Research Paper

Pdf file on Synergy

Download Chinese abstract

Acta Biochim Biophys Sin 2005,37:515-524

doi:10.1111/j.1745-7270.2005.00073.x

A Lipidomic Study of the Effects of N-methyl-N'-nitro-N-nitrosoguanidine on Sphingomyelin Metabolism

 

Yun HUANG&#, Jing SHEN#, Ting WANG, Yan-Ke YU, Fanqing F. CHEN1, and Jun YANG*

 

Department of Pathology and Pathophysiology, Center for Environmental Genomics, Zhejiang University School of Medicine, Hangzhou 310031, China;

1 Molecular Biology Branch, Life Science Division, Lawrence Berkeley National Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA

 

Received: March 20, 2005

Accepted: May 23, 2005

& Present address: Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA

# These authors contributed equally to this work

*Corresponding author: Tel/Fax, 86-571-87217149; E-mail, [email protected]

 

Abstract        Systems biology is a new and rapidly developing research area in which, by quantitatively describing the interaction among all the individual components of a cell, a systems-level understanding of a biological response can be achieved. Therefore, it requires high-throughput measurement technologies for biological molecules, such as genomic and proteomic approaches for DNA/RNA and protein, respectively. Recently, a new concept, lipidomics, which utilizes the mass spectrometry (MS) method for lipid analysis, has been proposed. Using this lipidomic approach, the effects of N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) on sphingomyelin metabolism, a major class of sphingolipids, were evaluated. Sphingomyelin molecules were extracted from cells and analyzed by matrix-assisted laser desorption ionization-time of flight MS. It was found that MNNG induced profound changes in sphingomyelin metabolism, including the appearance of some new sphingomyelin species and the disappearance of some others, and the concentrations of several sphingomyelin­ species also changed. This was accompanied by the redistribution of acid sphingomyelinase (ASM), a key player in sphingomyelin metabolism. On the other hand, imipramine, an inhibitor of ASM, caused the accumulation­ of sphingomyelin. It also prevented some of the effects of MNNG, as well as the redistribution of ASM. Taken together, these data suggested that the lipidomic approach is highly effective for the systematic­ analysis of cellular lipids metabolism.

 

Key words        lipidomics; mass spectrometry; ceramide; sphingomyelin; acid sphingomyelinase

 

The completion of the human genome project has led to a revolution in the world of biological science: the generation­ of "genomics". Following this event, "omics" in other disciplines also emerged, such as proteomics, metabonomics, toxicogenomics and pharmacogenomics [1,2]. All of these "omics", genomics and proteomics in particular, form the foundation for a new research field, systems biology. The goal of systems biology is to formulate a computational/mathematical model that describes­ the structure of the system and its response to individual perturbations through the monitoring of systematic changes of all cellular­ components­ (genes, proteins, or signaling pathways) in response­ to any type of perturbation (biological, genetic, or chemical) [3,4]. Therefore, it requires certain technical approaches which can define many cellular molecules at multiple levels; microarray for DNA analysis in genomics and 2-dimensional (2-D) gel electrophoresis combined with mass spectrometry (MS) for protein analysis in proteomics are just such methods. 

It has been gradually recognized that studying DNA and protein alone does not engender a full understanding of a complex biological response, as other major cellular constituents including lipids and carbohydrates are also involved­ in many physiological processes. Consequently, the lack of such information would hamper the construction of a computational model for systems biology. Recently, a new concept, "lipidomics", has been proposed [5,6]. Lipidomics is a comprehensive analysis of lipid molecules which, in combination with genomics and proteomics, is essential for the understanding of cellular physiology and pathology. Consequently, lipid biology has become a major­ research target of the postgenomic revolution and systems­ biology [7].

Lipids are crucial structural/functional components of cells. As structural material, they not only provide a physical­ barrier for cells, but also provide a platform (or lipid raft) for membrane protein-protein interaction. Even more importantly, many lipid species have distinct cellular functions. For example, diacylglycerol, ceramides, eicosanoids and lysolipids are all second messengers which participate in various cellular events such as growth, proliferation, differentiation and cell death [8]. Sphingolipids are a group of sphingoid-based lipids which are gaining increasing attention from researchers. They are the major components for lipid raft. Furthermore, sphingolipids and their metabolites are involved in many important signal transduction pathways which regulate such cellular processes­ as cell cycle arrest or apoptosis, proliferation and calcium homeostasis, as well as cancer development, multidrug resistance, and viral or bacterial infection processes­ [9]. Clearly, the importance of this group of lipids should not be underestimated.

Unfortunately, the study of lipids is far behind those of genes and proteins. One major obstacle is the lack of high-throughput­ technologies in lipid analysis. The traditional methods, such as isotope labeling, thin-layer chroma­to­graphy­ and high performance liquid chromatography, could provide some useful information, but are far from adequate. Nevertheless, until the application of MS in sphingolipid­ study does a great amount of information is generated. Compared with traditional methods, MS analysis­ is more accurate, less labor-intensive and, most of all, can identify the molecular species of each class of lipids [10,11]. In our previous studies, using isotope labeling methods­ as well as matrix-assisted laser desorption ionization­-time of flight (MALDI-TOF) MS, it has been shown that N-methyl­-N'-nitro-N-nitrosoguanidine (MNNG), an alkylating­ agent which is a potent carcinogen, can affect ceramide metabolism­ [12,13]. Sphingomyelin is another important sphingolipid species closely related to ceramide meta­bolism, for example, sphingomyelin can be hydrolyzed to generate­ ceramide [9]. Therefore, it is quite reasonable to speculate that MNNG would also affect­ sphingomyelin metabolism.

In this research, we investigated the effects of MNNG on sphingomyelin metabolism. In addition, the cellular distribution­ of acid sphingomyelinase (ASM), a key enzyme­ in sphingomyelin metabolism, was also determined. As reported here, MNNG induced the generation/loss of some sphingo­myelin species, as well as the increase/decrease of other sphingo­myelin species.

 

 

Materials and Methods

 

 

Cell culture and reagents

 

Human amnion FL cells were cultured in Eagle's minimum essential medium (EMEM; Invitrogen, Carlsbad, USA) containing 10% fetal bovine serum, supplemented with 100 U/ml penicillin, 100 U/ml streptomycin and 0.03% L-glutamine in a humidified incubator at 37 ºC with 5% CO2. MNNG (Sigma, St. Louis, USA) was dissolved­ in dimethylsulfoxide (DMSO) as a 10 mM stock. Imipramine (Sigma) was also dissolved in DMSO as a 50 mM stock. For MNNG treatment, cells were treated with 10 mM of MNNG for 20 min. DMSO-treated or untreated­ cells were used as solvent control or blank control, respectively.

D-sphingosine, N-acetyl-D-sphingosine (C2-ceramide), N-hexanoyl-D-sphingosine (C6-ceramide), N-octanoyl-D-sphingosine (C8-ceramide), D-threo-ceramide C8, dihydrosphingosine, C2-dihydroceramide, C6-dihydroceramide, and C8-dihydroceramide were all purchased from Sigma; and each was dissolved following the manufacturer's instructions.

 

Immunofluorescent microscopy

 

The translocation of ASM was observed by immuno­fluorescent microscopy as described before [14]. Briefly, 1´105 FL cells were seeded into a 6-well culture plate with a glass cover slip in each well. After MNNG (10 mM) treatment for 20 min, cells were fixed and permeated­ with 100% ice-cold methanol for 5 min, followed­ by blocking­ in a blocking solution (Zymed Laboratories Inc., San Francisco, USA) for 2 h. The plate was washed with PBS three times and the polyclonal rabbit anti-ASM antibody­ (1:200; Santa Cruz Biotechnology, Santa Cruz, USA) was added and incubated for 90 min. Cy3-labeled goat anti-rabbit secondary antibody (1:200; Boster Biological­ Technology Limited, Wuhan, China) was then added to the plate and incubated for 1 h. These cells were then washed and stained with 500 ng/ml FITC-cholera toxin B (Sigma) for 1 h. The cover slip was removed from the plate, mounted onto a glass slide, observed with an Olympus AX70 fluorescent microscope (Olympus, Tokyo, Japan), and analyzed using Image-Pro Plus software (MediaCybernetics, Silver Spring, USA). For imipramine treatment, cells were pre-incubated with 50 mM imipramine for 1 h before adding MNNG.

 

Sphingomyelin extraction and MALDI-TOF MS

 

Sphingomyelin was extracted as described before [12]. In short, approximately 4´107 cells were resolved in 500 ml chloroform:methanol (2:1, V/V). 1 ml H2O was then added to each sample. The mixed samples were centrifuged at 4770 g for 15 min and the lower phase was dried by vacuum centrifugation in a centrifugal evaporator (Speed-Vac, Thermo Savant, Holbrook, USA). Then, 500 ml methanol containing 0.1 M NaOH was added into each tube at 55 ºC for 1 h to decompose glycerophospholipids. After neutralization with 100 ml methanol containing 1 M HCl, 500 ml hexane and one drop of water were added to each sample. The mixture was then centrifuged again at 4770 g for 15 min and the lower phase was dried in a centrifugal evaporator after the upper phase was removed. The residue was mixed with 0.8 ml theoretical lower phase (chloroform:methanol:water, 86:14:1, V/V) and 0.2 ml theoretical upper phase (chloroform:methanol:water, 3:48:47, V/V) for the Folch partition, and centrifuged at 4770 g for 15 min. The lower phase was evaporated in a centrifugal evaporator after removing the upper phase to discard the salt. The residue crude sphingomyelin was stored at -70 ºC.

For MALDI-TOF MS analysis, each sample was dissolved­ in 5 ml chloroform:methanol (2:1, V/V), followed­ by the addition of 5 ml matrix solution, ethylacetate containing­ 0.5 M 2,5-dihydroxyl-benzoic acid (2,5-DHB; Sigma) and 0.1% TFA, in a 0.5 ml Eppendorf tube. The tube was agitated vigorously on a vortex mixer then centrifuged­ in a microcentrifuge for 1 min. Then, 1 ml of mixture was directly added to the sample plate and rapidly dried under a warm stream of air in order to remove­ the organic solvent within seconds.

All samples were analyzed using a Voyager-DE STR MALDI-TOF mass spectrometer (ABI Applied Biosystem, Framingham, USA) with a 337 nm N2 UV laser. The mass spectra of the samples were obtained in positive ion mode. Mass/charge (m/z) ratios were measured in the reflector/delayed extraction mode with an accelerating voltage of 20 kV, grid voltage of 67% and delay time of 100 ns. C2-dihydroceramide (MW 343.6) was used to calibrate the instrument. All sample lipid spectra were acquired using a low-mass gate at 400 Da. For each sample, 6 or 7 spectra were obtained; only when a peak appeared in at least 5 spectra with relatively stable intensity was it considered a candidate for analysis. All MS data were analyzed as described­ before [15,16].

 

 

Results

 

 

Establishment of MS data analysis protocol

 

In order to establish a working protocol for analyzing MS data for sphingolipids, up to 10 different sphingolipid species (natural or synthetic) were subjected to MS, and the major peaks from resulting mass spectra were calculated­ to deduce the possible chemical structures. The major­ peaks from two sphingolipids molecules, C8-ceramide and C8-dihydroceramide, are listed in Table 1 and Table 2, respectively. 25 major peaks were generated by C8-ceramide during the ionization process, of which most were from the matrix 2,5-DHB. m/z 425 (425.6896) corresponded­ to the intact C8-ceramide. However, the relative­ intensity of this peak was only 14.17%; on the other hand, m/z 407 (407.7233) had a relative intensity of 100%. Based on calculation it was concluded that m/z 407 could stand for the fragment of C8-ceramide with an H2O loss, suggesting that most C8-ceramide lost one molecule of water during ionization. Further chemical structural analysis gave two possible structures for this fragment [Fig. 1(A)]. The ionization process can even break the whole octal carbon sidechain away from a small portion­ (4.12%) of C8-ceramide, resulting in the formation of a D-sphingosine-like fragment, which corresponded to m/z 281 (281.3613). Two isotope peaks were also present for m/z 407 and one for 425 (Table 1). Similar analysis was also conducted for C8-dihydroceramide (Table 2), and the possible chemical structures for some fragments are depicted in Fig. 1(B). Together, these processes formulated the basic protocol for sphingolipids MS data analysis. 

 

MNNG induces dramatic changes in sphingomyelin metabolism

 

Previously we have shown that MNNG can induce changes in ceramide metabolism [12,13]. As sphingo­myelin is closely associated with ceramide, we further examined the cellular sphingomyelin metabolism using MALDI-TOF MS. The major sphingomyelin peaks obtained from control, DMSO-treated, and MNNG-treated cells are listed in Table 3. It was found that while DMSO had only a minor effect on sphingomyelin metabolism, there were significant differences between MNNG-treated and control­ samples for sphingomyelin. For example, m/z 778 was not present in control but appeared after MNNG treatment; whereas m/z 782 showed up in control­ but disappeared after MNNG treatment (Table 3). In addition, the concentrations of several sphingomyelin species, including m/z 770, 784, 805 and 814, were increased. The mass spectra data for some sphingomyelin species [Fig. 2(A), m/z 782, 784, 805 and 814] and possible structures for some of the identified sphingomyelin species are also presented­ (Table 4).

 

MNNG induces the redistribution of ASM

 

ASM is responsible for hydrolyzing sphingomyelin to generate ceramide, and its translocation is usually associated with its activation [17-19]. Using immunofluorescent microscopy, the distribution of ASM and its relationship with lipid rafts were studied. ASM exhibited a diffused, even distribution in control cells [Fig. 3(A)] and DMSO solvent control (data not shown). However, MNNG treatment caused the "polarization" of ASM, which concentrated on one side of the cell [Fig. 3(A)]. In addition, ASM colocalized with lipid raft, which was labeled by cholera toxin B. This observation implied that ASM might be involved­ in the altered sphingomyelin metabolism.

 

Imipramine induces the accumulation of sphingo­myelin and inhibits some of the effects of MNNG on sphingomyelin

 

Imipramine is known to inhibit ASM activity [20]. MNNG-induced changes in sphingomyelin may be a result­ of ASM activation, therefore cells were pre-incubated with imipramine followed with MNNG treatment and, after sphingomyelin extraction, the mass spectra were compared­ with those without imipramine. It was found that imipramine alone could cause the accumulation of several sphingomyelin species, indicating that it inhibited the hydrolysis of sphingomyelin (Table 3). Furthermore, it diminished some effects of MNNG on sphingomyelin. For example, the increases of m/z 770 and 784 by MNNG treatment were reversed by imipramine pre-incubation, and the disappeared­ m/z 782 was restored [Table 3, Fig. 2(B)]. Furthermore, imipramine also prevented the polarization of ASM induced by MNNG, implying the inactivation of ASM [Fig. 3(B)].

 

 

Discussion

 

Many sphingolipid molecules, such as ceramide, sphingosine and sphingosine-1-phosphate, are increasingly recognized­ as important modulators of many cellular processes. For example, ceramide has been shown to function­ as a second messenger for Fas, tumor necrosis factor, interleukin (IL-1) and other cytokines, as well as many other extracellular stimuli, usually with the result of either cell cycle arrest or apoptosis [9,13,21,22]. However, unlike nucleotides and proteins, lipids and sphingolipids, have long been a group of molecules that are difficult to study. Sphingolipids were named after the famous Egyptian statue "Sphinx" for their mystical properties. The bottleneck in research was due to the lack of suitable technologies for analyzing the vast number of lipid species, even less the high-throughput technology for systems biology.

The breakthrough came after the application of MS to lipid study, in which many molecules from the lipidome could be directly characterized and quantitated [8,10,11]. Using this method, we have shown that MNNG can affect­ the metabolism of a major sphingolipid species, ceramide [12]. This change of metabolism is associated with some of the cellular effects of MNNG, such as membrane receptor­ clustering [12]. In this study, we further evaluated­ the metabolism of sphingomyelin, which can be hydrolyzed by ASM to generate ceramide [9]. It was found that, similar to ceramide, sphingomyelin metabolism was also affected by MNNG treatment (Table 3), indicating that MNNG may have a global effect on sphingolipids metabolism.

More importantly, using this MS approach, the different­ sphingomyelin species could be identified, and the changes for each species measured. For instance, eight distinct m/z ratios were identified, with each m/z ratio representing one or more possible molecular structures (Table 3). The differences in sidechain length, as well as the number of unsaturated bonds, may influence the function of a specific­ lipid molecule. Therefore, this type of analysis provides invaluable information that cannot be obtained using traditional methods.

The applicability of this technique was further validated by the imipramine experiment. As an inhibitor for ASM, it was expected that imipramine would inhibit the hydro­lysis of sphingomyelin, thus increasing the cellular content­ of sphingomyelin. Indeed it was found that imipramine­ treatment­ caused the accumulation of several sphingomyelin­ species, particularly m/z 748 and 782 [Table 3, Fig. 2(B)]. In addition, imipramine pre-incubation interfered­ with the effect of MNNG on sphingomyelin, indicating that MNNG probably elicited its effect through the action of ASM. This was also supported by the immunfluorescent microscopy data, as MNNG treatment triggered the relocation of ASM, while imipramine prevented­ it (Fig. 3).

Some problems do exist for the MS method. For example, except for a few m/z ratios, exact chemical structures­ could not be deduced precisely; instead, several­ possibilities­ were formulated. Furthermore, when standard­ sphingolipids were subjected to MALDI-TOF analysis, several fragments were generated for each standard (for example, m/z 407 and 281 for C8-ceramide). It would be difficult to tell if these fragments were the original forms presented in the sample or just fragments generated from other molecules during the ionization process. The presence­ of matrix peaks complicates the analysis even further. Finally, the reproducibility of mass spectra should be carefully­ handled. Mass spectrometry is a very sensitive­ method and efforts should be taken to minimize the variations­ which might affect the analyses.

Compared with MALDI-TOF, liquid chromatography-electrospray ionization MS (LC-ESI MS) may prove to be a better solution. First, there is no need for matrices in the analysis. Secondly, its "soft" ionization process, generally, would not fragment the samples. Therefore, LC-ESI MS has far less "noise" than MALDI-TOF MS. Nevertheless, there is the possibility that two molecules have distinct structures but the same molecular weight. To solve this problem, Han and Cheng developed a 2-D ESI MS/MS method [10]. Through lipid class-selective intrasource ionization­ and subsequent analysis of 2-D cross-peak intensities, the chemical identity and mass composition of individual molecular species of most lipid classes can be determined [10].

In summary, the data presented here demonstrated that MALDI-TOF MS is a powerful tool in lipid research. Together­ with the 2-D ESI MS/MS method, these techniques­ provide a strong foundation for the automated analysis of lipid mass spectra data, which will help to push the study of systems biology to a new level.

 

 

Acknowledgements

 

The authors gratefully thank Dr. T. Taketomi for providing detailed instructions for analyzing the MALDI-TOF mass spectrometry data, and Dr. X. Han for the helpful discussion regarding lipidomics.

 

 

References

 

 1   Pognan F. Genomics, proteomics and metabonomics in toxicology: Hopefully­ not 'fashionomics'. Pharmacogenomics 2004, 5: 879-893

 2   Kramer R, Cohen D. Functional genomics to new drug targets. Nat Rev Drug Discov 2004, 3: 965-972

 3   Aggarwal K, Lee KH. Functional genomics and proteomics as a foundation for systems biology. Brief Funct Genomic Proteomic 2003, 2: 175-184

 4   Kell DB. Metabolomics and systems biology: Making sense of the soup. Curr Opin Microbiol 2004, 7: 296-307

 5   Han X, Gross RW. Global analyses of cellular lipidomes directly from crude extracts of biological samples by ESI mass spectrometry: A bridge to lipidomics. J Lipid Res 2003, 44: 1071-1079

 6   Lagarde M, Geloen A, Record M, Vance D, Spener F. Lipidomics is emerging. Biochim Biophys Acta 2003, 1634: 61

 7   Fahy E, Subramaniam S, Brown HA, Glass CK, Merrill AH Jr, Murphy RC, Raetz CR et al. A comprehensive classification system for lipids. J Lipid Res 2005, 46: 839-862

 8   Han X, Yang J, Cheng H, Ye H, Gross RW. Toward fingerprinting cellular lipidomes directly from biological samples by two-dimensional electrospray ionization mass spectrometry. Anal Biochem 2004, 330: 317-331

 9   Yang J, Yu Y, Sun S, Duerksen-Hughes PJ. Ceramide and other sphingolipids in cellular responses. Cell Biochem Biophys 2004, 40: 323-350

10  Han X, Cheng H. Characterization and direct quantitation of cerebroside molecular species from lipid extracts by shotgun lipidomics. J Lipid Res 2005, 46: 163-175

11  Han X, Gross RW. Shotgun lipidomics: Electrospray ionization mass spectrometric­ analysis and quantitation of cellular lipidomes directly from crude extracts of biological samples. Mass Spectrom Rev 2005, 24: 367-412

12  Huang Y, Yang J, Shen J, Chen F, Yu Y. Sphingolipids are involved in N-methyl-N'-nitro-N-nitrosoguanidine-induced epidermal growth factor receptor­ clustering. Biochem Biophys Res Commun 2005, 330: 430-438

13  Yang J, Duerksen-Hughes PJ. Activation of a p53-independent, sphingolipid-mediated cytolytic pathway in p53-negative mouse fibroblast cells treated with N-methyl-N-nitro-N-nitrosoguanidine. J Biol Chem 2001, 276: 27129-27135

14 Gao Z, Yang J, Huang Y, Yu Y. N-methyl-N'-nitro-N-nitrosoguanidine interferes­ with the epidermal growth factor receptor-mediated signaling pathway. Mutat Res 2005, 570: 175-184

15  Fujiwaki T, Yamaguchi S, Tasaka M, Sakura N, Taketomi T. Application of delayed extraction-matrix-assisted laser desorption ionization time-of-flight mass spectrometry for analysis of sphingolipids in pericardial fluid, peritoneal fluid and serum from Gaucher disease patients. J Chromatogr B Analyt Technol Biomed Life Sci 2002, 776: 115-123

16  Fujiwaki T, Yamaguchi S, Sukegawa K, Taketomi T. Application of delayed extraction matrix-assisted laser desorption ionization time-of-flight mass spectrometry­ for analysis of sphingolipids in cultured skin fibroblasts from sphingolipidosis patients. Brain Dev 2002, 24: 170-173

17  Grassme H, Cremesti A, Kolesnick R, Gulbins E. Ceramide-mediated clustering­ is required for CD95-DISC formation. Oncogene 2003, 22: 5457-5470

18  Gulbins E, Grassme H. Ceramide and cell death receptor clustering. Biochim Biophys Acta 2002, 1585: 139-145

19  Gulbins E. Regulation of death receptor signaling and apoptosis by ceramide. Pharmacol Res 2003, 47: 393-399

20  Lacour S, Hammann A, Grazide S, Lagadic-Gossmann D, Athias A, Sergent O, Laurent G et al. Cisplatin-induced CD95 redistribution into membrane lipid rafts of HT29 human colon cancer cells. Cancer Res 2004, 64: 3593-3598

21  Pru JK, Hendry IR, Davis JS, Rueda BR. Soluble Fas ligand activates the sphingomyelin pathway and induces apoptosis in luteal steroidogenic cells independently of stress-activated p38(MAPK). Endocrinology 2002, 143: 4350-4357

22  Luberto C, Hassler DF, Signorelli P, Okamoto Y, Sawai H, Boros E, Hazen-Martin DJ et al. Inhibition of tumor necrosis factor-induced cell death in MCF7 by a novel inhibitor of neutral sphingomyelinase. J Biol Chem 2002, 277: 41128-41139