Skip to main content
Cancer Control: Journal of the Moffitt Cancer Center logoLink to Cancer Control: Journal of the Moffitt Cancer Center
. 2020 Dec 14;27(1):1073274820976664. doi: 10.1177/1073274820976664

The Tumorigenic Effect of Sphingosine Kinase 1 and Its Potential Therapeutic Target

Xianwang Wang 1,2,, Yong Sun 1, Xiaochun Peng 2, Syed Manzar Abbas Shah Naqvi 2, Yue Yang 1, Jing Zhang 1, Meiwen Chen 2, Yuan Chen 2, Hongyue Chen 2, Huizi Yan 2, Guangliang Wei 2, Peng Hong 3, Yingying Lu 3,
PMCID: PMC8480355  PMID: 33317322

Abstract

Sphingosine kinase 1 (SPHK1) regulates cell proliferation and survival by converting sphingosine to the signaling mediator sphingosine 1-phosphate (S1P). SPHK1 is widely overexpressed in most cancers, promoting tumor progression and is associated with clinical prognosis. Numerous studies have explored SPHK1 as a promising target for cancer therapy. However, due to insufficient knowledge of SPHK1 oncogenic mechanisms, its inhibitors’ therapeutic potential in preventing and treating cancer still needs further investigation. In this review, we summarized the metabolic balance regulated by the SPHK1/S1P signaling pathway and highlighted the oncogenic mechanisms of SPHK1 via the upregulation of autophagy, proliferation, and survival, migration, angiogenesis and inflammation, and inhibition of apoptosis. Drug candidates targeting SPHK1 were also discussed at the end. This review provides new insights into the oncogenic effect of SPHK1 and sheds light on the future direction for targeting SPHK1 as cancer therapy.

Keywords: sphingosine kinase 1 (SPHK1/SK1/SPK1), oncogenic mechanisms, therapeutic inhibitors, S1P, cancer therapy

Introduction

Sphingosine kinase 1(SPHK1/SK1/SPK1) mediates the conversion of sphingosine (Sph) to sphingosine-1-phosphate (S1P), a pivotal sphingolipid signaling mediator involved in a wide variety of cellular processes, including cell growth, survival, differentiation, and motility.1,2 The activation of SPHK1 requires 2 events: phosphorylated by extracellular signal-regulated kinase 1/2 (ERK1/2) and translocation to plasmalemma.3,4 Two mammalian isoenzymes have been identified, SPHK1 and SPHK2.5,6 SPHK1 localizes predominantly in the cytosol as it contains a functional nuclear export sequence, whereas SPHK2 locates both in the nuclei and the cytoplasm, indicating their distinct biological roles.7 Since the first human SPHK1 structure was illustrated,8 significant advances have been made in understanding the composition and function of SPHK1.9 Human SPHK1 gene is localized to 17q25.2, and its protein products have 3 splice isoforms, SPHK1a, SPHK1b, and SPHK1c. SPHK1a is primarily involved in the extracellular signaling transduction, while SPHK1b and SPHK1c are mainly anchored on the cell membrane.10 The structure of SPHK1 adopts a 2-domain architecture that comprises 9 α helices, 17 β strands, and a 310-helix (Figure 1).8,11 Each motif has its specific binding sites associated closely with its functions, such as active sites, nucleotide-binding sites, calcium/calmodulin(Ca2+/CaM) coupling sites, magnesium ion combining sites, lipid associating sites, phosphorylation/dephosphorylation sites, etc. (Table 1). Although the structure has been characterized,8,9 molecular mechanisms of SPHK1 functions, such as its translocation activation, and interactions between 3 various isoforms and other molecules, are poorly understood. SPHK1 is a perplexing kinase with pivotal roles in various cellular processes (Figure 2). Cumulating evidence has revealed that SPHK1 is upregulated in cancer cells and closely correlated with tumors progression16-19 (Table 2), and high expression level or kinase activity of SPHK1 is associated with a poor prognosis in several types of cancers. Recently, oncogenic mechanisms of SPHK1 have been identified in multiple aspects, including tumor growth, tumor migration and angiogenesis, and so on. This review introduced the SPHK1/S1P signaling pathway and discussed the potential mechanism of SPHK1/S1P signaling in tumor progression. Besides, inhibitors of SPHK1 with therapeutic potentials were also summarized.

Figure 1.

Figure 1.

The basic structure of SPHK1. SPHK1 consists of NTD and CTD, 5 conserved motifs, C1-C5. C1-C3 involved in NTD, while C4-C5 in CTD (C1:19-36aa, C2:72-96aa, C3108-119aa, C4:165-198aa, C5:338-343aa). Every domain has specific motifs, 9 α helices, 17 β strands, and a 310-helix.

Table 1.

The Specific Binding Sites of SPHK1.a

Sites Position(s) Description/Functions References
Active site 81 Proton donor/acceptor, as a catalytically critical residue 8
Necleotide binding 17∼24, 54∼58, 79∼103
111∼113, 178,185,191,
341∼343
ATP and TRAf2 binding 11,12
Ca2+/CaM binding* Between 134 ∼153,290∼303
191-206
Acting as a cellular calcium sensor for SPHK1;
(197 and 198 are essential)
13
14
Magnesium ion binding 143, 343 Coordinating to the γ-phosphate group of ATP and the pyrophosphate moiety 13
Lipid biding β8∼β9, β10∼β11, β11∼β12 Binding with Sph 8,9,11
α7∼α8 Acting like a lipid gate controlling the in-and-out of lipid substrate and product
ERK1/2 225 Phosphorylation to SPHK1 15
PKC* 54,181,205,371 Phosphorylation to SPHK1 13
PP2A-B’α 225 Dephosphorylation to SPHK1 14

a SPHK1 contains multiple sites, such as active site (which is essential for the function of this enzyme), nucleotide-binding sites (bind ATP, as a substrate to phosphorylate Sph), Ca2+/CaM binding sites (bind Ca2+/CaM or act as a cellular calcium sensor for SPHK1), magnesium ion binding sites (coordinate to the γ-phosphate group of ATP and the pyrophosphate moiety), lipid biding sites (bind with Sph and act like a lipid gate controlling the in-and-out of lipid substrate and product), ERK1/2 and PKC phosphorylation sites (phosphorylate SPHK1) and PP2A-B’α dephosphorylation site (dephosphorylate SPHK1). *: Direct activation of the enzyme by Ca2+ and PKC has not been demonstrated so far.

Figure 2.

Figure 2.

The schematic of the SPHK1/S1P signaling pathway. SPHK1 catalyzes the formation of S1P from Sph and is pivotal regulators of the balance between Cer, Sph, and S1P. The gene expression of SPHK1 is regulated by a transcription factor (TF), such as transcription factor specificity protein 1 (Sp1), AP2, E2F transcription factor, hypoxia inducible factors (HIF), LIM-domain-only protein 2 (LMO2). After expression in the cytoplasm, SPHK1 is activated by ERK1/2, resulting in its phosphorylation and translocation to the cell membrane; CIB1 participates in this process. When SPHK1 is fitted on the cell membrane, it will catalyze the production of S1P, and the yielded S1P is secreted to the extracellular domain via the S1P channels (ABCA1/B1/C1, Spns2), and then the extracellular S1P engaged with the receptors (S1PR1-5) of other cells or itself cell membrane. Owning to S1PR is a G-protein coupled receptor (GPCR) that could transmit extracellular signaling into intracellular second message S1P, initiates the classical GPCR signaling pathway, and elicits serious effects, including cell metabolism, proliferation, migration, angiogenesis, autophagy, apoptosis, inflammation, etc.

Table 2.

The Expression and Potential Mechanism of SPHK1 in Human Cancers.a

Cancer types Expression Potential mechanism Related molecules References
Breast cancer ≥2.0folds Proliferation↑ˎ Angiogenesis↑ˎ Migration↑ˎ Apoptosis↓ EGF  ˎ ERK1/2  ˎ HDAC  ˎ RAS  ˎ JAK2  ˎ RTKs  ˎ Ca2+  ˎ MAPK   ˎ AKT  ˎ PI3K  ˎ ER  ˎ Wnt  ˎ miR-515-5p  ˎ NF-κB/FSCN1  ˎ SNAI2 20,21,22,23,24
Lung cancer ≥2.0 folds Innovation↑ˎ Migration↑ˎ Proliferation↑ EMT↑ˎ Apoptosis↓ˎ Metabolism↑ AKT  ˎ miRNA  ˎ Spns2  ˎ GSK-3β  ˎ STAT3  ˎ LncRNA HULC
25,26,27
Uterine cancer Apoptosis↓ˎ Invasion↑ˎ Proliferation↑ˎ Migration↑ˎ Angiogenesis↑ MMP-2  ˎ VEGF-A  ˎ PKC  ˎ ABCC1  ˎ COX2  ˎ ERK 28,29
Ovarian cancer ≥2.0 folds Angiogenesis↑ˎ Apoptosis↓ˎ Migration↑ˎ Invasion↑ VEGF, IL-8  ˎ IL-6  ˎ S1PR1/3  ˎ FMMP   ˎ CIB2  ˎ TGF-β  ˎ p38 MAPK  ˎ HIF1α and HIF2α 30,31,32,33,34
Liver cancer EMT↑ˎ Autophagy↑ˎ Apoptosis↓ˎ Proliferation↑ˎ Angiogenesis↑ CDH1  ˎ TRAF2  ˎ BECN1  ˎ JNK1  ˎ miR-506 35,36,37
Prostate cancer Invasion and metastasis↑
Proliferation↑
ROS  ˎ HIF-1α  ˎ VEGF  ˎ TP53  ˎ P21 38,39,40
Colon/colorectal cancer EMT↑ˎ Proliferation↑ˎ Migration↑ˎ Invasion↑ˎ Autophagy↑ˎ Apoptosis↓ ERK/12  ˎ Ras  ˎ miRNA  ˎ AKT  ˎ NF-κB  ˎ CD44  ˎ LncRNA MEG3  ˎ FAK 41,42,43,44,45
Kidney cancer 2.7 folds Innovation↑ˎ Angiogenesis↑ FAK  ˎ S1PR1/3  ˎ HIF-2α 46
Gastric cancer ≥ 2.0 folds Apoptosis↓ˎ Angiogenesis↑ˎ Invasion↑ˎ Migration↑ Bim  ˎ AKT  ˎ FOXO3a  ˎ BCL-2  ˎ ERK/12  ˎ STAT3  ˎ miR-330-3p  ˎ NF-κB 47,48
Chronic myeloid leukemia Proliferation↑ˎ Apoptosis↓ miR-659-3p  ˎ PI3K  ˎ AKT2  ˎ mTOR 49,50
Glioma ≥1.0 folds Angiogenesis↑ˎ Proliferation↑ˎ Invasion↑ˎ Migration↑ Ca2+  ˎ EGFR  ˎ S1PR1/2/3/5 51,52,53
Multiple myelomas Apoptosis↓ˎ Proliferation↑ EGCG   ˎ 67LR  ˎ RTKs  ˎ AKT  ˎ PI3K  ˎ IGF-1R 54
Osteosarcoma Autophagy↓ miR-506-3p 55
Thyroid cancer Apoptosis↓
Proliferation↑
Metastasis↑
miR-128 56
Clear cell renal cell carcinoma Proliferation↑
Migration↑
Akt/mTOR 19
Primary effusion lymphoma Apoptosis↓ ABC  ˎ NF-κB  ˎ MAPK 57

a SPHK1 is elevated in many tumors, mostly more than twice that of normal cells (non-tumor cells). And lots of associated molecules participate in the process of cancer. EMT: epithelial-mesenchymal transition; 67LR: 67-kDa laminin receptors; ER: estrogen receptor; TRAF2: TNF-receptor-associated factor.

SPHK1/S1P/S1PR Signaling Pathway

SPHK1 is one of the versatile kinases that catalyze the synthesis of S1P, and it performs an increasingly essential role in regulating the metabolic balance of sphingolipids such as ceramide (Cer), sphingosine (Sph), and S1P.58 Cer plays a vital role in apoptosis, cell senescence, differentiation, and cellular stress responses,58-60 and Sph is an anti-growth signaling molecule.61 In contrast, S1P promotes cell proliferation, survival and inhibits apoptosis.62-64 The SPHK1/S1P signaling plays a crucial role in inflammation, cell migration, and vascular development,16 which has been inextricably linked to tumorigenesis. SPHK1 also acts on D-erythro-sphingosine in vitro 8 and, to a lesser extent, sphinganine65 through phosphorylation. Moreover, SPHK1 has serine acetyltransferase activity on cyclooxygenase 2 (COX2) in an acetyl-CoA dependent manner, which contributes to pathogenesis in a model of Alzheimer’s disease (AD).66

SPHK1 has been identified as an important regulator in regulating extracellular and intracellular S1P levels (Figure 2). In normal conditions, SPHK1 is a predominantly cytosolic enzyme.9 The expression of SPHK1 is regulated by the transcription factors (TF), such as transcription factor specificity protein 1 (Sp1), AP2, E2F transcription factor, hypoxia-inducible factors (HIF), LIM-domain-only protein 2 (LMO2)4 (Figure 2). When exposure to several stimuli, such as growth factors, cytokines, and hormones, SPHK1 is activated and translocated to the cell membrane, catalyzes the production of S1P, thereby raising S1P contents.67-69 As a second messager, S1P is a biologically active lipid catalyzed by either SPHK1 or SPHK2, which can be released from cells via specific channels such as ATP-binding cassette (ABC-A1/B1/C1), major facilitator superfamily transporter 2b (Mfsd2b), and spinster homolog 2 (Spns2)70-72 to target to the G protein-coupled receptor family (S1PR1-5) proteins.10 In the cytoplasm, S1P can also couple with specific partners, such as histone deacetylases (HDAC1/2), human telomerase reverse transcriptase (hTERT), TNF-receptor-associated factor(TRAF2)73-75 and prohibitin 2 (PHB2),76 to trigger a variety of cellular responses.

On the other hand, the activity of SPHK1 is also crucial for the clearance of sphingolipids. Once S1P comes into being, S1P is hydrolyzed by S1P lyase in an irreversible reaction to yield hexadecenal and phosphoethanolamine (Figure 2), which is an important exit of sphingolipid metabolic cycle.58 The oncogenic signaling of SPHK1 is dependent on its phosphorylation activation at Ser225 by ERK1/2 and shifts from the cytoplasm to the plasma membrane.3,77 ERK1/2 not only stimulates the catalytic activity but also is necessary for SPHK1 plasma membrane trafficking.78 Finally, inactivation of SPHK1 is dephosphorylated by specific protein phosphatase, such as protein phosphatase 2A (PP2A, Table 1). It has been demonstrated that B’α (B56α/PR61α/PPP2R5A), a regulatory subunit of PP2A, interacts with the c-terminus of SPHK1, then decrease SPHK1 phosphorylation.14 SPHK1 is degraded through either the proteasome or lysosomal pathways in response to several stimuli, such as DNA damage, tumor protein 53 (TP53), tumor necrosis factor α (TNF-α), and even its inhibitors.4

Nevertheless, the SPHK1 membrane localization mechanism is still unclear, which might be associated with the activation of ERK1/2 or other partners like Ca2+/CaM and PKC.79,80 The activation of SPHK1 and its subsequent translocation to the plasma membrane was observed in response to the PKC activator, PMA (Phorbol 12-myristate 13-acetate).80 Although SPHK1 contains 4 putative PKC phosphorylation sites (shown in Table 1), purified PKC does not affect SPHK1 activity in vitro,81 indicating an indirect mechanism of PKC on SPHK1 activation. As for Ca2+, chelation of intracellular Ca2+ inhibits S1P production, whereas increasing intracellular Ca2+ enhances S1P formation.82 It has been shown that Ca2+ plays an essential role for CaM in the Ca2+-dependent translocation of SPHK1 to the plasma membrane.14 However, although several putative CaM-binding sites have been identified by sequence analysis of SPHK1 (Table 1), direct activation of SPHK1 by Ca2+ has not been demonstrated so far. Besides, several pieces of evidence demonstrated that the translocation of SPHK1 to the cell membrane is mediated by calcium and integrin-binding protein (CIB1) through its Ca2+ myristoyl switch function, and the SPHK1 signal transduction may be achieved through the Ras pathway.83-85 To a large extent, CIB1 is associated with the perplexing structure of SPHK1,9 while CIB2 has opposite actions.32 Based on these findings, Ca2+ may perform its functions by binding their partner, especially CaM and CIB1 (Figure 2).

Interestingly, Adams et al. proposed that the translocation of SPHK1 to the plasma membrane also attributes to SPHK1 dimerization.9,86 Specifically, SPHK1 forms dimers and interacts with plasma membranes through a single contiguous interface that would elongate the interface and strengthen the SPHK1-phospholipids interaction.4 However, the kinetics and dynamics underlying SPHK1 dimerization and how that would affect membrane binding and function in cells need to be further investigated.

Of note, cytokines such as TNF-α, interleukin-1 (IL-1), or growth factors have been reported to regulate the SPHK1/S1P signaling pathway, affecting the formation of S1P.18,87 Similarly, hormones are also fundamental stimulation signals, impacting SPHK1, and S1P metabolism, thus controlling cell life activities.88 After activation, SPHK1 promotes tumorigenesis by modulating various processes, such as apoptosis, autophagy, proliferation, migration, invasiveness, angiogenesis, and inflammation (Table 2). As shown in Figure 2, these diversified processes benefit from the significant position of SPHK1 in the SPHK1/S1P signaling pathway.30,44,89

The Potential Oncogenic Mechanism of SPHK1

SPHK1/S1P axis is implicated in numerous pathophysiological conditions and diseases, such as deafness, hepatitis, diabetes, obesity, atherosclerosis, osteoporosis, Alzheimer’s disease, multiple sclerosis, and even cancer.90 SPHK1 is widely upregulated across a diverse range of human cancers (Table 2), such as breast cancer, lung cancer, uterine cancer, ovarian cancer, gastric cancer, kidney cancer, liver cancer, prostate cancer, colorectal cancer, small bowel cancer, chronic myeloid leukemia, glioblastoma, and lymphoma.19,51,91,92 After being activated by phosphorylation, SPHK1 accelerates the synthesis of S1P on the cell membrane. Then the generated S1P as a second messenger, in an autocrine or paracrine manner, engages with S1PRs, to evoke a range of biological functions, such as apoptosis, autophagy, proliferation, migration, invasion, angiogenesis, and inflammation.

Effect of SPHK1 on autophagy and apoptosis

Recent studies have shown that SPHK1 is involved in autophagy and apoptosis. Dysregulation of autophagy was associated with various human disorders, such as neurodegenerative diseases, obesity, infectious diseases, cardiomyopathy, type 2 diabetes, and cancer,93,94 while apoptosis also plays vital roles in the pathogenesis of these diseases.95 Hence, both autophagy and apoptosis have been recognized as promising therapeutic targets. In particular, it is proposed that SPHK1 promotes tumorigenesis by simultaneously upregulating autophagy and suppressing apoptosis (Figure 3).

Figure 3.

Figure 3.

SPHK1 coordinates autophagy and apoptosis. A, The relationship between SPHK and autophagy. On the one hand, autophagy is a self-protection process that provides sufficient energy supply for cell growth by recycling useless raw materials; on the other hand, cancer cell takes advantage of autophagy for tumorigenesis by some mechanisms, such as cell survival, EMT and protecting cell under starvation. SPHK1 is a potential target that inhibiting SPHK1 can induce autophagy-induced death via some special mechanism, for example, TP53. B, The role of SPHK1 in apoptosis. SPHK1 performs an anti-apoptosis effect through PI3K/Akt/NF-κB(or only PI3K/Akt), Akt/FoxO3a/Bim, or RTKs. Targeting SPHK to inducing apoptosis has been a focus on cancer therapy. Many inhibitors can induce apoptosis, including SK1-Ⅰ/Ⅱ ˎ Ski/ DHS ˎ FTY720 ˎ CB5468139 ˎ PF-543 ˎ Safingol, and so on (Table 3). To sum up, there are 4 ways to induce apoptosis by inhibiting SPHK1: promoting hydrolysis, inhibiting translocation to plasmalemma, regulatory RNA, and even a direct inhibition of inhibitor. C, The role of SPHK1 between autophagy and apoptosis; cancer therapy targeting SPHK1. In principle, the relationship between autophagy and apoptosis is antagonistic. But SPHK1 correlates autophagy and apoptosis, and targeting SPHK1 could induce cell death via autophagy and apoptosis.

Autophagy is regulated by more than 30 autophagy-related genes (ATG) and requires functional late endosomes and lysosomes to participate.96,97 It is unraveled that SK1-I (a specific SPHK1 inhibitor, Table 3) inhibits SPHK1 activity, promotes the fusion of endosomal membranes to accumulate dysfunctional enlarged late endosomes, and blocks autophagy flux.98 Whereas inhibition of SPHK1 by SK1-I greatly increases autophagic flux and induces TP53-dependent cell death mediated by the autophagic regulators BECN1 and ATG5.17 In neuronal cells, overexpression of SPHK1 enhances the formation of the pre-autophagocytic Beclin1-positive structure and strengthens autophagy flux, whereas S1PPase and S1PLase have opposite effects on regulating neuronal autophagy.99 Interestingly, it has been reported that SPHK1 accelerates lysosomal degradation of CDH1 (cadherin family members) to induce epithelial-mesenchymal transition (EMT), which depended on TRAF2-mediated autophagy activation.100 These findings have revealed a novel mechanism responsible for regulating the EMT via SPHK1/TRAF2/BECN1/CDH1 signal cascades in hepatocellular carcinoma (HCC) cells.100 MiR506-3p has been demonstrated to act as a tumor suppressor through MET initiation and autophagy inhibition, which can be reversed by SPHK1overexprssion.55 SPHK1 expression and activity were significantly augmented by TGF-β1 stimulation, resulted in increased expression of autophagy-related genes ATG5/12 and Beclin 1 and subsequent autophagy induction.101 Moreover, SPHK1 could activate the ERK pathway in colon cancer,45 which also elicits autophagy. In MDCK cells with high expression of K-Ras, S1P and S1PR2 receptors are down-regulated by K-Ras, while SPHK1 and S1PPase were not affected. The lack of S1P was linked with increased autophagy.102 It is noteworthy that SPHK1 may also interact with other biologically active substances such as Cer and S1P.103 It is, therefore, a promising approach to investigate the regulation of sphingolipids (S1P, Cer, and SPHK1 were included) on autophagy and cell death (Figure 3C).

Table 3.

The Potential SPHK1 Inhibitors.a

Inhibitors Targeted molecules Cells or cancer models Dose Route of administration Potential mechanism Effects Advantages and disadvantages References
SK1-I
SKI-I
SPHK1, ERK2, AKT HCT116 U937 and Jurkat
C57BL/6 mice
10μM 5μM,10μM
5 mg/kg
Intranasal injection Concomitant increase Cer, decreases in ERK1/2 and Akt pro-survival signaling Autophagy↑, apoptosis↑, cell growth ↓, cell death↑, S1P↓ Water-soluble, an SPHK1-selective inhibitor



a sphingosine-competitive inhibitor of SPHK1
Lima S et al., 2018 Paugh SW et al., 2008
Price MM et al., 2013
SPHK1/2ERK2, PKC, PI3 K, CERK, DAGK BALB/c mice
50 mg/kg Intraperitoneal injection Induce apoptosis, decrease cellular S1P, increase cellular Cer and Sph Cell growth ↓, cell death↑, apoptosis↑, S1P↓, inflammation ↓, tumor burden↓, lymph node, and lung metastases↓, hemangiogenesis, and lymphangiogenesis↓ French KJ et al., 2006
SKI-II(SKi) SPHK1/2 Huh7 A549, MDA-MB-468
BALB/c mice
10/20/50 μμ
50 mg/kg
Intraperitoneal injection Cause apoptosis, decrease cellular S1P, increase cellular Cer and Sph, and induce the degradation of SK1, inhibits dihydroceramide desaturase In response to ROS in cardiac cells, enhance the effects of other anti-cancer agents on cell lines, including chemoresistant, growth↓, pulmonary fibrosis↓, inflammation, and hyperalgesia↓ Non-toxic to normal liver cells, but targeting SPHK2 over SPHK1; shows oral bioavailability and anti-tumor activity Chatzakos V et al., 2012; Pitman MR et al., 2016
French KJ et al., 2006
SKI-III/IV/V SPHK1/2, ERK2 and PI3K MDA-MB-231, T24 20 μg/ml Inhibit cancer cell proliferation and induce apoptosis, inhibit Sphk activity, thereby decreasing S1P levels, increasing Cer levels Cell growth ↓, cell death↑, S1P↓ Not be specific for Sphk, causing undesired inhibition for other ATP-dependent enzymes French KJ et al., 2003
French KJ et al., 2006
SKI-V BALB/c mice 75 mg/kg Intraperitoneal injection Lack of overt toxicity
SKI-178 SPHK1 MDA-MB-231(Human) and 4T1(Murine) 5 μM Inhibit effect on SPHK1, proliferation, and anti-tumor activity Cell growth ↓, viability↓, tumor volume↓ Useful both in vitro and in vivo Has a milder toxicity profile an SPHK1-selective inhibitor Alshaker H et al., 2018
BALB/c nude mice 5mg/kg Intraperitoneal injection
SK-F SPHK1 MDA-MB-231,4T1
BALB/c nude mice
1 μM
5mg/kg
Intraperitoneal injection induce significant loss of proliferation Cell growth ↓, cell viability↓, tumor volume↓ a selective SPHK1 inhibitor and a competitive inhibitor of SPHK1, without significant whole-body toxicity Alshaker H et al., 2018
DMS SPHK1/2, PKC ALF of the liver in mice 50μmol /L Intraperitoneal injection Inhibition of SPHK1 activation downregulates inflammatory cytokine and HMGB1 levels Cell growth ↓, proliferation↓, migration↓, HMGB1 cytoplasmic translocation↓ Inhibit SPHK1 activity in vivo Lei YC et al., 2015
FTY720 SPHK1/2, S1PRs, PI3K/Akt, PKC Diverse cancer and normal cells
mice*1
50uM
5/10mg/kg
Oral administration and intraperitoneal injection Inhibit or degrade SPHK1 Apoptosis↑, transfer↓, drug sensitivity↑, regulation of tumor immune microenvironment Low cytotoxicity, many targets and good synergistic effect with other drugs, good prospects for clinical application White C et al., 2016
Wallington-Beddoe CT et al., 2012
B-5354c SPHK1 PC-3 and Lncap NMRI/Nu mice 10 μmol/L 20 mg/kg Intraperitoneal injection Inhibit SPHK1 and sensitizes prostate cancer cells to chemotherapy-induced apoptosis; increase the efficacy of irinotecan Caspase activity↑, SPHK1↓, Cer/S1P rheostat toward Cer Low drugs sensitive, non-competitive, strong specificity, non-toxic Pchejetski D et al., 2008
F-12509a SPHK1/2 LAMA84-s and LAMA84-r (CML) 10μM Activate caspase-3, induce apoptosis Cell death↓, viability↑, ceramide↑, S1P↓ Dose- and time-dependent cytotoxicity, overcome resistance to imatinib in LAMA84-r cells, as effective as imatinib in killing CML primary cells Bonhoure E et al., 2008
CB5468139 SPHK1, DAG, AKT, CLK1, FYN, Met, MST2, PIM2, SYK and TNK2 A498 2 μMα Target the sphingosine binding site, decrease S1P, reduce the expression of p53 Cell growth ↓, proliferation↓, migration↓, proliferation↓, apoptosis↑ An SPHK1-selective inhibitor, an ATP mimetic chemical Gao P et al., 2012
PF-543 (Compound 22a) SPHK1 1483*2 , A549, LN229, Jurkat, U937, MCF-7 IC50 = 26.7 nM in human whole bloodβ Inhibiting SPHK1-catalyzed sphingosine phosphorylation in a reversible competitive way Does not affect cell proliferation and survival, but S1P/Sph↓, apoptosis↑, autophagy↑ The most effective SPHK1 inhibitor, an SPHK1-selective inhibitor Schnute ME et al., 2012, 2017
Safingol SPHK1/2, PKC SH-SY5Y
HCT-116
10 μM
12 μM
Inhibit SPHK1, effect MAPK, and p38 pathway
Induces cell death of an exclusively autophagic character, inhibit PKCβ-I/δ/∊ and phosphorylation of critical components of the PI3k/Akt/mTOR pathway and MAPK pathway; Promote the prevention of RTK phosphorylation and activation of DAPK1
Autophagy↑, ceramide↑, survival↓ A competitive inhibitor but low selective for SPHK1, first enter clinical trials as an anti-cancer agent, without affecting normal cells Tavarini S et al., 2000
Coward J et al., 2009
U266, ARH-77, RPMI8226 (human) and MPC-11 (mouse) BALB/c mice 1μM
5 mg/kg
Intraperitoneal injection Apoptosis↑, ceramide↑, tumor size↓, tumor growth↓, the survival rate↑ Tsukamoto S et al,2015; Acharya S et al., 2019
Compound 82 SPHK1 MDA-MB-231 WM266.4 B16F10 IC50 = 90 nM IC50 = 63 nM IC50 = 250 nM Inhibit SPHK1 activity, deplete S1P, inhibit proliferation Cell viability ↑, S1P↓ An SPHK1-selective inhibitor Gustin DJ et al., 2013
Athymic nude mice, C57Bl/6 mice 100 mg/kg Oral gavage Tumor volume↓
SLP7111228 (compound36a) SPHK1 Sprague-Dawley strain rats (200-300 g) 10 mg/kg Intraperitoneal injection decreased S1P levels in vitro and in vivo blood S1P levels↓ An SPHK1-selective inhibitor, affording >200-fold selectivity Patwardhan NN et al., 2015
Compound 51 SPHK1, CDKs A-673, SK-Br-3, SK-ES-1 and T-47D 1.5 µM suppressed CDK-specific phosphorylation of the Rb protein and induced activation of caspase-3 inhibited the proliferation/survival of cancer cells High selectivity for CDKs Xiang YB et al., 2010; Schonbrunn E et al., 2013

a All dose are the significant differences : Clinical phase inhibitors; *1 : Human ALL xenografts in NOD/ SCIDγc-/- Mice; *2: 1483 Head/Neck carcinoma cells; α: The dose results in ≥50% inhibition of 12 of 65 protein kinase (including SPHK1); β: Ki = 3.6 nM; CML: Chronic myeloid leukemia cells, DAPK1: death-associated protein kinase 1, RTK: receptor tyrosine kinase; CERK: ceramide kinase, DAGK: diacylglycerol kinase.

Interestingly, the correlation between autophagy and SPHK1 varies in different tumor types and stages. Overexpression of SPHK1 induces autophagy in neurons and protects against starvation in star glial cells.104 SPHK1 promotes autophagy in neurons by translocating to endocytic and autophagic vesicles, while in SH-SY5Y neuroblastoma cells, SPHK1 exclusively locates on the endosomal membrane.105 Autophagy is a double-edged sword in tumor biology. Previous studies demonstrated that autophagy inhibits tumorigenesis in the early stage, while more advanced cancer cells utilize autophagy as a pro-survival tool to enhance tumor growth.106-108 SPHK1 knock-out protects TP53 -/- and TP53 +/- mice from developing thymic lymphoma,109 suggesting that SPHK1 is required for spontaneous tumorigenesis in TP53 -/- mice. Thus, p53 is functionally associated with SPHK1, and SPHK1 could be a downstream target of p53. Furthermore, SPHK1-induced autophagy promotes tumor cell survival, induces EMT, and protects against starvation. These processes are strongly associated with tumorigenesis. Besides, SPHK1 inhibition could induce autophagic cell death, a novel anti-cancer strategy (Figure 3A/C).

SPHK1 also plays a vital role in apoptosis. Inhibition of SPHK1 results in lower S1P levels and apoptosis of prostate cancer cells.110 Similar results have been reproduced in other types of cancer (Table 2). Recent reports show that p53-mediated caspase-2 activation is required for SPHK1 proteolysis. Caspase-2 activation is downstream of the checkpoint kinase 1 (CHK1, a serine/threonine-protein kinase) inhibitory pathway in TP53-mutated cells, and inhibition of CHK1 leads to caspase-2 activation and apoptosis.111 The aforementioned p53 activation by inhibition of SPHK1 leads to pro-apoptotic BCL2 family members’ upregulation, including BAD, BAK1, and BID.17 CIB2 blocks the translocation of SPHK1 to the plasma membrane and impairs its subsequent signaling, including induction of apoptosis by TNF-α and limiting Ras-induced tumorigenesis.32 In contrast, CIB1 mediates the translocation of SPHK1 to cell membranes.83-85 but whether CIB1 participates in apoptosis is unclear.

A recent study in non-small cell lung cancer (NSCLC) has revealed that the upregulation of SPHK1 boosts the PI3K/AKT/NF-kB pathway, and suppressing this pathway abolishes the anti-apoptotic effect of SPHK1.112 However, neither SPHK1 nor SPHK2 regulates NF-kB activation.113 Suppression of SPHK1 induces apoptosis in glioblastoma cells and limits tumor growth. SK1-I specifically restrains phosphorylation and activation of AKT in an S1P-dependent manner, while inhibition of JNK attenuates SK1-I-mediated cell death,114 implying that targeting SPHK1 to induce cancer cells apoptosis requires cooperation with the JNK pathway. In gastric cancer cells, SPHK1 inhibits apoptosis by downregulation of Bim via stimulating Akt/FoxO3a signaling.47 Through inhibiting SPHK1 by Ski (also SKI-II), SPHK1 protects multiple myeloma (MM) cells against apoptosis by inhibition of cancer-specific RTKs.54 Interestingly, inhibition of SPHK1 also promotes the synthesis of a novel pro-apoptotic molecule diadenosine 5′,5′′′-P1, P3-triphosphate (Ap3A), which is associated with tumor suppressor of fragile histidine triad (FHIT), then triggering apoptosis.110

Some non-coding RNA is associated with apoptosis, such as miRNA and LncRNA, and their mechanisms may be different. Overexpression of miR-515-5p in breast cancer cells could weaken SPHK1 activity, decreased cell proliferation, and elevated caspase-dependent apoptosis.21 It has been shown that SPHK1-siRNA not only accelerates apoptosis but also restricts proliferation in ovarian cancer cells,115 which could be a potential targeted therapy.116 LncRNA HULC inhibits apoptosis by upregulating SPHK1 and its downstream PI3K/Akt pathway.25 Downregulating lncRNA MEG3 inhibits apoptosis by upregulating TGF-β1 and its downstream SPHK1,41 suggesting that the SPHK1-dependent inhibition of apoptosis in cancer cells could be a therapeutic target by different approaches, such as promoting hydrolysis, inhibiting translocation to plasmalemma, regulatory RNA, and chemical inhibition (Figure 3B).

In summary, SPHK1 serves as both a stimulant of autophagy and an inhibitor of apoptosis (Figure 3A/B/C). Autophagy protects cells from accumulating toxins, improper molecules, and damaged organelles and facilitates tissue development and cell differentiation.117 Autophagy also supplies nutrition for cell metabolism by recycling the materials mentioned above. Under adverse conditions such as severe inflammation, hypoxia, or malnutrition, autophagy may become excessive and cause autophagic cell death.108 SPHK1 may regulate autophagy and related signaling pathways in response to changes in the environment, leading to cell survival or death. Thus, targeting SPHK1-mediated autophagic cell death could be a promising therapeutic approach for cancer treatment.

Since SPHK1 also exerts anti-apoptosis effects, inhibitors targeting SPHK1 have been tested in various cancer models (Table 3). Furthermore, SPHK1-mediated autophagy may directly affect apoptotic signals (Figure 3C). It is widely believed that SPHK1-mediated autophagy and apoptosis are mutually exclusive.118 For example, SPHK1-induced autophagy protects against apoptosis during nutrient starvation, while Cer-induced autophagy does not.119 Autophagy usually precedes apoptosis and activates apoptosis via various mechanisms, but autophagy can also protect cells from apoptosis. Apoptosis and autophagy have distinct signaling pathways and cellular processes but also share some functions.120,121 These 2 processes can occur simultaneously or independently, and the result depends on the environment and cellular response.120,122 Altogether, either SPHK1-mediated autophagy or apoptosis can be targeted in cancer therapy to induce autophagic or apoptotic cell death (Figure 3C).

SPHK1 inducing tumor proliferation and survival

The proliferation of tumor cells is regulated by numerous cellular factors.123 Cumulating studies have shown that SPHK1 promotes cell proliferation and tumor growth18,88 (Figure 4).

Figure 4.

Figure 4.

The mechanism of SPHK1 in proliferation, migration, invasion, angiogenesis, and inflammation.

SPHK1 is a positive regulator of cell proliferation and survival. Knocked-out of SPHK1 suppresses AKT signaling pathway and inhibits cell proliferation.124 SPHK1 regulates tumorigenesis and tumor growth in early colon cancer,125 glioma,126 breast cancer, and chronic lymphocyte by modulating calcium signaling.127,128,129 Overexpression of SPHK1 enhances triple-negative breast cancer proliferation via PI3K/AKT signaling,130 whereas TNF-α regulates breast cancer cell proliferation by modulating Cer.131 Elevated SPHK1 also augments colon cancer cell proliferation by regulating the MAPK/MMP-2/9 pathway.71 Gene expression array has shown that SPHK1 regulates cell survival, proliferation, and tumor transformation by upregulation of transferrin receptor 1 (TFR1).3 TFR1 is a novel target of SPHK1, which provides new insights into the regulatory mechanism of SPHK1-dependent tumorigenesis. Epoxyeicosatrienoic acid (EET) is a product of cytochrome P450 (CYP) peroxidase. Studies have shown that SPHK1 augments EET-stimulated endothelial cell proliferation, which requires S1PR1 and S1PR3.132 Vascular endothelial growth factor (VEGF) can induce endothelial cell proliferation through its receptors.133 Activated SPHK1 in breast cancer upregulates VEGF expression, and both VEGF and SPHK1 are independently regulated by the mammalian target of rapamycin C1 (mTORC1).134 VEGF and follicle-stimulating hormone (FSH) could upregulate S1P synthesis through phosphorylation of SPHK1, accelerating cell proliferation.135 Recent studies have revealed that inhibition of SPHK1 attenuates proliferation and survival in cancer cells by impairing PKC activity and cytokinesis.62 Leptin is an essential adipokine that plays a key role in regulating energy balance, body weight, metabolism, and endocrine function.136 Studies have shown that leptin can escalate the proliferation and activation of SPHK1 via ERK1/2 and Src family kinase (SFK) pathways.137

MiRNAs such as miR-515-5p and miR-124 can inhibit apoptosis in tumor cells, and miRNAs can control the activity and expression of SPHK1 to suppress proliferation, which is closely related to WNT pathway.21,31,138 In bladder cancer, miR-613 inhibits bladder cancer cells’ proliferation by targeting SPHK1 expression and function.71,139 MiR-128 also suppresses the growth of thyroid carcinoma by downregulating SPHK1.56 Thus, SPHK1 is a target for miRNA with anti-cancer potential,44 but further investigation of the miRNA-dependent regulation mechanism of SPHK1 is needed. Apart from miRNAs, lncRNAs with distinct functions significantly contribute to SPHK1 signaling as well. Particularly, lncRNAs HULC induces NSCLC proliferation and inhibits apoptosis by enhancing SPHK1 and PI3K/Akt signaling.25 In contrast, suppression of lncRNA MEG3 accelerates colorectal adenocarcinoma cell proliferation and abolishes apoptosis by upregulating TGF-β1/ SPHK1 pathway.41

Cancerous proliferation is often caused by cell cycle regulation abnormalities and genetic changes.123,140 However, SPHK1 and SPHK2 activity are not required for tumor cell viability.141 Altogether, it still needs to explore further the contribution of SPHK1 to tumor proliferation and cell survival. In summary, SPHK1 promotes cell proliferation and survival in cancer, whereas its inhibitors such as miRNA and chemical inhibitors suppress proliferation. These data provide a strategy to inhibit tumor proliferation by targeting SPHK1.

SPHK1 promoting tumor migration and invasion

SPHK1 has been implicated in tumor metastasis as well as invasiveness.38 SPHK1 is overexpressed in many tumor types (Table 2) and plays an important role in tumor migration and invasion. Previous studies have found that SPHK1 is overexpressed in NSCLC and enhances the migratory and invasive ability of NSCLC through the AKT pathway.26 SPHK1 is also overexpressed in triple-negative breast cancer (TNBC).23 SPHK1 knock-out attenuates migration and invasion of TNBC by controlling PI3K/AKT signaling pathway.130 Tumorigenesis is not only triggered by intracellular factors but also closely related to the extracellular environment.142 Hormones and growth factors can regulate SPHK1 and S1P production and facilitate the migration of endothelial cells.143 Hepatocyte growth factor (HGF) could induce the phosphorylation of SPHK1 and drive the production of S1P, thereby promoting the migration of lung endothelial cells.144 SPHK1/S1P regulates sirtuin 1 (SIRT1) expression through P38 MAPK, ERK, and AKT signals, and SIRT1 knock-out blocks the migration of human umbilical vein endothelial cells (HUVEC).145 Death receptor 5 (DR5) is another SPHK1 functional partner. When DR5 is suppressed, SPHK1/S1P may be involved in TRAF2-mediated activation of JNK/AP-1 to stimulate tumor invasiveness.146 SPHK1-dependent migration may also be associated with myristoylated alanine-rich C-kinase substrate (MARCKS)-related proteins.147 These results show that SPHK1 promotes tumor migration and invasion in cooperation with other proteins. In response to irradiation, head and neck squamous cell carcinoma (HNSCC) migration may be associated with EGFR.148 SPHK1 is also strongly linked to colon cancer cells (CRC), which advances CRC migration and invasion, and may be connected to E-cadherin and vimentin to induce EMT.149 Similarly, SPHK1 can promote CRC migration and EMT by regulating the expression of p-FAK.150 It was unveiled that SPHK1 facilitates colorectal cancer’s metastasis facilitates the metastasis of colorectal cancer by stimulating EMT via the FAK/AKT/MMPs axis.42

The production of S1P and its receptor S1PR are also associated with migration and invasion regulated by SPHK1. When S1P is produced in a large amount, it also provokes migration of oral squamous cell carcinoma (OSCC), at least through targeting S1PR2.151 SPHK1-mediated renal clear cell carcinoma (ccRCC) invasion occurs by S1PR2-dependent FAK phosphorylation and a FAK-independent mechanism through S1PR1/3.46 And HNSCC invasion possibly associates with S1PR1.152 By the cancer genome atlas (TCGA) RNA database analysis, SPHK1 stimulates invasion in von Hippel-Lindau (VHL) mutant ccRCC via S1PR2-dependent FAK phosphorylation and FAK-dependent S1PR1/3 mechanism.46

Viruses are also implicated in SPHK1-mediated tumor invasion.153 Hepatitis B virus (HBV) contributes to tumor growth by upregulating AP2α and SPHK1,154 and SPHK1 can trigger the proliferation, growth, and migration of HCC through S1P/endothelial differentiation G-protein coupled receptor 1 (EDG1) and nuclear factor Kappa B subunit 1 (NF-κB) pathways.155 Moreover, Epstein-Barr virus (EBV) activates AKT via the SPHK1/S1P pathway to promote the migration of undifferentiated nasopharyngeal carcinoma (NPC).156 These findings suggest that pathogenic microorganisms should be considered as a novel regulator of SPHK1 in tumorigenesis.

SPHK1-related miRNAs also are associated with tumor migration and invasion. Overexpression of SPHK1 upregulates miRNAs that play different roles in tumorigenesis. In particular, the miR-144-3p/fibronectin 1 (FN1) pathway may be involved in the pro-invasive role of SPHK1 in thyroid papillary cancer cells (PTC).157 Of interest, some miRNAs show distinct effects. For example, miR-613 protect against migration by targeting SPHK1 in bladder cancer139 and PTC,158 while miRNA-124 targets SPHK1 and down-regulates SPHK1 expression to arrest tumor proliferation and migration.138

SPHK1 is currently explored as a therapeutic target to restrain tumor growth and migration. Dimethylsphingosine (DMS, SPHK inhibitor) can govern tumor invasiveness through NF-κB, alleviating tumor permeation.159 CB5468139 also inhibits migration by targeting SPHK1 (Table 3). These results provide an anti-metastasis therapeutic strategy by targeting SPHK1. In conclusion, SPHK1 promotes tumor migration and invasion with viral infection as an important factor, which calls for further study into the detailed regulatory mechanism.

SPHK1 prompting tumor angiogenesis

To ensure tumor growth, the tumor tissue will establish a vascular system, in which the tumor or other cells secrete pro-angiogenic factors and facilitate angiogenesis.160 SPHK1 promotes tumor angiogenesis by several mechanisms (Figure 4).

SPHK1 and its metabolite S1P significantly contribute to tumor angiogenesis and survival.71 In epithelial ovarian cancer, inhibition of SPHK1 or S1PR1/3 attenuates angiogenic potential and angiogenic factor secretion.161 When S1P is added, angiogenic potential and angiogenic factor secretion can be restored, suggesting that the SPHK1/S1P/S1PR1/3 pathway plays an essential role in angiogenesis.30 The identical mechanism was also characterized in clear cell renal cell carcinoma (ccRCC).46 Angiogenesis is associated with cancer metastasis and invasion, while the molecular mechanism is not yet clear.162 Enhancing the activity of SPHK1 triggers angiogenesis and facilitates invasion and metastasis of cervical cancer.163 Increased SPHK1 leads to upgraded S1P production, resulting in angiogenesis and metastasis in triple-negative breast cancer23,128 and undermine breast cancer migration and survival.164 S1P targets corresponding receptors in hematologic malignancies, activates Ras/MEK/ERK1/2, PI3K/AKT/mTOR, Rac, Rho, and PLC to govern angiogenesis.165 Besides, SPHK1 promotes angiogenesis, metastasis, and invasiveness in head and neck squamous cell carcinoma (HNSCC).166

Besides, non-coding RNAs, such as miRNAs and lncRNAs, are also involved in SPHK1-mediated angiogenesis. As an example, miR-506 can suppress the expression of SPHK1 in hepatocellular carcinoma HepG2 cells through binding to the 3′-UTR of SPHK1-mRNA and reduce S1P production to impair angiogenesis.37 Another lncRNA has also been demonstrated to promote tumor angiogenesis in liver cancer by upregulating SPHK1.167

In summary, SPHK1 prompts tumor angiogenesis, which provides new capillaries to deliver oxygen and nutrients to the tumor (Figure 4) and facilitates metastasis.

The relationship between SPHK1 and inflammation

SPHK1 and S1P are closely associated with inflammation and inflammatory factors.168,169 Especially, inflammatory factors such as cytokines trigger inflammation and tumorigenesis by activating SPHK1.88 Therefore, SPHK1 could be viewed as a mediator of inflammation in the tumor microenvironment.

The SPHK1/S1P pathway is involved in inflammatory responses to cytokines, such as TNF-α and IL-1.18 SPHK1 can also be activated by inflammatory signaling molecules such as IL-1b, IFN-g, and IgE.170 However, SPHK1 knock-out mice lack systemic inflammatory response.171 TNF-α activates SPHK1 via a TRAF2-dependent mechanism and affects cell inflammation and survival through AKT and NF-κB signaling.18 In a mouse model, SPHK1 and TRAF2 regulate TNF signaling, and inhibition of NF-κB and TNF signaling may be beneficial to certain diseases.113 Prior research suggests that SPHK1 functions through the p38MAPK pathway and was not required to activate the canonical NF-κB pathway.172 Therefore, the molecular mechanism of SPHK1 in inflammation and tumors needs further investigation. Of course, SPHK1 can promote the release of pro-inflammatory cytokines such as IL-6 in turn.18,173 It is reported that SPHK1 mediated inflammation and colon cancer by modulating various molecules, including cyclooxygenase 2 (COX2), prostaglandin E2 (PGE2), NF-κB, IL-6, and signal transducer and activator of transcription 3 (STAT3).174 For COX2, it suppresses the anti-tumor response by CTL-, Th1-, and NK cell-mediated type-1 immunity.175 But whether the role of COX2 in inflammation is caused by SPHK1 phosphorylation or acetylation is not clear since SPHK1 acetylates COX2 in an acetyl-CoA dependent manner.66 STAT3 is a key transcription factor of inflammation and cancer.166 SPHK1/S1P is implicated in the regulation of STAT3 and the development of intestinal inflammation and relevant cancers,176 and it upregulates CD44 in human colon cancer cells through ERK signaling.43 Targeting S1P signaling and metabolism might be a new strategy for treating inflammatory bowel disease (IBD), which may lower colon cancer risk.177 In the mouse intestinal cancer model, the upregulation of SPHK1 and S1P production is beneficial to developing inflammation and colon cancer via the signaling network of S1P/S1PR1/STAT3/NF-κB/IL-6/TNF-α.178 SPHK1 is also associated with the development and progression of inflammatory gastrointestinal (GI) diseases, leading to GI cancer through immunologic mechanisms,179 suggesting SPHK1 as a pivotal regulator of inflammation-induced tumorigenesis (Figure 4).

SPHK1 is originally recognized as a pro-inflammatory enzyme capable of activating TNF-α and NF-κB,180,181 but later studies have identified that in the mouse bone marrow macrophage (BMDM) model, SPHK1 is not essential for inflammatory response.182 Aquaporin 4 (AQP4) is functionally associated with SPHK1 directly or indirectly. AQP4 deficiency was associated with the SPHK1/MAPK/AKT pathway and could alleviate the release of pro-inflammatory cytokines from astrocytes.183 Targeting the SPHK1/S1P/S1PR1 pathway, which influences the progression of inflammation and breast cancer, is dependent on the activation of STAT3 and upregulation of IL-6.184

In conclusion, SPHK1 strongly correlates with inflammation. In general, pro-inflammatory molecules activate the SPHK1/S1P signaling pathway that upregulates STAT3, NF-κB, IL, TNF-α, MAPK, AKT, ERK, COX2, etc. which in turn promote cytokines production and release. Last, the amplification loop promotes prolonged and excessive inflammation that facilitates tumorigenesis. Here we speculate positive feedback between SPHK1 and inflammation. Inflammatory signals activate SPHK1 that subsequently promotes the synthesis and release of inflammatory molecules to the tumor microenvironment, which could promote tumorigenesis and cancer cell survival (Figure 4).

The Recent Development of Potential SPHK1 Inhibitors

Due to the association of SPHK1 with various processes of tumorigenesis, inhibition of SPHK1 is an attractive approach to limit tumor growth and metastasis. A wide variety of potent agents targeting SPHK1 have been developed, including SK1-I, SKI-I∼V, DMS, FTY720, Safingol, SKI-178, SK-F, B-5354c, F-12509a, CB5468139, SLP7111228, Compound 23/51/82, and PF-543 (Table 3, Figure 5). The summary of these drug candidates is described below.

Figure 5.

Figure 5.

The structure of Sph, S1P, and SPHK1 inhibitors. A, The structure of Sph, S1P. Both Sph and S1P are sphingolipids. The former is the substrate of the SPHK1 enzyme, and the latter is the production of SPHK1. B, The structure of dual SPHK1/SPHK2 inhibitors. C, The structure of SPHK1-selective inhibitors.

Dimethylsphingosine (DMS), the first direct SPHK inhibitor, suppresses cancer cell growth, and induces apoptosis while also potently blocks PKC signaling.185 Meanwhile, DMS has been shown to inhibit both SPHK2 and ceramide kinase (CERK), making it unable to decipher the role of SPHK1.11 (2 R, 3 S, 4E)-N-methyl-5-(4-pentylphenyl)-2-aminopent-4-ene-1, 3-diol (SK1-I), an Sph-competitive SPHK1 inhibitor, has been widely implemented in vivo to elucidate the role of SPHK1 in cancer.186 SK1-I decreases serum S1P levels, stimulates cancer cell apoptosis, and prevents lymph node and lung metastasis in a murine breast cancer model.187 SK1-I is regarded as a selective SPHK1 inhibitor that does not suppress SPHK2,188 but it is also reported to restrain SPHK2 with a similar affinity.189 Although SKI-I (N-[(2-hydroxy-1-naphthyl)methylene]-3(2naphthyl)-1Hpyrazole-5-carbohydrazide) is also an Sph-competitive inhibitor of SPHK1, it targets SPHK2 and cross-reacts with ERK2, PKC, and PI3 K.186 Similar to SKI-I, 2-(p-hydroxyanilino)-4-(p-chlorophenyl) thiazole (SKI-II, also SKi) was revealed to attenuate SPHK1 signaling by triggering the lysosomal degradation of SPHK1 in various cell types rather than direct inhibition.190 However, SKI-II targets not only SPHK1 but also impairs SPHK2 activation.186 Certainly, it was more effective to suppress SPHK1 than SPHK2 based on their Ki (Ki -SPHK1 = 16, Ki-SPHK2 = 8). Moreover, SKI-II decreases cellular S1P levels and increases cellular Cer and Sph, leading to apoptosis and suppression of proliferation and migration.191 Another pharmacologic inhibitor, SKI-III∼V, an analog of SKI-I/II, has been unveiled by a series of modifications of Sph.192

Fingolimod (FTY720), identified by Billich et al. in 2003,193 is an FDA-approved immunomodulating drug for multiple sclerosis by antagonizing S1PR1.194,195 FTY720 inhibits S1P generation by competition with Sph as a substrate for both SPHK1 and SPHK2,69 with a lower efficiency with SPHK1 than SPHK2.196 FTY720 also promotes the degradation of SPHK1 via ubiquitination and proteasomal pathway in cell lines.18,196 In vitro and in vivo studies demonstrate the anti-growth and pro-apoptosis ability of FTY720 in various normal and cancer cells.196 In a model of human ALL xenografts in mice, FTY720 remarkably reduces the incidence of leukemia, which may be a potential treatment for Ph+ ALL but not Ph- B-ALL.197 L-threo-dihydrosphingosine (Safingol, also DHS) showed potent SPHK-inhibiting properties198 and was a promising anti-cancer agent, but it lacks specificity, with SPHK1, SPHK2, and PKC as targets.199

More novel and potent inhibitors of SPHK1 are in the pipeline (Figure 5). It is reported that SKI-178 suppresses the functions of SPHK1 in tumorigenesis and cancer progression in vitro and in vivo.198 It is SPHK1-selective and has low toxicity (Table 3). SK-F is another selective inhibitor of SPHK1, which potently decreases cancer cell viability in vitro and sensitized mouse breast tumors to docetaxel (DTX) in vivo without significant whole-body toxicity.198 F-12509a, a new sphingosine kinase inhibitor, led to ceramide accumulation, reduction in S1P content, and apoptosis.200 F-12509a functions by mimicking the conformation of Sph to bind to the active site of SPHK1. In contrast, B-5354c, a non-competitive inhibitor for Sph, maybe interact with domains distinct from the Sph associating sites to regulate SPHK1 activity.185 Of note, B-5354c and F-12509a are isolated from a marine bacterium and discomycete Trichopezizella barbata, respectively.201

The ATP-competitive SPHK1 selective inhibitor, CB5468139, was identified from a small molecule library.191 Unfortunately, CB5468139 blocks various protein kinases such as CDC like kinase 1 (CLK1), Src family tyrosine kinase (FYN), Met (tyrosine kinase), MST2 (histone acetyltransferase), PIM2 (serine/threonine kinase), SYK (spleen associated tyrosine kinase) and tyrosine kinase non-receptor 2 (TNK2) (with IC50 values<2 μM).186 Thus, the phenotypes observed after treatment may be due to off-target effects. Compound 23 was identified from a series of structure-based SPHK1 and SPHK2 inhibitors. It has a lower IC50 of 20 nM to SPHK1 than other compounds and makes key hydrogen bonding interactions with D81 and D178 of SPHK1.202 Similar with compound 23, compound 82 has the same IC50 to SPHK1 and reduces plasma S1P levels by approximately 3.5 fold.202 Unfortunately, its effects on endogenous lipids or protein kinases have not been evaluated.186 SLP7111228 (compound 36a, Ki = 48 nM) is a potent and selective SPHK1 inhibitor, which decreases S1P levels in U937 cells. Administration of SLP7111228 in rats also reduces blood S1P levels.203 Compound 51 is an SPHK1-selective inhibitor with improved aqueous solubility and ADME properties while maintain or enhance enzyme potency.192 Notably, a novel cell-permeant inhibitor of SPHK1, PF-543, is the most potent inhibitor of SPHK1 with 130-fold selectivity over SPHK2.204,205 PF-543 has been co-crystallized with SPHK1, which gives structural insights into the Sph pocket and suggests that Phe302 in the SPHK1b isoform is not conserved in SPHK2, confers selectivity for SPHK1.206 The development of PF-543 has shown that even if S1P is strongly reduced, there is no effect on cancer cell proliferation.

Altogether, most SPHK inhibitors lack specificity to SPHK1 (Table 3, Figure 5). Recently, novel molecules such as SKI-178, B-5335c, PF-543, and compound 51/82 are designed to target SPHK1 (Table 3) selectively. However, these second-generation inhibitors would need validation in tumor growth and metastasis models first.

Currently, there are no FDA-approved drugs that target SPHK1. Most SPHK1 inhibitors remain at the pre-clinical stage, except Safingol (Table 3). Safingol has concluded Phase I trial with non-toxic and a maximally tolerated dose.185 Of note, S1PR1 antagonist FTY720 was approved by the FDA in 2010 for treating multiple sclerosis.194

Conclusion

In summary, SPHK1 is a crucial sphingolipid metabolic kinase with versatile functions, especially in tumor biology. SPHK1 regulates tumorigenesis and cancer progression by modulating various cellular processes, including apoptosis, autophagy, proliferation, migration, invasion, angiogenesis, and inflammation, suggesting SPHK1 as a promising target for cancer therapy. Further investigation into the detailed oncogenic mechanism of SPHK1 and the development of potent SPHK1 inhibitors with improved specificity may offer a novel direction in cancer therapy.

Footnotes

Author Contributions: Xianwang Wang, PhD, Yong Sun, MM and Xiaochun Peng, PhD are authors contributed equally to this work. XW W, XC P, YY L, and SY conceived the study. YS performed the literature search and data analysis. YS, SM Abbas, YY, JZ, MW C, YC, HY C, HY Y, and GL W produced the figures and tables. XW W, PH, YY L, and SY wrote the manuscript. All authors read and approved the manuscript.

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by grants from the National Natural Science Foundation of China (31700736, 31501116), China Scholarship Council (201908420102), Hubei Province Scientific and Technological Research Project (D20201306), Hubei Medical Youth Tip-Top Talent, Leading Talent Program of Yangtze Talent Project and the College Students Innovative Entrepreneurial Training Program in Yangtze University (2018184, 2019372, Yz2020334).

ORCID iD: Xianwang Wang, PhD Inline graphic https://orcid.org/0000-0002-2112-1608

Xiaochun Peng, PhD Inline graphic https://orcid.org/0000-0001-9443-0439

References

  • 1.Olivera A, Kohama T, Edsall L, et al. Sphingosine kinase expression increases intracellular sphingosine-1-phosphate and promotes cell growth and survival. J Cell Biol. 1999;147(3):545–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Olivera A, Spiegel S. Sphingosine-1-phosphate as second messenger in cell proliferation induced by PDGF and FCS mitogens. Nature. 1993;365(6446):557–560. [DOI] [PubMed] [Google Scholar]
  • 3.Pham DH, Powell JA, Gliddon BL, et al. Enhanced expression of transferrin receptor 1 contributes to oncogenic signalling by sphingosine kinase 1. Oncogene. 2014;33(48):5559–5568. [DOI] [PubMed] [Google Scholar]
  • 4.Pulkoski-Gross MJ, Obeid LM. Molecular mechanisms of regulation of sphingosine kinase 1. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863(11):1413–1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kohama T, Olivera A, Edsall L, Nagiec MM, Dickson R, Spiegel S. Molecular cloning and functional characterization of murine sphingosine kinase. J Biol Chem. 1998;273(37):23722–23728. [DOI] [PubMed] [Google Scholar]
  • 6.Liu H, Sugiura M, Nava VE, et al. Molecular cloning and functional characterization of a novel mammalian sphingosine kinase type 2 isoform. J Biol Chem. 2000;275(26):19513–19520. [DOI] [PubMed] [Google Scholar]
  • 7.Igarashi N, Okada T, Hayashi S, Fujita T, Jahangeer S, Nakamura S. Sphingosine kinase 2 is a nuclear protein and inhibits DNA synthesis. J Biol Chem. 2003;278(47):46832–46839. [DOI] [PubMed] [Google Scholar]
  • 8.Wang Z, Min X, Xiao SH, et al. Molecular basis of sphingosine kinase 1 substrate recognition and catalysis. Structure (London, England: 1993). 2013;21(5):798–809. [DOI] [PubMed] [Google Scholar]
  • 9.Adams DR, Pyne S, Pyne NJ. Sphingosine kinases: emerging structure-function insights. Trends Biochem Sci. 2016;41(5):395–409. [DOI] [PubMed] [Google Scholar]
  • 10.Haddadi N, Lin Y, Simpson AM, Nassif NT, McGowan EM. “Dicing and splicing” sphingosine kinase and relevance to cancer. Int J Mol Sci. 2017;18(9):1891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Pulkoski-Gross MJ, Donaldson JC, Obeid LM. Sphingosine-1-phosphate metabolism: a structural perspective. Crit Rev Biochem Mol Biol. 2015;50(4):298–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Pitson SM, Moretti PA, Zebol JR, et al. The nucleotide-binding site of human sphingosine kinase 1. J Biol Chem. 2002;277(51):49545–49553. [DOI] [PubMed] [Google Scholar]
  • 13.Pitson SM, D’Andrea RJ, Vandeleur L, et al. Human sphingosine kinase: purification, molecular cloning and characterization of the native and recombinant enzymes. Biochem J. 2000;350(Pt 2):429–441. [PMC free article] [PubMed] [Google Scholar]
  • 14.Pitman MR, Barr RK, Gliddon BL, Magarey AM, Moretti PA, Pitson SM. A critical role for the protein phosphatase 2A B’alpha regulatory subunit in dephosphorylation of sphingosine kinase 1. Int J Biochem Cell Biol. 2011;43(3):342–347. [DOI] [PubMed] [Google Scholar]
  • 15.Pitson SM, Xia P, Leclercq TM, et al. Phosphorylation-dependent translocation of sphingosine kinase to the plasma membrane drives its oncogenic signalling. J Exp Med. 2005;201(1):49–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Jin L, Liu WR, Tian MX, Fan J, Shi YH. The SphKs/S1P/S1PR1 axis in immunity and cancer: more ore to be mined. World J Surg Oncol. 2016;14(1):131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lima S, Takabe K, Newton J, et al. TP53 is required for BECN1- and ATG5-dependent cell death induced by sphingosine kinase 1 inhibition. Autophagy. 2018;14(6):942–957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Alshaker H, Sauer L, Monteil D, et al. Therapeutic potential of targeting SK1 in human cancers. Adv Cancer Res. 2013;117:143–200. [DOI] [PubMed] [Google Scholar]
  • 19.Xu Y, Dong B, Wang J, Zhang J, Xue W, Huang Y. Sphingosine kinase 1 overexpression contributes to sunitinib resistance in clear cell renal cell carcinoma. Oncoimmunology. 2018;7(12):e1502130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Geffken K, Spiegel S. Sphingosine kinase 1 in breast cancer. Adv Biol Regul. 2018;67(undefined):59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pinho FG, Frampton AE, Nunes J, et al. downregulation of microRNA-515-5p by the estrogen receptor modulates sphingosine kinase 1 and breast cancer cell proliferation. Cancer Res. 2013;73(19):5936–5948. [DOI] [PubMed] [Google Scholar]
  • 22.Yamada A, Nagahashi M, Aoyagi T, et al. ABCC1-exported sphingosine-1-phosphate, produced by sphingosine kinase 1, shortens survival of mice and patients with breast cancer. Mol Cancer Res. 2018;16(6):1059–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Acharya S, Yao J, Li P, et al. Sphingosine-kinase-1 signaling promotes metastasis of triple-negative breast cancer. Cancer Res. 2019;79(16):4211–4226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wang W, Hind T, Lam BWS, Herr DR. Sphingosine 1-phosphate signaling induces SNAI2 expression to promote cell invasion in breast cancer cells. FASEB J. 2019;33(6):7180–7191. [DOI] [PubMed] [Google Scholar]
  • 25.Liu L, Zhou XY, Zhang JQ, et al. LncRNA HULC promotes non-small cell lung cancer cell proliferation and inhibits the apoptosis by upregulating sphingosine kinase 1 (SPHK1) and its downstream PI3K/Akt pathway. Eur Rev Med Pharmacol Sci. 2018;22(24):8722–8730. [DOI] [PubMed] [Google Scholar]
  • 26.Zhu L, Wang Z, Lin Y, et al. Sphingosine kinase 1 enhances the invasion and migration of non-small cell lung cancer cells via the AKT pathway. Oncol Rep. 2015;33(3):1257–1263. [DOI] [PubMed] [Google Scholar]
  • 27.Bradley E, Dasgupta S, Jiang X, et al. Critical role of Spns2, a Sphingosine-1-phosphate transporter, in lung cancer cell survival and migration. PLoS One. 2014;9(10):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kim H-S, Yoon G, Ryu J-Y, et al. Sphingosine kinase 1 is a reliable prognostic factor and a novel therapeutic target for uterine cervical cancer. Oncotarget. 2015;6(29):26746–26756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tanfin Z, Serrano-Sanchez M, Leiber D. ATP-binding cassette ABCC1 is involved in the release of sphingosine 1-phosphate from rat uterine leiomyoma ELT3 cells and late pregnant rat myometrium. Cell Signalling. 2011;23(12):1997–2004. [DOI] [PubMed] [Google Scholar]
  • 30.Dai L, Liu Y, Xie L, Wu X, Qiu L, Di W. Sphingosine kinase 1/sphingosine-1-phosphate (S1P)/S1P receptor axis is involved in ovarian cancer angiogenesis. Oncotarget. 2017;8(43):74947–74961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhang Y, Cai H, Chen S, Sun D, Zhang D, He Y. Exosomal transfer of miR-124 inhibits normal fibroblasts to cancer-associated fibroblasts transition by targeting sphingosine kinase 1 in ovarian cancer. J Cell Biochem. 2019;120(8):13187–13201. [DOI] [PubMed] [Google Scholar]
  • 32.Zhu W, Jarman KE, Lokman NA, et al. CIB2 negatively regulates oncogenic signaling in ovarian cancer via sphingosine kinase 1. Cancer Res. 2017;77(18):4823–4834. [DOI] [PubMed] [Google Scholar]
  • 33.Beach JA, Aspuria P-JP, Cheon D-J, et al. Sphingosine kinase 1 is required for TGF-beta mediated fibroblast-to-myofibroblast differentiation in ovarian cancer. Oncotarget. 2016;7(4):4167–4182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hart PC, Chiyoda T, Liu X, et al. SPHK1 is a novel target of metformin in ovarian cancer. Mol Cancer Res. 2019;17(4):870–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Liu H, Ma Y, He H-W, Zhao W-L, Shao R-G. SPHK1 (sphingosine kinase 1) induces epithelial-mesenchymal transition by promoting the autophagy-linked lysosomal degradation of CDH1/E-cadherin in hepatoma cells. Autophagy. 2017;13(5):900–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lu P-H, Chen M-B, Liu Y-Y, et al. Identification of sphingosine kinase 1 (SphK1) as a primary target of icaritin in hepatocellular carcinoma cells. Oncotarget. 2017;8(14):22800–22810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lu Z, Zhang W, Gao S, et al. MiR-506 suppresses liver cancer angiogenesis through targeting sphingosine kinase 1 (SPHK1) mRNA. Biochem Biophys Res Commun. 2015;468(1-2):8–13. [DOI] [PubMed] [Google Scholar]
  • 38.Lee CF, Dang A, Hernandez E, et al. Activation of sphingosine kinase by lipopolysaccharide promotes prostate cancer cell invasion and metastasis via SphK1/S1PR4/matriptase. Oncogene. 2019;38(28):5580–5598. [DOI] [PubMed] [Google Scholar]
  • 39.Lee S-O, Kim J-S, Lee M-S, Lee H-J. Anti-cancer effect of pristimerin by inhibition of HIF-1 alpha involves the SPHK-1 pathway in hypoxic prostate cancer cells. BMC Cancer. 2016;16(1):701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.McNaughton M, Pitman M, Pitson SM, Pyne NJ, Pyne S.Proteasomal degradation of sphingosine kinase 1 and inhibition of dihydroceramide desaturase by the sphingosine kinase inhibitors, SKi or ABC294640, induces growth arrest in androgen-independent LNCaP-AI prostate cancer cells. Oncotarget. 2016;7(13):16663–16675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Dong X, Wang J, Li T, Xu YP, Li SY. Down regulation of lncRNA MEG3 promotes colorectal adenocarcinoma cell proliferation and inhibits the apoptosis by upregulating TGF-beta1 and its downstream sphingosine kinase 1. Eur Rev Med Pharmacol Sci. 2018;22(23):8265–8272. [DOI] [PubMed] [Google Scholar]
  • 42.Liu SQ, Xu CY, Wu WH, et al. Sphingosine kinase 1 promotes the metastasis of colorectal cancer by inducing the epithelialmesenchymal transition mediated by the FAK/AKT/MMPs axis. Int J Oncol. 2019;54(1):41–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kawahara S, Otsuji Y, Nakamura M, et al. Sphingosine kinase 1 plays a role in the upregulation of CD44 expression through extracellular signal-regulated kinase signaling in human colon cancer cells. Anticancer Drugs. 2013;24(5):473–483. [DOI] [PubMed] [Google Scholar]
  • 44.Bao Y, Guo Y, Zhang C, Fan F, Yang W. Sphingosine kinase 1 and sphingosine-1-phosphate signaling in colorectal cancer. Int J Mol Sci. 2017;18(10):2109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Xu C, Zhang W, Liu S, Wu W, Qin M, Huang J. Activation of the SphK1/ERK/p-ERK pathway promotes autophagy in colon cancer cells. Oncol Lett. 2018;15(6):9719–9724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Salama MF, Carroll B, Adada M, Pulkoski-Gross M, Hannun YA, Obeid LM. A novel role of sphingosine kinase-1 in the invasion and angiogenesis of VHL mutant clear cell renal cell carcinoma. FASEB J. 2015;29(7):2803–2813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Xiong H, Wang J, Guan H, et al. SphK1 confers resistance to apoptosis in gastric cancer cells by downregulating Bim via stimulating Akt/FoxO3a signaling. Oncol Rep. 2014;32(4):1369–1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang Z, Qu H, Gong W, Liu A. Up-regulation and tumor-promoting role of SPHK1 were attenuated by miR-330-3p in gastric cancer. IUBMB Life. 2018;70(11):1164–1176. [DOI] [PubMed] [Google Scholar]
  • 49.Liu ZG, He C, Qu Y, Chen XC, Zhu HL, Xiang B. MiR-659-3p regulates the progression of chronic myeloid leukemia by targeting SPHK1. Int J Clin Exp Pathol. 2018;11(5):2470–2478. [PMC free article] [PubMed] [Google Scholar]
  • 50.Marfe G, Di Stefano C, Gambacurta A, et al. Sphingosine kinase 1 overexpression is regulated by signaling through PI3 K, AKT2, and mTOR in imatinib-resistant chronic myeloid leukemia cells. Exp Hematol. 2011;39(6):653–665. e656. [DOI] [PubMed] [Google Scholar]
  • 51.Abuhusain HJ, Matin A, Qiao Q, et al. A metabolic shift favoring sphingosine 1-phosphate at the expense of ceramide controls glioblastoma angiogenesis. J Biol Chem. 2013;288(52):37355–37364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cattaneo MG, Vanetti C, Samarani M, et al. Cross-talk between sphingosine-1-phosphate and EGFR signaling pathways enhances human glioblastoma cell invasiveness. FEBS Lett. 2018;592(6):949–961. [DOI] [PubMed] [Google Scholar]
  • 53.Quint K, Stiel N, Neureiter D, et al. The role of sphingosine kinase isoforms and receptors S1P1, S1P2, S1P3, and S1P5 in primary, secondary, and recurrent glioblastomas. Tumour Biol. 2014;35(9):8979–8989. [DOI] [PubMed] [Google Scholar]
  • 54.Tsukamoto S, Huang YH, Kumazoe M, et al. Sphingosine kinase-1 protects multiple myeloma from apoptosis driven by cancer-specific inhibition of RTKs. Mol Cancer Ther. 2015;14(10):2303–2312. [DOI] [PubMed] [Google Scholar]
  • 55.Wang D, Bao F, Teng Y, Li Q, Li J. MicroRNA-506-3p initiates mesenchymal-to-epithelial transition and suppresses autophagy in osteosarcoma cells by directly targeting SPHK1. Biosci Biotechnol Biochem. 2019;83(5):836–844. [DOI] [PubMed] [Google Scholar]
  • 56.Cao XZ, Bin H, Zang ZN. MiR-128 suppresses the growth of thyroid carcinoma by negatively regulating SPHK1. Biomed Pharmacother. 2019;109:1960–1966. [DOI] [PubMed] [Google Scholar]
  • 57.Qin Z, Dai L, Trillo-Tinoco J, et al. Targeting sphingosine kinase induces apoptosis and tumor regression for KSHV-associated primary effusion lymphoma. Mol Cancer Ther. 2014;13(1):154–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ogretmen B. Sphingolipid metabolism in cancer signalling and therapy. Nat Rev Cancer. 2017;18(1):33–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Uchida Y. Ceramide signaling in mammalian epidermis. Biochim Biophys Acta. 2014;1841(3):453–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Coant N, Sakamoto W, Mao C, Hannun YA. Ceramidases, roles in sphingolipid metabolism and in health and disease. Adv Biol Regul. 2017;63:122–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Taha TA, El-Alwani M, Hannun YA, Obeid LM. Sphingosine kinase-1 is cleaved by cathepsin B in vitro: identification of the initial cleavage sites for the protease. FEBS Lett. 2006;580(26):6047–6054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kotelevets N, Fabbro D, Huwiler A, Zangemeister-Wittke U. Targeting sphingosine kinase 1 in carcinoma cells decreases proliferation and survival by compromising PKC activity and cytokinesis. PLoS One. 2012;7(6):e39209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Pchejetski D, Böhler T, Stebbing J, Waxman J. Therapeutic potential of targeting sphingosine kinase 1 in prostate cancer. Nat Rev Urol. 2011;8(10):569–678. [DOI] [PubMed] [Google Scholar]
  • 64.Maceyka M, Spiegel S. Sphingolipid metabolites in inflammatory disease. Nature. 2014;510(7503):58–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gijsbers S, Asselberghs S, Herdewijn P, Van Veldhoven PP. 1-O-Hexadecyl-2-desoxy-2-amino-sn-glycerol, a substrate for human sphingosine kinase. Biochim Biophys Acta. 2002;1580(1):1–8. [DOI] [PubMed] [Google Scholar]
  • 66.Lee JY, Han SH, Park MH, et al. Neuronal SphK1 acetylates COX2 and contributes to pathogenesis in a model of Alzheimer’s disease. Nat Commun. 2018;9(1):1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Anelli V, Gault CR, Snider AJ, Obeid LM. Role of sphingosine kinase-1 in paracrine/transcellular angiogenesis and lymphangiogenesis in vitro. FASEB J. 2010;24(8):2727–2738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Pyne NJ, El Buri A, Adams DR, Pyne S. Sphingosine 1-phosphate and cancer. Adv Biol Regul. 2018;68:97–106. [DOI] [PubMed] [Google Scholar]
  • 69.Pitman MR, Pitson SM. Inhibitors of the sphingosine kinase pathway as potential therapeutics. Curr Cancer Drug Targets. 2010;10(4):354–367. [DOI] [PubMed] [Google Scholar]
  • 70.Takabe K, Spiegel S. Export of sphingosine-1-phosphate and cancer progression. J Lipid Res. 2014;55(9):1839–1846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Zheng X, Li W, Ren L, et al. The sphingosine kinase-1/sphingosine-1-phosphate axis in cancer: potential target for anti-cancer therapy. Pharmacol Ther. 2018;195:85–99. [DOI] [PubMed] [Google Scholar]
  • 72.Vu TM, Ishizu AN, Foo JC, et al. Mfsd2b is essential for the sphingosine-1-phosphate export in erythrocytes and platelets. Nature. 2017;550(7677):524–528. [DOI] [PubMed] [Google Scholar]
  • 73.Pyne NJ, McNaughton M, Boomkamp S, et al. Role of sphingosine 1-phosphate receptors, sphingosine kinases and sphingosine in cancer and inflammation. Adv Biol Regul. 2016;60:151–159. [DOI] [PubMed] [Google Scholar]
  • 74.Hait NC, Allegood J, Maceyka M, et al. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science (New York, NY). 2009;325(5945):1254–1257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Alvarez SE, Harikumar KB, Hait NC, et al. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature. 2010;465(7301):1084–1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Strub GM, Paillard M, Liang J, et al. Sphingosine-1-phosphate produced by sphingosine kinase 2 in mitochondria interacts with prohibitin 2 to regulate complex IV assembly and respiration. FASEB J. 2011;25(2):600–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Nishino S, Yamashita H, Tamori M, et al. Translocation and activation of sphingosine kinase 1 by ceramide-1-phosphate. J Cell Biochem. 2018;120(4):5396–5408. [DOI] [PubMed] [Google Scholar]
  • 78.Wallington-Beddoe CT, Bradstock KF, Bendall LJ. Oncogenic properties of sphingosine kinases in haematological malignancies. Br J Haematol. 2013;161(5):623–638. [DOI] [PubMed] [Google Scholar]
  • 79.Johnson KR, Becker KP, Facchinetti MM, Hannun YA, Obeid LM.PKC-dependent activation of sphingosine kinase 1 and translocation to the plasma membrane. Extracellular release of sphingosine-1-phosphate induced by phorbol 12-myristate 13-acetate (PMA). J Biol Chem. 2002;277(38):35257–35262. [DOI] [PubMed] [Google Scholar]
  • 80.Sutherland CM, Moretti PA, Hewitt NM, Bagley CJ, Vadas MA, Pitson SM. The calmodulin-binding site of sphingosine kinase and its role in agonist-dependent translocation of sphingosine kinase 1 to the plasma membrane. J Biol Chem. 2006;281(17):11693–11701. [DOI] [PubMed] [Google Scholar]
  • 81.Pyne S, Lee SC, Long J, Pyne NJ. Role of sphingosine kinases and lipid phosphate phosphatases in regulating spatial sphingosine 1-phosphate signalling in health and disease. Cell Signal. 2009;21(1):14–21. [DOI] [PubMed] [Google Scholar]
  • 82.Alemany R, van Koppen CJ, Danneberg K, Ter Braak M, Meyer Zu Heringdorf D. Regulation and functional roles of sphingosine kinases. Naunyn Schmiedebergs Arch Pharmacol. 2007;374(5-6):413–428. [DOI] [PubMed] [Google Scholar]
  • 83.Jarman KE, Moretti PA, Zebol JR, Pitson SM. Translocation of sphingosine kinase 1 to the plasma membrane is mediated by calcium- and integrin-binding protein 1. J Biol Chem. 2010;285(1):483–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Zhu W, Gliddon BL, Jarman KE, et al. CIB1 contributes to oncogenic signalling by Ras via modulating the subcellular localisation of sphingosine kinase 1. Oncogene. 2017;36(18):2619–2627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wang X, Peng X, Zhang X, et al. The emerging roles of CIB1 in cancer. Cell Physiol Biochem. 2017;43(4):1413–1424. [DOI] [PubMed] [Google Scholar]
  • 86.Bayraktar O, Ozkirimli E, Ulgen K. Sphingosine kinase 1 (SK1) allosteric inhibitors that target the dimerization site. Comput Biol Chem. 2017;69:64–76. [DOI] [PubMed] [Google Scholar]
  • 87.Marfe G, Mirone G, Shukla A, Di Stefano C. Sphingosine kinases signalling in carcinogenesis. Mini Rev Med Chem. 2015;15(4):300–314. [DOI] [PubMed] [Google Scholar]
  • 88.Heffernan-Stroud LA, Obeid LM. Sphingosine kinase 1 in cancer. Adv Cancer Res. 2013;117:201–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zheng X, Li W, Ren L, et al. The sphingosine kinase-1/sphingosine-1-phosphate axis in cancer: potential target for anti-cancer therapy. Pharmacol Ther. 2019;195:85–99. [DOI] [PubMed] [Google Scholar]
  • 90.Maceyka M, Harikumar KB, Milstien S, Spiegel S. Sphingosine-1-phosphate signaling and its role in disease. Trends Cell Biol. 2012;22(1):50–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhang Y, Wang Y, Wan Z, Liu S, Cao Y, Zeng Z. Sphingosine kinase 1 and cancer: a systematic review and meta-analysis. PLoS One. 2014;9(2):e90362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Xia P, Gamble JR, Wang L, et al. An oncogenic role of sphingosine kinase. Curr Biol. 2000;10(23):1527–1530. [DOI] [PubMed] [Google Scholar]
  • 93.Saha S, Panigrahi DP, Patil S, Bhutia SK.Autophagy in health and disease: a comprehensive review. Biomed Pharmacother. 2018;104:485–495. [DOI] [PubMed] [Google Scholar]
  • 94.Mizushima N. A brief history of autophagy from cell biology to physiology and disease. Nat Cell Biol. 2018;20(5):521–527. [DOI] [PubMed] [Google Scholar]
  • 95.Xu X, Lai Y, Hua ZC. Apoptosis and apoptotic body: disease message and therapeutic target potentials. Biosci Rep. 2019;39(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Feng Y, He D, Yao Z, Klionsky DJ. The machinery of macroautophagy. Cell Res. 2014;24(1):24–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wang X, Song J, Wu Z, Fan B, Mode X. Dual roles of CD38 in autophagy. Yangtze Med. 2017;1(1):8–19. [Google Scholar]
  • 98.Young MM, Takahashi Y, Fox TE, Yun JK, Kester M, Wang HG. Sphingosine kinase 1 cooperates with autophagy to maintain endocytic membrane trafficking. Cell Rep. 2016;17(6):1532–1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Moruno Manchon JF, Uzor NE, Dabaghian Y, Furr-Stimming EE, Finkbeiner S, Tsvetkov AS. Cytoplasmic sphingosine-1-phosphate pathway modulates neuronal autophagy. Sci Rep. 2015;5:15213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Liu H, Ma Y, He HW, Zhao WL, Shao RG. SPHK1 (sphingosine kinase 1) induces epithelial-mesenchymal transition by promoting the autophagy-linked lysosomal degradation of CDH1/E-cadherin in hepatoma cells. Autophagy. 2017;13(5):900–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Du C, Ren Y, Yao F, et al. Sphingosine kinase 1 protects renal tubular epithelial cells from renal fibrosis via induction of autophagy. Int J Biochem Cell Biol. 2017;90:17–28. [DOI] [PubMed] [Google Scholar]
  • 102.Slattum G, Gu Y, Sabbadini R, Rosenblatt J. Autophagy in oncogenic K-Ras promotes basal extrusion of epithelial cells by degrading S1P. Curr Biol. 2014;24(1):19–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Jiang W, Ogretmen B. Autophagy paradox and ceramide. Biochim Biophys Acta. 2014;1841(5):783–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Moruno-Manchon JF, Uzor NE, Ambati CR, et al. Sphingosine kinase 1-associated autophagy differs between neurons and astrocytes. Cell Death Dis. 2018;9(5):521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Moruno Manchon JF, Uzor NE, Finkbeiner S, Tsvetkov AS. SPHK1/sphingosine kinase 1-mediated autophagy differs between neurons and SH-SY5Y neuroblastoma cells. Autophagy. 2016;12(8):1418–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lorin S, Codogno P, Djavaheri-Mergny M. Autophagy: a new concept in cancer research [in French]. Bull Cancer. 2008;95(1):43–50. [DOI] [PubMed] [Google Scholar]
  • 107.Choi KS. Autophagy and cancer. Exp Mol Med. 2012;44(2):109–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Jin S, Wei J, You L, Liu H, Qian W.Autophagy regulation and its dual role in blood cancers: a novel target for therapeutic development (Review). Oncol Rep. 2018;39(6):2473–2481. [DOI] [PubMed] [Google Scholar]
  • 109.Heffernan-Stroud LA, Helke KL, Jenkins RW, De Costa AM, Hannun YA, Obeid LM. Defining a role for sphingosine kinase 1 in p53-dependent tumors. Oncogene. 2012;31(9):1166–1175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Watson DG, Tonelli F, Alossaimi M, et al. The roles of sphingosine kinases 1 and 2 in regulating the Warburg effect in prostate cancer cells. Cell Signal. 2013;25(4):1011–1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Carroll BL, Bonica J, Shamseddine AA, Hannun YA, Obeid LM. A role for caspase-2 in sphingosine kinase 1 proteolysis in response to doxorubicin in breast cancer cells—implications for the CHK1-suppressed pathway. FEBS Open Bio. 2018;8(1):27–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Song L, Xiong H, Li J, et al. Sphingosine kinase-1 enhances resistance to apoptosis through activation of PI3K/Akt/NF-kappaB pathway in human non-small cell lung cancer. Clin Cancer Res. 2011;17(7):1839–1849. [DOI] [PubMed] [Google Scholar]
  • 113.Etemadi N, Chopin M, Anderton H, et al. TRAF2 regulates TNF and NF-kappaB signalling to suppress apoptosis and skin inflammation independently of Sphingosine kinase 1. Elife. 2015;4:e10592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kapitonov D, Allegood JC, Mitchell C, et al. Targeting sphingosine kinase 1 inhibits Akt signaling, induces apoptosis, and suppresses growth of human glioblastoma cells and xenografts. Cancer Res. 2009;69(17):6915–6923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Yang YL, Ji C, Cheng L, et al. Sphingosine kinase-1 inhibition sensitizes curcumin-induced growth inhibition and apoptosis in ovarian cancer cells. Cancer Sci. 2012;103(8):1538–1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Tam C, Wong JH, Cheung RCF, Zuo T, Ng TB. Therapeutic potentials of short interfering RNAs. Appl Microbiol Biotechnol. 2017;101(19):7091–7111. [DOI] [PubMed] [Google Scholar]
  • 117.Costas MA, Rubio MF.Autophagy. A strategy for cell survival [in Spanish]. Medicina. 2017;77(4):314–320. [PubMed] [Google Scholar]
  • 118.Kroemer G, Marino G, Levine B. Autophagy and the integrated stress response. Molecular Cell. 2010;40(2):280–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Lavieu G, Scarlatti F, Sala G, et al. Regulation of autophagy by sphingosine kinase 1 and its role in cell survival during nutrient starvation. J Biol Chem. 2006;281(13):8518–8527. [DOI] [PubMed] [Google Scholar]
  • 120.Su Z, Yang Z, Xu Y, Chen Y, Yu Q. Apoptosis, autophagy, necroptosis, and cancer metastasis. Mol Cancer. 2015;14:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Delou JM, Biasoli D, Borges HL. The complex link between apoptosis and autophagy: a promising new role for RB. An Acad Bras Cienc. 2016;88(4):2257–2275. [DOI] [PubMed] [Google Scholar]
  • 122.Bhat P Kriel J Shubha Priya B Basappa, Shivananju NS Loos B.. Modulating autophagy in cancer therapy: advancements and challenges for cancer cell death sensitization. Biochem Pharmacol. 2018;147:170–182. [DOI] [PubMed] [Google Scholar]
  • 123.Ruijtenberg S, van den Heuvel S.Coordinating cell proliferation and differentiation: antagonism between cell cycle regulators and cell type-specific gene expression. Cell Cycle. 2016;15(2):196–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Shirai K, Kaneshiro T, Wada M, et al. A role of sphingosine kinase 1 in head and neck carcinogenesis. Cancer Prev Res (Philadelphia, Pa). 2011;4(3):454–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Furuya H, Shimizu Y, Tamashiro PM, et al. Sphingosine kinase 1 expression enhances colon tumor growth. J Transl Med. 2017;15(1):120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zhang H, Liu L, Shi M, Liu X, Tang H. Sphingosine kinase 1 promotes glioma cell proliferation under hypoxia via calcium signaling [in Chinese]. Nan Fang Yi Ke Da Xue Xue Bao. 2015;35(7):1014–1018. [PubMed] [Google Scholar]
  • 127.Datta A, Loo SY, Huang B, et al. SPHK1 regulates proliferation and survival responses in triple-negative breast cancer. Oncotarget. 2014;5(15):5920–5933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Nazouri AS, Asadpour O, Dabiri S, Pourseyedi B, Lashkarizadeh MR, Zianalinejad H. High expression of sphingosine kinase 1 in estrogen and progesterone receptors-negative breast cancer. Iran J Pathol. 2017;12(3):218–224. [PMC free article] [PubMed] [Google Scholar]
  • 129.Almejun MB, Borge M, Colado A, et al. Sphingosine kinase 1 participates in the activation, proliferation and survival of chronic lymphocytic leukemia cells. Haematologica. 2017;102(7):e257–e260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Li J, Song Z, Wang Y, et al. Overexpression of SphK1 enhances cell proliferation and invasion in triple-negative breast cancer via the PI3K/AKT signaling pathway. Tumour Biol. 2016;37(8):10587–10593. [DOI] [PubMed] [Google Scholar]
  • 131.Lyu Y, Xu X, Yun J, et al. TNF-alpha regulates the proliferation of human breast cancer cells via regulation of ceramide content [in Chinese]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2017;33(10):1303–1309. [PubMed] [Google Scholar]
  • 132.Yan G, Chen S, You B, Sun J. Activation of sphingosine kinase-1 mediates induction of endothelial cell proliferation and angiogenesis by epoxyeicosatrienoic acids. Cardiovasc Res. 2008;78(2):308–314. [DOI] [PubMed] [Google Scholar]
  • 133.Goel HL, Mercurio AM. VEGF targets the tumour cell. Nature Rev Cancer. 2013;13(12):871–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Alshaker H, Wang Q, Bohler T, et al. Combination of RAD001 (everolimus) and docetaxel reduces prostate and breast cancer cell VEGF production and tumour vascularisation independently of sphingosine-kinase-1. Sci Rep. 2017;7(1):3493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Hernandez-Coronado CG, Guzman A, Rodriguez A, et al. Sphingosine-1-phosphate, regulated by FSH and VEGF, stimulates granulosa cell proliferation. Gen Comp Endocrinol. 2016;236:1–8. [DOI] [PubMed] [Google Scholar]
  • 136.Paz-Filho G, Mastronardi CA, Licinio J. Leptin treatment: facts and expectations. Metabolism. 2015;64(1):146–156. [DOI] [PubMed] [Google Scholar]
  • 137.Alshaker H, Krell J, Frampton AE, et al. Leptin induces upregulation of sphingosine kinase 1 in oestrogen receptor-negative breast cancer via Src family kinase-mediated, janus kinase 2-independent pathway. Breast Cancer Res: BCR. 2014;16(5):426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Zhou Y, Han Y, Zhang Z, et al. MicroRNA-124 upregulation inhibits proliferation and invasion of osteosarcoma cells by targeting sphingosine kinase 1. Hum Cell. 2017;30(1):30–40. [DOI] [PubMed] [Google Scholar]
  • 139.Yu H, Duan P, Zhu H, Rao D. miR-613 inhibits bladder cancer proliferation and migration through targeting SphK1. Am J Transl Res. 2017;9(3):1213–1221. [PMC free article] [PubMed] [Google Scholar]
  • 140.Wiman KG, Zhivotovsky B. Understanding cell cycle and cell death regulation provides novel weapons against human diseases. J Int Med. 2017;281(5):483–495. [DOI] [PubMed] [Google Scholar]
  • 141.Rex K, Jeffries S, Brown ML, et al. Sphingosine kinase activity is not required for tumor cell viability. PLoS One. 2013;8(7):e68328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Albinet V, Bats ML, Huwiler A, et al. Dual role of sphingosine kinase-1 in promoting the differentiation of dermal fibroblasts and the dissemination of melanoma cells. Oncogene. 2014;33(26):3364–3373. [DOI] [PubMed] [Google Scholar]
  • 143.Sukocheva OA. Expansion of sphingosine kinase and sphingosine-1-phosphate receptor function in normal and cancer cells: from membrane restructuring to mediation of estrogen signaling and stem cell programming. Int J Mol Sci. 2018;19(2):420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Fu P, Ebenezer DL, Berdyshev EV, et al. Role of sphingosine kinase 1 and S1P transporter Spns2 in HGF-mediated lamellipodia formation in lung endothelium. J Biol Chem. 2016;291(53):27187–27203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Gao Z, Wang H, Xiao FJ, et al. SIRT1 mediates Sphk1/S1P-induced proliferation and migration of endothelial cells. Int J Biochem Cell Biol. 2016;74:152–160. [DOI] [PubMed] [Google Scholar]
  • 146.Oh YT, Yue P, Sun SY. DR5 suppression induces sphingosine-1-phosphate-dependent TRAF2 polyubiquitination, leading to activation of JNK/AP-1 and promotion of cancer cell invasion. Cell Commun Signal. 2017;15(1):18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Yagoub D, Wilkins MR, Lay AJ, et al. Sphingosine kinase 1 isoform-specific interactions in breast cancer. Mol Endocrinol (Baltimore, Md). 2014;28(11):1899–1915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Schiefler C, Piontek G, Doescher J, et al. Inhibition of SphK1 reduces radiation-induced migration and enhances sensitivity to cetuximab treatment by affecting the EGFR / SphK1 crosstalk. Oncotarget. 2014;5(20):9877–9888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Long J, Xie Y, Yin J, Lu W, Fang S. SphK1 promotes tumor cell migration and invasion in colorectal cancer. Tumour Biol. 2016;37(5):6831–6836. [DOI] [PubMed] [Google Scholar]
  • 150.Xu CY, Liu SQ, Qin MB, et al. SphK1 modulates cell migration and EMT-related marker expression by regulating the expression of p-FAK in colorectal cancer cells. Int J Mol Med. 2017;39(5):1277–1284. [DOI] [PubMed] [Google Scholar]
  • 151.Patmanathan SN, Johnson SP, Lai SL, et al. Aberrant expression of the S1P regulating enzymes, SPHK1 and SGPL1, contributes to a migratory phenotype in OSCC mediated through S1PR2. Sci Rep. 2016;6:25650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Tamashiro PM, Furuya H, Shimizu Y, Kawamori T. Sphingosine kinase 1 mediates head & neck squamous cell carcinoma invasion through sphingosine 1-phosphate receptor 1. Cancer Cell Inte. 2014;14(1):76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Vijayan M, Seo YJ, Pritzl CJ, Squires SA, Alexander S, Hahm B. Sphingosine kinase 1 regulates measles virus replication. Virology. 2014;450-451:55–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lu ZP, Xiao ZL, Yang Z, et al. Hepatitis B virus X protein promotes human hepatoma cell growth via upregulation of transcription factor AP2alpha and sphingosine kinase 1. Acta Pharmacol Sin. 2015;36(10):1228–1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Bao M, Chen Z, Xu Y, et al. Sphingosine kinase 1 promotes tumour cell migration and invasion via the S1P/EDG1 axis in hepatocellular carcinoma. Liver Int. 2012;32(2):331–338. [DOI] [PubMed] [Google Scholar]
  • 156.Lee HM, Lo KW, Wei W, et al. Oncogenic S1P signalling in EBV-associated nasopharyngeal carcinoma activates AKT and promotes cell migration through S1P receptor 3. J Pathol. 2017;242(1):62–72. [DOI] [PubMed] [Google Scholar]
  • 157.Liang W, Xie Z, Cui W, et al. Comprehensive gene and microRNA expression profiling reveals a role for miRNAs in the oncogenic roles of SphK1 in papillary thyroid cancer. J Cancer Res Clin Oncol. 2017;143(4):601–611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Qiu W, Yang Z, Fan Y, Zheng Q. MicroRNA-613 inhibits cell growth, migration and invasion of papillary thyroid carcinoma by regulating SphK2. Oncotarget. 2016;7(26):39907–39915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Zhang Z, Yan Z, Yuan Z, Sun Y, He H, Mai C. SPHK1 inhibitor suppresses cell proliferation and invasion associated with the inhibition of NF-kappaB pathway in hepatocellular carcinoma. Tumour Biol. 2015;36(3):1503–1509. [DOI] [PubMed] [Google Scholar]
  • 160.Viallard C, Larrivee B. Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis. 2017;20(4):409–426. [DOI] [PubMed] [Google Scholar]
  • 161.Lee JW, Ryu JY, Yoon G, et al. Sphingosine kinase 1 as a potential therapeutic target in epithelial ovarian cancer. Int J Cancer. 2015;137(1):221–229. [DOI] [PubMed] [Google Scholar]
  • 162.Steeg PS. Targeting metastasis. Nat Rev Cancer. 2016;16(4):201–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Kim HS, Yoon G, Ryu JY, et al. Sphingosine kinase 1 is a reliable prognostic factor and a novel therapeutic target for uterine cervical cancer. Oncotarget. 2015;6(29):26746–26756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Mukhopadhyay P, Ramanathan R, Takabe K. S1P promotes breast cancer progression by angiogenesis and lymphangiogenesis. Breast Cancer Manag. 2015;4(5):241–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Evangelisti C, Evangelisti C, Buontempo F, et al. Therapeutic potential of targeting sphingosine kinases and sphingosine 1-phosphate in hematological malignancies. Leukemia. 2016;30(11):2142–2151. [DOI] [PubMed] [Google Scholar]
  • 166.Nema R, Vishwakarma S, Agarwal R, Panday RK, Kumar A. Emerging role of sphingosine-1-phosphate signaling in head and neck squamous cell carcinoma. Onco Targets Ther. 2016;9:3269–3280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Lu Z, Xiao Z, Liu F, et al. Long non-coding RNA HULC promotes tumor angiogenesis in liver cancer by upregulating sphingosine kinase 1 (SPHK1). Oncotarget. 2016;7(1):241–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Huang WC, Nagahashi M, Terracina KP, Takabe K.Emerging role of sphingosine-1-phosphate in inflammation, cancer, and lymphangiogenesis. Biomolecules. 2013;3(3):408–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Schwiebs A, Herrero San Juan M, Schmidt KG, et al. Cancer-induced inflammation and inflammation-induced cancer in colon: a role for S1P lyase. Oncogene. 2019;38(24):4788–4803. [DOI] [PubMed] [Google Scholar]
  • 170.Alvarez SE, Milstien S, Spiegel S. Autocrine and paracrine roles of sphingosine-1-phosphate. Trends Endocrinol Metab. 2007;18(8):300–307. [DOI] [PubMed] [Google Scholar]
  • 171.Snider AJ, Kawamori T, Bradshaw SG, et al. A role for sphingosine kinase 1 in dextran sulfate sodium-induced colitis. FASEB J. 2009;23(1):143–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Adada MM, Orr-Gandy KA, Snider AJ, et al. Sphingosine kinase 1 regulates tumor necrosis factor-mediated RANTES induction through p38 mitogen-activated protein kinase but independently of nuclear factor kappaB activation. J Biol Chem. 2013;288(38):27667–27679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Pchejetski D, Nunes J, Coughlan K, et al. The involvement of sphingosine kinase 1 in LPS-induced Toll-like receptor 4-mediated accumulation of HIF-1α protein, activation of ASK1 and production of the pro-inflammatory cytokine IL-6. Immunol Cell Biol. 2011;89(2):268–274. [DOI] [PubMed] [Google Scholar]
  • 174.Pyne NJ, Ohotski J, Bittman R, Pyne S. The role of sphingosine 1-phosphate in inflammation and cancer. Adv Biol Regul. 2014;54:121–129. [DOI] [PubMed] [Google Scholar]
  • 175.Nagahashi M, Abe M, Sakimura K, Takabe K, Wakai T. The role of sphingosine-1-phosphate in inflammation and cancer progression. Cancer Sci. 2018;109(12):3671–3678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Liang J, Nagahashi M, Kim EY, et al. Sphingosine-1-phosphate links persistent STAT3 activation, chronic intestinal inflammation, and development of colitis-associated cancer. Cancer Cell. 2013;23(1):107–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Degagne E, Saba JD.S1pping fire: Sphingosine-1-phosphate signaling as an emerging target in inflammatory bowel disease and colitis-associated cancer. Clin Exp Gastroenterol. 2014;7:205–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Xie Z, Liu H, Geng M. Targeting sphingosine-1-phosphate signaling for cancer therapy. Sci China Life Sci. 2017;60(6):585–600. [DOI] [PubMed] [Google Scholar]
  • 179.Sukocheva OA, Furuya H, Ng ML, et al. Sphingosine kinase and sphingosine-1-phosphate receptor signaling pathway in inflammatory gastrointestinal disease and cancers: a novel therapeutic target. Pharmacol Ther. 2020;207:107464. [DOI] [PubMed] [Google Scholar]
  • 180.Hammad SM, Crellin HG, Wu BX, Melton J, Anelli V, Obeid LM. Dual and distinct roles for sphingosine kinase 1 and sphingosine 1 phosphate in the response to inflammatory stimuli in RAW macrophages. Prostaglandins Other Lipid Mediat. 2008;85(3-4):107–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Pettus BJ, Bielawski J, Porcelli AM, et al. The sphingosine kinase 1/sphingosine-1-phosphate pathway mediates COX-2 induction and PGE2 production in response to TNF-alpha. FASEB J. 2003;17(11):1411–1421. [DOI] [PubMed] [Google Scholar]
  • 182.Xiong Y, Lee HJ, Mariko B, et al. Sphingosine kinases are not required for inflammatory responses in macrophages. J Biol Chem. 2016;291(21):11465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Dai W, Yan J, Chen G, Hu G, Zhou X, Zeng X. AQP4knockout alleviates the lipopolysaccharide-induced inflammatory response in astrocytes via SPHK1/MAPK/AKT signaling. Int J Mol Med. 2018;42(3):1716–1722. [DOI] [PubMed] [Google Scholar]
  • 184.Nagahashi M, Yamada A, Katsuta E, et al. Targeting the SphK1/S1P/S1PR1 Axis that links obesity, chronic inflammation, and breast cancer metastasis. Cancer Res. 2018;78(7):1713–1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Cuvillier O. Downregulating sphingosine kinase-1 for cancer therapy. Expert Opin Ther Targets. 2008;12(8):1009–1020. [DOI] [PubMed] [Google Scholar]
  • 186.Pitman MR, Costabile M, Pitson SM. Recent advances in the development of sphingosine kinase inhibitors. Cell Signal. 2016;28(9):1349–1363. [DOI] [PubMed] [Google Scholar]
  • 187.Nagahashi M, Ramachandran S, Kim EY, et al. Sphingosine-1-phosphate produced by sphingosine kinase 1 promotes breast cancer progression by stimulating angiogenesis and lymphangiogenesis. Cancer Res. 2012;72(3):726–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Madhunapantula SV, Hengst J, Gowda R, Fox TE, Yun JK, Robertson GP. Targeting sphingosine kinase-1 to inhibit melanoma. Pigment Cell Melanoma Res. 2012;25(2):259–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Ren S, Xin C, Pfeilschifter J, Huwiler A. A novel mode of action of the putative sphingosine kinase inhibitor 2-(p-hydroxyanilino)-4-(p-chlorophenyl) thiazole (SKI II): induction of lysosomal sphingosine kinase 1 degradation. Cell Physiol Biochem. 2010;26(1):97–104. [DOI] [PubMed] [Google Scholar]
  • 191.Gao P, Peterson YK, Smith RA, Smith CD. Characterization of isoenzyme-selective inhibitors of human sphingosine kinases. PLoS One. 2012;7(9):e44543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Xiang Y, Hirth B, Kane JL, Jr, et al. Discovery of novel sphingosine kinase-1 inhibitors. Part 2. Bioorg Med Chem Lett. 2010;20(15):4550–4554. [DOI] [PubMed] [Google Scholar]
  • 193.Billich A, Bornancin F, Devay P, Mechtcheriakova D, Urtz N, Baumruker T. Phosphorylation of the immunomodulatory drug FTY720 by sphingosine kinases. J Biol Chem. 2003;278(48):47408–47415. [DOI] [PubMed] [Google Scholar]
  • 194.Brinkmann V, Billich A, Baumruker T, et al. Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat Rev Drug Discov. 2010;9(11):883–897. [DOI] [PubMed] [Google Scholar]
  • 195.Chaudhry BZ, Cohen JA, Conway DS. Sphingosine 1-phosphate receptor modulators for the treatment of multiple sclerosis. Neurotherapeutics. 2017;14(4):859–873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.White C, Alshaker H, Cooper C, Winkler M, Pchejetski D. The emerging role of FTY720 (Fingolimod) in cancer treatment. Oncotarget. 2016;7(17):23106–23127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Wallington-Beddoe CT, Don AS, Hewson J, et al. Disparate in vivo efficacy of FTY720 in xenograft models of Philadelphia positive and negative B-lineage acute lymphoblastic leukemia. PLoS One. 2012;7(5):e36429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Alshaker H, Srivats S, Monteil D, Wang Q, Low CMR, Pchejetski D. Field template-based design and biological evaluation of new sphingosine kinase 1 inhibitors. Breast Cancer Res Treat. 2018;172(1):33–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Coward J, Ambrosini G, Musi E, et al. Safingol (L-threo-sphinganine) induces autophagy in solid tumor cells through inhibition of PKC and the PI3-kinase pathway. Autophagy. 2009;5(2):184–193. [DOI] [PubMed] [Google Scholar]
  • 200.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. [DOI] [PubMed] [Google Scholar]
  • 201.Cuvillier O, Ader I, Bouquerel P, et al. Activation of sphingosine kinase-1 in cancer: implications for therapeutic targeting. Curr Mol Pharmacol. 2010;3(2):53–65. [DOI] [PubMed] [Google Scholar]
  • 202.Gustin DJ, Li Y, Brown ML, et al. Structure guided design of a series of sphingosine kinase (SphK) inhibitors. Bioorg Med Chem Lett. 2013;23(16):4608–4616. [DOI] [PubMed] [Google Scholar]
  • 203.Patwardhan NN, Morris EA, Kharel Y, et al. Structure-activity relationship studies and in vivo activity of guanidine-based sphingosine kinase inhibitors: discovery of SphK1- and SphK2-selective inhibitors. J Med Chem. 2015;58(4):1879–1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Schnute ME, McReynolds MD, Kasten T, et al. modulation of cellular S1P levels with a novel, potent and specific inhibitor of sphingosine kinase-1. Biochem J. 2012;444(1):79–88. [DOI] [PubMed] [Google Scholar]
  • 205.McReynolds MD, Carroll J, Chrencik J, et al. Discovery of a potent and selective sphingosine kinase 1 inhibitor through the molecular combination of chemotype-distinct screening hits. J Med Chem. 2017;60(6):2562–2572. [DOI] [PubMed] [Google Scholar]
  • 206.Wang J, Knapp S, Pyne NJ, Pyne S, Elkins JM. Crystal structure of sphingosine kinase 1 with PF-543. ACS Med Chem Lett. 2014;5(12):1329–1333. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cancer Control : Journal of the Moffitt Cancer Center are provided here courtesy of SAGE Publications

RESOURCES