Skip to main content

Sphingosine kinase 2 supports the development of BCR/ABL-independent acute lymphoblastic leukemia in mice

Abstract

Background

Sphingosine kinase (SphK) 2 has been implicated in the development of a range of cancers and inhibitors of this enzyme are currently in clinical trial. We have previously demonstrated a role for SphK2 in the development of acute lymphoblastic leukemia (ALL).

Methods

In this and our previous study we use mouse models: in the previous study the disease was driven by the proto-oncogene BCR/ABL1, while in this study cancer risk was elevated by deletion of the tumor suppressor ARF.

Results

Mice lacking ARF and SphK2 had a significantly reduced incidence of ALL compared mice with wild type SphK2.

Conclusions

These results show that the role of SphK2 in ALL development is not limited to BCR/ABL1 driven disease extending the potential use of inhibitors of this enzyme to ALL patients whose disease have driver mutations other than BCR/ABL1.

Background

There are two forms of sphingosine kinase (SphK), SphK1 and SphK2. SphK1 has an established role in malignant biology with overexpression being associated with poor survival in patients with solid tumors [1,2,3,4,5,6,7,8,9,10] and resistance to therapy [11,12,13,14]. Furthermore, inhibitors of SphK1 have demonstrated preclinical activity in acute myeloid leukemia (AML) [15, 16]. The role of SphK2 has been more controversial but it is increasingly being shown to play a role in malignant disease and has been associated with poor patient outcome [17]. Knockdown of SphK2 expression increases the sensitivity of cancer cells to chemotherapy [18,19,20], while chemical inhibition can reduce cancer cell growth in vitro [21,22,23,24,25,26,27,28] and in pre-clinical animal models [21, 24, 26]. SphK2 inhibitors are now in phase II clinical trials for a number of cancers including B cell malignancies, following successful completion of phase I studies [29]. We have recently shown that chemical inhibition of SphK2 can reduce acute lymphoblastic leukemia (ALL) cell growth, induce cell death in vitro and extend the survival of mice bearing human ALL xenografts. Furthermore, cells lacking SphK2 had a reduced capacity to induce ALL driven by the BCR/ABL1 fusion gene in WT mice, while SphK2 inhibition synergized with imatinib treatment of BCR/ABL1+ ALL in vitro and in vivo [30].

Mice deficient in the tumor suppressor gene ARF are prone to malignancies, with undifferentiated sarcomas predominating (~ 38%), followed by lymphomas (~ 23%), carcinomas (~ 15%) and neurological tumors (~ 10%), with a latency of around 266 days [31]. Genetic loss of material at the 9p21 locus, which includes ARF, is common in ALL, being reported in up to 45% of B lineage disease [32,33,34], making this a biologically relevant model. The development of tumors in these mice appears to be dependent on the aquisition of additional genetic changes as treatment with radiation or the mutagen DMBA significantly reduces latency. Here we show that blockade of T and B cell maturation by crossing ARF deficient mice onto a Rag1−/− background [35] resulted in an incidence of ALL of over 60%. Further crossing of these mice onto SphK2 deficient animals [36] permitted the examination of the role of SphK2 in the development of ALL, demonstrating a significant reduction in disease incidence.

Methods

Development of mouse model

Mice lacking the p19ARF product of the INK4a/ARF locus (ARF−/−) develop malignancies at a high penetrance with 80% of animals dying within the first year of life [31]. To facilitate breeding we used mice where the ARF gene had been floxed (ARFfl/fl) (B6.129-Cdkn2atm4Cjs/Nci, [37]) obtained from Graham Walker (QIMR, Queensland Australia). In order to produce an ALL model we crossed these mice with those lacking Rag1tm1Mom from The Jackson Laboratory (Bar Harbour, ME, USA) [35]. The resulting Mx1.Cre.ARFfl/fl.Rag1−/− (MAR) mice were then crossed onto animals lacking SphK2 (Richard Proia (Bethesda, USA) [36]) to produce Mx1.Cre.ARFfl/fl.Rag1−/−.SphK2−/− animals (MARS2 mice). The deletion of the ARF gene was undertaken at 6 weeks of age by intraperitoneal injection of 15 mg/kg of PolyI:polyC every second day for a total of 3 doses and confirmed by PCR (Additional file 1: Figure S1). All mice were obtained or were backcrossed onto on a C57Bl6 background. Experimental mice were monitored for up to 400 days. Mice were defined as having ALL when at the time of death the bone marrow and spleen primarily consisted of B220+CD19+Gr1− cells. Survival was analysed using the Kaplan-Meier method and SPSS Statistics software.

Mice were genotyped by PCR on genomic DNA obtained from ear punches using DirectPCR Lysis Reagent (Ear) (Viagen Biotech, Los Angeles CA) with 0.4 mg/mL proteinase K (Promega, Alexandria, NSW, Australia) (complete lysis solution). Ear punches from mice were incubated in complete lysis solution for 2 h at 56 °C and proteinase K was inactivated for 30 min at 85 °C prior to PCR. Deletion of ARF was detected in genomic DNA obtained from spleen cells recovered from culled mice. PCR reactions were performed using MyTaq DNA polymerase (Bioline, Eveleigh NSW Australia) and specific primers as indicated in Additional file 1: Table S1. The IL-2 PCR was used as a positive DNA control for the Mx1.Cre reaction. The PCR conditions were 95 °C for 1″, then 95 °C for 15″, 58 °C for 15″, 72 °C for 20″ for 35 cycles, 72 °C for 5′. Amplified products were separated on a 2% agarose (Sigma-Aldrich) gel stained with Midori Green Nucleic Acid solution (Bulldog Bio Inc., Portsmouth NH) and visualised using ChemiDoc MP Imaging System (Bio-Rad, Hercules, CA).

Flow cytometry

Flow cytometry was performed using a FACSCanto 6-colour flow cytometer (BD Biosciences, San Jose CA). The following antibodies were purchased: Sca-1-PE-Cy7, c-Kit-APC, CD43-APC, IgM-PCP.Cy5.5, IgM-Biotin (Australian Biosearch, WangarraWA, Australia); B220-APC.Cy7, B220 PE-Cy5, CD11b-PE, CD11b-FITC, CD19-PE, CD19-APC.Cy7, Gr1-FITC, Streptavidin APC and Lineage Cocktail of biotinylated CD3, Gr-1, Ter119, B220 and CD11b (BD Biosciences, San Jose CA), and Streptavidin Pacific Blue (Thermofisher Scientific, North Ryde, NSW, Australia). Cells were labelled with antibodies as previously described [30].

Histology and image acquisition

Blood films were prepared and stained with a Romanowsky stain. Tissues were fixed in 10% formalin, embedded, sectioned and stained as previously described [38]. Femurs were decalcified prior to embedding as previously described [38]. Images were obtained using a NanoZoomer Slide Scanner (SDR Scientific, Sydney Australia) or an Olympus BX51 microscope with images captured using a Spot RT slider camera (Diagnostic Instruments, Sterling Heights, MI) and SPOT Advanced software. Composite figures prepared using Adobe Photoshop software.

Results

Deletion of ARF in Rag1 deficient mice predisposes to ALL

Mice lacking ARF are known to develop malignancies with an increased incidence [31]. To generate an ALL model we bred Mx1.Cre.ARFfl/fl mice with Rag1−/− mice to generate Mx1.Cre.ARFfl/fl.Rag1−/− mice. At 6 weeks of age mice received 3 injections of polyI:polyC to delete the ARF gene producing Mx1.Cre.ARF−/−.Rag1−/− (MAR) mice.

Rag1−/− mice with deleted ARF (MAR mice) survived for up to 304 days (median 193 days) (Fig. 1a). The most common cause of death was B lineage ALL, which occurred in 61% of mice between 119 and 243 days with a median of 192 days. The remaining animals succumbed to a number of causes including other haematological malignancies, with the most common feature of non-ALL deaths being massively enlarged pale livers that sometimes contained defined tumors (Fig. 1b). However the origin of the tumors could not be determined with certainty. Many appeared to be haematological in origin based on morphology but the bone marrows mostly appeared normal (Additional file 1: Figure S2). Flow cytometric analysis of cells recovered from the bone marrow and spleens of these animals was generally uninformative.

Fig. 1
figure 1

MAR mice develop malignancies with B lineage ALL predominating. a Kalpan-Meyer analysis showing the survival of MAR mice. b Mouse culled due to disease other than ALL showing tumors in the liver (white arrows) and an enlarged spleen (black arrow). c Mouse culled due to ALL showing enlarged spleen (black arrow). d Blood film from a mouse with ALL showing circulating lymphoblasts. Image acquired using a slide scanner and size bar represents 100 μm. Lower imaged taken on a Spot camera, original magnification 600×. e Flow cytometric analysis of bone marrow and spleen cells from mice culled due to ALL. Upper panels are from the same mouse. Central panels show the lowest and highest CD11b expression detected. Lower panels show typical expression of maturation markers B220, CD19, CD43 and surface IgM. Quadrants were set based on control stained cells from the same animal. f Section of liver from a mouse culled due to ALL showing both perivascular (thin arrow) and diffuse (thick arrow) infiltration by ALL cells. The degree of infiltration in this animal was typical. Image acquired using slide scanner and size bar indicates 250 μm

Mice that developed ALL were easily identified, demonstrating weight loss, reduced activity and/or impaired use of hind limbs and tail. One displayed hydrocephaly, with fitting. Necropsy findings were consistent with B lineage ALL with enlarged spleens and often enlarged livers, without evidence of tumors and a normal dark red colour (Fig. 1c). Mice with ALL also had elevated WBC for immune-compromised mice (median 15.2, range 2.1-286.5 cells/mL) with significant numbers of lymphoblasts present in blood smears (Fig. 1d). Lymph nodes were rarely involved with only 2 mice having visible nodes on cull and only 1 of those having significant lymphadenopathy (Additional file 1: Figure S3). Cells in the spleen and bone marrow were mostly B220 and CD19 positive (average of 73%, range 56-87 and 86%, range 73-97 respectively), lacking staining for the myeloid marker Gr1 and the T cell marker CD3, however CD11b was detected on cells from some animals (Fig. 1e). Cells from all mice with ALL were positive for immature marker CD43 and most expressed IgM on at least a proportion of the cells (Fig. 1e). The lack of lymph node involvement in the vast majority of animals, near complete replacement of the bone marrow by lymphoblasts as well as the expression of the immature marker CD43 and low expression of IgM indicate a pro- to pre-B classification of these leukemias. Other organs, primarily the liver, were infiltrated with lymphoblasts (Fig. 1f). ALL induced death tended to be earlier compared to non-ALL deaths, with the latter occurring between 68 and 304 days with a median of 229 days, although this was not statistically significant, p = 0.06) (Additional file 1: Figure S4). Animals that did not develop ALL mostly presented with solid tumors at a slightly later time point.

Deletion of SphK2 reduced the incidence of B ALL

A cohort of mice lacking ARF and Rag1 was also generated using the same methodology on an SphK2−/− background (MARS2 mice). ARF was similarly deleted at 6 weeks of age by 3 injections of polyI:polyC. These mice also largely succumbed to conditions consistent with malignant diseases but compared to MAR mice had significantly increased overall survival with deaths occurring between 120 and > 400 days (one mouse was electively culled disease free at 400 days) with a median of 234 days (p < 0.05) (Fig. 2a). Notably there were fewer deaths resulting from ALL in MARS2 animals with only 43% of deaths being due to ALL, resulting in a significant increase in leukemia free survival in MARS2 mice (p = 0.044) (Fig. 2b).

Fig. 2
figure 2

Loss of SphK2 reduces the incidence of B lineage ALL. a-c Kaplan-Meier plots showing all (a) and ALL-induced (b) deaths. Deaths due to causes other than ALL are illustrated in (c). Total WBC (d, left panel) and ALL blast counts (d, right panel) at the time of sacrifice are shown. # indicates p < 0.05. e Mouse culled due to ALL showing enlarged spleen (black arrow). f Blood film from a mouse with ALL showing circulating lymphoblasts. Image acquired using a slide scanner and size bar represents 100 μm. g Section of liver from a mouse culled due to ALL showing both perivascular (thin arrow) and diffuse (thick arrow) infiltration by ALL cells. The degree of infiltration in this animal was typical. Image acquired using slide scanner and size bar indicates 250 μm

The absence of SphK2 did not alter the nature of the ALL that developed, with latency, phenotype and disease dissemination being similar. Death due to ALL was slightly delayed in MARS2 mice (range 169 – 253, median 219.5 days), however this was not significantly different from that of MAR mice (Fig. 2c). Interestingly the WBC in the leukemic MARS2 mice was significantly lower than in the MAR mice, as was the number of circulating blasts (Fig. 2d), however the blast percentage was similar between the two groups. Otherwise the disease was identical in MARS2 and MAR mice, with similar enlargement of spleen and liver and infiltration of other organs (Fig. 2e–g).

Discussion

Inhibition of sphingosine kinases has recently become of interest for the treatment of a number of conditions including malignant disease [39]. Clinical trials for the SphK2 inhibitor ABC294640, are well under way with phase I studies complete [29] and phase I/II and phase II trials examining hepatocellular carcinoma, Kaposi sarcoma as well as the haematological malignancies multiple myeloma and diffuse large B cell lymphoma ongoing (NCT02229981, NCT02939807 and NCT02757326). These trials have been supported by recent preclinical data from a number of groups [23, 24, 26, 30, 40,41,42,43,44]. The majority of these studies have focussed on solid tumors, however there are reports in haematological malignancies including multiple myeloma [26] and T-ALL [45], and we have previously reported a role for SphK2 in B lineage ALL [30] using a BCR/ABL1-dependent model. In this study, we examined the effects of SphK2 gene deletion on the development of ALL in a model that is not dependent on forced expression of BCR/ABL1 and demonstrated that genetic deletion of SphK2 also inhibits the development of B lineage ALL independent of BCR/ABL1 expression. The similar latency and features of the disease in MAR and MARS2 mice suggests that the principal effect of SphK2 loss was on leukemia initiation rather than rate of disease progression. However, we previously demonstrated that the SphK2 inhibitor ABC294640 impedes disease progression in a xenograft model of Ph− human ALL, suggesting that SphK2 loss/inhibition has some effect on disease progression [30].

The reason why loss of SphK2 decreases the incidence of ALL is not entirely clear. However SphK2 has a well-established role in promoting malignant cell survival [46] making it possible that in the absence of SphK2, cells with newly acquired potentially oncogenic changes are more susceptible to cell death. While precise mechanisms are yet to be determined, one potential explanation relates to CDKN1A expression. CDKN1A is an inhibitor of apoptosis induced in response to DNA damage whose expression is increased by SphK2-mediated effects on histone acetylation [47]. In the absence of SphK2, induction of CDKN1A expression following DNA damage could be reduced increasing the probability of cell death. Another possible mechanism relating loss of SphK2 to the reduced incidence of ALL concerns the localization of SphK2 to the endoplasmic reticulum (ER) membrane and its involvement in sphingolipid metabolism at this site. We have recently demonstrated that inhibition of SphK2 induces unrecoverable ER stress leading to apoptosis of multiple myeloma cells and this ER stress-inducing mechanism is most likely also applicable to a range of cell types, including those of ALL, thus impacting on its development in our model [48].

The lower WBC in leukemic MARS2 was interesting and although altered trafficking of lymphoid cells in SphK2−/− animals might be an explanation for this observation, previous reports have demonstrated increased plasma sphingosine-1-phosphate (S1P) and resultant increased lymphocyte mobilization in SphK2−/− mice [49]. All but one MARS2 mouse that did not develop ALL went on to develop solid tumors at a time closer to the previously reported latency (median of 266 days) for solid tumors in ARF deficient animals [31]. Since the tumors that emerged in this study could not be definitively classified, it is not possible to comment on the effects of SphK2 loss on the development of other malignancies.

Conclusions

We have previously demonstrated the role of SphK2 in ALL driven by BCR/ABL1 and the potential therapeutic application of SphK2 inhibitors in this disease. In this study we demonstrate that SphK2 also plays a role in the development of BCR/ABL1 negative ALL with genetic deletion of SphK2 reducing disease incidence. These findings further support and broaden the potential application of SphK2 inhibitors in the treatment of ALL.

Abbreviations

ALL:

Acute lymphoblastic leukemia

AML:

Acute myeloid leukemia

ARF−/− :

Mice lacking the p19ARF product of the INK4a/ARF locus

ARFfl/fl :

Mice where the ARF gene had been floxed

ER:

Endoplasmic reticulum

MAR:

Mx1.Cre.ARFfl/fl.Rag1−/−

MARS2:

Mx1.Cre.ARFfl/fl.Rag1−/−.SphK2−/−

S1P:

Sphingosine-1-phosphate

SphK:

Sphingosine kinase

References

  1. Cai H, Xie X, Ji L, Ruan X, Zheng Z. Sphingosine kinase 1: a novel independent prognosis biomarker in hepatocellular carcinoma. Oncol Lett. 2017;13(4):2316–22.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Chen MH, Yen CC, Cheng CT, Wu RC, Huang SC, Yu CS, et al. Identification of SPHK1 as a therapeutic target and marker of poor prognosis in cholangiocarcinoma. Oncotarget. 2015;6(27):23594–608.

    PubMed  PubMed Central  Google Scholar 

  3. Facchinetti MM, Gandini NA, Fermento ME, Sterin-Speziale NB, Ji Y, Patel V, et al. The expression of sphingosine kinase-1 in head and neck carcinoma. Cells Tissues Organs. 2010;192(5):314–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kim HS, Yoon G, Ryu JY, Cho YJ, Choi JJ, Lee YY, et al. Sphingosine kinase 1 is a reliable prognostic factor and a novel therapeutic target for uterine cervical cancer. Oncotarget. 2015;6(29):26746–56.

    PubMed  PubMed Central  Google Scholar 

  5. Li W, Yu CP, Xia JT, Zhang L, Weng GX, Zheng HQ, et al. Sphingosine kinase 1 is associated with gastric cancer progression and poor survival of patients. Clin Cancer Res. 2009;15(4):1393–9.

    Article  CAS  PubMed  Google Scholar 

  6. Liu G, Zheng H, Zhang Z, Wu Z, Xiong H, Li J, et al. Overexpression of sphingosine kinase 1 is associated with salivary gland carcinoma progression and might be a novel predictive marker for adjuvant therapy. BMC Cancer. 2010;10:495.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Meng XD, Zhou ZS, Qiu JH, Shen WH, Wu Q, Xiao J. Increased SPHK1 expression is associated with poor prognosis in bladder cancer. Tumour Biol. 2014;35(3):2075–80.

    Article  CAS  PubMed  Google Scholar 

  8. Li J, Guan HY, Gong LY, Song LB, Zhang N, Wu J, et al. Clinical significance of sphingosine kinase-1 expression in human astrocytomas progression and overall patient survival. Clin Cancer Res. 2008;14(21):6996–7003.

    Article  CAS  PubMed  Google Scholar 

  9. Ruckhaberle E, Rody A, Engels K, Gaetje R, von Minckwitz G, Schiffmann S, et al. Microarray analysis of altered sphingolipid metabolism reveals prognostic significance of sphingosine kinase 1 in breast cancer. Breast Cancer Res Treat. 2008;112(1):41–52.

    Article  PubMed  Google Scholar 

  10. Van Brocklyn JR, Jackson CA, Pearl DK, Kotur MS, Snyder PJ, Prior TW. Sphingosine kinase-1 expression correlates with poor survival of patients with glioblastoma multiforme: roles of sphingosine kinase isoforms in growth of glioblastoma cell lines. J Neuropathol Exp Neurol. 2005;64(8):695–705.

    Article  CAS  PubMed  Google Scholar 

  11. Salas A, Ponnusamy S, Senkal CE, Meyers-Needham M, Selvam SP, Saddoughi SA, 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):5941–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bonhoure E, Lauret A, Barnes DJ, Martin C, Malavaud B, Kohama T, et al. Sphingosine kinase-1 is a downstream regulator of imatinib-induced apoptosis in chronic myeloid leukemia cells. Leukemia. 2008;22(5):971–9.

    Article  CAS  PubMed  Google Scholar 

  13. Baran Y, Salas A, Senkal CE, Gunduz U, Bielawski J, Obeid LM, et al. 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):10922–34.

    Article  CAS  PubMed  Google Scholar 

  14. Sobue S, Nemoto S, Murakami M, Ito H, Kimura A, Gao S, et al. 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):266–75.

    Article  CAS  PubMed  Google Scholar 

  15. Paugh SW, Paugh BS, Rahmani M, Kapitonov D, Almenara JA, Kordula T, et al. A selective sphingosine kinase 1 inhibitor integrates multiple molecular therapeutic targets in human leukemia. Blood. 2008;112(4):1382–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Powell JA, Lewis AC, Zhu W, Toubia J, Pitman MR, Wallington-Beddoe CT, et al. Targeting sphingosine kinase 1 induces MCL1-dependent cell death in acute myeloid. Leukemia. 2017;129(6):771–82.

    CAS  Google Scholar 

  17. Wang Q, Li J, Li G, Li Y, Xu C, Li M, et al. Prognostic significance of sphingosine kinase 2 expression in non-small cell lung cancer. Tumour Biol. 2014;35(1):363–8.

    Article  CAS  PubMed  Google Scholar 

  18. Sankala HM, Hait NC, Paugh SW, Shida D, Lepine S, Elmore LW, et al. Involvement of sphingosine kinase 2 in p53-independent induction of p21 by the chemotherapeutic drug doxorubicin. Cancer Res. 2007;67(21):10466–74.

    Article  CAS  PubMed  Google Scholar 

  19. Nemoto S, Nakamura M, Osawa Y, Kono S, Itoh Y, Okano Y, et al. Sphingosine kinase isoforms regulate oxaliplatin sensitivity of human colon cancer cells through ceramide accumulation and Akt activation. J Biol Chem. 2009;284(16):10422–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Schnitzer SE, Weigert A, Zhou J, Brune B. Hypoxia enhances sphingosine kinase 2 activity and provokes sphingosine-1-phosphate-mediated chemoresistance in A549 lung cancer cells. Mol Cancer Res. 2009;7(3):393–401.

    Article  CAS  PubMed  Google Scholar 

  21. Weigert A, Schiffmann S, Sekar D, Ley S, Menrad H, Werno C, et al. Sphingosine kinase 2 deficient tumor xenografts show impaired growth and fail to polarize macrophages towards an anti-inflammatory phenotype. Int J Cancer. 2009;125(9):2114–21.

    Article  CAS  PubMed  Google Scholar 

  22. Beljanski V, Knaak C, Smith CD. A novel sphingosine kinase inhibitor induces autophagy in tumor cells. J Pharmacol Exp Ther. 2010;333(2):454–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. French KJ, Zhuang Y, Maines LW, Gao P, Wang W, Beljanski V, et al. Pharmacology and antitumor activity of ABC294640, a selective inhibitor of sphingosine kinase-2. J Pharmacol Exp Ther. 2010;333(1):129–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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):524–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. White MD, Chan L, Antoon JW, Beckman BS. Targeting ovarian cancer and chemoresistance through selective inhibition of sphingosine kinase-2 with ABC294640. Anticancer Res. 2013;33(9):3573–9.

    CAS  PubMed  Google Scholar 

  26. Venkata JK, An N, Stuart R, Costa LJ, Cai H, Coker W, et al. Inhibition of sphingosine kinase 2 downregulates the expression of c-Myc and Mcl-1 and induces apoptosis in multiple myeloma. Blood. 2014;124(12):1915–25.

    Article  CAS  PubMed  Google Scholar 

  27. Chu JH, Gao ZH, Qu XJ. Down-regulation of sphingosine kinase 2 (SphK2) increases the effects of all-trans-retinoic acid (ATRA) on colon cancer cells. Biomed Pharmacother. 2014;68(8):1089–97.

    Article  CAS  PubMed  Google Scholar 

  28. Yang J, Yang C, Zhang S, Mei Z, Shi M, Sun S, et al. ABC294640, a sphingosine kinase 2 inhibitor, enhances the antitumor effects of TRAIL in non-small cell lung cancer. Cancer Biol Ther. 2015;16(8):1194–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Britten CD, Garrett-Mayer E, Chin SH, Shirai K, Ogretmen B, Bentz TA, et al. A phase I study of ABC294640, a first-in-class sphingosine kinase-2 inhibitor, in patients with advanced solid tumors. Clin Cancer Res. 2017;23(16):4642–50.

    Article  CAS  PubMed  Google Scholar 

  30. Wallington-Beddoe CT, Powell JA, Tong D, Pitson SM, Bradstock KF, Bendall LJ. Sphingosine kinase 2 promotes acute lymphoblastic leukemia by enhancing MYC expression. Cancer Res. 2014;74(10):2803–15.

    Article  CAS  PubMed  Google Scholar 

  31. Kamijo T, Bodner S, van de Kamp E, Randle DH, Sherr CJ. Tumor spectrum in ARF-deficient mice. Cancer Res. 1999;59(9):2217–22.

    CAS  PubMed  Google Scholar 

  32. Faderl S, Estrov Z, Kantarjian HM, Thomas D, Cortes J, Manshouri T, et al. The incidence of chromosome 9p21 abnormalities and deletions of tumor suppressor genes p15(INK4b)/p16(INK4a)/p14(ARF) in patients with acute lymphoblastic leukemia. Cytokines Cell Mol Ther. 1999;5(3):159–63.

    CAS  PubMed  Google Scholar 

  33. Bertin R, Acquaviva C, Mirebeau D, Guidal-Giroux C, Vilmer E, Cave H. CDKN2A, CDKN2B, and MTAP gene dosage permits precise characterization of mono- and bi-allelic 9p21 deletions in childhood acute lymphoblastic leukemia. Genes Chromosomes Cancer. 2003;37(1):44–57.

    Article  CAS  PubMed  Google Scholar 

  34. Gardiner RB, Morash BA, Riddell C, Wang H, Fernandez CV, Yhap M, et al. Using MS-MLPA as an efficient screening tool for detecting 9p21 abnormalities in pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer. 2012;58(6):852–9.

    Article  PubMed  Google Scholar 

  35. Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S, Papaioannou VE. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 1992;68(5):869–77.

    Article  CAS  PubMed  Google Scholar 

  36. Mizugishi K, Yamashita T, Olivera A, Miller GF, Spiegel S, Proia RL. Essential role for sphingosine kinases in neural and vascular development. Mol Cell Biol. 2005;25(24):11113–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gromley A, Churchman ML, Zindy F, Sherr CJ. Transient expression of the Arf tumor suppressor during male germ cell and eye development in Arf-Cre reporter mice. Proc Natl Acad Sci U S A. 2009;106(15):6285–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Crazzolara R, Cisterne A, Thien M, Hewson J, Baraz R, Bradstock KF, et al. Potentiating effects of RAD001 (Everolimus) on vincristine therapy in childhood acute lymphoblastic leukemia. Blood. 2009;113(14):3297–306.

    Article  CAS  PubMed  Google Scholar 

  39. Pyne S, Adams DR, Pyne NJ. Sphingosine 1-phosphate and sphingosine kinases in health and disease: recent advances. Prog Lipid Res. 2016;62:93–106.

    Article  CAS  PubMed  Google Scholar 

  40. Antoon JW, White MD, Driver JL, Burow ME, Beckman BS. Sphingosine kinase isoforms as a therapeutic target in endocrine therapy resistant luminal and basal-A breast cancer. Exp Biol Med (Maywood). 2012;237(7):832–44.

    Article  CAS  Google Scholar 

  41. Liu K, Guo TL, Hait NC, Allegood J, Parikh HI, Xu W, et al. Biological characterization of 3-(2-amino-ethyl)-5-[3-(4-butoxyl-phenyl)-propylidene]-thiazolidine-2,4-dione (K145) as a selective sphingosine kinase-2 inhibitor and anticancer agent. PLoS One. 2013;8(2):e56471.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Xun C, Chen MB, Qi L, Tie-Ning Z, Peng X, Ning L, et al. Targeting sphingosine kinase 2 (SphK2) by ABC294640 inhibits colorectal cancer cell growth in vitro and in vivo. J Exp Clin Cancer Res. 2015;34:94.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Schrecengost RS, Keller SN, Schiewer MJ, Knudsen KE, Smith CD. Downregulation of critical oncogenes by the selective SK2 inhibitor ABC294640 hinders prostate cancer progression. Mol Cancer Res. 2015;13(12):1591–601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Venant H, Rahmaniyan M, Jones EE, Lu P, Lilly MB, Garrett-Mayer E, et al. The sphingosine kinase 2 inhibitor ABC294640 reduces the growth of prostate cancer cells and results in accumulation of dihydroceramides in vitro and in vivo. Mol Cancer Ther. 2015;14(12):2744–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Evangelisti C, Evangelisti C, Teti G, Chiarini F, Falconi M, Melchionda F, 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):7886–901.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Wallington-Beddoe CT, Bradstock KF, Bendall LJ. Oncogenic properties of sphingosine kinases in haematological malignancies. Br J Haematol. 2013;161(5):623–38.

    Article  CAS  PubMed  Google Scholar 

  47. Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, et al. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science. 2009;325(5945):1254–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wallington-Beddoe CT, Bennett MK, Vandyke K, Davies L, Zebol JR, Moretti PAB, et al. Sphingosine kinase 2 inhibition synergises with bortezomib to target myeloma by enhancing endoplasmic reticulum stress. Oncotarget. 2017;8(27):43602–16.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Adamiak M, Chelvarajan L, Lynch K, Santos W, Abdel-Latif A, Ratajczak M. Mobilization studies in mice deficient in sphingosine kinase 2 support a crucial role of the plasma level of sphingosine-1-phosphate in the egress of hematopoietic stem progenitor cells. Oncotarget. 2017;8(39):65588–600.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Flow cytometry was performed in the Flow Cytometry Core Facility that is supported by Westmead Institute for Medical Research, Westmead Research Hub, Cancer Institute New South Wales and National Health and Medical Research Council. Histology was performed in the Histology Platform at Westmead Institute for Medical Research with the assistance of Virginia James.

Funding

This work was supported by an NHMRC Senior Research Fellowship (1042305) and Cancer Institute NSW Fellowship.

Availability of data and materials

All data generated or analysed during this study are included in this published article [and its supplementary information files]. The mice are available through Australian BioResources as cryopreserved embryos.

Author information

Authors and Affiliations

Authors

Contributions

LB, CW-B and KB made substantial contributions to conception and design of the study. DT, LB and CW-B designed the breeding strategies required for the development of the animals used in this study. VX and LB were responsible for data acquisition, analysis and interpretation of data. LB drafted the manuscript and all authors made significant contributions to revising the final document. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Linda J. Bendall.

Ethics declarations

Ethics approval and consent to participate

The experiments reported here were conducted with the approval of the Animal Ethics Committee of the Western Sydney Local Health District - approval number 5107.

Consent for publication

Not Applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Additional file

Additional file 1:

Additional Data: Table S1, Figures S1-4. (DOCX 6406 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xie, V., Tong, D., Wallington-Beddoe, C.T. et al. Sphingosine kinase 2 supports the development of BCR/ABL-independent acute lymphoblastic leukemia in mice. Biomark Res 6, 6 (2018). https://doi.org/10.1186/s40364-018-0120-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-018-0120-4

Keywords