Sphingolipid and Glycosphingolipid Metabolic Pathways in the Era of SphingolipidomicsClick to copy article linkArticle link copied!
SPECIAL ISSUE
This article is part of the
Introduction
An Overview of Sphingolipid Structure and Function
Backbone and Headgroup Nomenclature
Variation in the Lipid Moieties
Sphingoid Base Diversity
N-Acyl-sphingoid Bases (Ceramides)
Variation of the Complex Sphingolipid Headgroups
Phosphosphingolipids
Glycosphingolipids
Other Types of Compounds
Sphingolipid Metabolic Pathways
Biosynthesis of the Lipid Moieties de Novo
Formation of the Sphingoid Base Backbones
Ceramide Synthases
Desaturation and Hydroxylation of Dihydroceramide to Form Ceramides and 4-Hydroxyceramides (Phytoceramides)
Complex Sphingolipid Biosynthesis
Sphingomyelin, Ceramide Phosphoethanolamine, and Ceramide Phosphate
Other Non-Glycan Headgroups
Glycosphingolipids
Biosynthesis of GlcCer
Biosynthesis of LacCer
Biosynthesis of Ganglio-Series Glycosphingolipids
Biosynthesis of Lacto-/Neolacto-Series Glycosphingolipids
Biosynthesis of Globo-/Isoglobo-Series Glycosphingolipids
Biosynthesis of GalCer
Sulfated Glycosphingolipids
Integration of Backbone and Headgroup Biosynthetic Pathways
Sphingolipid Turnover, Trafficking, and Recycling
Metabolic Turnover
Sphingolipid Trafficking and Membrane Dynamics
Sphingolipid Recycling (Salvage Pathways)
Analysis of Sphingolipid Metabolism by “Omic” Technologies
Use of Mass Spectrometry for Sphingolipidomics
Tissue-Imaging Mass Spectrometry of Sphingolipids
Integration of “Omic” Data Sets for a Systems Biology of Sphingolipid Metabolism and Function
Visualization Tools
Mathematical Modeling
Perspective on the Current State of Sphingolipid Research
Biography
Acknowledgment
This chapter draws heavily on ideas that the author has absorbed from laboratory colleagues (students and postdoctoral fellows, long-time associate Elaine Wang, and collaborators M. Cameron Sullards and May Wang), and gleaned from works by, and fruitful conversations with, Tony Futerman, Yusuf Hannun, Konrad Sandhoff, Walt Shaw, Jim Shayman, Sarah Spiegel, Akemi Suzuki and too many other experts on the topics to cite them adequately. Therefore, heartfelt thanks are due to them all, as well as to the reviewers of this manuscript for Chemical Reviews. Any errors are, of course, the author’s fault. Support from the NIH is gratefully acknowledged (NIH GM069338 “Lipid Maps,” GM76217 and CA137812), as is the Smithgall Institute endowment for the chair in Molecular Cell Biology at Georgia Tech that has facilitated some of the studies described herein.
References
This article references 639 other publications.
- 1Thudichum, J. L. W. A Treatise on the Chemical Constitution of Brain; Bailliere, Tindall, and Cox: London, 1884.Google ScholarThere is no corresponding record for this reference.
- 2Carter, H. E.; Haines, W. J.; Ledyard, W. E.; Norris, W. P. J. Biol. Chem. 1947, 169, 77Google ScholarThere is no corresponding record for this reference.
- 3Merrill, A. H., Jr.; Wang, M. D.; Park, M.; Sullards, M. C. Trends Biochem. Sci. 2007, 32, 457Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtF2lsbrJ&md5=b890217b97e9ecb075367b925f6f4529(Glyco)sphingolipidology: an amazing challenge and opportunity for systems biologyMerrill, Alfred H.; Wang, May Dongmei; Park, Meeyoung; Sullards, M. CameronTrends in Biochemical Sciences (2007), 32 (10), 457-468CODEN: TBSCDB; ISSN:0968-0004. (Elsevier B.V.)A review. Sphingolipids are found in essentially all eukaryotes and in some prokaryotes and viruses, where they influence cell structure, signaling and interactions with the extracellular environment. Because of the combinatorial nature of their biosynthesis, the sphingolipidome comprises untold thousands of species that encompass bioactive backbones and complex phospho- and glycolipids. Mass spectrometry is able to analyze a growing fraction of the sphingolipidome and is beginning to provide information about localization. Use of these structure specific, quant. methods is producing insights, and surprises, regarding sphingolipid structure, metab., function and disease. Dealing with such large data sets poses special challenges for systems biol., but the intrinsic and elegant interrelationships among these compds. might provide a key to dealing with the complexity of the sphingolipidome.
- 4Sonnino, S.; Prinetti, A.; Mauri, L.; Chigorno, V.; Tettamanti, G. Chem. Rev. 2006, 106, 2111Google ScholarThere is no corresponding record for this reference.
- 5Bartke, N.; Hannun, Y. A. J. Lipid Res. 2009, 50 (Suppl) S91Google ScholarThere is no corresponding record for this reference.
- 6Strub, G. M.; Maceyka, M.; Hait, N. C.; Milstien, S.; Spiegel, S. Adv. Exp. Med. Biol. 2010, 688, 141Google ScholarThere is no corresponding record for this reference.
- 7Lopez, P. H.; Schnaar, R. L. Curr. Opin. Struct. Biol. 2009, 19, 549Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1ylsL3N&md5=484bd0ed90820692b5d94663962264ccGangliosides in cell recognition and membrane protein regulationLopez, Pablo H. H.; Schnaar, Ronald L.Current Opinion in Structural Biology (2009), 19 (5), 549-557CODEN: COSBEF; ISSN:0959-440X. (Elsevier B.V.)A review. Gangliosides, sialic acid-bearing glycosphingolipids, are expressed on all vertebrate cells, and are the major glycans on nerve cells. They are anchored to the plasma membrane through their ceramide lipids with their varied glycans extending into the extracellular space. Through sugar-specific interactions with glycan-binding proteins on apposing cells, gangliosides function as receptors in cell-cell recognition, regulating natural killer cell cytotoxicity via Siglec-7, myelin-axon interactions via Siglec-4 (myelin-assocd. glycoprotein), and inflammation via E-selectin. Gangliosides also interact laterally in their own membranes, regulating the responsiveness of signaling proteins including the insulin, epidermal growth factor, and vascular endothelial growth factor receptors. In these ways, gangliosides act as regulatory elements in the immune system, in the nervous system, in metabolic regulation, and in cancer progression.
- 8Yu, R. K.; Nakatani, Y.; Yanagisawa, M. J. Lipid Res. 2009, 50 (Suppl) S440Google ScholarThere is no corresponding record for this reference.
- 9Eckhardt, M. Mol. Neurobiol. 2008, 37, 93Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXptFOrurw%253D&md5=afb7cc79922c9071771c952427726760The role and metabolism of sulfatide in the nervous systemEckhardt, MatthiasMolecular Neurobiology (2008), 37 (2-3), 93-103CODEN: MONBEW; ISSN:0893-7648. (Humana Press Inc.)A review. 3-O-sulfogalactosylceramide or sulfatide is a major component of the myelin sheath in the central and peripheral nervous system. The examn. of mice deficient in the sulfatide-synthesizing enzyme, cerebroside sulfotransferase, has provided new insight into the role of sulfatide in the differentiation of myelinating cells, formation of the paranodal junction, and myelin maintenance. Although in general regarded as a marker for oligodendrocytes and Schwann cells, sulfatide is also present in astrocytes and neurons. The relatively low amt. of sulfatide in neurons can dramatically increase in the absence of the sulfatide-degrading enzyme, arylsulfatase A, as in metachromatic leukodystrophy. Recent advances in the understanding of this disease comes from studies on new transgenic mouse models. Significant changes in sulfatide levels have also been obsd. more recently in Alzheimer's disease and other diseases, suggesting that sulfatide might be involved in the pathogenesis of these diseases as well. This review summarizes recent studies on the physiol. and pathophysiol. role of sulfatide using transgenic mice deficient in its synthesis or degrdn.
- 10Lingwood, C. A.; Binnington, B.; Manis, A.; Branch, D. R. FEBS Lett. 2010, 584, 1879Google ScholarThere is no corresponding record for this reference.
- 11Pruett, S. T.; Bushnev, A.; Hagedorn, K.; Adiga, M.; Haynes, C. A.; Sullards, M. C.; Liotta, D. C.; Merrill, A. H., Jr. J. Lipid Res. 2008, 49, 1621Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXovVCrs7Y%253D&md5=960698c280f321d4a7010ad139a5436cBiodiversity of sphingoid bases (sphingosines) and related amino alcoholsPruett, Sarah T.; Bushnev, Anatoliy; Hagedorn, Kerri; Adiga, Madhura; Haynes, Christopher A.; Sullards, M. Cameron; Liotta, Dennis C.; Merrill, Alfred H., Jr.Journal of Lipid Research (2008), 49 (8), 1621-1639CODEN: JLPRAW; ISSN:0022-2275. (American Society for Biochemistry and Molecular Biology, Inc.)A review. Sphingosin was first described by J. L. W. Thudichum in 1884 and structurally characterized as 2S,3R,4E-2-aminooctadec-4-ene-1,3-diol in 1947 by Herb Carter, who also proposed the designation of lipids derived from sphingosine as sphingolipids. This category of amino alcs. is now known to encompass hundreds of compds. that are referred to as sphingoid bases and sphingoid base-like compds., which vary in chain length, no., position, and stereochem. of double bonds, hydroxyl groups, and other functionalities. Some have esp. intriguing features, such as the tail-to-tail combination of 2 sphingoid bases in the α,ω-sphingoids produced by sponges. Most of these compds. participate in cell structure and regulation, and some (such as the fumonisins) disrupt normal sphingolipid metab. and cause plant and animal disease. Many of the naturally occurring and synthetic sphingoid bases are cytotoxic for cancer cells and pathogenic microorganisms or have other potentially useful bioactivities; hence, they offer promise as pharmaceutical leads. This thematic review gives an overview of the biodiversity of the backbones of sphingolipids and the broader field of naturally occurring and synthetic sphingoid base-like compds.
- 12Varki, A.; Cummings, R. D.; Esko, J. D.; Freeze, H. H.; Stanley, P.; Marth, J. D.; Bertozzi, C. R.; Hart, G. W.; Etzler, M. E. Proteomics 2009, 9, 5398Google ScholarThere is no corresponding record for this reference.
- 13Varki, A. Trends Mol. Med. 2008, 14, 351Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXpsVCltrk%253D&md5=fc4d8eb51bcc4509dc8b03cf7533a1c1Sialic acids in human health and diseaseVarki, AjitTrends in Molecular Medicine (2008), 14 (8), 351-360CODEN: TMMRCY; ISSN:1471-4914. (Elsevier B.V.)A review. The surfaces of all vertebrate cells are decorated with a dense and complex array of sugar chains, which are mostly attached to proteins and lipids. Most sol. secreted proteins are also similarly decorated with such glycans. Sialic acids are a diverse family of sugar units with a nine-carbon backbone that are typically found attached to the outermost ends of these chains. Given their location and ubiquitous distribution, sialic acids can mediate or modulate a wide variety of physiol. and pathol. processes. This review considers some examples of their established and newly emerging roles in aspects of human physiol. and disease.
- 14Kolter, T.; Proia, R. L.; Sandhoff, K. J. Biol. Chem. 2002, 277, 25859Google ScholarThere is no corresponding record for this reference.
- 15Chester, M. A. Eur. J. Biochem. 1998, 257, 293Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXntFWmurc%253D&md5=cdb9d2ada5fa0a093f985b6e8fc9eb38IUPAC-IUB Joint Commission on Biochemical Nomenclature (JCBN) Nomenclature of glycolipids Recommendations 1997Chester, M. AlanEuropean Journal of Biochemistry (1998), 257 (2), 293-298CODEN: EJBCAI; ISSN:0014-2956. (Springer-Verlag)Glycolipids are glycosyl derivs. of lipids. They are collectively part of a larger family of substances known as glycoconjugates. The structures of glycolipids are often complex and difficult to reproduce in the text of articles and cannot be referred to in oral discussions without a nomenclature that implies specific chem. structural features. The nomenclature needs to be convenient and practical, as well as extensible, to accommodate newly discovered structures. The 1997 recommendations for nomenclature of glycolipids from the IUPAC-IUB Joint Commission on Biochem. Nomenclature are presented.
- 16Fahy, E.; Subramaniam, S.; Brown, H. A.; Glass, C. K.; Merrill, A. H., Jr.; Murphy, R. C.; Raetz, C. R.; Russell, D. W.; Seyama, Y.; Shaw, W.; Shimizu, T.; Spener, F.; van Meer, G.; VanNieuwenhze, M. S.; White, S. H.; Witztum, J. L.; Dennis, E. A. J. Lipid Res. 2005, 46, 839Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXktFaqtLY%253D&md5=1ca24f3b244e28edc6509cd08057072fA comprehensive classification system for lipidsFahy, Eoin; Subramaniam, Shankar; Brown, H. Alex; Glass, Christopher K.; Merrill, Alfred H., Jr.; Murphy, Robert C.; Raetz, Christian R. H.; Russell, David W.; Seyama, Yousuke; Shaw, Walter; Shimizu, Takao; Spener, Friedrich; van Meer, Gerrit; VanNieuwenhze, Michael S.; White, Stephen H.; Witztum, Joseph L.; Dennis, Edward A.Journal of Lipid Research (2005), 46 (5), 839-861CODEN: JLPRAW; ISSN:0022-2275. (American Society for Biochemistry and Molecular Biology, Inc.)Lipids are produced, transported, and recognized by the concerted actions of numerous enzymes, binding proteins, and receptors. A comprehensive anal. of lipid mols., "lipidomics," in the context of genomics and proteomics is crucial to understanding cellular physiol. and pathol.; consequently, lipid biol. has become a major research target of the postgenomic revolution and systems biol. To facilitate international communication about lipids, a comprehensive classification of lipids with a common platform that is compatible with informatics requirements has been developed to deal with the massive amts. of data that will be generated by the lipid community. As an initial step in this development, the authors divide lipids into 8 categories (fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, sterol lipids, prenol lipids, saccharolipids, and polyketides) contg. distinct classes and subclasses of mols., devise a common manner of representing the chem. structures of individual lipids and their derivs., and provide a 12 digit identifier for each unique lipid mol. The lipid classification scheme is chem. based and driven by the distinct hydrophobic and hydrophilic elements that compose the lipid. This structured vocabulary will facilitate the systematization of lipid biol. and enable the cataloging of lipids and their properties in a way that is compatible with other macromol. databases.
- 17Schauer, R. Curr. Opin. Struct. Biol. 2009, 19, 507Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1ylsLzO&md5=394b118ae7fa757883ae6532e6889ebcSialic acids as regulators of molecular and cellular interactionsSchauer, RolandCurrent Opinion in Structural Biology (2009), 19 (5), 507-514CODEN: COSBEF; ISSN:0959-440X. (Elsevier B.V.)A review. The wide occurrence of sialic acids (Sia) in various chem. forms linked as monomers or polymers in an outstanding position in a multitude of complex carbohydrates of animals and microorganisms renders them as most versatile function modulators in cell biol. and pathol. A survey is presented of recent advances in the study of the influences that Sias have as bulky hydrophilic and electronegatively charged monosaccharides on animal cells and on their interaction with microorganisms. Some highlights are: sialylation leads to increased anti-inflammatory activity of IgG antibodies, facilitates the escape of microorganisms from the host's immune system, and in polymeric form is involved in the regulation of embryogenesis and neuronal growth and function. The role of siglecs in immunoregulation, the dynamics of lymphocyte binding to selectins and the interactions of toxins, viruses, and other microorganisms with the host's Sia are now better understood. N-Glycolylneuraminic acid from food is antigenic in man and seems to have pathogenic potential. Sia O-acetylation mediated by various eukaryotic and prokaryotic O-acetyltransferases modulates the affinity of these monosaccharides to mammalian and microbial receptors and hinders apoptosis. The functionally versatile O-acetylated ganglioside GD3 is an onco-fetal antigen.
- 18Cantu, L.; Corti, M.; Casellato, R.; Acquotti, D.; Sonnino, S. Chem. Phys. Lipids 1991, 60, 111Google ScholarThere is no corresponding record for this reference.
- 19Yu, R. K.; Yanagisawa, M.; Ariga, T. In Comprehensive Glycoscience. From Chemistry to Systems Biology; Kamerling, J. P., Ed.; Elsevier: Oxford, U.K., 2007; Vol. 1.Google ScholarThere is no corresponding record for this reference.
- 20Nystrom, K.; Grahn, A.; Lindh, M.; Brytting, M.; Mandel, U.; Larson, G.; Olofsson, S. Glycobiology 2007, 17, 355Google ScholarThere is no corresponding record for this reference.
- 21Simons, K.; Gerl, M. J. Nat. Rev. Mol. Cell Biol. 2010, 11, 688Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtFylur%252FI&md5=7dfb5143b2ffb8844954be2b8e270f5eRevitalizing membrane rafts: New tools and insightsSimons, Kai; Gerl, Mathias J.Nature Reviews Molecular Cell Biology (2010), 11 (10), 688-699CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Ten years ago, the authors wrote a review on lipid rafts and signaling. At the time, this field was suffering from ambiguous methodol. and imprecise nomenclature. Now, however, new techniques are deepening the insight into the dynamics of membrane organization. Here, the authors discuss how the field has matured and present an evolving model in which membranes are occupied by fluctuating nanoscale assemblies of sphingolipids, cholesterol, and proteins that can be stabilized into platforms that are important in signaling, viral infection, and membrane trafficking.
- 22Hakomori, Si, S. I. Proc. Natl. Acad. Sci. U. S. A. 2002, 99, 225Google ScholarThere is no corresponding record for this reference.
- 23Goni, F. M.; Alonso, A. Biochim. Biophys. Acta 2009, 1788, 169Google ScholarThere is no corresponding record for this reference.
- 24Luberto, C.; Yoo, D. S.; Suidan, H. S.; Bartoli, G. M.; Hannun, Y. A. J. Biol. Chem. 2000, 275, 14760Google ScholarThere is no corresponding record for this reference.
- 25Singh, R. D.; Marks, D. L.; Pagano, R. E. Curr. Protoc. Cell Biol. 2007, Chapter 24, Unit 24 1.Google ScholarThere is no corresponding record for this reference.
- 26Fyrst, H.; Saba, J. D. Nat. Chem. Biol. 2010, 6, 489Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnsFKitLk%253D&md5=6128ea6a4f1a910deed32f7db8827840An update on sphingosine-1-phosphate and other sphingolipid mediatorsFyrst, Henrik; Saba, Julie D.Nature Chemical Biology (2010), 6 (7), 489-497CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)A review. Sphingolipids comprise a complex family of naturally occurring mols. that are enriched in lipid rafts and contribute to their unique biochem. properties. Membrane sphingolipids also serve as a reservoir for bioactive metabolites including sphingosine, ceramide, sphingosine 1-phosphate (S1P) and ceramide 1-phosphate. Among these, S1P has emerged as a central regulator of mammalian biol. S1P is essential for mammalian brain and cardiac development and for maturation of the systemic circulatory system and lymphatics. In addn., S1P contributes to trafficking and effector functions of lymphocytes and other hematopoietic cells and protects against various forms of tissue injury. However, S1P is also an oncogenic lipid that promotes tumor growth and progression. Recent preclin. and clin. investigations using pharmacol. agents that target S1P, its receptors, and the enzymes required for its biosynthesis and degrdn. demonstrate the promise and potential risks of modulating S1P signaling in treatment strategies for autoimmunity, cancer, cardiovascular disease, and other pathol. conditions.
- 27Milhas, D.; Clarke, C. J.; Hannun, Y. A. FEBS Lett. 2010, 584, 1887Google ScholarThere is no corresponding record for this reference.
- 28Gault, C. R.; Obeid, L. M.; Hannun, Y. A. Adv. Exp. Med. Biol. 2010, 688, 1Google ScholarThere is no corresponding record for this reference.
- 29Merrill, A. H., Jr. J. Biol. Chem. 2002, 277, 25843Google ScholarThere is no corresponding record for this reference.
- 30Ledeen, R. W.; Wu, G. J. Lipid Res. 2008, 49, 1176Google ScholarThere is no corresponding record for this reference.
- 31Zeidan, Y. H.; Hannun, Y. A. Curr. Mol. Med. 2010, 10, 454Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXos1OjsLs%253D&md5=6782ce4346167f30296142138f73d724The acid sphingomyelinase/ceramide pathway: Biomedical significance and mechanisms of regulationZeidan, Y. H.; Hannun, Y. A.Current Molecular Medicine (2010), 10 (5), 454-466CODEN: CMMUBP; ISSN:1566-5240. (Bentham Science Publishers Ltd.)A review. One of the most intriguing enzymes of sphingolipid biol. is acid sphingomyelinase (ASMase). In a phospholipase C reaction, ASMase catalyzes the cleavage of the phosphocholine head group of sphingomyelin to generate ceramide. Cumulative efforts of various labs. over the past 40 years have placed ASMase and its product ceramide at the forefront of lipid research. Activation of the ASMase/ceramide pathway is a shared response to an ever-growing list of receptor and non-receptor mediated forms of cellular stress including: death ligands (TNFα, TRAIL, Fas ligand), cytokines (IL-1, IFNγ), radiation, pathogenic infections, cytotoxic agents and others. The strategic role of ASMase in lipid metab. and cellular stress response has sparked interest in investigating the mol. mechanisms underlying ASMase activation. In this article, we review the translational role of the ASMase/ceramide pathway and recent advances on its mechanisms of regulation.
- 32Nikolova-Karakashian, M.; Morgan, E. T.; Alexander, C.; Liotta, D. C.; Merrill, A. H., Jr. J. Biol. Chem. 1997, 272, 18718Google ScholarThere is no corresponding record for this reference.
- 33Maceyka, M.; Milstien, S.; Spiegel, S. Prostaglandins Other Lipid Mediators 2005, 77, 15Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXnslylu7Y%253D&md5=80c153e9c3f0d95dac2ffa7c50dd0ebbSphingosine kinases, sphingosine-1-phosphate and sphingolipidomicsMaceyka, Michael; Milstien, Sheldon; Spiegel, SarahProstaglandins & Other Lipid Mediators (2005), 77 (1-4), 15-22CODEN: POLMFL; ISSN:1098-8823. (Elsevier B.V.)A review. It has become abundantly clear over the past decade that sphingolipids and their metabolites are key signaling mols. Ceramide, the backbone of all sphingolipids, predominantly inhibits cell growth and induces apoptosis, while its metabolite, sphingosine-1-phosphate promotes growth and survival. Given the interconvertibility of these two opposing signaling mols., it is essential that any study that examines the effects of one also look at the other. The newly available technol. of liq. chromatog.-tandem mass spectroscopy (LC-MS/MS) is increasingly being applied for this purpose, as it can quickly identify and measure many different sphingolipids simultaneously. An added benefit of LC-MS/MS is that it is several orders of magnitude more sensitive than enzymic methods or more traditional chromatog. techniques, allowing smaller sample sizes and increased throughput. Here, we briefly discuss the importance of LC-MS/MS for measuring sphingolipid metabolites and some future directions researchers may take given the increasingly accessibility to this technol.
- 34Sullards, M. C.; Merrill, A. H., Jr. Sci STKE 2001, 2001, l1Google ScholarThere is no corresponding record for this reference.
- 35Sullards, M. C.; Liu, Y.; Chen, Y.; Merrill, A. H., Jr. Biochim. Biophys. Acta 2011, DOI: 10.1016/j.bbalip.2011.06.027Google ScholarThere is no corresponding record for this reference.
- 36Karlsson, K. A. Chem. Phys Lipids 1970, 5, 6Google ScholarThere is no corresponding record for this reference.
- 37Karlsson, K. A. Lipids 1970, 5, 878Google ScholarThere is no corresponding record for this reference.
- 38Samuelsson, B.; Samuelsson, L. J. Lipid Res. 1969, 10, 47Google ScholarThere is no corresponding record for this reference.
- 39Shaner, R. L.; Allegood, J. C.; Park, H.; Wang, E.; Kelly, S.; Haynes, C. A.; Sullards, M. C.; Merrill, A. H., Jr. J. Lipid Res. 2009, 50, 1692Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXovV2lsr8%253D&md5=d5c56e8b3b4310bd75ce766d5dd47c50Quantitative analysis of sphingolipids for lipidomics using triple quadrupole and quadrupole linear ion trap mass spectrometersShaner, Rebecca L.; Allegood, Jeremy C.; Park, Hyejung; Wang, Elaine; Kelly, Samuel; Haynes, Christopher A.; Sullards, M. Cameron; Merrill, Alfred H., Jr.Journal of Lipid Research (2009), 50 (8), 1692-1707CODEN: JLPRAW; ISSN:0022-2275. (American Society for Biochemistry and Molecular Biology, Inc.)Sphingolipids are a highly diverse category of bioactive compds. This article describes methods that have been validated for the extn., liq. chromatog. (LC) sepn., identification and quantitation of sphingolipids by electrospray ionization, tandem mass spectrometry (ESI-MS/MS) using triple quadrupole (QQQ, API 3000) and quadrupole-linear-ion trap (API 4000 QTrap, operating in QQQ mode) mass spectrometers. Advantages of the QTrap included: greater sensitivity, similar ionization efficiencies for sphingolipids with ceramide vs. dihydroceramide backbones, and the ability to identify the ceramide backbone of sphingomyelins using a pseudo-MS3 protocol. Compds. that can be readily quantified using an internal std. cocktail developed by the LIPID MAPS Consortium are: sphingoid bases and sphingoid base 1-phosphates, more complex species such as ceramides, ceramide 1-phosphates, sphingomyelins, mono- and di-hexosylceramides, and these complex sphingolipids with dihydroceramide backbones. With minor modifications, glucosylceramides and galactosylceramides can be distinguished, and more complex species such as sulfatides can also be quantified, when the internal stds. are available. LC ESI-MS/MS can be utilized to quantify a large no. of structural and signaling sphingolipids using com. available internal stds. The application of these methods is illustrated with RAW264.7 cells, a mouse macrophage cell line. These methods should be useful for a wide range of focused (sphingo)lipidomic investigations.
- 40Quehenberger, O.; Armando, A. M.; Brown, A. H.; Milne, S. B.; Myers, D. S.; Merrill, A. H.; Bandyopadhyay, S.; Jones, K. N.; Kelly, S.; Shaner, R. L.; Sullards, C. M.; Wang, E.; Murphy, R. C.; Barkley, R. M.; Leiker, T. J.; Raetz, C. R.; Guan, Z.; Laird, G. M.; Six, D. A.; Russell, D. W.; McDonald, J. G.; Subramaniam, S.; Fahy, E.; Dennis, E. A. J. Lipid Res. 2010, 51, 3299Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVWgt7vN&md5=158d2770e092103e9aaa14171bd023a9Lipidomics reveals a remarkable diversity of lipids in human plasmaQuehenberger, Oswald; Armando, Aaron M.; Brown, Alex H.; Milne, Stephen B.; Myers, David S.; Merrill, Alfred H.; Bandyopadhyay, Sibali; Jones, Kristin N.; Kelly, Samuel; Shaner, Rebecca L.; Sullards, Cameron M.; Wang, Elaine; Murphy, Robert C.; Barkley, Robert M.; Leiker, Thomas J.; Raetz, Christian R. H.; Guan, Ziqiang; Laird, Gregory M.; Six, David A.; Russell, David W.; McDonald, Jeffrey G.; Subramaniam, Shankar; Fahy, Eoin; Dennis, Edward A.Journal of Lipid Research (2010), 51 (11), 3299-3305CODEN: JLPRAW; ISSN:0022-2275. (American Society for Biochemistry and Molecular Biology)The focus of the present study was to define the human plasma lipidome and to establish novel anal. methodologies to quantify the large spectrum of plasma lipids. Partial lipid anal. is now a regular part of every patient's blood test and physicians readily and regularly prescribe drugs that alter the levels of major plasma lipids such as cholesterol and triglycerides. Plasma contains many thousands of distinct lipid mol. species that fall into six main categories including fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, sterols, and prenols. The physiol. contributions of these diverse lipids and how their levels change in response to therapy remain largely unknown. As a first step toward answering these questions, we provide herein an in-depth lipidomics anal. of a pooled human plasma obtained from healthy individuals after overnight fasting and with a gender balance and an ethnic distribution that is representative of the US population. In total, we quant. assessed the levels of over 500 distinct mol. species distributed among the main lipid categories. As more information is obtained regarding the roles of individual lipids in health and disease, it seems likely that future blood tests will include an ever increasing no. of these lipid mols.
- 41Tanaka, K.; Yamada, M.; Tamiya-Koizumi, K.; Kannagi, R.; Aoyama, T.; Hara, A.; Kyogashima, M. Glycoconj. J. 2011, 28, 67Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXktVCmtrs%253D&md5=7c42d669fda409f2f7fb25326b9a6de3Systematic analyses of free ceramide species and ceramide species comprising neutral glycosphingolipids by MALDI-TOF MS with high-energy CIDTanaka, Kouji; Yamada, Masaki; Tamiya-Koizumi, Keiko; Kannagi, Reiji; Aoyama, Toshifumi; Hara, Atsushi; Kyogashima, MamoruGlycoconjugate Journal (2011), 28 (2), 67-87CODEN: GLJOEW; ISSN:0282-0080. (Springer)Free ceramides and glycosphingolipids (GSLs) are important components of the membrane microdomain and play significant roles in cell survival. Recent studies have revealed that both fatty acids and long-chain bases (LCBs) are more diverse than expected, in terms of i) alkyl chain length, ii) hydroxylation and iii) the presence or absence of double bonds. Electrospray ionization mass spectrometry and matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) have been well utilized to characterize sphingolipids with high throughput, but reports to date have not fully characterized various types of ceramide species such as hydroxyl fatty acids and/or trihydroxy-LCBs of both free ceramides and the constituent ceramides in neutral GSLs. We performed a systematic anal. of both ceramide species, including LCBs with nona-octadeca lengths using MALDI-TOF MS with high-energy collision-induced dissocn. (CID) at 20 keV. Using both protonated and sodiated ions, this technique enabled us to propose general rules to discriminate between isomeric and isobaric ceramide species, unrelated to the presence or absence of sugar chains. In addn., this high-energy CID generated 3,5A ions, indicating Hex1-4Hex linkage in the sugar chains. Using this method, we demonstrated distinct differences among ceramide species, including free ceramides, sphingomyelins, and neutral GSLs of glucosylceramides, galactosylceramides, lactosylceramides, globotriaosylceramides and Forssman glycolipids in the equine kidneys.
- 42Scherer, M.; Bottcher, A.; Schmitz, G.; Liebisch, G. Biochim. Biophys. Acta 2011, 1811, 68Google ScholarThere is no corresponding record for this reference.
- 43Fujino, Y.; Fujishima, T. J. Dairy Res. 1972, 39, 11Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE38Xht1Gmsbk%253D&md5=698e49827f0aeb512d1cd707546e5715Nature of ceramide in bovine milkFujino, Y.; Fujishima, T.Journal of Dairy Research (1972), 39 (1), 11-14CODEN: JDRSAN; ISSN:0022-0299.Milk was added to 4% Cl3CCO2H and the ppts. were centrifuged, lyophilized, and extd. with CHCl3-MeOH. The total lipid obtained was chromatographed on silicic acid and the eluted fraction was treated with 0.4N KOH in MeOH and rechromatographed. The lipid in the eluted fraction was recrystd. from ether to purify the ceramide. The Rf value of the single spot obtained from thin-layer chromatog. on silicic acid and the ir spectrum agreed with that of a std. prepn. Sixteen component fatty acids were found upon hydrolysis of the ceramide, among which C23:0 (38.1%), C24:0 (29.5%), C22:0 (17.9%), and C16:0 (7.2%) were predominant. Seven component long-chain bases were detected. The principal bases were C18-sphingosine (35.0%) and C16-sphingosine (31.6%).
- 44Byrdwell, W. C.; Perry, R. H. J. Chromatogr. A 2007, 1146, 164Google ScholarThere is no corresponding record for this reference.
- 45Sonnino, S.; Chigorno, V. Biochim. Biophys. Acta 2000, 1469, 63Google ScholarThere is no corresponding record for this reference.
- 46Keranen, A. Chem. Phys. Lipids 1976, 17, 14Google ScholarThere is no corresponding record for this reference.
- 47Kyogashima, M.; Tadano-Aritomi, K.; Aoyama, T.; Yusa, A.; Goto, Y.; Tamiya-Koizumi, K.; Ito, H.; Murate, T.; Kannagi, R.; Hara, A. J. Biochem. 2008, 144, 95Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXps1Ojurk%253D&md5=c1289ebbbb0a74387f1115bd6d08ae95Chemical and apoptotic properties of hydroxy-ceramides containing long-chain bases with unusual alkyl chain lengthsKyogashima, Mamoru; Tadano-Aritomi, Keiko; Aoyama, Toshifumi; Yusa, Akiko; Goto, Yoshiko; Tamiya-Koizumi, Keiko; Ito, Hiromi; Murate, Takashi; Kannagi, Reiji; Hara, AtsushiJournal of Biochemistry (2008), 144 (1), 95-106CODEN: JOBIAO; ISSN:0021-924X. (Japanese Biochemical Society)We analyzed four types of free ceramides (Cer 1, Cer 2, Cer 3 and Cer 4) from equine kidneys by electrospray ionization mass spectrometry. Cer 1 was composed of dihydroxy long-chain bases (dLCBs) of (4E)-sphingenine (d18:1), sphinganine and non-hydroxy fatty acids (NFAs); Cer 2 was composed of trihydroxy LCBs (tLCBs) of 4-hydroxysphinganine, t16:0, t18:0, t19:0 and t20:0, and NFAs; Cer 3 was composed of dLCBs, d16:1, d17:1, d18:1, d19:1 and d20:1, and hydroxy FAs (HFAs); and Cer 4 was composed of tLCBs, t16:0, t17:0, t18:0, t19:0 and t20:0, and HFAs. The results indicate all ceramide species contg. LCBs with non-octadeca lengths (NOD-LCBs) can be classified into hydroxy-ceramides since these species always consist of tLCBs, and/or HFAs. Furthermore, such species tend to contain FAs with longer acyl chains but contain neither palmitate (C16:0) nor its hydroxylated form (C16:0h). The apoptosis-inducing activities of these hydroxyl-ceramides towards tumor cell lines were compared with that of non-hydroxy-ceramides, dLCB-NFA (Cer 1). Monohydroxy-ceramides, tLCB-NFA (Cer 2) and dLCB-HFA (Cer 3), exhibited stronger activities, whereas dihydroxy-ceramides, tLCB-HFA (Cer 4), exhibited similar or weaker activity than dLCB-NFA (Cer 1), depending on cell lines.
- 48Kyogashima, M.; Tamiya-Koizumi, K.; Ehara, T.; Li, G.; Hu, R.; Hara, A.; Aoyama, T.; Kannagi, R. Glycobiology 2006, 16, 719Google ScholarThere is no corresponding record for this reference.
- 49Merrill, A. H., Jr.; Wang, E.; Wertz, P. W. Lipids 1986, 21, 529Google ScholarThere is no corresponding record for this reference.
- 50Fyrst, H.; Herr, D. R.; Harris, G. L.; Saba, J. D. J. Lipid Res. 2004, 45, 54Google ScholarThere is no corresponding record for this reference.
- 51Goni, F. M.; Alonso, A. Biochim. Biophys. Acta 2006, 1758, 1902– 21Google ScholarThere is no corresponding record for this reference.
- 52Ogawa-Goto, K.; Funamoto, N.; Abe, T.; Nagashima, K. J. Neurochem. 1990, 55, 1486Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXitFehsw%253D%253D&md5=e1624d0840c03478e55fc08e66341b27Different ceramide compositions of gangliosides between human motor and sensory nervesOgawa-Goto, K.; Funamoto, N.; Abe, T.; Nagashima, K.Journal of Neurochemistry (1990), 55 (5), 1486-93CODEN: JONRA9; ISSN:0022-3042.Ganglioside anal. of human motor and sensory nerves revealed that ceramide compns. of sensory nerve GD1a, GD1b, and GM1 apparently differed from those in the motor nerve. These gangliosides from sensory nerve contained a large amt. of long-chain fatty acids and C18-sphingosine as a major long-chain base. On the contrary, the motor nerve gangliosides contained C16-18 fatty acids and a large amt. of d20:1 besides C18-sphingosine. Furthermore, these gangliosides were enriched more in the axon fraction than in the myelin fraction. LM1, which was a major ganglioside in myelin from human peripheral nerve, was composed of similar ceramide compns. in the 2 nerves. Apparently, the characteristic ceramide species of nerve gangliosides may reflect in part properties of their own neurons.
- 53Sugiura, Y.; Shimma, S.; Konishi, Y.; Yamada, M. K.; Setou, M. PLoS One 2008, 3, e3232Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1cnhtFagug%253D%253D&md5=15fc8c9132ccaf0acf1c833c2576fb2dImaging mass spectrometry technology and application on ganglioside study; visualization of age-dependent accumulation of C20-ganglioside molecular species in the mouse hippocampusSugiura Yuki; Shimma Shuichi; Konishi Yoshiyuki; Yamada Maki K; Setou MitsutoshiPloS one (2008), 3 (9), e3232 ISSN:.Gangliosides are particularly abundant in the central nervous system (CNS) and thought to play important roles in memory formation, neuritogenesis, synaptic transmission, and other neural functions. Although several molecular species of gangliosides have been characterized and their individual functions elucidated, their differential distribution in the CNS are not well understood. In particular, whether the different molecular species show different distribution patterns in the brain remains unclear. We report the distinct and characteristic distributions of ganglioside molecular species, as revealed by imaging mass spectrometry (IMS). This technique can discriminate the molecular species, raised from both oligosaccharide and ceramide structure by determining the difference of the mass-to-charge ratio, and structural analysis by tandem mass spectrometry. Gangliosides in the CNS are characterized by the structure of the long-chain base (LCB) in the ceramide moiety. The LCB of the main ganglioside species has either 18 or 20 carbons (i.e., C18- or C20-sphingosine); we found that these 2 types of gangliosides are differentially distributed in the mouse brain. While the C18-species was widely distributed throughout the frontal brain, the C20-species selectively localized along the entorhinal-hippocampus projections, especially in the molecular layer (ML) of the dentate gyrus (DG). We revealed development- and aging-related accumulation of the C-20 species in the ML-DG. Thus it is possible to consider that this brain-region specific regulation of LCB chain length is particularly important for the distinct function in cells of CNS.
- 54Umesaki, Y.; Takamizawa, K.; Ohara, M. Biochim. Biophys. Acta 1989, 1001, 157Google ScholarThere is no corresponding record for this reference.
- 55Holleran, W. M.; Takagi, Y.; Uchida, Y. FEBS Lett. 2006, 580, 5456Google ScholarThere is no corresponding record for this reference.
- 56Stewart, M. E.; Downing, D. T. J. Invest. Dermatol. 1995, 105, 613Google ScholarThere is no corresponding record for this reference.
- 57Stewart, M. E.; Downing, D. T. J. Lipid Res. 1999, 40, 1434Google ScholarThere is no corresponding record for this reference.
- 58Chun, J.; Byun, H. S.; Bittman, R. J. Org. Chem. 2003, 68, 348Google ScholarThere is no corresponding record for this reference.
- 59Renkonen, O.; Hirvisalo, E. L. J. Lipid Res. 1969, 10, 687Google ScholarThere is no corresponding record for this reference.
- 60Panganamala, R. V.; Geer, J. C.; Cornwell, D. G. J. Lipid Res. 1969, 10, 445Google ScholarThere is no corresponding record for this reference.
- 61Touboul, D.; Roy, S.; Germain, D. P.; Baillet, A.; Brion, F.; Prognon, P.; Chaminade, P.; Laprevote, O. Anal. Bioanal. Chem. 2005, 382, 1209Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXlvVCitrs%253D&md5=617732a4d92a8dd3ae70e000ca9773e4Fast fingerprinting by MALDI-TOF mass spectrometry of urinary sediment glycosphingolipids in Fabry diseaseTouboul, David; Roy, Sandrine; Germain, Dominique P.; Baillet, Arlette; Brion, Francoise; Prognon, Patrice; Chaminade, Pierre; Laprevote, OlivierAnalytical and Bioanalytical Chemistry (2005), 382 (5), 1209-1216CODEN: ABCNBP; ISSN:1618-2642. (Springer)Fabry disease (FD) is an X-linked inborn error of glycosphingolipid (GSL) metab., caused by a deficiency of the lysosomal α-galactosidase A, which results in high levels in lysosomes and biol. fluids of globotriaosylceramide (Gb3) and digalactosylceramide (Ga2), also known as galabiosylceramide. The authors report here a detailed study of the mol. species of GSLs in urinary samples obtained from hemizygous and heterozygous patients by use of matrix-assisted laser desorption ionization and tandem mass spectrometry (MALDI-MS-MS). Twenty-two and fifteen mol. species were identified in the globotriaosylceramide and digalabiosylceramide series, resp. The major sphingoid base was sphingosine (d18:1), and dihydrosphingosine (C18:0) and sphingadienine (d18:2) were also present. The mol. profiles obtained by MALDI-TOF-MS enabled the authors to show significant differences between GSLs compn. for young, adult or atypic hemizygote and heterozygote patients. Thus, MALDI-TOF-MS and MS-MS proved a powerful tool for screening a population of patients with clin. signs suggestive of FD by direct and rapid GSL fingerprinting and identification, and for study of the biol. processes occurring in glycosphingolipid accumulation.
- 62Lynch, D. V.; Caffrey, M.; Hogan, J. L.; Steponkus, P. L. Biophys. J. 1992, 61, 1289Google ScholarThere is no corresponding record for this reference.
- 63Sullards, M. C.; Lynch, D. V.; Merrill, A. H., Jr.; Adams, J. J. Mass Spectrom. 2000, 35, 347Google ScholarThere is no corresponding record for this reference.
- 64Bartke, N.; Fischbeck, A.; Humpf, H. U. Mol. Nutr. Food Res. 2006, 50, 1201Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmsFensQ%253D%253D&md5=c3aea2174c8acd3248ba23f139c75d49Analysis of sphingolipids in potatoes (Solanum tuberosum L.) and sweet potatoes (Ipomoea batatas (L.) Lam.) by reversed phase high-performance liquid chromatography electrospray ionization tandem mass spectrometry (HPLC-ESI-MS/MS)Bartke, Nana; Fischbeck, Anne; Humpf, Hans-UlrichMolecular Nutrition & Food Research (2006), 50 (12), 1201-1211CODEN: MNFRCV; ISSN:1613-4125. (Wiley-VCH Verlag GmbH & Co. KGaA)Ceramides and glucocerebrosides of potatoes (Solanum tuberosum L.) and sweet potatoes (Ipomoea batatas (L.) Lam.) were analyzed using RP-HPLC-ESI-MS/MS. Ceramides and glucocerebrosides contg. the 3 different long-chain bases 4,8-sphingadienine (d18:2Δ4,Δ8), 4-hydroxy-8-sphingenine (t18:1Δ8), and 8-sphingenine (d18:1Δ8) acylated to satd. and unsatd. hydroxy- and nonhydroxy fatty acids with 16-26 C atoms were detected. For ceramides and glucocerebrosides 4,8-sphingadienine (d18:2Δ4,Δ8) was found as the major long-chain base, with lesser amts. of 4-hydroxy-8-sphingenine (t18:1Δ8) and 8-sphingenine (d18:1Δ8). 2-(α-)Hydroxypalmitic acid (C16:0h) was the major fatty acid, which was found to be acylated to the long-chain bases. For quantification of these compds., an RP-HPLC-ESI-MS/MS method with an "echo-peak"-technique simulating internal std. injection was developed. The analyzed samples of potatoes and sweet potatoes showed amts. of ∼0.1-8 μg/kg single ceramides and amts. up to 500 μg/kg glucocerebrosides, with C16:0h-glucosyl-4,8-sphingadienine as the major component.
- 65Sugawara, T.; Duan, J.; Aida, K.; Tsuduki, T.; Hirata, T.Lipids, 45, 451.Google ScholarThere is no corresponding record for this reference.
- 66Blaas, N.; Schuurmann, C.; Bartke, N.; Stahl, B.; Humpf, H. U. J. Agric. Food Chem. 2011, 59, 6018Google ScholarThere is no corresponding record for this reference.
- 67Zitomer, N. C.; Mitchell, T.; Voss, K. A.; Bondy, G. S.; Pruett, S. T.; Garnier-Amblard, E. C.; Liebeskind, L. S.; Park, H.; Wang, E.; Sullards, M. C.; Merrill, A. H., Jr.; Riley, R. T. J. Biol. Chem. 2009, 284, 4786Google ScholarThere is no corresponding record for this reference.
- 68Penno, A.; Reilly, M. M.; Houlden, H.; Laura, M.; Rentsch, K.; Niederkofler, V.; Stoeckli, E. T.; Nicholson, G.; Eichler, F.; Brown, R. H., Jr.; von Eckardstein, A.; Hornemann, T. J. Biol. Chem. 2010, 285, 11178Google ScholarThere is no corresponding record for this reference.
- 69Fyrst, H.; Saba, J. D. Biochim. Biophys. Acta 2008, 1781, 448Google ScholarThere is no corresponding record for this reference.
- 70Ohashi, Y.; Tanaka, T.; Akashi, S.; Morimoto, S.; Kishimoto, Y.; Nagai, Y. J. Lipid Res. 2000, 41, 1118Google ScholarThere is no corresponding record for this reference.
- 71Carter, G. T.; Rinehart, K. L. J. Am. Chem. Soc. 1978, 100, 7441Google ScholarThere is no corresponding record for this reference.
- 72Umemura, T.; Mori, K. Agric. Biol. Chem. 1987, 51, 217Google Scholar72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2sXmtlOjtLs%253D&md5=58c0761fb331b998894d453959ae31e7Synthesis of sphingosine relatives. Part V. Synthesis of both 2,3-erythro and 2,3-threo isomers of aplidiasphingosine, a marine terpenoidUmemura, Takeaki; Mori, KenjiAgricultural and Biological Chemistry (1987), 51 (1), 217-24CODEN: ABCHA6; ISSN:0002-1369.Stereoisomeric 2,3-erythro and 2,3-threo isomers of aplidiasphingosine (I) were synthesized in many steps from dithianeacetaldehyde (II). 13C NMR data confirmed their 2,3-threo and 13,14-erythro relative configurations.
- 73Molinski, T. F. Curr. Med. Chem.: Anti-Infect. Agents 2000, 3, 197Google ScholarThere is no corresponding record for this reference.
- 74Garrido, L.; Zubia, E.; Ortega, M. J.; Naranjo, S.; Salva, J. Tetrahedron 2001, 57, 4579Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXjsFWrsb4%253D&md5=b54dfb45cfccee723e0de408ad214f96Obscuraminols, new unsaturated amino alcohols from the tunicate Pseudodistoma obscurum: structure and absolute configurationGarrido, L.; Zubia, E.; Ortega, M. J.; Naranjo, S.; Salva, J.Tetrahedron (2001), 57 (21), 4579-4588CODEN: TETRAB; ISSN:0040-4020. (Elsevier Science Ltd.)The study of the ascidian Pseudodistoma obscurum from Tarifa Island (Cadiz, Spain) has led to the characterization of six new unsatd. 2-amino-3-ol compds., the obscuraminols A-F (e.g. I, obscuraminol A). Five of them, the obscuraminols B-F, were isolated as their corresponding diacetyl derivs. Their structures were established by spectroscopic anal., their relative configurations by NOEDS study of oxazolidinone derivs., and their abs. configurations by application of Mosher's method to N-acetyl derivs.
- 75Zhou, B.-N.; Mattern, M. P.; Johnson, R. K.; Kingston, D. G. I. Tetrahedron 2001, 57, 9549Google Scholar75https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXotlWqs7w%253D&md5=9dea5cca91ef9a34c4f0526e78ac9958Structure and stereochemistry of a novel bioactive sphingolipid from a Calyx sp.Zhou, Bing-Nan; Mattern, Michael P.; Johnson, Randall K.; Kingston, David G. I.Tetrahedron (2001), 57 (47), 9549-9554CODEN: TETRAB; ISSN:0040-4020. (Elsevier Science Ltd.)Bioassay-directed fractionation of a sponge of the genus Calyx using a yeast bioassay for DNA-damaging agents yielded the novel sphingolipid calyxoside (I) as the major bioactive constituent. The structure of I was assigned as 1,3,26-trihydroxy-2,27-diaminooctacosan-18-one-1-β-D-glucoside by 1H- and 13C NMR, DEPT, DQCOSY, HMQC, and HMBC spectra. The carbonyl group was located at C-18 by anal. of the EI-MS fragmentation of the amino deriv. of its aglycon pentaacetate. Its abs. configuration was detd. as 2S,3R,26S,27S by anal. of the 1H NMR and CD spectra of its aglycon pentabenzoate.
- 76Cuadros, R.; de Garcini, E. M.; Wandosell, F.; Faircloth, G.; Fernandez-Sousa, J. M.; Avila, J. Cancer Lett. 2000, 152, 23Google Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXit1Whu7o%253D&md5=582a0efea754a97caef2bebc5707cbf0The marine compound spisulosine, an inhibitor of cell proliferation, promotes the disassembly of actin stress fibersCuadros, R.; Montejo de Garcini, E.; Wandosell, F.; Faircloth, G.; Fernandez-Sousa, J. M.; Avila, J.Cancer Letters (Shannon, Ireland) (2000), 152 (1), 23-29CODEN: CALEDQ; ISSN:0304-3835. (Elsevier Science Ireland Ltd.)Spisulosine is a novel antiproliferative (antitumoral) compd. of marine origin. In this work the mol. target for this toxic agent has been analyzed. In the presence of spisulosine, cultured cells change their morphol., first acquiring a fusiform morphol., and later becoming rounded without focal adhesions. Anal. of the cytoskeleton of treated cells indicate the absence of actin stress fibers.
- 77Gelderblom, W. C.; Jaskiewicz, K.; Marasas, W. F.; Thiel, P. G.; Horak, R. M.; Vleggaar, R.; Kriek, N. P. Appl. Environ. Microbiol. 1988, 54, 1806Google Scholar77https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1cXlt1CitLo%253D&md5=5e4163f541ae6675d9a441bc1b95338eFumonisins-novel mycotoxins with cancer-promoting activity produced by Fusarium moniliformeGelderblom, W. C. A.; Jaskiewicz, K.; Marasas, W. F. O.; Thiel, P. G.; Horak, R. M.; Vleggaar, R.; Kriek, N. P. J.Applied and Environmental Microbiology (1988), 54 (7), 1806-11CODEN: AEMIDF; ISSN:0099-2240.Culture material of F. moniliforme isolate exhibits cancer-promoting activity in a short-term cancer initiation-promotion bioassay with diethylnitrosamine-initiated rats and induces γ-glutamyltranspeptidase-pos. (GGT+) foci as an endpoint after 4 wk of promotion. This bioassay was used as a monitoring system to isolate cancer-promoting compds. from cultures of F. moniliforme MRC 826. Culture material was successively extd. with Et acetate and MeOH-H2O (3:1). Most of the cancer-promoting activity was recovered in the MeOH-H2O ext. and remained in the aq. phase following partitioning of this ext. between MeOH-H2O (1:3) and CHCl3. The MeOH-H2O fraction was chromatographed on an Amberlite XAD-2 column, and the active fraction was eluted with MeOH. This fraction was chromatographed on a silica gel column with CHCl3-MeOH-MeCO2H (6:3:1) as eluent and further purified on a C18 reverse-phase column. Two pure compds. were isolated, and these have been chem. characterized and given the trivial names fumonisin B1 and B2. At least 2 g of the major compd. fumonisin B1 was purified from 1 kg of culture material. Fumonisin B1 in the diet (0.1%) significantly induced the formation of GGT+ foci in the livers of initiated as well as noninitiated rats. The cancer-promoting effect of fumonisin B1 in rats was assocd. with a toxic effect, as evidenced by a significant redn. in wt. gain during the 4-wk promoting treatment. The principal pathol. change in rats treated with fumonisin B1 was an insidious and progressive toxic hepatitis similar to that induced by toxic culture material of F. moniliforme MRC 826.
- 78Wang, E.; Norred, W. P.; Bacon, C. W.; Riley, R. T.; Merrill, A. H., Jr. J. Biol. Chem. 1991, 266, 14486Google Scholar78https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXlt12luro%253D&md5=cc1b3ed76f6c11b4be62b94aebfdbdfdInhibition of sphingolipid biosynthesis by fumonisins. Implications for diseases associated with Fusarium moniliformeWang, Elaine; Norred, William P.; Bacon, Charles W.; Riley, Ronald T.; Merrill, Alfred H., Jr.Journal of Biological Chemistry (1991), 266 (22), 14486-90CODEN: JBCHA3; ISSN:0021-9258.Incubation of rat hepatocytes with fumonisins inhibited incorporation of [14C]serine into the sphingosine moiety of cellular sphingolipids with an IC50 of 0.1 μM for fumonisin B1 (I). In contrast, I increased the amt. of the biosynthetic intermediate sphinganine, which suggests that fumonisins inhibit the conversion of [14C]sphinganine to N-acyl-[14C]sphinganines, a step that is thought to precede introduction of the 4,5-trans double bond of sphingosine (1986). In agreement with this mechanism, I inhibited the activity of sphingosine N-acyltransferase (ceramide synthase) in rat liver microsomes with 50% inhibition at approx. 0.1 μM and reduced the conversion of [3H]sphingosine to [3H]ceramide by intact hepatocytes. As far as the authors are aware, this is the 1st discovery of a naturally occurring inhibitor of this step of sphingolipid metab. These findings suggest that disruption of the de novo pathway of sphingolipid biosynthesis may be a crit. event in the diseases that have been assocd. with consumption of fumonisins.
- 79Marasas, W. F.; Riley, R. T.; Hendricks, K. A.; Stevens, V. L.; Sadler, T. W.; Gelineau-van Waes, J.; Missmer, S. A.; Cabrera, J.; Torres, O.; Gelderblom, W. C.; Allegood, J.; Martinez, C.; Maddox, J.; Miller, J. D.; Starr, L.; Sullards, M. C.; Roman, A. V.; Voss, K. A.; Wang, E.; Merrill, A. H., Jr. J. Nutr. 2004, 134, 711Google Scholar79https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjtFWmtro%253D&md5=75d5e627de87164c80a2e0620989a7a5Fumonisins disrupt sphingolipid metabolism, folate transport, and neural tube development in embryo culture and in vivo: a potential risk factor for human neural tube defects among populations consuming fumonisin-contaminated maizeMarasas, Walter F. O.; Riley, Ronald T.; Hendricks, Katherine A.; Stevens, Victoria L.; Sadler, Thomas W.; Gelineau-van Waes, Janee; Missmer, Stacey A.; Cabrera, Julio; Torres, Olga; Gelderblom, Wentzel C. A.; Allegood, Jeremy; Martinez, Carolina; Maddox, Joyce; Miller, J. David; Starr, Lois; Sullards, M. Cameron; Roman, Ana Victoria; Voss, Kenneth A.; Wang, Elaine; Merrill, Alfred H., Jr.Journal of Nutrition (2004), 134 (4), 711-716CODEN: JONUAI; ISSN:0022-3166. (American Society for Nutritional Sciences)A review. Fumonisins are a family of toxic and carcinogenic mycotoxins produced by Fusarium verticillioides (formerly Fusarium moniliforme), a common fungal contaminant of maize. Fumonisins inhibit ceramide synthase, causing accumulation of bioactive intermediates of sphingolipid metab. (sphinganine and other sphingoid bases and derivs.) as well as depletion of complex sphingolipids, which interferes with the function of some membrane proteins, including the folate-binding protein (human folate receptor α). Fumonisin causes neural tube and craniofacial defects in mouse embryos in culture. Many of these effects are prevented by supplemental folic acid. Recent studies in LMBc mice found that fumonisin exposure in utero increases the frequency of developmental defects and administration of folate or a complex sphingolipid is preventive. High incidences of neural tube defects (NTD) occur in some regions of the world where substantial consumption of fumonisins has been documented or plausibly suggested (Guatemala, South Africa, and China); furthermore, a recent study of NTD in border counties of Texas found a significant assocn. between NTD and consumption of tortillas during the first trimester. Hence, the authors propose that fumonisins are potential risk factors for NTD, craniofacial anomalies, and other birth defects arising from neural crest cells because of their apparent interference with folate utilization.
- 80Merrill, A. H., Jr.; Liotta, D. C.; Riley, R. T. Trends Cell Biol. 1996, 6, 218Google Scholar80https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xktlylsrg%253D&md5=00bcb7d977075a79853e0c10e17c513bFumonisins: Fungal toxins that shed light on sphingolipid functionMerrill, Alfred H. Jr.; Liotta, Dennis C.; Riley, Ronald T.Trends in Cell Biology (1996), 6 (6), 218-223CODEN: TCBIEK; ISSN:0962-8924. (Elsevier Trends Journals)A review and discussion with 52 refs. Fumonisins are sphinganine analogs produced by Fusarium moniliforme and related fungi. They inhibit ceramide synthase and block the biosynthesis of complex sphingolipids, promoting accumulation of sphinganine and sphinganine 1-phosphate. Disruption of sphingolipid metab. by fumonisin B1 alters cell-cell interactions, the behavior of cell-surface proteins, the activity of protein kinases, the metab. of other lipids, and cell growth and viability. This multitude of effects probably accounts for the toxicity and carcinogenicity of these mycotoxins. Naturally occurring inhibitors of sphingolipid metab. such as fumonisins are proving to be powerful tools for studying the diverse roles of sphingolipids in cell regulation and disease.
- 81Miyake, Y.; Kozutsumi, Y.; Nakamura, S.; Fujita, T.; Kawasaki, T. Biochem. Biophys. Res. Commun. 1995, 211, 396Google ScholarThere is no corresponding record for this reference.
- 82Hanada, K.; Nishijima, M.; Fujita, T.; Kobayashi, S. Biochem. Pharmacol. 2000, 59, 1211Google ScholarThere is no corresponding record for this reference.
- 83Ikushiro, H.; Hayashi, H.; Kagamiyama, H. Biochemistry 2004, 43, 1082Google ScholarThere is no corresponding record for this reference.
- 84Fujita, T.; Hirose, R.; Yoneta, M.; Sasaki, S.; Inoue, K.; Kiuchi, M.; Hirase, S.; Chiba, K.; Sakamoto, H.; Arita, M. J. Med. Chem. 1996, 39, 4451Google ScholarThere is no corresponding record for this reference.
- 85Thangada, S; Khanna, K. M.; Blaho, V. A.; Oo, M. L.; Im, D. S.; Guo, C; Lefrancois, L; Hla, T. J Exp. Med. 2010, 207, 1475Google ScholarThere is no corresponding record for this reference.
- 86Pyne, S.; Pyne, N. J. Trends Mol. Med. 2011, 17, 463Google ScholarThere is no corresponding record for this reference.
- 87Ter Braak, M.; Claas, R. F.; Hegen, B.; Labocha, S.; Ferreiros, N.; Pfeilschifter, J.; Huwiler, A.; van Echten-Deckert, G.; Meyer Zu Heringdorf, D. Biochem. Pharmacol. 2011, 81, 617Google ScholarThere is no corresponding record for this reference.
- 88van Echten-Deckert, G.; Zschoche, A.; Bar, T.; Schmidt, R. R.; Raths, A.; Heinemann, T.; Sandhoff, K. J. Biol. Chem. 1997, 272, 15825Google ScholarThere is no corresponding record for this reference.
- 89Schwartz, G. K.; Ward, D.; Saltz, L.; Casper, E. S.; Spiess, T.; Mullen, E.; Woodworth, J.; Venuti, R.; Zervos, P.; Storniolo, A. M.; Kelsen, D. P. Clin. Cancer Res. 1997, 3, 537Google Scholar89https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXislGitLc%253D&md5=dab6b7b4e13b06881a87f6b4a733f148A pilot clinical/pharmacological study of the protein kinase C-specific inhibitor safingol alone and in combination with doxorubicinSchwartz, Gary K.; Ward, David; Saltz, Leonard; Casper, Ephraim S.; Spiess, Tara; Mullen, Eillen; Woodworth, Jim; Venuti, Robert; Zervos, Peter; et al.Clinical Cancer Research (1997), 3 (4), 537-543CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)We performed a pilot clin. trial with safingol (L-threo-dihydrosphingosine), a protein kinase C-specific inhibitor that potentiates the effect of doxorubicin (DOX) in tumor-bearing animals. Safingol was initially administered as a 1-h infusion at escalating doses. Fourteen days later, patients received the same dose of safingol in combination with a fixed dose of DOX. The combination was repeated at 3-wk intervals. Safingol dose levels ranged from 15 to 120 mg/m2. The plasma levels achieved at the final dose level were comparable to those assocd. with potentiation of DOX in animals. The mean Cmax and area under the curve for safingol at the 120 mg/m2 dose level were 1040 ± 196 ng/mL and 1251 ± 317 mg × h/mL, resp. The mean plasma half-life for safingol was 3.97 ± 2.51 h, the mean estd. clearance was 3140 ± 765 mL/min, and the mean vol. of distribution was of 995 ± 421 L. Coadministration of a fixed dose of DOX did not significantly change the pharmacokinetics of safingol, nor did increasing doses of safingol significantly affect the pharmacokinetics of DOX. Minor responses were obsd. in three patients with pancreatic cancer and one patient with angiosarcoma of the scalp. This pilot Phase I study indicates that the protein kinase C inhibitor safingol can be given safely with 45 mg/m2 of DOX at a dose that is potentially pharmacol. active without dose-limiting toxicity.
- 90Coward, J.; Ambrosini, G.; Musi, E.; Truman, J. P.; Haimovitz-Friedman, A.; Allegood, J. C.; Wang, E.; Merrill, A. H., Jr.; Schwartz, G. K. Autophagy 2009, 5, 184Google ScholarThere is no corresponding record for this reference.
- 91Dickson, M. A.; Carvajal, R. D.; Merrill, A. H., Jr.; Gonen, M.; Cane, L. M.; Schwartz, G. K. Clin. Cancer Res. 2011, 17, 2484Google Scholar91https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXkslCgt7g%253D&md5=fb09e8c3a4903928616a53f942b7a6b9A Phase I Clinical Trial of Safingol in Combination with Cisplatin in Advanced Solid TumorsDickson, Mark A.; Carvajal, Richard D.; Merrill, Alfred H., Jr.; Gonen, Mithat; Cane, Lauren M.; Schwartz, Gary K.Clinical Cancer Research (2011), 17 (8), 2484-2492CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Sphingosine 1-phosphate (S1P) is an important mediator of cancer cell growth and proliferation. Prodn. of S1P is catalyzed by sphingosine kinase 1 (SphK). Safingol, (L-threo-dihydrosphingosine) is a putative inhibitor of SphK. We conducted a phase I trial of safingol (S) alone and in combination with cisplatin (C). A 3 + 3 dose escalation was used. For safety, S was given alone 1 wk before the combination. S + C were then administered every 3 wk. S was given over 60 to 120 min, depending on dose. Sixty minutes later, C was given over 60 min. The C dose of 75 mg/m2 was reduced in cohort 4 to 60 mg/m2 due to excessive fatigue. Forty-three patients were treated, 41 were evaluable for toxicity, and 37 for response. The max. tolerated dose (MTD) was S 840 mg/m2 over 120 min C 60 mg/m2, every 3 wk. Dose-limiting toxicity (DLT) attributed to cisplatin included fatigue and hyponatremia. DLT from S was hepatic enzyme elevation. S pharmacokinetic parameters were linear throughout the dose range with no significant interaction with C. Patients treated at or near the MTD achieved S levels of more than 20 μmol/L and maintained levels greater than and equal to 5 μmol/L for 4 h. The best response was stable disease in 6 patients for on av. 3.3 mo (range 1.8-7.2 m). One patient with adrenal cortical cancer had significant regression of liver and lung metastases and another had prolonged stable disease. S was assocd. with a dose-dependent redn. in S1P in plasma. Safingol, the first putative SphK inhibitor to enter clin. trials, can be safely administered in combination with cisplatin. Reversible dose-dependent hepatic toxicity was seen, as expected from preclin. data. Target inhibition was achieved with downregulation of S1P. The recommended phase II dose is S 840 mg/m2 and C 60 mg/m2, every 3 wk. Clin Cancer Res; 17(8); 2484-92.
- 92Symolon, H.; Bushnev, A.; Peng, Q.; Ramaraju, H.; Mays, S. G.; Allegood, J. C.; Pruett, S. T.; Sullards, M. C.; Dillehay, D. L.; Liotta, D. C.; Merrill, A. H., Jr. Mol. Cancer Ther. 2011, 10, 648Google Scholar92https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXkslSrtLo%253D&md5=ddfa116c8344f76b548d45a27081b0d1Enigmol: A Novel Sphingolipid Analogue with Anticancer Activity against Cancer Cell Lines and In vivo Models for Intestinal and Prostate CancerSymolon, Holly; Bushnev, Anatoliy; Peng, Qiong; Ramaraju, Harsha; Mays, Suzanne G.; Allegood, Jeremy C.; Pruett, Sarah T.; Sullards, M. Cameron; Dillehay, Dirck L.; Liotta, Dennis C.; Merrill, Alfred H., Jr.Molecular Cancer Therapeutics (2011), 10 (4), 648-657CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)Sphingoid bases are cytotoxic for many cancer cell lines and are thought to contribute to suppression of intestinal tumorigenesis in vivo by ingested sphingolipids. This study explored the behavior of a sphingoid base analog, (2S,3S,5S)-2-amino-3,5-dihydroxyoctadecane (Enigmol), that cannot be phosphorylated by sphingosine kinases and is slowly N-acylated and therefore is more persistent than natural sphingoid bases. Enigmol had potential anticancer activity in a National Cancer Institute (NCI-60) cell line screen and was confirmed to be more cytotoxic and persistent than naturally occurring sphingoid bases using HT29 cells, a colon cancer cell line. Although the mol. targets of sphingoid bases are not well delineated, Enigmol shared one of the mechanisms that has been found for naturally occurring sphingoid bases: normalization of the aberrant accumulation of β-catenin in the nucleus and cytoplasm of colon cancer cells due to defect(s) in the adenomatous polyposis coli (APC)/β-catenin regulatory system. Enigmol also had antitumor efficacy when administered orally to Min mice, a mouse model with a truncated APC gene product (C57Bl/6JMin/+ mice), decreasing the no. of intestinal tumors by half at 0.025% of the diet (wt./wt.), with no evidence of host toxicity until higher dosages. Enigmol was also tested against the prostate cancer cell lines DU145 and PC-3 in nude mouse xenografts and suppressed tumor growth in both. Thus, Enigmol represents a novel category of sphingoid base analog that is orally bioavailable and has the potential to be effective against multiple types of cancer. Mol Cancer Ther; 10(4); 648-57.
- 93Baird, R. D.; Kitzen, J.; Clarke, P. A.; Planting, A.; Reade, S.; Reid, A.; Welsh, L.; Lopez Lazaro, L.; de las Heras, B.; Judson, I. R.; Kaye, S. B.; Eskens, F.; Workman, P.; deBono, J. S.; Verweij, J. Mol. Cancer Ther. 2009, 8, 1430Google Scholar93https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXntVKgsLk%253D&md5=2739e092eca6c63b2a53afe31c220d40Phase I safety, pharmacokinetic, and pharmacogenomic trial of ES-285, a novel marine cytotoxic agent, administered to adult patients with advanced solid tumorsBaird, Richard D.; Kitzen, Jos; Clarke, Paul A.; Planting, Andre; Reade, Sarah; Reid, Alison; Welsh, Lyndsey; Lopez Lazaro, Luis; de las Heras, Begona; Judson, Ian R.; Kaye, Stan B.; Eskens, Ferry; Workman, Paul; de Bono, Johann S.; Verweij, JaapMolecular Cancer Therapeutics (2009), 8 (6), 1430-1437CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)A dose-escalation, phase I study evaluated the safety, pharmacokinetics, pharmacogenomics, and efficacy of ES-285, a novel agent isolated from a marine mollusc, in adult cancer patients. Patients received a 24-h i.v. infusion of ES-285 once every 3 wk until disease progression or unacceptable toxicity. The starting dose was 4 mg/m2. Dose escalation in cohorts of at least three patients proceeded according to the worst toxicity obsd. in the previous cohort. Twenty-eight patients were treated with 72 courses of ES-285 across eight dose levels. No dose-limiting toxicities were seen between 4 and 128 mg/m2. Two of four patients treated at 256 mg/m2 had dose-limiting reversible grade 3 transaminitis; one patient at 256 mg/m2 also had transient grade 3 central neurotoxicity. One of three patients subsequently treated at 200 mg/m2 died following drug-related central neurotoxicity. Other toxicities included phlebitis, nausea, fatigue, and fever. Pharmacokinetic studies indicated dose proportionality with high vol. of distribution (median Vss at 256 mg/m2 was 2,389 L; range, 1,615-4,051 L) and long elimination half life (median t1/2 at 256 mg/m2 was 28 h; range, 21-32 h). The three patients with dose-limiting toxicity had the highest drug exposure. Pharmacogenomic studies of paired surrogate tissue samples identified changes in gene expression following treatment that correlated with increasing dose. Disease stabilization for 6 to 18 wk was recorded in nine patients. Using this schedule, 128 mg/m2 was considered safe and feasible. At this dose, pharmacol. relevant concns. of the drug were safely achieved with pharmacogenomic studies indicating changes in the expression of genes of potential mechanistic relevance.
- 94Schoffski, P.; Dumez, H.; Ruijter, R.; Miguel-Lillo, B.; Soto-Matos, A.; Alfaro, V.; Giaccone, G. Cancer Chemother. Pharmacol. 2011, DOI: 10.1007/s00280-011-1612-1Google ScholarThere is no corresponding record for this reference.
- 95Faircloth, G.; Cuevas, C. Prog. Mol. Subcell. Biol. 2006, 43, 363Google Scholar95https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xos1ymt7s%253D&md5=3391732e41a2ec96ee072d0dcc28c2f6Kahalalide F and ES285: potent anticancer agents from marine molluscsFaircloth, G.; Cuevas, C.Progress in Molecular and Subcellular Biology (2006), 43 (Molluscs), 363-379CODEN: PMSBA4; ISSN:0079-6484. (Springer-Verlag)A review. The marine environment is proving to be a very rich source of unique compds. with significant activities against cancer of several types. Finding the sources of these new chem. entities has made it necessary for marine and medical scientists to find enterprising ways to collaborate in order to sample the great variety of intertidal, shallow and deep-water sea life. Recently these efforts resulted in a first generation of drugs from the sea undergoing clin. trials. These include PharmaMar compds.: Yondelis, Aplidin, kahalalide F, ES285 and Zalypsis. Two of these compds., kahalalide F and ES285, have been isolated from the Indopacific mollusc Elysia rufescens and the North Atlantic mollusc Spisula polynyma, resp.
- 96Menaldino, D. S.; Bushnev, A.; Sun, A.; Liotta, D. C.; Symolon, H.; Desai, K.; Dillehay, D. L.; Peng, Q.; Wang, E.; Allegood, J.; Trotman-Pruett, S.; Sullards, M. C.; Merrill, A. H., Jr. Pharmacol. Res. 2003, 47, 373Google Scholar96https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXisFWlsLY%253D&md5=a67a39be727ddcdbb2694b0f02389497Sphingoid bases and de novo ceramide synthesis: enzymes involved, pharmacology and mechanisms of actionMenaldino, David S.; Bushnev, Anatoliy; Sun, Aiming; Liotta, Dennis C.; Symolon, Holly; Desai, Kena; Dillehay, Dirck L.; Peng, Qiong; Wang, Elaine; Allegood, Jeremy; Trotman-Pruett, Sarah; Sullards, M. Cameron; Merrill, Alfred H.Pharmacological Research (2003), 47 (5), 373-381CODEN: PHMREP; ISSN:1043-6618. (Elsevier Science Ltd.)A review. The sphingoid base backbones of sphingolipids (sphingosines, sphinganines, 4-hydroxysphinganines and others) are highly bioactive species directly and-in most cases-as their metabolites, the N-acyl-sphingoid bases (ceramides) and sphingoid base 1-phosphates. The complexity of these compds. affords many opportunities to prep. synthetic analogs for studies of sphingolipid metab. and the functions of the sphingoid bases and metabolites. Described in this review are methods for the prepn. of libraries of sphingoid bases, including a series of 1-deoxy-analogs, as well as information about their metab. and biol. activities. Findings with these compds. have uncovered some of the complications of working with compds. that mimic a naturally occurring biomodulator-such as that they are sometimes metabolized by enzymes that handle the endogenous compds. and the products may have potent (and unexpected) biol. activities. Through studying such compds., there is now a greater understanding of the metab. and mechanism(s) of action of naturally occurring sphingoid bases as well as of these analogs.
- 97Zheng, 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.; Merrill, A. H., Jr. Biochim. Biophys. Acta 2006, 1758, 1864Google ScholarThere is no corresponding record for this reference.
- 98Fyrst, H.; Oskouian, B.; Bandhuvula, P.; Gong, Y.; Byun, H. S.; Bittman, R.; Lee, A. R.; Saba, J. D. Cancer Res. 2009, 69, 9457Google ScholarThere is no corresponding record for this reference.
- 99Liu, K.; Zhang, X.; Sumanasekera, C.; Lester, R. L.; Dickson, R. C. Biochem. Soc. Trans. 2005, 33, 1170Google Scholar99https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtFCqs7fJ&md5=4beee0312f73c001b9cae2e8540c1f12Signalling functions for sphingolipid long-chain bases in Saccharomyces cerevisiaeLiu, K.; Zhang, X.; Sumanasekera, C.; Lester, R. L.; Dickson, R. C.Biochemical Society Transactions (2005), 33 (5), 1170-1173CODEN: BCSTB5; ISSN:0300-5127. (Portland Press Ltd.)Over the past several years, studies of sphingolipid functions in the baker's yeast Saccharomyces cerevisiae have revealed that the sphingid LCBs (long-chain bases), dihydrosphingosine and PHS (phytosphingosine), are important signaling mols. or second messengers under heat stress and during non-stressed conditions. LCBs are now recognized as regulators of AGC-type protein kinase (where AGC stands for protein kinases A, G and C) Pkh1 and Pkh2, which are homologues of mammalian phosphoinositide-dependent protein kinase 1. LCBs were previously shown to activate Pkh1 and Pkh2, which then activate the downstream protein kinase Pkc1. The authors have demonstrated that PHS stimulates Pkh1 to activate addnl. downstream kinases including Ypk1, Ypk2 and Sch9. They have also found that PHS acts downstream of Pkh1 and partially activates Ypk1, Ypk2 and Sch9. These kinases control a wide range of cellular processes including growth, cell wall integrity, stress resistance, endocytosis and aging. As we learn more about the cellular processes controlled by Ypk1, Ypk2 and Sch9, we will have a far greater appreciation of LCBs as second messengers.
- 100Wertz, P. W. Acta Derm. Venereol. Suppl. (Stockh.) 2000, 208, 7Google Scholar100https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3M%252Fjt1Oqtg%253D%253D&md5=c84a398e9d1bce411393fba1050bf1aaLipids and barrier function of the skinWertz P WActa dermato-venereologica. Supplementum (2000), 208 (), 7-11 ISSN:0365-8341.The purpose of the present manuscript is to review the chemical and physical properties of epidermal lipids and to relate these properties to the formation and function of the permeability barrier of the skin. Lipids accumulate in small organelles known as lamellar granules as epidermal keratinocytes differentiate. This lipid is extruded into the intercellular spaces where it undergoes enzymatic processing to produce a lipid mixture consisting of ceramides, cholesterol and fatty acids. This intercellular lipid is uniquely organized into a multilamellar complex that fills most of the intercellular space of the stratum corneum. The barrier properties of the stratum corneum are related to the phase behavior of the intercellular lipids. It has been proposed that a structurally unusual acylglucosylceramide is thought to be involved in assembly of the lamellar granules, and a related acylceramide may have a major influence on the organization of the lamellae in the stratum corneum.
- 101Wartewig, S.; Neubert, R. H. Skin Pharmacol. Physiol. 2007, 20, 220Google Scholar101https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXpsVarsrw%253D&md5=d9127bd39d202219e1c2786c840b20a4Properties of ceramides and their impact on the stratum corneum structureWartewig, S.; Neubert, R. H. H.Skin Pharmacology and Physiology (2007), 20 (5), 220-229CODEN: SPPKE6; ISSN:1660-5527. (S. Karger AG)A review. The lipid matrix of the stratum corneum (SC) is the major diffusion-rate-limiting pathway by which most drugs intracellularly pass the SC. The major lipid classes extd. from the SC are ceramides, cholesterol, and free fatty acids. Ceramides that comprise 9 subclasses play a crucial role in maintaining the barrier function of the skin. A profound knowledge of the phys. properties of ceramides is essential for a deeper understanding of the impact of each ceramide species on the barrier function. Here, the authors summarize the thermotropic and/or lyotropic behavior of sphingosine-type ceramides (CER AS, CER NS) and phytosphingosine-type ceramides (CER AP, CER NP) revealed by DSC, x-ray diffraction, FTIR spectroscopy, and FT-Raman spectroscopy in past decades. Polymorphism is a characteristic feature of ceramides. At physiol. temps., all cryst. phases of ceramides exhibit lamellar structures with highly ordered hydrocarbon chains. The differential behavior of the head groups of ceramides may be an important determinant for the skin barrier function.
- 102Feingold, K. R. J. Lipid Res. 2007, 48, 2531Google Scholar102https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsValtL7L&md5=5f15317b5ea11938138dc83f18f18de6The role of epidermal lipids in cutaneous permeability barrier homeostasisFeingold, Kenneth R.Journal of Lipid Research (2007), 48 (12), 2531-2546CODEN: JLPRAW; ISSN:0022-2275. (American Society for Biochemistry and Molecular Biology, Inc.)A review. The permeability barrier is required for terrestrial life and is localized to the stratum corneum, where extracellular lipid membranes inhibit water movement. The lipids that constitute the extracellular matrix have a unique compn. and are 50% ceramides, 25% cholesterol, and 15% free fatty acids. Essential fatty acid deficiency results in abnormalities in stratum corneum structure function. The lipids are delivered to the extracellular space by the secretion of lamellar bodies, which contain phospholipids, glucosylceramides, sphingomyelin, cholesterol, and enzymes. In the extracellular space, the lamellar body lipids are metabolized by enzymes to the lipids that form the lamellar membranes. The lipids contained in the lamellar bodies are derived from both epidermal lipid synthesis and extracutaneous sources. Inhibition of cholesterol, fatty acid, ceramide, or glucosylceramide synthesis adversely affects lamellar body formation, thereby impairing barrier homeostasis. Studies have further shown that the elongation and desatn. of fatty acids is also required for barrier homeostasis. The mechanisms that mediate the uptake of extracutaneous lipids by the epidermis are unknown, but keratinocytes express LDL and scavenger receptor class B type 1, fatty acid transport proteins, and CD36. Topical application of physiol. lipids can improve permeability barrier homeostasis and has been useful in the treatment of cutaneous disorders.
- 103Walden, C. M.; Sandhoff, R.; Chuang, C. C.; Yildiz, Y.; Butters, T. D.; Dwek, R. A.; Platt, F. M.; van der Spoel, A. C. J. Biol. Chem. 2007, 282, 32655Google ScholarThere is no corresponding record for this reference.
- 104Rabionet, M.; van der Spoel, A. C.; Chuang, C. C.; von Tumpling-Radosta, B.; Litjens, M.; Bouwmeester, D.; Hellbusch, C. C.; Korner, C.; Wiegandt, H.; Gorgas, K.; Platt, F. M.; Grone, H. J.; Sandhoff, R. J. Biol. Chem. 2008, 283, 13357Google ScholarThere is no corresponding record for this reference.
- 105Van Overloop, H.; Denizot, Y.; Baes, M.; Van Veldhoven, P. P. Biol. Chem. 2007, 388, 315Google ScholarThere is no corresponding record for this reference.
- 106Lee, T. C.; Ou, M. C.; Shinozaki, K.; Malone, B.; Snyder, F. J. Biol. Chem. 1996, 271, 209Google ScholarThere is no corresponding record for this reference.
- 107Bielawska, A.; Linardic, C. M.; Hannun, Y. A. FEBS Lett. 1992, 307, 211Google ScholarThere is no corresponding record for this reference.
- 108Bielawska, A.; Crane, H. M.; Liotta, D.; Obeid, L. M.; Hannun, Y. A. J. Biol. Chem. 1993, 268, 26226Google ScholarThere is no corresponding record for this reference.
- 109Bielawska, A.; Greenberg, M. S.; Perry, D.; Jayadev, S.; Shayman, J. A.; McKay, C.; Hannun, Y. A. J. Biol. Chem. 1996, 271, 12646Google ScholarThere is no corresponding record for this reference.
- 110van Echten-Deckert, G.; Giannis, A.; Schwarz, A.; Futerman, A. H.; Sandhoff, K. J. Biol. Chem. 1998, 273, 1184Google ScholarThere is no corresponding record for this reference.
- 111Senkal, C. E.; Ponnusamy, S.; Rossi, M. J.; Sundararaj, K.; Szulc, Z.; Bielawski, J.; Bielawska, A.; Meyer, M.; Cobanoglu, B.; Koybasi, S.; Sinha, D.; Day, T. A.; Obeid, L. M.; Hannun, Y. A.; Ogretmen, B. J. Pharmacol. Exp. Ther. 2006, 317, 1188Google Scholar111https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xlt1eqtb8%253D&md5=28c9ad1ac69883317a02adf97f9f42e6Potent antitumor activity of a novel cationic pyridinium-ceramide alone or in combination with gemcitabine against human head and neck squamous cell carcinomas in vitro and in vivoSenkal, Can E.; Ponnusamy, Suriyan; Rossi, Michael J.; Sundararaj, Kamala; Szulc, Zdzislaw; Bielawski, Jacek; Bielawska, Alicja; Meyer, Mario; Cobanoglu, Bengu; Koybasi, Serap; Sinha, Debajyoti; Day, Terry A.; Obeid, Lina M.; Hannun, Yusuf A.; Ogretmen, BesimJournal of Pharmacology and Experimental Therapeutics (2006), 317 (3), 1188-1199CODEN: JPETAB; ISSN:0022-3565. (American Society for Pharmacology and Experimental Therapeutics)In this study, a cationic water-sol. ceramide analog L-threo-C6-pyridinium-ceramide-bromide (L-t-C6-Pyr-Cer), which exhibits high soly. and bioavailability, inhibited the growth of various human head and neck squamous cell carcinoma (HNSCC) cell lines at low IC50 concns., independent of their p53 status. Consistent with its design to target neg. charged intracellular compartments, L-t-C6-Pyr-Cer accumulated mainly in mitochondria-, and nuclei-enriched fractions upon treatment of human UM-SCC-22A cells [human squamous cell carcinoma (SCC) of the hypopharynx] at 1 to 6 h. In addn. to its growth-inhibitory function as a single agent, the supra-additive interaction of L-t-C6-Pyr-Cer with gemcitabine (GMZ), a chemotherapeutic agent used in HNSCC, was detd. using isobologram studies. Then, the effects of this ceramide, alone or in combination with GMZ, on the growth of UM-SCC-22A xenografts in SCID mice was assessed following the detn. of preclin. parameters, such as max. tolerated dose, clearance from the blood, and bioaccumulation. Results demonstrated that treatment with L-t-C6-Pyr-Cer in combination with GMZ significantly prevented the growth of HNSCC tumors in vivo. The therapeutic efficacy of L-t-C6-Pyr-Cer/GMZ combination against HNSCC tumors was approx. 2.5-fold better than that of the combination of 5-fluorouracil/cisplatin. In addn., liq. chromatog./mass spectroscopy anal. showed that the levels of L-t-C6-Pyr-Cer in HNSCC tumors were significantly higher than its levels in the liver and intestines; interestingly, the combination with GMZ increased the sustained accumulation of this ceramide by approx. 40%. Moreover, treatment with L-t-C6-Pyr-Cer/GMZ combination resulted in a significant inhibition of telomerase activity and decrease in telomere length in vivo, which are among downstream targets of ceramide.
- 112Struckhoff, A. P.; Bittman, R.; Burow, M. E.; Clejan, S.; Elliott, S.; Hammond, T.; Tang, Y.; Beckman, B. S. J. Pharmacol. Exp. Ther. 2004, 309, 523Google Scholar112https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjs1yhtbk%253D&md5=b0222816b08be3c1b59c29ac982fe74aNovel ceramide analogs as potential chemotherapeutic agents in breast cancerStruckhoff, Amanda P.; Bittman, Robert; Burow, Matthew E.; Clejan, Sanda; Elliott, Steven; Hammond, Timothy; Tang, Yan; Beckman, Barbara S.Journal of Pharmacology and Experimental Therapeutics (2004), 309 (2), 523-532CODEN: JPETAB; ISSN:0022-3565. (American Society for Pharmacology and Experimental Therapeutics)Recent evidence suggests a role for aberrant ceramide levels in the pathogenesis of cancer and chemoresistance and indicates that manipulation of tumor ceramide levels may be a useful strategy in the fight against breast cancer. This study demonstrates that alterations in the degree and position of unsatn. of bonds in the sphingoid backbone of D-erythro-N-octanoyl-sphingosine (Cer) affect the antiproliferative ability of ceramide analogs in breast cancer cells. The most potent analog of Cer we tested is (2S,3R)-(4E,6E)-2-octanoylamidoocta- decadiene-1,3-diol (4,6-diene-Cer), which contains an addnl. trans double bond at C(6)-C(7) of the sphingoid backbone. 4,6-Diene-Cer exhibited higher potency than Cer in tumor necrosis factor (TNF)-α-resistant (IC50 of 11.3 vs. 32.9 μM) and TNF-α-sensitive (IC50 of 13.7 vs. 37.7 μM) MCF-7 cells. 4,6-Diene-Cer was also more potent than Cer in inducing cell death in MDA-MB-231 and NCI/ADR-RES breast cancer cell lines (IC50 of 3.7 vs. 11.3 μM, and 24.1 vs. 86.9 μM, resp.). 4,6-Diene-Cer caused a prolonged elevation of intracellular ceramide levels in MCF-7 cells, which may contribute to its enhanced cytotoxicity. Furthermore, treatment of MCF-7 cells with Cer or 4,6-diene-Cer resulted in induction of apoptosis by 8 h via the mitochondrial pathway, as demonstrated by release of cytochrome c, loss of membrane asymmetry (measured by Annexin V staining), and a decrease in the mitochondrial membrane potential. Importantly, both Cer and 4,6-diene-Cer displayed selectivity toward transformed breast cells over nontransformed breast epithelial cells. These data suggest that these and other novel ceramide analogs represent potential therapeutic agents in breast cancer treatment.
- 113Bittman, R.; Li, Z.; Samadder, P.; Arthur, G. Cancer Lett. 2007, 251, 53Google Scholar113https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXksVyjsrk%253D&md5=f4261b165a682634751b7a1651342adfAnticancer activity of a ceramide analog containing a disulfide linkageBittman, Robert; Li, Zaiguo; Samadder, Pranati; Arthur, GilbertCancer Letters (Amsterdam, Netherlands) (2007), 251 (1), 53-58CODEN: CALEDQ; ISSN:0304-3835. (Elsevier B.V.)The effects of exogenous short-chain ceramide (1) on the arrest of growth of cancer cells in vitro and induction of apoptosis have been well documented. In the present study, an analog of 1 with a disulfide linkage, N-(4',5'-dithiaheptanoyl)-D-erythro-ceramide (2), was synthesized and found to be significantly more antiproliferative and cytotoxic than 1 in BT549, A549, and DU145 cancer cells. The activity was correlated with a redn. in cellular glutathione (GSH) level. Ceramide analogs with a tri- and tetra-sulfide moiety were also prepd., but they did not deplete cellular GSH levels and did not possess antiproliferative or cytotoxic properties.
- 114Stover, T. C.; Kim, Y. S.; Lowe, T. L.; Kester, M. Biomaterials 2008, 29, 359Google ScholarThere is no corresponding record for this reference.
- 115Liu, X.; Ryland, L.; Yang, J.; Liao, A.; Aliaga, C.; Watts, R.; Tan, S. F.; Kaiser, J.; Shanmugavelandy, S. S.; Rogers, A.; Loughran, K.; Petersen, B.; Yuen, J.; Meng, F.; Baab, K. T.; Jarbadan, N. R.; Broeg, K.; Zhang, R.; Liao, J.; Sayers, T. J.; Kester, M.; Loughran, T. P., Jr. Blood 2010, 116, 4192Google ScholarThere is no corresponding record for this reference.
- 116Tagaram, H. R.; Divittore, N. A.; Barth, B. M.; Kaiser, J. M.; Avella, D.; Kimchi, E. T.; Jiang, Y.; Isom, H. C.; Kester, M.; Staveley-O’Carroll, K. F. Gut 2011, 60, 695Google ScholarThere is no corresponding record for this reference.
- 117Megha; Sawatzki, P.; Kolter, T.; Bittman, R.; London, E. Biochim. Biophys. Acta 2007, 1768, 2205Google ScholarThere is no corresponding record for this reference.
- 118Goni, F. M.; Contreras, F. X.; Montes, L. R.; Sot, J.; Alonso, A. Biochem. Soc. Symp. 2005, 177Google Scholar118https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtF2mu7c%253D&md5=891b8bc575c6d8f2ac6ec6c60a94de55Biophysics (and sociology) of ceramidesGoni, Felix M.; Contreras, F.-Xabier; Montes, L.-Ruth; Sot, Jesus; Alonso, AliciaBiochemical Society Symposia (2005), 72 (Lipids, Rafts and Traffic), 177-188CODEN: BSSYAT; ISSN:0067-8694. (Portland Press Ltd.)A review. In the past decade, the long-neglected ceramides (N-acylsphingosines) have become one of the most attractive lipid mols. in mol. cell biol., because of their involvement in essential structures (stratum corneum) and processes (cell signalling). Most natural ceramides have a long (16-24 C atoms) N-acyl chain, but short N-acyl chain ceramides (two to six C atoms) also exist in Nature, apart from being extensively used in experimentation, because they can be dispersed easily in water. Long-chain ceramides are among the most hydrophobic mols. in Nature, they are totally insol. in water and they hardly mix with phospholipids in membranes, giving rise to ceramide-enriched domains. In situ enzymic generation, or external addn., of long-chain ceramides in membranes has at least three important effects: (i) the lipid monolayer tendency to adopt a neg. curvature, e.g. through a transition to an inverted hexagonal structure, is increased, (ii) bilayer permeability to aq. solutes is notoriously enhanced, and (iii) transbilayer (flip-flop) lipid motion is promoted. Short-chain ceramides mix much better with phospholipids, promote a pos. curvature in lipid monolayers, and their capacities to increase bilayer permeability or transbilayer motion are very low or non-existent.
- 119Contreras, F. X.; Basanez, G.; Alonso, A.; Herrmann, A.; Goni, F. M. Biophys. J. 2005, 88, 348Google ScholarThere is no corresponding record for this reference.
- 120Siskind, L. J.; Kolesnick, R. N.; Colombini, M. Mitochondrion 2006, 6, 118Google ScholarThere is no corresponding record for this reference.
- 121Hannun, Y. A.; Obeid, L. M. J. Biol. Chem. 2002, 277, 25847Google ScholarThere is no corresponding record for this reference.
- 122Hannun, Y. A.; Obeid, L. M. Nat. Rev. Mol. Cell Biol. 2008, 9, 139Google Scholar122https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXovFOntg%253D%253D&md5=0bee4e560d083ef4c43f7d0b591a0997Principles of bioactive lipid signalling: lessons from sphingolipidsHannun, Yusuf A.; Obeid, Lina M.Nature Reviews Molecular Cell Biology (2008), 9 (2), 139-150CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. It has become increasingly difficult to find an area of cell biol. in which lipids do not have important, if not key, roles as signaling and regulatory mols. The rapidly expanding field of bioactive lipids is exemplified by many sphingolipids, such as ceramide, sphingosine, sphingosine-1-phosphate (S1P), ceramide-1-phosphate, and lyso-sphingomyelin, which have roles in the regulation of cell growth, death, senescence, adhesion, migration, inflammation, angiogenesis, and intracellular trafficking. Deciphering the mechanisms of these varied cell functions necessitates an understanding of the complex pathways of sphingolipid metab. and the mechanisms that regulate lipid generation and lipid action.
- 123Gangoiti, P.; Camacho, L.; Arana, L.; Ouro, A.; Granado, M. H.; Brizuela, L.; Casas, J.; Fabrias, G.; Abad, J. L.; Delgado, A.; Gomez-Munoz, A. Prog. Lipid Res. 2010, 49, 316Google Scholar123https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtFWqsLvL&md5=b4b81f8cb184d6ec25acde919518948dControl of metabolism and signaling of simple bioactive sphingolipids: implications in diseaseGangoiti, Patricia; Camacho, Luz; Arana, Lide; Ouro, Alberto; Granado, Maria H.; Brizuela, Leyre; Casas, Josefina; Fabrias, Gemma; Abad, Jose Luis; Delgado, Antonio; Gomez-Munoz, AntonioProgress in Lipid Research (2010), 49 (4), 316-334CODEN: PLIRDW; ISSN:0163-7827. (Elsevier Ltd.)A review. Simple bioactive sphingolipids include ceramide, sphingosine and their phosphorylated forms sphingosine 1-phosphate and ceramide 1-phosphate. These mols. are crucial regulators of cell functions. In particular, they play important roles in the regulation of angiogenesis, apoptosis, cell proliferation, differentiation, migration, and inflammation. Decoding the mechanisms by which these cellular functions are regulated requires detailed understanding of the signaling pathways that are implicated in these processes. Most importantly, the development of inhibitors of the enzymes involved in their metab. may be crucial for establishing new therapeutic strategies for treatment of disease.
- 124Ogretmen, B.; Hannun, Y. A. Nat. Rev. Cancer 2004, 4, 604Google Scholar124https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmtVOhsb4%253D&md5=1147c2d9741a1da2a22ac28fa5a279e7Biologically active sphingolipids in cancer pathogenesis and treatmentOgretmen, Besim; Hannun, Yusuf A.Nature Reviews Cancer (2004), 4 (8), 604-616CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Biol. active sphingolipids have key roles in the regulation of several fundamental biol. processes that are integral to cancer pathogenesis. Recent significant progress in understanding biol. active sphingolipid synthesis, specifically within ceramide and sphingosine-1-phosphate (S1P)-mediated pathways, has identified crucial roles for these mols. both in cancer development and progression. Ceramide - a central mol. in sphingolipid metab. - in effect functions as a tumor-suppressor lipid, inducing antiproliferative and apoptotic responses in various cancer cells. Conversely, S1P induces responses that, on aggregate, render S1P a tumor-promoting lipid. These discoveries are paving the way for the advancement of anticancer therapies.
- 125Hannun, Y. A.; Obeid, L. M. J. Biol. Chem. 2011, 286, 27855Google ScholarThere is no corresponding record for this reference.
- 126Oskouian, B.; Saba, J. D. Adv. Exp. Med. Biol. 2010, 688, 185Google ScholarThere is no corresponding record for this reference.
- 127Fox, T. E.; Finnegan, C. M.; Blumenthal, R.; Kester, M. Cell. Mol. Life Sci. 2006, 63, 1017Google Scholar127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XlsFagsrk%253D&md5=f37c79d3070127554175fe8d95d55e3aThe clinical potential of sphingolipid-based therapeuticsFox, T. E.; Finnegan, C. M.; Blumenthal, R.; Kester, M.Cellular and Molecular Life Sciences (2006), 63 (9), 1017-1023CODEN: CMLSFI; ISSN:1420-682X. (Birkhaeuser Verlag)A review. The era of sphingolipid-based therapeutics is upon us. A large body of work has been accumulating that demonstrates the distinct biol. roles of sphingolipids in maintaining a homeostatic environment and in responding to environmental stimuli to regulate cellular processes. It is thus necessary to further investigate alterations in sphingolipid-metab. in pathol. conditions and, in turn, try to exploit altered sphingolipid-metabolizing enzymes and their metabolites as therapeutic targets. This review will examine how advances in the fields of drug delivery, drug discovery, synthetic chem., enzyme replacement therapy, immunobiol., infectious disease and nanotechnol. have delivered the potential and promise of utilizing and/or targeting sphingolipid metabolites as therapies for diverse diseases.
- 128Daido, S.; Kanzawa, T.; Yamamoto, A.; Takeuchi, H.; Kondo, Y.; Kondo, S. Cancer Res. 2004, 64, 4286Google ScholarThere is no corresponding record for this reference.
- 129Scarlatti, F.; Bauvy, C.; Ventruti, A.; Sala, G.; Cluzeaud, F.; Vandewalle, A.; Ghidoni, R.; Codogno, P. J. Biol. Chem. 2004, 279, 18384Google ScholarThere is no corresponding record for this reference.
- 130Zauner, S.; Ternes, P.; Warnecke, D. Adv. Exp. Med. Biol. 2010, 688, 249Google ScholarThere is no corresponding record for this reference.
- 131Jennemann, R.; Geyer, R.; Sandhoff, R.; Gschwind, R. M.; Levery, S. B.; Grone, H. J.; Wiegandt, H. Eur. J. Biochem. 2001, 268, 1190Google Scholar131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXhvFSks7c%253D&md5=4c227b24141e0060b8eaffd70b75d104Glycoinositolphosphosphingolipids (basidiolipids) of higher mushroomsJennemann, Richard; Geyer, Rudolf; Sandhoff, Roger; Gschwind, Ruth M.; Levery, Steven B.; Grone, Hermann-Josef; Wiegandt, HerbertEuropean Journal of Biochemistry (2001), 268 (5), 1190-1205CODEN: EJBCAI; ISSN:0014-2956. (Blackwell Science Ltd.)The basidiolipids of 6 mushroom spp., i.e. the basidiomycetes Amanita virosa (engl., death cup), Calvatia exipuliformis (engl., puffball), Cantharellus cibarius (engl., chanterelle), Leccinum scabrum (engl., red birch boletus), Lentinus edodes (jap., Shiitake), and Pleurotus ostreatus (engl., oystermushroom), were isolated, and their chem. structures investigated. All glycolipids are structurally related to those of the Agaricales (engl., field mushroom). They are glycoinositolphosphosphingolipids, their ceramide moiety consisting of t18:0-trihydroxysphinganine and an α-hydroxy long-chain fatty acid. In contrast to a previous study, the glycoside anomery of the hexose (mannose) connected to the inositol of all investigated basidiomycete glycolipids, including the basidiolipids of Agaricus bisporus, was detd. unequivocally to be alpha. Therefore, the root structure of all basidiolipids consists of α-D-Manp-2Ins1-[PO4]-Cer. In addn., for some mushroom spp., the occurrence of an inositol substitution position variant, α-Manp-4Ins1-[PO4]-Cer, is shown. The carbohydrate of chanterelle basidiolipids consists solely of mannose, i.e. Cc1, Manα-3 or -6Manα; Cc2, Manα-3(Manα-6)Manα-. All other spp. investigated show extension of the α-mannoside in the 6-position by β-galactoside, which, in some instances, is α-fucosylated in 2-position (Fucα-2)Galβ-6Manα-. Further sugar chain elongation at the β-galactoside may be in 3- and/or 6-position by α-galactoside, e.g. Ce4, Po2, Galα-3-(Galα-6)(Fucα-2)Galβ-6Manα-, whereas A. virosa, Av-3, has a more complex, highly α-fucosylated terminus, Galα-3 (Fucα-2)(Fucα-6)Galα-2(Galα-3)Galβ-6Manα-. L. edodes basidiolipids show further elongation by α-mannoside, e.g. Le3, Manα-2Manα-6Galα-3(Fucα-2)Galβ-6Manα-, C. exipuliformis glycolipid by α-glucoside, i.e. Ce3, Glcα-6Galβ-6Manα-. Basidiolipid Ls1 from L. scabrum, notably, has a 3-α-mannosylated α-fucose, i.e. Galα-6(Manα-3Fucα-2)Galα-6Galβ-6Manα-. In conclusion, basidiolipids, though identical in their ceramide constitution, display wide and systematic mushroom species dependent variabilities of their chem. structures.
- 132Levery, S. B.; Toledo, M. S.; Straus, A. H.; Takahashi, H. K. Rapid Commun. Mass Spectrom. 2001, 15, 2240Google Scholar132https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXptFOmtL0%253D&md5=eed4f2ee23bdd24cf9a559dc44011fd0Comparative analysis of glycosylinositol phosphorylceramides from fungi by electrospray tandem mass spectrometry with low-energy collision-induced dissociation of Li+ adduct ionsLevery, Steven B.; Toledo, Marcos S.; Straus, Anita H.; Takahashi, Helio K.Rapid Communications in Mass Spectrometry (2001), 15 (23), 2240-2258CODEN: RCMSEF; ISSN:0951-4198. (John Wiley & Sons Ltd.)Glycosylinositol phosphorylceramides (GIPCs) are a class of acidic glycosphingolipids (GSLs) expressed by fungi, plants, and certain parasitic organisms, but not found in cells or tissues of mammals or other higher animals. Recent characterizations of fungal GIPCs point to an emerging diversity which could rival that already known for mammalian GSLs, and which can be expected to present a multitude of challenges for the anal. chemist. Previously, the use of Li+ cationization, in conjunction with electrospray ionization mass spectrometry (ESI-MS) and low-energy collision-induced dissocn. tandem mass spectrometry (ESI-MS/CID-MS), was found to be particularly effective for detailed structural anal. of monohexosylceramides (cerebrosides) from a variety of sources, including fungi, esp. minor components present in mixts. at extremely low abundance. In applying Li+ cationization to characterization of GIPCs, a substantial increase in both sensitivity and fragmentation was obsd. on collision-induced dissocn. of [M + Li]+ vs. [M + Na]+ for the same components analyzed under similar conditions, similar to results obtained previously with cerebrosides. Mol. adduct fragmentation patterns were found to be systematic and characteristic for both the glycosylinositol and ceramide moieties with or without phosphate. Interestingly, significant differences were obsd. in fragmentation patterns when comparing GIPCs having Manα1→2 vs. Manα1→6Ins core linkages. In addn., it was useful to perform tandem product ion scans on primary fragments generated in the orifice region, equiv. to ESI-(CID-MS)2 mode. Finally, precursor ion scanning from appropriate glycosylinositol phosphate product ions yielded clean mol. ion profiles in the presence of obscuring impurity peaks. The methods were applied to detailed characterization of GIPC fractions of increasing structural complexity from a variety of fungi, including a non-pathogenic Basidiomycete (mushroom), Agaricus blazei, and pathogenic Euascomycete species such as Aspergillus fumigatus, Histoplasma capsulatum, and Sporothrix schenckii. The anal. confirmed a remarkable diversity of GIPC structures synthesized by the dimorphic S. schenckii, as well as differential expression of both glycosylinositol and ceramide structures in the mycelium and yeast forms of this mycopathogen. Mass spectrometry also established that the ceramides of some A. fumigatus GIPC fractions contain very little 2-hydroxylation of the long-chain fatty-N-acyl moiety, a feature that is not generally obsd. with fungal GIPCs.
- 133Levery, S. B. Methods Enzymol. 2005, 405, 300Google Scholar133https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmt1SntL4%253D&md5=5de0568070f8dbe4663fa8138712d8b4Glycosphingolipid structural analysis and glycosphingolipidomicsLevery, Steven B.Methods in Enzymology (2005), 405 (Mass Spectrometry: Modified Proteins and Glycoconjugates), 300-369CODEN: MENZAU; ISSN:0076-6879. (Elsevier)A review. Sphingosines, or sphingoids, are a family of naturally occurring long-chain hydrocarbon derivs. sharing a common 1,3-dihydroxy-2-amino-backbone motif. The majority of sphingolipids, as their derivs. are collectively known, can be found in cell membranes in the form of amphiphilic conjugates, each composed of a polar head group attached to an N-acylated sphingoid, or ceramide. Glycosphingolipids (GSLs), which are the glycosides of either ceramide or myo-inositol-(1-O)-phosphoryl-(O-1)-ceramide, are a structurally and functionally diverse sphingolipid subclass; GSLs are ubiquitously distributed among all eukaryotic species and are found in some bacteria. Since GSLs are secondary metabolites, direct and comprehensive anal. (metabolomics) must be considered an essential complement to genomic and proteomic approaches for establishing the structural repertoire within an organism and deducing its possible functional roles. The glycosphingolipidome clearly comprises an important and extensive subset of both the glycome and the lipidome, but the complexities of GSL structure, biosynthesis, and function form the outlines of a considerable anal. problem, esp. since their structural diversity confers by extension an enormous variability with respect to physicochem. properties. This chapter covers selected developments and applications of techniques in mass spectrometric (MS) that have contributed to GSL structural anal. and glycosphingolipidomics since 1990. Sections are included on basic characteristics of ionization and fragmentation of permethylated GSLs and of lithium-adducted nonderivatized GSLs under pos.-ion electrospray ionization mass spectrometry (ESI-MS) and collision-induced mass spectrometry (CID-MS) conditions; on the anal. of sulfatides, mainly using neg.-ion techniques; and on selected applications of ESI-MS and matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) to emerging GSL structural, functional, and anal. issues. The latter section includes a particular focus on evolving techniques for anal. of gangliosides, GSLs contg. sialic acid, as well as on characterizations of GSLs from selected nonmammalian eukaryotes, such as dipterans, nematodes, cestodes, and fungi. Addnl. sections focus on the issue of whether it is better to leave GSLs intact or remove the ceramide; on development and uses of thin-layer chromatog. (TLC) blotting and TLC-MS techniques; and on emerging issues of high-throughput anal., including the use of flow injection, liq. chromatog. mass spectrometry (LC-MS), and capillary electrophoresis mass spectrometry (CE-MS).
- 134Itonori, S.; Sugita, M. In Comprehansive Glycoscience. From Chemistry to Systems Biology; Kamerling, J. P., Ed.; Elsevier: Oxford, U.K., 2007; Vol. 3.Google ScholarThere is no corresponding record for this reference.
- 135Laine, R. A. Glycobiology 1994, 4, 759Google ScholarThere is no corresponding record for this reference.
- 136Levery, S. B.; Nudelman, E. D.; Salyan, M. E.; Hakomori, S. Biochemistry 1989, 28, 7772Google ScholarThere is no corresponding record for this reference.
- 137Fukuda, M.; Fukuda, M. N.; Papayannopoulou, T.; Hakomori, S. Proc. Natl. Acad. Sci. U.S.A. 1980, 77, 3474Google ScholarThere is no corresponding record for this reference.
- 138Wijesinghe, D. S.; Allegood, J. C.; Gentile, L. B.; Fox, T. E.; Kester, M.; Chalfant, C. E. J. Lipid Res. 2010, 51, 641Google ScholarThere is no corresponding record for this reference.
- 139Lamour, N. F.; Stahelin, R. V.; Wijesinghe, D. S.; Maceyka, M.; Wang, E.; Allegood, J. C.; Merrill, A. H., Jr.; Cho, W.; Chalfant, C. E. J. Lipid Res. 2007, 48, 1293Google ScholarThere is no corresponding record for this reference.
- 140Hinkovska-Galcheva, V.; Boxer, L. A.; Kindzelskii, A.; Hiraoka, M.; Abe, A.; Goparju, S.; Spiegel, S.; Petty, H. R.; Shayman, J. A. J. Biol. Chem. 2005, 280, 26612Google ScholarThere is no corresponding record for this reference.
- 141Gomez-Munoz, A. Biochim. Biophys. Acta 2006, 1758, 2049Google ScholarThere is no corresponding record for this reference.
- 142Gomez-Munoz, A.; Kong, J. Y.; Salh, B.; Steinbrecher, U. P. J. Lipid Res. 2004, 45, 99Google ScholarThere is no corresponding record for this reference.
- 143Gangoiti, P.; Arana, L.; Ouro, A.; Granado, M. H.; Trueba, M.; Gomez-Munoz, A. Cell Signal 2011, 23, 27Google Scholar143https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht12lu7zN&md5=b03a1c52f1fc541f5c9352caf0bd4afdActivation of mTOR and RhoA is a major mechanism by which ceramide 1-phosphate stimulates macrophage proliferationGangoiti, Patricia; Arana, Lide; Ouro, Alberto; Granado, Maria H.; Trueba, Miguel; Gomez-Munoz, AntonioCellular Signalling (2011), 23 (1), 27-34CODEN: CESIEY; ISSN:0898-6568. (Elsevier)This study tested the hypothesis that Ceramide 1-phosphate (C1P) stimulates macrophage proliferation through activation of the mammalian target of rapamycin (mTOR). We first reported that C1P is mitogenic for fibroblasts and macrophages, but the mechanisms whereby it stimulates cell proliferation are incompletely understood. Here we demonstrate that C1P causes phosphorylation of mTOR in primary (bone marrow-derived) macrophages. Activation of this kinase was tested my measuring the phosphorylation state of its downstream target p70S6K after treatment with C1P. These actions were dependent upon prior activation of phosphoinositide 3 kinase (PI3-K), as selective inhibition of this kinase blocked mTOR phosphorylation and activation. In addn., C1P caused phosphorylation of PRAS40, a component of the mTOR complex 1 (mTORC1) that is absent in mTORC2. Furthermore, inhibition of the small G protein Ras homolog enriched in brain (Rheb), which is also a specific component of mTORC1, with FTI277, completely blocked C1P-stimulated mTOR phosphorylation, DNA synthesis and macrophage growth. In addn., C1P caused phosphorylation of another Ras homolog gene family member, RhoA, which is also involved in cell proliferation. Interestingly, inhibition of the RhoA downstream effector RhoA-assocd. kinase (ROCK) also blocked C1P-stimulated mTOR and cell proliferation. It can be concluded that mTORC1, and RhoA/ROCK are essential components of the mechanism whereby C1P stimulates macrophage proliferation.
- 144Lamour, N. F.; Subramanian, P.; Wijesinghe, D. S.; Stahelin, R. V.; Bonventre, J. V.; Chalfant, C. E. J. Biol. Chem. 2009, 284, 26897Google ScholarThere is no corresponding record for this reference.
- 145Pettus, B. J.; Chalfant, C. E.; Hannun, Y. A. Curr. Mol. Med. 2004, 4, 405Google Scholar145https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksFGjt70%253D&md5=06cce3c165c1fdd1526c39dc0b6919beSphingolipids in inflammation: Roles and implicationsPettus, B. J.; Chalfant, C. E.; Hannun, Y. A.Current Molecular Medicine (2004), 4 (4), 405-418CODEN: CMMUBP; ISSN:1566-5240. (Bentham Science Publishers Ltd.)A review. Sphingolipids, historically described as potential reservoirs for bioactive lipids, presently define a new family of cellular mediators, joining the well-established glycerolipid-derived mediators of signal transduction such as diacylglycerol, phosphatidylinositides, and eicosanoids. Sphingolipid metab. is clearly involved in the regulation of cell growth, differentiation, and programmed cell death. Indeed, a majority of the greater than four thousand studies conducted on sphingolipids during the past five years were investigations of the role of sphingolipids as cellular bioregulators. Studies spanning more than a decade have shown multiple interactions and intersections of the sphingolipid-mediated pathways and the eicosanoid pathway. This review will discuss the emerging mechanisms by which sphingolipids induce inflammatory responses via the eicosanoid pathway in addn. to linking previous literature on sphingolipids and inflammation with newer findings of distinct roles for sphingosine-1-phosphate in regulating cyclooxygenase-2 and ceramide-1-phosphate in the regulation of cytosolic phospholipase A2α. Finally, the relationship between bioactive sphingolipids and inflammation is discussed.
- 146Gubern, A.; Barcelo-Torns, M.; Barneda, D.; Lopez, J. M.; Masgrau, R.; Picatoste, F.; Chalfant, C. E.; Balsinde, J.; Balboa, M. A.; Claro, E. J. Biol. Chem. 2009, 284, 32359Google ScholarThere is no corresponding record for this reference.
- 147Barenholz, Y.; Thompson, T. E. Chem. Phys Lipids 1999, 102, 29Google ScholarThere is no corresponding record for this reference.
- 148Barenholz, Y. Subcell Biochem 2004, 37, 167Google Scholar148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhs1ahug%253D%253D&md5=de98603be77740d97a014a623853cd34Sphingomyelin and cholesterol: from membrane biophysics and rafts to potential medical applicationsBarenholz, YechezkelSubcellular Biochemistry (2004), 37 (Membrane Dynamics and Domains), 167-215CODEN: SBCBAG; ISSN:0306-0225. (Kluwer Academic/Plenum Publishers)A review. The preferential sphingomyelin-cholesterol interaction which results from the structure and the mol. properties of these two lipids seems to be the physicochem. basis for the formation and maintenance of cholesterol/sphingolipid-enriched nano- and micro-domains (referred to as membrane "rafts") in the plane of plasma and other organelle (i.e., Golgi) membranes. This claim is supported by much exptl. evidence and also by theor. considerations. However, although there is a large vol. of information about these rafts regarding their lipid and protein compn., their size, and their dynamics, there is still much to be clarified on these issues, as well as on how rafts are formed and maintained. It is well accepted now that the lipid phase of the rafts is the liq. ordered (LO) phase. However, other (non-raft) parts of the membrane may also be in the LO phase. There are indications that the raft LO phase domains are more tightly packed than the non-raft LO phase, possibly due to intermol. hydrogen bonding involving sphingolipid and cholesterol. This also explains why the former are detergent-resistant membranes (DRM), while the non-raft LO phase domains are detergent-sol. (sensitive) membranes (DSM). Recent findings suggest that protein-protein interactions such as crosslinking can be controlled by protein distribution between raft and non-raft domains, and, as well, these interactions affect raft size distribution. The cholesterol/sphingomyelin-enriched rafts seem to be involved in many biol. processes, mediated by various receptors, as exemplified by various lipidated glycosylphosphatidylinositol (GPI)- and acyl chain-anchored proteins that reside in the rafts. The rafts serve as signaling platforms in the cell. Various pathogens (viruses and toxins) utilize the raft domains on the host cell membrane as a port of entry, site of assembly (viruses), and port of exit (viral budding). Existence and maintenance of cholesterol-sphingomyelin rafts are dependent on the level of membrane cholesterol and sphingomyelin. This explains why redn. of cholesterol level - either through reverse cholesterol transport, using cholesterol acceptors such as β-cyclodextrin, or through cholesterol biosynthesis inhibition using statins interferes with many processes which involve rafts and can be applied to treating raft-related infections and diseases. Detailed elucidation of raft structure and function will improve understanding of biol. membrane compn.-structure-function relationships and also may serve as a new avenue for the development of novel treatments for major diseases, including viral infections, neurodegenerative diseases (Alzheimer's), atherosclerosis, and tumors.
- 149Koivusalo, M.; Jansen, M.; Somerharju, P.; Ikonen, E. Mol. Biol. Cell 2007, 18, 5113Google Scholar149https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVeis7zE&md5=807523a4f27c18deb1d7fb580b905290Endocytic trafficking of sphingomyelin depends on its acyl chain lengthKoivusalo, Mirkka; Jansen, Maurice; Somerharju, Pentti; Ikonen, ElinaMolecular Biology of the Cell (2007), 18 (12), 5113-5123CODEN: MBCEEV; ISSN:1059-1524. (American Society for Cell Biology)To study the principles of endocytic lipid trafficking, we introduced pyrene sphingomyelins (PyrSMs) with varying acyl chain lengths and domain partitioning properties into human fibroblasts or HeLa cells. We found that a long-chain, ordered-domain preferring PyrSM was targeted Hrs and Tsg101 dependently to late endosomal compartments and recycled to the plasma membrane in an NPC1- and cholesterol-dependent manner. A short-chain, disordered domain preferring PyrSM recycled more effectively, by using Hrs-, Tsg101- and NPC1-independent routing that was insensitive to cholesterol loading. Similar chain length-dependent recycling was obsd. for unlabeled sphingomyelins (SMs). The findings (1) establish acyl chain length as an important determinant in the endocytic trafficking of SMs, (2) implicate ESCRT complex proteins and NPC1 in the endocytic recycling of ordered domain lipids to the plasma membrane, and (3) introduce long-chain PyrSM as the first fluorescent lipid tracing this pathway.
- 150Shogomori, H.; Kobayashi, T. Biochim. Biophys. Acta 2008, 1780, 612Google ScholarThere is no corresponding record for this reference.
- 151Kiyokawa, E.; Baba, T.; Otsuka, N.; Makino, A.; Ohno, S.; Kobayashi, T. J. Biol. Chem. 2005, 280, 24072Google ScholarThere is no corresponding record for this reference.
- 152Muehlenberg, B. A.; Sribney, M.; Duffe, M. K. Can. J. Biochem. 1972, 50, 166Google Scholar152https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE38XhtV2msbc%253D&md5=8d56ec0ff956c7e0df623c7537adf709Occurrence and biosynthesis of ceramide phosphorylethanolamine in chicken and rat liverMuehlenberg, Bernd A.; Sribney, Michael; Duffe, Marilyn K.Canadian Journal of Biochemistry (1972), 50 (2), 166-73CODEN: CJBIAE; ISSN:0008-4018.Ceramide phosphorylethanolamine occurs in chicken and rat liver, and an enzyme (CDP-ethanolamine:ceramide ethanol-aminephosphotransferase) (I) has been found in a no. of tissues which catalyzes the biosynthesis of this lipid. The enzyme catalyzes the transfer of the phosphorylethanolamine moiety of CDP-ethanolamine to the free primary hydroxyl group of a ceramide (N-acylsphingosine). The chicken liver enzyme requires 0.010M Mn2+ for optimal activity and has a pH optimum of 7.7. The Km for the substrate N-octanoyl-threo-sphingosine was 2.5 × 10-4 M. A study of the effect of increasing CDP-ethanolamine concn. on the reaction rate indicates from sigmoid kinetics that the coenzyme modulates and possibly regulates I activity. The enutilize the unnatural threo isomer of N-acylsphingosines (threoceramides) as acceptors for the phosphorylethanolamine moiety of CDP-ethanolamine.
- 153Ternes, P.; Brouwers, J. F.; van den Dikkenberg, J.; Holthuis, J. C. J. Lipid Res. 2009, 50, 2270Google ScholarThere is no corresponding record for this reference.
- 154Vacaru, A. M.; Tafesse, F. G.; Ternes, P.; Kondylis, V.; Hermansson, M.; Brouwers, J. F.; Somerharju, P.; Rabouille, C.; Holthuis, J. C. J. Cell Biol. 2009, 185, 1013Google ScholarThere is no corresponding record for this reference.
- 155Masood, M. A.; Yuan, C.; Acharya, J. K.; Veenstra, T. D.; Blonder J. Anal. Biochem. 2010, 400, 259Google Scholar155https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjs1Ohurs%253D&md5=4989f3c11b4128b6e435ded27f80b705Quantitation of ceramide phosphorylethanolamines containing saturated and unsaturated sphingoid base coresMasood, M. Athar; Yuan, Changqing; Acharya, Jairaj K.; Veenstra, Timothy D.; Blonder, JosipAnalytical Biochemistry (2010), 400 (2), 259-269CODEN: ANBCA2; ISSN:0003-2697. (Elsevier B.V.)Sphingomyelin (SM) and ceramide-phosphoethanolamines (cer-PEs) are related lipids present in mammals and insects, resp. Owing to the crit. roles that cer-PEs play in eukaryotic cellular function, there is a need to develop methods that provide accurate quantitation of these compds. Results obtained in this study demonstrate that Drosophila contains cer-PEs with unsatd. sphingoid base cores as well as low levels of cer-PEs that possess satd. sphingoid base cores. Specifically, the method developed in this study enabled the quantitation of picogram amts. of cer-PE contg. both unsatd. d14:1Δ4 and d16:1Δ4 and satd. d14:0 sphingoid base cores. Using this method, cer-PE compds. with both satd. and unsatd. sphingoid base cores were initially identified by neutral loss scanning, followed by quantitation using selected reaction monitoring (SRM) scans. The SRM scans measured a product ion originating from the sphingoid base backbone, rather than from the head group, increasing the specificity and sensitivity of the quantitation measurement.
- 156Lester, R. L.; Dickson, R. C. Adv Lipid Res 1993, 26, 253Google ScholarThere is no corresponding record for this reference.
- 157Dickson, R. C.; Lester, R. L. Biochim. Biophys. Acta 1999, 1426, 347Google ScholarThere is no corresponding record for this reference.
- 158Warnecke, D.; Heinz, E. Cell. Mol. Life Sci. 2003, 60, 919Google Scholar158https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXlt1aitLo%253D&md5=9d43d2542d0c1b0f3a9f5b09e7273cd8Recently discovered functions of glucosylceramides in plants and fungiWarnecke, D.; Heinz, E.Cellular and Molecular Life Sciences (2003), 60 (5), 919-941CODEN: CMLSFI; ISSN:1420-682X. (Birkhaeuser Verlag)A review. Glycosphingolipids are ubiquitous membrane lipids of eukaryotic organisms and a few bacteria. Whereas inositol-contg. glycosphingolipids are restricted to plants and fungi, galactosylceramide occurs only in fungi and animals. In contrast, glucosylceramide is the unique glycosphingolipid which plants, fungi, and animals have in common. However, there are specific differences in the structure of the ceramide backbone of glucosylceramides from these organisms. A comparison of the structural features and the biosynthesis of glucosylceramides from plants, fungi and animals has contributed to an understanding of their functions, which so far have been analyzed mainly in animals. The availability of nearly all genes involved in the biosynthesis of glucosylceramides has enabled the specific manipulation of glycosphingolipid metab. by techniques of forward and reverse genetics. The application of this approach to unicellular organisms such as yeasts, multicellular filamentous fungi, as well as to complex organisms like plants has revealed common and different glucosylceramide functions in these organisms. These glycolipids play a role both in intracellular processes and in cell-to-cell interactions. These interactions may occur between cells of a multicellular organism or between cells of different species, as in host-pathogen interactions.
- 159Sillence, D. J.; Puri, V.; Marks, D. L.; Butters, T. D.; Dwek, R. A.; Pagano, R. E.; Platt, F. M. J. Lipid Res. 2002, 43, 1837Google ScholarThere is no corresponding record for this reference.
- 160van Meer, G.; Wolthoorn, J.; Degroote, S. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2003, 358, 869Google Scholar160https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXlsFeru7k%253D&md5=946f2bf7d891dc011927bfa8df25b498The fate and function of glycosphingolipid glucosylceramidevan Meer, Gerrit; Wolthoorn, Jasja; Degroote, SophiePhilosophical Transactions of the Royal Society of London, Series B: Biological Sciences (2003), 358 (1433), 869-873CODEN: PTRBAE; ISSN:0962-8436. (Royal Society)A review. In higher eukaryotes, glucosylceramide is the simplest member and precursor of a fascinating class of membrane lipids, the glycosphingolipids. These lipids display an astounding variation in their carbohydrate head groups, suggesting that glycosphingolipids serve specialized functions in recognition processes. It is now realized that they are organized in signaling domains on the cell surface. They are of vital importance as, in their absence, embryonal development is inhibited at an early stage. Remarkably, individual cells can live without glycolipids, perhaps because their survival does not depend on glycosphingolipid-mediated signaling mechanisms. Still, these cells suffer from defects in intracellular membrane transport. Various membrane proteins do not reach their intracellular destination, and, indeed, some intracellular organelles do not properly differentiate to their mature stage. The fact that glycosphingolipids are required for cellular differentiation suggests that there are human diseases resulting from defects in glycosphingolipid synthesis. In addn., the same cellular differentiation processes may be affected by defects in the degrdn. of glycosphingolipids. At the cellular level, the pathol. of glycosphingolipid storage diseases is not completely understood. Cell biol. studies on the intracellular fate and function of glycosphingolipids may open new ways to understand and defeat not only lipid storage diseases, but perhaps other diseases that have not been connected to glycosphingolipids so far.
- 161Messner, M. C.; Cabot, M. C. Adv. Exp. Med. Biol. 2010, 688, 156Google ScholarThere is no corresponding record for this reference.
- 162Lavie, Y.; Cao, H.; Bursten, S. L.; Giuliano, A. E.; Cabot, M. C. J. Biol. Chem. 1996, 271, 19530Google ScholarThere is no corresponding record for this reference.
- 163Liu, Y. Y.; Patwardhan, G. A.; Xie, P.; Gu, X.; Giuliano, A. E.; Cabot, M. C. Int. J. Oncol. 2011, 39, 425Google Scholar163https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpvVegsb0%253D&md5=5f74e33aaf3d17296552f1d6af09f117Glucosylceramide synthase, a factor in modulating drug resistance, is overexpressed in metastatic breast carcinomaLiu, Yong-Yu; Patwardhan, Gauri A.; Xie, Ping; Gu, Xin; Giuliano, Armando E.; Cabot, Myles C.International Journal of Oncology (2011), 39 (2), 425-431CODEN: IJONES; ISSN:1019-6439. (International Journal of Oncology)Drug resistance causes treatment failure in approx. 50% of breast cancer patients with chemotherapy. Overexpression of glucosylceramide synthase (GCS) confers drug resistance in cancer cells, and suppression of GCS sensitizes cancers to chemotherapy in preclin. studies. Thus, GCS becomes a potential target to reverse drug resistance; however, little is known about GCS expression levels in normal tissues and whether GCS overexpression is assocd. with metastatic cancers. Herewith, we report our studies in GCS expression levels and breast cancer from patients. GCS levels were analyzed using cancer profiling arrays, breast cancer histo-arrays and quant. RT-PCR in tumor tissues. We found that breast (18 exp. index) and other hormone-dependent organs (testis, cervix, ovary, prostate) displayed the lowest levels of GCS mRNA, whereas liver (52 exp. index) and other organs (kidney, bladder, stomach) displayed the highest levels of GCS. GCS mRNA levels were significantly elevated in tumors of breast, cervix, rectum and small intestine, as compared to each paired normal tissue. In mammary tissue, GCS overexpression was detected in breast cancers with metastasis, but not in benign fibroadenoma or primary tumors. GCS overexpression was coincident with HER2 expression (γ2 = 0.84) in ER-neg. breast adenocarcinoma. In tumor specimens, GCS mRNA was elevated by 4-fold and significantly assocd. with stage III (5/7), lymph node-pos. (7/8) and estrogen receptor-pos. breast cancers (7/9). GCS expression was significantly and selectively elevated in breast cancer, in particular in metastatic disease. GCS overexpression was highly assocd. with ER-pos. and HER2-pos. breast cancer with metastasis. Although a small study, these data suggest that GCS may be a prognostic indicator and potential target for the treatment of chemotherapy-refractory breast cancer.
- 164Lalazar, G.; Ben Ya’acov, A.; Livovsky, D. M.; El Haj, M.; Pappo, O.; Preston, S.; Zolotarov, L.; Ilan, Y. Am. J. Pathol. 2009, 174, 1390Google Scholar164https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXksVOktbs%253D&md5=2819a669fcc785bd7fd777c749b89f13β-glycoglycosphingolipid-induced alterations of the STAT signaling pathways are dependent on CD1d and the lipid raft protein flotillin-2Lalazar, Gadi; Ben Ya'acov, Ami; Livovsky, Dan M.; El Haj, Madi; Pappo, Orit; Preston, Sarah; Zolotarov, Lidya; Ilan, YaronAmerican Journal of Pathology (2009), 174 (4), 1390-1399CODEN: AJPAA4; ISSN:0002-9440. (American Society for Investigative Pathology)β-Glucosylceramide has been shown to affect natural killer T cell function in models of inflammation. We, therefore, investigated the effects of different β-glycosphingolipids, including β-glucosylceramide, on STAT (signal transducers and activators of transcription) signaling pathways and detd. whether these effects were mediated by lipid raft microdomains and/or CD1d mols. The effects of α- and β-structured ligands on the lipid raft protein flotillin-2 were studied in both natural killer T hybridoma cells and leptin-deficient mice. To det. whether CD1d was involved in the effects of the β-glycosphingolipids, an anti-CD1d blocking antibody was used in a cell proliferation assay system. The downstream effects on the protein phosphorylation levels of STAT1, STAT3, and STAT6 were examd. in both immune-mediated hepatitis and hepatoma models. The effects of β-glycosphingolipids on the STAT signaling pathways were found to be dependent on CD1d. Lipid rafts were affected by both the dose and ratio of the β-glycosphingolipids and the acyl chain length, and these effects were followed by downstream effects on STAT proteins. Our results show that β-glycosphingolipids have beneficial effects in natural killer T cell-dependent immune-mediated metabolic and malignant animal models in vivo.
- 165Boggs, J. M.; Gao, W.; Zhao, J.; Park, H. J.; Liu, Y.; Basu, A. FEBS Lett. 2010, 584, 1771Google ScholarThere is no corresponding record for this reference.
- 166Venkataswamy, M. M.; Porcelli, S. A. Semin. Immunol. 2010, 22, 68Google Scholar166https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjt1Witbo%253D&md5=ec5bea20f0dc4d4d621280159366bc34Lipid and glycolipid antigens of CD1d-restricted natural killer T cellsVenkataswamy, Manjunatha M.; Porcelli, Steven A.Seminars in Immunology (2010), 22 (2), 68-78CODEN: SEIME2; ISSN:1044-5323. (Elsevier B.V.)A review. In spite of their relatively limited antigen receptor repertoire, CD1d-restricted NKT cells recognize a surprisingly diverse range of lipid and glycolipid antigens. Recent studies of natural and synthetic CD1d-presented antigens provide an increasingly detailed picture of how the specific structural features of these lipids and glycolipids influence their ability to be presented to NKT cells and stimulate their diverse immunol. functions. Particularly for synthetic analogs of α-galactosylceramides which have been the focus of intense recent investigation, it is becoming clear that the design of glycolipid antigens with the ability to precisely control the specific immunol. activities of NKT cells is likely to be feasible. The emerging details of the mechanisms underlying the structure-activity relationship of NKT cell antigens will assist greatly in the design and prodn. of immunomodulatory agents for the precise manipulation of NKT cells and the many other components of the immune system that they influence.
- 167Goff, R. D.; Gao, Y.; Mattner, J.; Zhou, D.; Yin, N.; Cantu, C., 3rd; Teyton, L.; Bendelac, A.; Savage, P. B. J. Am. Chem. Soc. 2004, 126, 13602Google ScholarThere is no corresponding record for this reference.
- 168Yu, K. O.; Im, J. S.; Molano, A.; Dutronc, Y.; Illarionov, P. A.; Forestier, C.; Fujiwara, N.; Arias, I.; Miyake, S.; Yamamura, T.; Chang, Y. T.; Besra, G. S.; Porcelli, S. A. Proc. Natl. Acad. Sci. U.S.A. 2005, 102, 3383Google ScholarThere is no corresponding record for this reference.
- 169Ishizuka, I. Prog. Lipid Res. 1997, 36, 245Google Scholar169https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXisVOku7o%253D&md5=15e559a87c9518b9c8b556cd60977255Chemistry and functional distribution of sulfoglycolipidsIshizuka, IneoProgress in Lipid Research (1997), 36 (4), 245-319CODEN: PLIRDW; ISSN:0163-7827. (Elsevier Science Ltd.)A review, with 667 refs. The topics discussed include: isolation and purifn.; anal. methods; structure; functional distribution; biosynthesis and biodegrdn.; and interaction with biomols.
- 170Popovic, Z. V.; Sandhoff, R.; Sijmonsma, T. P.; Kaden, S.; Jennemann, R.; Kiss, E.; Tone, E.; Autschbach, F.; Platt, N.; Malle, E.; Grone, H. J. J. Immunol. 2007, 179, 6770Google ScholarThere is no corresponding record for this reference.
- 171Merten, M.; Thiagarajan, P. Z. Kardiol. 2004, 93, 855Google Scholar171https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhsFSnug%253D%253D&md5=0820870057eee7fb59c0e13bdc019abeP-selectin in arterial thrombosisMerten, M.; Thiagarajan, P.Zeitschrift fuer Kardiologie (2004), 93 (11), 855-863CODEN: ZKRDAX; ISSN:0300-5860. (Steinkopff Verlag)A review. P-selectin is a transmembrane protein present in the a granules of platelets and the Weibel-Palade bodies of endothelial cells. Following activation, it is rapidly translocated to the cell surface. P-selectin expression in platelets has been shown to be elevated in disorders assocd. with arterial thrombosis such as coronary artery disease, acute myocardial infarction, stroke, and peripheral artery disease. P-selectin mediates rolling of platelets and leukocytes on activated endothelial cells as well as interactions of platelets with leukocytes. Platelet P-selectin interacts with P-selectin glycoprotein ligand-1 (PSGL-1) on leukocytes to form platelet-leukocyte aggregates. Furthermore, this interaction of P-selectin with PSGL-1 induces the upregulation of tissue factor, several cytokines in leukocytes and the prodn. of procoagulant microparticles, thereby contributing to a prothrombotic state. P-selectin is also involved in platelet-platelet interactions, i.e. platelet aggregation which is a major factor in arterial thrombosis. P-selectin interacts with platelet sulfatides, thereby stabilizing initial platelet aggregates formed.
- 172Shikata, K.; Suzuki, Y.; Wada, J.; Hirata, K.; Matsuda, M.; Kawashima, H.; Suzuki, T.; Iizuka, M.; Makino, H.; Miyasaka, M. J. Pathol. 1999, 188, 93Google Scholar172https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXjtF2ksb0%253D&md5=fece36b3b2937d87d2a0d0ba4c31e3efL-selectin and its ligands mediate infiltration of mononuclear cells into kidney interstitium after ureteric obstructionShikata, Kenichi; Suzuki, Yasuo; Wada, Jun; Hirata, Kyoji; Matsuda, Mitsuhiro; Kawashima, Hiroto; Suzuki, Takashi; Iizuka, Masako; Makino, Hirofumi; Miyasaka, MasayukiJournal of Pathology (1999), 188 (1), 93-99CODEN: JPTLAS; ISSN:0022-3417. (John Wiley & Sons Ltd.)It was previously reported that the L-selectin ligands detected by a rat L-selectin and human IgG chimeric mol. (rLEC-IgG) are expressed in the distal tubules of the kidney, where no leukocyte traffic is seen under physiol. conditions. In the present study, the role of L-selectin ligands in leukocyte infiltration into the kidney interstitium was investigated using a rat ureteric obstruction model. After ligation of the ureter, ligands for L-selectin rapidly disappeared from tubular epithelial cells and were relocated to the interstitium and peritubular capillary walls, where infiltration of monocytes and CD8+ T cells subsequently occurred. Mononuclear cell infiltration was significantly inhibited by i.v. injection of a neutralizing monoclonal antibody (MAb) against L-selectin, indicating the possible involvement of an L-selectin-mediated pathway. Interestingly, immunohistochem. studies with a MAb against sulfatide showed that the distribution of sulfatide, known to be one of the candidates of L-selectin ligand, was almost indistinguishable from the staining pattern of rLEC-IgG in both normal and ureteric obstructed kidneys, suggesting that sulfatide and/or related mol.(s) relocated to the renal interstitium were recognized by leukocyte L-selectin, leading to interstitial leukocyte infiltration. In line with this notion, i.v. injection of sulfatide markedly inhibited leukocyte infiltration, suggesting that L-selectin-sulfatide interaction may play a pivotal role in interstitial leukocyte infiltration in the kidney following ureteric obstruction.
- 173Kobayashi, T.; Honke, K.; Miyazaki, T.; Matsumoto, K.; Nakamura, T.; Ishizuka, I.; Makita, A. J. Biol. Chem. 1994, 269, 9817Google ScholarThere is no corresponding record for this reference.
- 174Don, A. S.; Rosen, H. Biochem. Biophys. Res. Commun. 2009, 380, 87Google ScholarThere is no corresponding record for this reference.
- 175Morichika, H.; Hamanaka, Y.; Tai, T.; Ishizuka, I. Cancer 1996, 78, 43Google ScholarThere is no corresponding record for this reference.
- 176Hiraiwa, N.; Fukuda, Y.; Imura, H.; Tadano-Aritomi, K.; Nagai, K.; Ishizuka, I.; Kannagi, R. Cancer Res. 1990, 50, 2917Google ScholarThere is no corresponding record for this reference.
- 177Sakakibara, N.; Gasa, S.; Kamio, K.; Makita, A.; Nonomura, K.; Togashi, M.; Koyanagi, T.; Hatae, Y.; Takeda, K. Cancer Lett. 1991, 57, 187Google Scholar177https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXksVKqur0%253D&md5=468f38864ffd7360947aed8a58803f4cDistinctive glycolipid patterns in Wilms' tumor and renal cell carcinomaSakakibara, N.; Gasa, Shinsei; Kamio, K.; Makita, A.; Nonomura, K.; Togashi, M.; Koyanagi, T.; Hatae, Y.; Takeda, K.Cancer Letters (Shannon, Ireland) (1991), 57 (3), 187-92CODEN: CALEDQ; ISSN:0304-3835.Glycolipid patterns were analyzed chromatog. in Wilms' tumor and renal-cell carcinoma tissues and compared with those of uninvolved tissue. Ganglioside GM3 was increased in both cancer tissues, whereas sulfatides accumulated only in renal-cell carcinoma, as reported earlier. Neolactotetraosylceramide was detected in both cancer tissues, but not in the uninvolved kidney tissues. In four cases of Wilms' tumors, only a low level of sulfotransferase towards galactosylceramide was found in one case, while no activity was detected in the three other cases. Present results show that the increased sulfatide(s) in the renal cell carcinoma and the deficiency of the sulfatides in Wilms' tumors appear to be biochem. characteristics of histol. differentt carcinomas.
- 178Li, J.; Pearl, D. K.; Pfeiffer, S. E.; Yates, A. J. J. Neurosci. Res. 1994, 39, 148Google Scholar178https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXmsFaksrY%253D&md5=9afb9838eeb3f21b04c5462418141579Patterns of reactivity with anti-glycolipid antibodies in human primary brain tumorsLi, J.; Pearl, D. K.; Pfeiffer, S. E.; Yates, A. J.Journal of Neuroscience Research (1994), 39 (2), 148-58CODEN: JNREDK; ISSN:0360-4012.Antibodies against carbohydrates of three glycolipids were used to det. patterns of immunohistochem. reactivity of histol. identifiable cell subpopulations in 101 human primary brain tumors. For all tumor types fibrillary cells, polar cells, and gemistocytes (commonly seen in astrocytomas and ependymomas) stained more frequently for galactosylcerebroside with mAbO1 than small tumor cells and macrophages. Frequency of staining for sulfatide with mAbO4 was fibrillary > polar > small cells = macrophages. Gemistocytes stained more frequently with mAbO4 than polar cells in all tumors except low grade astrocytomas. These data indicate that tumors classified on histol. grounds as astrocytic are often stained with antibodies that recognize oligodendrocytes and their progenitors. Thus, anti-glycolipid antibodies used in the study of developmental lineage may offer useful tools for classification of human brain tumors. Staining of fibrillary cells, polar cells, and gemistocytes for paragloboside directly with mAb F1H11 was much less common than with mAbO1, but this increased by pretreatment of the tissues with neuraminidase (F1H11+N). Of particular note was the finding that small tumor cells frequently stained with F1H11+N. Evidence that these were not macrophages was obtained using double immunostaining with F1H11+N and anti-macrophage antibodies. In astrocytomas the frequency of small tumor cells immunostained with F1H11+N was high grade > anaplastic > low grade, demonstrating a correlation of this tumor cell population with more aggressive astrocytomas. Thus, immunostaining with F1H11+N may be of value in identifying small, anaplastic tumor cells, esp. in small biopsies or tissue taken adjacent to the main tumor mass.
- 179Gnewuch, C.; Jaques, G.; Havemann, K.; Wiegandt, H. Int. J. Cancer Suppl 1994, 8, 125Google Scholar179https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK2c3lt1Ogtg%253D%253D&md5=d8b81235504c58aa40224bcd500d986fRe-assessment of acidic glycosphingolipids in small-cell-lung-cancer tissues and cell linesGnewuch C; Jaques G; Havemann K; Wiegandt HInternational journal of cancer. Supplement = Journal international du cancer. Supplement (1994), 8 (), 125-6 ISSN:0898-6924.The occurrence of tumor-associated glycosphingolipids (GSLs) has been documented in a variety of cancer tissues (Hakomori, 1984, 1985, 1989). In the case of small-cell lung cancer (SCLC), the monosialoganglioside IV2Fuc-II3NeuAc-Gg4Cer (Fuc-GM1; short notations of gangliosides are according to Svennerholm, 1963), first described from bovine liver (Wiegandt, 1973), was found to be a unique tumor-associated GSL (Nilsson et al., 1984). It is present in up to 90% of all SCLC cases as compared with 25% frequency in non-SCLC, and no occurrence in normal lung (Brezicka et al., 1989, 1992). Thus, Fuc-GM1 may represent a suitable target antigen for immunotherapy of SCLC, and successful experiments have been performed showing tumor-cell killing by monoclonal antibodies (MAbs) against Fuc-GM1, both in vitro and, in a mouse model, in vivo (Brezicka et al., 1991). However, an effective tumor vaccination in humans would require this antigen to be expressed by the primary tumor and also by all metastases. The co-expression of Fuc-GM1 has already been reported in primary tumors and in most but not all metastases of SCLC (Hanquing et al., 1986; Nilsson et al., 1986; Brezicka et al., 1989). In view of the significance this ganglioside may have for possible immunotherapeutical approaches to SCLC and of the difficulty in obtaining a sufficient number of samples for analysis, a re-assessment of Fuc-GM1 expression was made in SCLC primary tumors and their metastases, as well as in established SCLC cell lines. In addition, the possible presence of such gangliosides, that might help to explain the selective tetanus-toxin binding of SCLC cells (Critchley et al., 1986; Heymanns et al., 1989) was investigated. Finally, the typical occurrence of sulfatide in all SCLC tissues and cell lines could be established.
- 180Liu, Y.; Chen, Y.; Momin, A.; Shaner, R.; Wang, E.; Bowen, N. J.; Matyunina, L. V.; Walker, L. D.; McDonald, J. F.; Sullards, M. C.; Merrill, A. H., Jr. Mol. Cancer 2010, 9, 186Google Scholar180https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3cjhvFGrsw%253D%253D&md5=24a811e26d9f81400db52a07c560b06eElevation of sulfatides in ovarian cancer: an integrated transcriptomic and lipidomic analysis including tissue-imaging mass spectrometryLiu Ying; Chen Yanfeng; Momin Amin; Shaner Rebecca; Wang Elaine; Bowen Nathan J; Matyunina Lilya V; Walker L Deette; McDonald John F; Sullards M Cameron; Merrill Alfred H JrMolecular cancer (2010), 9 (), 186 ISSN:.BACKGROUND: Sulfatides (ST) are a category of sulfated galactosylceramides (GalCer) that are elevated in many types of cancer including, possibly, ovarian cancer. Previous evidence for elevation of ST in ovarian cancer was based on a colorimetric reagent that does not provide structural details and can also react with other lipids. Therefore, this study utilized mass spectrometry for a structure-specific and quantitative analysis of the types, amounts, and tissue localization of ST in ovarian cancer, and combined these findings with analysis of mRNAs for the relevant enzymes of ST metabolism to explore possible mechanisms. RESULTS: Analysis of 12 ovarian tissues graded as histologically normal or having epithelial ovarian tumors by liquid chromatography electrospray ionization-tandem mass spectrometry (LC ESI-MS/MS) established that most tumor-bearing tissues have higher amounts of ST. Because ovarian cancer tissues are comprised of many different cell types, histological tissue slices were analyzed by matrix-assisted laser desorption ionization-tissue-imaging MS (MALDI-TIMS). The regions where ST were detected by MALDI-TIMS overlapped with the ovarian epithelial carcinoma as identified by H & E staining and histological scoring. Furthermore, the structures for the most prevalent species observed via MALDI-TIMS (d18:1/C16:0-, d18:1/C24:1- and d18:1/C24:0-ST) were confirmed by MALDI-TIMS/MS, whereas, a neighboring ion(m/z 885.6) that was not tumor specific was identified as a phosphatidylinositol. Microarray analysis of mRNAs collected using laser capture microdissection revealed that expression of GalCer synthase and Gal3ST1 (3'-phosphoadenosine-5'-phosphosulfate:GalCer sulfotransferase) were approximately 11- and 3.5-fold higher, respectively, in the ovarian epithelial carcinoma cells versus normal ovarian stromal tissue, and they were 5- and 2.3-fold higher in comparison with normal surface ovarian epithelial cells, which is a likely explanation for the higher ST. CONCLUSIONS: This study combined transcriptomic and lipidomic approaches to establish that sulfatides are elevated in ovarian cancer and should be evaluated further as factors that might be important in ovarian cancer biology and, possibly, as biomarkers.
- 181Harris, J. F.; Beaton, D. W. Clin. Exp. Metastasis 1990, 8, 361Google Scholar181https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXksVOj&md5=61917b9fc3cf61dee71ad167364e40feSulfated glycoconjugate determinants recognized by monoclonal antibody, SG-1, correlate with the experimental metastatic ability of KHT fibrosarcoma cellsHarris, J. F.; Beaton, D. W.Clinical & Experimental Metastasis (1990), 8 (4), 361-79CODEN: CEXMD2; ISSN:0262-0898.The binding and functional activity was examd. of monoclonal antibody (MAb) SG-1 that was raised by immunization against embryonal carcinoma cells and screened using KHT fibrosarcoma cells. Quant. absorption, binding and in situ immunochem. staining assays indicate that the MAb SG-1-defined epitopes are expressed preferentially by the highly metastatic KHT35-L1 cells relative to the weakly metastatic, parental KHTp cells. Furthermore, there was a significant correlation between the expression of MAb SG-1-defined antigen on the cells, following trypsin treatment, and their metastatic ability. Binding of MAb SG-1 to antigen was inhibited by specific sulfated polysaccharides including cerebroside sulfate (brain sulfatide), fucoidan, and dextran sulfate (500 kD) but not by heparin, chondroitin, keratan, or dextran (5 kD) sulfates. Initial characterization of antigen from KHT cells indicates that the epitope of MAb SG-1 is defined by sulfated glycoconjugates contg. galactose and sulfate but not N-acetylglucosamine. In the total lipid exts. of KHT35-L1 cells the antigen was detected in the delipidated protein fraction as well as in the chloroform/methanol fraction. Thus, the sulfated glycoconjugate determinants identified by MAb SG-1 may be relevant to the metastatic process of KHT fibrosarcoma cells.
- 182Iwabuchi, K.; Nagaoka, I. Blood 2002, 100, 1454Google ScholarThere is no corresponding record for this reference.
- 183Chatterjee, S.; Pandey, A. Biochim. Biophys. Acta 2008, 1780, 370Google ScholarThere is no corresponding record for this reference.
- 184Won, J. S.; Singh, A. K.; Singh, I. J. Neurochem. 2007, 103 (Suppl 1) 180Google Scholar184https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtl2jurrF&md5=46147041f1c071bd37a01ea9bef1c757Lactosylceramide: a lipid second messenger in neuroinflammatory diseaseWon, Je-Seong; Singh, Avtar K.; Singh, InderjitJournal of Neurochemistry (2007), 103 (Suppl. 1), 180-191CODEN: JONRA9; ISSN:0022-3042. (Blackwell Publishing Ltd.)A review. Inflammatory disease plays a crit. role in the pathogenesis of many neurol. disorders. Astrogliosis and induction of pro-inflammatory mediators such as chemokines, cytokines and inducible nitric oxide synthase (iNOS) are the 'hallmarks' of inflammatory disease. Increased activity of lactosylceramide (LacCer) synthase and increased synthesis of LacCer during glial proliferation, and induction of pro-inflammatory cytokines and iNOS suggests a role for LacCer in these cellular signaling pathways. Studies using complementary techniques of inhibitors and antisense reported that inhibition of LacCer synthesis inhibits glial proliferation, as well as the induction of pro-inflammatory mediators (cytokines and iNOS). This inhibition was bypassed by exogenous LacCer, but not by other related lipids (e.g. glucosylceramide, galactocerebroside, GD1, GM1), indicating a role for LacCer in inflammatory signaling pathways. Furthermore, inhibition of glial proliferation and induction of inflammatory mediators by antisense to Ras GTPase, PI3-Kinase and inhibitors of mitogen-activated protein kinase indicate the participation of the phosphoinositide 3-kinase (PI3Kinas)/Ras/mitogen-activated protein kinase/nuclear factor-κB (NF-κB) signaling pathways in LacCer-mediated inflammatory events thus exposing addnl. targets for therapeutics for inflammatory disease conditions.
- 185Nakayama, H.; Yoshizaki, F.; Prinetti, A.; Sonnino, S.; Mauri, L.; Takamori, K.; Ogawa, H.; Iwabuchi, K. J. Leukocyte Biol. 2008, 83, 728Google Scholar185https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXjtVaqtbw%253D&md5=6d722ed2e4edb68664adab30bdf71572Lyn-coupled LacCer-enriched lipid rafts are required for CD11b/CD18-mediated neutrophil phagocytosis of nonopsonized microorganismsNakayama, Hitoshi; Yoshizaki, Fumiko; Prinetti, Alessandro; Sonnino, Sandro; Mauri, Laura; Takamori, Kenji; Ogawa, Hideoki; Iwabuchi, KazuhisaJournal of Leukocyte Biology (2008), 83 (3), 728-741CODEN: JLBIE7; ISSN:0741-5400. (Federation of American Societies for Experimental Biology)The integrin CD11b/CD18 plays a central role in neutrophil phagocytosis. Although CD11b/CD18 binds a wide range of ligands, including C3bi and β-glucan, and transmits outside-in signaling, the mechanism of this signaling responsible for phagocytosis remains obscure. Here, we report that lactosylceramide (LacCer)-enriched lipid rafts are required for CD11b/CD18-mediated phagocytosis of nonopsonized zymosans (NOZs) by human neutrophils. Anti-CD11b and anti-LacCer antibodies inhibited the binding of NOZs to neutrophils and the phagocytosis of NOZs. During phagocytosis of NOZ, CD11b and LacCer were accumulated and colocalized in the actin-enriched phagocytic cup regions. Immunopptn. expts. suggested that CD11b/CD18 was mobilized into the LacCer-enriched lipid rafts during phagocytosis of NOZs. DMSO-treated, neutrophil-like HL-60 cells (D-HL-60 cells) lacking Lyn-coupled, LacCer-mediated signaling showed little phagocytosis of NOZs. However, loading of D-HL-60 cells with C24 fatty acid chain-contg. LacCer (C24-LacCer) reconstructed functional Lyn-assocd., LacCer-enriched lipid rafts, and restored D-HL-60 cell NOZ phagocytic activity, which was inhibited by anti-LacCer and anti-CD11b antibodies. Lyn knockdown by small interfering RNA blocked the effect of C24:1-LacCer loading on D-HL-60 cell phagocytosis of NOZs. CD11b/CD18 activation expts. indicated phosphorylation of LacCer-assocd. Lyn by activation of CD11b. Taken together, these observations suggest that CD11b activation causes translocation of CD11b/CD18 into Lyn-coupled, LacCer-enriched lipid rafts, allowing neutrophils to phagocytose NOZs via CD11b/CD18.
- 186Sonnino, S.; Prinetti, A.; Nakayama, H.; Yangida, M.; Ogawa, H.; Iwabuchi, K. Glycoconj. J. 2009, 26, 615Google Scholar186https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXovVOiu7k%253D&md5=d8b798d538518e2918fb33416cb938a9Role of very long fatty acid-containing glycosphingolipids in membrane organization and cell signaling: The model of lactosylceramide in neutrophilsSonnino, Sandro; Prinetti, Alessandro; Nakayama, Hitoshi; Yangida, Mitsuaki; Ogawa, Hideoki; Iwabuchi, KazuhisaGlycoconjugate Journal (2009), 26 (6), 615-621CODEN: GLJOEW; ISSN:0282-0080. (Springer)A review. Glycosphingolipids are highly enriched in specialized membrane microdomains ("lipid rafts", caveolar domains and glycosynapses), and they participate to the process of transduction of information across the membrane. Lactosylceramide (LacCer) is specifically coupled with the Src family kinase Lyn in plasma membrane microdomains of human neutrophils. Ligand binding to LacCer activates Lyn, resulting in neutrophil functions, such as superoxide generation and migration. The β-Gal-(1-4)-β-Glc disaccharide structure of LacCer is necessary, but it is not sufficient for LacCer-mediated Lyn activation. For this function, the presence of a LacCer mol. species with ceramide contg. a very long fatty acid chain is also required. In this manuscript, the authors discuss the importance of interdigitation within the membrane, promoted by the presence of glycosphingolipid species with very long fatty acyl chains as determinants for membrane organization, instrumental to the signaling process.
- 187Iwabuchi, K.; Nakayama, H.; Iwahara, C.; Takamori, K. FEBS Lett. 2010, 584, 1642Google ScholarThere is no corresponding record for this reference.
- 188Seah, N.; Santacroce, P. V.; Basu, A. Org. Lett. 2009, 11, 559Google ScholarThere is no corresponding record for this reference.
- 189Bremer, E. G.; Schlessinger, J.; Hakomori, S. J. Biol. Chem. 1986, 261, 2434Google ScholarThere is no corresponding record for this reference.
- 190Zhou, Q.; Hakomori, S.; Kitamura, K.; Igarashi, Y. J. Biol. Chem. 1994, 269, 1959Google ScholarThere is no corresponding record for this reference.
- 191Yoon, S. J.; Nakayama, K.; Hikita, T.; Handa, K.; Hakomori, S. I. Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 18987Google ScholarThere is no corresponding record for this reference.
- 192Coskun, U.; Grzybek, M.; Drechsel, D.; Simons, K. Proc. Natl. Acad. Sci. U.S.A. 2011, 108, 9044Google ScholarThere is no corresponding record for this reference.
- 193Bremer, E. G.; Hakomori, S.; Bowen-Pope, D. F.; Raines, E.; Ross, R. J. Biol. Chem. 1984, 259, 6818Google ScholarThere is no corresponding record for this reference.
- 194von Gunten, S.; Bochner, B. S. Ann. N.Y. Acad. Sci. 2008, 1143, 61Google ScholarThere is no corresponding record for this reference.
- 195Bekri, S.; Lidove, O.; Jaussaud, R.; Knebelmann, B.; Barbey, F. Cardiovasc. Hematol. Agents Med. Chem. 2006, 4, 289Google ScholarThere is no corresponding record for this reference.
- 196Utskarpen, A.; Massol, R.; van Deurs, B.; Lauvrak, S. U.; Kirchhausen, T.; Sandvig, K. PLoS One 2010, 5, e10944Google ScholarThere is no corresponding record for this reference.
- 197Adlercreutz, D.; Weadge, J. T.; Petersen, B. O.; Duus, J. O.; Dovichi, N. J.; Palcic, M. M. Carbohydr. Res. 2010, 345, 1384Google ScholarThere is no corresponding record for this reference.
- 198Falguieres, T.; Maak, M.; von Weyhern, C.; Sarr, M.; Sastre, X.; Poupon, M. F.; Robine, S.; Johannes, L.; Janssen, K. P. Mol. Cancer Ther. 2008, 7, 2498Google Scholar198https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtVWksLzP&md5=5f68dcc23423c53d82ad63bb9dae8fc7Human colorectal tumors and metastases express Gb3 and can be targeted by an intestinal pathogen-based delivery toolFalguieres, Thomas; Maak, Matthias; von Weyhern, Claus; Sarr, Marianne; Sastre, Xavier; Poupon, Marie-France; Robine, Sylvie; Johannes, Ludger; Janssen, Klaus-PeterMolecular Cancer Therapeutics (2008), 7 (8), 2498-2508CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)The targeting of solid tumors requires delivery tools that resist intracellular and extracellular inactivation, and that are taken up specifically by tumor cells. We have shown previously that the recombinant nontoxic B-subunit of Shiga toxin (STxB) can serve as a delivery tool to target digestive tumors in animal models. The aim of this study was to expand these expts. to human colorectal cancer. Tissue samples of normal colon, benign adenomas, colorectal carcinomas, and liver metastases from 111 patients were obtained for the quantification of the expression of the cellular STxB receptor, the glycosphingolipid globotriaosyl ceramide (Gb3 or CD77). We found that compared with normal tissue, the expression of Gb3 was strongly increased in colorectal adenocarcinomas and their metastases, but not in benign adenomas. Short-term primary cultures were prepd. from samples of 43 patients, and STxB uptake was studied by immunofluorescence microscopy. Of a given tumor sample, on av., 80% of the cells could visibly bind STxB, and upon incubation at 37°C, STxB was transported to the Golgi app., following the retrograde route. This STxB-specific intracellular targeting allows the mol. to avoid recycling and degrdn., and STxB could consequently be detected on tumor cells even 5 days after initial uptake. In conclusion, the targeting properties of STxB could be diverted for the delivery of contrast agents to human colorectal tumors and their metastases, whose early detection and specific targeting remains one of the principal challenges in oncol.
- 199Johannes, L.; Romer, W. Nat. Rev. Microbiol. 2010, 8, 105Google Scholar199https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFyhsLnP&md5=f2e9fd1b0ff7ab92efe8bf647745e7fdShiga toxins: From cell biology to biomedical applicationsJohannes, Ludger; Romer, WinfriedNature Reviews Microbiology (2010), 8 (2), 105-116CODEN: NRMACK; ISSN:1740-1526. (Nature Publishing Group)A review. Shiga toxin-producing Escherichia coli is an emergent pathogen that can induce hemolytic uremic syndrome. The toxin has received considerable attention not only from microbiologists but also in the field of cell biol., where it has become a powerful tool to study intracellular trafficking. In this Review, we summarize the Shiga toxin family members and their structures, receptors, trafficking pathways and cellular targets. We discuss how Shiga toxin affects cells not only by inhibiting protein biosynthesis but also through the induction of signalling cascades that lead to apoptosis. Finally, we discuss how Shiga toxins might be exploited in cancer therapy and immunotherapy.
- 200Engedal, N.; Skotland, T.; Torgersen, M. L.; Sandvig, K. Microb. Biotechnol. 2011, 4, 32Google Scholar200https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFyjtLY%253D&md5=3be8d6de083c48ca213f1df8d89e5ab6Shiga toxin and its use in targeted cancer therapy and imagingEngedal, Nikolai; Skotland, Tore; Torgersen, Maria L.; Sandvig, KirstenMicrobial Biotechnology (2010), 4 (1), 32-46CODEN: MBIIB2; ISSN:1751-7915. (Wiley-Blackwell)A review. Shiga and the Shiga-like toxins are related protein toxins produced by Shigella dysenteriae and certain strains of Escherichia coli. These toxins are composed of two non-covalently attached, modular parts: the A moiety (StxA) contg. the enzymically active A1 fragment, and the non-toxic, pentameric binding moiety (StxB). Stx binds specifically to the glycosphingolipid globotriaosylceramide (Gb3) at the surface of target cells and is then internalized by endocytosis. Subsequently, in toxin-sensitive cells, the Stx/Gb3 complex is transported in a retrograde manner via the Golgi app. to the endoplasmic reticulum, where the enzymically active part of Stx is translocated to the cytosol, enabling it to irreversibly inhibit protein synthesis via modification of ribosomal 28S RNA. Whereas Gb3 shows a relatively restricted expression in normal human tissues, it has been reported to be highly expressed in many types of cancers. This review gives a brief introduction to Stx and its intracellular transport. Furthermore, after a description of Gb3 and the methods that are currently used to detect its cellular expression, we provide an updated overview of the published reports on Gb3 overexpression in human cancers. Finally, we discuss the possibility of utilizing Stx or StxB coupled to therapeutic compds. or contrast agents in targeted cancer therapy and imaging.
- 201Christiansen, D.; Milland, J.; Mouhtouris, E.; Vaughan, H.; Pellicci, D. G.; McConville, M. J.; Godfrey, D. I.; Sandrin, M. S. PLoS Biol. 2008, 6, e172Google ScholarThere is no corresponding record for this reference.
- 202Biellmann, F.; Hulsmeier, A. J.; Zhou, D.; Cinelli, P.; Hennet, T. BMC Dev. Biol. 2008, 8, 109Google ScholarThere is no corresponding record for this reference.
- 203Kuan, C. T.; Chang, J.; Mansson, J. E.; Li, J.; Pegram, C.; Fredman, P.; McLendon, R. E.; Bigner, D. D. BMC Dev. Biol. 2010, 10, 114Google ScholarThere is no corresponding record for this reference.
- 204Westerlund, B.; Slotte, J. P. Biochim. Biophys. Acta 2009, 1788, 194Google ScholarThere is no corresponding record for this reference.
- 205Wennekes, T.; van den Berg, R. J.; Boot, R. G.; van der Marel, G. A.; Overkleeft, H. S.; Aerts, J. M. Angew. Chem., Int. Ed. Engl. 2009, 48, 8848Google ScholarThere is no corresponding record for this reference.
- 206Degroote, S.; Wolthoorn, J.; van Meer, G. Semin. Cell Dev. Biol. 2004, 15, 375Google Scholar206https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXltVCisLw%253D&md5=9a9b4195c6790c778ea02e48d041b89dThe cell biology of glycosphingolipidsDegroote, Sophie; Wolthoorn, Jasja; Van Meer, GerritSeminars in Cell & Developmental Biology (2004), 15 (4), 375-387CODEN: SCDBFX; ISSN:1084-9521. (Elsevier Science B.V.)A review. Glycosphingolipids, a family of heterogeneous lipids with biophys. properties conserved from fungi to mammals, are key components of cellular membranes. Because of their tightly packed backbone, they have the ability to assoc. with other sphingolipids and cholesterol to form microdomains called lipid rafts, with which a variety of proteins assoc. These microdomains are thought to originate in the Golgi app., where most sphingolipids are synthesized, and are enriched at the plasma membrane. They are involved in an increasing no. of processes, including sorting of proteins by allowing selectivity in intracellular membrane transport. Apart from being involved in recognition and signaling on the cell surface, glycosphingolipids may fulfill unexpected roles on the cytosolic surface of cellular membranes.
- 207Ledeen, R. W.; Wu, G. Biochim. Biophys. Acta 2006, 1761, 588Google ScholarThere is no corresponding record for this reference.
- 208Dyatlovitskaya, E. V. Biochemistry (Moscow) 2007, 72, 479Google Scholar208https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXlslOgt7c%253D&md5=bc0185fd6e7636e32e3cccd3595e1d2cThe role of lysosphingolipids in the regulation of biological processesDyatlovitskaya, E. V.Biochemistry (Moscow) (2007), 72 (5), 479-484CODEN: BIORAK; ISSN:0006-2979. (Pleiades Publishing, Ltd.)A review. This review summarizes data on the role of lysosphingolipids (glucosyl-and galactosylsphingosines, sphingosine-1-phosphate, sphingosine-1-phosphocholine) in the regulation of various biol. processes in normal and pathol. states.
- 209Meyer zu Heringdorf, D.; Jakobs, K. H. Biochim. Biophys. Acta 2007, 1768, 923Google ScholarThere is no corresponding record for this reference.
- 210Nixon, G. F.; Mathieson, F. A.; Hunter, I. Prog. Lipid Res. 2008, 47, 62Google Scholar210https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVSmurbK&md5=99250499a57eabb319c5e729113a35f3The multi-functional role of sphingosylphosphorylcholineNixon, Graeme F.; Mathieson, Fiona A.; Hunter, IreneProgress in Lipid Research (2008), 47 (1), 62-75CODEN: PLIRDW; ISSN:0163-7827. (Elsevier B.V.)A review. The sphingomyelin metabolite, sphingosylphosphorylcholine (SPC) has been the subject of much recent interest and controversy. Studies have indicated that SPC naturally occurs in plasma and a constituent of lipoproteins. Synthesis is also increased in some pathol. conditions. Research has demonstrated that SPC is a potentially important lipid mediator of cell type specific functions in major tissues, such as heart, blood vessels, skin, brain and immune system. These effects are regulated via a no. of different intracellular signalling cascades, also dependent upon cell type. Initial reports identifying high affinity SPC receptors at first appeared to reinforce the physiol. relevance of this sphingolipid. However, these studies have now been retracted. Some SPC effects have been shown be occur via plasma membrane receptors for the related sphingolipid, sphingosine 1-phosphate (S1P). Despite a lack of well-defined receptor signal transduction mechanisms and sparse pharmacol. data, several key characteristics of SPC are now emerging. SPC can act as a mitogen in several different cell types and in certain circumstances, may also be a pro-inflammatory mediator. In this review, these actions of SPC are discussed with a view to understanding the potential physiol. relevance of this sphingolipid.
- 211Okamoto, R.; Arikawa, J.; Ishibashi, M.; Kawashima, M.; Takagi, Y.; Imokawa, G. J. Lipid Res. 2003, 44, 93Google ScholarThere is no corresponding record for this reference.
- 212Imokawa, G. J. Dermatol. Sci. 2009, 55, 1Google Scholar212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXms12jtLw%253D&md5=ba310f8a159f5c7534f9b5f8568316c3A possible mechanism underlying the ceramide deficiency in atopic dermatitis: Expression of a deacylase enzyme that cleaves the N-acyl linkage of sphingomyelin and glucosylceramideImokawa, GenjiJournal of Dermatological Science (2009), 55 (1), 1-9CODEN: JDSCEI; ISSN:0923-1811. (Elsevier Ireland Ltd.)A review. A deficiency of ordinary ceramides in the stratum corneum is an essential etiol. factor for the dry and barrier-disrupted skin of patients with atopic dermatitis (AD). We have proposed that the mechanism underlying that deficiency involves a novel sphingolipid metabolizing enzyme, termed sphingomyelin (SM) glucosylceramide (GCer) deacylase, which hydrolyzes SM or GCer at the acyl site to yield their lysoforms sphingosylphosphorylcholine (SPC) or glucosylsphingosine (GSP) instead of ceramide, leading to the ceramide deficiency in the AD skin. The enzymic characteristics obsd. showed a pH dependency of catalytic activity with a peak at pH 5.0 and a mol. wt. of 40,000. Anal. isoelec. focusing (IEF) chromatog. demonstrated that the pI values of SM deacylase, GlcCDase, SMase and ceramidase were 4.2, 7.4, 7.0 and 5.7, resp. Those enzymic characteristics of SM-GCer deacylase are completely distinct from ceramidase as well as the other known deacylases. Our enzymic measurements demonstrated that SM-GCer deacylase activity is enhanced more than 5-fold in involved stratum corneum, more than 3-fold in uninvolved stratum corneum and approx. 3-fold in the involved epidermis from patients with AD compared with healthy controls. Our findings suggest that the novel enzyme, SM-GCer deacylase, is expressed in situ at significant levels in the epidermis of AD patients. This results in the prodn. of SPC and GSP, instead of ceramides, which leads in turn to the ceramide deficiency seen in the stratum corneum of those patients. It is likely that the biogenesis of SM-GCer deacylase may be crit. to the pathogenesis of AD.
- 213Suzuki, K. J. Neurochem. Res. 1998, 23, 251Google Scholar213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXosFGhug%253D%253D&md5=8fdada81983efdf275b4ea756529eecbTwenty five years of the "psychosine hypothesis": a personal perspective of its history and present statusSuzuki, KunihikoNeurochemical Research (1998), 23 (3), 251-259CODEN: NEREDZ; ISSN:0364-3190. (Plenum Publishing Corp.)A review, with 54 refs. Twenty five years ago in 1972, a hypothesis was introduced to explain the pathogenetic mechanism underlying the unusual cellular and biochem. characteristics of globoid cell leukodystrophy (Krabbe disease). It postulated that galactosylsphingosine (psychosine), which cannot be degraded due to the underlying genetic defect, is responsible for the very rapid loss of the oligodendrocytes and the consequent paradoxical anal. finding, the lack of accumulation of the primary substrate, galactosylceramide, in patients' brain. It took nearly ten years before the actual accumulation of psychosine was demonstrated in human Krabbe patients and also in the brain of twitcher mice, an equiv. murine mutant. Meanwhile this "psychosine hypothesis" has been extended to Gaucher disease and then to a more general hypothesis encompassing all sphingolipidoses that the "lyso-derivs." of the primary sphingolipid substrates of the defective enzymes in resp. disorders play a key role in their pathogenesis. Some of these extensions not only remain speculative without conclusive factual evidence but may eventually turn out to be an overstretching. This article attempts, from my personal perspective, at tracing historical development of the "psychosine hypothesis" and examg. its current status and possible future directions.
- 214Carter, H. E.; Nalbandov, O.; Tavormina, P. A. J. Biol. Chem. 1951, 192, 197Google ScholarThere is no corresponding record for this reference.
- 215Kisic, A.; Tsuda, M.; Kulmacz, R. J.; Wilson, W. K.; Schroepfer, G. J., Jr. J. Lipid Res. 1995, 36, 787Google ScholarThere is no corresponding record for this reference.
- 216Igarashi, Y.; Hakomori, S. Biochem. Biophys. Res. Commun. 1989, 164, 1411Google ScholarThere is no corresponding record for this reference.
- 217Morales, P. R.; Dillehay, D. L.; Moody, S. J.; Pallas, D. C.; Pruett, S.; Allgood, J. C.; Symolon, H.; Merrill, A. H., Jr. Drug Chem. Toxicol. 2007, 30, 197Google Scholar217https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXptV2hsbo%253D&md5=a4ad9d68028ca843b86587ebe2d51c8bSafingol toxicology after oral administration to TRAMP mice: demonstration of safingol uptake and metabolism by N-acylation and N-methylationMorales, Pablo R.; Dillehay, Dirck L.; Moody, Steven J.; Pallas, David C.; Pruett, Sarah; Allgood, Jeremy C.; Symolon, Holly; Merrill, Alfred H., Jr.Drug and Chemical Toxicology (1977) (2007), 30 (3), 197-216CODEN: DCTODJ; ISSN:0148-0545. (Informa Healthcare)Safingol [(2S,3S)-2-amino-1,3-octadecanediol] is an unnatural L-threo-stereoisomer of sphinganine that is cytotoxic for cancer cells in culture and is being tested in phase 1 human clin. trials. To det. if safingol can be absorbed orally and if it affects prostate cancer in a mouse strain used in prostate cancer studies, safingol was fed to TRAMP (transgenic adenocarcinoma of mouse prostate) mice for 2 wk at 0.0125% to 0.1% wt./wt. of the diet. Anal. of safingol and safingol metabolites in blood and tissues by liq. chromatog. electrospray ionization tandem mass spectrometry revealed uptake in tissue and extensive conversion of safingol to N-acyl species (comparable to natural "ceramides") and mono-, di-, and tri-N-Me metabolites that have not been obsd. previously. Safingol caused significant hepatotoxicity at all dosages, as reflected in elevated liver alanine aminotransferase, and at the highest dose (0.1 %) caused changes in liver histol. (appearance of autophagosomal vacuoles) and renal toxicity (based on elevation of blood urea nitrogen) and decreases in packed blood cell vol. and body wt. Safingol did not inhibit the prostate pre-neoplastic lesion (prostate intraepithelial neoplasia) in TRAMP mice; however, addnl. studies at lower dosages for longer time were not pursued due to host toxicity. Safingol and its N-Me metabolites were cytotoxic to both a human prostate cell line (DU145) and mouse BALB 3T3 cells; therefore, the host and potential antitumor toxicity may be due to multiple mol. species of safingol.
- 218Doering, T.; Proia, R. L.; Sandhoff, K. FEBS Lett. 1999, 447, 167Google ScholarThere is no corresponding record for this reference.
- 219Stewart, M. E.; Downing, D. T. J. Lipid Res. 2001, 42, 1105Google ScholarThere is no corresponding record for this reference.
- 220Farwanah, H.; Pierstorff, B.; Schmelzer, C. E.; Raith, K.; Neubert, R. H.; Kolter, T.; Sandhoff, K. J. Chromatogr., B: Anal. Technol. Biomed. Life Sci. 2007, 852, 562Google Scholar220https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmtVGlsL4%253D&md5=a62744ee36727fdffff09daca8af4f2bSeparation and mass spectrometric characterization of covalently bound skin ceramides using LC/APCI-MS and Nano-ESI-MS/MSFarwanah, Hany; Pierstorff, Barbara; Schmelzer, Christian E. H.; Raith, Klaus; Neubert, Reinhard H. H.; Kolter, Thomas; Sandhoff, KonradJournal of Chromatography, B: Analytical Technologies in the Biomedical and Life Sciences (2007), 852 (1-2), 562-570CODEN: JCBAAI; ISSN:1570-0232. (Elsevier B.V.)Ceramides covalently bound to keratinocytes are essential for the barrier function of the skin, which can be disturbed in diseases, such as psoriasis and atopic dermatitis. These ceramides of the classes ω-hydroxyacyl-sphingosine and ω-hydroxyacyl-6-hydroxysphingosine contain an ω-hydroxy fatty acid. For their sepn. and identification, a new anal. approach based on normal phase liq. chromatog. coupled to atm. pressure chem. ionization mass spectrometry and tandem nano-electrospray mass spectrometry, resp., is presented here. Tandem mass spectrometry provided structural information about the sphingoid base as well as the fatty acid moieties. The chain lengths of the bases ranged from C12 to C22, the chain lengths of the fatty acids varied between C28 and C36. In total, 67 ceramide species have been identified in human skin. The anal. methods presented in this work can be helpful for investigating alterations in the ceramide compn. of the skin as seen in psoriasis, atopic dermatitis, and diseases with impaired epidermal barrier function.
- 221Bosson, R.; Guillas, I.; Vionnet, C.; Roubaty, C.; Conzelmann, A. Eukaryot. Cell 2009, 8, 306Google Scholar221https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlsFyjtrg%253D&md5=e72654fe825576ecca2e6b1d290834eeIncorporation of ceramides into Saccharomyces cerevisiae glycosylphosphatidylinositol-anchored proteins can be monitored in vitroBosson, Regine; Guillas, Isabelle; Vionnet, Christine; Roubaty, Carole; Conzelmann, AndreasEukaryotic Cell (2009), 8 (3), 306-314CODEN: ECUEA2; ISSN:1535-9786. (American Society for Microbiology)After glycosylphosphatidylinositols (GPIs) are added to GPI proteins of Saccharomyces cerevisiae, a fatty acid of the diacylglycerol moiety is exchanged for a C26:0 fatty acid through the subsequent actions of Per1 and Gup1. In most GPI anchors this modified diacylglycerol-based anchor is subsequently transformed into a ceramide-contg. anchor, a reaction which requires Cwh43. Here we show that the last step of this GPI anchor lipid remodeling can be monitored in microsomes. The assay uses microsomes from cells that have been grown in the presence of myriocin, a compd. that blocks the biosynthesis of dihydrosphingosine (DHS) and thus inhibits the biosynthesis of ceramide-based anchors. Such microsomes, when incubated with [3H]DHS, generate radiolabeled, ceramide-contg. anchor lipids of the same structure as made by intact cells. Microsomes from cwh43Δ or mcd4Δ mutants, which are unable to make ceramide-based anchors in vivo, do not incorporate [3H]DHS into anchors in vitro. Moreover, gup1Δ microsomes incorporate [3H]DHS into the same abnormal anchor lipids as gup1Δ cells synthesize in vivo. Thus, the in vitro assay of ceramide incorporation into GPI anchors faithfully reproduces the events that occur in mutant cells. Incorporation of [3H]DHS into GPI proteins is obsd. with microsomes alone, but the reaction is stimulated by cytosol or bovine serum albumin, ATP plus CoA, or C26:0-CoA, particularly if microsomes are depleted of acyl-CoA. Thus, [3H]DHS cannot be incorporated into proteins in the absence of acyl-CoA.
- 222Pinto, W. J.; Srinivasan, B.; Shepherd, S.; Schmidt, A.; Dickson, R. C.; Lester, R. L. J. Bacteriol. 1992, 174, 2565Google Scholar222https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38Xks1KnsLo%253D&md5=60ef304d3644e4395abd186a226e4b72Sphingolipid long-chain-base auxotrophs of Saccharomyces cerevisiae: genetics, physiology, and a method for their selectionPinto, William J.; Srinivasan, Bharath; Shepherd, Sherri; Schmidt, Ann; Dickson, Robert C.; Lester, Robert L.Journal of Bacteriology (1992), 174 (8), 2565-74CODEN: JOBAAY; ISSN:0021-9193.A selection method for sphingolipid long-chain-base auxotrophs of S. cerevisiae was devised after observing that strains that require a long-chain base for growth become denser when starved for this substance. Genetic anal. of >60 such strains indicated only 2 complementation classes, lcb1 and lcb2. Mutant strains from each class grew equally well with 3-ketodihydrophingosine, erythrodihydrosphingosine or threodihydrosphingosine, or phytosphingosine. Since these metabolites represent the first, second, and last components, resp., of the long-chain-base biosynthetic pathway, it is likely that the LCB1 and LCB2 genes are involved in the first step of long-chain-based synthesis. The results of long-chain-base starvation in the Lcb- strains suggest that ≥1 sphingolipids have a vital role in S. cerevisiae. Immediate sequelae of long-chain-base starvation were loss of viability, exacerbated in the presence of α-cyclodextrin, and loss of phosphoinositol sphingolipid synthesis but not phosphatidylinositol synthesis. Loss of viability with long-chain-base starvation could be prevented by also blocking either protein or nucleic acid synthesis. Without a long-chain-base, cell division, dry mass accumulation, and protein synthesis continued at a diminished rate and were further inhibited by the detergent Tergitol. The cell d. increase induced by long-chain-base starvation is thus explained as a differential loss of cell division and mass accumulation. Long-chain-base starvation in Lcb- S. cerevisiae and inositol starvation of Inos- S. cerevisiae share common features: an increase in cell d. and a loss of cell viability overcome by blocking macromol. synthesis.
- 223Hanada, K.; Nishijima, M.; Akamatsu, Y. J. Biol. Chem. 1990, 265, 22137Google ScholarThere is no corresponding record for this reference.
- 224Hojjati, M. R.; Li, Z.; Jiang, X. C. Biochim. Biophys. Acta 2005, 1737, 44Google ScholarThere is no corresponding record for this reference.
- 225Adachi-Yamada, T.; Gotoh, T.; Sugimura, I.; Tateno, M.; Nishida, Y.; Onuki, T.; Date, H. Mol. Cell. Biol. 1999, 19, 7276Google Scholar225https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXmtlaltr0%253D&md5=1ff9cb8ced368b1796fadb5d2248e0edDe novo synthesis of sphingolipids is required for cell survival by down-regulating c-Jun N-terminal kinase in Drosophila imaginal discsAdachi-Yamada, Takashi; Gotoh, Tomokazu; Sugimura, Isamu; Tateno, Minoru; Nishida, Yasuyoshi; Onuki, Tomoya; Date, HideyukiMolecular and Cellular Biology (1999), 19 (10), 7276-7286CODEN: MCEBD4; ISSN:0270-7306. (American Society for Microbiology)Mitogen-activated protein kinase (MAPK) is a conserved eukaryotic signaling factor that mediates various signals, cumulating in the activation of transcription factors. Extracellular signal-regulated kinase (ERK), a MAPK, is activated through phosphorylation by the kinase MAPK/ERK kinase (MEK). To elucidate the extent of the involvement of ERK in various aspects of animal development, we searched for a Drosophila mutant which responds to elevated MEK activity and herein identified a lace mutant. Mutants with mild lace alleles grow to become adults with multiple aberrant morphologies in the appendages, compd. eye, and bristles. These aberrations were suppressed by elevated MEK activity. Structural and transgenic analyses of the lace cDNA have revealed that the lace gene product is a membrane protein similar to the yeast protein LCB2, a subunit of serine palmitoyltransferase (SPT), which catalyzes the 1st step of sphingolipid biosynthesis. In fact, SPT activity in the fly expressing epitope-tagged Lace was absorbed by epitope-specific antibody. The no. of dead cells in various imaginal disks of a lace hypomorph was considerably increased, thereby ectopically activating c-Jun N-terminal kinase (JNK), another MAPK. These results account for the adult phenotypes of the lace mutant and suppression of the phenotypes by elevated MEK activity: we hypothesize that mutation of lace causes decreased de novo synthesis of sphingolipid metabolites, some of which are signaling mols., and ≥1 of these changes activates JNK to elicit apoptosis. The ERK pathway may be antagonistic to the JNK pathway in the control of cell survival.
- 226Nilsson, A. Biochim. Biophys. Acta 1968, 164, 575Google ScholarThere is no corresponding record for this reference.
- 227Nilsson, A. Biochim. Biophys. Acta 1969, 187, 113Google ScholarThere is no corresponding record for this reference.
- 228Schmelz, E. M.; Crall, K. J.; Larocque, R.; Dillehay, D. L.; Merrill, A. H., Jr. J. Nutr. 1994, 124, 702Google Scholar228https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXjt1WqtLo%253D&md5=30d164f5a58ea261be2cc425e244e2afUptake and metabolism of sphingolipids in isolated intestinal loops of miceSchmelz, Eva Maria; Crall, Kara J.; Larocque, Regina; Dillehay, Dirck L.; Merrill, Alfred H., Jr.Journal of Nutrition (1994), 124 (5), 702-12CODEN: JONUAI; ISSN:0022-3166.In this study, radiolabeled sphingolipids were placed in isolated intestinal segments of female CF1 mice, and the metab. and distribution of the radiolabel were followed. Most of the sphingomyelin was degraded to ceramide and other products in all regions of the intestine, and increasing amts. of several [3H]-labeled sphingolipids appeared in the tissues. The results establish that some of the sphingomyelin that enters the gastrointestinal tract is hydrolyzed and taken up by the intestine, with the lipid backbone being degraded or reutilized for complex sphingolipid synthesis; however, at least a portion passes into the large intestine. The appearance of bioactive compds. throughout the gastrointestinal tract may alter the behavior of intestinal cells.
- 229Nilsson, A.; Duan, R. D. J. Lipid Res. 2006, 47, 154Google ScholarThere is no corresponding record for this reference.
- 230Vesper, H.; Schmelz, E. M.; Nikolova-Karakashian, M. N.; Dillehay, D. L.; Lynch, D. V.; Merrill, A. H., Jr. J. Nutr. 1999, 129, 1239Google Scholar230https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXkt12nt7k%253D&md5=67f8c8e1674c27986dd701ade3fbdc15Sphingolipids in food and the emerging importance of sphingolipids to nutritionVesper, Hubert; Schmelz, Eva-Maria; Nikolova-Karakashian, Mariana N.; Dillehay, Dirck L.; Lynch, Daniel V.; Merrill, Alfred H., Jr.Journal of Nutrition (1999), 129 (7), 1239-1250CODEN: JONUAI; ISSN:0022-3166. (American Society for Nutritional Sciences)A review with 181 refs. Eukaryotes and some prokaryotes and viruses contain sphingolipids defined by common backbone sphingoid base, such as sphingosine. The sphingolipids of mammalian tissues, lipoproteins, and milk include ceramides, sphingomyelins, cerebrosides, gangliosides, and sulfatides; plants, fungi, and yeast have mainly cerebrosides and phosphoinositides. The total amts. of sphingolipids in foods vary considerably, from a few micromoles per kg in fruits to several millimoles per kg in dairy products, eggs, and soybeans. The per capita sphingolipid consumption in the US is estd. at 150-180 mmol (∼115-140 g) per yr, or 0.3-0.4 g/day. There is no known nutritional requirement for sphingolipids, but they are hydrolyzed in the gastrointestinal tract to the same metabolites (ceramides and sphingoid bases) that are used by cells to regulate growth, differentiation, apoptosis, and other cellular functions. Studies in exptl. animals have shown that feeding sphingolipids inhibits colon carcinogenesis, decreases blood serum LDL cholesterol and elevates HDL, suggesting that sphingolipids may be a functional food constituent. Sphingolipid metab. can also be modified by dietary constituents, such as cholesterol, fatty acids, and mycotoxins (fumonisins), with consequences for cell regulation and disease. Assocns. among diet, sphingolipids, and health may emerge as more is learned about these compds.
- 231Dillehay, D. L.; Webb, S. K.; Schmelz, E. M.; Merrill, A. H., Jr. J. Nutr. 1994, 124, 615Google Scholar231https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXivFOgurY%253D&md5=2540d8642369c1dc9a5311b0444148bfDietary sphingomyelin inhibits 1,2-dimethylhydrazine-induced colon cancer in CF1 miceDillehay, Dirck L.; Webb, Sonji K.; Schmelz, Eva Maria; Merrill, Alfred H., Jr.Journal of Nutrition (1994), 124 (5), 615-20CODEN: JONUAI; ISSN:0022-3166.Sphingolipids are in all eukaryotic cells and modulate cell growth, differentiation, and transformation; however, little is known about the physiol. effects of their consumption. Mice were fed diets supplemented with milk sphingomyelin to det. effects on colon carcinogenesis. Cancer was initiated in CF1 mice by 1,2-dimethylhydrazine. Mice were then fed AIN76A diets supplemented with 0.025-0.1 g sphingomyelin/100 g for 28 wk until the supply of sphingomyelin was depleted and then fed unsupplemented diet for 24 wk. Sphingomyelin did not affect wt. gain. Mice fed sphingomyelin had a 20% incidence of colon tumors compared with 47% in controls. Tumors were adenomas or adenocarcinomas and located in the distal third of the colon. In shorter-term studies, colonic epithelial cell proliferation was significantly greater than controls in mice fed 0.025 g sphingomyelin/100 g diet, but not in those fed higher amts. of sphingomyelin. The no. of aberrant crypts was significantly lower in 1,2-dimethylhydrazine-treated mice fed 0.05 g sphingomyelin/100 g diet than in controls. These results demonstrate that consumption of sphingomyelin affects the behavior of colonic cells. Because sphingolipids are present in food, the redn. in 1,2-dimethylhydrazine-induced premalignant lesions and the incidence of colon tumors in CF1 mice implies that these compds. may be another important class of nutritional modulators of carcinogenesis.
- 232Schmelz, E. M.; Roberts, P. C.; Kustin, E. M.; Lemonnier, L. A.; Sullards, M. C.; Dillehay, D. L.; Merrill, A. H., Jr. Cancer Res. 2001, 61, 6723Google ScholarThere is no corresponding record for this reference.
- 233Symolon, H.; Schmelz, E. M.; Dillehay, D. L.; Merrill, A. H., Jr. J. Nutr. 2004, 134, 1157Google Scholar233https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjvFGqu74%253D&md5=371f666153307628249ae69beaa3b9d4Dietary soy sphingolipids suppress tumorigenesis and gene expression in 1,2-dimethylhydrazine-treated CF1 mice and ApcMin/+ miceSymolon, Holly; Schmelz, Eva M.; Dillehay, Dirck L.; Merrill, Alfred H., Jr.Journal of Nutrition (2004), 134 (5), 1157-1161CODEN: JONUAI; ISSN:0022-3166. (American Society for Nutritional Sciences)Dietary supplementation with milk sphingolipids inhibits colon tumorigenesis in CF1 mice treated with a colon carcinogen [1,2-dimethylhydrazine (DMH)] and in multiple intestinal neoplasia (Min) mice, which develop intestinal tumors spontaneously. Plant sphingolipids differ structurally from those of mammals [soy glucosylceramide (GlcCer) consists predominantly of a 4,8-sphingadiene backbone and α-hydroxy-palmitic acid], which might affect their bioactivity. Soy GlcCer was added to the AIN-76A diet (which contains <0.005% sphingolipid) to investigate whether it would also suppress tumorigenesis in these mouse models. Soy GlcCer reduced colonic cell proliferation in the upper half of the crypts in mice treated with DMH by 50 and 56% (P < 0.05) at 0.025 and 0.1% of the diet (wt/wt), resp., and reduced the no. of aberrant colonic crypt foci (an early marker of colon carcinogenesis) by 38 and 52% (P < 0.05). Min mice fed diets contg. 0.025 and 0.1% (wt/wt) soy GlcCer developed 22 and 37% fewer adenomas (P < 0.05), resp. The effects of dietary sphingolipids on gene expression in the intestinal mucosal cells of Min mice were analyzed using Affymetrix GeneChip microarrays. Soy GlcCer affected the expression of 96 genes by ≥2-fold in a dose-dependent manner, increasing 32 and decreasing 64. Decreases in the mRNA expression of 2 transcription factors assocd. with cancer, hypoxia-induced factor 1α (HIF1α) and transcription factor 4 (TCF4), were confirmed by quant. RT-PCR. In conclusion, soy GlcCer suppressed colon tumorigenesis in 2 mouse models; hence, plant sphingolipids warrant further investigation as inhibitors of colon cancer. Because soy contains relatively high amts. of GlcCer, sphingolipids may partially account for the anticancer benefits attributed to soy-based foods.
- 234Inamine, M.; Suzui, M.; Morioka, T.; Kinjo, T.; Kaneshiro, T.; Sugishita, T.; Okada, T.; Yoshimi, N. Cancer Sci. 2005, 96, 876Google Scholar234https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XotFWiuw%253D%253D&md5=a5c525235b21a21b17458f2ab8b138d5Inhibitory effect of dietary monoglucosylceramide 1-O-β-glucosyl-N-2'-hydroxyarachidoyl-4,8-sphingadienine on two different categories of colon preneoplastic lesions induced by 1,2-dimethylhydrazine in F344 ratsInamine, Morihiko; Suzui, Masumi; Morioka, Takamitsu; Kinjo, Tatsuya; Kaneshiro, Tatsuya; Sugishita, Tomoko; Okada, Tadashi; Yoshimi, NaokiCancer Science (2005), 96 (12), 876-881CODEN: CSACCM; ISSN:1347-9032. (Blackwell Publishing Asia Pty Ltd.)Sphingolipids display a wide spectrum of biol. activities, including cell growth, differentiation and apoptosis. However, precise mechanisms by which these compds. exert anticancer or cancer-preventive effects are not known. In the present study, we evaluated the preventive efficacy of enriched dietary monoglucosylceramide 1-O-β-glucosyl-N-2'-hydroxyarachidoyl-4,8-sphingadienine (G1CM) on 1,2-dimethylhydrazine (DMH)-induced aberrant crypt foci (ACF) and β-catenin-accumulated crypt (BCAC) formation in F344 rats during initiation stage. We also examd. whether G1CM affects cell proliferation and apoptosis in these lesions. Pure G1CM was isolated from rice bran. Forty-two rats were divided randomly into five exptl. groups. Rats in groups 1-3 were given s.c. injections of DMH (40 mg/kg body wt.) once a week for 2 wk. One week before the first injection of DMH, rats in groups 2 and 3 were fed a diet contg. 200 and 1000 ppm G1CM, resp., for 5 wk. Rats in group 4 were fed a diet contg. 1000 ppm G1CM. Rats in group 5 were given the basal diet alone and served as untreated controls. The expt. was terminated 5 wk after the start. Dietary G1CM at both doses (groups 2 and 3) significantly inhibited the induction of ACF and BCAC (P < 0.001) when compared to group 1 treated with DMH alone. In groups 2 and 3, the proliferating cell nuclear antigen labeling indexes of epithelial cells in ACF and BCAC were also lower than in group 1 (P < 0.0001 for ACF, P < 0.05 for BCAC). These results, that dietary G1CM has possible chemopreventive effects in the present short-term colon carcinogenesis bioassays, suggest that longer exposure may cause suppression of tumor development.
- 235Simon, K. W.; Roberts, P. C.; Vespremi, M. J.; Manchen, S.; Schmelz, E. M. Mol. Nutr. Food Res. 2009, 53, 332Google Scholar235https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXktlSgsL0%253D&md5=aad48160df24c2dec6f178556997c982Regulation of β-catenin and connexin-43 expression: targets for sphingolipids in colon cancer preventionSimon, Kirk W.; Roberts, Paul C.; Vespremi, Michael J.; Manchen, Steven; Schmelz, Eva M.Molecular Nutrition & Food Research (2009), 53 (3), 332-340CODEN: MNFRCV; ISSN:1613-4125. (Wiley-VCH Verlag GmbH & Co. KGaA)Sphingolipid metabolites are generated throughout the intestinal tract after hydrolysis of orally administered complex sphingolipids and significantly suppress colon cancer in carcinogen-treated CF1 mice. In the present study, the mechanisms of tumor suppression by dietary sphingolipids were investigated. Changes in select genes that are crit. in early stages of colon cancer were analyzed in the colonic mucosa of dimethylhydrazine-treated CF1 mice fed AIN76A diet with or without 0.05% sphingomyelin (SM). Supplementation with SM did not significantly alter mRNA levels of most of the selected genes. However, a downregulation of β-catenin (p = 0.007) and increased protein levels of connexin-43 (p = 0.017) and Bcl-2 (p = 0.033) were obsd. in SM-fed animals. This suggests that sphingolipids may be regulating specific post-transcriptional events to reverse aberrant expression of individual proteins. Since the dysregulation of β-catenin metab. and its transcriptional activity in addn. to a decreased intercellular communication was causally linked to the development of colon cancer while a low Bcl-2 expression is assocd. with a worse prognosis in colon cancer, the reversal of these early changes may be important events in the prevention of colon cancer by orally administered sphingolipids, and may provide specific mol. biomarkers for sphingolipid efficacy in vivo.
- 236Mazzei, J. C.; Zhou, H.; Brayfield, B. P.; Hontecillas, R.; Bassaganya-Riera, J.; Schmelz, E. M. J Nutr Biochem 2011, DOI: 10.1016/j.jnutbio.2010.09.017Google ScholarThere is no corresponding record for this reference.
- 237Fujiwara, K.; Kitatani, K.; Fukushima, K.; Yazama, H.; Umehara, H.; Kikuchi, M.; Igarashi, Y.; Kitano, H.; Okazaki, T. Int. J. Clin. Oncol. 2011, 16, 133Google Scholar237https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXktlyqurw%253D&md5=fb333c5d2b6175da0105e79f7c8f8e9bInhibitory effects of dietary glucosylceramides on squamous cell carcinoma of the head and neck in NOD/SCID miceFujiwara, Kazunori; Kitatani, Kazuyuki; Fukushima, Kei; Yazama, Hiroaki; Umehara, Hisanori; Kikuchi, Mitsunori; Igarashi, Yasuyuki; Kitano, Hiroya; Okazaki, ToshiroInternational Journal of Clinical Oncology (2011), 16 (2), 133-140CODEN: IJCOF6; ISSN:1341-9625. (Springer Japan)Background: Sphingolipids, components of cellular membranes in eukaryotic cells, have roles in the regulation of tumor growth, inflammation, angiogenesis, and immunity. We investigated the effects of dietary glucosylceramides, sphingolipids isolated from rice bran, on tumor growth of human head and neck squamous cell carcinoma. Methods: The tumor cell line SCCKN cells isolated from well-differentiated human head and neck cancer were s.c. inoculated into the right flank of NOD/SCID mice, to establish an SCCKN xenograft model. Rice bran glucosylceramides (300 mg/kg/day) were administered orally to the mice for 14 consecutive days. Results: Dietary glucosylceramides significantly inhibited the growth of the xenograft tumor in comparison with the control group. The TUNEL stain revealed that treatment of mice with glucosylceramides increased the no. of apoptotic cells in the implanted tumor tissues and that apoptosis induction was accompanied by the formation of active/cleaved caspase-3. Conclusion: These results suggest that dietary glucosylceramides possibly exert anti-tumor activity by inducing apoptosis of head and neck squamous cell carcinoma. Therefore, their potential usefulness in treatment and prevention of human head and neck squamous cell carcinoma warrants further investigation.
- 238Carter, H. E.; Glick, F. J.; Norris, W. P.; Phillips, G. E. J. Biol. Chem. 1947, 170, 285Google ScholarThere is no corresponding record for this reference.
- 239Brady, R. O.; Koval, G. J. J. Biol. Chem. 1958, 233, 26Google ScholarThere is no corresponding record for this reference.
- 240Brady, R. O.; Formica, J. V.; Koval, G. J. J. Biol. Chem. 1958, 233, 1072Google ScholarThere is no corresponding record for this reference.
- 241Braun, P. E.; Snell, E. E. Proc. Natl. Acad. Sci. U.S.A. 1967, 58, 298Google ScholarThere is no corresponding record for this reference.
- 242Stoffel, W.; LeKim, D.; Sticht, G. Hoppe Seylers Z. Physiol. Chem. 1968, 349, 664Google ScholarThere is no corresponding record for this reference.
- 243Buede, R.; Rinker-Schaffer, C.; Pinto, W. J.; Lester, R. L.; Dickson, R. C. J. Bacteriol. 1991, 173, 4325Google Scholar243https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XhvVyrt7c%253D&md5=ae4cbafc3a9377075a71e8867ff64d47Cloning and characterization of LCB1, a Saccharomyces gene required for biosynthesis of the long-chain base component of sphingolipidsBuede, Rebecca; Rinker-Schaffer, Carrie; Pinto, William J.; Lester, Robert L.; Dickson, Robert C.Journal of Bacteriology (1991), 173 (14), 4325-32CODEN: JOBAAY; ISSN:0021-9193.The existence of auxotrophic mutants of S. cerevisiae having an abs. requirement for the long-chain base (lcb) component of sphingolipids suggests that sphingolipids are crucial for viability and growth. One mutant, termed the lcb1-1 mutant, lacks the activity of serine palmitoyltransferase, the first enzyme in the pathway for long-chain base synthesis. Here, an evidence is presented that LCB1 has been molecularly cloned. The size of the LCB1 transcript, the direction of transcription, the transcription initiation sites were detd. In addn., the coding region and its 5' and 3' flanking regions were sequenced. Anal. of the DNA sequence revealed a single open reading frame of 1674 nucleotides, encoding a predicted peptide of 558 amino acids. The hydropathy profile of the predicted peptide suggests a hydrophobic, globular, membrane-assocd. protein with two potential transmembrane helices. Comparison of the predicted amino acid sequence to known protein sequences revealed homol. to 5-aminolevulinic acid synthase and to 2-amino-3-ketobutyrate CoA ligase. These homologies, the similarity of the chem. reactions catalyzed by the 3 enzymes, and the finding that LCB1 restores serine palmitoyltransferase activity to an lcb1-defective strain indicate that serine palmitoyltransferase or a subunit of the enzyme is the most likely product of LCB1. Homol. of the LCB1 predicted protein to the Escherichia coli biotin synthetase was also obsd., but the biol. significance of this observation is not clear. A role for sphingolipid in sporulation is implicated by the finding that diploids homozygous for lch1 failed to sporulate.
- 244Nagiec, M. M.; Baltisberger, J. A.; Wells, G. B.; Lester, R. L.; Dickson, R. C. Proc. Natl. Acad. Sci. U.S.A. 1994, 91, 7899Google ScholarThere is no corresponding record for this reference.
- 245Weiss, B.; Stoffel, W. Eur. J. Biochem. 1997, 249, 239Google Scholar245https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXmvVKnurs%253D&md5=6cee22704c670b862dcc1fbaebe18f78Human and murine serine-palmitoyl-CoA transferase Cloning, expression and characterization of the key enzyme in sphingolipid synthesisWeiss, Bertram; Stoffel, WilhelmEuropean Journal of Biochemistry (1997), 249 (1), 239-247CODEN: EJBCAI; ISSN:0014-2956. (Springer)Serine palmitoyltransferase (SPT, EC 2.3.1.50) is the key enzyme in sphingolipid biosynthesis. It catalyzes the pyridoxal-5'-phosphate-dependent condensation of L-serine and palmitoyl-CoA to 3-oxosphinganine. Human expressed-sequence-tag (EST) clones are similar to the two yeast genes for synthesis of long-chain bases, LCB1 and LCB2, which are believed to encode two subunits of SPT [Buede, R., Pinto, W. J., Lester, R. L. & Dickson, R. C. (1991) J. Bacteriol. 173, 4325-5332; Nagiec, M. M., Baltisberger, J. A., Wells, G. B., Lester, R. L. & Dickson, R. C. (1994) Proc. Natl. Acad. Sci. USA 91, 7899-7902]. We have cloned and characterized two complete human and murine cDNA sequences named hLCB1 & mLCB1 and hLCB2 & mLCB2, resp., similar to the yeast LCB1 and LCB2 genes. Human embryonic kidney cells (HEK 293) transfected with murine sequences of LCB1 (mLCB1) and LCB2 (mLCB2) independently and in coexpression showed an overexpression of the transcripts on the mRNA and protein level. The enzymic activity of cells expressing mLCB2 alone or coexpressed with mLCB1 was three times higher than the activity of untransfected HEK cells. MLCB1 expression was not required for the synthesis of 3-oxo-sphinganine in mammalian cells. Transcription/translation in vitro yielded mLCB1 (53 kDa) and mLCB2 (63 kDa). The two proteins do not contain a signal peptide nor are they glycosylated. The endogenous and overexpressed SPT activity were both sensitive to common SPT inhibitors. Labeling studies with [1-14C]palmitic acid indicated that cell lines transfected with mLCB2 preferentially use the excess sphingoid bases for glucocerebroside and galactocerebroside synthesis. Our results provide conclusive genetic and biochem. evidence that the human and murine LCB2 genes described here encode serine palmitoyltransferase. Further studies will be required to unravel the function of the LCB1 gene in mammalian cells.
- 246Nagiec, M. M.; Lester, R. L.; Dickson, R. C. Gene 1996, 177, 237Google ScholarThere is no corresponding record for this reference.
- 247Hanada, K.; Hara, T.; Nishijima, M.; Kuge, O.; Dickson, R. C.; Nagiec, M. M. J. Biol. Chem. 1997, 272, 32108Google ScholarThere is no corresponding record for this reference.
- 248Williams, R. D.; Wang, E.; Merrill, A. H., Jr. Arch. Biochem. Biophys. 1984, 228, 282Google ScholarThere is no corresponding record for this reference.
- 249Hanada, K. Biochim. Biophys. Acta 2003, 1632, 16Google ScholarThere is no corresponding record for this reference.
- 250Hornemann, T.; Wei, Y.; von Eckardstein, A. Biochem. J. 2007, 405, 157Google ScholarThere is no corresponding record for this reference.
- 251Wei, J.; Yerokun, T.; Leipelt, M.; Haynes, C. A.; Radhakrishna, H.; Momin, A.; Kelly, S.; Park, H.; Wang, E.; Carton, J. M.; Uhlinger, D. J.; Merrill, A. H., Jr. Biochim. Biophys. Acta 2009, 1791, 746Google ScholarThere is no corresponding record for this reference.
- 252Carton, J. M.; Uhlinger, D. J.; Batheja, A. D.; Derian, C.; Ho, G.; Argenteri, D.; D’Andrea, M. R. J. Histochem. Cytochem. 2003, 51, 715Google Scholar252https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXktlWit7g%253D&md5=dcfb91140d57512c69ad794332136b9eEnhanced serine palmitoyltransferase expression in proliferating fibroblasts, transformed cell lines, and human tumorsCarton, Jill M.; Uhlinger, David J.; Batheja, Ameesha D.; Derian, Claudia; Ho, George; Argenteri, Dennis; D'Andrea, Michael R.Journal of Histochemistry and Cytochemistry (2003), 51 (6), 715-726CODEN: JHCYAS; ISSN:0022-1554. (Histochemical Society, Inc.)Metastatic processes, including cell invasion, extracellular matrix degrdn., and tissue remodeling, require cellular reorganization and proliferation. The cell signaling mols. required and the proteins involved in cell restructuring were not completely elucidated. We were studying the role of sphingolipids in normal cell activity and in several pathophysiol. states. In this study the authors used immunohistochem. to observe the presence of the 2 known subunits of serine palmitoyltransferase (SPT) in proliferating cells, in an in vitro model of wound repair, and in human malignant tissue. We report increased expression of the two subunits, SPT1 and SPT2, in the proliferating cells in these models. We also demonstrate a change in subcellular localization of the SPT subunits from predominantly cytosolic in quiescent cells to nuclear in proliferating cells. In addn., the authors obsd. SPT1 and SPT2 immunoreactivity in reactive stromal fibroblasts surrounding the carcinoma cells of some of the tumors. This enhanced SPT expression was absent in the stromal fibroblasts surrounding normal epithelial cells. These results suggest a potential role for overexpression of SPT in the processes of cell metastasis.
- 253Mandon, E. C.; Ehses, I.; Rother, J.; van Echten, G.; Sandhoff, K. J. Biol. Chem. 1992, 267, 11144– 8Google ScholarThere is no corresponding record for this reference.
- 254Yasuda, S.; Nishijima, M.; Hanada, K. J. Biol. Chem. 2003, 278, 4176Google ScholarThere is no corresponding record for this reference.
- 255Gable, K.; Slife, H.; Bacikova, D.; Monaghan, E.; Dunn, T. M. J. Biol. Chem. 2000, 275, 7597Google ScholarThere is no corresponding record for this reference.
- 256Hanada, K.; Hara, T.; Nishijima, M. J. Biol. Chem. 2000, 275, 8409Google ScholarThere is no corresponding record for this reference.
- 257Han, G.; Gupta, S. D.; Gable, K.; Niranjanakumari, S.; Moitra, P.; Eichler, F.; Brown, R. H., Jr.; Harmon, J. M.; Dunn, T. M. Proc. Natl. Acad. Sci. U.S.A. 2009, 106, 8186Google ScholarThere is no corresponding record for this reference.
- 258Inuzuka, M.; Hayakawa, M.; Ingi, T. J. Biol. Chem. 2005, 280, 35776Google ScholarThere is no corresponding record for this reference.
- 259Breslow, D. K.; Collins, S. R.; Bodenmiller, B.; Aebersold, R.; Simons, K.; Shevchenko, A.; Ejsing, C. S.; Weissman, J. S. Nature 2010, 463, 1048Google ScholarThere is no corresponding record for this reference.
- 260Han, S.; Lone, M. A.; Schneiter, R.; Chang, A. Proc. Natl. Acad. Sci. U.S.A. 2010, 107, 5851Google ScholarThere is no corresponding record for this reference.
- 261Gavin, A. C.; Bosche, M.; Krause, R.; Grandi, P.; Marzioch, M.; Bauer, A.; Schultz, J.; Rick, J. M.; Michon, A. M.; Cruciat, C. M.; Remor, M.; Hofert, C.; Schelder, M.; Brajenovic, M.; Ruffner, H.; Merino, A.; Klein, K.; Hudak, M.; Dickson, D.; Rudi, T.; Gnau, V.; Bauch, A.; Bastuck, S.; Huhse, B.; Leutwein, C.; Heurtier, M. A.; Copley, R. R.; Edelmann, A.; Querfurth, E.; Rybin, V.; Drewes, G.; Raida, M.; Bouwmeester, T.; Bork, P.; Seraphin, B.; Kuster, B.; Neubauer, G.; Superti-Furga, G. Nature 2002, 415, 141Google ScholarThere is no corresponding record for this reference.
- 262Giot, L.; Bader, J. S.; Brouwer, C.; Chaudhuri, A.; Kuang, B.; Li, Y.; Hao, Y. L.; Ooi, C. E.; Godwin, B.; Vitols, E.; Vijayadamodar, G.; Pochart, P.; Machineni, H.; Welsh, M.; Kong, Y.; Zerhusen, B.; Malcolm, R.; Varrone, Z.; Collis, A.; Minto, M.; Burgess, S.; McDaniel, L.; Stimpson, E.; Spriggs, F.; Williams, J.; Neurath, K.; Ioime, N.; Agee, M.; Voss, E.; Furtak, K.; Renzulli, R.; Aanensen, N.; Carrolla, S.; Bickelhaupt, E.; Lazovatsky, Y.; DaSilva, A.; Zhong, J.; Stanyon, C. A.; Finley, R. L., Jr.; White, K. P.; Braverman, M.; Jarvie, T.; Gold, S.; Leach, M.; Knight, J.; Shimkets, R. A.; McKenna, M. P.; Chant, J.; Rothberg, J. M. Science 2003, 302, 1727Google ScholarThere is no corresponding record for this reference.
- 263Yard, B. A.; Carter, L. G.; Johnson, K. A.; Overton, I. M.; Dorward, M.; Liu, H.; McMahon, S. A.; Oke, M.; Puech, D.; Barton, G. J.; Naismith, J. H.; Campopiano, D. J. J. Mol. Biol. 2007, 370, 870Google ScholarThere is no corresponding record for this reference.
- 264Raman, M. C.; Johnson, K. A.; Yard, B. A.; Lowther, J.; Carter, L. G.; Naismith, J. H.; Campopiano, D. J. J. Biol. Chem. 2009, 284, 17328Google ScholarThere is no corresponding record for this reference.