Structural Characteristics, Classification, and Nomenclature of Glycosphingolipids: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , , ,

Glycosphingolipids (GSLs) are a glycolipid subtype which plays vital roles in numerous biological processes, cell–cell interactions, as well as oncogenesis and ontogenesis. They are ubiquitous molecules found mostly in cell membranes.

  • glycosphingolipids (GSLs)
  • MALDI
  • mass spectrometry (MS)
  • healthy human tissues
  • pathological human tissues

1. Introduction

Glycosphingolipids (GSLs) represent essential lipid components embedded within the cell plasma membrane of mammalian tissues as well as that of invertebrates, plants, bacteria, and fungi. They constitute a glycolipid subtype which contains the amino alcohol (long-chain base or sphingoid base) sphingosine or its derivatives. GSLs can also be considered sphingolipids with an attached carbohydrate. Cell–cell interactions and cell adhesion make good use of the orientation of the hydrophilic oligosaccharide core, to the cell membrane exterior, while the hydrophobic moiety, the ceramide, positioned within the plasma membrane lipid bilayer, is made up of the sphingoid base replaced by a fatty acid at the amino group. The primary hydroxyl group is bound to the carbohydrate moiety [1][2][3][4][5].
GSLs are not distributed homogeneously within the plasma membrane, clustered together with cholesterol, sphingomyelin, and selected proteins (e.g., glycosylphosphatidylinositol (GPI)-anchored proteins, cell signaling transmembrane proteins such as the receptor tyrosine kinases (RTKs), endothelin receptors, Src family kinases, caveolin, flotillin, low molecular weight and heterotrimeric G proteins (Gα subunits), mitogen-activated protein (MAP)-kinase, protein-kinase C (PKC), Grb2, Shc-adaptor protein, p85 regulatory subunit of PI 3-kinase) [6][7][8][9][10][11][12] in lipid rafts, minute short-lived self-associating membrane molecule microdomains. Glycosyl epitopes in microdomains interact with various receptors and modulate lipid-raft-associated signaling events through molecular assemblies called glycosynapses, with GSLs acting as hydrogen bond donors and acceptors [13][14].
Thus, GSLs modulate biological functions by their interaction with the complementary molecules on other cell membranes (trans recognition) in order to communicate with nearby cells, or within the same cell membrane (cis recognition) with other components (proteins) of the membrane [4][15][16].
Each component of a GSL is involved in mediating various biological processes and physiological functions. While the ceramide functions as an anchor of the glycan headgroup and modulates its antigenic and assembling properties [17][18], the carbohydrate moiety acts as a receptor (for bacteria and viruses), an antigen (in several autoimmune diseases), is involved in protein interactions, receptor regulation, cell recognition, differentiation and adhesion, cell signaling, apoptosis, and formation of the myelin sheath [19][20][21][22]. Hence, GSLs participate in embryogenesis [19][21], brain development, synaptogenesis [23][24][25][26][27][28][29], antigenicity and immune response [30][31][32][33][34], kidney function [25][29], and hemostasis/thrombosis [29].
A wide range of literature data to date have revealed that changes in both moieties of GSL molecules can impact their normal biological functions and will result in abnormal GSL expression which is in close relation with numerous pathological conditions such as central nervous system-related/neurodegenerative diseases [35][36][37][38][39][40][41][42][43][44][45][46][47][48][49][50], cancer [41][42][43][44][45][46][47][48][49][50][51][52][53][54][55][56][57][58], infectious diseases [29][59], autoimmune diseases [60], GSL storage diseases [61][62], and diabetes mellitus [29].
Over time, the GSLs physiological roles have been studied using various biophysical, genetic, cell biology, and biochemical methods. However, the GSLs involvement in the etiopathogenesis of these diseases is not extensively known and, therefore, this represents a field with much promise for the future.
Mass spectrometry (MS) has had a great impact in the structural elucidation and quantitative profiling of GSLs within the recent past. Based on innovative MS-approaches coupled with chromatographic separation [63][64][65], intact GSLs can be investigated to provide at the same time the carbohydrate core sequence and the ceramide moiety composition. A first analytical challenge in investigating GSLs is represented by their structural complexity, which results in highly variable physicochemical properties. In addition, the quality of the laborious GSL extraction/purification procedures from biological matrices greatly influences their ionization (to remove the interference of other lipids, proteins, and nucleic acids). Another obstacle is the lack of commercially available standards and the reduced availability and quantity of normal and pathological tissue sample, making it difficult to achieve a quantitative, comprehensive, and large-scale profiling of GSLs in various tissues/organisms under normal or pathological conditions.
Among the soft ionization methods for mass spectrometric GLS analysis, matrix-assisted laser desorption ionization (MALDI) offers significant advantages, such as the simplicity and speed of the analysis; an adequate molecular mass lipids range, which is high enough that background matrix signal interferences are minimal; and the tolerance of higher quantities of impurities in comparison to other MS-based methods, reducing the necessity for sample purification, and offering high reproducibility [66]. Moreover, choosing the right matrix plays a pivotal analysis role, as a potential risk is represented by the post-source decay of GSLs, which is associated with noticeable decarboxylation and desialylation reactions, especially for sialic acid-containing GSLs [67][68].

2. Structural Characteristics, Classification, and Nomenclature of GSLs

Because of the existing variations in structure in both the ceramide and carbohydrate parts, GSLs are characterized by great complexity and diversity. The GSL glycan core expresses its specific species, while GSLs with different ceramide structures but the same glycan are considered different lipoforms (lipid forms) of the same species of GSL [69][70]. A single lipoform, homogeneous regarding the fatty acid, sphingoid base, and glycan, is considered equivalent to what is sometimes stated as a “molecular species” in GSL literature.
Every species of GSL can present a multitude of lipoforms, varied in the ceramide structure, such as the fatty acid chain length (C14 to C30 or greater, although the fatty acids most common in mammalian GSL ceramides are C18:0, C16:0, and C20:0), unsaturation degree, branching pattern, and hydroxylation possibility for both the fatty acid and the sphingoid base.
Removal from the ceramide moiety of the fatty acid residue, most commonly under physiological conditions using acid ceramidases, results in the formation of lyso-GSLs, which can be connected to various human diseases [71].
The basic chemical structure for the sphingoid base is represented by sphinganine or dihydrosphingosine, denoted as d18:0 (d stands for ‘di’ (two) hydroxyl groups at positions 1 and 3, 18 represents the number of carbon atoms, and 0 the C-C double bond number). In mammals, the most commonly encountered sphingoid base is, however, sphingosine (d18:1), possessing a double bond between C-4 and C-5, in addition to the structure of sphinganine. Phytosphingosine (t18:0) possesses an additional hydroxyl on C-4 while lacking the double bond.
In animals, ceramides which contain sphinganine and phytosphingosine are less abundant, while those which present phytosphingosine are found very widely in the GSLs of plants and fungi [1][72][73][74][75][76][77].
Other sphingoid bases containing a different number of C atoms, d20:0, d20:1, d16:0, d16:1, are also present in eukaryotes [65][78][79] and additional double bonds of carbon occur regularly at different positions in the hydrocarbon chain, generating a variety of sphingetrienes and sphingedienes. Several modifications of sphingosine, such as creating hydroxyl or oxo groups at different positions in the carbon backbone by the oxidation of carbons, or the addition of methylene or methyl groups to form rings or branches, are usually found within the tree of life’s low species [70].
Even though ceramide variations result in a substantial variety of GSL structures, most of the important structural and even functional classifications are owing to the structure of the carbohydrate core. The carbohydrate part can contain different types and numbers of monosaccharide residues, different linkages connecting them, or can be modified with various functional groups.
In vertebrates, the first monosaccharide attached to ceramide can be glucose (Glc-Cer) or galactose (Gal-Cer), but relatively reduced numbers of Gal-Cer-derived GSLs are found because, usually, extending the Gal-Cer glycan is constrained. The result is that the majority of mammalian GSLs are generated from Glc-Cer, while in invertebrates Gal-Cer derivatives are found.
GSLs have been divided into two classes on the basis of their glycans’ physicochemical properties: acidic GSLs and neutral (nonionic) GSLs. Acidic GSLs are mainly made up of two groups: the sialosyl-GSLs (which contain one or more than one sialic acid residue) or gangliosides and the sulfo-GSLs (which contain sulfate monoesters) or sulfatides [7]. In humans, only two forms of sialic acid usually exist, namely, N-glycolylneuraminic (Neu5Gc) and N-acetylneuraminic (Neu5Ac), the former only being present in trace amounts, originating either from diet [80][81], or produced by some malignant cells [82][83][84][85][86][87][88][89][90][91][92][93][94][95][96].
An unusual and rare form of sialic acid, deaminoneuraminic (KDN), and its glycoconjugates are only abundant in pathogenic bacteria and lower vertebrates; nevertheless, KDN was identified in some human tumors and in different animal organs, its presence being enhanced in hypoxic conditions [97][98][99].
GSLs are further classified according to the number, sequence, configuration, and linkages between the constituent monosaccharides as ganglio-, isoganglio-, globo-, isoglobo-, lacto-, neolacto-, lactoganglio-, muco-, gala-, neogala-, mollu-, arthro-, schisto-, and spirometo-series [100]. GSLs are divided into three key groups in vertebrates, containing the lacto-/neolacto-series, the globo-/isoglobo-series, and the ganglio-/isoganglio-series, and are expressed in tissue-specific patterns (Table 1). This diversity probably reveals important differences in the functions of GSLs. Conventionally, all sialylated GSLs are called “gangliosides” if they are derived not just from the ganglio-series, but also from the lacto- and globo-series of neutral GSLs [101][102]. In invertebrates, the GSLs found are in the mollu- and arthro-series (Table 1).
Table 1. GSL classification according to the core carbohydrate structure.
In addition to the high structural variety of GSLs as a result of the existent variations in their glycan and lipid portions, chemical modifications such as O-acetylation, fucosylation, lactonization [64][103][104], and the uncommon O-ketalation [105] may also occur in the structure of glycans.
In 1997, the IUPAC-IUB Joint Commission on Biochemical Nomenclature proposed a nomenclature for GSL based on a set of core structures which are pre-defined (characterizing the linkages between the monosaccharide components and the composition of the glycan) for serving as a base name [106] and includes all modifications within the glycan headgroup (Table 2).
Table 2. The Svennerholm system of GSL nomenclature and the IUPAC-IUB Joint Commission nomenclature.
An example is the name III2-α-Fuc-Gb3Cer (d18:1/18:0) for the GSL having the composition Fucα1-2Galα1-4Galβ1-4Glcβ1–1′Cer. In this example, the carbohydrate core structure Galα1-4Galβ1-4Glcβ1–1′ is denoted Gb3 (the base name). Since the GSL contains a monosaccharide extending beyond this core structure, it is used as prefix to this name, which contains (1) a Roman numeral indicating the monosaccharide which is modified, counting the monosaccharide which is closest to the ceramide moiety as “I” (in this case Fuc is attached to Gal at position III); (2) a superscript on the Roman numeral indicating which hydroxyl on that monosaccharide is modified (2 in the above example); (3) the configuration of the linkage (α); and (4) the abbreviated name of the attached monosaccharide unit (Fuc). While this terminology resolves confusion within the chemical literature, particularly when addressing numerous GSL isomers, it proves overly intricate for everyday application and lacks a framework for naming the lipid component.
The established approach for GSL naming, initially introduced by Svennerholm and still prevalent today [107][108], relies on their series designations along with the type, number, bonding arrangement, and position of sugar units within the glycan structure (Table 2). As an illustration, gangliosides are commonly abbreviated using a combination of two letters and a numeral, for instance, GM1, GD1, GT1, GQ. Here, G signifies the ganglio-series; the second letter (M, D, T, Q, P) indicates the count of sialic acid units (single, double, triple, quadruple, pentuple) found in the glycans; and the numeral (1, 2, 3, 4) corresponds to their elution sequence in thin-layer chromatography (TLC) (Rf values: GM4 > GM3 > GM2 > GM1), relative to the starting point. The latter property depends on the composition and length of the glycan, and the neutral core oligosaccharides were shown to move farther in TLC as the number increased. The indexes such as a, b, c show the molecular synthesis pathway, with the sialic acids binding position being different for all three pathways: e.g., in GM1b, the residue of sialic acid is linked to a Gal residue on the oligosaccharide chain end, while in the case of GM1a, the inner Gal residue is bound to the sialic acid.
Due to the varying degrees of molecular structural insight provided by diverse MS techniques, the Lipid MAPS consortium proposed an extensive classification system in 2005. They also created a comprehensive structural database encompassing biologically significant lipids, GSLs included. This database features entries for each GSL species, presenting both the systematic nomenclature and the commonly used name. It also incorporates information about the identifiable sphingoid base and N-linked fatty acids for each GSL species. A shorthand notation based on Liebisch et al. [109] is used for annotating MS-data-derived lipid structures at three levels: species level, molecular species level, and full structure level. When considering the ganglioside GM3 (d18:1/24:0), it is named according to this shorthand notation:
(1) NeuAcHex2Cer, 42:1;O2, at the species level (when the number of hydroxyl groups within the sphingoid base is uncertain, the total count of N-linked fatty acyl and sphingoid bases should be identified as the total carbon number; double bonds total; ceramide moiety oxygen atoms number; unidentified monosaccharide); (2) NeuAcHex2Cer, 18:1;O2/24:0, at the molecular species level (if the fatty acid and long-chain base structure are both known, with the exception of the double bond or stereochemistry and position); (3) NeuAc-Gal-Glc-Cer, 18:1(4E);3OH/24:0 (GM3), at the full structure level (if both stereochemistry or double bond and position and are identified). This widely acknowledged representation applies exclusively to the oligosaccharide headgroups comprising a maximum of two monosaccharide units; it does not encompass the more intricate glycosphingolipids (GSLs).

This entry is adapted from the peer-reviewed paper 10.3390/app13179922

References

  1. Schnaar, R.L.; Sandhoff, R.; Tiemeyer, M.; Kinoshita, T. Glycosphingolipids. In Essentials of Glycobiology, 4th ed.; Varki, A., Cummings, R.D., Esko, J.D., Eds.; Cold Spring Harbor Laboratory Press: New York, NY, USA, 2022; Chapter 11.
  2. Hakomori, S.I. Structure and Function of Sphingoglycolipids in Transmembrane Signalling and Cell-Cell Interactions. Biochem. Soc. Trans. 1993, 21, 583–595.
  3. Hakomori, S.I.; Igarashi, Y. Functional Role of Glycosphingolipids in Cell Recognition and Signaling. J. Biochem. 1995, 118, 1091–1103.
  4. Kojima, N.; Hakomori, S.I. Cell Adhesion, Spreading, and Motility of G(M3)-Expressing Cells Based on Glycolipid-Glycolipid Interaction. J. Biol. Chem. 1991, 266, 17552–17558.
  5. Hakomori, S. Structure and Function of Glycosphingolipids and Sphingolipids: Recollections and Future Trends. Biochim. Biophys. Acta-Gen. Subj. 2008, 1780, 325–346.
  6. Smart, E.J.; Graf, G.A.; Mcniven, M.A.; Sessa, W.C.; Engelman, J.A.; Scherer, P.E.; Okamoto, T.; Lisanti, M.P. Caveolins, Liquid-Ordered Domains, and Signal Transduction overview: Caveolae and caveola-related domains are liquid-ordered microdomains. Mol. Cell. Biol. 1999, 19, 7289–7304.
  7. Brown, D.A.; Rose, J.K. Sorting of GPI-Anchored Proteins to Glycolipid-Enriched Membrane Subdomains during Transport to the Apical Cell Surface. Cell 1992, 68, 533–544.
  8. Liu, P.; Ying, Y.; Ko, Y.G.; Anderson, R.G.W. Localization of Platelet-Derived Growth Factor-Stimulated Phosphorylation Cascade to Caveolae. J. Biol. Chem. 1996, 271, 10299–10303.
  9. Mineo, C.; James, G.L.; Smart, E.J.; Anderson, R.G.W. Localization of Epidermal Growth Factor-Stimulated Ras/Raf-1 Interaction to Caveolae Membrane. J. Biol. Chem. 1996, 271, 11930–11935.
  10. Chun, M.; Liyanage, U.K.; Lisanti, M.P.; Lodish, H.F. Signal Transduction of a G Protein-Coupled Receptor in Caveolae: Colocalization of Endothelin and Its Receptor with Caveolin. Proc. Natl. Acad. Sci. USA 1994, 91, 11728–11732.
  11. Gorodinsky, A.; Harris, D.A. Glycolipid-Anchored Proteins in Neuroblastoma Cells Form Detergent- Resistant Complexes without Caveolin. J. Cell Biol. 1995, 129, 619–627.
  12. Bickel, P.E.; Scherer, P.E.; Schnitzer, J.E.; Oh, P.; Lisanti, M.P.; Lodish, H.F. Flotillin and Epidermal Surface Antigen Define a New Family of Caveolae- Associated Integral Membrane Proteins. J. Biol. Chem. 1997, 272, 13793–13802.
  13. Hakomori, S.I. The Glycosynapse. Proc. Natl. Acad. Sci. USA 2002, 99, 225–232.
  14. Hakomori, S. Glycosynapses: Microdomains Controlling Carbohydrate-Dependent Cell Adhesion and Signaling. An. Acad. Bras. Cienc. 2004, 76, 553–572.
  15. Handa, K.; Hakomori, S.I. Carbohydrate to Carbohydrate Interaction in Development Process and Cancer Progression. Glycoconj. J. 2012, 29, 627–637.
  16. Santacroce, P.V.; Basu, A. Probing Specificity in Carbohydrate-Carbohydrate Interactions with Micelles and Langmuir Monolayers. Angew. Chemie-Int. Ed. 2003, 42, 95–98.
  17. Garcia-Ruiz, C.; Morales, A.; Fernández-Checa, J.C. Glycosphingolipids and Cell Death: One Aim, Many Ways. Apoptosis 2015, 20, 607–620.
  18. Iwabuchi, K.; Nakayama, H.; Oizumi, A.; Suga, Y.; Ogawa, H.; Takamori, K. Role of Ceramide from Glycosphingolipids and Its Metabolites in Immunological and Inflammatory Responses in Humans. Mediat. Inflamm. 2015, 2015, 120748.
  19. Lingwood, C.A. Glycosphingolipid Functions. Cold Spring Harb. Perspect. Biol. 2011, 3, a004788.
  20. Ravindran, M.S.; Tanner, L.B.; Wenk, M.R. Sialic Acid Linkage in Glycosphingolipids Is a Molecular Correlate for Trafficking and Delivery of Extracellular Cargo. Traffic 2013, 14, 1182–1191.
  21. Hakomori, S. Traveling for the Glycosphingolipid Path. Glycoconj. J. 2000, 17, 627–647.
  22. Frey, S.L.; Lee, K.Y.C. Number of Sialic Acid Residues in Ganglioside Headgroup Affects Interactions with Neighboring Lipids. Biophys. J. 2013, 105, 1421–1431.
  23. Ohmi, Y.; Ohkawa, Y.; Yamauchi, Y.; Tajima, O.; Furukawa, K.; Furukawa, K. Essential Roles of Gangliosides in the Formation and Maintenance of Membrane Microdomains in Brain Tissues. Neurochem. Res. 2012, 37, 1185–1191.
  24. Walkley, S.U.; Zervas, M.; Wiseman, S. Gangliosides as Modulators of Dendritogenesis in Normal and Storage Disease-Affected Pyramidal Neurons. Cereb. Cortex 2000, 10, 1028–1037.
  25. Schachner, M.; Bartsch, U. Multiple Functions of the Myelin-Associated Glycoprotein MAG (Siglec-4a) in Formation and Maintenance of Myelin. Glia 2000, 29, 154–165.
  26. Vyas, A.A.; Schnaar, R.L. Brain Gangliosides: Functional Ligands for Myelin Stability and the Control of Nerve Regeneration. Biochimie 2001, 83, 677–682.
  27. Hilbush, B.S.; Levine, J.M. Modulation of a Ca2+ Signaling Pathway by G(M1) Ganglioside in PC12 Cells. J. Biol. Chem. 1992, 267, 24789–24795.
  28. Kim, J.Y.H.; Goldenring, J.R.; DeLorenzo, R.J.; Yu, R.K. Gangliosides Inhibit Phospholipid-sensitive Ca2+-dependent Kinase Phosphorylation of Rat Myelin Basic Proteins. J. Neurosci. Res. 1986, 15, 159–166.
  29. Takahashi, T.; Suzuki, T. Role of Sulfatide in Normal and Pathological Cells and Tissues. J. Lipid Res. 2012, 53, 1437–1450.
  30. Porubsky, S.; Speak, A.O.; Salio, M.; Jennemann, R.; Bonrouhi, M.; Zafarulla, R.; Singh, Y.; Dyson, J.; Luckow, B.; Lehuen, A.; et al. Globosides but Not Isoglobosides Can Impact the Development of Invariant NKT Cells and Their Interaction with Dendritic Cells. J. Immunol. 2012, 189, 3007–3017.
  31. Nakayama, H.; Nagafuku, M.; Suzuki, A.; Iwabuchi, K.; Inokuchi, J.I. The Regulatory Roles of Glycosphingolipid-Enriched Lipid Rafts in Immune Systems. FEBS Lett. 2018, 592, 3921–3942.
  32. Zhang, T.; De Waard, A.A.; Wuhrer, M.; Spaapen, R.M. The Role of Glycosphingolipids in Immune Cell Functions. Front. Immunol. 2019, 10, 90.
  33. Haslam, S.M.; Julien, S.; Burchell, J.M.; Monk, C.R.; Ceroni, A.; Garden, O.A.; Dell, A. Characterizing the Glycome of the Mammalian Immune System. Immunol. Cell Biol. 2008, 86, 564–573.
  34. Nakayama, H.; Ogawa, H.; Takamori, K.; Iwabuchi, K. GSL-Enriched Membrane Microdomains in Innate Immune Responses. Arch. Immunol. Ther. Exp. 2013, 61, 217–228.
  35. Yu, R.K.; Ariga, T. The role of glycosphingolipids in neurological disorders. Mechanisms of immune action. Ann. N. Y. Acad. Sci. 1998, 845, 285–306.
  36. Dodge, J.C.; Treleaven, C.M.; Pacheco, J.; Cooper, S.; Bao, C.; Abraham, M.; Cromwell, M.; Sardi, S.P.; Chuang, W.L.; Sidman, R.L.; et al. Glycosphingolipids Are Modulators of Disease Pathogenesis in Amyotrophic Lateral Sclerosis. Proc. Natl. Acad. Sci. USA 2015, 112, 8100–8105.
  37. Schnaar, R.L. Gangliosides of the Vertebrate Nervous System. J. Mol. Biol. 2016, 428, 3325–3336.
  38. Han, X. Lipid Alterations in the Earliest Clinically Recognizable Stage of Alzheimers Disease: Implication of the Role of Lipids in the Pathogenesis of Alzheimers Disease. Curr. Alzheimer Res. 2005, 2, 65–77.
  39. Wallom, K.L.; Fernández-Suárez, M.E.; Priestman, D.A.; Vruchte, D.; Huebecker, M.; Hallett, P.J.; Isacson, O.; Platt, F.M. Glycosphingolipid Metabolism and Its Role in Ageing and Parkinson’s Disease. Glycoconj. J. 2022, 39, 39–53.
  40. Zöller, I.; Meixner, M.; Hartmann, D.; Büssow, H.; Meyer, R.; Gieselmann, V.; Eckhardt, M. Absence of 2-Hydroxylated Sphingolipids Is Compatible with Normal Neural Development but Causes Late-Onset Axon and Myelin Sheath Degeneration. J. Neurosci. 2008, 28, 9741–9754.
  41. Sasaki, N.; Toyoda, M.; Ishiwata, T. Gangliosides as Signaling Regulators in Cancer. Int. J. Mol. Sci. 2021, 22, 5076.
  42. Hakomori, S.I.; Zhang, Y. Glycosphingolipid Antigens and Cancer Therapy. Chem. Biol. 1997, 4, 97–104.
  43. Park, D.D.; Xu, G.; Wong, M.; Phoomak, C.; Liu, M.; Haigh, N.E.; Wongkham, S.; Yang, P.; Maverakis, E.; Lebrilla, C.B. Membrane Glycomics Reveal Heterogeneity and Quantitative Distribution of Cell Surface Sialylation. Chem. Sci. 2018, 9, 6271–6285.
  44. Bellis, S.L.; Reis, C.A.; Varki, A.; Kannagi, R.; Stanley, P. Glycosylation Changes in Cancer. In Essentials of Glycobiology, 4th ed.; Varki, A., Cummings, R.D., Esko, J.D., Eds.; Cold Spring Harbor Laboratory Press: New York, NY, USA, 2022; Chapter 47.
  45. Li, B.; Qin, Y.; Yu, X.; Xu, X.; Yu, W. Lipid Raft Involvement in Signal Transduction in Cancer Cell Survival, Cell Death and Metastasis. Cell Prolif. 2022, 55, e13167.
  46. Mirkin, B.L.; Clark, S.H.; Zhang, C. Inhibition of Human Neuroblastoma Cell Proliferation and EGF Receptor Phosphorylation by Gangliosides GM1, GM3, GD1A and GT1B. Cell Prolif. 2002, 35, 105–115.
  47. Furukawa, K.; Ohmi, Y.; Ohkawa, Y.; Bhuiyan, R.H.; Zhang, P.; Tajima, O.; Hashimoto, N.; Hamamura, K.; Furukawa, K. New Era of Research on Cancer-Associated Glycosphingolipids. Cancer Sci. 2019, 110, 1544–1551.
  48. Kovbasnjuk, O.; Mourtazina, R.; Baibakov, B.; Wang, T.; Elowsky, C.; Choti, M.A.; Kane, A.; Donowitz, M. The Glycosphingolipid Globotriaosylceramide in the Metastatic Transformation of Colon Cancer. Proc. Natl. Acad. Sci. USA 2005, 102, 19087–19092.
  49. Hamamura, K.; Furukawa, K.; Hayashi, T.; Hattori, T.; Nakano, J.; Nakashima, H.; Okuda, T.; Mizutani, H.; Hattori, H.; Ueda, M.; et al. Ganglioside GD3 Promotes Cell Growth and Invasion through P130Cas and Paxillin in Malignant Melanoma Cells. Proc. Natl. Acad. Sci. USA 2005, 102, 11041–11046.
  50. Yoshida, S.; Fukumoto, S.; Kawaguchi, H.; Sato, S.; Ueda, R.; Furukawa, K. Ganglioside GD2 in Small Cell Lung Cancer Cell Lines: Enhancement of Cell Proliferation and Mediation of Apoptosis. Cancer Res. 2001, 61, 4244–4252.
  51. Wang, L.; Wang, Y.; Sato, T.; Yamagata, S.; Yamagata, T. Ganglioside GD1a Suppresses TNFα Expression via Pkn1 at the Transcriptional Level in Mouse Osteosarcoma-Derived FBJ Cells. Biochem. Biophys. Res. Commun. 2008, 371, 230–235.
  52. Ohkawa, Y.; Miyazaki, S.; Miyata, M.; Hamamura, K.; Furukawa, K.; Furukawa, K. Essential Roles of Integrin-Mediated Signaling for the Enhancement of Malignant Properties of Melanomas Based on the Expression of GD3. Biochem. Biophys. Res. Commun. 2008, 373, 14–19.
  53. Shibuya, H.; Hamamura, K.; Hotta, H.; Matsumoto, Y.; Nishida, Y.; Hattori, H.; Furukawa, K.; Ueda, M.; Furukawa, K. Enhancement of Malignant Properties of Human Osteosarcoma Cells with Disialyl Gangliosides GD2/GD3. Cancer Sci. 2012, 103, 1656–1664.
  54. Hamamura, K.; Tsuji, M.; Hotta, H.; Ohkawa, Y.; Takahashi, M.; Shibuya, H.; Nakashima, H.; Yamauchi, Y.; Hashimoto, N.; Hattori, H.; et al. Functional Activation of Src Family Kinase Yes Protein Is Essential for the Enhanced Malignant Properties of Human Melanoma Cells Expressing Ganglioside GD3. J. Biol. Chem. 2011, 286, 18526–18537.
  55. Hyuga, S.; Yamagata, S.; Tai, T.; Yamagata, T. Inhibition of Highly Metastatic FBJ-LL Cell Migration by Ganglioside GD1a Highly Expressed in Poorly Metastatic FBJ-S1 Cells. Biochem. Biophys. Res. Commun. 1997, 231, 340–343.
  56. Cao, T.; Zhang, T.; Wang, L.; Zhang, L.; Adachi, T.; Sato, T.; Yamagata, S.; Yamagata, T. Ganglioside GD1a Suppression of NOS2 Expression via ERK1 Pathway in Mouse Osteosarcoma FBJ Cells. J. Cell. Biochem. 2010, 110, 1165–1174.
  57. Furukawa, K.; Hamamura, K.; Ohkawa, Y.; Ohmi, Y.; Furukawa, K. Disialyl Gangliosides Enhance Tumor Phenotypes with Differential Modalities. Glycoconj. J. 2012, 29, 579–584.
  58. Mitsuzuka, K.; Handa, K.; Satoh, M.; Arai, Y.; Hakomori, S. A Specific Microdomain (“glycosynapse 3”) Controls Phenotypic Conversion and Reversion of Bladder Cancer Cells through GM3-Mediated Interaction of A3β1 Integrin with CD9. J. Biol. Chem. 2005, 280, 35545–35553.
  59. Lewis, A.L.; Szymanski, C.M.; Schnaar, R.L.; Aebi, M. Bacterial and Viral Infections. In Essentials of Glycobiology, 4th ed.; Varki, A., Cummings, R.D., Esko, J.D., Eds.; Cold Spring Harbor Laboratory Press: New York, NY, USA, 2022; Chapter 42.
  60. Sackstein, R.; Hoffmeister, K.M.; Stowell, S.R.; Kinoshita, T.; Varki, A.; Freeze, H.H. Glycans in Acquired Human Diseases. In Essentials of Glycobiology, 4th ed.; Varki, A., Cummings, R.D., Esko, J.D., Eds.; Cold Spring Harbor Laboratory Press: New York, NY, USA, 2022; Chapter 46.
  61. Freeze, H.H.; Steet, R.; Suzuki, T.; Kinoshita, T.; Schnaar, R.L. Genetic Disorders of Glycan Degradation. In Essentials of Glycobiology, 4th ed.; Varki, A., Cummings, R.D., Esko, J.D., Eds.; Cold Spring Harbor Laboratory Press: New York, NY, USA, 2022; Chapter 44.
  62. Breiden, B.; Sandhoff, K. Lysosomal Glycosphingolipid Storage Diseases. Annu. Rev. Biochem. 2019, 88, 461–485.
  63. Rustam, Y.H.; Reid, G.E. Analytical Challenges and Recent Advances in Mass Spectrometry Based Lipidomics. Anal. Chem. 2018, 90, 374–397.
  64. Farwanah, H.; Kolter, T. Lipidomics of Glycosphingolipids. Metabolites 2012, 2, 134–164.
  65. Merrill, A.H.; Sullards, M.C.; Allegood, J.C.; Kelly, S.; Wang, E. Sphingolipidomics: High-Throughput, Structure-Specific, and Quantitative Analysis of Sphingolipids by Liquid Chromatography Tandem Mass Spectrometry. Methods 2005, 36, 207–224.
  66. Fuchs, B.; Schiller, J. MALDI-TOF MS, Analysis of Lipids from Cells, Tissues and Body Fluids. In Lipids in Health and Disease. Subcellular Biochemistry; Quinn, P.J., Wang, X., Eds.; Springer: Berlin/Heidelberg, Germany, 2008; Chapter 21; p. 49.
  67. Barrientos, R.C.; Zhang, Q. Recent Advances in the Mass Spectrometric Analysis of Glycosphingolipidome—A Review. Anal. Chim. Acta 2020, 1132, 134–155.
  68. Leopold, J.; Popkova, Y.; Engel, K.M.; Schiller, J. Recent Developments of Useful MALDI Matrices for the Mass Spectrometric Characterization of Lipids. Biomolecules 2018, 8, 173.
  69. Levery, S.B. Studies of the Primary and Secondary Structure of Glycosphingolipids. Ph.D. Thesis, University of Washington, Seattle, WA, USA, 1993.
  70. Guo, Z. The Structural Diversity of Natural Glycosphingolipids (GSLs). J. Carbohydr. Chem. 2022, 41, 63–154.
  71. Van Eijk, M.; Ferra, M.J.; Boot, R.G.; Aerts, J.M.F.G. Lyso-Glycosphingolipids: Presence and Consequences. Essays Biochem. 2020, 64, 565–578.
  72. Pruett, S.T.; Bushnev, A.; Hagedorn, K.; Adiga, M.; Haynes, C.A.; Sullards, M.C.; Liotta, D.C.; Merrill, A.H. Biodiversity of Sphingoid Bases (“sphingosines”) and Related Amino Alcohols. J. Lipid Res. 2008, 49, 1621–1639.
  73. Fernandes, C.M.; Goldman, G.H.; Del Poeta, M. Biological Roles Played by Sphingolipids in Dimorphic and Filamentous Fungi. mBio 2018, 9, e00642-18.
  74. Hannich, J.T.; Umebayashi, K.; Riezman, H. Distribution and Functions of Sterols and Sphingolipids. Cold Spring Harb. Perspect. Biol. 2011, 3, a004762.
  75. Ishibashi, Y.; Kohyama-Koganeya, A.; Hirabayashi, Y. New Insights on Glucosylated Lipids: Metabolism and Functions. Biochim. Biophys. Acta-Mol. Cell Biol. Lipids 2013, 1831, 1475–1485.
  76. Guimaratildes, L.L.; Toledo, M.S.; Ferreira, F.A.S.; Straus, A.H.; Takahashi, H.K. Structural Diversity and Biological Significance of Glycosphingolipids in Pathogenic and Opportunistic Fungi. Front. Cell. Infect. Microbiol. 2014, 4, 138.
  77. Gault, C.R.; Obeid, L.M.; Hannun, Y.A. An Overview of Sphingolipid Metabolism: From Synthesis to Breakdown. Adv. Exp. Med. Biol. 2010, 688, 1–23.
  78. Merrill, A.H. Sphingolipid and Glycosphingolipid Metabolic Pathways in the Era of Sphingolipidomics. Chem. Rev. 2011, 111, 6387–6422.
  79. Han, X. Lipidomics for Studying Metabolism. Nat. Rev. Endocrinol. 2016, 12, 668–679.
  80. de Haan, N.; Yang, S.; Cipollo, J.; Wuhrer, M. Glycomics Studies Using Sialic Acid Derivatization and Mass Spectrometry. Nat. Rev. Chem. 2020, 4, 229–242.
  81. Schnaar, R.L.; Gerardy-Schahn, R.; Hildebrandt, H. Sialic Acids in the Brain: Gangliosides and Polysialic Acid in Nervous System Development, Stability, Disease, and Regeneration. Physiol. Rev. 2014, 94, 461–518.
  82. Szlasa, W.; Zendran, I.; Zalesińska, A.; Tarek, M.; Kulbacka, J. Lipid Composition of the Cancer Cell Membrane. J. Bioenerg. Biomembr. 2020, 52, 321–342.
  83. Krengel, U.; Bousquet, P.A. Molecular Recognition of Gangliosides and Their Potential for Cancer Immunotherapies. Front. Immunol. 2014, 5, 352.
  84. Hayashi, N.; Chiba, H.; Kuronuma, K.; Go, S.; Hasegawa, Y.; Takahashi, M.; Gasa, S.; Watanabe, A.; Hasegawa, T.; Kuroki, Y.; et al. Detection of N-Glycolyated Gangliosides in Non-Small-Cell Lung Cancer Using GMR8 Monoclonal Antibody. Cancer Sci. 2013, 104, 43–47.
  85. Blanco, R.; Domínguez, E.; Morales, O.; Blanco, D.; Martínez, D.; Rengifo, C.E.; Viada, C.; Cedeño, M.; Rengifo, E.; Carr, A. Prognostic Significance of N-Glycolyl GM3 Ganglioside Expression in Non-Small Cell Lung Carcinoma Patients: New Evidences. Patholog. Res. Int. 2015, 2015, 132326.
  86. Blanco, R.; Rengifo, C.E.; Cedeño, M.; Frómeta, M.; Rengifo, E.; Carr, A. Immunoreactivity of the 14F7 Mab (Raised against N-Glycolyl GM3 Ganglioside) as a Positive Prognostic Factor in Non-Small-Cell Lung Cancer. Patholog. Res. Int. 2012, 2012, 235418.
  87. van Cruijsen, H.; Ruiz, M.; van der Valk, P.; de Gruijl, T.D.; Giaccone, G. Tissue Micro Array Analysis of Ganglioside N-Glycolyl GM3 Expression and Signal Transducer and Activator of Transcription (STAT)-3 Activation in Relation to Dendritic Cell Infiltration and Microvessel Density in Non-Small Cell Lung Cancer. BMC Cancer 2009, 9, 180.
  88. Oliva, J.P.; Valdés, Z.; Casacó, A.; Pimentel, G.; González, J.; Álvarez, I.; Osorio, M.; Velazco, M.; Figueroa, M.; Ortiz, R.; et al. Clinical Evidences of GM3 (NeuGc) Ganglioside Expression in Human Breast Cancer Using the 14F7 Monoclonal Antibody Labelled with 99mTc. Breast Cancer Res. Treat. 2006, 96, 115–121.
  89. Zhong, Y.; Wu, Y.; Li, C.; Tang, J.; Wang, X.; Ren, G.; Carr, A.; Pérez, R.; Guo, W. N-Glycolyl GM3 Ganglioside Immunoexpression in Oral Mucosal Melanomas of Chinese. Oral Dis. 2012, 18, 741–747.
  90. Blanco, R.; Quintana, Y.; Blanco, D.; Cedeño, M.; Rengifo, C.E.; Frómeta, M.; Ríos, M.; Rengifo, E.; Carr, A. Tissue Reactivity of the 14F7 Mab Raised against N-Glycolyl GM3 Ganglioside in Tumors of Neuroectodermal, Mesodermal, and Epithelial Origin. J. Biomarkers 2013, 2013, 602417.
  91. Blanco, R.; Rengifo, E.; Cedeño, M.; Rengifo, C.E.; Alonso, D.F.; Carr, A. Immunoreactivity of the 14F7 Mab Raised against N-Glycolyl GM3 Ganglioside in Epithelial Malignant Tumors from Digestive System. ISRN Gastroenterol. 2011, 2011, 645641.
  92. Blanco, R.; Cedeño, M.; Escobar, X.; Blanco, D.; Rengifo, C.E.; Frómeta, M.; Alvarez, R.I.; Rengifo, E.; Carr, A. Immunorecognition of the 14F7 Mab Raised against N-Glycolyl GM3 Ganglioside in Some Normal and Malignant Tissues from Genitourinary System. ISRN Pathol. 2011, 2011, 953803.
  93. Blanco, R.; Blanco, D.; Quintana, Y.; Escobar, X.; Rengifo, C.E.; Osorio, M.; Gutiérrez, Z.; Lamadrid, J.; Cedeño, M.; Frómeta, M.; et al. Immunoreactivity of the 14F7 Mab Raised against N-Glycolyl GM3 Ganglioside in Primary Lymphoid Tumors and Lymph Node Metastasis. Patholog. Res. Int. 2013, 2013, 920972.
  94. Scursoni, A.M.; Galluzzo, L.; Camarero, S.; Lopez, J.; Lubieniecki, F.; Sampor, C.; Segatori, V.I.; Gabri, M.R.; Alonso, D.F.; Chantada, G.; et al. Detection of N-Glycolyl GM3 Ganglioside in Neuroectodermal Tumors by Immunohistochemistry: An Attractive Vaccine Target for Aggressive Pediatric Cancer. Clin. Dev. Immunol. 2011, 2011, 245181.
  95. Sampor, C.; Guthmann, M.D.; Scursoni, A.; Cacciavillano, W.; Torbidoni, A.; Galluzzo, L.; Camarero, S.; Lopez, J.; de Dávila, M.T.G.; Fainboim, L.; et al. Immune Response to Racotumomab in a Child with Relapsed Neuroblastoma. Front. Oncol. 2012, 2, 195.
  96. Scursoni, A.M.; Galluzzo, L.; Camarero, S.; Pozzo, N.; Gabri, M.R.; De Acosta, C.M.; Vázquez, A.M.; Alonso, D.F.; De Davila, M.T.G. Detection and Characterization of N-Glycolyated Gangliosides in Wilms Tumor by Immunohistochemistry. Pediatr. Dev. Pathol. 2010, 13, 18–23.
  97. Sites, J.B.C.A. Carcinoma Cells: CHEMICAL MAMMALS Identification of 2-Keto-3-Deoxy-D-Glycero-D-Galacto Nononic Acid Acid) Residues in Mammalian Tissues and Human Lung. Histochemistry 1996, 549, 2–6.
  98. Go, S.; Sato, C.; Yin, J.; Kannagi, R.; Kitajima, K. Hypoxia-Enhanced Expression of Free Deaminoneuraminic Acid in Human Cancer Cells. Biochem. Biophys. Res. Commun. 2007, 357, 537–542.
  99. Inoue, S.; Lin, S.L.; Chang, T.; Wu, S.H.; Yao, C.W.; Chu, T.Y.; Troy, F.A.; Inoue, Y. Identification of Free Deaminated Sialic Acid (2-Keto-3-Deoxy-D-Glycero-D-Galacto-Nononic Acid) in Human Red Blood Cells and Its Elevated Expression in Fetal Cord Red Blood Cells and Ovarian Cancer Cells. J. Biol. Chem. 1998, 273, 27199–27204.
  100. Yu, R.K.; Tsai, Y.T.; Ariga, T.; Yanagisawa, M. Structures, Biosynthesis, and Functions of Gangliosides-an Overview. J. Oleo Sci. 2011, 60, 537–544.
  101. Hakomori, S. Structure, Organization, and Function of Glycosphingolipids in Membrane. Curr. Opin. Hematol. 2003, 10, 16–24.
  102. Hakomori, S.; Ishizuka, I. Glycolipid: Animal. In Encyclopedia of Life Sciences; Nature Publishing Group: London, UK, 2001.
  103. Levery, S.B.; Nudelman, E.D.; Hakomori, S. Novel Modification of Glycosphingolipids by Long-Chain Cyclic Acetals: Isolation and Characterization of Plasmalocerebroside from Human Brain. Biochemistry 1992, 31, 5335–5340.
  104. Kolter, T. Ganglioside Biochemistry. ISRN Biochem. 2012, 2012, 506160.
  105. Mlinac, K.; Fabris, D.; Vukelić, Ž.; Rožman, M.; Heffer, M.; Bognar, S.K. Structural Analysis of Brain Ganglioside Acetylation Patterns in Mice with Altered Ganglioside Biosynthesis. Carbohydr. Res. 2013, 382, 1–8.
  106. Chester, M.A. IUPAC-IUB Joint Commission on Biochemical Nomenclature (JCBN). Nomenclature of Glycolipids--Recommendations 1997. Eur. J. Biochem. 1998, 257, 293–298.
  107. Svennerholm, L. The Gangliosides. J. Lipid Res. 1964, 5, 145–155.
  108. Svennerholm, L. Chromatographic Separation of Human Brain Gangliosides. J. Neurochem. 1963, 10, 613–623.
  109. Liebisch, G.; Fahy, E.; Aoki, J.; Dennis, E.A.; Durand, T.; Ejsing, C.S.; Fedorova, M.; Feussner, I.; Griffiths, W.J.; Köfeler, H.; et al. Update on LIPID MAPS Classification, Nomenclature, and Shorthand Notation for MS-Derived Lipid Structures. J. Lipid Res. 2020, 61, 1539–1555.
More
This entry is offline, you can click here to edit this entry!
Video Production Service