Vol 9, No 4 (2018)
Review paper
Published online: 2019-03-06

open access

Page views 763
Article views/downloads 2380
Get Citation

Connect on Social Media

Connect on Social Media

Role and therapeutic potential of sphingolipids signaling in hematological malignances

Marzena Wątek12, Bonita Durnaś2, Tomasz Wollny2, Małgorzata Żendzian-Piotrowska3, Marcin Pasiarski2, Stanisław Góźdź2
Hematologia 2018;9(4):318-329.

Abstract

One of the key obstacles in the progress of cancer treatment is the lack of balance between the
uncontrolled proliferation and cell apoptosis. It is now known that sphingolipids are essential
molecules regulating the processes of growth, differentiation and death of living cells. Depending
on their chemical nature, sphingolipids may have a stimulatory (S1P, sphingosine-1-phosphate)
or inhibitory (ceramide) effect on cellular proliferation. A number of different studies have shown
that the generation of ceramide in response to cytotoxic therapy is an important element leading
to cell death. Cancer cells use different methods limiting the production of ceramides that leads to
their removal. The effect of oncogenic S1P results from its stimulating effect on DNA synthesis
and chemotactic mobility of the vascular endothelial cells and angiogenesis. The use of monoclonal
anti-S1P antibodies is potentially a valuable therapeutic option for inhibiting angiogenesis
determining the growth of tumors. It was additionally demonstrated that S1P beyond the direct
and indirect by stimulating the release of vascular endothelial growth factor and basic fibroblast
growth factor angiogenic action has an effect on tumor growth and its metastatic potential. Among
the sphingolipids, ceramide was identified first as inducing differentiation and the death of human
HL-60 promyelocytic leukemia cells. Progress in understanding the role of sphingolipids was
regarded until recently as the only structural component of cell membranes allowing the use in the
treatment of complex properties of this group of signaling molecules. Thus, it has become important
to clarify the role of sphingolipids in the regulation of the balance between proliferation signals/
/survival rate and death of cells in order to develop new therapies for neoplastic diseases of myeloid
and lymphoid origin.

Article available in PDF format

View PDF (Polish) Download PDF file

References

  1. Ogretmen B, Hannun YA. Biologically active sphingolipids in cancer pathogenesis and treatment. Nat Rev Cancer. 2004; 4(8): 604–616.
  2. Kolesnick RN. 1,2-Diacylglycerols but not phorbol esters stimulate sphingomyelin hydrolysis in GH3 pituitary cells. J Biol Chem. 1987; 262(35): 16759–16762.
  3. Kolesnick RN. Thyrotropin-releasing hormone and phorbol esters induce phosphatidylcholine synthesis in GH3 pituitary cells. Evidence for stimulation via protein kinase C. J Biol Chem. 1987; 262(30): 14525–14530.
  4. Kolesnick RN, Paley AE. 1, 2-Diacylglycerols and phorbol esters stimulate phosphatidylcholine metabolism in GH3 pituitary cells. Evidence for separate mechanisms of action. J Biol Chem. 1987; 262(19): 9204–9210.
  5. Watters RJ, Wang HG, Sung SS, et al. Targeting sphingosine-1-phosphate receptors in cancer. Anticancer Agents Med Chem. 2011; 11(9): 810–817.
  6. Huang WC, Chen CL, Lin YS, et al. Apoptotic sphingolipid ceramide in cancer therapy. J Lipids. 2011; 2011: 565316.
  7. Brizuela L, Ader I, Mazerolles C, et al. First evidence of sphingosine 1-phosphate lyase protein expression and activity downregulation in human neoplasm: implication for resistance to therapeutics in prostate cancer. Mol Cancer Ther. 2012; 11(9): 1841–1851.
  8. Liu Y, Deng J, Wang L, et al. S1PR1 is an effective target to block STAT3 signaling in activated B cell-like diffuse large B-cell lymphoma. Blood. 2012; 120(7): 1458–1465.
  9. Sanna MG, Liao J, Jo E, et al. Sphingosine 1-phosphate (S1P) receptor subtypes S1P1 and S1P3, respectively, regulate lymphocyte recirculation and heart rate. J Biol Chem. 2004; 279(14): 13839–13848.
  10. Shimizu T. Lipid mediators in health and disease: enzymes and receptors as therapeutic targets for the regulation of immunity and inflammation. Annu Rev Pharmacol Toxicol. 2009; 49: 123–150.
  11. Clarke CJ, Wu BX, Hannun YA. The neutral sphingomyelinase family: identifying biochemical connections. Adv Enzyme Regul. 2011; 51(1): 51–58.
  12. Hammad SM. Blood sphingolipids in homeostasis and pathobiology. Adv Exp Med Biol. 2011; 721: 57–66.
  13. Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol. 2008; 9(2): 139–150.
  14. Ohanian J, Ohanian V. Sphingolipids in mammalian cell signalling. Cell Mol Life Sci. 2001; 58(14): 2053–2068.
  15. Gault CR, Obeid LM, Hannun YA. An overview of sphingolipid metabolism: from synthesis to breakdown. Adv Exp Med Biol. 2010; 688: 1–23.
  16. Wątek M, Durnaś B, Wollny T, et al. Unexpected profile of sphingolipid contents in blood and bone marrow plasma collected from patients diagnosed with acute myeloid leukemia. Lipids Health Dis. 2017; 16(1): 235.
  17. 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. 2001; 98(9): 2828–2836.
  18. Rodriguez-Lafrasse C, Alphonse G, Aloy MT, et al. Increasing endogenous ceramide using inhibitors of sphingolipid metabolism maximizes ionizing radiation-induced mitochondrial injury and apoptotic cell killing. Int J Cancer. 2002; 101(6): 589–598.
  19. Bonhoure E, Pchejetski D, Aouali N, et al. Overcoming MDR-associated chemoresistance in HL-60 acute myeloid leukemia cells by targeting sphingosine kinase-1. Leukemia. 2006; 20(1): 95–102.
  20. Powell JA, Lewis AC, Zhu W, et al. Targeting sphingosine kinase 1 induces MCL1-dependent cell death in acute myeloid leukemia. Blood. 2017; 129(6): 771–782.
  21. Baldari CT. S1PR2 deficiency in DLBCL: a FOXy connection. Blood. 2016; 127(11): 1380–1381.
  22. Paugh SW, Paugh BS, Rahmani M, et al. A selective sphingosine kinase 1 inhibitor integrates multiple molecular therapeutic targets in human leukemia. Blood. 2008; 112(4): 1382–1391.
  23. Sabbadini RA. Targeting sphingosine-1-phosphate for cancer therapy. Br J Cancer. 2006; 95(9): 1131–1135.
  24. Lee OH, Kim YM, Lee YM, et al. Sphingosine 1-phosphate induces angiogenesis: its angiogenic action and signaling mechanism in human umbilical vein endothelial cells. Biochem Biophys Res Commun. 1999; 264(3): 743–750.
  25. Argraves KM, Wilkerson BA, Argraves WS, et al. Sphingosine-1-phosphate signaling promotes critical migratory events in vasculogenesis. J Biol Chem. 2004; 279(48): 50580–50590.
  26. Wang G, Silva J, Krishnamurthy K, et al. Direct binding to ceramide activates protein kinase Czeta before the formation of a pro-apoptotic complex with PAR-4 in differentiating stem cells. J Biol Chem. 2005; 280(28): 26415–26424.
  27. Kolesnick R, Fuks Z. Radiation and ceramide-induced apoptosis. Oncogene. 2003; 22(37): 5897–5906.
  28. Castillo SS, Levy M, Thaikoottathil JV, et al. Reactive nitrogen and oxygen species activate different sphingomyelinases to induce apoptosis in airway epithelial cells. Exp Cell Res. 2007; 313(12): 2680–2686.
  29. Hetz CA. Caspase-dependent initiation of apoptosis and necrosis by the Fas receptor in lymphoid cells: onset of necrosis is associated with delayed ceramide increase. J Cell Sci. 2002; 115(23): 4671–4683.
  30. Oh JiE, So KS, Lim SeJ, et al. Induction of apoptotic cell death by a ceramide analog in PC-3 prostate cancer cells. Arch Pharm Res. 2006; 29(12): 1140–1146.
  31. MacRae VE, Burdon T, Ahmed SF, et al. Ceramide inhibition of chondrocyte proliferation and bone growth is IGF-I independent. J Endocrinol. 2006; 191(2): 369–377.
  32. Bartke N, Hannun YA. Bioactive sphingolipids: metabolism and function. J Lipid Res. 2009; 50(Suppl): S91–S96.
  33. Dobrzyn A, Chocian G. Sfingomielinowy szlak transmisjii sygnalow. Med Metabol. 2003; 7(1): 75–80.
  34. Morales A, Lee H, Goñi FM, et al. Sphingolipids and cell death. Apoptosis. 2007; 12(5): 923–939.
  35. Hearps AC, Burrows J, Connor CE, et al. Mitochondrial cytochrome c release precedes transmembrane depolarisation and caspase-3 activation during ceramide-induced apoptosis of Jurkat T cells. Apoptosis. 2002; 7(5): 387–394.
  36. Mullen TD, Obeid L. Ceramide and apoptosis: exploring the enigmatic connections between sphingolipid metabolism and programmed cell death. Anticancer Agents Med Chem. 2012; 12(4): 340–363.
  37. Jayadev S, Liu B, Bielawska AE, et al. Role for ceramide in cell cycle arrest. J Biol Chem. 1995; 270(5): 2047–2052.
  38. Kitatani K, Taniguchi M, Okazaki T. Role of sphingolipids and metabolizing enzymes in hematological malignancies. Mol Cells. 2015; 38(6): 482–495.
  39. Cuvillier O, Nava VE, Murthy SK, et al. Sphingosine generation, cytochrome c release, and activation of caspase-7 in doxorubicin-induced apoptosis of MCF7 breast adenocarcinoma cells. Cell Death Differ. 2001; 8(2): 162–171.
  40. Wallington-Beddoe CT, Bradstock KF, Bendall LJ. Oncogenic properties of sphingosine kinases in haematological malignancies. Br J Haematol. 2013; 161(5): 623–638.
  41. Xu XQ, Huang CM, Zhang YF, et al. S1PR1 mediates anti‑apoptotic/pro‑proliferative processes in human acute myeloid leukemia cells. Mol Med Rep. 2016; 14(4): 3369–3375.
  42. Bednarek J, Wesierska-Gadek J, Kiliańska ZM. [New face of antiapoptotic proteins. I. Protein Mcl-1]. Postepy Biochem. 2007; 53(3): 228–238.
  43. Salas A, Ponnusamy S, Senkal CE, et al. Sphingosine kinase-1 and sphingosine 1-phosphate receptor 2 mediate Bcr-Abl1 stability and drug resistance by modulation of protein phosphatase 2A. Blood. 2011; 117(22): 5941–5952.
  44. Li QF, Wu CT, Duan HF, et al. Activation of sphingosine kinase mediates suppressive effect of interleukin-6 on human multiple myeloma cell apoptosis. Br J Haematol. 2007; 138(5): 632–639.
  45. Tian H, Yu Z. Resveratrol induces apoptosis of leukemia cell line K562 by modulation of sphingosine kinase-1 pathway. Int J Clin Exp Pathol. 2015; 8(3): 2755–2762.
  46. Li QF, Wu CT, Guo Q, et al. Sphingosine 1-phosphate induces Mcl-1 upregulation and protects multiple myeloma cells against apoptosis. Biochem Biophys Res Commun. 2008; 371(1): 159–162.
  47. García-Bernal D, Redondo-Muñoz J, Dios-Esponera A, et al. Sphingosine-1-phosphate activates chemokine-promoted myeloma cell adhesion and migration involving α4β1 integrin function. J Pathol. 2013; 229(1): 36–48.
  48. Nishimura H, Akiyama T, Monobe Y, et al. Expression of sphingosine-1-phosphate receptor 1 in mantle cell lymphoma. Mod Pathol. 2010; 23(3): 439–449.
  49. Bode C, Berlin M, Röstel F, et al. Evaluating sphingosine and its analogues as potential alternatives for aggressive lymphoma treatment. Cell Physiol Biochem. 2014; 34(5): 1686–1700.
  50. Flori M, Schmid CA, Sumrall ET, et al. The hematopoietic oncoprotein FOXP1 promotes tumor cell survival in diffuse large B-cell lymphoma by repressing S1PR2 signaling. Blood. 2016; 127(11): 1438–1448.
  51. Koresawa R, Yamazaki K, Oka D, et al. Sphingosine-1-phosphate receptor 1 as a prognostic biomarker and therapeutic target for patients with primary testicular diffuse large B-cell lymphoma. Br J Haematol. 2016; 174(2): 264–274.
  52. Evangelisti C, Evangelisti C, Teti G, et al. Assessment of the effect of sphingosine kinase inhibitors on apoptosis,unfolded protein response and autophagy of T-cell acute lymphoblastic leukemia cells; indications for novel therapeutics. Oncotarget. 2014; 5(17): 7886–7901.
  53. Xu L, Zhang Y, Gao M, et al. Concurrent targeting Akt and sphingosine kinase 1 by A-674563 in acute myeloid leukemia cells. Biochem Biophys Res Commun. 2016; 472(4): 662–668.
  54. Tan SF, Liu X, Fox TE, et al. Acid ceramidase is upregulated in AML and represents a novel therapeutic target. Oncotarget. 2016; 7(50): 83208–83222.
  55. Beider K, Rosenberg E, Bitner H, et al. The sphingosine-1-phosphate modulator FTY720 targets multiple myeloma via the CXCR4/CXCL12 pathway. Clin Cancer Res. 2017; 23(7): 1733–1747.
  56. Chen L, Luo LF, Lu J, et al. FTY720 induces apoptosis of M2 subtype acute myeloid leukemia cells by targeting sphingolipid metabolism and increasing endogenous ceramide levels. PLoS One. 2014; 9(7): e103033.
  57. Smith P, O'Sullivan C, Gergely P. Sphingosine 1-phosphate signaling and its pharmacological modulation in allogeneic hematopoietic stem cell transplantation. Int J Mol Sci. 2017; 18(10).
  58. Vrzalikova K, Ibrahim M, Vockerodt M, et al. S1PR1 drives a feedforward signalling loop to regulate BATF3 and the transcriptional programme of Hodgkin lymphoma cells. Leukemia. 2018; 32(1): 214–223.



Hematology in Clinical Practice