Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jun 6.
Published in final edited form as: Anticancer Agents Med Chem. 2011 Nov;11(9):794–798. doi: 10.2174/187152011797655122

Targeting Sphingosine-1-Phosphate in Hematologic Malignancies

Christina E Stevenson 1, Kazuaki Takabe 1, Masayuki Nagahashi 1, Sheldon Milstien 2, Sarah Spiegel 2,*
PMCID: PMC3368248  NIHMSID: NIHMS379407  PMID: 21707492

Abstract

Sphingosine-1-phosphate (S1P) is a pleiotropic bioactive lipid mediator that regulates several processes important for hematologic cancer progression. S1P is generated by two sphingosine kinases, SphK1 and SphK2, and is exported outside the cell, where it activates specific cell surface S1P G-protein coupled receptors in autocrine/paracrine manner, coined “inside-out signaling”. In this review, we highlight the importance of SphK1 and inside-out signaling by S1P in hematologic malignancy. We also summarize the results of studies targeting the SphK1/S1P/S1P receptor axis and the effects of the S1P receptor modulator, FTY720, in hematologic malignancy.

Keywords: sphingosine-1-phosphate, sphingosine kinase, sphingosine-1-phosphate receptors, leukemia, lymphoma, hematologic malignancies

INTRODUCTION

Sphingosine-1-phosphate (S1P) is a pleiotropic bioactive lipid mediator that regulates numerous processes important for cancer progression including cell migration, growth, survival, and angiogenesis to name just a few [1, 2]. In contrast to S1P, which has been associated with cell growth and survival, its precursors, sphingosine and ceramide, have been shown to play a role in growth arrest and apoptosis. The balance between these counter-acting sphingolipid metabolites has been termed the “sphingolipid rheostat” and has been implicated in cell fate determination [3].

S1P is produced from sphingosine by sphingosine kinases, of which there are two isoenzymes (SphK1 and SphK2). Many growth factors, hormones, and cytokines stimulate SphK1 inducing its translocation to the plasma membrane leading to restricted formation of S1P which then can be exported by ATP-binding cassette transporters and subsequently activates its specific receptors in an autocrine/paracrine manner, defined as “inside-out signaling” [4]. S1P signals through five S1P specific G protein-coupled receptors (GPCR), known as S1P1–5 to exert many of its pleiotropic effects. The specific repertoire of S1P receptors that is expressed, together with their differential coupling to various heterotrimeric G-proteins that regulate numerous downstream signaling pathways, is responsible for the ability of S1P to regulate diverse physiological processes in a highly specific manner. A recent study demonstrated that STAT3 induces S1P1 expression and activation of S1P1 reciprocally regulates STAT3 activity, thus acting as a major positive feedback loop for persistent STAT3 activation in cancer cells and the tumor microenvironment for malignant progression [5].

S1P produced by SphK1 also has recently been shown to have direct intracellular actions. S1P binds to TRAF2 and is a required cofactor for its E3 ubiquitin ligase activity. This leads to lysine 63-linked polyubiquitination of RIP1, a critical event in NF-κB activation and provides a mechanistic explanation for the numerous observations of the importance of SphK1 in inflammatory, anti-apoptotic and immune processes [6].

Elevated expression of SphK1 has been observed in multiple types of cancer, including acute leukemia [7, 8]. Upregulation of SphK1 is associated with the tumorigenic phenotype of erythroleukemia, and overexpression of SphK1 in non-tumorigenic pro-erythroblasts increased their clonogenicity as well as resistance to apoptosis. They also acquired tumorigenicity when engrafted in vivo [9]. These results suggest that high expression of Sphk1 may be an oncogenic event required for progression of erythroleukemia.

In contrast to overexpression of SphK1, SphK2 overexpression suppresses cell proliferation [10]. Moreover, whereas expression of SphK1 reduces ceramide level, SphK2 increases it [11]. SphK1 and SphK2 have distinct and sometimes overlapping functions in cells, unique localizations depending on cell type, and the cellular location of S1P production may determine its actions. In many cancer cells SphK2 is localized to the nucleus where it is associated with the histone deacetylases, HDAC1 and HDAC2, in repressor complexes and inhibits their enzymatic activity, preventing the removal of acetyl groups from lysine residues within histone tails. This results in enhanced transcription of specific genes, such as the cyclin-dependent kinase inhibitor p21 or the transcriptional regulator c-fos. This links nuclear S1P and sphingolipid metabolism in the nucleus to epigenetic regulation of gene expression [12]. Vorinostat, an HDAC inhibitor, was recently approved for the treatment of cutaneous T-cell lymphoma by US Food and Drug Administration [13]. Understanding how S1P regulates HDAC activity in vivo may help in the development of new therapeutics that interfere with HDAC functions in a highly specific manner.

This review summarizes research to date on the involvement and mechanisms of action of S1P and the kinases that produce it in growth and progression of the hematologic malignancies leukemia and lymphoma. We will also discuss potential new options for therapy that target S1P signaling and function in these malignancies.

ROLE OF SPHK1, S1P, AND ITS RECEPTORS IN HEMATOLOGIC MALIGNANCIES

Abundant evidence indicates that S1P stimulates growth and survival of leukemia and lymphoma cells [3, 14, 15]. The initial observation in this field was that S1P stimulates the extracellular signal-regulated kinase (ERK)/MAPK pathway and counteracts ceramide-induced activation of stress-activated protein kinase (SAPK/JNK), resulting in decreased apoptosis [3]. S1P has been shown to prevent apoptosis in several human leukemia cells lines, including U937 and HL-60 cells, counteracting activation of caspases likely by inhibiting release of cytochrome c and Smac/DIABLO from mitochondria to the cytosol [15]. Furthermore, inhibition of SphKs induced ceramide accumulation, decreased S1P, and caused apoptosis equally in chemosensitive and chemoresistant cell lines that was reversed by exogenous treatment with S1P or by overexpression of SphK1 [16]. S1P mobilized intracellular calcium in the human leukemic cell line, U937, which was required for activation of NF-κB, a transcription factor important for their survival [14]. In agreement, SphK1 and NF-κB were found to be essential for the long-term survival of cytotoxic T lymphocytes in T cell large granular lymphocyte leukemia which features clonal expansion of antigen-primed competent cytotoxic T lymphocytes [17]. The Runx genes (Runx1, 2, and 3) regulate cell fate in development and can be oncogenic. Transgenic mice expressing Runx together with Myc develop lymphoma. The oncogenic potential of Runx was linked to key enzymes of sphingolipid metabolism (S1P phosphatase 1, UDP-glucose ceramide glycosyltransferase, and GM3 synthase) as direct targets for Runx transcriptional regulation in a manner consistent with survival and apoptosis resistance. The survival advantage conferred by ectopic Runx could be partially recapitulated by exogenous S1P and was accompanied by reduced phosphorylation of p38 MAPK [18]. In multiple myeloma cells, SphK1 is stimulated by IL-6, which influences their growth and survival. Moreover, SphK1 is involved in IL-6-mediated upregulation of myeloid cell leukaemia-1 (Mcl-1), leading to increased cell proliferation and survival [19], suggesting that SphK1 may contribute to this type of leukemia.

Elevation of S1P and SphK1 is an important factor that determines resistance to chemotherapy. It was reported that SphK1 expression was increased in Bcr-Abl-overexpressing leukemic cells, [20, 21]. Imatinib, a tyrosine kinase inhibitor, has been used in the treatment of certain leukemias because it blocks the formation of Bcr-Abl, which is crucial in the development of some types of leukemia. Imatinib has been shown to induce apoptosis in K562 cells, a human myelogenous leukemia cell line. Expression of SphK1 and generation of S1P were found to be increased significantly in Imatinib-resistant K562 cells [20]. Partial inhibition of SphK1 by siRNA reduced S1P levels and increased sensitivity to Imatinib-induced apoptosis in the resistant cells. Forced expression of SphK1 increased the ratio of S1P to C18-ceramide about six-fold, and prevented apoptosis significantly in response to Imatinib. This implies a role for SphK1 and S1P in the upregulation of Bcr-Abl expression at the post-transcriptional level, suggesting a mechanism for resistance to Imatinib-mediated apoptosis [20]. Similarly, it was shown that apoptosis of Imatinib-sensitive and resistant primary cells from chronic myeloid leukemia patients was induced by an inhibitor of SphK1 [22]. This study also substantiated the involvement of SphK1 in regulating Imatinib-induced apoptosis and established SphK1 as a downstream effector of the Bcr-Abl/Ras/ERK pathway inhibited by Imatinib but an upstream regulator of Bcl-2 family members [22].

Despite evidence that inside-out signaling by S1P plays an important role in cancer progression, much less is known of the importance of specific S1P receptors in hematological malignancies. T-lymphoma cell invasion is regulated by binding of S1P to its cell surface receptors that activates RhoA, phospholipase C and calcium signaling pathways leading to pseudopod formation and enhanced infiltration [23]. Yet the S1P receptors involved have not been identified. Multiple myeloma cells have been shown to express S1P1, S1P2, and S1P3. S1P, which protects multiple myeloma cells from apoptosis, upregulates Mcl-1, an anti-apoptotic protein that is a member of the Bcl-2 family [24]. Because this effect was blocked by pertussis toxin, which inhibits Gi protein signaling, it was suggested that one of these S1P receptors was involved [24]. More work is needed to unravel the importance of the S1P receptors in leukemia.

Interestingly, S1P5 receptor was identified as one of the differentially expressed genes in large granular lymphocyte leukemia [25]. This type of leukemia is often associated with autoimmune disease and is characterized by dysregulation of apoptosis. It is possible that this alteration in the apoptosis pathway may be due to activation of S1P5 receptors by S1P; however, this hypothesis has yet to be explored. Microarray-based immunohistochemistry with tissue samples from mantle cell lymphoma patients demonstrated that S1P1 was expressed on the surface of mantle cell lymphoma cells. PCR analysis of mantle cell lymphoma lines confirmed that S1P1 expression was upregulated [26]. Unfortunately, the role of S1P1 in mantle cell lymphoma has not been established yet. T-cell precursor malignancies, T-lymphoblastic lymphoma (T-LBL) and acute T-lymphoblastic leukemia (T-ALL), are known to be closely related, but it was not known why T-LBL remains highly localized with accumulation of those cells in mediastinal masses in some patients while disseminating rapidly as T-ALL in others. Based on a zebrafish model, it was recently suggested that expression of S1P1 in T-LBL cells may prevent their progression to T-ALL [27]. Feng et al reported that expression of S1P1 and its putative target ICAM-1 (Inter-Cellular Adhesion Molecule 1), which binds integrin, is higher in human T-LBL cells than in human T-ALL cells. They suggested that this T-LBL phenotype promotes homotypic cell-cell adhesion and blocks intravasation. Inhibition of S1P1 signaling in T-LBL cells led to decreased homotypic adhesion in vitro and increased tumor cell intravasation in vivo [27]. Thus, it seems that blockade of intravasation and hematologic dissemination in T-LBL may be due to elevated S1P1 signaling and increased expression of ICAM1 that augments homotypic cell-cell adhesion [27].

Interestingly S1P2 null mice develop germinal center-derived diffuse large B-cell lymphomas upon ageing, resulting in about half of all animals having this neoplasm by 1.5 to 2 years. Using histologic, immunophenotypic, and molecular analyses, a uniform tumor phenotype was noted with features of germinal center (GC)-derived diffuse large B-cell lymphoma (DLBCL). Tumor formation was preceded by increases in GC B cells, CD69 positive T cells, and increased formation of spontaneous GCs. Moreover, 26% of patients with DLBCL had multiple somatic mutations in the 5' sequences of the s1p2 gene. This work suggests that S1P2 signaling may play a critical role in suppression of diffuse large B-cell lymphomas derived from GCs [28].

SPHK1 TARGETED THERAPIES

Based upon increasing evidence that indicates a significant role for SphK1 and S1P in growth and survival of leukemia and lymphoma cells, it is not surprising that they have been the focus of some investigations as a target for therapy. N,N-Dimethylsphingosine (DMS), a pan-sphingosine kinase inhibitor that inhibits both SphK1 and SphK2, was shown to sensitize Jurkat, U937, and HL-60 cells to apoptosis triggered by ceramide, TNF-α, or serum deprivation [15, 29]. A caveat of this work is that DMS also inhibits protein kinase C. However, activation of protein kinase C itself leads to increased S1P levels and in some cells, DMS inhibits only SphKs and not protein kinase C [29]. Therefore, it is still possible that sensitization of cells to apoptosis by DMS is mediated by inhibition of SphKs. Because DMS is a toxic compound with very low aqueous solubility, its application as a therapeutic is severely limited. A related compound, N,N-dimethylphytosphingosine, was shown to inhibit sphingosine kinase activity in vitro [30]. N,N-Dimethylphytosphingosine decreased S1P by inhibiting SphK activity and induced caspase-dependent apoptosis of human leukemia cells which was dependent on increased reactive oxygen species [30].

F-12509a is a structurally complicated natural product inhibitor of SphK1 and SphK2 that was isolated from a culture broth of a discomycete [31]. F-12509a induced apoptosis and enhanced susceptibility to Imatinib of both sensitive and resistant chronic myeloid leukemia cells [22]. Moreover, treatment of human leukemia HL-60 cells with F-12509a, decreased S1P, increased ceramide accumulation, and caused apoptosis in both chemo-sensitive and multi-drug resistant lines [16]. As expression of multidrug resistant proteins is among the strongest prognostic factors in acute myelogenous leukemia, SphK inhibitors by overcoming apoptosis resistance and restoring sensitivity to chemotherapeutics, regardless of multidrug resistant status, may have potential for clinical development [16].

Another pan SphK inhibitor, 2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole, which is a non-ATP competitive inhibitor and thus expected to be more specific for SphKs, was used to assess the importance of S1P formation in daunorubicin sensitive and resistant leukemia cell lines [7]. This study showed that daunorubicin decreased S1P more in the sensitive cell lines than the resistant ones with corresponding inverse effects on ceramide levels. Furthermore, this SphK inhibitor also restored daunorubicin sensitivity in the resistant leukemia cell lines suggesting that combination of the SphK inhibitor and an anti-cancer drug such as daunorubicin might also be useful for future clinical development [7]. 4-[[4-(4-chlorophenyl)-2-thiazoyl]amino]phenol, called SKI-II, which inhibits both SphK1 and SphK2, significantly induced apoptosis of T cell large granular lymphocytic leukemia cells in a dose-dependent manner but not of normal peripheral blood mononuclear cells [17]. A specific SphK2 inhibitor has been shown to inhibit growth of several types of human tumor xenografts [3234], but its effects in leukemia have not yet been examined.

An issue that has hindered further development of SphK inhibitors is that their specificity has not been unequivocally established, and many inhibit both SphK1 and SphK2. As discussed, these two isoenzymes are distinctively different in localization and function; therefore, a pharmacological inhibitor that specifically inhibits one of the kinases was needed. (2R,3S,4E)-N-methyl-5-(4'-pentylphenyl)-2-aminopent-4-ene-1,3-diol, designated SK1-I, a potent, water soluble, isozyme-specific inhibitor of SphK1, was shown to decrease growth and survival of U937 and Jurkat cells and enhance apoptosis and cleavage of Bcl-2 [35]. SK1-I decreased S1P and concomitantly increased levels of its pro-apoptotic precursor ceramide. SK1-I also induced multiple perturbations in activation of signaling and survival-related proteins, including diminished phosphorylation of ERK1/2 and Akt. The importance of Akt survival signaling was confirmed by overexpression of constitutively active Akt which protected against SK1-I-induced apoptosis. Remarkably, SK1-I potently induced apoptosis in leukemic blasts isolated from patients with acute myelogenous leukemia but had little effects on normal peripheral blood mononuclear leukocytes. Moreover, SK1-I markedly reduced growth of AML xenograft tumors [35]. Taken together, these studies suggest that specific inhibitors of SphK1 warrant attention as potential additions to the therapeutic armamentarium in leukemia.

TARGETING S1P RECEPTORS WITH FTY720

FTY720, also known as fingolimod, is a fungal metabolite with immunosuppressive properties that has been approved by the FDA for treatment of multiple sclerosis [36]. It is a structural analogue of sphingosine and a prodrug, which is phosphorylated in vivo by SphK2 to phospho-FTY720, a S1P mimetic that binds to all of the S1P receptors except S1P2. FTY720 sequesters lymphocytes in lymph nodes by inducing internalization and degradation of S1P1, whose expression is crucial for their egress [37]. Hence, phospho-FTY720 is a “functional antagonist” of S1P1. There is no doubt that downregulation of specific S1P receptors is one of the major actions of this pro-drug. A number of reports have surfaced indicating that FTY720 reduces growth and induces apoptosis of tumor cells in culture and also reduces xenograft tumor growth. For example, proliferation of HL-60 and U937 leukemia cells was suppressed by FTY720 in a dose-dependent manner and activation of ERK1/2 was reduced as well as its translocation to nucleus during FTY720-induced apoptosis [38]. FTY720 was also shown to induce apoptosis of T-cell large granular lymphocyte leukemia cells and sensitize them to Fas-mediated apoptotic cell death [39]. FTY 720 has also been shown to mediate cytotoxic effects in different B-cell malignancies independent of activation of caspases [40]. Moreover, in CLL B-cells, FTY720 also induced downregulation of Mcl-1 but not Bcl-2. However, overexpression of Bcl-2 failed to protect these transformed B-cells from FTY720-induced apoptosis, suggesting a Bcl-2-independent mechanism. FTY720 treatment resulted in significantly prolonged survival in a xenograft SCID mouse model of disseminated B-cell lymphoma/leukemia [40], revealing a potential use of FTY720 as a therapeutic agent in B-cell malignancies.

FTY720 may also have intracellular actions that are independent of the S1P receptors [4144]. Several studies have investigated the mechanism by which FTY720 induces apoptosis and cell cycle arrest. Apoptosis of Jurkat T cells was triggered by intracellular accumulation of the phosphorylated compound acting through an yet unidentified intracellular target [42]. This led to the suggestion that sphingoid base substrates for SphK2 acting intracellularly could be useful in the treatment of lymphoproliferative diseases [42]. FTY720 induced dephosphorylation of Akt in Jurkat T cells and in human B-cell leukemia cells. In these cells, okadaic acid suppressed FTY720-induced dephosphorylation of Akt, suggesting that FTY720 promotes phosphoserine/phosphothreonine protein phosphatase activity [41]. Indeed, FTY720 was found to activate purified PP2A. This suggests that FTY720 activates PP2A phosphatase which dephosphorylates Akt, thus inhibiting this survival pathway resulting in the enhancement of apoptosis via the mitochondrial apoptotic pathway [41]. It also potently inhibited blast crisis chronic myelogenous leukemia and Philadelphia chromosome–positive acute lymphocytic leukemia cells from two fatal BCR-ABL leukemias (against which Abl kinase inhibitors fail to induce a long-term response) by impairing BCR-ABL activity and expression via activation of PP2A [44]. Furthermore, pharmacologic doses of FTY720 suppressed in vivo BCR-ABL-driven leukemogenesis without exerting any toxicity [44]. This suggests that FTY720 may be useful for treating patients who are resistant to Imatinib, Dasatinib and Nilotinib, and would complement drugs that target the BCR-ABL fusion protein. Recent findings show that activation of PP2A by FTY720 also caused the dephosphorylation of the c-KIT receptor and its downstream signaling targets pAkt, pSTAT5, and pERK1/2. Additionally, administration of FTY720 to mice delayed the growth of c-KIT tumors, inhibited splenic and bone marrow infiltration, and prolonged survival [45].

CONCLUSIONS

It has been well demonstrated that S1P plays an important role in multiple hematologic malignancies. Based upon studies that utilized pharmacological inhibitors and molecular interventions, targeting the SphK1/S1P/S1P receptor axis might be beneficial in treatment of hematologic malignancies and those that develop resistance to therapies. The promising results using FTY720 raise the intriguing notion that this compound deserves consideration as a drug for therapy of hematologic malignancies.

ACKNOWLEDGEMENTS

This work was supported by National Institutes of Health Grants R01CA61774 R37GM043880, R01AI50094, 1U19AI077435 (S. S.), and K12HD055881 (K. T.) and Susan G. Komen for the Cure Career Catalyst Research Grant KG090510 (K. T.). M. N. was supported by the SUMITOMO Life Social Welfare Services Foundation.

REFERENCES

  • 1.Milstien S, Spiegel S. Targeting sphingosine-1-phosphate: A novel avenue for cancer therapeutics. Cancer Cell. 2006;9(3):148–150. doi: 10.1016/j.ccr.2006.02.025. [DOI] [PubMed] [Google Scholar]
  • 2.Pyne NJ, Pyne S. Sphingosine 1-phosphate and cancer. Nat. Rev. Cancer. 2010;10(7):489–503. doi: 10.1038/nrc2875. [DOI] [PubMed] [Google Scholar]
  • 3.Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Coso OA, Gutkind S, Spiegel S. Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature. 1996;381(6585):800–803. doi: 10.1038/381800a0. [DOI] [PubMed] [Google Scholar]
  • 4.Takabe K, Kim RH, Allegood JC, Mitra P, Ramachandran S, Nagahashi M, Harikumar KB, Hait NC, Milstien S, Spiegel S. Estradiol induces export of sphingosine 1-phosphate from breast cancer cells via ABCC1 and ABCG2. J. Biol. Chem. 2010;285(14):10477–10486. doi: 10.1074/jbc.M109.064162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lee H, Deng J, Kujawski M, Yang C, Liu Y, Herrmann A, Kortylewski M, Horne D, Somlo G, Forman S, Jove R, Yu H. STAT3-induced S1PR1 expression is crucial for persistent STAT3 activation in tumors. Nat. Med. 2010 doi: 10.1038/nm.2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Alvarez SE, Harikumar KB, Hait NC, Allegood J, Strub GM, Kim EY, Maceyka M, Jiang H, Luo C, Kordula T, Milstien S, Spiegel S. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature. 2010;465(7301):1084–1088. doi: 10.1038/nature09128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sobue S, Nemoto S, Murakami M, Ito H, Kimura A, Gao S, Furuhata A, Takagi A, Kojima T, Nakamura M, Ito Y, Suzuki M, Banno Y, Nozawa Y, Murate T. Implications of sphingosine kinase 1 expression level for the cellular sphingolipid rheostat: relevance as a marker for daunorubicin sensitivity of leukemia cells. Int. J. Hematol. 2008;87(3):266–275. doi: 10.1007/s12185-008-0052-0. [DOI] [PubMed] [Google Scholar]
  • 8.Sobue S, Iwasaki T, Sugisaki C, Nagata K, Kikuchi R, Murakami M, Takagi A, Kojima T, Banno Y, Akao Y, Nozawa Y, Kannagi R, Suzuki M, Abe A, Naoe T, Murate T. Quantitative RT-PCR analysis of sphingolipid metabolic enzymes in acute leukemia and myelodysplastic syndromes. Leukemia. 2006;20(11):2042–2046. doi: 10.1038/sj.leu.2404386. [DOI] [PubMed] [Google Scholar]
  • 9.Le Scolan E, Pchejetski D, Banno Y, Denis N, Mayeux P, Vainchenker W, Levade T, Moreau-Gachelin F. Overexpression of sphingosine kinase 1 is an oncogenic event in erythroleukemic progression. Blood. 2005;106(5):1808–1816. doi: 10.1182/blood-2004-12-4832. [DOI] [PubMed] [Google Scholar]
  • 10.Igarashi N, Okada T, Hayashi S, Fujita T, Jahangeer S, Nakamura SI. Sphingosine kinase 2 is a nuclear protein and inhibits DNA synthesis. J. Biol. Chem. 2003;278(47):46832–46839. doi: 10.1074/jbc.M306577200. [DOI] [PubMed] [Google Scholar]
  • 11.Maceyka M, Sankala H, Hait NC, Le Stunff H, Liu H, Toman R, Collier C, Zhang M, Satin L, Merrill AH, Jr, Milstien S, Spiegel S. Sphk1 and Sphk2: Sphingosine kinase isoenzymes with opposing functions in sphingolipid metabolism. J. Biol. Chem. 2005;280(44):37118–37129. doi: 10.1074/jbc.M502207200. [DOI] [PubMed] [Google Scholar]
  • 12.Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, Luo C, Marmorstein R, Kordula T, Milstien S, Spiegel S. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science. 2009;325(5945):1254–1257. doi: 10.1126/science.1176709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lane AA, Chabner BA. Histone deacetylase inhibitors in cancer therapy. J. Clin. Oncol. 2009;27(32):5459–5468. doi: 10.1200/JCO.2009.22.1291. [DOI] [PubMed] [Google Scholar]
  • 14.Shatrov VA, Lehmann V, Chouaib S. Sphingosine-1-phosphate mobilizes intracellular calcium and activates transcription factor NF-kB in U937 cells. Biochem. Biophys. Res. Commun. 1997;234(1):121–124. doi: 10.1006/bbrc.1997.6598. [DOI] [PubMed] [Google Scholar]
  • 15.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. doi: 10.1182/blood.v98.9.2828. [DOI] [PubMed] [Google Scholar]
  • 16.Bonhoure E, Pchejetski D, Aouali N, Morjani H, Levade T, Kohama T, Cuvillier O. Overcoming MDR-associated chemoresistance in HL-60 acute myeloid leukemia cells by targeting sphingosine kinase-1. Leukemia. 2006;20(1):95–102. doi: 10.1038/sj.leu.2404023. [DOI] [PubMed] [Google Scholar]
  • 17.Zhang R, Shah MV, Yang J, Nyland SB, Liu X, Yun JK, Albert R, Loughran TP., Jr Network model of survival signaling in large granular lymphocyte leukemia. Proc. Natl. Acad. Sci. U.S.A. 2008;105(42):16308–16313. doi: 10.1073/pnas.0806447105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kilbey A, Terry A, Jenkins A, Borland G, Zhang Q, Wakelam MJ, Cameron ER, Neil JC. Runx regulation of sphingolipid metabolism and survival signaling. Cancer Res. 2010;70(14):5860–5869. doi: 10.1158/0008-5472.CAN-10-0726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Li QF, Wu CT, Duan HF, Sun HY, Wang H, Lu ZZ, Zhang QW, Liu HJ, Wang LS. Activation of sphingosine kinase mediates suppressive effect of interleukin-6 on human multiple myeloma cell apoptosis. Br. J. Haematol. 2007;138(5):632–639. doi: 10.1111/j.1365-2141.2007.06711.x. [DOI] [PubMed] [Google Scholar]
  • 20.Baran Y, Salas A, Senkal CE, Gunduz U, Bielawski J, Obeid LM, Ogretmen B. Alterations of ceramide/sphingosine 1-phosphate rheostat involved in the regulation of resistance to imatinib-induced apoptosis in K562 human chronic myeloid leukemia cells. J. Biol. Chem. 2007;282(15):10922–10934. doi: 10.1074/jbc.M610157200. [DOI] [PubMed] [Google Scholar]
  • 21.Li QF, Huang WR, Duan HF, Wang H, Wu CT, Wang LS. Sphingosine kinase-1 mediates BCR/ABL-induced upregulation of Mcl-1 in chronic myeloid leukemia cells. Oncogene. 2007;26(57):7904–7908. doi: 10.1038/sj.onc.1210587. [DOI] [PubMed] [Google Scholar]
  • 22.Bonhoure E, Lauret A, Barnes DJ, Martin C, Malavaud B, Kohama T, Melo JV, Cuvillier O. Sphingosine kinase-1 is a downstream regulator of imatinib-induced apoptosis in chronic myeloid leukemia cells. Leukemia. 2008;22(5):971–979. doi: 10.1038/leu.2008.95. [DOI] [PubMed] [Google Scholar]
  • 23.Stam JC, Michiels F, Kammen RA, Moolenaar WH, Collard JG. Invasion of T-lymphoma cells: cooperation between Rho family GTPases and lysophospholipid receptor signaling. EMBO J. 1998;17(14):4066–4074. doi: 10.1093/emboj/17.14.4066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Li QF, Wu CT, Guo Q, Wang H, Wang LS. Sphingosine 1-phosphate induces Mcl-1 upregulation and protects multiple myeloma cells against apoptosis. Biochem. Biophys. Res. Commun. 2008;371(1):159–162. doi: 10.1016/j.bbrc.2008.04.037. [DOI] [PubMed] [Google Scholar]
  • 25.Kothapalli R, Kusmartseva I, Loughran TP. Characterization of a human sphingosine-1-phosphate receptor gene (S1P5) and its differential expression in LGL leukemia. Biochim. Biophys. Acta. 2002;1579(2–3):117–123. doi: 10.1016/s0167-4781(02)00529-8. [DOI] [PubMed] [Google Scholar]
  • 26.Nishimura H, Akiyama T, Monobe Y, Matsubara K, Igarashi Y, Abe M, Sugihara T, Sadahira Y. Expression of sphingosine-1-phosphate receptor 1 in mantle cell lymphoma. Mod. Pathol. 2010;23(3):439–449. doi: 10.1038/modpathol.2009.194. [DOI] [PubMed] [Google Scholar]
  • 27.Feng H, Stachura DL, White RM, Gutierrez A, Zhang L, Sanda T, Jette CA, Testa JR, Neuberg DS, Langenau DM, Kutok JL, Zon LI, Traver D, Fleming MD, Kanki JP, Look AT. T-lymphoblastic lymphoma cells express high levels of BCL2, S1P1, and ICAM1, leading to a blockade of tumor cell intravasation. Cancer Cell. 2010;18(4):353–366. doi: 10.1016/j.ccr.2010.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cattoretti G, Mandelbaum J, Lee N, Chaves AH, Mahler AM, Chadburn A, Dalla-Favera R, Pasqualucci L, MacLennan AJ. Targeted disruption of the S1P2 sphingosine 1-phosphate receptor gene leads to diffuse large B-cell lymphoma formation. Cancer Res. 2009;69(22):8686–8692. doi: 10.1158/0008-5472.CAN-09-1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Edsall LC, Van Brocklyn JR, Cuvillier O, Kleuser B, Spiegel S. N,N-Dimethylsphingosine is a potent competitive inhibitor of sphingosine kinase but not of protein kinase C: modulation of cellular levels of sphingosine 1-phosphate and ceramide. Biochemistry. 1998;37(37):12892–12898. doi: 10.1021/bi980744d. [DOI] [PubMed] [Google Scholar]
  • 30.Kim BM, Choi YJ, Han Y, Yun YS, Hong SH. N,N-dimethyl phytosphingosine induces caspase-8-dependent cytochrome c release and apoptosis through ROS generation in human leukemia cells. Toxicol. Appl. Pharmacol. 2009;239(1):87–97. doi: 10.1016/j.taap.2009.05.020. [DOI] [PubMed] [Google Scholar]
  • 31.Kono K, Sugiura M, Kohama T. Inhibition of recombinant sphingosine kinases by novel inhibitors of microbial origin, F-12509A and B-5354c. J. Antibiot. (Tokyo) 2002;55(1):99–103. doi: 10.7164/antibiotics.55.99. [DOI] [PubMed] [Google Scholar]
  • 32.French KJ, Zhuang Y, Maines LW, Gao P, Wang W, Beljanski V, Upson JJ, Green CL, Keller SN, Smith CD. Pharmacology and antitumor activity of ABC294640, a selective inhibitor of sphingosine kinase-2. J. Pharmacol. Exp. Ther. 2010;333(1):129–139. doi: 10.1124/jpet.109.163444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Beljanski V, Lewis CS, Smith CD. Antitumor activity of sphingosine kinase 2 inhibitor ABC294640 and sorafenib in hepatocellular carcinoma xenografts. Cancer Biol. Ther. 2011;11(5):524–534. doi: 10.4161/cbt.11.5.14677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Antoon JW, White MD, Slaughter EM, Driver JL, Khalili HS, Elliott S, Smith CD, Burow ME, Beckman BS. Targeting NFkB mediated breast cancer chemoresistance through selective inhibition of sphingosine kinase-2. Cancer Biol. Ther. 2011;11(7):678–689. doi: 10.4161/cbt.11.7.14903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Paugh SW, Paugh BS, Rahmani M, Kapitonov D, Almenara JA, Kordula T, Milstien S, Adams JK, Zipkin RE, Grant S, Spiegel S. A selective sphingosine kinase 1 inhibitor integrates multiple molecular therapeutic targets in human leukemia. Blood. 2008;112(4):1382–1391. doi: 10.1182/blood-2008-02-138958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Brinkmann V, Billich A, Baumruker T, Heining P, Schmouder R, Francis G, Aradhye S, Burtin P. Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat. Rev. Drug Discov. 2010;9(11):883–897. doi: 10.1038/nrd3248. [DOI] [PubMed] [Google Scholar]
  • 37.Mandala S, Hajdu R, Bergstrom J, Quackenbush E, Xie J, Milligan J, Thornton R, Shei GJ, Card D, Keohane C, Rosenbach M, Hale J, Lynch CL, Rupprecht K, Parsons W, Rosen H. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science. 2002;296(5566):346–349. doi: 10.1126/science.1070238. [DOI] [PubMed] [Google Scholar]
  • 38.Li D, Zhang Y, Hu X, Cao W, Huang W. Role of extracelluar regulated protein kinases in FTY720-induced apoptosis of leukemia cell lines HL-60 and U937. J. Huazhong Univ. Sci. Technolog. Med. Sci. 2004;24(1):45–47. doi: 10.1007/BF02830703. [DOI] [PubMed] [Google Scholar]
  • 39.Shah MV, Zhang R, Irby R, Kothapalli R, Liu X, Arrington T, Frank B, Lee NH, Loughran TP., Jr Molecular profiling of LGL leukemia reveals role of sphingolipid signaling in survival of cytotoxic lymphocytes. Blood. 2008;112(3):770–781. doi: 10.1182/blood-2007-11-121871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Liu Q, Zhao X, Frissora F, Ma Y, Santhanam R, Jarjoura D, Lehman A, Perrotti D, Chen CS, Dalton JT, Muthusamy N, Byrd JC. FTY720 demonstrates promising preclinical activity for chronic lymphocytic leukemia and lymphoblastic leukemia/lymphoma. Blood. 2008;111(1):275–284. doi: 10.1182/blood-2006-10-053884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Matsuoka Y, Nagahara Y, Ikekita M, Shinomiya T. A novel immunosuppressive agent FTY720 induced Akt dephosphorylation in leukemia cells. Br. J. Pharmacol. 2003;138(7):1303–1312. doi: 10.1038/sj.bjp.0705182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Don AS, Martinez-Lamenca C, Webb WR, Proia RL, Roberts E, Rosen H. Essential requirement for sphingosine kinase 2 in a sphingolipid apoptosis pathway activated by FTY720 analogs. J. Biol. Chem. 2007;282:15833–15842. doi: 10.1074/jbc.M609124200. [DOI] [PubMed] [Google Scholar]
  • 43.Payne SG, Oskeritzian CA, Griffiths R, Subramanian P, Barbour SE, Chalfant CE, Milstien S, Spiegel S. The immunosuppressant drug FTY720 inhibits cytosolic phospholipase A2 independently of sphingosine-1-phosphate receptors. Blood. 2007;109:1077–1085. doi: 10.1182/blood-2006-03-011437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Neviani P, Santhanam R, Oaks JJ, Eiring AM, Notari M, Blaser BW, Liu S, Trotta R, Muthusamy N, Gambacorti-Passerini C, Druker BJ, Cortes J, Marcucci G, Chen CS, Verrills NM, Roy DC, Caligiuri MA, Bloomfield CD, Byrd JC, Perrotti D. FTY720, a new alternative for treating blast crisis chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphocytic leukemia. J. Clin. Invest. 2007;117(9):2408–2421. doi: 10.1172/JCI31095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Roberts KG, Smith AM, McDougall F, Carpenter H, Horan M, Neviani P, Powell JA, Thomas D, Guthridge MA, Perrotti D, Sim AT, Ashman LK, Verrills NM. Essential requirement for PP2A inhibition by the oncogenic receptor c-KIT suggests PP2A reactivation as a strategy to treat c-KIT+ cancers. Cancer Res. 2010;70(13):5438–5447. doi: 10.1158/0008-5472.CAN-09-2544. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES