatment were not accessible to exogenous SMase, highlighting a multipronged approach to interrogate SMase flux. There is a plethora of inhibitors for the SMases that can be employed to evaluate activation and roles of specific SMases. Many of these inhibitors are structurally unrelated to SM, including desipramine, SR33557, NB6, C11AG, and GW4869, though some are rather unselective. Of these, GW4869 is a selective noncompetitive inhibitor with an IC50 of 1 μM for nSMase activity, with no effect on aSmase (120). This inhibitor is typically utilized in cells at 10–20 μM (120, 213). Recently, 2,6-Dimethoxy-4-(5-Phenyl-4-Thiophen-2-yl-1H-Imidazol-2-yl)-Phenol (DPTIP), was identified as a potent inhibitor of nSMase2 through a high throughput screen. DPTIT demonstrated better solubility then previous nSMase inhibitors which contributed to its ability to cross the blood-brain barrier (171). aSMase is commonly inhibited by desipramine and related tricyclic compounds by disrupting aSMase binding to lipid bilayers in the lysosome, thereby inducing proteolytic degradation of the enzyme (83, 98). This class of compounds has been utilized in cells, normally at 25 μM for 1–2 h (Fig 3)(83, 89). However, these molecules also inhibit acid CDase (245) and probably have additional effects on lysosomal proteins. It has been demonstrated that both aSMase and nSMase can also be inhibited by cellular metabolites. Testai et al. observed non-competitive inhibition of aSMase with phosphatidyl-myo-inositol 3,4,5-triphosphate [PtdIns (3,4,5)P] (83), with a IC50 of 3 μM for Cer generation by SMase activity (214). Scaphostatin, one of the original molecules used to inhibit SMase activity, still remains one of the most potent inhibitors available for nSmase activity with an IC50 value of 1 μM for nSMase and 49.7 μM for aSMase (137). At 5mM, glutathione exhibited greater than 95% inhibition of nMSase in vitro (due to changes in buffer pH), but also exhibited inhibitory effects at 10mM in human airway epithelial cells and in hepatocytes (107, 113, 173). As these molecules are pivotal players in cellular metabolism, they cannot be utilized as pharmacological inhibitors, but they may constitute a means for cellular regulation of these enzymes.
4.2. Glucosylcerebrosidases - probes and inhibitors
The generation and catabolism of the structurally diverse complex glycosphingolipids is regulated by many enzymes, though the final degradation and clearance of the majority of these species (those based on GluCer) funnels down to the catabolic action of Glucosylcerebrosidases (GCases). The majority of GCases action occurs in the lysosomes through GBA1 activity, though hydrolysis of GluCer at the PM has been documented via the membrane-bound GBA2 (30, 244). Defective lysosomal activity of GBA1 results in toxic accumulations of GluCer and glucosylsphingosine that have been linked to the pathogenesis of Gaucher’s disease, an inherited disorder (32). Multiple NBD derivatives of the substrate have been utilized in the literature, including C6, C12 and C16-NBD-GluCer, indicating that GCase has no specificity for N-acyl chain length. In cells, C6-NBD-GluCer is typically utilized between 5–50μM for 0.5–3 h (Fig 2). Non-lysosomal GBA2 activity was initially tested in COS-7 cells using multiple florescent probes, including both C6-NBD-GluCer and 4-methylumbelliferyl-β-gluco-side. In combination with GCase and lysosomal inhibitors these experiments were able to take advantage of the different localization of the GCase enzymes to tease their activities apart (30). Most studies of these enzymes focus on GBA1 activity as it pertains to Gaucher’s disease. Indeed, both NBD and BODIPY Cer analogs along with 2-Hexadecanoylamino-4-nitrophenyl β-D-glucopyranoside and 1-O-Glucosyl-2-N-(dimethylaminonaphthalene-5-sulfonyl) sphingosine have been utilized as in vitro fluorescent substrates to diagnose Gaucher’s disease (51, 131). However, a major issue when utilizing these fluorescent molecules in cells, is that flux through GBA1 is confounded by exogenous substrate degradation at the PM through GBA2 activity, as well as apparent activity occurring in the cytosol, perhaps through a neutral GCase (76).
Multiple groups have published crystal structures of GCase with different inhibitors bound to the active site, such as the imminosugars isofagomine and DNJ analogues, which have led to structural modifications of the compounds to increase potency (34, 110). Specifically, modifications to DNJ structure have led to more potent inhibitors, such as the N-(5′-adamantane-1′-yl-methoxy)-pentyl-1-deoxynojirimycin (AMP-DNM) compound, which is a hydrophobic derivative of DNJ. AMP-DNM also displayed increased selectivity with an IC50 of 1 nM for GBA2 activity, compared to a 200 nM IC50 for GBA1 (238). Of note, this inhibitor can also antagonize GCS activity at an IC50 of 25 nM (238). Towards the inhibition of GBA1, modifications to Isofagomine have led to the development of an N-alkylated group of Isofagomine derivatives, of which the longest hydrocarbon chain (nonyl) was noted to have an IC50 of 0.6 nM for GBA1 activity (248). Therefore, extremely effective inhibitors are available for the distinction of GluCer hydrolysis by GBA1 versus GBA2.
4.3. Ceramidases - probes and inhibitors
As the first step in the clearance of Cer from cellular pools, ceramidases (CDases) represent a crucial regulatory point in sphingolipid metabolism. The five CDase enzymes are characterized by their pH optimum for activity, acid (aCDase), neutral (nCDase) and three alkaline CDases (ACERs), and by their specific subcellular localization and substrate specificity (39). Thus, analyzing these enzymes in cells with a single probe can be difficult. Initial studies utilized radio tracers ([3H] or [14C] labeled ceramide) to follow production of the radioactive fatty acid from Cer hydrolysis via CDase activity (132, 243). Fluorescent assays have utilized C6-NBD-Cer at 6 μM for 12 h (140), but it was realized that C12-NBD-Cer was more effectively processed by ACERs & nCDase (211). In a comprehensive in vitro study performed by Tani et al. utilizing enzymes from each of the pH groups with C12-NBD-Cer at concentrations between 5 and 50 μM for 0.5 to 3 h (Fig 2), Kms of 22, 59, and 111 μM for nCDase, aCDase, and ACER were measured, respectively (211). In another study by the same group, C12-NBD-Cer and C6-NBD-Cer were utilized in B16 melanoma cells at a concentration of 5μM for 1 h to demonstrate CDase preference for the longer chain length in situ (209). It has also been noted that BODIPY derivatives of Cer are more effectively hydrolyzed by aCDase (78). Bhabak et al. developed a FRET compatible Cer analog combining NBD and Nile Red fluorophores into (NBD)C12CerC7(NR). In vitro, the probe demonstrated Km values of 182 μM and 142 μM for aCDase and nCDase, respectively (19). After a 60 min incubation in HeLa cells, the probe localized to the Golgi, though no cleavage of the probe was observed (19). Optimization of these FRET compounds is ongoing (20). A high-throughput assay has been developed by Fabrias and coworkers that employs a coumarin dye attached to the Cer substrate. The hydrolytic CDase activity releases the amino diol group, which is subsequently chemically oxidized by periodate, generating the fluorescent molecule umbelliferone (17). Finally, reverse CDase activity, which is FB1 insensitive, has also been evaluated in lymphoid cells after a 2 h treatment utilizing 20μM C12-NBD-fatty acid and 10μM Sph to produce fluorescent Cer in conditions were CerS activity is inhibited (145).
CDases have been targeted by multiple inhibitors with ongoing development in this area of research. The initial inhibitor of CDase activity was N-Oleoylethanolamide (NOE), which took advantage of structural and stereochemical modifications to directly inhibit the CDases, though it suffered from poor selectivity (198) and low efficacy (Ki of 500 μM) (61). ACERs can be inhibited with the ceramidase inhibitor D-erythro-MAPP ((1S,2R)-N-myristoylaminophenylpropanol-1) which has an IC50 of approximately 1–5μM in vitro and can induce growth arrest in HL-60 cells in a dose and time dependent manner (22). Inhibition of aCDase has also been pursued, and D-e-MAPP served as the basis for subsequent work that developed B-13 (1R,2R)-N-myristoylamino-4’-nitro-phenylpropandiol-1,3) analogs (12, 20). As ceramidases are localized to specific subcellular compartments, inhibition of these enzymes should ideally be achieved by targeting these specific compartments. As an illustration, second and third generation B13 analogs targeting the lysosome were developed. LCL204 inhibited aCDase with IC50 ~40μM for MCF-7 cell growth (21). More recently LCL521 was developed as a prodrug that is deacylated to B13 in the lysosome (10). This compound exhibited the most potent effects in cells, but because of its design as a prodrug, it showed little effects in vitro. The tricyclic anti-depressant desipramine has also been found to inhibit aCDase, likely by the same indirect lysosomal mechanism by which it inhibits aSMase (245). Finally, Carmofur, a 5-fluorouracil-releasing drug, has demonstrated greater potency (IC50 of 29 nM) and specificity then other currently available aCDase inhibitors, both in vitro and in vivo (166).
Recently molecules that are structurally unrelated to Cer have been employed as inhibitors. The most potent and stable of these are a class of benzoxazolone carboxamides that covalently bind to cysteine-143 in the catalytic site of aCDase (157). In particular, Compound 17a demonstrated an IC50 of 79 nM in vitro, which remained below 1 μM when examined in both human and mouse cells (156). Importantly, this class of inhibitors is very efficient as they covalently bind to the enzyme, and inhibition occurs in a time dependent manner.
Inhibition of nCDase has recently been achieved with the ceramide analog C6 ureaceramide. This competitive inhibitor of nCDase was developed using structure-activity analysis (142, 220), and has been shown to maximally inhibit nCDase at 25 μM in cells with treatments up to 24 h (Fig 3) (54). This inhibitor did not modulate ceramide levels in epithelial fibroblasts derived from nCDase knock out mice, thus demonstrating its selectivity among the ceramidases (54). Taken together these inhibitors offer a comprehensive array of tools to probe CDase activity throughout the cell.
4.4. Sphingosine Kinases - probes and inhibitors
Two known isoforms of SK, SK1 and SK2, are responsible for the production of S1P from Sph. This bioactive lipid has been implicated in numerous biologies mostly due to its activation of one of five G protein-coupled receptors (S1PRs1–5) (95), but additional intracellular targets have been proposed (200, 216). SK activity and S1P generation have primarily been probed through the use of radioactive substrates, though fluorescent and odd chain sphingoid bases are becoming more popular. Initial studies utilized radiolabeled ATP as a substrate in vitro and radioactive phosphate in cells (to label cellular ATP) often between 1–5 uCi for up to 1 h (147, 192). Though these assays are highly accurate, they are also labor intensive and not high throughput. Safer and faster alternatives have come from the use of odd chain sphingoid bases. Similar to the measurement of CerS activity, 17CSph is added to cells to assess SK activity at concentrations between 0.5 and 2μM for 15–30 min (Fig 2), as 17CS1P is rapidly generated in response to this exogenous substrate (52, 162). This accurately assesses SK activity and requires MS for detection. However, fluorometric detection of NBD-Sph lends itself to high-throughput assays for the screening of potential SK inhibitors, and indeed this is how it has been employed. Lima et al. developed a 384 well-plate format assay to monitor the phosphorylation of NBD-sphingosine by SK activity, owed to an increase in emission once the probe has been phosphorylated (111).
Numerous inhibitors of SK have been utilized in the literature, and many of them (mostly those mimicking the substrate Sph) also result in proteosomal degradation of the protein in the cell (53). Classically, inhibition of SKs was achieved with dimethylsphingosine (DMS), which acts as a competitive inhibitor of SK1 (Ki=5μM) and a non-competitive inhibitor of SK2 (Ki=12μM) (163, 227). SKI-I, with a Ki of 10μM for SK1 has been used to inhibit growth of leukemia cells (149). SKI-II is perhaps the best characterized, but it is rather non-specific and has a Ki of ~17μM (148). Most of these inhibitors suffer from lack of specificity for SK1 vs SK2 and have significant off target effects. In particular, SKI-II has recently been shown to inhibit DeS1, a component of de novo sphingolipid synthesis (8, 118, 155). The recent identification of the SK1 structure has aided the development of the potent SK1 specific inhibitor PF543 with a Ki of 3.6 nM (179). Later this molecule was co-crystallized with SK1 and led to insights regarding its 130-fold difference in selectivity for SK1 over SK2 (234). Importantly, many of the studies with PF-543 effects on cell proliferation have utilized much higher concentrations of PF-543 (10 and 25μM) which are several fold higher than the Ki (Fig 3) (66, 91). Inhibition of SK1 is a promising therapeutic strategy given the important biologies associated with S1P, but it requires further study and development.
4.5. Sphingosine-1-Phosphate Lyase - probes and inhibitors
The gatekeeper to clearance of metabolites from sphingolipid metabolism is sphingosine-1-phosphate lyase (SPL). Regulation of SPL activity is partially responsible for flux through S1P and has been correlated with resistance to specific anticancer therapies (40, 138, 229). The observation of defects in lymphocytic egress from lymph nodes in SPL knockout mice has increased interest in SPL as a druggable target to suppress the immune response (181, 229). Fluorescent probes have been developed that allow rapid assays for S1PL activity. Bandhuyula et al. developed an in vitro assay for the analysis of SPL activity utilizing 5 nmol per sample of a NBD-S-1-P molecule for 15 min. A Bodipy probe has also been developed, though with a slightly higher Km of 35 μM compared to the NBD derivative (14.6 μM) (13). Very recently, the fluorescent substrate RBM148 was used and effectively added to intact cells to probe S1P metabolism. While it is an exciting step forward in the analysis of this critical enzyme, the authors did utilize RBM148 at a seemingly high concentration of 580 μM and had to encapsulate it in anionic liposomes for delivery (177). SPL inhibition has primarily been explored in vivo. The SPL inhibitor 2-acetyl-4-tetrahydroxybutyl imidazole (THI) is a component of caramel food coloring and induces lymphopenia by altering the S1P gradient in vivo (57). Though there has been controversy over the mechanism of action of THI, Ohtoyo et.al argued that this might be due to the fact that previously attempted in vitro SPL assays did not adequately represent SPL activity because they lacked pyridoxal 5’-phosphate, a key factor in SPL activity (144). Another potent SPL inhibitor is the sphingolipid analog 2-vinylsphinganine-1-phosphate (2VS1P) which has an IC50 of 2.4 μM (31). In a comprehensive approach to develop SPL inhibitors, Billich et al screened over 200,000 compounds, which eventually resulted in the development of compounds C and D, with IC50s of 0.21 and 0.024 μM, respectively (27, 115, 237). Compound C was later co-crystallized with SPL, revealing structural details about fitment in the substrate binding site (42). The pyridoxal 5′-phosphate analog 4′-deoxypyridoxine (DOP) is a nonspecific inhibitor of SPL. As DOP also modulates the activity of multiple enzymes involved in DNA synthesis, it can be highly toxic over time (26). Endogenous activation of SPL may also prove to be a more useful therapeutic target, instead of inhibition, as activation of this enzyme would clear S1P from the sphingolipid network and could be used to augment apoptosis in response to chemotherapy.
5. Unresolved Questions and General Considerations
In the last two decades, the field of sphingolipidomics has extensively moved forward, with improving detection of lipid species, sensitivity, and methods for probing metabolism. However, there are several aspects of sphingolipid analysis that could still benefit from improvements. The use of NBD probes for fluorescent detection of sphingolipid metabolites can affect the biophysical properties of downstream metabolites, as the label is bulky and typically located on the omega carbon of the N-acyl chain. Indeed, the need to adapt extraction procedures for the analysis of the much less hydrophobic C6-NBD-C1P molecule has been documented (122). This large change in hydrophobicity may also have metabolic consequences, potentially providing inaccurate flux information. While radioactive probes for assessing flux are more hazardous, they do constitute/convert into virtually identical molecules as endogenous sphingolipids, yet TLC separation must be utilized instead of HPLC. All this considered, radioactive assays typically used for the detection of flux in sphingolipid metabolites are being replaced thanks to the increased number and improved detection of stable isotope labeled sphingolipids. However, analysis of stable isotope probes can require extensive bioinformatic strategies due to mass overlap of probes that fall within the isotopic distribution window of the corresponding sphingolipid (247). Finally, the recent development of probes that utilize click chemistry to visualize sphingolipid flux presents an exciting opportunity to add spatial context to sphingolipid flux; however, quantitation with fluorescent microscopy is still under development (65).
When probing sphingolipid metabolism, it is also important to consider the potential biological effects of the probes even at low concentrations. Many of the probes that enter sphingolipid metabolism after the SPT reaction are bioactive molecules. However, because of detection limits of current technologies, they are rarely used at truly trace levels. This becomes particularly important when probing metabolites with receptors, such as flux in SK activity, where the addition of exogenous substrate provides a large generation of S1P proximal to the S1P receptors at the PM.
6. Conclusion
Advancements in the understanding of the structural specificity involved in sphingolipid metabolism have led to the development of many complimentary molecular tools for the interrogation of sphingolipid flux and mass. Moreover, this has led to the development of inhibitors and molecular probes for nearly every step central to sphingolipid generation and clearance. In the future, labeling techniques that can define regulation of distinct pools of lipids, combined with genetic manipulation of distinctly localized enzymes will hopefully add some much needed clarity to the bioactive roles of the “sphinx” of lipids.
Highlights:
Sphingolipids metabolism is complex and thus requires specific labeling and inhibitor strategies to probe
The combination of inhibitors and molecular probes for in-cell assays allows for the most authentic representation of sphingolipid regulation
Overview of commonly utilized techniques for the analysis of sphingolipid metabolism, along with parameters on the use of labels and inhibitors to probe sphingolipid homeostasis in mammalian cell lines
Acknowledgements
This work was supported by NIH grant R35 GM118128 to YAH and by NCI grant P01-CA97132 to YAH (project#1) and project #4 to CL. The authors declare no conflicts of interest.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Adachi R, Ogawa K, Matsumoto SI, Satou T, Tanaka Y, Sakamoto J, Nakahata T, Okamoto R, Kamaura M, and Kawamoto T. Discovery and characterization of selective human sphingomyelin synthase 2 inhibitors. Eur J Med Chem 136: 283–293, 2017. [DOI] [PubMed] [Google Scholar]
- 2.Andrieu-Abadie N, Carpentier S, Salvayre R, and Levade T. The tumour necrosis factor-sensitive pool of sphingomyelin is resynthesized in a distinct compartment of the plasma membrane. Biochem J 333 (Pt 1): 91–97, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Andrieu-Abadie N, and Levade T. Sphingomyelin hydrolysis during apoptosis. Biochimica et biophysica acta 1585: 126–134, 2002. [DOI] [PubMed] [Google Scholar]
- 4.Andrieu N, Salvayre R, and Levade T. Evidence against involvement of the acid lysosomal sphingomyelinase in the tumor-necrosis-factor- and interleukin-1-induced sphingomyelin cycle and cell proliferation in human fibroblasts. Biochem J 303 (Pt 2): 341–345, 1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Apraiz A, Idkowiak-Baldys J, Nieto-Rementeria N, Boyano MD, Hannun YA, and Asumendi A. Dihydroceramide accumulation and reactive oxygen species are distinct and nonessential events in 4-HPR-mediated leukemia cell death. Biochem Cell Biol 90: 209–223, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Apraiz A, Idkowiak-Baldys JK, Boyano MD, Perez-Yarza G, Hannun YA, and Asumendi A. Evaluation of bioactive sphingolipids in 4-HPR-resistant leukemia cells. BMC Cancer 11: 477, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Asano S, Kitatani K, Taniguchi M, Hashimoto M, Zama K, Mitsutake S, Igarashi Y, Takeya H, Kigawa J, Hayashi A, Umehara H, and Okazaki T. Regulation of cell migration by sphingomyelin synthases: sphingomyelin in lipid rafts decreases responsiveness to signaling by the CXCL12/CXCR4 pathway. Mol Cell Biol 32: 3242–3252, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Aurelio L, Scullino CV, Pitman MR, Sexton A, Oliver V, Davies L, Rebello RJ, Furic L, Creek DJ, Pitson SM, and Flynn BL. From Sphingosine Kinase to Dihydroceramide Desaturase: A Structure-Activity Relationship (SAR) Study of the Enzyme Inhibitory and Anticancer Activity of 4-((4-(4-Chlorophenyl)thiazol-2-yl)amino)phenol (SKI-II). J Med Chem 59: 965–984, 2016. [DOI] [PubMed] [Google Scholar]
- 9.Babia T, Kok JW, van der Haar M, Kalicharan R, and Hoekstra D. Transport of biosynthetic sphingolipids from Golgi to plasma membrane in HT29 cells: involvement of different carrier vesicle populations. Eur J Cell Biol 63: 172–181, 1994. [PubMed] [Google Scholar]
- 10.Bai A, Mao C, Jenkins RW, Szulc ZM, Bielawska A, and Hannun YA. Anticancer actions of lysosomally targeted inhibitor, LCL521, of acid ceramidase. PLoS One 12: e0177805, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bai A, Meier GP, Wang Y, Luberto C, Hannun YA, and Zhou D. Prodrug modification increases potassium tricyclo[5.2.1.0(2,6)]-decan-8-yl dithiocarbonate (D609) chemical stability and cytotoxicity against U937 leukemia cells. J Pharmacol Exp Ther 309: 1051–1059, 2004. [DOI] [PubMed] [Google Scholar]
- 12.Bai A, Szulc ZM, Bielawski J, Mayroo N, Liu X, Norris J, Hannun YA, and Bielawska A. Synthesis and bioevaluation of omega-N-amino analogs of B13. Bioorg Med Chem 17: 1840–1848, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bandhuvula P, Fyrst H, and Saba JD. A rapid fluorescence assay for sphingosine-1-phosphate lyase enzyme activity. J Lipid Res 48: 2769–2778, 2007. [DOI] [PubMed] [Google Scholar]
- 14.Baran Y, Salas A, Senkal CE, Gunduz U, Bielawski J, Obeid LM, and Ogretmen B. Alterations of ceramide/sphingosine 1-phosphate rheostat involved in the regulation of resistance to imatinib-induced apoptosis in K562 human chronic myeloid leukemia cells. J Biol Chem 282: 10922–10934, 2007. [DOI] [PubMed] [Google Scholar]
- 15.Beauchamp E, Tekpli X, Marteil G, Lagadic-Gossmann D, Legrand P, and Rioux V. NMyristoylation targets dihydroceramide Delta4-desaturase 1 to mitochondria: partial involvement in the apoptotic effect of myristic acid. Biochimie 91: 1411–1419, 2009. [DOI] [PubMed] [Google Scholar]
- 16.Becker KP, Kitatani K, Idkowiak-Baldys J, Bielawski J, and Hannun YA. Selective inhibition of juxtanuclear translocation of protein kinase C betaII by a negative feedback mechanism involving ceramide formed from the salvage pathway. J Biol Chem 280: 2606–2612, 2005. [DOI] [PubMed] [Google Scholar]
- 17.Bedia C, Camacho L, Abad JL, Fabrias G, and Levade T. A simple fluorogenic method for determination of acid ceramidase activity and diagnosis of Farber disease. J Lipid Res 51: 3542–3547, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Berdyshev EV, Gorshkova I, Skobeleva A, Bittman R, Lu X, Dudek SM, Mirzapoiazova T, Garcia JG, and Natarajan V. FTY720 inhibits ceramide synthases and up-regulates dihydrosphingosine 1-phosphate formation in human lung endothelial cells. J Biol Chem 284: 5467–5477, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bhabak KP, Hauser A, Redmer S, Banhart S, Heuer D, and Arenz C. Development of a novel FRET probe for the real-time determination of ceramidase activity. Chembiochem 14: 1049–1052, 2013. [DOI] [PubMed] [Google Scholar]
- 20.Bhabak KP, Kleuser B, Huwiler A, and Arenz C. Effective inhibition of acid and neutral ceramidases by novel B-13 and LCL-464 analogues. Bioorg Med Chem 21: 874–882, 2013. [DOI] [PubMed] [Google Scholar]
- 21.Bielawska A, Bielawski J, Szulc ZM, Mayroo N, Liu X, Bai A, Elojeimy S, Rembiesa B, Pierce J, Norris JS, and Hannun YA. Novel analogs of D-e-MAPP and B13. Part 2: signature effects on bioactive sphingolipids. Bioorg Med Chem 16: 1032–1045, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Bielawska A, Greenberg MS, Perry D, Jayadev S, Shayman JA, McKay C, and Hannun YA. (1S,2R)-D-erythro-2-(N-myristoylamino)-1-phenyl-1-propanol as an inhibitor of ceramidase. J Biol Chem 271: 12646–12654, 1996. [DOI] [PubMed] [Google Scholar]
- 23.Bielawski J, Pierce JS, Snider J, Rembiesa B, Szulc ZM, and Bielawska A. Comprehensive quantitative analysis of bioactive sphingolipids by high-performance liquid chromatography-tandem mass spectrometry. Methods in molecular biology 579: 443–467, 2009. [DOI] [PubMed] [Google Scholar]
- 24.Bielawski J, Pierce JS, Snider J, Rembiesa B, Szulc ZM, and Bielawska A. Sphingolipid analysis by high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Adv Exp Med Biol 688: 46–59, 2010. [DOI] [PubMed] [Google Scholar]
- 25.Bilal F, Peres M, Le Faouder P, Dupuy A, Bertrand-Michel J, Andrieu-Abadie N, Levade T, Badran B, Daher A, and Segui B. Liquid Chromatography-High Resolution Mass Spectrometry Method to Study Sphingolipid Metabolism Changes in Response to CD95L. Methods Mol Biol 1557: 213–217, 2017. [DOI] [PubMed] [Google Scholar]
- 26.Billich A, and Baumruker T. Sphingolipid metabolizing enzymes as novel therapeutic targets. Subcell Biochem 49: 487–522, 2008. [DOI] [PubMed] [Google Scholar]
- 27.Billich A, Beerli C, Bergmann R, Bruns C, and Loetscher E. Cellular assay for the characterization of sphingosine-1-phosphate lyase inhibitors. Anal Biochem 434: 247–253, 2013. [DOI] [PubMed] [Google Scholar]
- 28.Boath A, Graf C, Lidome E, Ullrich T, Nussbaumer P, and Bornancin F. Regulation and traffic of ceramide 1-phosphate produced by ceramide kinase: comparative analysis to glucosylceramide and sphingomyelin. J Biol Chem 283: 8517–8526, 2008. [DOI] [PubMed] [Google Scholar]
- 29.Boldin S, and Futerman AH. Glucosylceramide synthesis is required for basic fibroblast growth factor and laminin to stimulate axonal growth. J Neurochem 68: 882–885, 1997. [DOI] [PubMed] [Google Scholar]
- 30.Boot RG, Verhoek M, Donker-Koopman W, Strijland A, van Marle J, Overkleeft HS, Wennekes T, and Aerts JM. Identification of the non-lysosomal glucosylceramidase as beta-glucosidase 2. J Biol Chem 282: 1305–1312, 2007. [DOI] [PubMed] [Google Scholar]
- 31.Aea Boumendjel. Synthesis of an inhibitor of Sphingosine- 1-phosphate lyase Tetrahedron lett 35: 819–822, 1994. [Google Scholar]
- 32.Brady RO, Kanfer JN, and Shapiro D. Metabolism of Glucocerebrosides. Ii. Evidence of an Enzymatic Deficiency in Gaucher’s Disease. Biochem Biophys Res Commun 18: 221–225, 1965. [DOI] [PubMed] [Google Scholar]
- 33.Brenkert A, and Radin NS. Synthesis of galactosyl ceramide and glucosyl ceramide by rat brain: assay procedures and changes with age. Brain Res 36: 183–193, 1972. [DOI] [PubMed] [Google Scholar]
- 34.Brumshtein B, Greenblatt HM, Butters TD, Shaaltiel Y, Aviezer D, Silman I, Futerman AH, and Sussman JL. Crystal structures of complexes of N-butyl- and N-nonyl-deoxynojirimycin bound to acid beta-glucosidase: insights into the mechanism of chemical chaperone action in Gaucher disease. J Biol Chem 282: 29052–29058, 2007. [DOI] [PubMed] [Google Scholar]
- 35.Canals D, Roddy P, and Hannun YA. Protein phosphatase 1alpha mediates ceramide-induced ERM protein dephosphorylation: a novel mechanism independent of phosphatidylinositol 4, 5-biphosphate (PIP2) and myosin/ERM phosphatase. J Biol Chem 287: 10145–10155, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Canela N, Herrero P, Marine S, Nadal P, Ras MR, Rodriguez MA, and Arola L. Analytical methods in sphingolipidomics: Quantitative and profiling approaches in food analysis. J Chromatogr A 1428: 16–38, 2016. [DOI] [PubMed] [Google Scholar]
- 37.Casasampere M, Ordonez YF, Casas J, and Fabrias G. Dihydroceramide desaturase inhibitors induce autophagy via dihydroceramide-dependent and independent mechanisms. Biochimica et biophysica acta 1861: 264–275, 2017. [DOI] [PubMed] [Google Scholar]
- 38.Clarke CJ, Cloessner EA, Roddy PL, and Hannun YA. Neutral sphingomyelinase 2 (nSMase2) is the primary neutral sphingomyelinase isoform activated by tumour necrosis factor-alpha in MCF-7 cells. Biochem J 435: 381–390, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Coant N, Sakamoto W, Mao C, and Hannun YA. Ceramidases, roles in sphingolipid metabolism and in health and disease. Adv Biol Regul 63: 122–131, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Colie S, Van Veldhoven PP, Kedjouar B, Bedia C, Albinet V, Sorli SC, Garcia V, Djavaheri-Mergny M, Bauvy C, Codogno P, Levade T, and Andrieu-Abadie N. Disruption of sphingosine 1-phosphate lyase confers resistance to chemotherapy and promotes oncogenesis through Bcl-2/Bcl-xL upregulation. Cancer Res 69: 9346–9353, 2009. [DOI] [PubMed] [Google Scholar]
- 41.Compain P, and Martin OR. Design, synthesis and biological evaluation of iminosugar-based glycosyltransferase inhibitors. Curr Top Med Chem 3: 541–560, 2003. [DOI] [PubMed] [Google Scholar]
- 42.Cosconati S, and Novellino E. The first sphingosine 1-phosphate lyase inhibitors against multiple sclerosis: a successful drug discovery tale. J Med Chem 57: 5072–5073, 2014. [DOI] [PubMed] [Google Scholar]
- 43.Cowart LA, Szulc Z, Bielawska A, and Hannun YA. Structural determinants of sphingolipid recognition by commercially available anti-ceramide antibodies. J Lipid Res 43: 204–2048, 2002. [DOI] [PubMed] [Google Scholar]
- 44.Delgado A, Casas J, Llebaria A, Abad JL, and Fabrias G. Inhibitors of sphingolipid metabolism enzymes. Biochimica et biophysica acta 1758: 1957–1977, 2006. [DOI] [PubMed] [Google Scholar]
- 45.Deng X, Lin F, Zhang Y, Li Y, Zhou L, Lou B, Li Y, Dong J, Ding T, Jiang X, Wang R, and Ye D. Identification of small molecule sphingomyelin synthase inhibitors. Eur J Med Chem 73: 1–7, 2014. [DOI] [PubMed] [Google Scholar]
- 46.Desai K, Sullards MC, Allegood J, Wang E, Schmelz EM, Hartl M, Humpf HU, Liotta DC, Peng Q, and Merrill AH Jr. Fumonisins and fumonisin analogs as inhibitors of ceramide synthase and inducers of apoptosis. Biochimica et biophysica acta 1585: 188–192, 2002. [DOI] [PubMed] [Google Scholar]
- 47.Dolgachev V, Farooqui MS, Kulaeva OI, Tainsky MA, Nagy B, Hanada K, and Separovic D. De novo ceramide accumulation due to inhibition of its conversion to complex sphingolipids in apoptotic photosensitized cells. J Biol Chem 279: 23238–23249, 2004. [DOI] [PubMed] [Google Scholar]
- 48.Don AS, and Rosen H. A fluorescent plate reader assay for ceramide kinase. Anal Biochem 375: 265–271, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fuchs B, Suss R, Teuber K, Eibisch M, and Schiller J. Lipid analysis by thin-layer chromatography-a review of the current state. J Chromatogr A 1218: 2754–2774, 2011. [DOI] [PubMed] [Google Scholar]
- 50.Fukasawa M, Nishijima M, Itabe H, Takano T, and Hanada K. Reduction of sphingomyelin level without accumulation of ceramide in Chinese hamster ovary cells affects detergent-resistant membrane domains and enhances cellular cholesterol efflux to methyl-beta -cyclodextrin. J Biol Chem 275: 34028–34034, 2000. [DOI] [PubMed] [Google Scholar]
- 51.Gal AE, Pentchev PG, and Fash FJ. A novel chromogenic substrate for assaying glucocerebrosidase activity. Proc Soc Exp Biol Med 153: 363–366, 1976. [DOI] [PubMed] [Google Scholar]
- 52.Gandy KA, Canals D, Adada M, Wada M, Roddy P, Snider AJ, Hannun YA, and Obeid LM. Sphingosine 1-phosphate induces filopodia formation through S1PR2 activation of ERM proteins. Biochem J 449: 661–672, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gandy KA, and Obeid LM. Regulation of the sphingosine kinase/sphingosine 1-phosphate pathway. Handb Exp Pharmacol 275–303, 2013. [DOI] [PubMed] [Google Scholar]
- 54.Garcia-Barros M, Coant N, Kawamori T, Wada M, Snider AJ, Truman JP, Wu BX, Furuya H, Clarke CJ, Bialkowska AB, Ghaleb A, Yang VW, Obeid LM, and Hannun YA. Role of neutral ceramidase in colon cancer. FASEB J 30: 4159–4171, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Garcia-Ruiz C, Colell A, Mari M, Morales A, and Fernandez-Checa JC. Direct effect of ceramide on the mitochondrial electron transport chain leads to generation of reactive oxygen species. Role of mitochondrial glutathione. J Biol Chem 272: 11369–11377, 1997. [DOI] [PubMed] [Google Scholar]
- 56.Gencer EB, Ural AU, Avcu F, and Baran Y. A novel mechanism of dasatinib-induced apoptosis in chronic myeloid leukemia; ceramide synthase and ceramide clearance genes. Ann Hematol 90: 1265–1275, 2011. [DOI] [PubMed] [Google Scholar]
- 57.Gobin SJ, Legg RF, and Paine AJ. The role of lymphocyte production and migration in the lymphopenia caused by 2-acetyl-4-tetrahydroxybutyl imidazole. Int J Immunopharmacol 14: 687–697, 1992. [DOI] [PubMed] [Google Scholar]
- 58.Gomez-Munoz A, Kong JY, Parhar K, Wang SW, Gangoiti P, Gonzalez M, Eivemark S, Salh B, Duronio V, and Steinbrecher UP. Ceramide-1-phosphate promotes cell survival through activation of the phosphatidylinositol 3-kinase/protein kinase B pathway. FEBS Lett 579: 3744–3750, 2005. [DOI] [PubMed] [Google Scholar]
- 59.Graf C, Klumpp M, Habig M, Rovina P, Billich A, Baumruker T, Oberhauser B, and Bornancin F. Targeting ceramide metabolism with a potent and specific ceramide kinase inhibitor. Mol Pharmacol 74: 925–932, 2008. [DOI] [PubMed] [Google Scholar]
- 60.Graf C, Rovina P, Tauzin L, Schanzer A, and Bornancin F. Enhanced ceramide-induced apoptosis in ceramide kinase overexpressing cells. Biochem Biophys Res Commun 354: 309–314, 2007. [DOI] [PubMed] [Google Scholar]
- 61.Grijalvo S, Bedia C, Triola G, Casas J, Llebaria A, Teixido J, Rabal O, Levade T, Delgado A, and Fabrias G. Design, synthesis and activity as acid ceramidase inhibitors of 2-oxooctanoyl and N-oleoylethanolamine analogues. Chem Phys Lipids 144: 69–84, 2006. [DOI] [PubMed] [Google Scholar]
- 62.Gross RW, Jenkins CM, Yang J, Mancuso DJ, and Han X. Functional lipidomics: the roles of specialized lipids and lipid-protein interactions in modulating neuronal function. Prostaglandins Other Lipid Mediat 77: 52–64, 2005. [DOI] [PubMed] [Google Scholar]
- 63.Gu M, Kerwin JL, Watts JD, and Aebersold R. Ceramide profiling of complex lipid mixtures by electrospray ionization mass spectrometry. Anal Biochem 244: 347–356, 1997. [DOI] [PubMed] [Google Scholar]
- 64.Gupta V, Patwardhan GA, Zhang QJ, Cabot MC, Jazwinski SM, and Liu YY. Direct quantitative determination of ceramide glycosylation in vivo: a new approach to evaluate cellular enzyme activity of glucosylceramide synthase. Journal of lipid research 51: 866–874, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Haberkant P, Stein F, Hoglinger D, Gerl MJ, Brugger B, Van Veldhoven PP, Krijgsveld J, Gavin AC, and Schultz C. Bifunctional Sphingosine for Cell-Based Analysis of Protein-Sphingolipid Interactions. ACS Chem Biol 11: 222–230, 2016. [DOI] [PubMed] [Google Scholar]
- 66.Hamada M, Kameyama H, Iwai S, and Yura Y. Induction of autophagy by sphingosine kinase 1 inhibitor PF-543 in head and neck squamous cell carcinoma cells. Cell Death Discov 3: 17047, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Han X, Rozen S, Boyle SH, Hellegers C, Cheng H, Burke JR, Welsh-Bohmer KA, Doraiswamy PM, and Kaddurah-Daouk R. Metabolomics in early Alzheimer’s disease: identification of altered plasma sphingolipidome using shotgun lipidomics. PLoS One 6: e21643, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hanada K Serine palmitoyltransferase, a key enzyme of sphingolipid metabolism. Biochimica et biophysica acta 1632: 16–30, 2003. [DOI] [PubMed] [Google Scholar]
- 69.Hanada K, Hara T, Nishijima M, Kuge O, Dickson RC, and Nagiec MM. A mammalian homolog of the yeast LCB1 encodes a component of serine palmitoyltransferase, the enzyme catalyzing the first step in sphingolipid synthesis. J Biol Chem 272: 32108–32114, 1997. [DOI] [PubMed] [Google Scholar]
- 70.Hanada K, Nishijima M, Fujita T, and Kobayashi S. Specificity of inhibitors of serine palmitoyltransferase (SPT), a key enzyme in sphingolipid biosynthesis, in intact cells. A novel evaluation system using an SPT-defective mammalian cell mutant. Biochem Pharmacol 59: 1211–1216, 2000. [DOI] [PubMed] [Google Scholar]
- 71.Hannun YA, and Luberto C. Lipid metabolism: ceramide transfer protein adds a new dimension. Curr Biol 14: R163–165, 2004. [PubMed] [Google Scholar]
- 72.Hannun YA, and Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol 9: 139–150, 2008. [DOI] [PubMed] [Google Scholar]
- 73.Hannun YA, and Obeid LM. Sphingolipids and their metabolism in physiology and disease. Nat Rev Mol Cell Biol 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Hao M, and Maxfield FR. Characterization of rapid membrane internalization and recycling. J Biol Chem 275: 15279–15286, 2000. [DOI] [PubMed] [Google Scholar]
- 75.Hatch GM, and Vance DE. Stimulation of sphingomyelin biosynthesis by brefeldin A and sphingomyelin breakdown by okadaic acid treatment of rat hepatocytes. J Biol Chem 267: 12443–12451, 1992. [PubMed] [Google Scholar]
- 76.Hayashi Y, Okino N, Kakuta Y, Shikanai T, Tani M, Narimatsu H, and Ito M. Klotho-related protein is a novel cytosolic neutral beta-glycosylceramidase. J Biol Chem 282: 30889–30900, 2007. [DOI] [PubMed] [Google Scholar]
- 77.Haynes CA, Allegood JC, Wang EW, Kelly SL, Sullards MC, and Merrill AH Jr. Factors to consider in using [U-C]palmitate for analysis of sphingolipid biosynthesis by tandem mass spectrometry. J Lipid Res 52: 1583–1594, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.He X, Li CM, Park JH, Dagan A, Gatt S, and Schuchman EH. A fluorescence-based high-performance liquid chromatographic assay to determine acid ceramidase activity. Anal Biochem 274: 264–269, 1999. [DOI] [PubMed] [Google Scholar]
- 79.Heinrich M, Wickel M, Schneider-Brachert W, Sandberg C, Gahr J, Schwandner R, Weber T, Saftig P, Peters C, Brunner J, Kronke M, and Schutze S. Cathepsin D targeted by acid sphingomyelinase-derived ceramide. EMBO J 18: 5252–5263, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hernandez-Corbacho MJ, Canals D, Adada MM, Liu M, Senkal CE, Yi JK, Mao C, Luberto C, Hannun YA, and Obeid LM. Tumor Necrosis Factor-alpha (TNFalpha)-induced Ceramide Generation via Ceramide Synthases Regulates Loss of Focal Adhesion Kinase (FAK) and Programmed Cell Death. J Biol Chem 290: 25356–25373, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hu W, Bielawski J, Samad F, Merrill AH Jr., and Cowart LA. Palmitate increases sphingosine-1-phosphate in C2C12 myotubes via upregulation of sphingosine kinase message and activity. J Lipid Res 50: 1852–1862, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Huang S, Huang S, Wang X, Zhang Q, Liu J, and Leng Y. Downregulation of lipin-1 induces insulin resistance by increasing intracellular ceramide accumulation in C2C12 myotubes. Int J Biol Sci 13: 1–12, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hurwitz R, Ferlinz K, and Sandhoff K. The tricyclic antidepressant desipramine causes proteolytic degradation of lysosomal sphingomyelinase in human fibroblasts. Biol Chem Hoppe Seyler 375: 447–450, 1994. [DOI] [PubMed] [Google Scholar]
- 84.Idkowiak-Baldys J, Apraiz A, Li L, Rahmaniyan M, Clarke CJ, Kraveka JM, Asumendi A, and Hannun YA. Dihydroceramide desaturase activity is modulated by oxidative stress. Biochem J 427: 265–274, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Igarashi N, Okada T, Hayashi S, Fujita T, Jahangeer S, and Nakamura S. Sphingosine kinase 2 is a nuclear protein and inhibits DNA synthesis. J Biol Chem 278: 46832–46839, 2003. [DOI] [PubMed] [Google Scholar]
- 86.Iwamori M, Costello C, and Moser HW. Analysis and quantitation of free ceramide containing nonhydroxy and 2-hydroxy fatty acids, and phytosphingosine by high-performance liquid chromatography. J Lipid Res 20: 86–96, 1979. [PubMed] [Google Scholar]
- 87.Jarvis WD, Grant S, and Kolesnick RN. Ceramide and the induction of apoptosis. Clinical cancer research : an official journal of the American Association for Cancer Research 2: 1–6, 1996. [PubMed] [Google Scholar]
- 88.Jenkins RW, Canals D, and Hannun YA. Roles and regulation of secretory and lysosomal acid sphingomyelinase. Cell Signal 21: 836–846, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Jenkins RW, Idkowiak-Baldys J, Simbari F, Canals D, Roddy P, Riner CD, Clarke CJ, and Hannun YA. A novel mechanism of lysosomal acid sphingomyelinase maturation: requirement for carboxyl-terminal proteolytic processing. J Biol Chem 286: 3777–3788, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Johnson KR, Becker KP, Facchinetti MM, Hannun YA, and Obeid LM. PKC-dependent activation of sphingosine kinase 1 and translocation to the plasma membrane. Extracellular release of sphingosine-1-phosphate induced by phorbol 12-myristate 13-acetate (PMA). The Journal of biological chemistry 277: 35257–35262, 2002. [DOI] [PubMed] [Google Scholar]
- 91.Ju T, Gao D, and Fang ZY. Targeting colorectal cancer cells by a novel sphingosine kinase 1 inhibitor PF-543. Biochem Biophys Res Commun 470: 728–734, 2016. [DOI] [PubMed] [Google Scholar]
- 92.Kawase M, Watanabe M, Kondo T, Yabu T, Taguchi Y, Umehara H, Uchiyama T, Mizuno K, and Okazaki T. Increase of ceramide in adriamycin-induced HL-60 cell apoptosis: detection by a novel anti-ceramide antibody. Biochimica et biophysica acta 1584: 104–114, 2002. [DOI] [PubMed] [Google Scholar]
- 93.Kelleher JK, and Nickol GB. Isotopomer Spectral Analysis: Utilizing Nonlinear Models in Isotopic Flux Studies. Methods Enzymol 561: 303–330, 2015. [DOI] [PubMed] [Google Scholar]
- 94.Kerwin JL, Tuininga AR, and Ericsson LH. Identification of molecular species of glycerophospholipids and sphingomyelin using electrospray mass spectrometry. J Lipid Res 35: 1102–1114, 1994. [PubMed] [Google Scholar]
- 95.Kluk MJ, and Hla T. Signaling of sphingosine-1-phosphate via the S1P/EDG-family of G-protein-coupled receptors. Biochimica et biophysica acta 1582: 72–80, 2002. [DOI] [PubMed] [Google Scholar]
- 96.Kok JW, Babia T, Klappe K, and Hoekstra D. Fluorescent, short-chain 6-NBD-sphingomyelin, but not C6-NBD-glucosylceramide, is subject to extensive degradation in the plasma membrane: implications for signal transduction related to cell differentiation. Biochem J 309 (Pt 3): 905–912, 1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kok JW, Nikolova-Karakashian M, Klappe K, Alexander C, and Merrill AH Jr. Dihydroceramide biology. Structure-specific metabolism and intracellular localization. J Biol Chem 272: 21128–21136, 1997. [DOI] [PubMed] [Google Scholar]
- 98.Kolzer M, Werth N, and Sandhoff K. Interactions of acid sphingomyelinase and lipid bilayers in the presence of the tricyclic antidepressant desipramine. FEBS Lett 559: 96–98, 2004. [DOI] [PubMed] [Google Scholar]
- 99.Koval M, and Pagano RE. Sorting of an internalized plasma membrane lipid between recycling and degradative pathways in normal and Niemann-Pick, type A fibroblasts. J Cell Biol 111: 429–442, 1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kraveka JM, Li L, Szulc ZM, Bielawski J, Ogretmen B, Hannun YA, Obeid LM, and Bielawska A. Involvement of dihydroceramide desaturase in cell cycle progression in human neuroblastoma cells. J Biol Chem 282: 16718–16728, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Krebs S, Medugorac I, Rother S, Strasser K, and Forster M. A missense mutation in the 3-ketodihydrosphingosine reductase FVT1 as candidate causal mutation for bovine spinal muscular atrophy. Proc Natl Acad Sci U S A 104: 6746–6751, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Krishnamurthy K, Dasgupta S, and Bieberich E. Development and characterization of a novel anti-ceramide antibody. J Lipid Res 48: 968–975, 2007. [DOI] [PubMed] [Google Scholar]
- 103.Lahiri S, Lee H, Mesicek J, Fuks Z, Haimovitz-Friedman A, Kolesnick RN, and Futerman AH. Kinetic characterization of mammalian ceramide synthases: determination of K(m) values towards sphinganine. FEBS Lett 581: 5289–5294, 2007. [DOI] [PubMed] [Google Scholar]
- 104.Lee AY, Lee JW, Kim JE, Mock HJ, Park S, Kim S, Hong SH, Kim JY, Park EJ, Kang KS, Kim KP, and Cho MH. Dihydroceramide is a key metabolite that regulates autophagy and promotes fibrosis in hepatic steatosis model. Biochem Biophys Res Commun 494: 460–469, 2017. [DOI] [PubMed] [Google Scholar]
- 105.Lee JY, Hannun YA, and Obeid LM. Ceramide inactivates cellular protein kinase Calpha. J Biol Chem 271: 13169–13174, 1996. [DOI] [PubMed] [Google Scholar]
- 106.Lee L, Abe A, and Shayman JA. Improved inhibitors of glucosylceramide synthase. J Biol Chem 274: 14662–14669, 1999. [DOI] [PubMed] [Google Scholar]
- 107.Levy M, Castillo SS, and Goldkorn T. nSMase2 activation and trafficking are modulated by oxidative stress to induce apoptosis. Biochem Biophys Res Commun 344: 900–905, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Li M, Yang L, Bai Y, and Liu H. Analytical methods in lipidomics and their applications. Anal Chem 86: 161–175, 2014. [DOI] [PubMed] [Google Scholar]
- 109.Li YL, Qi XY, Jiang H, Deng XD, Dong YP, Ding TB, Zhou L, Men P, Chu Y, Wang RX, Jiang XC, and Ye DY. Discovery, synthesis and biological evaluation of 2-(4-(N-phenethylsulfamoyl)phenoxy)acetamides (SAPAs) as novel sphingomyelin synthase 1 inhibitors. Bioorg Med Chem 23: 6173–6184, 2015. [DOI] [PubMed] [Google Scholar]
- 110.Lieberman RL, Wustman BA, Huertas P, Powe AC Jr., Pine CW, Khanna R, Schlossmacher MG, Ringe D, and Petsko GA. Structure of acid beta-glucosidase with pharmacological chaperone provides insight into Gaucher disease. Nat Chem Biol 3: 101–107, 2007. [DOI] [PubMed] [Google Scholar]
- 111.Lima S, Milstien S, and Spiegel S. A real-time high-throughput fluorescence assay for sphingosine kinases. J Lipid Res 55: 1525–1530, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lipsky NG, and Pagano RE. Intracellular translocation of fluorescent sphingolipids in cultured fibroblasts: endogenously synthesized sphingomyelin and glucocerebroside analogues pass through the Golgi apparatus en route to the plasma membrane. J Cell Biol 100: 27–34, 1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Liu B, and Hannun YA. Inhibition of the neutral magnesium-dependent sphingomyelinase by glutathione. J Biol Chem 272: 16281–16287, 1997. [DOI] [PubMed] [Google Scholar]
- 114.Liu YY, Gupta V, Patwardhan GA, Bhinge K, Zhao Y, Bao J, Mehendale H, Cabot MC, Li YT, and Jazwinski SM. Glucosylceramide synthase upregulates MDR1 expression in the regulation of cancer drug resistance through cSrc and beta-catenin signaling. Mol Cancer 9: 145, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Loetscher E, Schneider K, Beerli C, and Billich A. Assay to measure the secretion of sphingosine-1-phosphate from cells induced by S1P lyase inhibitors. Biochem Biophys Res Commun 433: 345–348, 2013. [DOI] [PubMed] [Google Scholar]
- 116.Loidl A, Claus R, Deigner HP, and Hermetter A. High-precision fluorescence assay for sphingomyelinase activity of isolated enzymes and cell lysates. J Lipid Res 43: 815–823, 2002. [PubMed] [Google Scholar]
- 117.Lopez DJ, Egido-Gabas M, Lopez-Montero I, Busto JV, Casas J, Garnier M, Monroy F, Larijani B, Goni FM, and Alonso A. Accumulated bending energy elicits neutral sphingomyelinase activity in human red blood cells. Biophys J 102: 2077–2085, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Loveridge C, Tonelli F, Leclercq T, Lim KG, Long JS, Berdyshev E, Tate RJ, Natarajan V, Pitson SM, Pyne NJ, and Pyne S. The sphingosine kinase 1 inhibitor 2-(p-hydroxyanilino)-4-(pchlorophenyl)thiazole induces proteasomal degradation of sphingosine kinase 1 in mammalian cells. J Biol Chem 285: 38841–38852, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Luberto C, and Hannun YA. Sphingomyelin synthase, a potential regulator of intracellular levels of ceramide and diacylglycerol during SV40 transformation. Does sphingomyelin synthase account for the putative phosphatidylcholine-specific phospholipase C? J Biol Chem 273: 14550–14559, 1998. [DOI] [PubMed] [Google Scholar]
- 120.Luberto C, Hassler DF, Signorelli P, Okamoto Y, Sawai H, Boros E, Hazen-Martin DJ, Obeid LM, Hannun YA, and Smith GK. Inhibition of tumor necrosis factor-induced cell death in MCF7 by a novel inhibitor of neutral sphingomyelinase. J Biol Chem 277: 41128–41139, 2002. [DOI] [PubMed] [Google Scholar]
- 121.Martinez-Montanes F, and Schneiter R. Tools for the analysis of metabolic flux through the sphingolipid pathway. Biochimie 130: 76–80, 2016. [DOI] [PubMed] [Google Scholar]
- 122.Matsuzaki W, Takahashi H, Nakamura H, and Murayama T. Effects of Glycerophospholipids on Ceramide Kinase Activity: Cardiolipin-Affected Cellular Formation of Ceramide-1-phosphate. Biol Pharm Bull 39: 1708–1717, 2016. [DOI] [PubMed] [Google Scholar]
- 123.McNaughton M, Pitman M, Pitson SM, Pyne NJ, and Pyne S. Proteasomal degradation of sphingosine kinase 1 and inhibition of dihydroceramide desaturase by the sphingosine kinase inhibitors, SKi or ABC294640, induces growth arrest in androgen-independent LNCaP-AI prostate cancer cells. Oncotarget 7: 16663–16675, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Medlock KA, and Merrill AH Jr., Inhibition of serine palmitoyltransferase in vitro and long-chain base biosynthesis in intact Chinese hamster ovary cells by beta-chloroalanine. Biochemistry 27: 7079–7084, 1988. [DOI] [PubMed] [Google Scholar]
- 125.Meng A, Luberto C, Meier P, Bai A, Yang X, Hannun YA, and Zhou D. Sphingomyelin synthase as a potential target for D609-induced apoptosis in U937 human monocytic leukemia cells. Exp Cell Res 292: 385–392, 2004. [DOI] [PubMed] [Google Scholar]
- 126.Merrill AH Jr. Sphingolipid and glycosphingolipid metabolic pathways in the era of sphingolipidomics. Chemical reviews 111: 6387–6422, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Merrill AH Jr., Sullards MC, Allegood JC, Kelly S, and Wang E. Sphingolipidomics: high-throughput, structure-specific, and quantitative analysis of sphingolipids by liquid chromatography tandem mass spectrometry. Methods 36: 207–224, 2005. [DOI] [PubMed] [Google Scholar]
- 128.Merrill AH Jr., and Wang E. Biosynthesis of long-chain (sphingoid) bases from serine by LM cells. Evidence for introduction of the 4-trans-double bond after de novo biosynthesis of N-acylsphinganine(s). J Biol Chem 261: 3764–3769, 1986. [PubMed] [Google Scholar]
- 129.Merrill AH Jr., and Wang E. Enzymes of ceramide biosynthesis. Methods Enzymol 209: 427–437, 1992. [DOI] [PubMed] [Google Scholar]
- 130.Merrill AH Jr., Wang E, and Mullins RE. Kinetics of long-chain (sphingoid) base biosynthesis in intact LM cells: effects of varying the extracellular concentrations of serine and fatty acid precursors of this pathway. Biochemistry 27: 340–345, 1988. [DOI] [PubMed] [Google Scholar]
- 131.Midorikawa M, Okada S, Yutaka T, Yabuuchi H, Naoi M, Kiuchi K, and Yagi K. Assay of glucocerebrosidase using a fluorescent analogue of glucocerebroside for the diagnosis of Gaucher disease. Biochem Int 11: 327–332, 1985. [PubMed] [Google Scholar]
- 132.Mitsutake S, Kita K, Okino N, and Ito M. [14C]ceramide synthesis by sphingolipid ceramide N-deacylase: new assay for ceramidase activity detection. Anal Biochem 247: 52–57, 1997. [DOI] [PubMed] [Google Scholar]
- 133.Miyake Y, Kozutsumi Y, Nakamura S, Fujita T, and Kawasaki T. Serine palmitoyltransferase is the primary target of a sphingosine-like immunosuppressant, ISP-1/myriocin. Biochem Biophys Res Commun 211: 396–403, 1995. [DOI] [PubMed] [Google Scholar]
- 134.Mullen TD, Jenkins RW, Clarke CJ, Bielawski J, Hannun YA, and Obeid LM. Ceramide synthase-dependent ceramide generation and programmed cell death: involvement of salvage pathway in regulating postmitochondrial events. J Biol Chem 286: 15929–15942, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Mullen TD, Spassieva S, Jenkins RW, Kitatani K, Bielawski J, Hannun YA, and Obeid LM. Selective knockdown of ceramide synthases reveals complex interregulation of sphingolipid metabolism. J Lipid Res 52: 68–77, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Munoz-Olaya JM, Matabosch X, Bedia C, Egido-Gabas M, Casas J, Llebaria A, Delgado A, and Fabrias G. Synthesis and biological activity of a novel inhibitor of dihydroceramide desaturase. ChemMedChem 3: 946–953, 2008. [DOI] [PubMed] [Google Scholar]
- 137.Nara F, Tanaka M, Masuda-Inoue S, Yamasato Y, Doi-Yoshioka H, Suzuki-Konagai K, Kumakura S, and Ogita T. Biological activities of scyphostatin, a neutral sphingomyelinase inhibitor from a discomycete, Trichopeziza mollissima. J Antibiot (Tokyo) 52: 531–535, 1999. [DOI] [PubMed] [Google Scholar]
- 138.Newbigging S, Zhang M, and Saba JD. Immunohistochemical analysis of sphingosine phosphate lyase expression during murine development. Gene Expr Patterns 13: 21–29, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Newcomb B, Rhein C, Mileva I, Ahmad R, Clarke CJ, Snider J, Obeid LM, and Hannun YA. Identification of an acid sphingomyelinase ceramide kinase pathway in the regulation of the chemokine CCL5. J Lipid Res 59: 1219–1229, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Nikolova-Karakashian M, Morgan ET, Alexander C, Liotta DC, and Merrill AH Jr. Bimodal regulation of ceramidase by interleukin-1beta. Implications for the regulation of cytochrome p450 2C11. J Biol Chem 272: 18718–18724, 1997. [DOI] [PubMed] [Google Scholar]
- 141.Norred WP, Wang E, Yoo H, Riley RT, and Merrill AH Jr. In vitro toxicology of fumonisins and the mechanistic implications. Mycopathologia 117: 73–78, 1992. [DOI] [PubMed] [Google Scholar]
- 142.Novgorodov SA, Wu BX, Gudz TI, Bielawski J, Ovchinnikova TV, Hannun YA, and Obeid LM.Novel pathway of ceramide production in mitochondria: thioesterase and neutral ceramidase produce ceramide from sphingosine and acyl-CoA. J Biol Chem 286: 25352–25362, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Obeid LM, Linardic CM, Karolak LA, and Hannun YA. Programmed cell death induced by ceramide. Science 259: 1769–1771, 1993. [DOI] [PubMed] [Google Scholar]
- 144.Ohtoyo M, Tamura M, Machinaga N, Muro F, and Hashimoto R. Sphingosine 1-phosphate lyase inhibition by 2-acetyl-4-(tetrahydroxybutyl)imidazole (THI) under conditions of vitamin B6 deficiency. Mol Cell Biochem 400: 125–133, 2015. [DOI] [PubMed] [Google Scholar]
- 145.Okino N, He X, Gatt S, Sandhoff K, Ito M, and Schuchman EH. The reverse activity of human acid ceramidase. J Biol Chem 278: 29948–29953, 2003. [DOI] [PubMed] [Google Scholar]
- 146.Oleinik NV, and Krupenko SA. Ectopic expression of 10-formyltetrahydrofolate dehydrogenase in A549 cells induces G1 cell cycle arrest and apoptosis. Mol Cancer Res 1: 577–588, 2003. [PubMed] [Google Scholar]
- 147.Olivera A, Barlow KD, and Spiegel S. Assaying sphingosine kinase activity. Methods Enzymol 311: 215–223, 2000. [DOI] [PubMed] [Google Scholar]
- 148.Orr Gandy KA, and Obeid LM. Targeting the sphingosine kinase/sphingosine 1-phosphate pathway in disease: review of sphingosine kinase inhibitors. Biochimica et biophysica acta 1831: 157–166, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Paugh SW, Paugh BS, Rahmani M, Kapitonov D, Almenara JA, Kordula T, Milstien S, Adams JK, Zipkin RE, Grant S, and Spiegel S. A selective sphingosine kinase 1 inhibitor integrates multiple molecular therapeutic targets in human leukemia. Blood 112: 1382–1391, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Peng B, Weintraub ST, Coman C, Ponnaiyan S, Sharma R, Tews B, Winter D, and Ahrends R. A Comprehensive High-Resolution Targeted Workflow for the Deep Profiling of Sphingolipids. Anal Chem 89: 12480–12487, 2017. [DOI] [PubMed] [Google Scholar]
- 151.Penno A, Reilly MM, Houlden H, Laura M, Rentsch K, Niederkofler V, Stoeckli ET, Nicholson G, Eichler F, Brown RH Jr., von Eckardstein A, and Hornemann T. Hereditary sensory neuropathy type 1 is caused by the accumulation of two neurotoxic sphingolipids. J Biol Chem 285: 11178–11187, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Perrotta C, Cervia D, De Palma C, Assi E, Pellegrino P, Bassi MT, and Clementi E. The emerging role of acid sphingomyelinase in autophagy. Apoptosis 20: 635–644, 2015. [DOI] [PubMed] [Google Scholar]
- 153.Perry DK, and Hannun YA. The use of diglyceride kinase for quantifying ceramide. Trends Biochem Sci 24: 226–227, 1999. [DOI] [PubMed] [Google Scholar]
- 154.Pettus BJ, Bielawska A, Spiegel S, Roddy P, Hannun YA, and Chalfant CE. Ceramide kinase mediates cytokine- and calcium ionophore-induced arachidonic acid release. J Biol Chem 278: 38206–38213, 2003. [DOI] [PubMed] [Google Scholar]
- 155.Pitman MR, Powell JA, Coolen C, Moretti PA, Zebol JR, Pham DH, Finnie JW, Don AS, Ebert LM, Bonder CS, Gliddon BL, and Pitson SM. A selective ATP-competitive sphingosine kinase inhibitor demonstrates anti-cancer properties. Oncotarget 6: 7065–7083, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Pizzirani D, Bach A, Realini N, Armirotti A, Mengatto L, Bauer I, Girotto S, Pagliuca C, De Vivo M, Summa M, Ribeiro A, and Piomelli D. Benzoxazolone carboxamides: potent and systemically active inhibitors of intracellular acid ceramidase. Angew Chem Int Ed Engl 54: 485–489, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Pizzirani D, Pagliuca C, Realini N, Branduardi D, Bottegoni G, Mor M, Bertozzi F, Scarpelli R, Piomelli D, and Bandiera T. Discovery of a new class of highly potent inhibitors of acid ceramidase: synthesis and structure-activity relationship (SAR). J Med Chem 56: 3518–3530, 2013. [DOI] [PubMed] [Google Scholar]
- 158.Platt FM, Neises GR, Dwek RA, and Butters TD. N-butyldeoxynojirimycin is a novel inhibitor of glycolipid biosynthesis. J Biol Chem 269: 8362–8365, 1994. [PubMed] [Google Scholar]
- 159.Platt FM, Neises GR, Karlsson GB, Dwek RA, and Butters TD. N-butyldeoxygalactonojirimycin inhibits glycolipid biosynthesis but does not affect N-linked oligosaccharide processing. J Biol Chem 269: 27108–27114, 1994. [PubMed] [Google Scholar]
- 160.Preiss J, Loomis CR, Bishop WR, Stein R, Niedel JE, and Bell RM. Quantitative measurement of sn-1,2-diacylglycerols present in platelets, hepatocytes, and ras- and sis-transformed normal rat kidney cells. J Biol Chem 261: 8597–8600, 1986. [PubMed] [Google Scholar]
- 161.Previati M, Bertolaso L, Tramarin M, Bertagnolo V, and Capitani S. Low nanogram range quantitation of diglycerides and ceramide by high-performance liquid chromatography. Anal Biochem 233: 108–114, 1996. [DOI] [PubMed] [Google Scholar]
- 162.Pulkoski-Gross MJ, Uys JD, Orr-Gandy KA, Coant N, Bialkowska AB, Szulc ZM, Bai A, Bielawska A, Townsend DM, Hannun YA, Obeid LM, and Snider AJ. Novel sphingosine kinase-1 inhibitor, LCL351, reduces immune responses in murine DSS-induced colitis. Prostaglandins Other Lipid Mediat 130: 47–56, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Pyne NJ, and Pyne S. Sphingosine 1-phosphate and cancer. Nat Rev Cancer 10: 489–503, 2010. [DOI] [PubMed] [Google Scholar]
- 164.Rahmaniyan M, Curley RW Jr., Obeid LM, Hannun YA, and Kraveka JM. Identification of dihydroceramide desaturase as a direct in vitro target for fenretinide. J Biol Chem 286: 24754–24764, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Rajagopalan V, Canals D, Luberto C, Snider J, Voelkel-Johnson C, Obeid LM, and Hannun YA. Critical determinants of mitochondria-associated neutral sphingomyelinase (MA-nSMase) for mitochondrial localization. Biochimica et biophysica acta 1850: 628–639, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Realini N, Solorzano C, Pagliuca C, Pizzirani D, Armirotti A, Luciani R, Costi MP, Bandiera T, and Piomelli D. Discovery of highly potent acid ceramidase inhibitors with in vitro tumor chemosensitizing activity. Sci Rep 3: 1035, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Ren J, Snider J, Airola MV, Zhong A, Rana NA, Obeid LM, and Hannun YA. Quantification of 3-ketodihydrosphingosine using HPLC-ESI-MS/MS to study SPT activity in yeast Saccharomyces cerevisiae. J Lipid Res 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Riboni L, Prinetti A, Bassi R, Viani P, and Tettamanti G. The effects of exogenous sphingosine on Neuro2a cells are strictly related to the overall capacity of cells to metabolize sphingosine. J Biochem 124: 900–904, 1998. [DOI] [PubMed] [Google Scholar]
- 169.Riboni L, Viani P, Bassi R, Giussani P, and Tettamanti G. Basic fibroblast growth factor-induced proliferation of primary astrocytes. evidence for the involvement of sphingomyelin biosynthesis. J Biol Chem 276: 12797–12804, 2001. [DOI] [PubMed] [Google Scholar]
- 170.Riley RT, Norred WP, Wang E, and Merrill AH. Alteration in sphingolipid metabolism: bioassays for fumonisin- and ISP-I-like activity in tissues, cells and other matrices. Nat Toxins 7: 407–414, 1999. [DOI] [PubMed] [Google Scholar]
- 171.Rojas C, Barnaeva E, Thomas AG, Hu X, Southall N, Marugan J, Chaudhuri AD, Yoo SW, Hin N, Stepanek O, Wu Y, Zimmermann SC, Gadiano AG, Tsukamoto T, Rais R, Haughey N, Ferrer M, and Slusher BS. DPTIP, a newly identified potent brain penetrant neutral sphingomyelinase 2 inhibitor, regulates astrocyte-peripheral immune communication following brain inflammation. Sci Rep 8: 17715, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Rotolo J, Stancevic B, Zhang J, Hua G, Fuller J, Yin X, Haimovitz-Friedman A, Kim K, Qian M, Cardo-Vila M, Fuks Z, Pasqualini R, Arap W, and Kolesnick R. Anti-ceramide antibody prevents the radiation gastrointestinal syndrome in mice. J Clin Invest 122: 1786–1790, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Rutkute K, Asmis RH, and Nikolova-Karakashian MN. Regulation of neutral sphingomyelinase-2 by GSH: a new insight to the role of oxidative stress in aging-associated inflammation. J Lipid Res 48: 2443–2452, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Ryland LK, Fox TE, Liu X, Loughran TP, and Kester M. Dysregulation of sphingolipid metabolism in cancer. Cancer Biol Ther 11: 138–149, 2011. [DOI] [PubMed] [Google Scholar]
- 175.Sabbadini RA. Sphingosine-1-phosphate antibodies as potential agents in the treatment of cancer and age-related macular degeneration. Br J Pharmacol 162: 1225–1238, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Sankala HM, Hait NC, Paugh SW, Shida D, Lepine S, Elmore LW, Dent P, Milstien S, and Spiegel S. Involvement of sphingosine kinase 2 in p53-independent induction of p21 by the chemotherapeutic drug doxorubicin. Cancer Res 67: 10466–10474, 2007. [DOI] [PubMed] [Google Scholar]
- 177.Sanllehi P, Casasampere M, Abad JL, Fabrias G, Lopez O, Bujons J, Casas J, and Delgado A. The first fluorogenic sensor for sphingosine-1-phosphate lyase activity in intact cells. Chem Commun (Camb) 53: 5441–5444, 2017. [DOI] [PubMed] [Google Scholar]
- 178.Schiffmann S, Hartmann D, Fuchs S, Birod K, Ferreiros N, Schreiber Y, Zivkovic A, Geisslinger G, Grosch S, and Stark H. Inhibitors of specific ceramide synthases. Biochimie 94: 558–565, 2012. [DOI] [PubMed] [Google Scholar]
- 179.Schnute ME, McReynolds MD, Kasten T, Yates M, Jerome G, Rains JW, Hall T, Chrencik J, Kraus M, Cronin CN, Saabye M, Highkin MK, Broadus R, Ogawa S, Cukyne K, Zawadzke LE, Peterkin V, Iyanar K, Scholten JA, Wendling J, Fujiwara H, Nemirovskiy O, Wittwer AJ, and Nagiec MM. Modulation of cellular S1P levels with a novel, potent and specific inhibitor of sphingosine kinase-1. Biochem J 444: 79–88, 2012. [DOI] [PubMed] [Google Scholar]
- 180.Schulz A, Mousallem T, Venkataramani M, Persaud-Sawin DA, Zucker A, Luberto C, Bielawska A, Bielawski J, Holthuis JC, Jazwinski SM, Kozhaya L, Dbaibo GS, and Boustany RM. The CLN9 protein, a regulator of dihydroceramide synthase. J Biol Chem 281: 2784–2794, 2006. [DOI] [PubMed] [Google Scholar]
- 181.Schwab SR, Pereira JP, Matloubian M, Xu Y, Huang Y, and Cyster JG. Lymphocyte sequestration through S1P lyase inhibition and disruption of S1P gradients. Science 309: 1735–1739, 2005. [DOI] [PubMed] [Google Scholar]
- 182.Schwarzmann G, Arenz C, and Sandhoff K. Labeled chemical biology tools for investigating sphingolipid metabolism, trafficking and interaction with lipids and proteins. Biochim Biophys Acta 1841: 1161–1173, 2014. [DOI] [PubMed] [Google Scholar]
- 183.Senkal CE, Ponnusamy S, Rossi MJ, Bialewski J, Sinha D, Jiang JC, Jazwinski SM, Hannun YA, and Ogretmen B. Role of human longevity assurance gene 1 and C18-ceramide in chemotherapy-induced cell death in human head and neck squamous cell carcinomas. Mol Cancer Ther 6: 712–722, 2007. [DOI] [PubMed] [Google Scholar]
- 184.Seufferlein T, and Rozengurt E. Sphingosine induces p125FAK and paxillin tyrosine phosphorylation, actin stress fiber formation, and focal contact assembly in Swiss 3T3 cells. The Journal of biological chemistry 269: 27610–27617, 1994. [PubMed] [Google Scholar]
- 185.Shamseddine AA, Clarke CJ, Carroll B, Airola MV, Mohammed S, Rella A, Obeid LM, and Hannun YA. P53-dependent upregulation of neutral sphingomyelinase-2: role in doxorubicin-induced growth arrest. Cell Death Dis 6: e1947, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Shaner RL, Allegood JC, Park H, Wang E, Kelly S, Haynes CA, Sullards MC, and Merrill AH Jr. Quantitative analysis of sphingolipids for lipidomics using triple quadrupole and quadrupole linear ion trap mass spectrometers. Journal of lipid research 50: 1692–1707, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Shayman JA. Targeting Glucosylceramide Synthesis in the Treatment of Rare and Common Renal Disease. Semin Nephrol 38: 183–192, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Shayman JA, and Abe A. 1-O-acylceramide synthase. Methods Enzymol 311: 105–117, 2000. [DOI] [PubMed] [Google Scholar]
- 189.Sillence DJ, Puri V, Marks DL, Butters TD, Dwek RA, Pagano RE, and Platt FM. Glucosylceramide modulates membrane traffic along the endocytic pathway. J Lipid Res 43: 1837–1845, 2002. [DOI] [PubMed] [Google Scholar]
- 190.Sims K, Haynes CA, Kelly S, Allegood JC, Wang E, Momin A, Leipelt M, Reichart D, Glass CK, Sullards MC, and Merrill AH Jr., Kdo2-lipid A, a TLR4-specific agonist, induces de novo sphingolipid biosynthesis in RAW264.7 macrophages, which is essential for induction of autophagy. J Biol Chem 285: 38568–38579, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Siow D, Sunkara M, Dunn TM, Morris AJ, and Wattenberg B. ORMDL/serine palmitoyltransferase stoichiometry determines effects of ORMDL3 expression on sphingolipid biosynthesis. J Lipid Res 56: 898–908, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Siow DL, and Wattenberg BW. An assay system for measuring the acute production of sphingosine 1-phosphate in intact monolayers. Anal Biochem 371: 184–193, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Siskind LJ. Mitochondrial ceramide and the induction of apoptosis. J Bioenerg Biomembr 37: 143–153, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Snider AJ. Sphingosine kinase and sphingosine-1-phosphate: regulators in autoimmune and inflammatory disease. Int J Clin Rheumtol 8: 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Snider JM, Snider AJ, Obeid LM, Luberto C, and Hannun YA. Probing de novo sphingolipid metabolism in mammalian cells utilizing mass spectrometry. J Lipid Res 59: 1046–1057, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Snider JM, Snider AJ, Obeid LM, Luberto C, and Hannun YA. Probing de novo sphingolipid metabolism in mammalian cells utilizing mass spectrometry. J Lipid Res 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Spassieva S, Bielawski J, Anelli V, and Obeid LM. Combination of C(17) sphingoid base homologues and mass spectrometry analysis as a new approach to study sphingolipid metabolism. Methods Enzymol 434: 233–241, 2007. [DOI] [PubMed] [Google Scholar]
- 198.Spinedi A, Di Bartolomeo S, and Piacentini M. N-Oleoylethanolamine inhibits glucosylation of natural ceramides in CHP-100 neuroepithelioma cells: possible implications for apoptosis. Biochem Biophys Res Commun 255: 456–459, 1999. [DOI] [PubMed] [Google Scholar]
- 199.Stahelin RV, Hwang JH, Kim JH, Park ZY, Johnson KR, Obeid LM, and Cho W. The mechanism of membrane targeting of human sphingosine kinase 1. J Biol Chem 280: 43030–43038, 2005. [DOI] [PubMed] [Google Scholar]
- 200.Strub GM, Maceyka M, Hait NC, Milstien S, and Spiegel S. Extracellular and intracellular actions of sphingosine-1-phosphate. Adv Exp Med Biol 688: 141–155, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Subathra M, Korrapati M, Howell LA, Arthur JM, Shayman JA, Schnellmann RG, and Siskind LJ. Kidney glycosphingolipids are elevated early in diabetic nephropathy and mediate hypertrophy of mesangial cells. Am J Physiol Renal Physiol 309: F204–215, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Sugita M, Iwamori M, Evans J, McCluer RH, Dulaney JT, and Moser HW. High performance liquid chromatography of ceramides: application to analysis in human tissues and demonstration of ceramide excess in Farber’s disease. J Lipid Res 15: 223–226, 1974. [PubMed] [Google Scholar]
- 203.Sugiura M, Kono K, Liu H, Shimizugawa T, Minekura H, Spiegel S, and Kohama T. Ceramide kinase, a novel lipid kinase. Molecular cloning and functional characterization. J Biol Chem 277: 23294–23300, 2002. [DOI] [PubMed] [Google Scholar]
- 204.Sundaram KS, and Lev M. Inhibition of sphingolipid synthesis by cycloserine in vitro and in vivo. J Neurochem 42: 577–581, 1984. [DOI] [PubMed] [Google Scholar]
- 205.Suzuki A, Miyazaki M, Matsuda J, and Yoneshige A. High-performance thin-layer chromatography/mass spectrometry for the analysis of neutral glycosphingolipids. Biochimica et biophysica acta 1811: 861–874, 2011. [DOI] [PubMed] [Google Scholar]
- 206.Tafesse FG, Huitema K, Hermansson M, van der Poel S, van den Dikkenberg J, Uphoff A, Somerharju P, and Holthuis JC. Both sphingomyelin synthases SMS1 and SMS2 are required for sphingomyelin homeostasis and growth in human HeLa cells. J Biol Chem 282: 17537–17547, 2007. [DOI] [PubMed] [Google Scholar]
- 207.Taha TA, Argraves KM, and Obeid LM. Sphingosine-1-phosphate receptors: receptor specificity versus functional redundancy. Biochimica et biophysica acta 1682: 48–55, 2004. [DOI] [PubMed] [Google Scholar]
- 208.Takeichi T, Torrelo A, Lee JYW, Ohno Y, Lozano ML, Kihara A, Liu L, Yasuda Y, Ishikawa J, Murase T, Rodrigo AB, Fernandez-Crehuet P, Toi Y, Mellerio J, Rivera J, Vicente V, Kelsell DP, Nishimura Y, Okuno Y, Kojima D, Ogawa Y, Sugiura K, Simpson MA, McLean WHI, Akiyama M, and McGrath JA. Biallelic Mutations in KDSR Disrupt Ceramide Synthesis and Result in a Spectrum of Keratinization Disorders Associated with Thrombocytopenia. J Invest Dermatol 137: 2344–2353, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Tani M, Kita K, Komori H, Nakagawa T, and Ito M. Enzymatic synthesis of omega-amino-ceramide: preparation of a sensitive fluorescent substrate for ceramidase. Anal Biochem 263: 183–188, 1998. [DOI] [PubMed] [Google Scholar]
- 210.Tani M, and Kuge O. Sphingomyelin synthase 2 is palmitoylated at the COOH-terminal tail, which is involved in its localization in plasma membranes. Biochem Biophys Res Commun 381: 328–332, 2009. [DOI] [PubMed] [Google Scholar]
- 211.Tani M, Okino N, Mitsutake S, and Ito M. Specific and sensitive assay for alkaline and neutral ceramidases involving C12-NBD-ceramide. J Biochem 125: 746–749, 1999. [DOI] [PubMed] [Google Scholar]
- 212.Taniguchi M, and Okazaki T. The role of sphingomyelin and sphingomyelin synthases in cell death, proliferation and migration-from cell and animal models to human disorders. Biochimica et biophysica acta 1841: 692–703, 2014. [DOI] [PubMed] [Google Scholar]
- 213.Testai FD, Landek MA, and Dawson G. Regulation of sphingomyelinases in cells of the oligodendrocyte lineage. J Neurosci Res 75: 66–74, 2004. [DOI] [PubMed] [Google Scholar]
- 214.Testai FD, Landek MA, Goswami R, Ahmed M, and Dawson G. Acid sphingomyelinase and inhibition by phosphate ion: role of inhibition by phosphatidyl-myo-inositol 3,4,5-triphosphate in oligodendrocyte cell signaling. J Neurochem 89: 636–644, 2004. [DOI] [PubMed] [Google Scholar]
- 215.Tidhar R, Sims K, Rosenfeld-Gur E, Shaw W, and Futerman AH. A rapid ceramide synthase activity using NBD-sphinganine and solid phase extraction. J Lipid Res 56: 193–199, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Tolan D, Conway AM, Rakhit S, Pyne N, and Pyne S. Assessment of the extracellular and intracellular actions of sphingosine 1-phosphate by using the p42/p44 mitogen-activated protein kinase cascade as a model. Cell Signal 11: 349–354, 1999. [DOI] [PubMed] [Google Scholar]
- 217.Torretta E, Fania C, Vasso M, and Gelfi C. HPTLC-MALDI MS for (glyco)sphingolipid multiplexing in tissues and blood: A promising strategy for biomarker discovery and clinical applications. Electrophoresis 37: 2036–2049, 2016. [DOI] [PubMed] [Google Scholar]
- 218.Triola G, Fabrias G, Dragusin M, Niederhausen L, Broere R, Llebaria A, and van Echten-Deckert G. Specificity of the dihydroceramide desaturase inhibitor N-[(1R,2S)-2-hydroxy-1-hydroxymethyl-2-(2-tridecyl-1-cyclopropenyl)ethyl]octanami de (GT11) in primary cultured cerebellar neurons. Mol Pharmacol 66: 1671–1678, 2004. [DOI] [PubMed] [Google Scholar]
- 219.Turner N, Lim XY, Toop HD, Osborne B, Brandon AE, Taylor EN, Fiveash CE, Govindaraju H, Teo JD, McEwen HP, Couttas TA, Butler SM, Das A, Kowalski GM, Bruce CR, Hoehn KL, Fath T, Schmitz-Peiffer C, Cooney GJ, Montgomery MK, Morris JC, and Don AS. A selective inhibitor of ceramide synthase 1 reveals a novel role in fat metabolism. Nat Commun 9: 3165, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Usta J, El Bawab S, Roddy P, Szulc ZM, Yusuf, Hannun A, and Bielawska A. Structural equirements of ceramide and sphingosine based inhibitors of mitochondrial ceramidase. Biochemistry 40: 9657–9668, 2001. [DOI] [PubMed] [Google Scholar]
- 221.Vacaru AM, Tafesse FG, Ternes P, Kondylis V, Hermansson M, Brouwers JF, Somerharju P, Rabouille C, and Holthuis JC. Sphingomyelin synthase-related protein SMSr controls ceramide homeostasis in the ER. J Cell Biol 185: 1013–1027, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Van Overloop H, Denizot Y, Baes M, and Van Veldhoven PP. On the presence of C2-ceramide in mammalian tissues: possible relationship to etherphospholipids and phosphorylation by ceramide kinase. Biol Chem 388: 315–324, 2007. [DOI] [PubMed] [Google Scholar]
- 223.Van Overloop H, Gijsbers S, and Van Veldhoven PP. Further characterization of mammalian ceramide kinase: substrate delivery and (stereo)specificity, tissue distribution, and subcellular localization studies. J Lipid Res 47: 268–283, 2006. [DOI] [PubMed] [Google Scholar]
- 224.Van Overloop H, Van der Hoeven G, and Van Veldhoven PP. A Nonradioactive Fluorimetric SPE-Based Ceramide Kinase Assay Using NBD-C(6)-Ceramide. J Lipids 2012: 404513, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Van Veldhoven PP, Bishop WR, and Bell RM. Enzymatic quantification of sphingosine in the picomole range in cultured cells. Anal Biochem 183: 177–189, 1989. [DOI] [PubMed] [Google Scholar]
- 226.Venable ME, Lee JY, Smyth MJ, Bielawska A, and Obeid LM. Role of ceramide in cellular senescence. The Journal of biological chemistry 270: 30701–30708, 1995. [DOI] [PubMed] [Google Scholar]
- 227.Vessey DA, Kelley M, Zhang J, Li L, Tao R, and Karliner JS. Dimethylsphingosine and FTY720 inhibit the SK1 form but activate the SK2 form of sphingosine kinase from rat heart. J Biochem Mol Toxicol 21: 273–279, 2007. [DOI] [PubMed] [Google Scholar]
- 228.Villani M, Subathra M, Im YB, Choi Y, Signorelli P, Del Poeta M, and Luberto C. Sphingomyelin synthases regulate production of diacylglycerol at the Golgi. Biochem J 414: 31–41, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Vogel P, Donoviel MS, Read R, Hansen GM, Hazlewood J, Anderson SJ, Sun W, Swaffield J, and Oravecz T. Incomplete inhibition of sphingosine 1-phosphate lyase modulates immune system function yet prevents early lethality and non-lymphoid lesions. PLoS One 4: e4112, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.von Gerichten J, Schlosser K, Lamprecht D, Morace I, Eckhardt M, Wachten D, Jennemann R, Grone HJ, Mack M, and Sandhoff R. Diastereomer-specific quantification of bioactive hexosylceramides from bacteria and mammals. J Lipid Res 58: 1247–1258, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Walden CM, Sandhoff R, Chuang CC, Yildiz Y, Butters TD, Dwek RA, Platt FM, and van der Spoel AC. Accumulation of glucosylceramide in murine testis, caused by inhibition of beta-glucosidase 2: implications for spermatogenesis. J Biol Chem 282: 32655–32664, 2007. [DOI] [PubMed] [Google Scholar]
- 232.Wang H, Maurer BJ, Liu YY, Wang E, Allegood JC, Kelly S, Symolon H, Liu Y, Merrill AH Jr., Gouaze-Andersson V, Yu JY, Giuliano AE, and Cabot MC. N-(4-Hydroxyphenyl)retinamide increases dihydroceramide and synergizes with dimethylsphingosine to enhance cancer cell killing. Mol Cancer Ther 7: 2967–2976, 2008. [DOI] [PubMed] [Google Scholar]
- 233.Wang H, Maurer BJ, Reynolds CP, and Cabot MC. N-(4-hydroxyphenyl)retinamide elevates ceramide in neuroblastoma cell lines by coordinate activation of serine palmitoyltransferase and ceramide synthase. Cancer Res 61: 5102–5105, 2001. [PubMed] [Google Scholar]
- 234.Wang J, Knapp S, Pyne NJ, Pyne S, and Elkins JM. Crystal Structure of Sphingosine Kinase 1 with PF-543. ACS Med Chem Lett 5: 1329–1333, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Watanabe M, Kitano T, Kondo T, Yabu T, Taguchi Y, Tashima M, Umehara H, Domae N, Uchiyama T, and Okazaki T. Increase of nuclear ceramide through caspase-3-dependent regulation of the “sphingomyelin cycle” in Fas-induced apoptosis. Cancer Res 64: 1000–1007, 2004. [DOI] [PubMed] [Google Scholar]
- 236.Wegner MS, Schiffmann S, Parnham MJ, Geisslinger G, and Grosch S. The enigma of ceramide synthase regulation in mammalian cells. Prog Lipid Res 63: 93–119, 2016. [DOI] [PubMed] [Google Scholar]
- 237.Weiler S, Braendlin N, Beerli C, Bergsdorf C, Schubart A, Srinivas H, Oberhauser B, and Billich A. Orally active 7-substituted (4-benzylphthalazin-1-yl)-2-methylpiperazin-1-yl]nicotinonitriles as active-site inhibitors of sphingosine 1-phosphate lyase for the treatment of multiple sclerosis. J Med Chem 57: 5074–5084, 2014. [DOI] [PubMed] [Google Scholar]
- 238.Wennekesa T, et al. Synthesis and evaluation of dimeric lipophilic iminosugars as inhibitors of glucosylceramide metabolism. Tetrahedron: Asymmetry 20: 836–846, 2009. [Google Scholar]
- 239.White-Gilbertson S, Mullen T, Senkal C, Lu P, Ogretmen B, Obeid L, and Voelkel-Johnson C. Ceramide synthase 6 modulates TRAIL sensitivity and nuclear translocation of active caspase-3 in colon cancer cells. Oncogene 28: 1132–1141, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Wu BX, Zeidan YH, and Hannun YA. Downregulation of neutral ceramidase by gemcitabine: Implications for cell cycle regulation. Biochimica et biophysica acta 1791: 730–739, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Yachi R, Uchida Y, Balakrishna BH, Anderluh G, Kobayashi T, Taguchi T, and Arai H. Subcellular localization of sphingomyelin revealed by two toxin-based probes in mammalian cells. Genes Cells 17: 720–727, 2012. [DOI] [PubMed] [Google Scholar]
- 242.Yano M, Kishida E, Muneyuki Y, and Masuzawa Y. Quantitative analysis of ceramide molecular species by high performance liquid chromatography. J Lipid Res 39: 2091–2098, 1998. [PubMed] [Google Scholar]
- 243.Yavin E, and Gatt S. Enzymatic hydrolysis of sphingolipids. 8. Further purification and properties of rat brain ceramidase. Biochemistry 8: 1692–1698, 1969. [DOI] [PubMed] [Google Scholar]
- 244.Yildiz Y, Matern H, Thompson B, Allegood JC, Warren RL, Ramirez DM, Hammer RE, Hamra FK, Matern S, and Russell DW. Mutation of beta-glucosidase 2 causes glycolipid storage disease and impaired male fertility. J Clin Invest 116: 2985–2994, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Zeidan YH, Pettus BJ, Elojeimy S, Taha T, Obeid LM, Kawamori T, Norris JS, and Hannun YA. Acid ceramidase but not acid sphingomyelinase is required for tumor necrosis factor-{alpha}-induced PGE2 production. J Biol Chem 281: 24695–24703, 2006. [DOI] [PubMed] [Google Scholar]
- 246.Zha X, Pierini LM, Leopold PL, Skiba PJ, Tabas I, and Maxfield FR. Sphingomyelinase treatment induces ATP-independent endocytosis. J Cell Biol 140: 39–47, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Zhang L, Diaz-Diaz N, Zarringhalam K, Hermansson M, Somerharju P, and Chuang J. Dynamics of the ethanolamine glycerophospholipid remodeling network. PLoS One 7: e50858, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Zhu X, Sheth KA, Li S, Chang HH, and Fan JQ. Rational design and synthesis of highly potent beta-glucocerebrosidase inhibitors. Angew Chem Int Ed Engl 44: 7450–7453, 2005. [DOI] [PubMed] [Google Scholar]
- 249.Zitomer NC, Mitchell T, Voss KA, Bondy GS, Pruett ST, Garnier-Amblard EC, Liebeskind LS, Park H, Wang E, Sullards MC, Merrill AH Jr., and Riley RT. Ceramide synthase inhibition by fumonisin B1 causes accumulation of 1-deoxysphinganine: a novel category of bioactive 1-deoxysphingoid bases and 1-deoxydihydroceramides biosynthesized by mammalian cell lines and animals. J Biol Chem 284: 4786–4795, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]



