Abstract
Traditional methods of cancer treatment are limited in their efficacy due to both inherent and acquired factors. Many different studies have shown that the generation of ceramide in response to cytotoxic therapy is generally an important step leading to cell death. Cancer cells employ different methods to both limit ceramide generation and to remove ceramide in order to become resistant to treatment. Furthermore, sphingosine kinase activity, which phosphorylates sphingosine the product of ceramide hydrolysis, has been linked to multidrug resistance, and can act as a strong survival factor. This review will examine several of the most frequently used cancer therapies and their effect on both ceramide generation and the mechanisms employed to remove it. The development and use of inhibitors of sphingosine kinase will be focused upon as an example of how targeting sphingolipid metabolism may provide an effective means to improve treatment response rates and reduce associated treatment toxicity.
Keywords: Cancer, ceramide, chemoresistance, sphingosine kinase, sphingosine-1-phosphate, inhibitors
Introduction
The first generally accepted case of a chemotherapeutic agent being successfully used to treat cancer was in 1865 by Lissauer using potassium arsenite on a patient with chronic myelogenous leukemia [1]. There is reference from texts over 1000 years old of the use of wild chervil roots (containing deoxypodophyllotoxin) for the treatment of cancer [2]. However, for the first half of the twentieth century, radiation therapy (along with surgery) was generally the most effective cancer therapy. The discovery that nitrogen mustard (from mustard gas) could be used in the treatment of lymphoma in 1943 began a new era in the development and use of more effective cancer therapies [3]. Unfortunately, follow ups of patients treated with nitrogen mustard showed that remission was both incomplete and short [4]. This has been a recurring problem faced during cancer therapy throughout medical history.
The loss of normal growth control in cancer cells provides one common characteristic that can be generally targeted by cancer therapy [5]. Radiation therapy produces DNA damage in the cancer cell leading to its death by apoptosis or replicative shutdown due to irreparable and widespread DNA damage [6]. The majority of traditional chemotherapeutic drugs used today target various cellular components vital for cellular division [7].
Many connections have been developing between cancer therapies and sphingolipid metabolism. Several cancer treatments often result in the generation of ceramide which has been implicated in mediating or at least regulating the cell death response. As such, abrogation of ceramide generation is one of the mechanisms exploited by cancer cells in order to evade treatment as first demonstrated with induction of glucosylceramide synthesis [8]. Another common survival strategy employed by cancer cells is the synthesis of sphingosine 1-phosphate (S1P), formed by phosphorylating the product of ceramide hydrolysis [9]. In this review the role of sphingolipids in the induction of apoptosis or resistance to some of the most commonly used cancer therapies will be examined. In addition, strategies inhibiting sphingosine kinase (SK) activity will be reviewed, along with their effects on chemotherapy and their effect on chemoresistance. It is hoped that this will highlight the potential of manipulating the sphingolipid pathway as a means to both increase the efficiency of treatment and potentially reduce its more serious side effects.
Part 1. Sphingolipids and the initiation of apoptosis
Sphingolipids comprise a broad group of both structural and bioactive lipids [10, 11], and they are generated de novo in the endoplasmic reticulum from non-sphingolipid precursors. Ceramide can be considered the central hub of the sphingolipid pathway, and its generation has been observed following diverse treatments that can induce many different cellular effects including apoptosis, growth arrest, senescence and differentiation [12]. Induction of ceramide can be achieved either through hydrolysis of sphingomyelin by sphingomyelinases, hydrolysis of cerebrosides, or via the de novo pathway by ceramide synthases [13, 14]. The sphingomyelinase and de novo pathways are the best studied so far.
1.1. Generation of ceramide
1.1.1. Sphingomyelinases
Sphingomyelinases exist as three major groups depending on the pH required for optimal activity, neutral, acid and alkaline, and can hydrolyze sphingomyelin to form ceramide [15]. The potential role of sphingomyelinases in cancer therapy remains to be properly elucidated. Studies have shown levels of alkaline SMase activity are reduced in human colorectal carcinomas, suggesting a role in the development of malignancy [16]. Treatment of several diverse cell lines (including multidrug resistant prostate cancer cell line DU-1. 45) with either Sunitinib or SU11652, both multitargeting-tyrosine kinase inhibitors, inhibited acid sphingomyelinase (ASMase) activity leading to lysosomal destabilization and cell death [17]. Another somewhat contradictory report showed that treatment of implanted hepatocellular carcinoma cells with both sorafenib (a multi-serine/threonine kinase inhibitor) and recombinant ASMase increased cell death relative to sorafenib alone [18]. This is backed up by a study showing that liver ASMase activity can inhibit the growth of metastatic colon cancer [19]. It therefore appears that the activity of ASMase in promoting cancer death may be tied to both the cell type and the protein kinases that are present.
At present three different neutral SMase (nSMase) isoforms, encoded in separate genes, have been identified in mammals [20]. In the mid 1990’s a role for nSMase activity in chemotherapy was reported in 1-β-D-Arabinofuranosylcytosine (Ara-C) treatment of HL-60 (human promyelocytic leukemia cells) [21]. A role for nSMase in cell growth was suggested when cells overexpressing nSMase 2 exhibited slower proliferation, while growth arrested MCF-7 breast cancer cells had increased levels of nSMase 2 [22, 23]. Conversely, treatment of human mammary epithelial cells 184B5/HER with either exogenous nSMase or C2 or C6 ceramide could increase both cyclooxygenase 2 gene and protein expression and increase proliferation [24]. Analysis of nSMase genes showed that 5% of human acute myeloid leukemias and 6% of acute lymphoid leukemias tested had inactivating mutations [25]. Furthermore, nSMase 2 has been reported to promote angiogenesis and regulate metastasis through regulation of exosomal microRNA secretion [26]. Different isoforms of nSMase have been found within the nuclear envelope, nuclear matrix and associated with chromatin [27]. SMase activity is associated with chromatin unwinding and the initiation of replication, although nuclear SMase activity can also induce an apoptotic response [27, 28]. Interestingly, SMase-treatment of RNAse-resistant RNA can render it more sensitive to degradation, suggesting a role for sphingomyelin in RNA stability [29].
1.1.2. Ceramide synthases
Ceramide synthases are integral membrane proteins localized in the endoplasmic reticulum, and 6 different enzymes have been identified and have been named CerS1-6 [30, 31]. Each CerS shows specificity towards a fatty acyl CoA of different chain length, resulting in the synthesis of ceramides of different chain length [31]. Ceramide generated by CerS can be transported to the Golgi by either vesicular trafficking or through ceramide transfer protein, CERT [32]. Studies in knockout (KO) mice have shown that while activity of other expressed CerS seems to increase to compensate for total cellular levels of ceramide, severe pathologies based on the particular CerS KO have been reported [33, 34]. The CerS family has yet to be fully characterized, although the formation of CerS homo- and hetero- dimers appears to be a factor in modulating their activity [35]. CerS expression may be altered in several different human cancers such as head and neck, and is often associated with enhanced cancer survival [31].
1.2. Mechanisms of ceramide-induced apoptosis
Ceramide formation (mostly through the activity of sphingomyelinases) can enhance the signaling of proapoptotic membrane molecules such as CD95, part of the extrinsic pathway [36, 37]. Ceramide can also initiate apoptosis through intrinsic pathways involving intracellular mechanisms. Through hydrophobic interactions, ceramide can form transmembrane channels in the mitochondria that may allow the release of apoptosis-inducing proteins [38] and reviewed in [39]. The formation of these channels can be regulated by members of the Bcl-2 family such as Bax and Bcl-xL [39].
Ceramide may initiate apoptotic signaling by down-regulating the activity of the serine/threonine protein kinase Akt, a kinase that can mediate many anti-apoptotic functions [40]. Inactivation or alteration of Akt signaling in cancer is associated with chemoresistance and generally indicates a poor prognosis [41]. Ceramide can activate apoptosis signal-regulating kinase 1 (ASK1 or MAP3K5), which in turn activates p38 and JNK, resulting in apoptosis [42]. Ceramide accumulation can also activate p53 which may lead to the upregulation of the proapoptotic Bax and the subsequent initiation of apoptosis [42] although other studies place ceramide as downstream of p53 [43]. This could be related to differences in studying the function of exogenous ceramides from that of endogenous ceramides, respectively.
Ceramide activated protein phosphatases (CAPP) represent another mechanism that ceramide can exert its effects, the best studied being CAPP1 and CAPP2A [44, 45]. Pretreatment of human squamous carcinoma cells A431 with okadaic acid, an inhibitor of protein phosphatase 2A (including CAPP2A), rendered the cells resistant to C6 ceramide or tumor necrosis factor α (TNFα)-induced apoptosis [46]. Another potential CAPP substrate is Akt, suggesting a link between CAPP activation and cell survival [40]. While CAPP activation is often associated with anti-apoptotic signaling, CAPP2A activation has been shown to dephosphorylate Bcl-2 and render it inactive [47].
The role of ceramide in senescence was first determined when high levels of ceramide were measured in high-passage fibroblasts [48]. Addition of exogenous C6 ceramide was able to recapitulate this finding [48]. Treatment of human pancreatic cancer cells with gemcitabine could produce ceramide at low levels that was associated with cell cycle arrest [49]. Intermediate levels of cellular ceramide through addition of C6 ceramide caused senescence, and at high levels (by adding exogenous sphingomyelin concomittantly with gemcitabine) caused apoptosis [49]. Senescent fibroblasts are more resistant to TNFα-induced apoptosis due at least in part to interrupted ceramide signaling, suggesting that senescence may be another strategy to escape apoptosis [50, 51].
Ceramide has also been associated with telomerase activity and telomere length [52]. Telomeres, long tandem repeats of guanine-rich sequences, protect the ends of chromosomes and are progressively shortened after each cell division [53]. Once the telomere decreases beyond a critical length, the cell undergoes replicative senescence or apoptosis. Many cancer cells upregulate telomerase activity in order to lengthen telomeres [54]. Increased cellular ceramide levels have been shown to both decrease telomerase activity and accelerate telomere shortening [52, 55, 56].
Thus, increased ceramide commonly (although not exclusively) leads to cell death. Levels of ceramide can be regulated through various different mechanisms to produce new sphingolipids that may also have different cellular effects. Reduction in the amount of ceramide present is therefore one mechanism that can mediate cell survival. Strategies that can inhibit ceramide metabolism may provide new avenues to both enhance specificity and enhance sensitivity.
1.3. Removal of ceramide
1.3.1. Ceramidases
There are three major groups of ceramidases which, like the sphingomyelinases, are characterized by their optimal pH, acid, neutral and alkaline [57]. Sphingosine, the product of ceramide hydrolysis by ceramidases, has been shown to induce apoptosis and also act as an opposing regulator of ERK and SAPK signaling pathways [58, 59]. Interestingly, sphingosine has also been reported to form small, short-lived channels in the outer mitochondrial membrane [60]. Both neutral and acid ceramidase (ACDase) activity have been detected within liver nuclear membranes, although their role is at present undetermined [61]. Elevated acid ceramidase expression has been reported in cancers such as prostate, breast, and head and neck and has been linked to increased malignancy and worse clinical outcomes to treatment [62–64]. Gemcitabine-treatment of polyoma middle T transformed mouse endothelial cells downregulated neutral CDase expression leading to cell cycle arrest [65]. Knock down of neutral CDase by siRNA confirmed its role in cell cycle progression [65]. Inhibition of ceramidase may be of therapeutic effect as treatment of colon cancer cells with ceramidase inhibitor B13 completely suppressed tumor growth in nude mice [66]. Additionally, overexpression of ACDase in prostate cancer cells was associated with increased proliferation and migration, while knock down increased sensitivity to several chemotherapeutic drugs including gemcitabine [67]. While sphingosine can cause cell death, its conversion by SK into sphingosine-1 phosphate (S1P) turns it into a potent survival-associated sphingolipid [68].
1.3.2. Glucosylceramide synthase
Glycosylation of ceramide can occur via glucosyl ceramide synthase (GCS). Glucosylceramide has been reported to play a role in cell proliferation, differentiation and metastasis [69, 70]. In addition, glucosylceramide synthesis has been reported in the development of multi-drug resistant cancers and in upregulating the expression of Multidrug Resistance protein 1 (MDR1) [8, 71–74]. Levels of GCS mRNA may also be a marker of disease progression as levels were most elevated in late stage breast cancer [75]. Use of DL-threo-1-phenyl-2-palmitoylamino-3-morpholino-1-propanol (PPMP), a GCS inhibitor, along with C6 ceramide-carrying liposomes has been shown to synergistically induce apoptosis in natural killer cell leukemias [76]. Suppression of GCS may also restore p53-dependent apoptosis of p53 mutant ovarian cancer cells [77]. However, while studies using PPMP have demonstrated chemosensitization effects in cancer cells, the non-specific inhibition of other cellular processes hide the true contribution of GCS to the development of multidrug resistance (MDR) [78]. To address this problem, GCS levels were knocked down using siRNA, which conferred sensitivity to both TNFα and doxorubicin [79]. Conversely, over-expression of GCS conferred resistance to both TNFα and doxorubicin-induced apoptosis through suppression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity and subsequent reactive oxygen species generation [79] Drugs designed to inhibit GCS in the context of Gaucher disease type 1 (lysosomal storage disorder) have been developed and show good specificity and are well tolerated [80]. However, caution in using GCS inhibitors is warranted as it has been reported that PPMP (along with RNAi) can cause cytokinesis failure and multinucleation in human cervical adenocarcinoma cells HeLa, which can lead to chemoresistance [81, 82].
1.3.3. Ceramide kinase
Ceramide kinase can phosphorylate ceramide to produce ceramide-1 phosphate (C1P) [83]. The synthesis of C1P is associated with inflammatory processes and with the development of metabolic syndrome [83]. Exogenous C1P can promote bone marrow-derived macrophage proliferation, while addition of C1P to growth factor deprived macrophages could protect them from apoptosis through inhibition of ASMase [84, 85]. The primary sequence of ceramide kinase has been to shown to possess both nuclear import and export sequences suggesting an as yet unknown nuclear role for C1P [86].
1.4. Sphingosine kinase
Sphingosine kinase has two major isoforms (SK1 and SK2) that are encoded by separate genes and which share 80% similarity of their amino acid sequences [87]. Along with GCS, SK1 and possibly SK2 have been majorly implicated in cellular survival pathways. The sphingosine kinases phosphorylate sphingosine to S1P which can exert its effects either through multiple G protein-coupled receptors (named S1PR1-5) or through direct intracellular effects [88]. The different S1P receptors may play diverse roles, such as S1PR1 being implicated in the trafficking of immune cells and its inhibition (through FTY-720, trade name Fingolimod) is now used to decrease the relapse rate of patients with relapsing-remitting multiple sclerosis [89, 90]. Cancer cells frequently generate S1P that can act though either an autocrine or paracrine manner to promote growth, survival, metastasis, angiogenesis or regulate immune function [91–93]. Exogenous S1P has been shown to upregulate matrix metalloproteinase 2 gene expression and protein, providing a mechanism on how S1P may promote both metastasis and angiogenesis [94, 95] Nuclear S1P has been associated with progression through the S phase of the cell cycle [96]. It has been shown that S1PR1 signaling can act in a positive feedback loop in increasing signal transducer and activator of transcription-3 activity and driving cancer initiation, development and progression [97]. Another important role of S1P is in mediating the Hippo-YAP pathway that can both regulate adult organ size and also modulate proliferation, apoptosis and differentiation [98]. Extracellular S1P has also potent angiogenic and inflammatory effects that can also affect tumor growth [99–101]. Elevated levels of S1PR1 promote the development of localized T lymphoblastic lymphoma into disseminated T lymphoblastic leukemia [102]. Of potential major importance to cancer research are the effects of S1P to suppress ceramide-induced apoptosis [103].
Once S1P is generated, it can be irreversibly cleaved through the action of S1P lyase or converted back to sphingosine by S1P phosphatase [104, 105]. Through the action of S1P Lyase, S1P provides a link between sphingolipids and glycerophospholipids [106]. Interestingly, over expression of S1P lyase in human embryonic kidney, A549 lung cancer, MCF-7 and DLD-1 colon cancer cells has been shown to sensitize cells to apoptotic stress, probably through reduction of intracellular S1P levels [107–109]. Conversely, S1P lyase has been reported to be significantly downregulated in human colon cancer and in prostate cancer when compared to normal tissues [110, 111]. In addition, the chromosomal region containing the gene for S1P lyase is often deleted in human cancers [112–114].
1.4.1. Sphingosine kinase 1
Of particular interest to potential cancer chemotherapy is SK1. Located primarily in the cytoplasm, SK1 can undergo translocation to the plasma membrane in response to activation of protein kinase C (PKC) and/or phospholipase D and after phosphorylation by ERK [115–117]. Through alternative splicing, SK1 had been identified as having three different forms in humans, named SK1a, SK1b, and SK1c, of which the isoform SK1a may be extracelullarly exported [118]. There may be a role for SK in regulating cellular ceramide levels by phosphorylating dihydrosphingosine, thus removing it from the ceramide biosynthetic pathway [119, 120]. By mediating oncogenic H-ras transformation, SK1 has been shown to act in some cases as an oncogene [121]. Furthermore, SK1 has been described in ‘oncogene addiction’ of human epidermal growth factor-2 expressing breast cancer cells [122]. The concept of ‘oncogene addiction’ is evoked when a cancer type requires expression of an oncogene to maintain its disease phenotype. Since SK1 does not usually display any mutations that change its function, it can also be referred to as ‘non-oncogene addiction’ [123]. Proteolytic cleavage of SK1 (and its inactivation) mediated by p53 activation in response to DNA damage appears to be an important step in initiating the apoptotic process [124]. Overexpression of SK1 has been described in many different cancer types including lung, kidney, breast, ovarian, stomach and glioblastoma [125–128]. The overexpression of SK1 has been linked to advanced disease progression, resistance to chemotherapeutic drugs like doxorubicin and also to a poor prognosis [100, 127, 129–132]. Colon cancer is often preceded by chronic inflammation, and levels of SK are upregulated in mouse models of ulcerative colitis [133]. Mice with SK1 KO appear resistant for the development of colorectal cancer [101]. It was later shown that and an increase in SK1 expression in both inflammation and mutagen-treated mice drove the development of colon cancer [134].
1.4.2. Sphingosine kinase 2
While less studied than SK1, overexpression of SK2 in vitro appears to have opposite effects to SK1 expression [135]. The subcellular localization of SK2 may alter its effects on growth. While in the nucleus, SK2 can halt DNA replication through inhibition of histone deacetylase 2 [136, 137]. Interestingly, when COS-7 fibroblasts become confluent, the amount of nuclear SK2 increases [136]. This raises the possibility that SK2 may contribute to epigenetic changes associated with cancer. However, localization of SK2 to the endoplasmic reticulum under stress conditions promotes apoptosis [135]. Underlining the importance of subcellular localization to function, artificial targeting of the normally anti-apoptotic SK1 to the endoplasmic reticulum or nucleus could also promote apoptosis [135, 136]. Interestingly, despite the apparent differences in SK1 and 2 function, mice with either SK1 or SK2 knocked out are both viable and fertile suggesting a level of functional redundancy between the two SK forms [138]. On the other hand, mice lacking both SK1 and SK2 were embryonically lethal, displaying widespread vascular and neural defects [138]. Studies using siRNA against SK2 in A498 kidney adenocarcinoma cells could inhibit their growth more effectively than siRNA against SK1 [139]. Interestingly, ablation of SK2 led to increased intracellular S1P and SK1 expression, while ablation of SK1 did not lead to any corresponding increase in SK2 or S1P [139].
1.4.3. The S1P/ceramide ‘rheostat’ model
Because of the mainly pro-apoptotic effects of ceramide versus the mainly pro-survival effects of S1P, the rheostat model has been conceived [140]. According to the model, if S1P levels were to go down relative to ceramide levels, the cell would undergo apoptosis, and vice versa. While this model may account for gross cellular changes in either S1P or ceramide, the subcellular compartmentalization of either sphingolipid and the diversity of ceramide structure and function are over-looked [12, 141].
Part 2. Mechanisms behind resistance to cancer therapy
Chemotherapeutic drug resistance can be either innate and/or acquired, and result in recurrent cancers often being more resistant to treatment [142]. Innate mechanisms are inherent features of the particular cancer that render it drug resistant, and share many of the same processes that that acquired resistance of relapsed cancer can utilize. The mechanisms that can make cancer resistant to chemotherapy can be mediated through several different pathways.
2.1. Principles of resistance: Innate and acquired resistance
Entry of chemotherapeutic drugs into a cancer cell can be reduced through either a loss of or a mutation of drug transporters [143]. Chemotherapeutic drugs can be removed from the cell, thus lowering the intracellular drug concentration through enhanced efflux by over-expressed ATP-binding cassette proteins (such ABCB1 or MDR1) [144]. Incomplete drug penetration into solid tumors presents another innate mechanism reducing chemotherapeutic drug efficiency. The tumor microenvironment can be either hypoxic or have defective vasculature preventing optimal chemotherapeutic drug delivery [145, 146]. Furthermore, some tumors of the central nervous system or the brain are protected by the blood/brain barrier which can severely limit the penetration of chemotherapeutic drugs [147]. Enhanced or acquired detoxification mechanisms or drug metabolism such as through superoxide dismutase, glutathione S-transferase, or cytochrome P450 can mitigate drug efficacy [148–150]. Mutation of cell cycle checkpoints or deregulation of apoptotic pathways can suppress drug-induced cell death reviewed in [151, 152]. For example, an important study based on an unbiased screen of RNAi data showed that resistance to several chemotherapeutic drugs was mediated by regulators of mitotic arrest and chromosomal instability [153, 154]. These identified CERT and GCS as potentially important regulators of responses to paclitaxel. Conversely, chemotherapy-induced DNA damage can be reduced through activation of DNA repair mechanisms [143]. Therefore, in order to increase the efficacy of chemotherapeutic drug-treatment, one or more of these mechanisms must be overcome.
Part 3. The effect of sphingolipids on cancer treatment
3.1. Radiation Therapy
For many years DNA damage was considered the primary focus behind radiation-induced cell death. However, in recent years it has been reported that cell membranes may also be a target [155]. Later research showed that elements within the cytoplasm, when targeted with microbeam radiation, could also induce radiosensitivity [156]. Agents that could disrupt the formation of lipid rafts (filipin) could inhibit the effect, showing again that membranes can be a target of ionizing radiation. Ionizing radiation has been shown to induce cell death via at least two separate mechanisms, one involving ASMase and one involving CerS. A major problem involved in radiotherapy is the radiation-induced damage of nearby non-cancerous tissue. Furthermore, radiation damage to neighboring tissues can increase the probability of future malignancies forming [157]. While new technologies such as stereotactic radiosurgery can lessen the radiation dosage delivered to normal tissues while increasing the delivered radiation dose, the inherent resistance of some tumors to radiation treatment remains a significant limiting factor [158].
3.1.1. Role of ASMase on radiation-induced apoptosis
The importance of membranes and ceramide in the cellular response to ionizing radiation was first shown in 1994 when ceramide generation was observed in irradiated nuclei-free plasma membranes isolated from endothelial cells [155]. Concomitantly with the ceramide increase, sphingomyelin levels decreased, suggested the activation of a SMase. Remarkably, the radiation-induced ceramide generation occurred within a span of around 2 minutes. Lymphoblasts lacking in ASMase are insensitive to ionizing radiation-induced apoptosis and can be sensitized by genetic reintroduction of ASMase [159]. However, ionizing radiation-induced ASMase activation has been mostly described in endothelial cells. Whether the rapid activation of ASMase after irradiation is due to its relatively high abundance in endothelial cells compared to other cell types is still unknown [160]. The importance of the endothelial response to radiation treatment was underlined when implanted tumors, responsive to radiation in a mouse expressing ASMase, were rendered radioresistant in an ASMase KO mouse, thus suggesting a role for ASMase in host tissue and not tumor [161]. Immunohistochemistry showed that blood vessels infiltrating the tumor were undamaged in the irradiated ASMase KO mouse. Mice with either B16-F1 melanomas or MCA/129 fibrosarcomas had their tumors radiosensitized after intravenous injection of an adenoviral vector containing ASMase under control of an endothelial cell-specific promoter [162]. Interestingly, while the total numbers of transduced tumor endothelial cells was relatively low, in vitro transduction of endothelial cells showed most ASMase activity to be in the conditioned medium, suggesting a role for secretory ASMase in initiating radiation-induced cell death [162]. Therefore, through modification of radiation-induced ASMase activation, it may be possible to both increase damage to tumor vasculature and through inhibition to lessen damage to normal blood vessels.
3.1.2. Role of de novo ceramide generation in radiation-induced apoptosis
The second ceramide-mediated pathway activated by ionizing radiation is via ceramide synthase. Studies on endothelial cells showed that CerS activation occurred 12 hours following radiation [163]. Inhibition of CerS activity with fumonisin B1 inhibited the radiation-induced increase in ceramide and the subsequent apoptosis. Interestingly, irradiation of phorbol ester (potent activator of conventional and novel PKC families)-pretreated prostate cancer cell line LNCaP did not produce any measurable increase in ASMase activation, but did lead to an increase in CerS activity and apoptosis. While the mechanism is unknown, the inhibition of radiation-induced CerS activity by ataxia-telengiectasia mutated protein has been reported [163, 164]. Interestingly, some reports suggest that the ceramide chain length generated in response to ionizing radiation may also play a role in determining radiation sensitivity [165]. While ionizing radiation activated CerS 2, 5 and 6, overexpression of CerS2, responsible for the generation of C24:0 ceramide, partially protected HeLa cells from radiation-induced apoptosis [165]. Conversely, overexpression of Cers5 led to increased C16:0 ceramide and greater radiation-induced apoptosis [165]. Thus, selectively suppressing or enhancing CerS activation in response to ionizing radiation may increase both selectivity and efficiency of the treatment.
3.1.3. Role of ACDase and SK1 in radiation-resistance
Some cancer cells such as prostate or head and neck are able to escape ceramide-induced cell death by over-expressing ceramidases [62, 166]. Recent studies have shown that radiation treatment of prostate cancer cells can upregulate ACDase expression [167]. Surviving prostate cancer clonogens had increased ACDase expression which mediated both their increased radiation resistance and increased proliferation [167]. Inhibition of ACDase activity following irradiation of U87-MG glioma cells or of the PPC-1 prostate cancer cell line could confer sensitivity to radiation-induced apoptosis [168, 169]. The origin of the generated ceramide (either through the activity of ASMase or CerS) is unknown. These results suggest that inhibition of ceramidase activity may confer increased radiation sensitivity, allowing for more efficient tumor control and for the lowering of the effective dose.
Analogues of S1P have been shown to reduce murine radiation-induced lung injury, a serious potential side-effect to thoracic radiation treatment [170]. Pre-treatment of female macaque monkeys with S1P one week before ionizing radiation treatment could maintain fertility after irradiation of the ovaries [171]. Furthermore, xenografts of human ovarian cortical tissue also showed a suppression of radiation-induced oocyte depletion after pretreatment with S1P [171]. Protection of male mouse germ-cells from radiation treatment by S1P has also been reported [172]. Through activation of the Akt pathway, S1P can also protect the small intestine from radiation-induced endothelial apoptosis [173].
3.2. Doxorubicin
Doxorubicin was first isolated from a strain of Streptomyces peucetius treated with the mutagen N-nitroso-N-methyl urethane. It has been used to treat a wide spectrum of cancers including Hodgkin’s lymphoma, breast, bladder and stomach [174]. Doxorubicin inhibits topoisomerase II activity through intercalation of DNA, inhibiting synthesis of DNA and RNA and causing DNA damage [175, 176]. The most serious adverse effect associated with doxorubicin chemotherapy is cardiotoxicity, where the mortality rate for induced congestive heart failure leading from cardiomyopathy can reach 50% [177, 178]. In addition, doxorubicin treatment has also been associated with male infertility [179, 180].
Treatment of follicular thyroid carcinoma cells with doxorubicin increased ceramide via the de novo pathway [181]. Interestingly, it has been shown that in resistant HL-60 cells, doxorubicin can increase GCS expression via activation of the transcription factor sp-1 [182]. Recapitulating this result, doses of doxorubicin (0.1µM – 0.7µM) in MCF-7 cells could also increase GCS protein levels through increased transcription mediated by the transcription factor sp1 [183, 184]. Furthermore, the addition of C6 ceramide to MCF-7 could upregulate transcription of GCS, showing a link between ceramide and GCS expression. Increased GCS resulting from sub-optimal treatment with chemotherapeutic drugs may represent one mechanism through which multi-drug resistant (MDR) cells could be selected for. Conversely, treatment of Burkitt lymphoma cells or mouse ex-vivo T cells with sub-optimal doses of doxorubicin could produce ASMase-generated ceramide which sensitized the cells to subsequent TRAIL-induced apoptosis [185]. This further underlines the importance of ceramide for the initiation of the apoptotic pathway. Doxorubicin treatment (along with paclitaxel, tamoxifen and methotrexate) has been shown to disrupt mitochondrial function and stimulate NADPH oxidase activity which was blocked with increased GCS function [79]. Treatment of neonatal rat cardiomyocytes with doxorubicin caused mitochondrial fragmentation in a manner similar to C2 ceramide, suggesting a role for ceramide in doxorubicin-induced cardiomyopathy [186]. Furthermore, adult rat cardiomyocytes treated with doxorubicin slowly accumulated ceramide, which by 7 days could initiate an apoptotic response [187]. In rats treated with 7.5mg/kg doxorubicin, an increase in C16 ceramide was observed in the testis 2 days following treatment [188]. This suggests that the level of ceramide generated in response to treatment determines, at least in part, whether the cell will undergo apoptosis. Addition of exogenous C6 ceramide can sensitize several different cancer cell lines to doxorubicin by promoting AMP-activated kinase activation and inhibiting mammalian target of rapamycin complex 1 [189]. High levels of SK1 in non small cell lung cancer significantly contributed to doxorubicin resistance [132]. Down-regulation of SK1 expression in gastric cancer cell lines and MCF-7 cells could sensitize them to doxorubicin-treatment [190, 191]. Interestingly, binding of high-density lipoprotein to S1PR2 (but not S1PR1 or S1PR3) could protect cardiomyocytes from doxorubicin-induced cell death [192].
3.3. Paclitaxel
Paclitaxel (often also referred to as Taxol) is a chemotherapeutic used in the treatment for breast, ovarian and lung cancer. Paclitaxel binds to β-tubulin, and as a result stabilizes microtubules and prevents their normal depolymerization/polymerization [193, 194]. Microtubule assembly and disassembly are essential during mitosis, thus paclitaxel treatment causes cell cycle arrest at G2/M and apoptosis [195]. At higher doses, paclitaxel has also been observed to promote the polymerization of stable microtubules [196]. In addition to its effects on microtubule stability, paclitaxel has also been shown to cause mitochondrial depolarization and calcium release [197, 198]. Seriously limiting paclitaxel’s usage is that up to 30% of patients treated with higher doses of paclitaxel experience neuropathy of sensory axons [199, 200].
Jurkat cells treated concomitantly with ceramide and paclitaxel displayed a higher level of apoptosis [201]. Nanoparticles containing paclitaxel and C6 ceramide were more effective at inhibiting the proliferation of aortic smooth muscle cells than with paclitaxel alone [202]. Direct addition of ceramide along with paclitaxel could also increase the apoptotic response of pancreatic cells (L3.6) [203]. Replicative senescence, a loss of cellular ability to proliferate, was induced in human lung cancer cells when C2 ceramide was added together with paclitaxel [204]. Resistance to paclitaxel treatment of ovarian carcinoma cells was associated with a lack of ceramide generation and not to its hydrolysis or glycosylation [205]. One mechanism perhaps underlying this is the finding that in several cancer types CERT is downregulated [153]. On the other hand, glioblastoma cell lines express high levels of GCS that renders these cell lines intrinsically chemoresistant to paclitaxel treatment [206]. Furthermore, paclitaxel-treated MCF-7 cells activated ASMase and the generated ceramide led to cell rounding and a reduction in cell motility, suggesting that ceramide may play an important role in metastasis [207].
3.4. Etoposide
Etoposide was developed in 1966 from analogues of modified podophyllum toxin [208]. It kills cancer cells through inducing single and double strand breaks in DNA by increasing the steady-state concentration of topoisomerase II-containing DNA cleavage complexes [208]. As topoisomerase II is mostly found in actively dividing cells, it has found use as a potent chemotherapeutic drug [209]. Etoposide is commonly used for the treatment of various cancers including testicular, small cell lung, lymphomas and ovarian cancer [2, 208]. One of the major side-effects of etoposide use is the later development of acute myeloid leukemia in some patients, possibly through oxidative DNA damage [210].
Ceramide generated through the de novo pathway, and through increases in serine palmitoyl transferase activity, was reported following etoposide treatment of acute lymphoblastic leukemia cells MOLT-4 [211, 212]. Interestingly, ceramide generation after treatment was biphasic, reaching a peak 4 hours and another at 24–48h post treatment, somewhat reminiscent of radiation-induced ceramide increase [213]. Underlining the importance of ceramide in inducing etoposide (and doxorubicin) cell death, pretreatment of MDR breast cancer cell line MCF-7TN-R cells with a ceramide analogue significantly increased sensitivity to treatment [214]. Additionally, the suppression of dihydroceramide desaturase activity, an enzyme that introduces a 4,5-trans-double bond into the sphinganine backbone, has been shown to confer resistance to etoposide-induced apoptosis in mouse embryonic fibroblast cultures [215]. Pretreatment of mouse T-cell hybridomas with lithium could inhibit etoposide and ceramide-induced protein phosphatase 2A activation and protect the cells from apoptosis [216]. Removal of ceramide by increased expression of GCS has also been linked to etoposide resistance [217]. Additionally, mice lacking S1P lyase are resistant to etoposide treatment, suggesting a role for S1P in mediating resistance [218].
3.5. Cisplatin
First approved by the FDA in 1978, cisplatin is either used as a first line treatment or in combination for bladder cancer, cervical cancer, ovarian cancer, non small cell lung cancer, squamous cell carcinoma of the head and neck and testicular cancer [219]. Cisplatin forms DNA intrastrand crosslinks causing DNA damage, and can also cause mitochondrial damage leading to the generation of reactive oxygen species [220]. While not life threatening, cisplatin can cause severe hearing loss, especially in children [221]. In addition, and more seriously, cisplatin can also cause nephrotoxicity [219].
Ceramide generation in cisplatin-induced apoptosis of glioma cells has been reported [222]. Inhibition of sphingomyelinases suppressed cisplatin-induced apoptosis, while inhibition of ceramidase activity with N-oleoylethanolamine (OE) potentiated the apoptotic effect [222]. Furthermore, treatment of resistant cell cultures with OE increased their sensitivity to cisplatin, suggesting that ceramidase activity is responsible for cisplatin resistance. Knock down of CerS2 led to a reduction of C24 ceramide and an increase in C16 ceramide which increased sensitivity of HeLa cells to cisplatin treatment, suggesting that C16 ceramide induces apoptosis while C24 promotes survival [223]. Other studies showed that cisplatin treatment of baby mouse kidney cells could activate CerS through a Bak-mediated pathway [224]. Interestingly, over-expression of CerS1 could sensitize human embryonic kidney HEK293 cells to cisplatin, while over-expression of SK1 (but not SK2) in those same cells abrogated sensitivity [225]. Plasma membrane redistribution of CD95 in HT-29 colon cancer cells following cisplatin treatment suggest a role for the extrinsic pathway of ceramide-induced apoptosis [226].
3.6. Fluoropyrimidines
The first fluoropyrimidine, 5-fluorouracil (5-FU), was synthesized in 1957, and represents one of the first ‘rationally designed’ chemotherapeutic agents [227]. For many years 5-FU was the only drug approved for use in the US for the treatment of colorectal cancer, although response rates of 10–15% and a median survival of 8–12 months limit its usefulness [227]. Other cancers treated with 5-FU include gastric cancer and breast cancer. Treatment with 5-FU can kill cells via two mechanisms. The first is inhibition of thymidylate synthase, which synthesizes de novo deoxythymidine monophosphate, which after double phosphorylation, is incorporated into DNA [228]. The second method is by competing with uridine triphosphate inhibiting RNA and protein synthesis. Common side effects include hand/foot syndrome, diarrhea and nausea, although severe cardiotoxicity has also been reported [229].
Inhibition of thymidylate synthase with folate analog GW1843 can increase activity of both ASMase and nSMase leading to apoptosis in MOLT-4 cells [230]. Interestingly, pretreatment with a low dose of the rotenoid deguelin, a potentially chemo-prevantative drug against several different cancers, increased ceramide which in turn sensitized the cells to further 5-FU treatment [231]. Another study showed that adding exogenous sphingomyelin could enhance the sensitivity of human colonic xenografts to 5-FU treatment [232]. It is possible that increasing SMase substrate could potentiate the amount of ceramide generated in response to treatment, thereby increasing apoptosis. The role of SMase activity on the extrinsic pathway of apoptotic signaling is suggested by the finding that 5-FU treatment could kill mouse thymocytes via the CD95 pathway [233]. Treatment with carmofur (1-hexylcarbamoyl-5-fluorouracil), a drug that can intracellularly release 5-FU, can significantly inhibit ACDase activity in human colon adenocarcinoma SW403 while increasing ceramide levels [234]. This is turn could sensitize the cells to 5-FU treatment, while pretreatment of ACDase overexpressing cells with carmofur showed no potentiating effect [234].
3.7. Sphingomyelinases vs de novo synthesis
Although several studies have found either activation of ASMase or the de novo pathway in response to specific chemotherapeutic agents, these two events may not be ‘contradictory’. First, these two pathways could be turned on independently of each other in the same cell. This is supported by the different kinetics observed usually with activation of ASMase (usually early) vs the de novo pathway, which is usually later. This was well illustrated in one study with a thymidylate synthase inhibitor, where ASMase activity first peaked 50 mins following treatment then slowly increased to a maximum after 48hrs [230]. On the other hand, nSMase activity first peaked at 20min then peaked again around 18–24hrs after treatment, before dropping [230]. However, total cellular ceramide levels rose 4-fold after 60mins treatment, then started increasing again at 24hrs to reach levels nearly 8-fold higher after 48hrs [230]. Second, our group has demonstrated regulation of the salvage pathway of sphingolipid metabolism whereby hydrolysis of sphingomyelin or cerebrosides in the lysosomes leads to the formation of ceramide which is then acted upon by ACDase. The liberated sphingosine can then be reincorporated into ceramide through the action of CerS. Thus, ASMase and CerS can be part of one coordinated pathway, the salvage pathway. Importantly, the accumulated ceramide in this pathway can be inhibited by Fumonisin B1 which therefore does not distinguish the de novo pathway from the salvage one. In contrast, myriocin (ISP1) would selectively inhibit de novo synthesis. Therefore, the conclusion from some studies that implicate activation of the de novo pathway based on using Fumonisin must be considered as inconclusive as to whether it is the de novo pathway or the salvage pathway.
3.8. Takeaway lessons from sphingolipid studies
It is therefore clear that for many of the different ‘traditional’ chemotherapeutic drugs, ceramide accumulation is important for the initiation of the apoptotic pathway. While chemotherapeutic drugs can raise ceramide levels in a specific way, cancer cells can also upregulate specific ceramide metabolizing enzymes to counteract the treatment (summarized in Figure 1). Therefore, a tailored approach specific for the cancer type (and maybe even to the individual) might provide a way to both sensitize cancer cells and also limit the dosage to mitigate serious side effects in clinical applications. This kind of approach is still in the early stages of testing, and with the advent of better, more selective inhibitors of sphingolipid metabolism, a potentially promising avenue to explore.
Part 4. Inhibitors of SK1 and their application
4.1.1. Early inhibitors
Of all the sphingolipid-modifying enzymes known, probably research on SK is the most developed at this stage. The role of S1P as a potent survival and resistance factor has further driven research into its role in cancer chemoresistance. With the recent publication of the structure of SK1, the design of specific inhibitors should in the future be facilitated [235]. Representative structures of SK1, SK2, and dual SK1 and 2 inhibitors are shown in Figure 2. Early inhibitors of SK were sphingosine analogues that competitively inhibited both SK1 and SK2 such as L-threo-dihydrosphingosine (DHS), D-erythro-N,N-dimethylsphingosine (DMS), and N,N,N-trimethylsphingosine. The first described SK inhibitor, DHS (also known as safingol), has a good affinity for SK1 but can also potently inhibit protein kinase C α [236–238]. A phase 1 clinical trial used safingol, in combination with cisplatin for the treatment of various different solid tumors either refractory for treatment or for which there was no treatment available [239]. Blood levels of S1P decreased in a dose-dependent manner, and disease was stabilized in 6 patients (16%) [239]. Interestingly, the best response was observed in patients with adrenal cortical cancer [239]. DMS has been shown to induce apoptosis in both leukemias and colon carcinoma cells and sensitize TNF-α treated LNCaP cells to ionizing radiation-induced apoptosis [240, 241]. However, these compounds are all non-specific for SK and have other diverse cellular targets [125].
4.1.2. FTY720 (Fingolimod)
FTY720 has emerged as a novel sphingoid analog that suppresses the S1P pathway. It was first described in 1992 after modification of myriocin, a potent inhibitor of serinepalmitoyl transferase [242]. When administered, FTY720 is phosphorylated by SK2. The FTY720-P is a potent functional inhibitor of S1PR1 (and of SIPR3, 4 and 5). It can also induce the proteolysis of SK1 [242]. Interestingly, treatment of ovarian cancer cells with FTY720 could cause both autophagic and necrotic cell death [243]. Treatment of several different prostate cancer cells with Fingolimod caused S1PR-independent apoptosis and increased radiosensitivity [244]. In addition, Fingolimod treatment of mice carrying orthotopic prostate cancer xenografts potentiated the radiation response [244]. (R)-FTY720 methyl ether has been developed to specifically target SK2, and in vitro was able to inhibit DNA synthesis of MCF-7 cells [245]. A role for FTY720 in cancer prevention was demonstrated when daily treatment reduced colitis in both SK2 KO and wild type mice [134].
It has been recently shown that FTY720 can also disrupt lysosomes through proteolysis of ASMase protein in a manner similar to that observed after treatment with the tricyclic antidepressant desipramine. Since desipramine-treatment can induce apoptosis in various cell lines possibly through endoplasmic stress, induction of apoptosis via FTY720-treatment may use a similar, non-S1PR/SK-specific pathway [246, 247]
4.1.3. SKI-II
More selective compounds with IC50 in the submicromolar range were described in 2003, the best was the non-isoform selective 2-(p-hydroxyanilino)-4-(p-chlorophenyl)-thiazole, designated SKi or SKI-II [125]. While designed to be a specific SK1 inhibitor, it has been subsequently shown to also inhibit SK2 activity through competitive inhibition [125]. The initial evaluation of the drug showed that it could inhibit cell proliferation in a number of different cell lines (including MCF-7), and could induce apoptosis in the T24 human bladder cancer cells [125]. Later studies showed that SKI-II could induce apoptosis of androgen-responsive prostate cancer cells [248]. In addition, SKI-II was orally bioavailable and could inhibit murine mammary adenocarcinoma tumor growth and suppress ulcerative colitis in dextran-sulfate sodium (DSS)-treated mice [249, 250]. Inhibition of the Ras/ERK pathway in HepG2 human hepatoma cells using U0126 could sensitize them to SKI-II induced cell death [251]. In combination with doxorubicin or etoposide, SKI-II can increase the percentage of dead MCF-7TN-R cells [252]. SKI-II treatment of MDA-MB-231, HCT-116 colon cancer and NCI-H358 lung cancer cells increased sensitivity to doxorubicin treatment and formation of reactive oxygen species [253]. Head and neck squamous cell carcinoma cell lines can also be radiosensitized with SKI-II [254].
4.1.4. Amidine-based inhibitors
Selective, amidine-based SK inhibitors 100 times more potent towards SK1 than SK2 have been used in U937 (human monocytic leukemia) and Jurkat (human T-cell leukemia) cells, of which Compound 1a demonstrated the highest potency [255]. When using Compound 1a, cellular S1P levels decreased within 10mins, indicating that S1P generated by SK1 has a very rapid turnover [255]. Interestingly, Compound 1a could induce apoptosis in Jurkat cells after 16hrs of treatment and at a dose of 10µM, far higher than the dose required to maximally inhibit SK1 activity [255].
4.1.5. PF-543
A more potent newly described competitive inhibitor PF-543 is 100 times more potent towards SK1 than SK2, and did not inhibit the activity of protein kinases from a panel of almost 50 tested [256]. It has an IC50 of approximately 2nM and was not significantly inactivated in blood due to metabolism or the presence of binding proteins, so can theoretically be injectable without losing potency. Another important characteristic is that sphingosine levels increased during treatment with PF-543, underlining the fact that virtually all SK activity was abolished [256].
4.2. Inhibitors of SK2
4.2.1. ABC294640
ABC294640 is a selective inhibitor of SK2 activity with good oral bioavailability [257]. Treatment of melanoma cells along with several other cancer cell lines including prostate, colon, and breast showed decreased proliferation. In several different cell lines the compound could induce autophagy [258]. Furthermore, oral treatment of mice carrying subcutaneous JC mouse mammary adenocarcinoma tumors demonstrated a significant reduction in palpable tumor growth [257]. Further studies showed that ABC294640 could inhibit NF-κB-mediated chemoresistance of MCF-7TN-R cells implanted in mice [259]. The potential utility of this drug as an adjuvant to established chemotherapy was demonstrated using in vivo mouse models [260, 261]. Adding ABC294640 to sorafenib treatment, a multi-kinase inhibitor, showed a modest but significant suppression of hepatocellular carcinoma, pancreatic adenocarcinoma and kidney carcinoma [260, 261]. In addition, inhibition of SK2 activity in Caov-3 ovarian cancer cells could potentiate the apoptotic effects of paclitaxel treatment [262].
4.2.2. K145
A Thiazolidine-2,3-dione analogue, K145 has an IC50 of 4.3µM, and at concentrations of 10µM did not demonstrate any inhibitory effect on SK1 [263]. Treatment of HEK293 cells with K145 resulted in significantly reduced intracellular S1P levels and could induce apoptotic cell death in both U937 and JC cells in vitro [263]. Furthermore, oral treatment of mice carrying U937 tumors could significantly suppress growth [263].
4.3. Selective inhibitors of both SK1 and SK2
Potent inhibitors of both SK1 and SK2 have also been recently described [264]. Named Compound A and Compound B, their use in several different cell lines led to a significant reduction in intracellular S1P levels to undetectable levels [264]. While Compound A was orally bioavailable, it could not significantly suppress growth of MDA-MB-231 xenografts when used alone [264].
4.4. Differences of effects of SK1 inhibitors versus SK1 knockdown
Treatment of MCF-7 cells with siRNA against SK1 and treatment of human bladder cancer cells with SK1-II can induce apoptosis [125, 265]. Other experiments have shown that knockdown of SK1 or SK2 expression suppresses cell cycle progression but do not induce apoptosis [139]. However, experiments using the newer generation of potent and specific SK1 inhibitors have failed to induce apoptosis in a number of different cancer cell lines [255, 256, 264]. In contrast, both ABC294640 and K145, specific SK2 inhibitors, have been shown to suppress proliferation and cause cell death in some cell lines [257, 263]. It is easy to speculate that the apoptotic effects of the older SK1 inhibitors are simply due to off-target effects, and are thus non-specific. However, the fact that knock down of SK1 expression has been reported to induce either cell cycle arrest or apoptosis raises a couple of interesting possibilities. Either the S1P/ceramide rheostat theory is wrong (or overly simplistic), or that SK1 protein has other effects in addition to its kinase activity. Methods inhibiting SK1 activity have been to date mostly focused on analyzing their effects on cancer cells as monotherapy. Cancer models where SK1 is involved in oncogene/non-oncogene addiction may turn out to be suitable targets for therapeutic drugs directed against SK1. More research needs to be done using SK1 inhibitors in combination with chemotherapeutics to fully assess their potential efficacy in the treatment of cancer.
4.5. Hurdles to overcome before clinical applications targeting sphingolipid metabolism
Sphingolipid metabolism holds may hold promise over established methods of chemotherapy as a more mechanistically-driven approach to cancer therapeutics; however at this point there exist a few hurdles to accomplish such goals. One problem to be overcome is the redundancy of function of different sphingolipid metabolizing enzymes, such as CerS [32]. Another problem to face is that simply raising total cellular ceramide levels may not be sufficient to induce apoptosis without some corresponding driving apoptogenic stimulus [266]. A third problem is that until recently many of the compounds used to inhibit sphingolipid metabolism exhibit non-specific effects or were not effective at clinically tolerable doses [125, 267, 268]. While there is accumulating evidence that targeting sphingolipid metabolism may be of use in cancer therapy, a better understanding of the role of sphingolipid metabolism in cancer cell survival and other key cancer cell functions is required [162]. This, coupled with better, more specific inhibitors will be required before they can be reliably used in a clinical setting.
Summary
While there are other pathways initiated to induce radiation or chemoresistance, sphingolipids play a major role. Commonly, C16 ceramide induction will lead to apoptosis, while strategies to remove ceramide lead to enhanced chemoresistance. Therefore, maintaining C16 abundance may increase the sensitivity of cells to chemotherapeutic drugs. The disappointment of initial results using SK1 inhibitors alone is tempered by the encouraging results obtained inhibiting either SK1 or SK2 alone or together in combination with chemotherapeutic drug treatment.
Highlights.
We review how common chemotherapeutic agents can affect sphingolipid metabolism.
We also review resistance mechanisms cancer cells employ to escape treatment.
Sphingosine kinase inhibitors are examined as potential modifiers to chemotherapy.
Targeting sphingolipid metabolism may improve treatment outcomes.
Abbreviations
- ACDase
Acid ceramidase
- ASMase
Acid sphingomyelinase
- C1P
Ceramide 1-phosphate
- CAPP
Ceramide activated protein phosphatase
- CerS
Ceramide synthase
- CERT
Ceramide transfer protein
- DHS
L-threo-dihydrosphingosine
- DMS
D-erythro-N,N-dimethylsphingosine
- GCS
Glucosyl ceramide synthase
- KO
Knockout
- MDR
Multi drug resistance
- NADPH
Nicotinamide adenine dinucleotide phosphate
- nSMase
Neutral sphingomyelinase
- OE
N-oleoylethanolamine
- PKC
Protein kinase C
- PPMP
DL-threo-1-phenyl-2-palmitoylamino-3-morpholino-1-propanol
- RNAi
Interfering RNA
- S1P
Sphingosine 1-phosphate
- S1PR
Sphingosine 1-phosphate receptor
- siRNA
Small interfering RNA
- SMase
Sphingomyelinase
- SK1/2
Sphingosine kinase 1/2
- SKI-II
2-(p-hydroxyanilino)-4-(p-chlorophenyl)-thiazole
- TNFα
Tumor necrosis factor α
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Contributor Information
Jean-Philip Truman, Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794, Stony Brook, NY, USA, Jean-Philip.Truman@stonybrookmedicine.edu.
Mónica García-Barros, Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794, Stony Brook, NY, USA, Monica.Garcia-Barros@stonybrookmedicine.edu.
Lina M. Obeid, Northport Veterans Affairs Medical Center, Northport, NY 11768, and Health Science Center, Stony Brook University, 100 Nicolls Road, L4, 178, 11794, Stony Brook, NY, USA, Lina.Obeid@stonybrookmedicine.edu
Bibliography
- 1.Papac RJ. Origins of cancer therapy. The Yale journal of biology and medicine. 2001;74:391–398. [PMC free article] [PubMed] [Google Scholar]
- 2.Slevin ML. The clinical pharmacology of etoposide. Cancer. 1991;67:319–329. doi: 10.1002/1097-0142(19910101)67:1+<319::aid-cncr2820671319>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
- 3.Goodman LS, Wintrobe MM, et al. Nitrogen mustard therapy; use of methyl-bis (beta-chloroethyl) amine hydrochloride and tris (beta-chloroethyl) amine hydrochloride for Hodgkin's disease, lymphosarcoma, leukemia and certain allied and miscellaneous disorders. Journal of the American Medical Association. 1946;132:126–132. doi: 10.1001/jama.1946.02870380008004. [DOI] [PubMed] [Google Scholar]
- 4.Gilman A. The initial clinical trial of nitrogen mustard. American journal of surgery. 1963;105:574–578. doi: 10.1016/0002-9610(63)90232-0. [DOI] [PubMed] [Google Scholar]
- 5.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 6.Hall EJ, Giaccia AJ. Radiobiology for the radiologist. 7th ed. Philadelphia: Wolters Kluwer Health/Lippincott Williams & Wilkins; 2012. [Google Scholar]
- 7.Malhotra V, Perry MC. Classical chemotherapy: mechanisms, toxicities and the therapeutic window. Cancer Biol Ther. 2003;2:S2–S4. [PubMed] [Google Scholar]
- 8.Lucci A, Cho WI, Han TY, Giuliano AE, Morton DL, Cabot MC. Glucosylceramide: a marker for multiple-drug resistant cancers. Anticancer Res. 1998;18:475–480. [PubMed] [Google Scholar]
- 9.Gault CR, Obeid LM. Still benched on its way to the bedside: sphingosine kinase 1 as an emerging target in cancer chemotherapy. Critical reviews in biochemistry and molecular biology. 2011;46:342–351. doi: 10.3109/10409238.2011.597737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Linn SC, Kim HS, Keane EM, Andras LM, Wang E, Merrill AH., Jr Regulation of de novo sphingolipid biosynthesis and the toxic consequences of its disruption. Biochem Soc Trans. 2001;29:831–835. doi: 10.1042/0300-5127:0290831. [DOI] [PubMed] [Google Scholar]
- 11.Hannun YA, Obeid LM. The Ceramide-centric universe of lipid-mediated cell regulation: stress encounters of the lipid kind. J Biol Chem. 2002;277:25847–25850. doi: 10.1074/jbc.R200008200. [DOI] [PubMed] [Google Scholar]
- 12.Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol. 2008;9:139–150. doi: 10.1038/nrm2329. [DOI] [PubMed] [Google Scholar]
- 13.Pewzner-Jung Y, Ben-Dor S, Futerman AH. When do Lasses (longevity assurance genes) become CerS (ceramide synthases)?: Insights into the regulation of ceramide synthesis. J Biol Chem. 2006;281:25001–25005. doi: 10.1074/jbc.R600010200. [DOI] [PubMed] [Google Scholar]
- 14.Lozano J, Menendez S, Morales A, Ehleiter D, Liao WC, Wagman R, Haimovitz-Friedman A, Fuks Z, Kolesnick R. Cell autonomous apoptosis defects in acid sphingomyelinase knockout fibroblasts. J Biol Chem. 2001;276:442–448. doi: 10.1074/jbc.M006353200. [DOI] [PubMed] [Google Scholar]
- 15.Jenkins RW, Canals D, Hannun YA. Roles and regulation of secretory and lysosomal acid sphingomyelinase. Cell Signal. 2009;21:836–846. doi: 10.1016/j.cellsig.2009.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hertervig E, Nilsson A, Nyberg L, Duan RD. Alkaline sphingomyelinase activity is decreased in human colorectal carcinoma. Cancer. 1997;79:448–453. [PubMed] [Google Scholar]
- 17.Ellegaard AM, Groth-Pedersen L, Oorschot V, Klumperman J, Kirkegaard T, Nylandsted J, Jaattela M. Sunitinib and SU11652 Inhibit Acid Sphingomyelinase, Destabilize Lysosomes, and Inhibit Multidrug Resistance. Mol Cancer Ther. 2013 doi: 10.1158/1535-7163.MCT-13-0084. [DOI] [PubMed] [Google Scholar]
- 18.Savic R, He X, Fiel I, Schuchman EH. Recombinant human acid sphingomyelinase as an adjuvant to sorafenib treatment of experimental liver cancer. PLoS One. 2013;8:e65620. doi: 10.1371/journal.pone.0065620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Osawa Y, Suetsugu A, Matsushima-Nishiwaki R, Yasuda I, Saibara T, Moriwaki H, Seishima M, Kozawa O. Liver acid sphingomyelinase inhibits growth of metastatic colon cancer. J Clin Invest. 2013;123:834–843. doi: 10.1172/JCI65188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wu BX, Clarke CJ, Hannun YA. Mammalian neutral sphingomyelinases: regulation and roles in cell signaling responses. Neuromolecular medicine. 2010;12:320–330. doi: 10.1007/s12017-010-8120-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bradshaw CD, Ella KM, Thomas AL, Qi C, Meier KE. Effects of Ara-C on neutral sphingomyelinase and mitogen- and stress-activated protein kinases in T-lymphocyte cell lines. Biochemistry and molecular biology international. 1996;40:709–719. doi: 10.1080/15216549600201313. [DOI] [PubMed] [Google Scholar]
- 22.Marchesini N, Luberto C, Hannun YA. Biochemical properties of mammalian neutral sphingomyelinase 2 and its role in sphingolipid metabolism. J Biol Chem. 2003;278:13775–13783. doi: 10.1074/jbc.M212262200. [DOI] [PubMed] [Google Scholar]
- 23.Marchesini N, Osta W, Bielawski J, Luberto C, Obeid LM, Hannun YA. Role for mammalian neutral sphingomyelinase 2 in confluence-induced growth arrest of MCF7 cells. J Biol Chem. 2004;279:25101–25111. doi: 10.1074/jbc.M313662200. [DOI] [PubMed] [Google Scholar]
- 24.Subbaramaiah K, Chung WJ, Dannenberg AJ. Ceramide regulates the transcription of cyclooxygenase-2. Evidence for involvement of extracellular signal-regulated kinase/c-Jun N-terminal kinase and p38 mitogen-activated protein kinase pathways. J Biol Chem. 1998;273:32943–32949. doi: 10.1074/jbc.273.49.32943. [DOI] [PubMed] [Google Scholar]
- 25.Kim WJ, Okimoto RA, Purton LE, Goodwin M, Haserlat SM, Dayyani F, Sweetser DA, McClatchey AI, Bernard OA, Look AT, Bell DW, Scadden DT, Haber DA. Mutations in the neutral sphingomyelinase gene SMPD3 implicate the ceramide pathway in human leukemias. Blood. 2008;111:4716–4722. doi: 10.1182/blood-2007-10-113068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kosaka N, Iguchi H, Hagiwara K, Yoshioka Y, Takeshita F, Ochiya T. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis. J Biol Chem. 2013;288:10849–10859. doi: 10.1074/jbc.M112.446831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lucki NC, Sewer MB. Nuclear sphingolipid metabolism. Annual review of physiology. 2012;74:131–151. doi: 10.1146/annurev-physiol-020911-153321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Watanabe M, Kitano T, Kondo T, Yabu T, Taguchi Y, Tashima M, Umehara H, Domae N, Uchiyama T, Okazaki T. Increase of nuclear ceramide through caspase-3-dependent regulation of the "sphingomyelin cycle" in Fas-induced apoptosis. Cancer Res. 2004;64:1000–1007. doi: 10.1158/0008-5472.can-03-1383. [DOI] [PubMed] [Google Scholar]
- 29.Divecha N, Banfic H, Irvine RF. The polyphosphoinositide cycle exists in the nuclei of Swiss 3T3 cells under the control of a receptor (for IGF-I) in the plasma membrane, and stimulation of the cycle increases nuclear diacylglycerol and apparently induces translocation of protein kinase C to the nucleus. EMBO J. 1991;10:3207–3214. doi: 10.1002/j.1460-2075.1991.tb04883.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kageyama-Yahara N, Riezman H. Transmembrane topology of ceramide synthase in yeast. Biochem J. 2006;398:585–593. doi: 10.1042/BJ20060697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Park JW, Park WJ, Futerman AH. Ceramide synthases as potential targets for therapeutic intervention in human diseases. Biochim Biophys Acta. 2013 doi: 10.1016/j.bbalip.2013.08.019. [DOI] [PubMed] [Google Scholar]
- 32.Tidhar R, Futerman AH. The complexity of sphingolipid biosynthesis in the endoplasmic reticulum. Biochim Biophys Acta. 2013;1833:2511–2518. doi: 10.1016/j.bbamcr.2013.04.010. [DOI] [PubMed] [Google Scholar]
- 33.Pewzner-Jung Y, Park H, Laviad EL, Silva LC, Lahiri S, Stiban J, Erez-Roman R, Brugger B, Sachsenheimer T, Wieland F, Prieto M, Merrill AH, Jr, Futerman AH. A critical role for ceramide synthase 2 in liver homeostasis: I. alterations in lipid metabolic pathways. J Biol Chem. 2010;285:10902–10910. doi: 10.1074/jbc.M109.077594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhao L, Spassieva SD, Jucius TJ, Shultz LD, Shick HE, Macklin WB, Hannun YA, Obeid LM, Ackerman SL. A deficiency of ceramide biosynthesis causes cerebellar purkinje cell neurodegeneration and lipofuscin accumulation. PLoS Genet. 2011;7:e1002063. doi: 10.1371/journal.pgen.1002063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Laviad EL, Kelly S, Merrill AH, Jr, Futerman AH. Modulation of ceramide synthase activity via dimerization. J Biol Chem. 2012;287:21025–21033. doi: 10.1074/jbc.M112.363580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Grassme H, Jekle A, Riehle A, Schwarz H, Berger J, Sandhoff K, Kolesnick R, Gulbins E. CD95 signaling via ceramide-rich membrane rafts. J Biol Chem. 2001;276:20589–20596. doi: 10.1074/jbc.M101207200. [DOI] [PubMed] [Google Scholar]
- 37.Miyaji M, Jin ZX, Yamaoka S, Amakawa R, Fukuhara S, Sato SB, Kobayashi T, Domae N, Mimori T, Bloom ET, Okazaki T, Umehara H. Role of membrane sphingomyelin and ceramide in platform formation for Fas-mediated apoptosis. J Exp Med. 2005;202:249–259. doi: 10.1084/jem.20041685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Siskind LJ, Kolesnick RN, Colombini M. Ceramide channels increase the permeability of the mitochondrial outer membrane to small proteins. J Biol Chem. 2002;277:26796–26803. doi: 10.1074/jbc.M200754200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Colombini M. Membrane channels formed by ceramide. Handbook of experimental pharmacology. 2013:109–126. doi: 10.1007/978-3-7091-1368-4_6. [DOI] [PubMed] [Google Scholar]
- 40.Sato S, Fujita N, Tsuruo T. Modulation of Akt kinase activity by binding to Hsp90. Proc Natl Acad Sci U S A. 2000;97:10832–10837. doi: 10.1073/pnas.170276797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Asakuma J, Sumitomo M, Asano T, Asano T, Hayakawa M. Selective Akt inactivation and tumor necrosis actor-related apoptosis-inducing ligand sensitization of renal cancer cells by low concentrations of paclitaxel. Cancer Res. 2003;63:1365–1370. [PubMed] [Google Scholar]
- 42.Chen CL, Lin CF, Chang WT, Huang WC, Teng CF, Lin YS. Ceramide induces p38 MAPK and JNK activation through a mechanism involving a thioredoxin-interacting protein-mediated pathway. Blood. 2008;111:4365–4374. doi: 10.1182/blood-2007-08-106336. [DOI] [PubMed] [Google Scholar]
- 43.Dbaibo GS, Pushkareva MY, Rachid RA, Alter N, Smyth MJ, Obeid LM, Hannun YA. p53-dependent ceramide response to genotoxic stress. J Clin Invest. 1998;102:329–339. doi: 10.1172/JCI1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dobrowsky RT, Hannun YA. Ceramide stimulates a cytosolic protein phosphatase. J Biol Chem. 1992;267:5048–5051. [PubMed] [Google Scholar]
- 45.Gangoiti P, Granado MH, Alonso A. Implication of ceramide, ceramide 1-phosphate and sphingosine 1-phosphate in tumorigenesis. Translational oncogenomics. 2008;3:81–98. [PMC free article] [PubMed] [Google Scholar]
- 46.Reyes JG, Robayna IG, Delgado PS, Gonzalez IH, Aguiar JQ, Rosas FE, Fanjul LF, Galarreta CM. c-Jun is a downstream target for ceramide-activated protein phosphatase in A431 cells. J Biol Chem. 1996;271:21375–21380. doi: 10.1074/jbc.271.35.21375. [DOI] [PubMed] [Google Scholar]
- 47.Ruvolo PP, Deng X, Ito T, Carr BK, May WS. Ceramide induces Bcl2 dephosphorylation via a mechanism involving mitochondrial PP2A. J Biol Chem. 1999;274:20296–20300. doi: 10.1074/jbc.274.29.20296. [DOI] [PubMed] [Google Scholar]
- 48.Venable ME, Lee JY, Smyth MJ, Bielawska A, Obeid LM. Role of ceramide in cellular senescence. J Biol Chem. 1995;270:30701–30708. doi: 10.1074/jbc.270.51.30701. [DOI] [PubMed] [Google Scholar]
- 49.Modrak DE, Leon E, Goldenberg DM, Gold DV. Ceramide regulates gemcitabine-induced senescence and apoptosis in human pancreatic cancer cell lines. Mol Cancer Res. 2009;7:890–896. doi: 10.1158/1541-7786.MCR-08-0457. [DOI] [PubMed] [Google Scholar]
- 50.Wright WE, Shay JW. Cellular senescence as a tumor-protection mechanism: the essential role of counting. Current opinion in genetics & development. 2001;11:98–103. doi: 10.1016/s0959-437x(00)00163-5. [DOI] [PubMed] [Google Scholar]
- 51.DeJesus V, Rios I, Davis C, Chen Y, Calhoun D, Zakeri Z, Hubbard K. Induction of apoptosis in human replicative senescent fibroblasts. Exp Cell Res. 2002;274:92–99. doi: 10.1006/excr.2001.5425. [DOI] [PubMed] [Google Scholar]
- 52.Ogretmen B, Schady D, Usta J, Wood R, Kraveka JM, Luberto C, Birbes H, Hannun YA, Obeid LM. Role of ceramide in mediating the inhibition of telomerase activity in A549 human lung adenocarcinoma cells. J Biol Chem. 2001;276:24901–24910. doi: 10.1074/jbc.M100314200. [DOI] [PubMed] [Google Scholar]
- 53.Vajen B, Thomay K, Schlegelberger B. Induction of Chromosomal Instability via Telomere Dysfunction and Epigenetic Alterations in Myeloid Neoplasia. Cancers. 2013;5:857–874. doi: 10.3390/cancers5030857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, Coviello GM, Wright WE, Weinrich SL, Shay JW. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266:2011–2015. doi: 10.1126/science.7605428. [DOI] [PubMed] [Google Scholar]
- 55.Kraveka JM, Li L, Bielawski J, Obeid LM, Ogretmen B. Involvement of endogenous ceramide in the inhibition of telomerase activity and induction of morphologic differentiation in response to all-trans-retinoic acid in human neuroblastoma cells. Arch Biochem Biophys. 2003;419:110–119. doi: 10.1016/j.abb.2003.08.034. [DOI] [PubMed] [Google Scholar]
- 56.Sundararaj KP, Wood RE, Ponnusamy S, Salas AM, Szulc Z, Bielawska A, Obeid LM, Hannun YA, Ogretmen B. Rapid shortening of telomere length in response to ceramide involves the inhibition of telomere binding activity of nuclear glyceraldehyde-3-phosphate dehydrogenase. J Biol Chem. 2004;279:6152–6162. doi: 10.1074/jbc.M310549200. [DOI] [PubMed] [Google Scholar]
- 57.Mao C, Obeid LM. Ceramidases: regulators of cellular responses mediated by ceramide, sphingosine, and sphingosine-1-phosphate. Biochim Biophys Acta. 2008;1781:424–434. doi: 10.1016/j.bbalip.2008.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Cuvillier O, Edsall L, Spiegel S. Involvement of sphingosine in mitochondria-dependent Fas-induced apoptosis of type II Jurkat T cells. J Biol Chem. 2000;275:15691–15700. doi: 10.1074/jbc.M000280200. [DOI] [PubMed] [Google Scholar]
- 59.Coroneos E, Wang Y, Panuska JR, Templeton DJ, Kester M. Sphingolipid metabolites differentially regulate extracellular signal-regulated kinase and stress-activated protein kinase cascades. Biochem J. 1996;316(Pt 1):13–17. doi: 10.1042/bj3160013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Siskind LJ, Fluss S, Bui M, Colombini M. Sphingosine forms channels in membranes that differ greatly from those formed by ceramide. Journal of bioenergetics and biomembranes. 2005;37:227–236. doi: 10.1007/s10863-005-6632-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Shiraishi T, Imai S, Uda Y. The presence of ceramidase activity in liver nuclear membrane. Biol Pharm Bull. 2003;26:775–779. doi: 10.1248/bpb.26.775. [DOI] [PubMed] [Google Scholar]
- 62.Seelan RS, Qian C, Yokomizo A, Bostwick DG, Smith DI, Liu W. Human acid ceramidase is overexpressed but not mutated in prostate cancer. Genes Chromosomes Cancer. 2000;29:137–146. doi: 10.1002/1098-2264(2000)9999:9999<::aid-gcc1018>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
- 63.Ruckhaberle E, Holtrich U, Engels K, Hanker L, Gatje R, Metzler D, Karn T, Kaufmann M, Rody A. Acid ceramidase 1 expression correlates with a better prognosis in ER-positive breast cancer. Climacteric : the journal of the International Menopause Society. 2009;12:502–513. doi: 10.3109/13697130902939913. [DOI] [PubMed] [Google Scholar]
- 64.Elojeimy S, Liu X, McKillop JC, El-Zawahry AM, Holman DH, Cheng JY, Meacham WD, Mahdy AE, Saad AF, Turner LS, Cheng J, T AD, Dong JY, Bielawska A, Hannun YA, Norris JS. Role of acid ceramidase in resistance to FasL: therapeutic approaches based on acid ceramidase inhibitors and FasL gene therapy. Mol Ther. 2007;15:1259–1263. doi: 10.1038/sj.mt.6300167. [DOI] [PubMed] [Google Scholar]
- 65.Wu BX, Zeidan YH, Hannun YA. Downregulation of neutral ceramidase by gemcitabine: Implications for cell cycle regulation. Biochim Biophys Acta. 2009;1791:730–739. doi: 10.1016/j.bbalip.2009.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Selzner M, Bielawska A, Morse MA, Rudiger HA, Sindram D, Hannun YA, Clavien PA. Induction of apoptotic cell death and prevention of tumor growth by ceramide analogues in metastatic human colon cancer. Cancer Res. 2001;61:1233–1240. [PubMed] [Google Scholar]
- 67.Saad AF, Meacham WD, Bai A, Anelli V, Elojeimy S, Mahdy AE, Turner LS, Cheng J, Bielawska A, Bielawski J, Keane TE, Obeid LM, Hannun YA, Norris JS, Liu X. The functional effects of acid ceramidase overexpression in prostate cancer progression and resistance to chemotherapy. Cancer Biol Ther. 2007;6:1455–1460. doi: 10.4161/cbt.6.9.4623. [DOI] [PubMed] [Google Scholar]
- 68.Kunkel GT, Maceyka M, Milstien S, Spiegel S. Targeting the sphingosine-1-phosphate axis in cancer, inflammation and beyond. Nat Rev Drug Discov. 2013;12:688–702. doi: 10.1038/nrd4099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hillig I, Leipelt M, Ott C, Zahringer U, Warnecke D, Heinz E. Formation of glucosylceramide and sterol glucoside by a UDP-glucose-dependent glucosylceramide synthase from cotton expressed in Pichia pastoris. FEBS Lett. 2003;553:365–369. doi: 10.1016/s0014-5793(03)01058-5. [DOI] [PubMed] [Google Scholar]
- 70.Ito M, Komori H. Homeostasis of cell-surface glycosphingolipid content in B16 melanoma cells. Evidence revealed by an endoglycoceramidase. J Biol Chem. 1996;271:12655–12660. doi: 10.1074/jbc.271.21.12655. [DOI] [PubMed] [Google Scholar]
- 71.Morjani H, Aouali N, Belhoussine R, Veldman RJ, Levade T, Manfait M. Elevation of glucosylceramide in multidrug-resistant cancer cells and accumulation in cytoplasmic droplets, International journal of cancer. Journal international du cancer. 2001;94:157–165. doi: 10.1002/ijc.1449. [DOI] [PubMed] [Google Scholar]
- 72.Liu YY, Han TY, Giuliano AE, Cabot MC. Ceramide glycosylation potentiates cellular multidrug resistance. FASEB J. 2001;15:719–730. doi: 10.1096/fj.00-0223com. [DOI] [PubMed] [Google Scholar]
- 73.Liu YY, Gupta V, Patwardhan GA, Bhinge K, Zhao Y, Bao J, Mehendale H, Cabot MC, Li YT, Jazwinski SM. Glucosylceramide synthase upregulates MDR1 expression in the regulation of cancer drug resistance through cSrc and beta-catenin signaling. Mol Cancer. 2010;9:145. doi: 10.1186/1476-4598-9-145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Song M, Zang W, Zhang B, Cao J, Yang G. GCS overexpression is associated with multidrug resistance of human HCT-8 colon cancer cells. Journal of experimental & clinical cancer research : CR. 2012;31:23. doi: 10.1186/1756-9966-31-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Liu YY, Patwardhan GA, Xie P, Gu X, Giuliano AE, Cabot MC. Glucosylceramide synthase, a factor in modulating drug resistance, is overexpressed in metastatic breast carcinoma. International journal of oncology. 2011;39:425–431. doi: 10.3892/ijo.2011.1052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Watters RJ, Fox TE, Tan SF, Shanmugavelandy S, Choby JE, Broeg K, Liao J, Kester M, Cabot MC, Loughran TP, Liu X. Targeting glucosylceramide synthase synergizes with C6-ceramide nanoliposomes to induce apoptosis in natural killer cell leukemia. Leuk Lymphoma. 2013;54:1288–1296. doi: 10.3109/10428194.2012.752485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Liu YY, Patwardhan GA, Bhinge K, Gupta V, Gu X, Jazwinski SM. Suppression of glucosylceramide synthase restores p53-dependent apoptosis in mutant p53 cancer cells. Cancer Res. 2011;71:2276–2285. doi: 10.1158/0008-5472.CAN-10-3107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chai L, McLaren RP, Byrne A, Chuang WL, Huang Y, Dufault MR, Pacheco J, Madhiwalla S, Zhang X, Zhang M, Teicher BA, Carter K, Cheng SH, Leonard JP, Xiang Y, Vasconcelles M, Goldberg MA, Copeland DP, Klinger KW, Lillie J, Madden SL, Jiang YA. The chemosensitizing activity of inhibitors of glucosylceramide synthase is mediated primarily through modulation of P-gp function. International journal of oncology. 2011;38:701–711. doi: 10.3892/ijo.2010.888. [DOI] [PubMed] [Google Scholar]
- 79.Barth BM, Gustafson SJ, Young MM, Fox TE, Shanmugavelandy SS, Kaiser JM, Cabot MC, Kester M, Kuhn TB. Inhibition of NADPH oxidase by glucosylceramide confers chemoresistance. Cancer Biol Ther. 2010;10:1126–1136. doi: 10.4161/cbt.10.11.13438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Shayman JA. The design and clinical development of inhibitors of glycosphingolipid synthesis: will invention be the mother of necessity? Transactions of the American Clinical and Climatological Association. 2013;124:46–60. [PMC free article] [PubMed] [Google Scholar]
- 81.Atilla-Gokcumen GE, Bedigian AV, Sasse S, Eggert US. Inhibition of glycosphingolipid biosynthesis induces cytokinesis failure. J Am Chem Soc. 2011;133:10010–10013. doi: 10.1021/ja202804b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zhang S, Mercado-Uribe I, Liu J. Tumor stroma and differentiated cancer cells can be originated directly from polyploid giant cancer cells induced by paclitaxel, International journal of cancer. Journal international du cancer. 2013 doi: 10.1002/ijc.28319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Yuyama K, Mitsutake S, Igarashi Y. Pathological roles of ceramide and its metabolites in metabolic syndrome and Alzheimer's disease. Biochim Biophys Acta. 2013 doi: 10.1016/j.bbalip.2013.08.002. [DOI] [PubMed] [Google Scholar]
- 84.Gangoiti P, Granado MH, Wang SW, Kong JY, Steinbrecher UP, Gomez-Munoz A. Ceramide 1-phosphate stimulates macrophage proliferation through activation of the PI3-kinase/PKB, JNK and ERK1/2 pathways. Cell Signal. 2008;20:726–736. doi: 10.1016/j.cellsig.2007.12.008. [DOI] [PubMed] [Google Scholar]
- 85.Gomez-Munoz A, Kong JY, Salh B, Steinbrecher UP. Ceramide-1-phosphate blocks apoptosis through inhibition of acid sphingomyelinase in macrophages. J Lipid Res. 2004;45:99–105. doi: 10.1194/jlr.M300158-JLR200. [DOI] [PubMed] [Google Scholar]
- 86.Rovina P, Schanzer A, Graf C, Mechtcheriakova D, Jaritz M, Bornancin F. Subcellular localization of ceramide kinase and ceramide kinase-like protein requires interplay of their Pleckstrin Homology domain-containing N-terminal regions together with C-terminal domains. Biochim Biophys Acta. 2009;1791:1023–1030. doi: 10.1016/j.bbalip.2009.05.009. [DOI] [PubMed] [Google Scholar]
- 87.Liu H, Sugiura M, Nava VE, Edsall LC, Kono K, Poulton S, Milstien S, Kohama T, Spiegel S. Molecular cloning and functional characterization of a novel mammalian sphingosine kinase type 2 isoform. J Biol Chem. 2000;275:19513–19520. doi: 10.1074/jbc.M002759200. [DOI] [PubMed] [Google Scholar]
- 88.Maceyka M, Harikumar KB, Milstien S, Spiegel S. Sphingosine-1-phosphate signaling and its role in disease. Trends in cell biology. 2012;22:50–60. doi: 10.1016/j.tcb.2011.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Schwab SR, Cyster JG. Finding a way out: lymphocyte egress from lymphoid organs. Nat Immunol. 2007;8:1295–1301. doi: 10.1038/ni1545. [DOI] [PubMed] [Google Scholar]
- 90.Brinkmann V, Billich A, Baumruker T, Heining P, Schmouder R, Francis G, Aradhye S, Burtin P. Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat Rev Drug Discov. 2010;9:883–897. doi: 10.1038/nrd3248. [DOI] [PubMed] [Google Scholar]
- 91.Salas A, Ponnusamy S, Senkal CE, Meyers-Needham M, Selvam SP, Saddoughi SA, Apohan E, Sentelle RD, Smith C, Gault CR, Obeid LM, El-Shewy HM, Oaks J, Santhanam R, Marcucci G, Baran Y, Mahajan S, Fernandes D, Stuart R, Perrotti D, Ogretmen B. Sphingosine kinase-1 and sphingosine 1-phosphate receptor 2 mediate Bcr-Abl1 stability and drug resistance by modulation of protein phosphatase 2A. Blood. 2011;117:5941–5952. doi: 10.1182/blood-2010-08-300772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Kim ES, Kim JS, Kim SG, Hwang S, Lee CH, Moon A. Sphingosine 1-phosphate regulates matrix metalloproteinase-9 expression and breast cell invasion through S1P3-Galphaq coupling. J Cell Sci. 2011;124:2220–2230. doi: 10.1242/jcs.076794. [DOI] [PubMed] [Google Scholar]
- 93.Anelli V, Gault CR, Snider AJ, Obeid LM. Role of sphingosine kinase-1 in paracrine/transcellular angiogenesis and lymphangiogenesis in vitro. FASEB J. 2010;24:2727–2738. doi: 10.1096/fj.09-150540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Wu WT, Chen CN, Lin CI, Chen JH, Lee H. Lysophospholipids enhance matrix metalloproteinase-2 expression in human endothelial cells. Endocrinology. 2005;146:3387–3400. doi: 10.1210/en.2004-1654. [DOI] [PubMed] [Google Scholar]
- 95.Young N, Pearl DK, Van Brocklyn JR. Sphingosine-1-phosphate regulates glioblastoma cell invasiveness through the urokinase plasminogen activator system and CCN1/Cyr61. Mol Cancer Res. 2009;7:23–32. doi: 10.1158/1541-7786.MCR-08-0061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kleuser B, Maceyka M, Milstien S, Spiegel S. Stimulation of nuclear sphingosine kinase activity by platelet-derived growth factor. FEBS Lett. 2001;503:85–90. doi: 10.1016/s0014-5793(01)02697-7. [DOI] [PubMed] [Google Scholar]
- 97.Lee H, Deng J, Kujawski M, Yang C, Liu Y, Herrmann A, Kortylewski M, Horne D, Somlo G, Forman S, Jove R, Yu H. STAT3-induced S1PR1 expression is crucial for persistent STAT3 activation in tumors. Nat Med. 2010;16:1421–1428. doi: 10.1038/nm.2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Yu FX, Zhao B, Panupinthu N, Jewell JL, Lian I, Wang LH, Zhao J, Yuan H, Tumaneng K, Li H, Fu XD, Mills GB, Guan KL. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell. 2012;150:780–791. doi: 10.1016/j.cell.2012.06.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lee OH, Kim YM, Lee YM, Moon EJ, Lee DJ, Kim JH, Kim KW, Kwon YG. Sphingosine 1-phosphate induces angiogenesis: its angiogenic action and signaling mechanism in human umbilical vein endothelial cells. Biochem Biophys Res Commun. 1999;264:743–750. doi: 10.1006/bbrc.1999.1586. [DOI] [PubMed] [Google Scholar]
- 100.Kawamori T, Osta W, Johnson KR, Pettus BJ, Bielawski J, Tanaka T, Wargovich MJ, Reddy BS, Hannun YA, Obeid LM, Zhou D. Sphingosine kinase 1 is up-regulated in colon carcinogenesis. FASEB J. 2006;20:386–388. doi: 10.1096/fj.05-4331fje. [DOI] [PubMed] [Google Scholar]
- 101.Kawamori T, Kaneshiro T, Okumura M, Maalouf S, Uflacker A, Bielawski J, Hannun YA, Obeid LM. Role for sphingosine kinase 1 in colon carcinogenesis. FASEB J. 2009;23:405–414. doi: 10.1096/fj.08-117572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Feng H, Stachura DL, White RM, Gutierrez A, Zhang L, Sanda T, Jette CA, Testa JR, Neuberg DS, Langenau DM, Kutok JL, Zon LI, Traver D, Fleming MD, Kanki JP, Look AT. T-lymphoblastic lymphoma cells express high levels of BCL2, S1P1, and ICAM1, leading to a blockade of tumor cell intravasation. Cancer cell. 2010;18:353–366. doi: 10.1016/j.ccr.2010.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Coso OA, Gutkind JS, Spiegel S. Suppression Of Ceramide-Mediated Programmed Cell Death By Sphingosine-1-Phosphate. Nature (London) 1996;381:800–803. doi: 10.1038/381800a0. [DOI] [PubMed] [Google Scholar]
- 104.Fyrst H, Saba JD. Sphingosine-1-phosphate lyase in development and disease: sphingolipid metabolism takes flight. Biochim Biophys Acta. 2008;1781:448–458. doi: 10.1016/j.bbalip.2008.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Mandala SM, Thornton R, Galve-Roperh I, Poulton S, Peterson C, Olivera A, Bergstrom J, Kurtz MB, Spiegel S. Molecular cloning and characterization of a lipid phosphohydrolase that degrades sphingosine-1- phosphate and induces cell death. Proc Natl Acad Sci U S A. 2000;97:7859–7864. doi: 10.1073/pnas.120146897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kihara A. Sphingosine 1-phosphate is a key metabolite linking sphingolipids to glycerophospholipids. Biochim Biophys Acta. 2013 doi: 10.1016/j.bbalip.2013.08.014. [DOI] [PubMed] [Google Scholar]
- 107.Reiss U, Oskouian B, Zhou J, Gupta V, Sooriyakumaran P, Kelly S, Wang E, Merrill AH, Jr, Saba JD. Sphingosine-phosphate lyase enhances stress-induced ceramide generation and apoptosis. J Biol Chem. 2004;279:1281–1290. doi: 10.1074/jbc.M309646200. [DOI] [PubMed] [Google Scholar]
- 108.Min J, Van Veldhoven PP, Zhang L, Hanigan MH, Alexander H, Alexander S. Sphingosine-1-phosphate lyase regulates sensitivity of human cells to select chemotherapy drugs in a p38-dependent manner. Mol Cancer Res. 2005;3:287–296. doi: 10.1158/1541-7786.MCR-04-0197. [DOI] [PubMed] [Google Scholar]
- 109.Oskouian B, Sooriyakumaran P, Borowsky AD, Crans A, Dillard-Telm L, Tam YY, Bandhuvula P, Saba JD. Sphingosine-1-phosphate lyase potentiates apoptosis via p53- and p38-dependent pathways and is down-regulated in colon cancer. Proc Natl Acad Sci U S A. 2006;103:17384–17389. doi: 10.1073/pnas.0600050103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Oskouian B, Saba J. Sphingosine-1-phosphate metabolism and intestinal tumorigenesis: lipid signaling strikes again. Cell Cycle. 2007;6:522–527. doi: 10.4161/cc.6.5.3903. [DOI] [PubMed] [Google Scholar]
- 111.Brizuela L, Ader I, Mazerolles C, Bocquet M, Malavaud B, Cuvillier O. First evidence of sphingosine 1-phosphate lyase protein expression and activity downregulation in human neoplasm: implication for resistance to therapeutics in prostate cancer. Mol Cancer Ther. 2012;11:1841–1851. doi: 10.1158/1535-7163.MCT-12-0227. [DOI] [PubMed] [Google Scholar]
- 112.Steck PA, Ligon AH, Cheong P, Yung WK, Pershouse MA. Two tumor suppressive loci on chromosome 10 involved in human glioblastomas. Genes Chromosomes Cancer. 1995;12:255–261. doi: 10.1002/gcc.2870120404. [DOI] [PubMed] [Google Scholar]
- 113.Fawole AS, Simpson DJ, Rajagopal R, Elder J, Holland TA, Fryer A, Deakin M, Elder JB, Farrell WE. Loss of heterozygosity on chromosome 10q is associated with earlier onset sporadic colorectal adenocarcinoma, International journal of cancer. Journal international du cancer. 2002;99:829–833. doi: 10.1002/ijc.10432. [DOI] [PubMed] [Google Scholar]
- 114.Wrage M, Ruosaari S, Eijk PP, Kaifi JT, Hollmen J, Yekebas EF, Izbicki JR, Brakenhoff RH, Streichert T, Riethdorf S, Glatzel M, Ylstra B, Pantel K, Wikman H. Genomic profiles associated with early micrometastasis in lung cancer: relevance of 4q deletion. Clin Cancer Res. 2009;15:1566–1574. doi: 10.1158/1078-0432.CCR-08-2188. [DOI] [PubMed] [Google Scholar]
- 115.Delon C, Manifava M, Wood E, Thompson D, Krugmann S, Pyne S, Ktistakis NT. Sphingosine kinase 1 is an intracellular effector of phosphatidic acid. J Biol Chem. 2004;279:44763–44774. doi: 10.1074/jbc.M405771200. [DOI] [PubMed] [Google Scholar]
- 116.Pitson SM, Moretti PA, Zebol JR, Lynn HE, Xia P, Vadas MA, Wattenberg BW. Activation of sphingosine kinase 1 by ERK1/2-mediated phosphorylation. EMBO J. 2003;22:5491–5500. doi: 10.1093/emboj/cdg540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.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;277:35257–35262. doi: 10.1074/jbc.M203033200. [DOI] [PubMed] [Google Scholar]
- 118.Venkataraman K, Lee YM, Michaud J, Thangada S, Ai Y, Bonkovsky HL, Parikh NS, Habrukowich C, Hla T. Vascular endothelium as a contributor of plasma sphingosine 1-phosphate. Circ Res. 2008;102:669–676. doi: 10.1161/CIRCRESAHA.107.165845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Berdyshev EV, Gorshkova IA, Usatyuk P, Zhao Y, Saatian B, Hubbard W, Natarajan V. De novo biosynthesis of dihydrosphingosine-1-phosphate by sphingosine kinase 1 in mammalian cells. Cell Signal. 2006;18:1779–1792. doi: 10.1016/j.cellsig.2006.01.018. [DOI] [PubMed] [Google Scholar]
- 120.Siow DL, Anderson CD, Berdyshev EV, Skobeleva A, Natarajan V, Pitson SM, Wattenberg BW. Sphingosine kinase localization in the control of sphingolipid metabolism. Advances in enzyme regulation. 2011;51:229–244. doi: 10.1016/j.advenzreg.2010.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Xia P, Gamble JR, Wang L, Pitson SM, Moretti PA, Wattenberg BW, D'Andrea RJ, Vadas MA. An oncogenic role of sphingosine kinase. Curr Biol. 2000;10:1527–1530. doi: 10.1016/s0960-9822(00)00834-4. [DOI] [PubMed] [Google Scholar]
- 122.Long JS, Edwards J, Watson C, Tovey S, Mair KM, Schiff R, Natarajan V, Pyne NJ, Pyne S. Sphingosine kinase 1 induces tolerance to human epidermal growth factor receptor 2 and prevents formation of a migratory phenotype in response to sphingosine 1-phosphate in estrogen receptor-positive breast cancer cells. Mol Cell Biol. 2010;30:3827–3841. doi: 10.1128/MCB.01133-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Vadas M, Xia P, McCaughan G, Gamble J. The role of sphingosine kinase 1 in cancer: oncogene or non-oncogene addiction? Biochim Biophys Acta. 2008;1781:442–447. doi: 10.1016/j.bbalip.2008.06.007. [DOI] [PubMed] [Google Scholar]
- 124.Taha TA, Osta W, Kozhaya L, Bielawski J, Johnson KR, Gillanders WE, Dbaibo GS, Hannun YA, Obeid LM. Down-regulation of sphingosine kinase-1 by DNA damage: dependence on proteases and p53. J Biol Chem. 2004;279:20546–20554. doi: 10.1074/jbc.M401259200. [DOI] [PubMed] [Google Scholar]
- 125.French KJ, Schrecengost RS, Lee BD, Zhuang Y, Smith SN, Eberly JL, Yun JK, Smith CD. Discovery and evaluation of inhibitors of human sphingosine kinase. Cancer Res. 2003;63:5962–5969. [PubMed] [Google Scholar]
- 126.Johnson KR, Johnson KY, Crellin HG, Ogretmen B, Boylan AM, Harley RA, Obeid LM. Immunohistochemical distribution of sphingosine kinase 1 in normal and tumor lung tissue. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2005;53:1159–1166. doi: 10.1369/jhc.4A6606.2005. [DOI] [PubMed] [Google Scholar]
- 127.Van Brocklyn JR, Jackson CA, Pearl DK, Kotur MS, Snyder PJ, Prior TW. Sphingosine kinase-1 expression correlates with poor survival of patients with glioblastoma multiforme: roles of sphingosine kinase isoforms in growth of glioblastoma cell lines. Journal of neuropathology and experimental neurology. 2005;64:695–705. doi: 10.1097/01.jnen.0000175329.59092.2c. [DOI] [PubMed] [Google Scholar]
- 128.Li W, Yu CP, Xia JT, Zhang L, Weng GX, Zheng HQ, Kong QL, Hu LJ, Zeng MS, Zeng YX, Li M, Li J, Song LB. Sphingosine kinase 1 is associated with gastric cancer progression and poor survival of patients. Clin Cancer Res. 2009;15:1393–1399. doi: 10.1158/1078-0432.CCR-08-1158. [DOI] [PubMed] [Google Scholar]
- 129.Bonhoure E, Pchejetski D, Aouali N, Morjani H, Levade T, Kohama T, Cuvillier O. Overcoming MDR-associated chemoresistance in HL-60 acute myeloid leukemia cells by targeting sphingosine kinase-1. Leukemia. 2006;20:95–102. doi: 10.1038/sj.leu.2404023. [DOI] [PubMed] [Google Scholar]
- 130.Sobue S, Iwasaki T, Sugisaki C, Nagata K, Kikuchi R, Murakami M, Takagi A, Kojima T, Banno Y, Akao Y, Nozawa Y, Kannagi R, Suzuki M, Abe A, Naoe T, Murate T. Quantitative RT-PCR analysis of sphingolipid metabolic enzymes in acute leukemia and myelodysplastic syndromes. Leukemia. 2006;20:2042–2046. doi: 10.1038/sj.leu.2404386. [DOI] [PubMed] [Google Scholar]
- 131.Liu G, Zheng H, Zhang Z, Wu Z, Xiong H, Li J, Song L. Overexpression of sphingosine kinase 1 is associated with salivary gland carcinoma progression and might be a novel predictive marker for adjuvant therapy. BMC cancer. 2010;10:495. doi: 10.1186/1471-2407-10-495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Song L, Xiong H, Li J, Liao W, Wang L, Wu J, Li M. 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;17:1839–1849. doi: 10.1158/1078-0432.CCR-10-0720. [DOI] [PubMed] [Google Scholar]
- 133.Snider AJ, Kawamori T, Bradshaw SG, Orr KA, Gilkeson GS, Hannun YA, Obeid LM. A role for sphingosine kinase 1 in dextran sulfate sodium-induced colitis. FASEB J. 2009;23:143–152. doi: 10.1096/fj.08-118109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Liang J, Nagahashi M, Kim EY, Harikumar KB, Yamada A, Huang WC, Hait NC, Allegood JC, Price MM, Avni D, Takabe K, Kordula T, Milstien S, Spiegel S. Sphingosine-1-phosphate links persistent STAT3 activation, chronic intestinal inflammation, and development of colitis-associated cancer. Cancer cell. 2013;23:107–120. doi: 10.1016/j.ccr.2012.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Maceyka M, Sankala H, Hait NC, Le Stunff H, Liu H, Toman R, Collier C, Zhang M, Satin LS, Merrill AH, Jr, Milstien S, Spiegel S. SphK1 and SphK2, sphingosine kinase isoenzymes with opposing functions in sphingolipid metabolism. J Biol Chem. 2005;280:37118–37129. doi: 10.1074/jbc.M502207200. [DOI] [PubMed] [Google Scholar]
- 136.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;278:46832–46839. doi: 10.1074/jbc.M306577200. [DOI] [PubMed] [Google Scholar]
- 137.Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, Luo C, Marmorstein R, Kordula T, Milstien S, Spiegel S. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science. 2009;325:1254–1257. doi: 10.1126/science.1176709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Mizugishi K, Yamashita T, Olivera A, Miller GF, Spiegel S, Proia RL. Essential role for sphingosine kinases in neural and vascular development. Mol Cell Biol. 2005;25:11113–11121. doi: 10.1128/MCB.25.24.11113-11121.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Gao P, Smith CD. Ablation of sphingosine kinase-2 inhibits tumor cell proliferation and migration. Mol Cancer Res. 2011;9:1509–1519. doi: 10.1158/1541-7786.MCR-11-0336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Spiegel S, Cuvillier O, Edsall LC, Kohama T, Menzeleev R, Olah Z, Olivera A, Pirianov G, Thomas DM, Tu Z, Van Brocklyn JR, Wang F. Sphingosine-1-phosphate in cell growth and cell death. Ann N Y Acad Sci. 1998;845:11–18. doi: 10.1111/j.1749-6632.1998.tb09658.x. [DOI] [PubMed] [Google Scholar]
- 141.Hannun YA, Luberto C, Argraves KM. Enzymes of sphingolipid metabolism: from modular to integrative signaling. Biochemistry. 2001;40:4893–4903. doi: 10.1021/bi002836k. [DOI] [PubMed] [Google Scholar]
- 142.Wang Z, Chen W. Emerging Roles of SIRT1 in Cancer Drug Resistance. Genes & cancer. 2013;4:82–90. doi: 10.1177/1947601912473826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58. doi: 10.1038/nrc706. [DOI] [PubMed] [Google Scholar]
- 144.Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat Rev Cancer. 2006;6:583–592. doi: 10.1038/nrc1893. [DOI] [PubMed] [Google Scholar]
- 145.Szakacs G, Gottesman MM. Comparing solid tumors with cell lines: implications for identifying drug resistance genes in cancer. Molecular interventions. 2004;4:323–325. doi: 10.1124/mi.4.6.5. [DOI] [PubMed] [Google Scholar]
- 146.Szakacs G, Annereau JP, Lababidi S, Shankavaram U, Arciello A, Bussey KJ, Reinhold W, Guo Y, Kruh GD, Reimers M, Weinstein JN, Gottesman MM. Predicting drug sensitivity and resistance: profiling ABC transporter genes in cancer cells. Cancer cell. 2004;6:129–137. doi: 10.1016/j.ccr.2004.06.026. [DOI] [PubMed] [Google Scholar]
- 147.Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier. Neurobiol Dis. 2010;37:13–25. doi: 10.1016/j.nbd.2009.07.030. [DOI] [PubMed] [Google Scholar]
- 148.Hayes JD, Pulford DJ. The glutathione S-transferase supergene family: regulation of GST and the contribution of the isoenzymes to cancer chemoprotection and drug resistance. Critical reviews in biochemistry and molecular biology. 1995;30:445–600. doi: 10.3109/10409239509083491. [DOI] [PubMed] [Google Scholar]
- 149.Thomas-Schoemann A, Blanchet B, Bardin C, Noe G, Boudou-Rouquette P, Vidal M, Goldwasser F. Drug interactions with solid tumour-targeted therapies. Critical reviews in oncology/hematology. 2013 doi: 10.1016/j.critrevonc.2013.08.007. [DOI] [PubMed] [Google Scholar]
- 150.Saeidnia S, Abdollahi M. Antioxidants: friends or foe in prevention or treatment of cancer: the debate of the century. Toxicology and applied pharmacology. 2013;271:49–63. doi: 10.1016/j.taap.2013.05.004. [DOI] [PubMed] [Google Scholar]
- 151.Gottesman MM. Mechanisms of cancer drug resistance. Annual review of medicine. 2002;53:615–627. doi: 10.1146/annurev.med.53.082901.103929. [DOI] [PubMed] [Google Scholar]
- 152.Patel NR, Pattni BS, Abouzeid AH, Torchilin VP. Nanopreparations to overcome multidrug resistance in cancer. Adv Drug Deliv Rev. 2013 doi: 10.1016/j.addr.2013.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Swanton C, Marani M, Pardo O, Warne PH, Kelly G, Sahai E, Elustondo F, Chang J, Temple J, Ahmed AA, Brenton JD, Downward J, Nicke B. Regulators of mitotic arrest and ceramide metabolism are determinants of sensitivity to paclitaxel and other chemotherapeutic drugs. Cancer cell. 2007;11:498–512. doi: 10.1016/j.ccr.2007.04.011. [DOI] [PubMed] [Google Scholar]
- 154.Swanton C, Szallasi Z, Brenton JD, Downward J. Functional genomic analysis of drug sensitivity pathways to guide adjuvant strategies in breast cancer. Breast Cancer Res. 2008;10:214. doi: 10.1186/bcr2159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Haimovitz-Friedman A, Kan CC, Ehleiter D, Persaud RS, McLoughlin M, Fuks Z, Kolesnick RN. Ionizing radiation acts on cellular membranes to generate ceramide and initiate apoptosis. J Exp Med. 1994;180:525–535. doi: 10.1084/jem.180.2.525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Shao C, Folkard M, Michael BD, Prise KM. Targeted cytoplasmic irradiation induces bystander responses. Proc Natl Acad Sci U S A. 2004;101:13495–13500. doi: 10.1073/pnas.0404930101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Curtis RE. U.S. Dept. of Health and Human Services. Washington, D.C.: National Institutes of Health, National Cancer Institute; 2006. New malignancies among cancer survivors : SEER cancer registries, 1973–2000. [Google Scholar]
- 158.Linkous AG, Yazlovitskaya EM. Novel radiosensitizing anticancer therapeutics. Anticancer Res. 2012;32:2487–2499. [PubMed] [Google Scholar]
- 159.Santana P, Peña LA, Haimovitz-Friedman A, Martin S, Green D, McLoughlin M, Cordon-Cardo C, Schuchman EH, Fuks Z, Kolesnick R. Acid sphingomyelinase deficient human lymphoblasts and mice are defective in radiation-induced apoptosis. Cell. 1996;86:189–199. doi: 10.1016/s0092-8674(00)80091-4. [DOI] [PubMed] [Google Scholar]
- 160.Tabas I. Secretory sphingomyelinase. Chem Phys Lipids. 1999;102:123–130. doi: 10.1016/s0009-3084(99)00080-8. [DOI] [PubMed] [Google Scholar]
- 161.Garcia-Barros M, Paris F, Cordon-Cardo C, Lyden D, Rafii S, Haimovitz-Friedman A, Fuks Z, Kolesnick R. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science. 2003;300:1155–1159. doi: 10.1126/science.1082504. [DOI] [PubMed] [Google Scholar]
- 162.Stancevic B, Varda-Bloom N, Cheng J, Fuller JD, Rotolo JA, Garcia-Barros M, Feldman R, Rao S, Weichselbaum RR, Harats D, Haimovitz-Friedman A, Fuks Z, Sadelain M, Kolesnick R. Adenoviral transduction of human Acid sphingomyelinase into neo-angiogenic endothelium radiosensitizes tumor cure. PLoS One. 2013;8:e69025. doi: 10.1371/journal.pone.0069025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Liao W-C, Haimovitz-Friedman A, Persaud R, McLoughlin M, Ehleiter D, Zhang N, Gatei M, Lavin M, Kolesnick R, Fuks Z. Ataxia Telangiectasia-mutated gene product inhibits DNA damage-induced apoptosis via ceramide synthase. J Biol Chem. 1999;274:17908–17917. doi: 10.1074/jbc.274.25.17908. [DOI] [PubMed] [Google Scholar]
- 164.Truman JP, Gueven N, Lavin M, Leibel S, Kolesnick R, Fuks Z, Haimovitz-Friedman A. Down-regulation of ATM protein sensitizes human prostate cancer cells to radiation-induced apoptosis. J Biol Chem. 2005;280:23262–23272. doi: 10.1074/jbc.M503701200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Mesicek J, Lee H, Feldman T, Jiang X, Skobeleva A, Berdyshev EV, Haimovitz-Friedman A, Fuks Z, Kolesnick R. Ceramide synthases 2, 5, and 6 confer distinct roles in radiation-induced apoptosis in HeLa cells. Cell Signal. 2010;22:1300–1307. doi: 10.1016/j.cellsig.2010.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Norris JS, Bielawska A, Day T, El-Zawahri A, ElOjeimy S, Hannun Y, Holman D, Hyer M, Landon C, Lowe S, Dong JY, McKillop J, Norris K, Obeid L, Rubinchik S, Tavassoli M, Tomlinson S, Voelkel-Johnson C, Liu X. Combined therapeutic use of AdGFPFasL and small molecule inhibitors of ceramide metabolism in prostate and head and neck cancers: a status report. Cancer Gene Ther. 2006;13:1045–1051. doi: 10.1038/sj.cgt.7700965. [DOI] [PubMed] [Google Scholar]
- 167.Cheng JC, Bai A, Beckham TH, Marrison ST, Yount CL, Young K, Lu P, Bartlett AM, Wu BX, Keane BJ, Armeson KE, Marshall DT, Keane TE, Smith MT, Jones EE, Drake RR, Jr, Bielawska A, Norris JS, Liu X. Radiation-induced acid ceramidase confers prostate cancer resistance and tumor relapse. J Clin Invest. 2013;123:4344–4358. doi: 10.1172/JCI64791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Hara S, Nakashima S, Kiyono T, Sawada M, Yoshimura S, Iwama T, Banno Y, Shinoda J, Sakai N. p53-Independent ceramide formation in human glioma cells during gamma-radiation-induced apoptosis. Cell Death Differ. 2004;11:853–861. doi: 10.1038/sj.cdd.4401428. [DOI] [PubMed] [Google Scholar]
- 169.Mahdy AE, Cheng JC, Li J, Elojeimy S, Meacham WD, Turner LS, Bai A, Gault CR, McPherson AS, Garcia N, Beckham TH, Saad A, Bielawska A, Bielawski J, Hannun YA, Keane TE, Taha MI, Hammouda HM, Norris JS, Liu X. Acid ceramidase upregulation in prostate cancer cells confers resistance to radiation: AC inhibition, a potential radiosensitizer. Mol Ther. 2009;17:430–438. doi: 10.1038/mt.2008.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Mathew B, Jacobson JR, Berdyshev E, Huang Y, Sun X, Zhao Y, Gerhold LM, Siegler J, Evenoski C, Wang T, Zhou T, Zaidi R, Moreno-Vinasco L, Bittman R, Chen CT, LaRiviere PJ, Sammani S, Lussier YA, Dudek SM, Natarajan V, Weichselbaum RR, Garcia JG. Role of sphingolipids in murine radiation-induced lung injury: protection by sphingosine 1-phosphate analogs. FASEB J. 2011;25:3388–3400. doi: 10.1096/fj.11-183970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Zelinski MB, Murphy MK, Lawson MS, Jurisicova A, Pau KY, Toscano NP, Jacob DS, Fanton JK, Casper RF, Dertinger SD, Tilly JL. In vivo delivery of FTY720 prevents radiation-induced ovarian failure and infertility in adult female nonhuman primates. Fertility and sterility. 2011;95:1440–1445. doi: 10.1016/j.fertnstert.2011.01.012. e1441–1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Otala M, Suomalainen L, Pentikainen MO, Kovanen P, Tenhunen M, Erkkila K, Toppari J, Dunkel L. Protection from radiation-induced male germ cell loss by sphingosine-1-phosphate. Biol Reprod. 2004;70:759–767. doi: 10.1095/biolreprod.103.021840. [DOI] [PubMed] [Google Scholar]
- 173.Bonnaud S, Niaudet C, Legoux F, Corre I, Delpon G, Saulquin X, Fuks Z, Gaugler MH, Kolesnick R, Paris F. Sphingosine-1-phosphate activates the AKT pathway to protect small intestines from radiation-induced endothelial apoptosis. Cancer Res. 2010;70:9905–9915. doi: 10.1158/0008-5472.CAN-10-2043. [DOI] [PubMed] [Google Scholar]
- 174.Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacological reviews. 2004;56:185–229. doi: 10.1124/pr.56.2.6. [DOI] [PubMed] [Google Scholar]
- 175.Momparler RL, Karon M, Siegel SE, Avila F. Effect of adriamycin on DNA, RNA, and protein synthesis in cell-free systems and intact cells. Cancer Res. 1976;36:2891–2895. [PubMed] [Google Scholar]
- 176.Fornari FA, Randolph JK, Yalowich JC, Ritke MK, Gewirtz DA. Interference by doxorubicin with DNA unwinding in MCF-7 breast tumor cells. Mol Pharmacol. 1994;45:649–656. [PubMed] [Google Scholar]
- 177.Chatterjee K, Zhang J, Honbo N, Karliner JS. Doxorubicin cardiomyopathy. Cardiology. 2010;115:155–162. doi: 10.1159/000265166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Aissiou M, Perie D, Cheriet F, Dahdah NS, Laverdiere C, Curnier D. Imaging of early modification in cardiomyopathy: the doxorubicin-induced model. The international journal of cardiovascular imaging. 2013 doi: 10.1007/s10554-013-0248-0. [DOI] [PubMed] [Google Scholar]
- 179.Shamberger RC, Sherins RJ, Rosenberg SA. The effects of postoperative adjuvant chemotherapy and radiotherapy on testicular function in men undergoing treatment for soft tissue sarcoma. Cancer. 1981;47:2368–2374. doi: 10.1002/1097-0142(19810515)47:10<2368::aid-cncr2820471008>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
- 180.Meistrich ML, Goldstein LS, Wyrobek AJ. Long-term infertility and dominant lethal mutations in male mice treated with adriamycin. Mutat Res. 1985;152:53–65. doi: 10.1016/0027-5107(85)90046-6. [DOI] [PubMed] [Google Scholar]
- 181.Rath G, Schneider C, Langlois B, Sartelet H, Morjani H, Btaouri HE, Dedieu S, Martiny L. De novo ceramide synthesis is responsible for the anti-tumor properties of camptothecin and doxorubicin in follicular thyroid carcinoma. Int J Biochem Cell Biol. 2009;41:1165–1172. doi: 10.1016/j.biocel.2008.10.021. [DOI] [PubMed] [Google Scholar]
- 182.Uchida Y, Itoh M, Taguchi Y, Yamaoka S, Umehara H, Ichikawa S, Hirabayashi Y, Holleran WM, Okazaki T. Ceramide reduction and transcriptional up-regulation of glucosylceramide synthase through doxorubicin-activated Sp1 in drug-resistant HL-60/ADR cells. Cancer Res. 2004;64:6271–6279. doi: 10.1158/0008-5472.CAN-03-1476. [DOI] [PubMed] [Google Scholar]
- 183.Liu YY, Yu JY, Yin D, Patwardhan GA, Gupta V, Hirabayashi Y, Holleran WM, Giuliano AE, Jazwinski SM, Gouaze-Andersson V, Consoli DP, Cabot MC. A role for ceramide in driving cancer cell resistance to doxorubicin. FASEB J. 2008;22:2541–2551. doi: 10.1096/fj.07-092981. [DOI] [PubMed] [Google Scholar]
- 184.Zhang X, Wu X, Su P, Gao Y, Meng B, Sun Y, Li L, Zhou Z, Zhou G. Doxorubicin influences the expression of glucosylceramide synthase in invasive ductal breast cancer. PLoS One. 2012;7:48492. doi: 10.1371/journal.pone.0048492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Dumitru CA, Carpinteiro A, Trarbach T, Hengge UR, Gulbins E. Doxorubicin enhances TRAIL-induced cell death via ceramide-enriched membrane platforms. Apoptosis. 2007;12:1533–1541. doi: 10.1007/s10495-007-0081-9. [DOI] [PubMed] [Google Scholar]
- 186.Parra V, Eisner V, Chiong M, Criollo A, Moraga F, Garcia A, Hartel S, Jaimovich E, Zorzano A, Hidalgo C, Lavandero S. Changes in mitochondrial dynamics during ceramide-induced cardiomyocyte early apoptosis. Cardiovasc Res. 2008;77:387–397. doi: 10.1093/cvr/cvm029. [DOI] [PubMed] [Google Scholar]
- 187.Delpy E, Hatem SN, Andrieu N, de Vaumas C, Henaff M, Rucker-Martin C, Jaffrezou JP, Laurent G, Levade T, Mercadier JJ. Doxorubicin induces slow ceramide accumulation and late apoptosis in cultured adult rat ventricular myocytes. Cardiovasc Res. 1999;43:398–407. doi: 10.1016/s0008-6363(99)00142-x. [DOI] [PubMed] [Google Scholar]
- 188.Zanetti SR, Maldonado EN, Aveldano MI. Doxorubicin affects testicular lipids with long-chain (C18-C22) and very long-chain (C24-C32) polyunsaturated fatty acids. Cancer Res. 2007;67:6973–6980. doi: 10.1158/0008-5472.CAN-07-0376. [DOI] [PubMed] [Google Scholar]
- 189.Ji C, Yang B, Yang YL, He SH, Miao DS, He L, Bi ZG. Exogenous cell-permeable C6 ceramide sensitizes multiple cancer cell lines to Doxorubicin-induced apoptosis by promoting AMPK activation and mTORC1 inhibition. Oncogene. 2010;29:6557–6568. doi: 10.1038/onc.2010.379. [DOI] [PubMed] [Google Scholar]
- 190.Fuereder T, Hoeflmayer D, Jaeger-Lansky A, Rasin-Streden D, Strommer S, Fisker N, Hansen BJ, Crevenna R, Wacheck V. Sphingosine kinase 1 is a relevant molecular target in gastric cancer. Anti-cancer drugs. 2011;22:245–252. doi: 10.1097/cad.0b013e328340bd95. [DOI] [PubMed] [Google Scholar]
- 191.Sarkar S, Maceyka M, Hait NC, Paugh SW, Sankala H, Milstien S, Spiegel S. Sphingosine kinase 1 is required for migration, proliferation and survival of MCF-7 human breast cancer cells. FEBS Lett. 2005;579:5313–5317. doi: 10.1016/j.febslet.2005.08.055. [DOI] [PubMed] [Google Scholar]
- 192.Frias MA, Lang U, Gerber-Wicht C, James RW. Native and reconstituted HDL protect cardiomyocytes from doxorubicin-induced apoptosis. Cardiovasc Res. 2010;85:118–126. doi: 10.1093/cvr/cvp289. [DOI] [PubMed] [Google Scholar]
- 193.Mitchison T, Kirschner M. Dynamic instability of microtubule growth. Nature. 1984;312:237–242. doi: 10.1038/312237a0. [DOI] [PubMed] [Google Scholar]
- 194.Nogales E, Wolf SG, Khan IA, Luduena RF, Downing KH. Structure of tubulin at 6.5 A and location of the taxol-binding site. Nature. 1995;375:424–427. doi: 10.1038/375424a0. [DOI] [PubMed] [Google Scholar]
- 195.Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer. 2004;4:253–265. doi: 10.1038/nrc1317. [DOI] [PubMed] [Google Scholar]
- 196.Schiff PB, Fant J, Horwitz SB. Promotion of microtubule assembly in vitro by taxol. Nature. 1979;277:665–667. doi: 10.1038/277665a0. [DOI] [PubMed] [Google Scholar]
- 197.Varbiro G, Veres B, Gallyas F, Jr, Sumegi B. Direct effect of Taxol on free radical formation and mitochondrial permeability transition. Free Radic Biol Med. 2001;31:548–558. doi: 10.1016/s0891-5849(01)00616-5. [DOI] [PubMed] [Google Scholar]
- 198.Kidd JF, Pilkington MF, Schell MJ, Fogarty KE, Skepper JN, Taylor CW, Thorn P. Paclitaxel affects cytosolic calcium signals by opening the mitochondrial permeability transition pore. J Biol Chem. 2002;277:6504–6510. doi: 10.1074/jbc.M106802200. [DOI] [PubMed] [Google Scholar]
- 199.Park SB, Lin CS, Krishnan AV, Friedlander ML, Lewis CR, Kiernan MC. Early, progressive, and sustained dysfunction of sensory axons underlies paclitaxel-induced neuropathy. Muscle Nerve. 2011;43:367–374. doi: 10.1002/mus.21874. [DOI] [PubMed] [Google Scholar]
- 200.Carlson K, Ocean AJ. Peripheral neuropathy with microtubule-targeting agents: occurrence and management approach. Clinical breast cancer. 2011;11:73–81. doi: 10.1016/j.clbc.2011.03.006. [DOI] [PubMed] [Google Scholar]
- 201.Myrick D, Blackinton D, Klostergaard J, Kouttab N, Maizel A, Wanebo H, Mehta S. Paclitaxel-induced apoptosis in Jurkat, a leukemic T cell line, is enhanced by ceramide. Leuk Res. 1999;23:569–578. doi: 10.1016/s0145-2126(99)00048-x. [DOI] [PubMed] [Google Scholar]
- 202.Deshpande D, Devalapally H, Amiji M. Enhancement in anti-proliferative effects of paclitaxel in aortic smooth muscle cells upon co-administration with ceramide using biodegradable polymeric nanoparticles. Pharmaceutical research. 2008;25:1936–1947. doi: 10.1007/s11095-008-9614-3. [DOI] [PubMed] [Google Scholar]
- 203.Qiu L, Zhou C, Sun Y, Di W, Scheffler E, Healey S, Wanebo H, Kouttab N, Chu W, Wan Y. Paclitaxel and ceramide synergistically induce cell death with transient activation of EGFR and ERK pathway in pancreatic cancer cells. Oncol Rep. 2006;16:907–913. [PubMed] [Google Scholar]
- 204.Chen JY, Hwang CC, Chen WY, Lee JC, Fu TF, Fang K, Chu YC, Huang YL, Lin JC, Tsai WH, Chang HW, Chen BH, Chiu CC. Additive effects of C(2)-ceramide on paclitaxel-induced premature senescence of human lung cancer cells. Life Sci. 2010;87:350–357. doi: 10.1016/j.lfs.2010.06.017. [DOI] [PubMed] [Google Scholar]
- 205.Prinetti A, Millimaggi D, D'Ascenzo S, Clarkson M, Bettiga A, Chigorno V, Sonnino S, Pavan A, Dolo V. Lack of ceramide generation and altered sphingolipid composition are associated with drug resistance in human ovarian carcinoma cells. Biochem J. 2006;395:311–318. doi: 10.1042/BJ20051184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Giussani P, Bassi R, Anelli V, Brioschi L, De Zen F, Riccitelli E, Caroli M, Campanella R, Gaini SM, Viani P, Riboni L. Glucosylceramide synthase protects glioblastoma cells against autophagic and apoptotic death induced by temozolomide and Paclitaxel. Cancer investigation. 2012;30:27–37. doi: 10.3109/07357907.2011.629379. [DOI] [PubMed] [Google Scholar]
- 207.Zeidan YH, Jenkins RW, Hannun YA. Remodeling of cellular cytoskeleton by the acid sphingomyelinase/ceramide pathway. J Cell Biol. 2008;181:335–350. doi: 10.1083/jcb.200705060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Hande KR. Etoposide: four decades of development of a topoisomerase II inhibitor. European journal of cancer. 1998;34:1514–1521. doi: 10.1016/s0959-8049(98)00228-7. [DOI] [PubMed] [Google Scholar]
- 209.Heck MM, Hittelman WN, Earnshaw WC. Differential expression of DNA topoisomerases I and II during the eukaryotic cell cycle. Proc Natl Acad Sci U S A. 1988;85:1086–1090. doi: 10.1073/pnas.85.4.1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Papiez MA. The influence of curcumin and (−)-epicatechin on the genotoxicity and myelosuppression induced by etoposide in bone marrow cells of male rats. Drug and chemical toxicology. 2013;36:93–101. doi: 10.3109/01480545.2012.726626. [DOI] [PubMed] [Google Scholar]
- 211.Perry DK, Carton J, Shah AK, Meredith F, Uhlinger DJ, Hannun YA. Serine palmitoyltransferase regulates de novo ceramide generation during etoposide-induced apoptosis. J Biol Chem. 2000;275:9078–9084. doi: 10.1074/jbc.275.12.9078. [DOI] [PubMed] [Google Scholar]
- 212.Sawada M, Nakashima S, Banno Y, Yamakawa H, Hayashi K, Takenaka K, Nishimura Y, Sakai N, Nozawa Y. Ordering of ceramide formation, caspase activation, and Bax/Bcl-2 expression during etoposide-induced apoptosis in C6 glioma cells. Cell Death Differ. 2000;7:761–772. doi: 10.1038/sj.cdd.4400711. [DOI] [PubMed] [Google Scholar]
- 213.Dai Q, Liu J, Chen J, Durrant D, McIntyre TM, Lee RM. Mitochondrial ceramide increases in UV-irradiated HeLa cells and is mainly derived from hydrolysis of sphingomyelin. Oncogene. 2004;23:3650–3658. doi: 10.1038/sj.onc.1207430. [DOI] [PubMed] [Google Scholar]
- 214.Antoon JW, Beckman BS. Anti-proliferative effects of the novel ceramide analog (S)-2-(benzylideneamino)-3-hydroxy-N-tetrade-cylpropanamide in chemoresistant cancer. Bioorg Med Chem Lett. 2012;22:2624–2628. doi: 10.1016/j.bmcl.2012.01.087. [DOI] [PubMed] [Google Scholar]
- 215.Siddique MM, Bikman BT, Wang L, Ying L, Reinhardt E, Shui G, Wenk MR, Summers SA. Ablation of dihydroceramide desaturase confers resistance to etoposide-induced apoptosis in vitro. PLoS One. 2012;7:e44042. doi: 10.1371/journal.pone.0044042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Chen CL, Lin CF, Chiang CW, Jan MS, Lin YS. Lithium inhibits ceramideand etoposide-induced protein phosphatase 2A methylation, Bcl-2 dephosphorylation, caspase-2 activation, and apoptosis. Mol Pharmacol. 2006;70:510–517. doi: 10.1124/mol.106.024059. [DOI] [PubMed] [Google Scholar]
- 217.Gouaze V, Yu JY, Bleicher RJ, Han TY, Liu YY, Wang H, Gottesman MM, Bitterman A, Giuliano AE, Cabot MC. Overexpression of glucosylceramide synthase and P-glycoprotein in cancer cells selected for resistance to natural product chemotherapy. Mol Cancer Ther. 2004;3:633–639. [PubMed] [Google Scholar]
- 218.Colie S, Van Veldhoven PP, Kedjouar B, Bedia C, Albinet V, Sorli SC, Garcia V, Djavaheri-Mergny M, Bauvy C, Codogno P, Levade T, Andrieu-Abadie N. Disruption of sphingosine 1-phosphate lyase confers resistance to chemotherapy and promotes oncogenesis through Bcl-2/Bcl-xL upregulation. Cancer Res. 2009;69:9346–9353. doi: 10.1158/0008-5472.CAN-09-2198. [DOI] [PubMed] [Google Scholar]
- 219.dos Santos NA, Carvalho Rodrigues MA, Martins NM, dos Santos AC. Cisplatin-induced nephrotoxicity and targets of nephroprotection: an update. Archives of toxicology. 2012;86:1233–1250. doi: 10.1007/s00204-012-0821-7. [DOI] [PubMed] [Google Scholar]
- 220.Stewart JD, Bolt HM. Cisplatin-induced nephrotoxicity. Archives of toxicology. 2012;86:1155–1156. doi: 10.1007/s00204-012-0887-2. [DOI] [PubMed] [Google Scholar]
- 221.Langer T, Am Zehnhoff-Dinnesen A, Radtke S, Meitert J, Zolk O. Understanding platinum-induced ototoxicity. Trends in pharmacological sciences. 2013;34:458–469. doi: 10.1016/j.tips.2013.05.006. [DOI] [PubMed] [Google Scholar]
- 222.Noda S, Yoshimura S, Sawada M, Naganawa T, Iwama T, Nakashima S, Sakai N. Role of ceramide during cisplatin-induced apoptosis in C6 glioma cells. Journal of neuro-oncology. 2001;52:11–21. doi: 10.1023/a:1010624823158. [DOI] [PubMed] [Google Scholar]
- 223.Sassa T, Suto S, Okayasu Y, Kihara A. A shift in sphingolipid composition from C24 to C16 increases susceptibility to apoptosis in HeLa cells. Biochim Biophys Acta. 2012;1821:1031–1037. doi: 10.1016/j.bbalip.2012.04.008. [DOI] [PubMed] [Google Scholar]
- 224.Siskind LJ, Mullen TD, Romero Rosales K, Clarke CJ, Hernandez-Corbacho MJ, Edinger AL, Obeid LM. The BCL-2 protein BAK is required for long-chain ceramide generation during apoptosis. J Biol Chem. 2010;285:11818–11826. doi: 10.1074/jbc.M109.078121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Min J, Mesika A, Sivaguru M, Van Veldhoven PP, Alexander H, Futerman AH, Alexander S. (Dihydro)ceramide synthase 1 regulated sensitivity to cisplatin is associated with the activation of p38 mitogen-activated protein kinase and is abrogated by sphingosine kinase 1. Mol Cancer Res. 2007;5:801–812. doi: 10.1158/1541-7786.MCR-07-0100. [DOI] [PubMed] [Google Scholar]
- 226.Lacour S, Hammann A, Grazide S, Lagadic-Gossmann D, Athias A, Sergent O, Laurent G, Gambert P, Solary E, Dimanche-Boitrel MT. Cisplatin-induced CD95 redistribution into membrane lipid rafts of HT29 human colon cancer cells. Cancer Res. 2004;64:3593–3598. doi: 10.1158/0008-5472.CAN-03-2787. [DOI] [PubMed] [Google Scholar]
- 227.Chu E. Clinical Colorectal Cancer: “Ode to 5-Fluorouracil”. Clinical colorectal cancer. 2007;6:609. [Google Scholar]
- 228.Mader RM, Muller M, Steger GG. Resistance to 5-fluorouracil. General pharmacology. 1998;31:661–666. doi: 10.1016/s0306-3623(98)00191-8. [DOI] [PubMed] [Google Scholar]
- 229.Molteni LP, Rampinelli I, Cergnul M, Scaglietti U, Paino AM, Noonan DM, Bucci EO, Gottardi O, Albini A. Capecitabine in breast cancer: the issue of cardiotoxicity during fluoropyrimidine treatment. The breast journal. 2010;16(Suppl 1):S45–48. doi: 10.1111/j.1524-4741.2010.01004.x. [DOI] [PubMed] [Google Scholar]
- 230.Laethem RM, Hannun YA, Jayadev S, Sexton CJ, Strum JC, Sundseth R, Smith GK. Increases in neutral, Mg2 + -dependent and acidic, Mg2 + -independent sphingomyelinase activities precede commitment to apoptosis and are not a consequence of caspase 3-like activity in Molt-4 cells in response to thymidylate synthase inhibition by GW1843. Blood. 1998;91:4350–4360. [PubMed] [Google Scholar]
- 231.Yang YL, Ji C, Bi ZG, Lu CC, Wang R, Gu B, Cheng L. Deguelin induces both apoptosis and autophagy in cultured head and neck squamous cell carcinoma cells. PLoS One. 2013;8:e54736. doi: 10.1371/journal.pone.0054736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Modrak DE, Rodriguez MD, Goldenberg DM, Lew W, Blumenthal RD. Sphingomyelin enhances chemotherapy efficacy and increases apoptosis in human colonic tumor xenografts. International journal of oncology. 2002;20:379–384. [PubMed] [Google Scholar]
- 233.Eichhorst ST, Muerkoster S, Weigand MA, Krammer PH. The chemotherapeutic drug 5-fluorouracil induces apoptosis in mouse thymocytes in vivo via activation of the CD95(APO-1/Fas) system. Cancer Res. 2001;61:243–248. [PubMed] [Google Scholar]
- 234.Realini N, Solorzano C, Pagliuca C, Pizzirani D, Armirotti A, Luciani R, Costi MP, Bandiera T, Piomelli D. Discovery of highly potent acid ceramidase inhibitors with in vitro tumor chemosensitizing activity. Scientific reports. 2013;3:1035. doi: 10.1038/srep01035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Wang Z, Min X, Xiao SH, Johnstone S, Romanow W, Meininger D, Xu H, Liu J, Dai J, An S, Thibault S, Walker N. Molecular basis of sphingosine kinase 1 substrate recognition and catalysis. Structure. 2013;21:798–809. doi: 10.1016/j.str.2013.02.025. [DOI] [PubMed] [Google Scholar]
- 236.Buehrer BM, Bell RM. Inhibition of sphingosine kinase in vitro and in platelets. Implications for signal transduction pathways. J Biol Chem. 1992;267:3154–3159. [PubMed] [Google Scholar]
- 237.Hannun YA, Loomis CR, Merrill AH, Jr, Bell RM. Sphingosine inhibition of protein kinase C activity and of phorbol dibutyrate binding in vitro and in human platelets. J Biol Chem. 1986;261:12604–12609. [PubMed] [Google Scholar]
- 238.Sachs CW, Safa AR, Harrison SD, Fine RL. Partial inhibition of multidrug resistance by safingol is independent of modulation of P-glycoprotein substrate activities and correlated with inhibition of protein kinase C. J Biol Chem. 1995;270:26639–26648. doi: 10.1074/jbc.270.44.26639. [DOI] [PubMed] [Google Scholar]
- 239.Dickson MA, Carvajal RD, Merrill AH, Jr, Gonen M, Cane LM, Schwartz GK. A phase I clinical trial of safingol in combination with cisplatin in advanced solid tumors. Clin Cancer Res. 2011;17:2484–2492. doi: 10.1158/1078-0432.CCR-10-2323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Sweeney EA, Sakakura C, Shirahama T, Masamune A, Ohta H, Hakomori S, Igarashi Y. Sphingosine and its methylated derivative N,N-dimethylsphingosine (DMS) induce apoptosis in a variety of human cancer cell lines, International journal of cancer. Journal international du cancer. 1996;66:358–366. doi: 10.1002/(SICI)1097-0215(19960503)66:3<358::AID-IJC16>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
- 241.Nava VE, Cuvillier O, Edsall LC, Kimura K, Milstien S, Gelmann EP, Spiegel S. Sphingosine enhances apoptosis of radiation-resistant prostate cancer cells. Cancer Res. 2000;60:4468–4474. [PubMed] [Google Scholar]
- 242.Adachi K, Chiba K. FTY720 story. Its discovery and the following accelerated development of sphingosine 1-phosphate receptor agonists as immunomodulators based on reverse pharmacology. Perspectives in medicinal chemistry. 2008;1:11–23. [PMC free article] [PubMed] [Google Scholar]
- 243.Zhang N, Qi Y, Wadham C, Wang L, Warren A, Di W, Xia P. FTY720 induces necrotic cell death and autophagy in ovarian cancer cells: a protective role of autophagy. Autophagy. 2010;6:1157–1167. doi: 10.4161/auto.6.8.13614. [DOI] [PubMed] [Google Scholar]
- 244.Pchejetski D, Bohler T, Brizuela L, Sauer L, Doumerc N, Golzio M, Salunkhe V, Teissie J, Malavaud B, Waxman J, Cuvillier O. FTY720 (fingolimod) sensitizes prostate cancer cells to radiotherapy by inhibition of sphingosine kinase-1. Cancer Res. 2010;70:8651–8661. doi: 10.1158/0008-5472.CAN-10-1388. [DOI] [PubMed] [Google Scholar]
- 245.Lim KG, Sun C, Bittman R, Pyne NJ, Pyne S. (R)-FTY720 methyl ether is a specific sphingosine kinase 2 inhibitor: Effect on sphingosine kinase 2 expression in HEK 293 cells and actin rearrangement and survival of MCF-7 breast cancer cells. Cell Signal. 2011;23:1590–1595. doi: 10.1016/j.cellsig.2011.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Dawson G, Qin J. Gilenya (FTY720) inhibits acid sphingomyelinase by a mechanism similar to tricyclic antidepressants. Biochem Biophys Res Commun. 2011;404:321–323. doi: 10.1016/j.bbrc.2010.11.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Ma J, Qiu Y, Yang L, Peng L, Xia Z, Hou LN, Fang C, Qi H, Chen HZ. Desipramine induces apoptosis in rat glioma cells via endoplasmic reticulum stress-dependent CHOP pathway. Journal of neuro-oncology. 2011;101:41–48. doi: 10.1007/s11060-010-0237-2. [DOI] [PubMed] [Google Scholar]
- 248.Tonelli F, Alossaimi M, Williamson L, Tate RJ, Watson DG, Chan E, Bittman R, Pyne NJ, Pyne S. The sphingosine kinase inhibitor 2-(p-hyroxyanilino)-4-(p-chlorophenyl)thiazole reduces androgen receptor expression via an oxidative stress-dependent mechanism. British journal of pharmacology. 2013;168:1497–1505. doi: 10.1111/bph.12035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.French KJ, Upson JJ, Keller SN, Zhuang Y, Yun JK, Smith CD. Antitumor activity of sphingosine kinase inhibitors. J Pharmacol Exp Ther. 2006;318:596–603. doi: 10.1124/jpet.106.101345. [DOI] [PubMed] [Google Scholar]
- 250.Maines LW, Fitzpatrick LR, French KJ, Zhuang Y, Xia Z, Keller SN, Upson JJ, Smith CD. Suppression of ulcerative colitis in mice by orally available inhibitors of sphingosine kinase. Dig Dis Sci. 2008;53:997–1012. doi: 10.1007/s10620-007-0133-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Zhang C, He H, Zhang H, Yu D, Zhao W, Chen Y, Shao R. The blockage of Ras/ERK pathway augments the sensitivity of SphK1 inhibitor SKI II in human hepatoma HepG2 cells. Biochem Biophys Res Commun. 2013;434:35–41. doi: 10.1016/j.bbrc.2013.03.070. [DOI] [PubMed] [Google Scholar]
- 252.Antoon JW, White MD, Burow ME, Beckman BS. Dual inhibition of sphingosine kinase isoforms ablates TNF-induced drug resistance. Oncol Rep. 2012;27:1779–1786. doi: 10.3892/or.2012.1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Huwiler A, Kotelevets N, Xin C, Pastukhov O, Pfeilschifter J, Zangemeister-Wittke U. Loss of sphingosine kinase-1 in carcinoma cells increases formation of reactive oxygen species and sensitivity to doxorubicin-induced DNA damage. British journal of pharmacology. 2011;162:532–543. doi: 10.1111/j.1476-5381.2010.01053.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Sinha UK, Schorn VJ, Hochstim C, Chinn SB, Zhu S, Masood R. Increased radiation sensitivity of head and neck squamous cell carcinoma with sphingosine kinase 1 inhibition. Head & neck. 2011;33:178–188. doi: 10.1002/hed.21418. [DOI] [PubMed] [Google Scholar]
- 255.Kharel Y, Mathews TP, Gellett AM, Tomsig JL, Kennedy PC, Moyer ML, Macdonald TL, Lynch KR. Sphingosine kinase type 1 inhibition reveals rapid turnover of circulating sphingosine 1-phosphate. Biochem J. 2011;440:345–353. doi: 10.1042/BJ20110817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Schnute ME, McReynolds MD, Kasten T, Yates M, Jerome G, Rains JW, Hall T, Chrencik J, Kraus M, Cronin CN, Saabye M, Highkin MK, Broadus R, Ogawa S, Cukyne K, Zawadzke LE, Peterkin V, Iyanar K, Scholten JA, Wendling J, Fujiwara H, Nemirovskiy O, Wittwer AJ, Nagiec MM. Modulation of cellular S1P levels with a novel, potent and specific inhibitor of sphingosine kinase-1. Biochem J. 2012;444:79–88. doi: 10.1042/BJ20111929. [DOI] [PubMed] [Google Scholar]
- 257.French KJ, Zhuang Y, Maines LW, Gao P, Wang W, Beljanski V, Upson JJ, Green CL, Keller SN, Smith CD. Pharmacology and antitumor activity of ABC294640, a selective inhibitor of sphingosine kinase-2. J Pharmacol Exp Ther. 2010;333:129–139. doi: 10.1124/jpet.109.163444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Beljanski V, Knaak C, Smith CD. A novel sphingosine kinase inhibitor induces autophagy in tumor cells. J Pharmacol Exp Ther. 2010;333:454–464. doi: 10.1124/jpet.109.163337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Antoon JW, White MD, Slaughter EM, Driver JL, Khalili HS, Elliott S, Smith CD, Burow ME, Beckman BS. Targeting NFkB mediated breast cancer chemoresistance through selective inhibition of sphingosine kinase-2. Cancer Biol Ther. 2011;11:678–689. doi: 10.4161/cbt.11.7.14903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Beljanski V, Knaak C, Zhuang Y, Smith CD. Combined anticancer effects of sphingosine kinase inhibitors and sorafenib. Investigational new drugs. 2011;29:1132–1142. doi: 10.1007/s10637-010-9452-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Beljanski V, Lewis CS, Smith CD. Antitumor activity of sphingosine kinase 2 inhibitor ABC294640 and sorafenib in hepatocellular carcinoma xenografts. Cancer Biol Ther. 2011;11:524–534. doi: 10.4161/cbt.11.5.14677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.White MD, Chan L, Antoon JW, Beckman BS. Targeting Ovarian Cancer and Chemoresistance Through Selective Inhibition of Sphingosine Kinase-2 with ABC294640. Anticancer Res. 2013;33:3573–3579. [PubMed] [Google Scholar]
- 263.Liu K, Guo TL, Hait NC, Allegood J, Parikh HI, Xu W, Kellogg GE, Grant S, Spiegel S, Zhang S. Biological characterization of 3-(2-amino-ethyl)-5-[3-(4-butoxyl-phenyl)-propylidene]-thiazolidine-2,4-dione (K145) as a selective sphingosine kinase-2 inhibitor and anticancer agent. PLoS One. 2013;8:e56471. doi: 10.1371/journal.pone.0056471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Rex K, Jeffries S, Brown ML, Carlson T, Coxon A, Fajardo F, Frank B, Gustin D, Kamb A, Kassner PD, Li S, Li Y, Morgenstern K, Plant M, Quon K, Ruefli-Brasse A, Schmidt J, Swearingen E, Walker N, Wang Z, Watson JE, Wickramasinghe D, Wong M, Xu G, Wesche H. Sphingosine kinase activity is not required for tumor cell viability. PLoS One. 2013;8:e68328. doi: 10.1371/journal.pone.0068328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Taha TA, Kitatani K, El-Alwani M, Bielawski J, Hannun YA, Obeid LM. Loss of sphingosine kinase-1 activates the intrinsic pathway of programmed cell death: modulation of sphingolipid levels and the induction of apoptosis. FASEB J. 2006;20:482–484. doi: 10.1096/fj.05-4412fje. [DOI] [PubMed] [Google Scholar]
- 266.Truman JP, Garcia-Barros M, Kaag M, Hambardzumyan D, Stancevic B, Chan M, Fuks Z, Kolesnick R, Haimovitz-Friedman A. Endothelial membrane remodeling is obligate for anti-angiogenic radiosensitization during tumor radiosurgery. PLoS One. 2010;5 doi: 10.1371/annotation/6e222ad5-b175-4a00-9d04-4d120568a897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Bleicher RJ, Cabot MC. Glucosylceramide synthase and apoptosis. Biochim Biophys Acta. 2002;1585:172–178. doi: 10.1016/s1388-1981(02)00338-4. [DOI] [PubMed] [Google Scholar]
- 268.Chuturgoon A, Phulukdaree A, Moodley D. Fumonisin B induces global DNA hypomethylation in HepG2 cells - An alternative mechanism of action. Toxicology. 2013 doi: 10.1016/j.tox.2013.11.004. [DOI] [PubMed] [Google Scholar]