DOI QR코드

DOI QR Code

Development of a Label-Free LC-MS/MS-Based Glucosylceramide Synthase Assay and Its Application to Inhibitors Screening for Ceramide-Related Diseases

  • Fu, Zhicheng (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Yun, So Yoon (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Won, Jong Hoon (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Back, Moon Jung (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Jang, Ji Min (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Ha, Hae Chan (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Lee, Hae Kyung (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Shin, In Chul (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Kim, Ju Yeun (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Kim, Hee Soo (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University) ;
  • Kim, Dae Kyong (Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University)
  • 투고 : 2018.07.04
  • 심사 : 2018.08.09
  • 발행 : 2019.03.01

초록

Ceramide metabolism is known to be an essential etiology for various diseases, such as atopic dermatitis and Gaucher disease. Glucosylceramide synthase (GCS) is a key enzyme for the synthesis of glucosylceramide (GlcCer), which is a main ceramide metabolism pathway in mammalian cells. In this article, we developed a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method to determine GCS activity using synthetic non-natural sphingolipid C8-ceramide as a substrate. The reaction products, C8-GlcCer for GCS, could be separated on a C18 column by reverse-phase high-performance liquid chromatography (HPLC). Quantification was conducted using the multiple reaction monitoring (MRM) mode to monitor the precursor-to-product ion transitions of m/z $588.6{\rightarrow}264.4$ for C8-GlcCer at positive ionization mode. The calibration curve was established over the range of 0.625-160 ng/mL, and the correlation coefficient was larger than 0.999. This method was successfully applied to detect GCS in the human hepatocellular carcinoma cell line (HepG2 cells) and mouse peripheral blood mononuclear cells. We also evaluated the inhibition degree of a known GCS inhibitor 1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP) on GCS enzymatic activity and proved that this method could be successfully applied to GCS inhibitor screening of preventive and therapeutic drugs for ceramide metabolism diseases, such as atopic dermatitis and Gaucher disease.

키워드

참고문헌

  1. Abe, A., Inokuchi, J., Jimbo, M., Shimeno, H., Nagamatsu, A., Shayman, J. A., Shukla, G. S. and Radin, N. S. (1992) Improved inhibitors of glucosylceramide synthase. J. Biochem. 111, 191-196. https://doi.org/10.1093/oxfordjournals.jbchem.a123736
  2. Abe, A., Radin, N. S., Shayman, J. A., Wotring, L. L., Zipkin, R. E., Sivakumar, R., Ruggieri, J. M., Carson, K. G. and Ganem, B. (1995) Structural and stereochemical studies of potent inhibitors of glucosylceramide synthase and tumor cell growth. J. Lipid Res. 36, 611-621. https://doi.org/10.1016/S0022-2275(20)39895-3
  3. Basu, S., Kaufman, B. and Roseman, S. (1973) Enzymatic synthesis of glucocerebroside by a glucosyltransferase from embryonic chicken brain. J. Biol. Chem. 248, 1388-1394. https://doi.org/10.1016/S0021-9258(19)44311-1
  4. Bennett, L. L. and Mohan, D. (2013) Gaucher disease and its treatment options. Ann. Pharmacother. 47, 1182-1193. https://doi.org/10.1177/1060028013500469
  5. Bleicher, R. J. and Cabot, M. C. (2002) Glucosylceramide synthase and apoptosis. Biochim. Biophys. Acta 1585, 172-178. https://doi.org/10.1016/S1388-1981(02)00338-4
  6. Chatterjee, S. and Alsaeedi, N. (2012) Lactosylceramide synthase as a therapeutic target to mitigate multiple human diseases in animal models. Adv. Exp. Med. Biol. 749, 153-169. https://doi.org/10.1007/978-1-4614-3381-1_11
  7. Choi, M. J. and Maibach, H. I. (2005) Role of ceramides in barrier function of healthy and diseased skin. Am. J. Clin. Dermatol. 6, 215-223. https://doi.org/10.2165/00128071-200506040-00002
  8. Chujor, C. S., Feingold, K. R., Elias, P. M. and Holleran, W. M. (1998) Glucosylceramide synthase activity in murine epidermis: quantitation, localization, regulation, and requirement for barrier homeostasis. J. Lipid Res. 39, 277-285. https://doi.org/10.1016/S0022-2275(20)33889-X
  9. Degroote, S., Wolthoorn, J. and van Meer, G. (2004) The cell biology of glycosphingolipids. Semin. Cell Dev. Biol. 15, 375-387. https://doi.org/10.1016/j.semcdb.2004.03.007
  10. Di Sano, F., Di Bartolomeo, S., Fazi, B., Fiorentini, C., Matarrese, P., Spinedi, A. and Piacentini, M. (2002) Antisense to glucosylceramide synthase in human neuroepithelioma affects cell growth but not apoptosis. Cell Death Differ. 9, 693-695. https://doi.org/10.1038/sj.cdd.4401040
  11. Duclos, R. I. (2001) The total syntheses of D-erythro-sphingosine, N-palmitoylsphingosine (ceramide), and glucosylceramide (cerebroside) via an azidosphingosine analog. Chem. Phys. Lipids 111, 111-138. https://doi.org/10.1016/S0009-3084(01)00152-9
  12. Ekiz, H. A. and Baran, Y. (2010) Therapeutic applications of bioactive sphingolipids in hematological malignancies. Int. J. Cancer 127, 1497-1506. https://doi.org/10.1002/ijc.25478
  13. Hakomori, S. I. (2008) Structure and function of glycosphingolipids and sphingolipids: recollections and future trends. Biochim. Biophys. Acta 1780, 325-346. https://doi.org/10.1016/j.bbagen.2007.08.015
  14. Hayashi, Y., Horibata, Y., Sakaguchi, K., Okino, N. and Ito, M. (2005) A sensitive and reproducible assay to measure the activity of glucosylceramide synthase and lactosylceramide synthase using HPLC and fluorescent substrates. Anal. Biochem. 345, 181-186. https://doi.org/10.1016/j.ab.2005.05.029
  15. Hospattankar, A. V. and Radin, N. S. (1981) A 2-phase liquid scintillation assay for glycolipid synthetases. Lipids 16, 764-766. https://doi.org/10.1007/BF02535347
  16. Ichikawa, S., Sakiyama, H., Suzuki, G., Hidari, K. I. and Hirabayashi, Y. (1996) Expression cloning of a cDNA for human ceramide glucosyltransferase that catalyzes the first glycosylation step of glycosphingolipid synthesis. Proc. Natl. Acad. Sci. U.S.A. 93, 12654.
  17. Imokawa, G. (2009) A possible mechanism underlying the ceramide deficiency in atopic dermatitis: expression of a deacylase enzyme that cleaves the N-acyl linkage of sphingomyelin and glucosylceramide. J. Dermatol. Sci. 55, 1-9. https://doi.org/10.1016/j.jdermsci.2009.04.006
  18. Kartal Yandim, M., Apohan, E. and Baran, Y. (2013) Therapeutic potential of targeting ceramide/glucosylceramide pathway in cancer. Cancer Chemother. Pharmacol. 71, 13-20. https://doi.org/10.1007/s00280-012-1984-x
  19. Kolesnick, R. (2002) The therapeutic potential of modulating the ceramide/ sphingomyelin pathway. J. Clin. Invest. 110, 3-8. https://doi.org/10.1172/JCI0216127
  20. Limgala, R. P., Ioanou, C., Plassmeyer, M., Ryherd, M., Kozhaya, L., Austin, L., Abidoglu, C., Unutmaz, D., Alpan, O. and Goker-Alpan, O. (2016) Time of initiating enzyme replacement therapy affects immune abnormalities and disease severity in patients with Gaucher disease. PLoS ONE 11, e0168135. https://doi.org/10.1371/journal.pone.0168135
  21. Liu, Y. Y., Han, T. Y., Yu, J. Y., Bitterman, A., Le, A., Giuliano, A. E. and Cabot, M. C. (2004) Oligonucleotides blocking glucosylceramide synthase expression selectively reverse drug resistance in cancer cells. J. Lipid Res. 45, 933-940. https://doi.org/10.1194/jlr.M300486-JLR200
  22. Liu, Y. Y., Hill, R. A. and Li, Y. T. (2013) Ceramide glycosylation catalyzed by glucosylceramide synthase and cancer drug resistance. Adv. Cancer Res. 117, 59-89. https://doi.org/10.1016/B978-0-12-394274-6.00003-0
  23. Maunula, S., Bjorkqvist, Y. J., Slotte, J. P. and Ramstedt, B. (2007) Differences in the domain forming properties of N-palmitoylated neutral glycosphingolipids in bilayer membranes. Biochim. Biophys. Acta 1768, 336-345. https://doi.org/10.1016/j.bbamem.2006.09.003
  24. Mishra, S. and Chatterjee, S. (2014) Lactosylceramide promotes hypertrophy through ROS generation and activation of ERK1/2 in cardiomyocytes. Glycobiology 24, 518-531. https://doi.org/10.1093/glycob/cwu020
  25. Miura, T., Kajimoto, T., Jimbo, M., Yamagishi, K., Inokuchi, J. C. and Wong, C. H. (1998) Synthesis and evaluation of morpholino- and pyrrolidinosphingolipids as inhibitors of glucosylceramide synthase. Bioorg. Med. Chem. 6, 1481-1489. https://doi.org/10.1016/S0968-0896(98)00077-7
  26. Mizutani, Y., Mitsutake, S., Tsuji, K., Kihara, A. and Igarashi, Y. (2009) Ceramide biosynthesis in keratinocyte and its role in skin function. Biochimie 91, 784-790. https://doi.org/10.1016/j.biochi.2009.04.001
  27. Nagral, A. (2014) Gaucher disease. J. Clin. Exp. Hepatol. 4, 37-50. https://doi.org/10.1016/j.jceh.2014.02.005
  28. Radin, N. S. (1996) Treatment of Gaucher disease with an enzyme inhibitor. Glycoconj. J. 13, 153-157. https://doi.org/10.1007/BF00731489
  29. Senchenkov, A., Litvak, D. A. and Cabot, M. C. (2001) Targeting ceramide metabolism-a strategy for overcoming drug resistance. J. Natl. Cancer Inst. 93, 347-357. https://doi.org/10.1093/jnci/93.5.347
  30. Shawky, R. M. and Elsayed, S. M. (2016) Treatment options for patients with Gaucher disease. Egypt. J. Med. Hum. Genet. 17, 281-285. https://doi.org/10.1016/j.ejmhg.2016.02.001
  31. Sietsma, H., Veldman, R. J. and Kok, J. W. (2001) The involvement of sphingolipids in multidrug resistance. J. Membr. Biol. 181, 153-162. https://doi.org/10.1007/s00232-001-0033-1
  32. Tuuf, J., Kjellberg, M. A., Molotkovsky, J. G., Hanada, K. and Mattjus, P. (2011) The intermembrane ceramide transport catalyzed by CERT is sensitive to the lipid environment. Biochim. Biophys. Acta 1808, 229-235. https://doi.org/10.1016/j.bbamem.2010.09.011
  33. van Meer, G. and Holthuis, J. C. (2000) Sphingolipid transport in eukaryotic cells. Biochim. Biophys. Acta 1486, 145-170. https://doi.org/10.1016/S1388-1981(00)00054-8
  34. Vielhaber, G., Pfeiffer, S., Brade, L., Lindner, B., Goldmann, T., Vollmer, E., Hintze, U., Wittern, K. P. and Wepf, R. (2001) Localization of ceramide and glucosylceramide in human epidermis by immunogold electron microscopy. J. Invest. Dermatol. 117, 1126-1136. https://doi.org/10.1046/j.0022-202x.2001.01527.x
  35. Vunnam, R. R. and Radin, N. S. (1980) Analogs of ceramide that inhibit glucocerebroside synthetase in mouse brain. Chem. Phys. Lipids 26, 265-278. https://doi.org/10.1016/0009-3084(80)90057-2
  36. Wang, J., Lv, X. W. and Du, Y. G. (2009) Potential mechanisms involved in ceramide-induced apoptosis in human colon cancer HT29 cells. Biomed. Environ. Sci. 22, 76-85. https://doi.org/10.1016/S0895-3988(09)60026-X
  37. Xie, P., Shen, Y. F., Shi, Y. P., Ge, S. M., Gu, Z. H., Wang, J., Mu, H. J., Zhang, B., Qiao, W. Z. and Xie, K. M. (2008) Overexpression of glucosylceramide synthase in associated with multidrug resistance of leukemia cells. Leuk. Res. 32, 475-480. https://doi.org/10.1016/j.leukres.2007.07.006
  38. Yoshizaki, F., Nakayama, H., Iwahara, C., Takamori, K., Ogawa, H. and Iwabuchi, K. (2008) Role of glycosphingolipid-enriched microdomains in innate immunity: microdomain-dependent phagocytic cell functions. Biochim. Biophys. Acta 1780, 383-392. https://doi.org/10.1016/j.bbagen.2007.11.004
  39. Zheng, W., Kollmeyer, J., Symolon, H., Momin, A., Munter, E., Wang, E., Kelly, S., Allegood, J. C., Liu, Y., Peng, Q., Ramaraju, H., Sullards, M. C., Cabot, M. and Merrill, A. H., Jr. (2006) Ceramides and other bioactive sphingolipid backbones in health and disease: lipidomic analysis, metabolism and roles in membrane structure, dynamics, signaling and autophagy. Biochim. Biophys. Acta 1758, 1864-1884. https://doi.org/10.1016/j.bbamem.2006.08.009

피인용 문헌

  1. Mapping Sphingolipid Metabolism Pathways during Phagosomal Maturation vol.16, pp.12, 2021, https://doi.org/10.1021/acschembio.1c00393