Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Biologically active sphingolipids in cancer pathogenesis and treatment

Key Points

  • The sphingolipids ceramide and sphingosine-1-phosphate (S1P) function as effector molecules, and have important roles in stimulus/agonist-mediated signalling and the regulation of many cellular processes.

  • Ceramide, the central molecule in sphingolipid metabolism, mediates antiproliferative responses such as growth inhibition, apoptosis, differentiation, modulation of telomerase activity and telomere length, and senescence. Conversely, S1P induces proliferation, transformation, angiogenesis and cell motility.

  • A network of specialized and compartmentalized enzymes regulates the levels of ceramide and S1P. These enzymes serve as transducers that couple the actions of numerous extra- and intracellular signals to sphingolipid-mediated responses.

  • Attenuation of ceramide levels and/or increased levels of S1P are increasingly implicated in various stages of cancer pathogenesis, including an anti-apoptotic phenotype, metastasis and escape from senescence.

  • Small-molecule inhibitors of enzymes involved in ceramide clearance specifically induce ceramide accumulation and could present a novel therapeutic modality for the treatment of human cancers, including the reversal of drug resistance.

Abstract

Biologically active sphingolipids have key roles in the regulation of several fundamental biological processes that are integral to cancer pathogenesis. Recent significant progress in understanding biologically active sphingolipid synthesis, specifically within ceramide and sphingosine-1-phosphate (S1P)-mediated pathways, has identified crucial roles for these molecules both in cancer development and progression. Ceramide — a central molecule in sphingolipid metabolism — in effect functions as a tumour-suppressor lipid, inducing antiproliferative and apoptotic responses in various cancer cells. Conversely, S1P induces responses that, on aggregate, render S1P a tumour-promoting lipid. These discoveries are paving the way for the advancement of anticancer therapies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Pathways of sphingolipid metabolism.
Figure 2: Ceramide-regulated targets and pathways.
Figure 3: Targets and pathways regulated by S1P.
Figure 4: Compartmentalized pathways of ceramide signalling.
Figure 5: Roles of ceramide/sphingosine-1-phosphate in specific cancers.

Similar content being viewed by others

References

  1. Hannun, Y. A. Functions of ceramide in coordinating cellular response to stress. Science 274, 1855–1859 (1996).

    Article  CAS  PubMed  Google Scholar 

  2. Hannun, Y. A. & Obeid, L. M. The ceramide-centric universe of lipid-mediated cell regulation: stress encounters of the lipid kind. J. Biol. Chem. 277, 25847–25850 (2002).

    Article  CAS  PubMed  Google Scholar 

  3. Ogretmen, B. & Hannun, Y. A. Updates on functions of ceramide in chemotherapy-induced cell death and in multidrug resistance. Drug Resist. Updat. 4, 368–377 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Andrieu-Abadie, N. et al. Ceramide in apoptosis signaling: relationship with oxidative stress. Free Radic. Biol. Med. 31, 717–718 (2001).

    Article  CAS  PubMed  Google Scholar 

  5. Liu, Y. Y. et al. Ceramide glycosylation potentiates cellular multidrug resistance. FASEB J. 15, 719–730 (2001). Describes the role for glucosylceramide synthase, which attenuates cellular ceramide, in the development of drug resistance.

    Article  CAS  PubMed  Google Scholar 

  6. Chalfant, C. E. et al. The structural requirements for ceramide activation of serine-threonine protein phosphatases. J. Lipid Res. 45, 496–506 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Heinrich, M. et al. Cathepsin D links TNF-induced acid sphingomyelinase to Bid-mediated caspase-9 and–3 activation. Cell Death Differ. 11, 550–563 (2004). Describes the mechanism of action of lysosomal A-SMase-generated ceramide in mediating apoptosis.

    Article  CAS  PubMed  Google Scholar 

  8. Payne, S. G., Milstien, S. & Spiegel, S. Sphingosine-1-phosphate: dual messenger functions. FEBS Lett. 531, 54–57 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Hla, T. Signaling and biological actions of sphingosine 1-phosphate. Pharmacol. Res. 47, 401–407 (2003).

    Article  CAS  PubMed  Google Scholar 

  10. Maceyka, M. et al. Sphingosine kinase, sphingosine-1-phosphate, and apoptosis. Biochim. Biophys. Acta 1585, 193–201 (2002).

    Article  CAS  PubMed  Google Scholar 

  11. Pettus, B. J. et al. The sphingosine kinase 1/sphingosine-1-phosphate pathway mediates COX-2 induction and PGE2 production in response to TNF-α. FASEB J. 17, 1411–1421 (2003). Demonstrates a role for SK1/S1P in the activation of COX2 and PGE 2 production in response to TNFα.

    Article  CAS  PubMed  Google Scholar 

  12. van Meer, G. & Lisman, Q. Sphingolipid transport: rafts and translocators. J. Biol. Chem. 277, 25855–25858 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Senchenkov, A., Litvak, D. A. & Cabot, M. C. Trageting ceramide metabolism — a strategy for overcoming drug resistance. J. Natl Cancer Inst. 93, 347–357 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Riboni, L. et al. Ceramide levels are inversely associated with malignant progression of human glial tumors. Glia 39, 105–113 (2002). Demonstrates the clinical relevance of ceramide in human gliomas.

    Article  PubMed  Google Scholar 

  15. Rylova, S. N., Somova, O. G. & Dyatlovitskaya, E. V. Comparative investigation of sphingoid bases and fatty acids in ceramides and sphingomyelins from human ovarian malignant tumors and normal ovary. Biochemistry (Mosc.) 63, 1057–1060 (1998).

    CAS  PubMed  Google Scholar 

  16. French, K. F. et al. Discovery and evaluation of inhibitors of human sphingosine kinase. Cancer Res. 63, 5962–5969 (2003).

    CAS  PubMed  Google Scholar 

  17. Pettus, B. J., Chalfant, C. E. & Hannun, Y. A. Ceramide in apoptosis: an overview and current perspectives. Biochim. Biophys. Acta 1585, 114–125 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Kroesen, B. J. et al. Induction of apoptosis through B-cell receptor cross-linking occurs via de novo generated C16-ceramide and involves mitochondria. J. Biol. Chem. 276, 13606–13614 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Eto, M. et al. C16 ceramide accumulates following androgen ablation in LNCaP prostate cancer cells. Prostate 57, 66–79 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Bose, R. et al. Ceramide synthase mediates daunorubicin-induced apoptosis: an alternative mechanism for generating death signals. Cell 82, 405–414 (1995). Describes for the first time the involvement of de-novo -generated ceramide in chemotherapy-induced apoptosis.

    Article  CAS  PubMed  Google Scholar 

  21. Chalfant, C. E. et al. FAS activation induces dephosphorylation of SR proteins; dependence on the de novo generation of ceramide and activation of protein phosphatase 1. J. Biol. Chem. 276, 44848–44855 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Perry, D. K. et al. Serine palmitoyltransferase regulates de novo ceramide generation during etoposide-induced apoptosis. J. Biol. Chem. 275, 9078–9084 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Chalfant, C. E. et al. De novo ceramide regulates the alternative splicing of caspase 9 and Bcl-x in A549 lung adenocarcinoma cells. Dependence on protein phosphatase-1. J. Biol. Chem. 277, 12587–12595 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Wang, H. et al. N-(4-hydroxyphenyl)retinamide elevates ceramide in neuroblastoma cell lines by coordinate activation of serine palmitoyltransferase and ceramide synthase. Cancer Res. 61, 5102–5105 (2001).

    CAS  PubMed  Google Scholar 

  25. del Pulgar, T. et al. De novo-synthesized ceramide is involved in cannabinoid-induced apoptosis. Biochem J. 363, 183–188 (2002).

    Article  Google Scholar 

  26. Sullards, M. C. Analysis of sphingomyelin, glucosylceramide, ceramide, sphingosine, and sphingosine 1-phosphate by tandem mass spectrometry. Methods Enzymol. 312, 32–45 (2000).

    Article  CAS  PubMed  Google Scholar 

  27. Santana, P. et al. Acid sphingomyelinase-deficient human lymphoblasts and mice are defective in radiation-induced apoptosis. Cell 86, 189–199 (1996).

    Article  CAS  PubMed  Google Scholar 

  28. Zhang, Y. et al. Involvement of the acid sphingomyelinase pathway in uva-induced apoptosis. J. Biol. Chem. 276, 11775–11782 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Luberto, C. et al. Inhibition of tumor-necrosis factor-induced cell death in MCF7 by a novel inhibitor of neutral sphingomyelinase. J. Biol. Chem. 277, 41128–41139 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Lee, J. T. et al. Amyloid-β-peptide induces oligodendrocyte death by activating the neutral sphingomyelinase-ceramide pathway. J. Cell Biol. 164, 123–131 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Liu, J. J. et al. Activation of neutral sphingomyelinase participates in ethanol-induced apoptosis in Hep G2 cells. Alcohol Alcohol. 35, 569–573 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Liu, B. et al. Glutathione regulation of neutral sphingomyelinase in tumor necrosis factor-α-induced cell death. J. Biol. Chem. 273, 11313–11320 (1998).

    Article  CAS  PubMed  Google Scholar 

  33. Segui, B. et al. Involvement of FAN in TNF-induced apoptosis. J. Clin. Invest. 108, 143–151 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Testai, R. D., Landek, M. A. & Dawson, G. Regulation of sphingomyelinases in cells of the oligodendrocyte lineage. J. Neurosci. Res. 75, 66–74 (2004).

    Article  CAS  PubMed  Google Scholar 

  35. Franzen, R. et al. Nitric oxide induces degradation of the neutral ceramidase in rat renal mesangial cells and is counterreguated by protein kinase C. J. Biol. Chem. 277, 46184–46190 (2002).

    Article  CAS  PubMed  Google Scholar 

  36. Acharya, U. et al. Modulating sphingolipid biosynthetic pathway rescues photoreceptor degeneration. Science 299, 1740–1743 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Dbaibo, G. S. et al. Ceramide generation by two distinct pathways in tumor necrosis factor α-induced cell death. FEBS Lett. 503, 7–12 (2001).

    Article  CAS  PubMed  Google Scholar 

  38. Watanabe, M. et al. Increase of nuclear ceramide through caspase-3-dependent regulation of the “sphingomyelin cycle” in Fas-induced apoptosis. Cancer Res. 64, 1000–1007 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Cremesti, A. et al. Ceramide enables fas to cap and kill. J. Biol. Chem. 276, 23954–23961 (2001).

    Article  CAS  PubMed  Google Scholar 

  40. Dbaibo, G. S. et al. Retinoblastoma gene product as a downstream target for a ceramide-dependent pathway of growth arrest. Proc. Natl Acad. Sci. USA 92, 1347–1351 (1995). Defines an RB-dependent mechanism by which ceramide induces growth arrest.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lee, J. Y., Bielawska, A. E. & Obeid, L. M. Regulation of cyclin-dependent kinase 2 activity by ceramide. Exp. Cell Res. 261, 303–311 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Zhu, X. F. et al. Ceramide induces cell cycle arrest and upregulates p27kip in nasopharyngeal carcinoma cells. Cancer Lett. 193, 149–154 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Marchesini, N., Luberto, C. & Hannun, Y. A. Biochemical properties of mammalian neutral sphingomyelinase 2 and its role in sphingolipid metabolism. J. Biol. Chem. 278, 13775–13783 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Marchesini, N. et al. Role for neutral sphingomyelinase 2 in confluence-induced growth arrest of MCF-7 cells. J. Biol. Chem. 279, 25101–25111 (2004).

    Article  CAS  PubMed  Google Scholar 

  45. Venable, M. E. et al. Role of ceramide in cellular senescence. J. Biol. Chem. 270, 30701–30708 (1995). Show for the first time the role for ceramide in cellular senescence.

    Article  CAS  PubMed  Google Scholar 

  46. Guillas, I. et al. C26-CoA-dependent ceramide synthesis of Saccharomyces cerevisiae is operated by Lag1p and Lac1p. EMBO J. 20, 2655–2665 (2001). Demonstrates that LAG1 and its homologue LAC1, which have important roles in lifespan, are key components of ceramide synthase.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Schorling, S. et al. Lag1p and Lac1p are essential for the Acyl-CoA-dependent ceramide synthase reaction in Saccharomyces cerevisae. Mol. Biol. Cell. 12, 3417–3427 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Venkataraman, K. et al. Upstream of growth and differentiation factor 1 (uog1), a mammalian homolog of the yeast longevity assurance gene 1 (LAG1), regulates N-Stearoyl-sphinganine (C18-(dihydro)ceramide) synthesis in a fumonisin B1-independent manner in mammalian cells. J. Biol. Chem. 277, 35642–35649 (2002).

    Article  CAS  PubMed  Google Scholar 

  49. Obeid, L. M. & Hannun, Y. A. Ceramide, stress, and a “LAG” in aging. Sci. Aging Knowledge Environ. 2003, PE27 (2003).

    Article  PubMed  Google Scholar 

  50. Ogretmen, B. et al. Role of ceramide in mediating the inhibition of telomerase activity in A549 human lung adenocarcinoma cells. J. Biol. Chem. 276, 24901–24910 (2001). This study, together with references 52 and 53, shows for the first time that ceramide is one of the upstream regulators of telomerase and telomere length.

    Article  CAS  PubMed  Google Scholar 

  51. Kraveka, J. M. et al. Involvement of endogenous ceramide in the inhibition of telomerase activity and induction of morphologic differentiation in response to all-trans-retinoic acid in human neuroblastoma cells. Arch. Biochem. Biophys. 419, 110–119 (2003).

    Article  CAS  PubMed  Google Scholar 

  52. Ogretmen, B. et al. Molecular mechanisms of ceramide-mediated telomerase inhibition in the A549 human lung adenocarcinoma cell line. J. Biol. Chem. 276, 32506–32514 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Sundararaj, K. P. et al. Rapid shortening of telomere length in response to ceramide involves the inhibition of telomere binding activity of nuclear glyceraldehyde-3-phosphate dehydrogenase. J. Biol. Chem. 279, 6152–6162 (2004). Demonstrates a novel role for nuclear glyceraldehyde-3-phosphate dehydrogenase in telomere binding and protection.

    Article  CAS  PubMed  Google Scholar 

  54. Xia, P. et al. An oncogenic role of sphingosine kinase. Curr. Biol. 10, 1527–1530 (2000). Describes for the first time a tumour-promoting role for SK1.

    Article  CAS  PubMed  Google Scholar 

  55. Cuvillier, O. et al. Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature 381, 800–803 (1996). Shows a specific role of S1P in the inhibition of apoptosis.

    Article  CAS  PubMed  Google Scholar 

  56. Olivera, A. et al. Sphingosine kinase type 1 induces G12/13-mediated stress fiber formation, yet promotes growth and survival independent of G protein-coupled receptors. J. Biol. Chem. 278, 46452–46460 (2003).

    Article  CAS  PubMed  Google Scholar 

  57. Van Brocklyn, J. R., Young, N. & Roof, R. Sphingosine-1-phosphate stimulates motility and invasiveness of human glioblastoma multiforme cells. Cancer Lett. 199, 53–60 (2003).

    Article  CAS  PubMed  Google Scholar 

  58. Nava, V. E. et al. Sphingosine kinase type 1 promotes estrogen-dependent tumorigenesis of breast cancer MCF-7 cells. Exp. Cell Res. 281, 115–127 (2002).

    Article  CAS  PubMed  Google Scholar 

  59. Johnson, K. R. et al. Role of human sphingosine-1-phosphate phosphatase 1 in the regulation of intra- and extracellular sphingosine-1-phosphate levels and cell viability. J. Biol. Chem. 278, 34541–34547 (2003).

    Article  CAS  PubMed  Google Scholar 

  60. Liu, F. et al. Differential regulation of sphingosine-1-phosphate- and VEGF-induced endothelial cell chemotaxis. Involvement of G(iα2)-linked Rho kinase activity. Am. J. Respir. Cell Mol. Biol. 24, 711–719 (2001).

    Article  CAS  PubMed  Google Scholar 

  61. Wu, W. et al. VEGF receptor expression and signaling in human bladder tumors. Oncogene 22, 3361–3370 (2003).

    Article  CAS  PubMed  Google Scholar 

  62. Licht, T. et al. Induction of pro-angiogenic signaling by a synthetic peptide derived from the second intracellular loop of S1P3 (EDG3). Blood 102, 2099–2107 (2003).

    Article  CAS  PubMed  Google Scholar 

  63. Kluk, M. J. & Hla, T. Signaling of sphingosine-1-phosphate via the S1P/EDG-family of G-protein-coupled receptors. Biochim. Biophys. Acta 1582, 72–80 (2002).

    Article  CAS  PubMed  Google Scholar 

  64. Okamoto, H. et al. Inhibitory regulation of Rac activation, membrane ruffling, and cell migration by the G protein-coupled sphingosine-1-phosphate receptor EDG5 but not EDG1 or EDG3. Mol. Cell Biol. 20, 9247–9261 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Liu, Y. et al. Edg-1, the G-protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation. J. Clin. Invest. 106, 951–961 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Xia, P. et al. Activation of sphingosine kinase by tumor necrosis factor-α inhibits apoptosis in human endothelial cells. J. Biol. Chem. 274, 34499–34505 (1999).

    Article  CAS  PubMed  Google Scholar 

  67. Xia, P. et al. Tumor necrosis factor-α induces adhesion molecule expression through the sphingosine kinase pathway. Proc. Natl Acad. Sci. USA 95, 14196–14201 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Xia, P. et al. Sphingosine kinase interacts with TRAF2 and dissects tumor necrosis factor-α signaling. J. Biol. Chem. 277, 7996–8003 (2002).

    Article  CAS  PubMed  Google Scholar 

  69. Perry, D. K. & Kolesnick, R. N. Ceramide and sphingosine 1-phosphate in anti-cancer therapies. Cancer Treat. Res. 115, 345–354 (2003).

    Article  CAS  PubMed  Google Scholar 

  70. Radin, N. S. Killing tumours by ceramide-induced apoptosis: a critique of available drugs. Biochem. J. 371, 243–256 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Chauvier, D., Morjani, H. & Manfait, M. Ceramide involvement in homocamptothecin- and camptothecin-induced cytotoxicity and apoptosis in colon HT29 cells. Int. J. Oncol. 20, 855–863 (2002).

    CAS  PubMed  Google Scholar 

  72. Biswal, S. S. et al. Changes in ceramide and sphingomyelin following fludarabine treatment of human chronic B-cell leukemia cells. Toxicology 154, 45–53 (2000).

    Article  CAS  PubMed  Google Scholar 

  73. Strum, J. C. et al. 1-β-D-Arabinofuranosylcytosine stimulates ceramide and digylceride formation in HL-60 cells. J. Biol. Chem. 269, 15493–15497 (1994).

    Article  CAS  PubMed  Google Scholar 

  74. Bezombes, C. et al. Oxidative stress-induced activation of Lyn recruits sphingomyelinase and is requisite for its stimulation by Ara-C. FASEB J. 15, 1583–1585 (2001).

    Article  CAS  PubMed  Google Scholar 

  75. Sawada, M. et al. Influence of bax or bcl-2 overexpression on the ceramide-dependent apoptotic pathway in glioma cells. Oncogene 19, 3508–3520 (2000).

    Article  CAS  PubMed  Google Scholar 

  76. Dbaibo, G. S. et al. p53-dependent ceramide response to genotoxic stress. J. Clin. Invest. 102, 329–339 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Garcia-Barros, M. et al. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science 300, 1155–1159 (2003).

    Article  CAS  PubMed  Google Scholar 

  78. Bleicher, R. J. & Cabot, M. C. Glucosylceramide synthase and apoptosis. Biochim. Biophys. Acta 1585, 172–178 (2002).

    Article  CAS  PubMed  Google Scholar 

  79. Liu, Y. Y. et al. Ceramide glycosylation potentiates cellular multidrug resistance. FASEB J. 15, 719–730 (2001).

    Article  CAS  PubMed  Google Scholar 

  80. Liu, Y. Y. et al. Oligonucleotides blocking glucosylceramide synthase expression selectively reverse drug resistance in cancer cells. J. Lipid Res. 45, 933–940 (2004).

    Article  CAS  PubMed  Google Scholar 

  81. Goulding, C. W., Giuliano, A. E. & Cabot, M. C. SDZ PSC 833, the drug resistance modulator, activates cellular ceramide formation by a pathway independent of P-glycoprotein. Cancer Lett. 149, 143–151 (2000).

    Article  CAS  PubMed  Google Scholar 

  82. Sietsma, H., Veldman, R. J. & Kok, J. W. The involvement of sphingolipids in multidrug resistance. J. Membr. Biol. 181, 153–162 (2001).

    Article  CAS  PubMed  Google Scholar 

  83. De Rosa, M. F. et al. Role of multiple drug resistance protein 1 in neutral but not acidic glycosphingolipid biosynthesis. J. Biol. Chem. 279, 7867–7876 (2004). Demonstrates a functional role for P-gp in the translocation of glucosylceramide across the Golgi membrane.

    Article  CAS  PubMed  Google Scholar 

  84. Lavie, Y. et al. Accumulation of glucosylceramides in multidrug-resistant cancer cells. J. Biol. Chem. 271, 19530–19536 (1996).

    Article  CAS  PubMed  Google Scholar 

  85. Veldman, R. J. et al. Altered sphingolipid metabolism in multidrug-resistant ovarian cancer cells is due to uncoupling of glycolipid biosynthesis in the Golgi apparatus. FASEB J. 16, 1111–1113 (2002).

    Article  CAS  PubMed  Google Scholar 

  86. Weiss, M. et al. Inhibition of melanoma tumor growth by a novel inhibitor of glucosylceramide synthase. Cancer Res. 63, 3654–3658 (2003).

    CAS  PubMed  Google Scholar 

  87. Maurer, B. J. et al. Synergistic cytotoxicity in solid tumor cell lines between N-(4-hydroxyphenyl)retinamide and modulators of ceramide metabolism. J. Natl Cancer Inst. 92, 1897–1909 (2000).

    Article  CAS  PubMed  Google Scholar 

  88. Maurer, B. J. et al. Increase of ceramide and induction of mixed apoptosis/necrosis by N-(4-hydroxyphenyl)-retinamide in neuroblastoma cell lines. J. Natl Cancer Inst. 91, 1138–1146 (1999).

    Article  CAS  PubMed  Google Scholar 

  89. Tepper, A. D. et al. Glucosylceramide synthase does not attenuate the ceramide pool accumulating during apoptosis induced by CD95 or anti-cancer regimens. J. Biol. Chem. 275, 34810–34817 (2000).

    Article  CAS  PubMed  Google Scholar 

  90. Veldman, R. J. et al. The absence of functional glucosylceramide synthase does not sensitize melanoma cells for anticancer drugs. FASEB J. 17, 1144–1146 (2003).

    Article  CAS  PubMed  Google Scholar 

  91. Selzner, M. et al. Induction of apoptotic cell death and prevention of tumor growth by ceramide analogues in metastatic human colon cancer. Cancer Res. 61, 1233–1240 (2001).

    CAS  PubMed  Google Scholar 

  92. Samsel, L. et al. The ceramide analog, B13, induces apoptosis in prostate cancer cell lines and inhibits tumor growth in prostate cancer xenografts. Prostate 58, 382–393 (2004).

    Article  CAS  PubMed  Google Scholar 

  93. Wieder, T., Orfanos, C. E. & Geilen, C. C. Inducton of ceramide-mediated apoptosis by the anticancer phospholipid analog, hexadecylphosphocholine. J. Biol. Chem. 273, 11025–11031 (1998).

    Article  CAS  PubMed  Google Scholar 

  94. Meng, A. et al. Sphingomyelin synthase as a potential target for D609-induced apoptosis in U937 human monocytic leukemia cells. Exp. Cell Res. 292, 385–392 (2004).

    Article  CAS  PubMed  Google Scholar 

  95. Bieberich, E., Kawaguchi, T. & Yu, R. K. N-acylated serinol is a novel ceramide mimic inducing apoptosis in neuroblastoma cells. J. Biol. Chem. 275, 177–181 (2000).

    Article  CAS  PubMed  Google Scholar 

  96. Bieberich, E. et al. Synthesis and characterization of novel ceramide analogs for induction of apoptosis in human cancer cells. Cancer Lett. 181, 55–64 (2002).

    Article  CAS  PubMed  Google Scholar 

  97. Struckhoff, A. P. et al. Novel ceramide analogs as potential chemothrapeutic agents in breast cancer. J. Pharmacol. Exp. Ther. 309, 523–532 (2004).

    Article  CAS  PubMed  Google Scholar 

  98. Crawford, K. W. et al. Novel ceramide analogs display selective cytotoxicity in drug-resistant breast tumor cell lines compared to normal breast epithelial cells. Cell. Mol. Biol. 49, 1017–1023 (2003).

    CAS  PubMed  Google Scholar 

  99. Stover, T. & Kester, M. Liposomal delivery enhances short-chain ceramide-induced apoptosis of breast cancer cells. J. Pharmacol. Exp. Ther. 307, 468–475 (2003).

    Article  CAS  PubMed  Google Scholar 

  100. Shabbits, J. A. & Mayer, L. D. High ceramide content liposomes with in vivo antitumor activity. Anticancer Res. 23, 3663–3669 (2003)

    CAS  PubMed  Google Scholar 

  101. Shirahama, T. et al. In vitro and in vivo induction of apoptosis by sphingosine and N,N-dimethylsphingosine in human epidermoid carcinoma KB-3-1 and its multidrug-resistant cells. Clin. Cancer Res. 3, 257–264 (1997).

    CAS  PubMed  Google Scholar 

  102. Sweeney, E. A. et al. Sphingosine and its methylated derivative N,N-dimethylsphingosine (DMS) induce apoptosis in a variety of human cancer cell lines. Int. J. Cancer 66, 358–366 (1996).

    Article  CAS  PubMed  Google Scholar 

  103. Cuvillier, O. & Levade, T. Sphingosine 1-phosphate antagonizes apoptosis of human leukemia cells by inhibiting release of cytochrome c and Smac/DIABLO from mitochondria. Blood 98, 2828–2836 (2001).

    Article  CAS  PubMed  Google Scholar 

  104. Tilly, J. L. & Kolesnick, R. N. Sphingolipids, apoptosis, cancer treatments and the ovary: investigating a crime against female fertility. Biochim. Biophys. Acta 1585, 135–138 (2002).

    Article  CAS  PubMed  Google Scholar 

  105. Suomalainen, L. et al. Sphingosine-1-phosphate in inhibition of male germ cell apoptosis in the human testis. J. Clin. Endocrinol. Metab. 88, 5572–5579 (2003).

    Article  CAS  PubMed  Google Scholar 

  106. Morita, Y. et al. Oocyte apoptosis is suppressed by disruption of the acid sphingomyelinase gene or by sphingosine-1-phosphate therapy. Nature Med. 6, 1109–1114 (2000).

    Article  CAS  PubMed  Google Scholar 

  107. Gräler, M. H. & Goetzl, E. J. The immunosuppressant FTY720 down-regulates sphingosine 1-phosphate G protein–coupled receptors. FASEB J. 10 (Epub) (2004).

  108. Billich, A. et al. Phosphorylation of the immunomodulatory drug FTY720 by sphingosine kinases. J. Biol. Chem. 278, 47408–47415 (2003).

    Article  CAS  PubMed  Google Scholar 

  109. Paugh, S. W. et al. The immunosuppressant FTY720 is phosphorylated by sphingosine kinase type 2. FEBS Lett. 554, 189–193 (2003).

    Article  CAS  PubMed  Google Scholar 

  110. Mandala, S. et al. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science 296, 346–349 (2002).

    Article  CAS  PubMed  Google Scholar 

  111. Matsuoka, Y et al. A novel immunosuppressive agent FTY720 induced Akt dephosphorylation in leukemia cells. Br. J. Pharmacol. 138, 1303–1312 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Azuma, H. et al. Induction of apoptosis in human bladder cancer cells in vitro and in vivo caused by FTY720 treatment. J. Urol. 169, 2372–2377 (2003).

    Article  CAS  PubMed  Google Scholar 

  113. Sonoda, Y. et al. FTY720, a novel immunosuppressive agent, induces apoptosis in human glioma cells. Biochem. Biophys. Res. Commun. 281, 282–288 (2001).

    Article  CAS  PubMed  Google Scholar 

  114. Wang, J. D. et al. Early induction of apoptosis in androgen-independent prostate cancer cell line by FTY720 requires caspase-3 activation. Prostate 40, 50–55 (1999).

    Article  CAS  PubMed  Google Scholar 

  115. Azuma, H. et al. Marked prevention of tumor growth and metastasis by a novel immunosuppressive agent, FTY720, in mouse breast cancer models. Cancer Res. 62, 1410–1419 (2202).

    Google Scholar 

  116. Ricchi, P. et al. Nonsteroidal anti-inflammatory drugs in colorectal cancer: from prevention to therapy. Br. J. Cancer 88, 803–807 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Symolon, H. et al. Dietary soy sphingolipids suppress tumorigenesis and gene expression in 1,2-dimethylhydrazine-terated CF1 mice and ApcMin/+ mice. J. Nutr. 134, 1157–1161 (2004).

    Article  CAS  PubMed  Google Scholar 

  118. Merrill, A. H. Jr. et al. Sphingolipid metabolism: roles in signal transduction and disruption by fumonisins. Environ. Health Perspect. 109, 283–289 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Huitema, K. et al. Identification of a family of animal sphingomyelin synthases. EMBO J. 23, 33–44 (2004).

    Article  CAS  PubMed  Google Scholar 

  120. Yamaoka, S. et al. Expression cloning of a human cDNA restoring sphingomyelin synthesis and cell growth in sphingomyelin synthase-defective lymphoid cells. J. Biol. Chem. 279, 18688–18693 (2004).

    Article  CAS  PubMed  Google Scholar 

  121. Sugiura, M. et al. Ceramide kinase, a novel lipid kinase. Molecular cloning and functional characterization. J. Biol. Chem. 277, 23294–23300 (2002).

    Article  CAS  PubMed  Google Scholar 

  122. El Bawab, S. et al. Ceramidases in the regulation of ceramide levels and function. Subcell. Biochem. 36, 187–205 (2002).

    Article  CAS  PubMed  Google Scholar 

  123. Mao, C. et al. Cloning and characterization of a novel human alkaline ceramidase. A mammalian enzyme that hydrolyzes phytoceramide. J. Biol. Chem. 276, 26577–26588 (2001).

    Article  CAS  PubMed  Google Scholar 

  124. Strelow, A. et al. Overexpression of acid ceramidase protects from tumor necrosis factor-induced cell death. J. Exp. Med. 192, 601–612 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Dobrowsky, R. T. et al. Ceramide activates heterotrimeric protein phosphatase 2A. J. Biol. Chem. 268, 15523–15530 (1993).

    Article  CAS  PubMed  Google Scholar 

  126. Bourbon, N. A. et al. Ceramide-induced inhibition of Akt is mediated through protein kinase Cζ: implications for growth arrest. J. Biol. Chem. 277, 3286–3292 (2002).

    Article  CAS  PubMed  Google Scholar 

  127. Pitson, S. M. et al. Activation of sphingosine kinase 1 by ERK1/2-mediated phosphorylation. EMBO J. 22, 5491–5500 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Spiegel, S. & Milstien, S. Sphingosine-1-phosphate: signaling inside and out. FEBS Lett. 476, 55–67 (2000).

    Article  CAS  PubMed  Google Scholar 

  129. Hanada, K. et al. Molecular machinery for non-vesicular trafficking of ceramide. Nature 426, 803–809 (2003). Describes the identification of a transporter protein, CERT, that transports ceramide from ER to Golgi specifically for the synthesis of sphingomyelin.

    Article  CAS  PubMed  Google Scholar 

  130. Ruvolo, P. P. et al. Ceramide induces Bcl2 dephosphorylation via a mechanism involving mitochondrial PP2A. J. Biol. Chem. 274, 20296–20300 (1999).

    Article  CAS  PubMed  Google Scholar 

  131. Alphonse, G. et al. Overcoming resistance to γ-rays in squamous carcinoma cells by poly-drug elevation of ceramide levels. Oncogene 23, 2703–2715 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by research grants from the National Institutes of Health/National Cancer Institute. We apologize to investigators whose manuscripts could not be cited in this article because of space limitations. We would like to thank L. Wooten, C. E. Senkal and M. Hinson for their assistance during the preparation of the manuscript. We also thank Z. Sculz, D. Perry and N. Marchesini for critically reviewing the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yusuf A. Hannun.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

Cancer.gov

breast cancer

colon cancer

head and neck cancer

kidney cancer

lung cancer

ovarian cancer

prostate cancer

stomach cancer

uterine cancer

Entrez Gene

AKT

BCL2

caspase-3

caspase 9

cathepsin D

CDK2

CDK4

COX2

c-RAF

FAN

kinase suppressor of RAS

KIP1

Lac1

Lag1

P-gp

phospholipase D

PKCζ

RB

SK1

TNFα

TP53

TRAF2

VEGF

WAF1

FURTHER INFORMATION

Ceramide Web

Glossary

LIPID RAFTS

Dynamic assemblies of cholesterol and sphingolipids in the plasma membrane that are important for amassing signalling complexes or specific aggregations of particular proteins.

MULTIDRUG RESISTANCE

Simultaneous resistance to several structurally unrelated drugs that do not have a common mechanism of action.

CASPASES

A family of cystein proteases that are activated in response to pro-apoptotic signals and cleave a number of specific protein targets, facilitating the rapid induction of apoptosis.

RNAI-MEDIATED KNOCKDOWN

Use of double-stranded RNA to target specific mRNAs for degradation, resulting in sequence-specific post-transcriptional gene silencing.

REACTIVE OXYGEN SPECIES

Highly reactive chemical radicals that are generated as products of oxygen degradation.

CRISIS

A terminal block in cell proliferation in which cells are destined to die because of collapse of telomeres at the end of chromosomes.

SCID MICE

Severe combined immuno-deficient mice used for the development of tumour xenografts.

VASCULAR SPROUTING

The vascular sprouting process is based on endothelial cell migration, proliferation and tube formation during vasculogenesis, which in situ is the differentiation and growth of blood vessels from mesodermal-derived haemangioblasts.

NEOVASCULARIZATION

A form of angiogenesis that is responsible for the remodelling and expansion of the vascular network.

OLIGODEOXYRIBONUCLEOTIDES

Synthetic, short, defined sequences of DNA.

ABC TRANSPORTER

A member of the membrane-spanning transporter protein family containing an ATP-binding cassette (ABC).

EPIDERMOID

A tissue structure that is similar to that of the flattened top layers of cells (squames) in the epidermis of the skin. It can cover tumours arising from the skin and upper oesophagus, and squamous-cell carcinoma of the lung, among others.

ASCITES

Excess fluid in the space between the membranes lining the abdomen and abdominal organs (the peritoneal cavity). Ascites can occur in patients with cancers affecting organs within the peritoneal cavity, that is, ovarian, liver and intestinal cancers.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ogretmen, B., Hannun, Y. Biologically active sphingolipids in cancer pathogenesis and treatment. Nat Rev Cancer 4, 604–616 (2004). https://doi.org/10.1038/nrc1411

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc1411

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing