Skip to main content

Signaling controversy and future therapeutical perspectives of targeting sphingolipid network in cancer immune editing and resistance to tumor necrosis factor-α immunotherapy

Abstract

Anticancer immune surveillance and immunotherapies trigger activation of cytotoxic cytokine signaling, including tumor necrosis factor-α (TNF-α) and TNF-related apoptosis-inducing ligand (TRAIL) pathways. The pro-inflammatory cytokine TNF-α may be secreted by stromal cells, tumor-associated macrophages, and by cancer cells, indicating a prominent role in the tumor microenvironment (TME). However, tumors manage to adapt, escape immune surveillance, and ultimately develop resistance to the cytotoxic effects of TNF-α. The mechanisms by which cancer cells evade host immunity is a central topic of current cancer research. Resistance to TNF-α is mediated by diverse molecular mechanisms, such as mutation or downregulation of TNF/TRAIL receptors, as well as activation of anti-apoptotic enzymes and transcription factors. TNF-α signaling is also mediated by sphingosine kinases (SphK1 and SphK2), which are responsible for synthesis of the growth-stimulating phospholipid, sphingosine-1-phosphate (S1P). Multiple studies have demonstrated the crucial role of S1P and its transmembrane receptors (S1PR) in both the regulation of inflammatory responses and progression of cancer. Considering that the SphK/S1P/S1PR axis mediates cancer resistance, this sphingolipid signaling pathway is of mechanistic significance when considering immunotherapy-resistant malignancies. However, the exact mechanism by which sphingolipids contribute to the evasion of immune surveillance and abrogation of TNF-α-induced apoptosis remains largely unclear. This study reviews mechanisms of TNF-α-resistance in cancer cells, with emphasis on the pro-survival and immunomodulatory effects of sphingolipids. Inhibition of SphK/S1P-linked pro-survival branch may facilitate reactivation of the pro-apoptotic TNF superfamily effects, although the role of SphK/S1P inhibitors in the regulation of the TME and lymphocyte trafficking should be thoroughly assessed in future studies.

Introduction

Remarkable innovations in cancer therapies have been achieved during the last few decades. Despite breakthroughs in treatment, cancer cells still manage to escape host immunity, survive, and progress towards treatment resistance in a subset of patients via multiple mechanisms, many of which remain unclear. One of the common reasons for inefficient cancer elimination is tumor immune evasion, the key mechanism that facilitates the failure of immune surveillance [1, 2]. During the efficient surveillance, cancer cells are designated for clearance if recognized as anomalous; and immune killing mechanisms are activated [1, 3]. The most successful endogenous death-initiating mechanisms rely on cytotoxic cytokines generated by natural killer (NK) T cells and/or phagocytes [1, 4, 5]. During acquisition of immune evasion strategies, the resistant cancer cell develops molecular tools which grant it immunity from NK-mediated cytotoxicity and cytokine attacks [6, 7], resulting in the activation of immunosenescence and promotion of an immunosuppressive tumor microenvironment (TME) [4].

A number of recently developed anticancer/immunotherapy pharmaceuticals aim to restore and strengthen internal surveillance capacity [1, 2]. The immune program relies on CD8+ and NK (CD3+ T lymphocytes) T cell subsets which can identify cancerous (as non-self) cells and delete them through complex clearance mechanisms, including release of inflammatory cytokines such as interferon-γ (IFN-γ) and tumor necrosis factors (TNF) [1, 8]. Initially defined as an endotoxin‐induced cytokine, TNF-α has demonstrated potent cancer-eradicating properties [9]. The ability to suppress cytotoxic cytokine signaling is a crucial survival adaptation for tumor cells. Notably, disruption of TNF-mediated cell death, normally initiated by CD8+ T cells, has been regarded as a major mechanism of immune evasion [1]. TNF-α is produced by the majority of immune cells, including macrophages, neutrophils, fibroblasts, keratinocytes, NK cells, T cells and B cells [5]. The cytokine activates apoptosis mainly through the death receptor (DR) pathway that is initiated by TNF-α receptor‐1 and -2 (TNFR1 and TNFR2) [10, 11]. TNF-α targets not only cancer cells, but also tumor-associated vasculature [6, 12, 13].

The internal tumor-related characteristics (cancer type and stage) and TME define the proapoptotic effects of TNF-α and its ability to inhibit tumor progression [6, 14]. For instance, human lymphoma is, generally, a TNF-sensitive type of cancer that demonstrates good immunotherapy response [15]. However, many solid tumors, including some breast malignancies, are intrinsically resistant to TNF-α effects. Cancer cell resistance to TNF-α cytotoxicity is a complex, multifactorial, and often unclear process. Several intrinsic factors and molecular mechanisms have been found responsible for the development of TNF-resistance, including mutation and downregulation of DR expression [10], activation of anti-apoptotic effectors (such as superoxide dismutase (MnSOD or SOD) [16, 17] and mitogen-activated protein kinase (MAPK)) [18]), diversion of nuclear transcription factor signaling (including nuclear factor kappa-light-chain-enhancer of activated B cells κB (NF-κB)) signaling [19]), and other pro-survival mechanisms. One of the survival pathways associated with anti-apoptotic and growth-promoting mechanisms is represented by the sphingolipid signaling axis [20, 21]. Sphingolipids are involved in the regulation of numerous intracellular mechanisms, both as mediators and effectors of signaling.

Besides regulation of cancer cell growth and metastasis, sphingolipids direct lymphocyte trafficking and cytokine responses, which are key factors in the resolution of inflammation [22,23,24]. The TNF-α/TNF receptors (TNFRs) network has been shown to trigger activation of sphingolipid signaling via sphingosine kinases 1 and 2 (SphK1 and SphK2). SphK1/2, “housekeeping” enzymes, are constitutively expressed and function to support the membrane metabolism in all cell types, including cancer and immune cells. These enzymes are responsible for the synthesis of sphingosine-1-phosphate (S1P), an established regulator of pro-survival machinery in multiple cancers. S1P and its transmembrane receptors (S1PRs) were found to be involved in the regulation of cytokine signaling and chronic inflammation [23,24,25]. Considering that sphingolipids, particularly those within the SphK/S1P/S1PR axis, are important effectors in the regulation of cancer cell survival and immune responses, these molecules may be considered as the key contributors to the development of immunotherapy resistance. However, the role of sphingolipids in the development of solid cancer resistance to immunotherapies and specifically to TNF-α-induced apoptosis remains to be clarified. Therefore, this review aims to discuss mechanisms of sphingolipid involvement in TNF-α-resistance in cancer cells and provide insights into the association of immune evasion with regards to SphK/S1P/S1PR axis.

TNF superfamily signaling network: the cell death gatekeeping system

The TNF superfamily and TNFR network are crucial regulators of the extrinsic cell death (apoptosis) pathway and cancer cell surveillance [10, 26, 27]. The superfamily consists of signaling molecules (referred to as cytokines) that bind 29 corresponding receptors, including TNFRs [14, 27, 28]. Respective of what stimuli and/or receptors are involved, TNFRs can trigger not only several types of programmed cell death (apoptosis, necrosis, and anoikis), but also cell differentiation, migration, and proliferation [28,29,30]. The death-triggering mechanisms have been extensively reviewed [26, 28]. Several TNFR subtypes have been grouped according to the presence or absence of intracellular death domains (DDs, six α-helical fold fragment) [29]) (Fig. 1). The DD-containing subfamily includes TNFR1 (often named as p55, or DR1, or TNFRSF1A), Fas (CD95/TNFRSF6), DR3 (TNFRSF25), TNF-α-related apoptosis-inducing ligands (TRAIL) receptor 1 (TRAIL-R1, 1TNFRSF10A, DR4, CD261), TRAIL receptor 2 (TRAIL-R2, TNFRSF10B, DR5, CD262) [2, 4], DR6 (TNFRSF1), and EDAR [28, 31].

Fig. 1
figure 1

Death receptors (DR) and their ligands intracellular network. Ligands (FasL, TRAIL, TNF-α, TL1) can activate signaling cascades required for the activation of apoptosis and other complex cell responses. TNF-α/TRAIL/FasLs (and other ligands) bind the corresponding receptors (Fas, TRAIL-R1, and TNFR1) and activate apoptosis and necroptosis through interactions between death domains (FADD), TRADD adapter, and various caspases. Both TNFR1 and TNFR2 can trigger the classical NF-κB signaling. Binding of TNF to TNFR1 results in the formation of protein Complex I. Recruitment of IKKα/β through NEMO promotes activation of NF-κB and TAK1 induces MAPK signaling. Activation of the alternative NF-κB pathway is also possible via multiple mechanisms, leading to induction of survival effectors (MAPK and FLIP) which may counterbalance apoptosis (conditional). Complex I formation may also trigger pro-inflammatory and survival gene expression through these signaling pathways. Complex II formation results in the activation of caspase-8 and apoptosis. Should caspase-8 be inhibited, necroptotic cell death can occur instead. Abbreviations: FasL, Fas ligands; TRAIL, TNF-related apoptosis-inducing ligand; TNF-α, tumor necrosis factor-α; TL1, a novel TNF-like cytokine; TNFR1, TNF-α receptor 1; TNFR2, TNF-α receptor 2; FADD, FAS-associated death domain protein; TRADD, TNF receptor type 1-associated death domain protein; NF-κB, nuclear factor-κB; IKKα/β, IκB kinase α/β; NEMO, NF-κB essential modulator; TAK1, TGFβ-activated kinase 1; MAPK, mitogen activated protein kinase; FLIP, FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein

The complexity of TNFR network is associated with the continuously expanding TNF superfamily of cytokines which presently includes 19 different ligands. TNF-α and TNF-β were identified several decades ago and have since been heavily studied. Other TNFR ligands, including homologous lymphotoxin (LT), ligands for Fas (or CD95), TRAIL (or APO2L), receptor activator of NF-κB ligand (RANKL), and osteoprotegerin ligand (OPGL) are relatively new members of this large family with poorly defined roles in cancer surveillance [14, 32, 33]. The full-length TNF-α is encoded by the TNF-α gene on human chromosome 6 [5]. TNF isoforms interact with TNFR1 and TNFR2 (defined as p75 or DR2) [11, 29, 32], leading to the formation of two signaling complexes I and II and cell death induction [3, 34] (Fig. 1). TRAIL can also induce apoptosis via binding to DR4 and DR5 in cancer cells [35, 36]. Fas and TRAIL receptors, the dual-signaling receptors, belong to the third DR subfamily with DD at their C-termini and TRAF recruitment domains at the opposing N-termini, providing them with the ability to activate NF-κB [34] (Fig. 2). Like the other members of the TNFR family, DR4/5 not only activate apoptosis, but can also regulate cell differentiation and proliferation [28, 30]. Functional TRAIL receptors (DR4/DR5) are widely expressed [37].

Fig. 2
figure 2

The dichotomy of TNF/TNFR effects is associated with activation of antagonizing effects, both promoting and counteracting cell death in immune cells. The resulting effect is defined by the active involvement of intracellular death machinery, which may be overruled by activation of pro-survival effectors. Both pathways lead to production of cytokines and propagation/differentiation of specific immune cells and their recruitment to the site of infection. Abbreviations: DAMPs, damage-associated molecular patterns; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor kappa B; PAMPs, pathogen-associated molecular pattern molecules; PRR, pattern recognition receptors; TNF-α, tumor necrosis factor-α; TNFR1, tumor necrosis factor-α (TNF-α) receptor-1; Ub, ubiquitin

Activation of TNFR1 by its ligand is followed by receptor solubilization, membrane shedding, and binding of TNFR-associated death domain (TRADD), TNFR-associated factor 2 (TRAF2), receptor-interacting protein (RIP) kinase (RIPK) and transforming growth factor-β-activated kinase 1 (TAK1) proteins, leading to the activation of the classical NF-κB pathway [10, 32]. Recruited TRAF triggers ubiquitin ligase complexes (the upstream activator of NF-κB, activator protein 1 (AP-1)), p53 (tumor suppressor), and other transcription factors (Fig. 1) [10, 14, 32]. The internalized TNFR1 complex may also activate growth-regulating MAPK signaling effectors, including c-Jun N-terminal kinase (JNK) and p38 cascades [30, 33], ERK1/2 pathway, Fas-associated death domain (FADD)-like IL-1β-converting enzyme (FLICE) inhibitory protein (FLIP) [38], Bcl-2 (B-cell lymphoma 2) and Bcl-xL, and nitric oxide (NO) production [18, 38, 39]. Both TNFRs can also activate the phosphatidylinositol 3-kinase (PI3K)/Akt (protein kinase B) anti-apoptotic pathway in a TRAF2-dependent manner [30, 40, 41]. During DR-dependent activation of NF-κB and p53, TNF-α triggers extensive downregulation of XIAP, as well as cellular inhibitors of apoptosis protein-1 and -2 (cIAP1 and cIAP2), resulting in DNA fragmentation [42]. For instance, TNF-α was shown to induce apoptosis and DNA fragmentation within 24 h of treatment in MCF-7 mammary adenocarcinoma cells [42]. The quick (non-genomic) effects of TNF-α in MCF-7 cells start with the internalization of TNFR1. This results in the activation of caspase 8 and the cleavage of BID (a pro-death Bcl-2 family protein) [43]. Truncated tBID migrates to the mitochondria, causing activation of Bcl2 associated X protein (Bax)/Bcl-2 antagonist killer 1 (Bak), and release of cytochrome c (cyt c) [37, 43]. Following this, mitochondrial damage, and excessive production of reactive oxygen species (ROS) were observed [44]. Together with activated caspase 9, cyt c induces formation of the cytoplasmic apoptosome and irreversible propagation of apoptosis [37]. Death receptor (DR) activation can also inhibit expression of anti-apoptotic Bcl-2 and Bcl-xL [34, 39, 45].

TNF expression is tightly regulated in normal cells and commonly induced during pro-inflammatory responses in various immune cells, fibroblasts, endothelial, and epithelial cells [46]. Macrophages and T cells are the major sources of secreted TNF-α, which targets all innate immunity cells responsible for pro-inflammatory effects in the TME, including differentiation of CD4+/CD8+ T cells [47, 48]. TNF-α may simultaneously activate both anti- and pro-apoptotic signals, that are required for the adaptation of immune system responses to dynamic intrinsic and/or extrinsic changes (Fig. 3). Using its non-apoptotic network, TNF-α induces differentiation of various immune cells, including monocytes/macrophages, microglia, Langerhans cells, and Kupffer cells [27]. Cancer spreading (metastasis) may also be triggered by TNF-α via the epithelial-to-mesenchymal transition (EMT) process [47, 49]. The activation of survival mechanisms was also noted during DR signaling in cancer cells and cells within the TME [5, 13, 40]. For instance, despite previously showing destruction of cancer-supporting blood vessels by TNF-α in cancer patients [12], intracellular TNFR1 signaling in endothelial cells activates two opposing pathways, one with pro-apoptotic effects [12], and another NF–κB-mediated pro-survival pathway [5, 13]. The multidirectional outcome of TNF-α signaling demonstrates the convoluted nature of this cytokine’s mechanistic actions, many of which remain largely unclear. The major anti-apoptotic effectors and pathways are discussed in this study, focusing on their connections to the sphingolipid network.

Fig. 3
figure 3

Involvement of TNF-α in the regulation of immune cell differentiation during inflammation and cancer progression. Promoting reprogramming of the TME, TNF-α was suggested to play central role as a connector of inflammation with cancer spreading. Abbreviations: INF-γ, Interferon‐γ; TH1, Type 1 T helper cell; TH2, Type 2 T helper cell; TNF-α, tumor necrosis factor-α

Conventional mechanisms of TNF-α/TRAIL resistance in cancer cells

Cancer cells adapt to avoid recognition and elimination by the immune system (referred to as cancer immune evasion) [41]. Resistance to TNF-α/TRAIL is cancer-specific and can be mediated by several anti-apoptotic mechanisms. Among the most prominent TNF-α-resistance mechanisms are abnormal DR expression and functioning [50, 51], stoichiometry of the relevant ligand, heterozygous mutations and/or post-translational modifications of DRs and their ligands [50], mitochondrial dysfunctions, deficiency (lower expression or silencing) of key pro-apoptotic proteins/apoptosis pathway effectors (tumor intrinsic and host-related factors), low immunogenic capacity of immune effectors in the TME, and activation of complex pro-survival machinery [39, 51, 52]. Inhibition of TNF-α/TNFR-associated apoptosis was also detected in cells with dysregulated oxidative phosphorylation (OXPHOS) and/or abnormal expression/signaling of energy metabolism regulators, such as MnSOD [17, 53]. Considering the high level of cancer heterogeneity, complex resistance mechanisms may be present within one cancer tissue. Accordingly, the outcome of TNF/TRAIL-induced responses is determined by the relative contribution of the combined apoptotic signals, transmitted by DRs and their downstream targets, and pro-survival actions of cIAPs and other pro-survival effectors, including the growth-promoting and immunomodulatory components of the sphingolipid network. In TNF-α-resistant cancer cells, the combined pro-apoptotic signals are overwhelmed by pro-survival machinery, leading to cancer progression (Fig. 4). Notably, sphingolipid signaling contributes to many of the forementioned mechanisms. In this study, immune evasion-linked mechanisms of TNF/TNFR interactions within the sphingolipid network will be covered. The sphingolipid signaling axis is a recent addition to the list of cancer resistance-promoting modalities. Notably, sphingolipids were shown to be part of many different pro-survival and growth-stimulating networks, and thus may contribute to TNF-α resistance at multiple levels [21, 54].

Fig. 4
figure 4

Interplay between cancer cell intrinsic factors, TME, and host-related factors that contribute towards the development of TNF/TRAIL resistance and metastasis. Abbreviations: Bcl-2, B-cell lymphoma 2; BMI, body mass index; DCs, dendritic cells; DR, death receptor; FLIP, FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein; IAPs, inhibitors of apoptosis; MiRs, micro ribonucleic acids; ROS, reactive oxygen species; TAMs, tumor-associated macrophages; TNF, tumor necrosis superfamily; TRAIL, TNF-related apoptosis-inducing ligand; Tregs, regulatory T cells

Sphingolipids as mediators, facilitators, and inhibitors of TNF-α-signaling

SphK/S1P/S1PR axis: focus on growth-promoting and anti-apoptotic effects

A significant role in the regulation of sphingolipid signaling and metabolism by TNF-α has been demonstrated in multiple studies [13, 21, 55,56,57] (Table 1). In turn, both pro-survival and pro-apoptotic sphingolipids were implicated in the regulation of TNF-α/DR-induced effects.

Table 1 The SphK1/S1P/S1PR axis mediates and directs TNF-α signaling

For instance, activation of apoptosis by DRs has previously been shown to downregulate SphK1 protein expression and activity via proteasomal degradation [158, 159]. Sphingolipids are not only structural components of all biological membranes, but also signaling and regulatory molecules. The variety of sphingolipid family members and their functions have been reviewed previously [160, 161]. The accumulation of apoptosis-inducing members, ceramide and sphingosine, was noted during DR-signaling [54, 160, 161] (Fig. 5) (Table 1). Key enzymes responsible for ATP-dependent metabolism of sphingosine and generation of S1P include SphK isoforms (SphK1 and SphK2), which are found in cytoplasmic, mitochondrial, and nuclear compartments [57, 153, 161, 162]. Activation of SphK1 and S1P synthesis are responsible for growth-stimulating and pro-survival effects in normal and cancer cells [21, 54, 57, 163]. The role of SphK1 will be considered as a counterbalancing anti-apoptotic force for DRs in this review [61, 164]. Both SphK1 and SphK2 were suggested to mediate numerous cellular responses to external stimuli and stress [21, 54, 151, 153]. Notably, SphK2 was reported to suppress proliferation and facilitate propagation of apoptosis, thus playing an opposite role to SphK1, although this hypothesis remains to be confirmed [25, 165]. The roles of SphK2 in DR signaling and propagation of apoptosis have been discussed previously [166].

S1P is a multifunctional messenger which can bind both intracellular targets and membrane-located (extracellular) receptors. Paracrine-, blood-, or lymph-released S1P binds transmembrane S1P receptors (G-protein coupled S1PRn (n = 1–5)), which are the established effectors of growth and survival [21, 167] (Fig. 5). S1PR1 is abundantly expressed in all cell types, including large variety of immune cells [168, 169], indicating the high importance of this receptor for the regulation of vital cell functions. The receptor signals via Gi/o heterotrimeric proteins which may inhibit adenylyl cyclase and activate potassium channels [170].

S1PR2 is also ubiquitously expressed [168, 171], although the receptor signaling remains less investigated. Notably, S1PR2 was shown to inhibit colorectal cancer tumorigenesis [172]. The activation of S1PR2 or S1PR3 was linked to the activation of Gi/o, Gq, and G12/13, suggesting the potential activation of large variety of downstream effectors [173]. Aside from normal cell types, S1PR3 is highly expressed in various cancer cells and was shown to stimulate cancer progression [21, 153, 168]. It is common to observe the co-expression of different S1P receptors, especially presence of S1PR1 and S1PR3 within one cell type which may indicate cooperation of signaling among the receptors [174]. In comparison to S1PR1 and S1PR3 effects, S1PR4 was found to be growth-inhibitory in some immune cells [175], while its role in the lymphocyte trafficking and expansion was extensively discussed [169]. The receptor may regulate the cytotoxicity of T cells towards cancerous tissues [176], although downstream signaling pathways of S1PR4 remain largely unclear. S1PR5 was also shown to regulate T cell subtype maturation and functions [177]. Gi/o and G12/13 were shown to transmit S1PR4 and S1PR5 signals in normal and malignant cells [178]. The expression of S1PRs in both cancer and immune cells represents a debatable phenomenon which was recently reviewed [169]. To complicate the problem, the level of S1PR expression may vary during morphogenesis, cell growth and differentiation [153]. The growth-promoting and/or pro-carcinogenic role of S1PR1 and S1PR3 seems confirmed. However, current knowledge does not provide unequivocal answer about the role of different S1PRs in specific cancer or immune cells. The problem is complicated by the high level of cancer and immune cell heterogeneity, the different combinations of S1PR expression, and diversity of S1PR downstream effectors.

S1P may bind other target molecules important for sphingolipid metabolism and signaling. For instance, phosphatases can bind, dephosphorylate approximately half of the intracellular S1P in endoplasmic reticulum, and direct this sphingolipid towards de novo ceramide synthesis during membrane metabolism and recycling [57, 160] (Fig. 5). S1P lyases can also bind S1P and degrade it into phosphoethanolamine and hexadecenal, which can be used for further glycerolipid and phosphatidylethanolamine syntheses [57, 160]. Large amounts of S1P were detected in the circulation where the this sphingolipid forms complexes with high-density lipo-proteins (HDL) [179]. Substantial extracellular levels of S1P are maintained by erythrocytes [180], platelets [181, 182], endothelial cells [183], and various immune and malignant cells [24, 167]. In majority of these cells, S1P secretion is mediated by ATP-binding cassette transporters (ABC-transporter) [180, 184]. S1P gradient, the difference between the intra- and extra-cellular concentrations of S1P, modulates S1PRs expression and represents a novel factor in the regulation of S1P signaling in the immune system and circulation [169, 184].

Fig. 5
figure 5

The sphingolipid signaling pathway. Various sphingolipids molecules (second messengers) can be derived from the membrane lipid sphingomyelin by sphingomyelinase (SMase) and metabolised by a “rheostat”-forming network which regulate homeostasis. Accumulation of ceramide and sphingosine can tip the balance towards apoptosis and other types of cell death. Activation of SphK1/2, production of S1P (and activation of S1P receptors), and/or S1P degradation by S1P lyase to hexadecenal and ethanolamine phosphate result in pro-survival and growth-promoting effects. Sphingomyelin membrane content can be restored through activation of sphingomyelin synthase (SM Synthase), which can also help to minimise the content of ceramide. The amount of sphingosine can be increased via inhibition of SphK1/2 and or through activation of S1P phosphatase

The proliferation-stimulating effect of the SphK/S1P/S1PR axis is mediated by growth factor network, including MAPK and epidermal growth factor receptor (EGFR) [21, 167, 185]. Various growth factor receptors, including EGFR and VEGFR, were also shown to induce SphK activation, increase the level of S1P production, and transactivate S1P receptors [21, 167, 185]. Aside from EGFR/ERK1/2 [185, 186], S1PR activation influences signaling patterns of various global targets, such as Notch [187], signal transducer and activator of transcription (STAT)3 [23], Akt/mammalian target of rapamycin (mTOR) [188, 189], NF-κB [186, 190], Hippo-YAP pathway [191], and cyclic-AMP responsive element binding protein (CREB) [192]. Cell-, tissue-, and disease-specific expression of S1PR is mediated by coupling to a range of G proteins [193] and/or other receptors (transactivation mechanisms) [21, 57]. S1PRs network interacts with growth factor receptors, including EGFR [167, 185], vascular endothelial growth factor (VEGF) receptors [22, 23], and IGF receptors [194]. Moreover, the SphK/S1P/S1PR axis may be activated by various hormones and cytokines during basic cell growth maintenance, cell differentiation, and metabolic transformations in cancer cells [21, 193]. The mutual transactivation of the network by growth factor effectors provides limitless opportunities to counterbalance apoptosis.

S1P may trigger S1PR-independent mechanisms via binding to other non-traditional receptors, including transcription factors. S1P was demonstrated to induce S1PR-independent activation of TRAF2 [71, 72], although the effect seems cell- and tissue-specific [195]. S1P can also stimulate gene transcription via binding to histone deacetylase 1/2 (HDAC1/2), an epigenetic regulatory enzyme [196]. Activation of endoplasmic reticulum stress and inflammation in keratinocytes was determined to be mediated by S1P binding to the endoplasmic chaperone protein GRP94, recruitment of TRAF2 to inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α), and NF-κB signaling. S1P binding to heat shock protein (HSP) 90α was also detected [197]. S1P binds and inhibits ceramide synthase 2 (CerS2), leading to blockade of ceramide (pro-apoptotic effector) synthesis [198]. There may be other not-yet-identified S1P receptors, including some lipid mediators. For instance, myristate, a component of milk fat, was shown to activate pro-inflammatory responses (such as release of TNF-α and induction of COX-2) in colon tissues. Observed effects of myristate were mediated by an unspecified, intracellular target of S1P and were not blocked by S1PR inhibition [163]. Thus, the S1PR-independent effects of S1P are not uncommon, indicating versatility of this sphingolipid signaling.

Regulation of apoptosis by the SphK/S1P/S1PRs axis

TNF-α-induced effects are not limited to S1P and instead are mediated by a variety of sphingolipids generated during distinct metabolic processes. It has been postulated that TNF-α triggers both pro-apoptotic (ceramide-related) [161, 185] and anti-apoptotic (SphK/S1PRs axis) signaling branches of the sphingolipid network [57, 72, 92]. Activation of apoptosis and autophagy by ceramide has been extensively reviewed elsewhere [147, 161, 199, 200]. A concept of dynamic sphingolipid-based regulation, called a “sphingolipid rheostat”, was suggested to describe a shift towards apoptosis triggered by increased production of ceramide; while a generation of S1P provides a more sustainable cell survival environment and shifts the balance toward anti-apoptotic effects [161, 199, 200].

Ceramide metabolism in normal and cancer cells is regulated by several enzymes, including glucosylceramide synthase, sphingomyelin synthase, ceramide kinase, ceramidases, and SphK [200]. These enzymes define cell life-to-death balance. However, other cell death regulators, including p53, are involved and often provoke unavoidable cell death [200, 201]. A complex relationship between p53 and ceramide has been described, accentuating the importance of ceramide accumulation during activation of stress responses and DNA damage [202]. Notably, ceramide and p53 can trigger signaling effectors upstream or downstream of each other, resulting in sometimes contradictory effects described elsewhere [200, 201].

The SphK/S1P/S1PRs axis is a powerful molecular tool for the regulation of cell survival. The ability of S1P to protect against apoptosis has been well documented in many normal and malignant cell types exposed to pro-apoptotic stimuli, such as TNF-α/Fas ligands [71, 75, 156], serum deprivation [203], ionizing radiation [204], and anticancer drugs [21, 54, 119, 157]. Inhibition of S1P signaling was shown to enhance apoptosis. For instance, treatment of HCC-38 and MDA-MB-468 cells with SphK1 inhibitor PF543 and doxorubicin resulted in synergistic apoptosis-enhancing effects [205]. During carcinogenesis, the SphK/S1Ps axis is highjacked by cancer cells to promote survival. Its role in the development of cancer drug resistance was extensively reviewed and is associated with transactivation of growth-factor networks, stem cells, and other molecular adaptations [21, 54]. Mechanisms of SphK/S1PR involvement in the regulation of TNF-α-induced cell death are complex and sometimes controversial. Sphingolipids trigger signal transduction branching at several different points of the network. There is a possibility that cancer-induced transformation of SphK/S1PRs signaling is responsible for the development of TNF/TRAIL resistance in cancers, although the hypothesis remains untested. Several interactive hotspots (molecular effectors and networks) between DRs and SphK/S1PR networks are discussed below.

Regulation of inflammation by the SphK/S1P/S1PRs axis

Inflammation is recognized as one of the contributing and promoting factors of carcinogenesis. The SphK1/S1P axis is part of a large signaling network formed by key pro-inflammatory cytokines, such as TNF-α [58, 72, 156], IL-6 [206], IL-1β [81, 207], CCL5 chemokine (regulated on activation, normal T cell expressed and secreted (RANTES)) [128], and others [54, 208]. Bacterial lipopolysaccharide (LPS) was shown to induce SphK/S1P/S1PR3 activation [209], accentuating the potential involvement of sphingolipids during antibacterial responses. The activation of the sphingolipid axis was accompanied by induction of major genes responsible for the propagation of inflammation (COX-2, IL-1β, IL-6, TNF-α, iNOS) [206, 209]. The effect is mediated by a two-way signal-propagating process. Inflammatory responses mediated by COX-2 also required activation of the SphK1/S1PRs axis during progression and resolution of infection [13, 207]. Accordingly, inhibition of S1PR3 by TY52156 resulted in the inhibition of pro-inflammatory gene signaling [209]. Pharmacological SphK1 inhibition (or genetic silencing) also helped to recover the metabolic characteristics of T cells and induced immune antitumor activity [210]. Sphk1 inhibition may improve immunotherapies and stimulate responses to anti-PD-1 and other immune checkpoint inhibitors (ICIs) [211].

Inflammation is a normal immune response by an organism facing infection. Various normal cells may be affected by inflammation and respond to stimulation by cytokines. Sphingolipids are important mediators of normal inflammatory responses in non-malignant cells. Crosstalk between the Fas network and endogenous sphingolipids was observed in various normal cells during pro-inflammatory processes, including osteoclasts from mice with rheumatoid arthritis (RA). Increased level of S1P was associated with osteoclast apoptosis during the development of RA [212]. Furthermore, COX-2, iNOS, prostaglandin E2 (PGE2), IL-1β, and TNF-α signaling pathways activated the S1P network in macrophages during LPS-induced inflammation [75, 93, 208]. S1P mediates various immune responses, including mast cell degranulation, migration of neutrophils, and maturation of lymphocytes [213]. Interestingly, an anti-inflammatory role of SphK1/S1P was also observed [214]. For instance, activation of S1PR2 prevented excessive macrophage recruitment in a peritonitis model in vivo [215], although the effect is macrophage type- and/or pathology-specific [216]. Levels of IL-1β and IL-18 in plasma of wild-type mice were reduced by application of JTE-013 (S1PR2 antagonist) [216]. In SphK1-null mice (SphK1−/−), SphK1 was found responsible for suppression of LPS-induced neutrophil oxidant production. Binding of SphK1 to JNK resulted in stabilization of JNK and inhibition of JNK binding to the JNK-interacting protein 3 (JIP3). The change of “partners” prevented the activation of nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidase and NF-κB activation, indicating a novel mechanism of anti-inflammatory signaling via SphK1/JNK interactions [217].

SphK1/2 is involved in the regulation of inflammation in other non-cancerous tissues, though its role is not straightforward. In an in vivo study of arthritis, downregulation of SphK1 decreased inflammation, while total knockdown of SphK2 resulted in a heightened inflammatory response [99]. Similar diversity of the effects of SphK1/2 knockdown was observed during induction of inflammation in the colon [23, 102]. In intestinal epithelial cells, SphK1 was involved in TNF-α/COX-2 pro-inflammatory signaling during exposure to myristate [151]. In neuronal tissue, acetylation of COX-2 via non-specific acetyltransferase activity was also linked to SphK1 activities [218]. Interestingly, triggering of the S1P network resulted in anti-inflammatory effects and suppression of IFN and STAT1 functions [205]. STAT1, a pro-apoptotic effector, controls expression of several cell cycle regulators, enhances death-promoting functions of Bak, and blocks transcription of anti-apoptotic Bcl-2 and Bcl-xL [219]. STAT1 may also induce expression of DR ligands, such as TNF-α, FAS, and TRAIL [45]. Conclusively, limited S1P production via SphK1 knockdown/inhibition may provide an effective tool for a re-activation of the STAT1/IFN pathway [220]. The role of SphK2 in this process remains controversial and should be clarified in future studies. There are reports which indicated contribution of SphK2 in the activation of pro-inflammatory processes [221], which can be (potentially) employed to facilitate anticancer therapies. It is essential to keep in mind the multifactorial role of S1P and provide only cell-targeted reduction of S1P levels and tissue-specific inhibition of SphK1/2 inhibition, as it would be counterproductive to eliminate the effect of sphingolipids on lymphocyte trafficking [54, 193] and anticancer activation. Considering the regulatory role of sphingolipids in T cells, the impact of the SphK/S1P axis should be considered during cancer progression (Fig. 6). The activation of Sphk1/S1PR may significantly change the ability of T cells to recognize and eliminate cancer cells (immunosuppressive effects). To make cancer cells susceptible to T cell recognition/killing, application of nanocarriers and epigenetic reprogramming of malignant cells was suggested as a promising therapeutic approach in this field [222, 223].

Fig. 6
figure 6

Dichotomy of TNF-α–induced signaling in cancers is hypothetically linked to sphingolipid balance where the relative amounts of ceramide and S1P cause cell proliferation, survival, or death. Stressed cells can increase ceramide in response to TNF-α, resulting in growth arrest and apoptosis. However, in some cells TNF-α can activate SphK and mitigate its pro-apoptotic ability via production of growth-stimulating S1P and activation of S1PR1-5. Abbreviations: CerS, ceramidase; GPCR, G-protein coupled receptor; HDAC1/2, histone deacetylase 1 and 2; hTERT, human telomerase reverse transcriptase gene; JNK, c-Jun NH2-terminal kinase; PPARγ, peroxisome proliferator-activated receptor-γ; SMase, sphingomyelinase

Role sphingolipids in the regulation of lipid metabolism and obesity-associated inflammation

Obesity is regarded as a powerful contributor in the development of cardiovascular diseases and cancer [224, 225]. For instance, obesity-driven inflammation was linked to colorectal cancer progression and metastasis [225]. Low levels of inflammation were found to mark increased fat deposition [226]. Inflammation is promoted in fat tissue by several mechanisms, including imbalanced metabolism, activation of pro-inflammatory immune cells, secretion of cytokines, and other immune mediators [227, 228]. Macrophages and neutrophils located in adipose tissue were shown to secrete pro-inflammatory cytokines (such as IL-1, IL-6, IL-8, C-reactive protein (CRP), TNF-α) [229] (Fig. 3). Accumulation of macrophages in fat tissue and increased secretion of adipokines (fat hormones) were linked to the activation of several signal transduction pathways (JAK/STAT, MAPK, PI3K, mTOR, and 5’AMPK signaling pathways), COX-2 downregulation, and dysregulation of mRNA expression [230]. Excessive saturated fatty acids (SFAs), which are generated in adipose tissues, induce pro-inflammatory signaling in many cell types, including adipose tissue macrophages. SFA deposition also results in enhanced expression of cytokines, such as TNF-α and IL-6 [231]. Interestingly, obesity-related inflammation may trigger carcinogenesis, promote metastasis, and promote cancer immune evasion [229].

The primary function of fat-regulating agents, or adipokines, is to control fat deposition and utilization [232]. Adipokine leptin can suppress appetite by acting upon several mediating effectors, including leptin receptor (LEPR) in neurons [233,234,235]. Surprisingly, cancer cells are also responsive to leptin and express adipokine receptors. Adipokines may activate pro-carcinogenic and metastasis-promoting effects [14, 233]. LEPR belongs to class 1 of the cytokine receptor family and is reported to play significant roles in carcinogenesis [236]. It has been shown that leptin induces expression of SphK1 in breast cancer [237]. In another study, leptin-activated SphK1 was demonstrated to trigger IL-6 secretion which maintained low levels of inflammation in the effected tissues [237]. Alternatively, SphK1 deficiency and pharmacological inhibition were associated with adipogenesis, increased expression of regulatory genes associated with adiposity, and production of anti-inflammatory molecules IL-10 and adiponectin. Inhibition of SphK1 resulted in lower recruitment of macrophages and reduced production of TNF-α and IL-6 in adipose tissues [238]. However, sphingolipid regulation of adipose tissue metabolism remains controversial [237, 238].

Interactions between the adipokine network and sphingolipids are delicately balanced by a feedback mechanism of signaling. The role of sphingolipid metabolizing enzymes in adipose tissue has been assessed in several recent studies [151, 239]. SFAs were reported to serve as substrates for ceramide synthases (CerS) and serine palmitoyl transferases (SPT). Both CerS and SPT can modify sphingolipid metabolism [151]. Accordingly, the level of pro-apoptotic ceramide was increased by SFAs (and high fat diet). Moreover, enhanced levels of sphingosine and S1P were found in the blood plasma, liver, and skeletal muscle of rodents following SFA (high fat diet) administration in vivo [237, 239]. In another study utilizing rats, increased expression of SphK2 (but not SphK1) was observed during consumption of fat [240]. However, these studies did not assess the level of pro-inflammatory signaling in those animals, and, therefore, it remains unclear whether these changes led to the propagation of inflammation or just aimed to minimize fat deposition.

The cancer-regulating role of CerS, the dual mediator of adipose tissue effects and sphingolipid metabolizing enzyme, is especially intriguing considering recent findings in breast adenocarcinoma cells. High level of CerS6 decreased phosphorylation of Akt and ERK in MCF-7 breast cancer cells. This effect was associated with inhibition of MCF-7 cell proliferation and activation of the mTOR pathway [241]. The study also analyzed public data using the Cancer Genome Atlas database. Investigators determined the presence of invasive breast carcinoma is negatively associated with CerS6/S1PR2 or CerS6/SphK1 expression. This study suggested that mTOR activity depends on the balance between the production of S1P (by SphK1) and C16-ceramide (by CerS6) [241]. However, it was not tested whether adipose tissue metabolism or adipokines are involved in the regulation of CerS6 and mTOR signaling in breast cancer tissues. The association of these effectors with inflammation and resistance to immunoediting was also not assessed.

A recent study utilized a mice model to show a myristate-enriched milk fat-based diet (MFBD) increased the expression of TNF-α in colonic tissues [151]. MFBD also elevated S1P levels in intestinal epithelium via regulation of SphK1 and JNK [151]. Thus, this data established a link between fat-based diet, activation of SphK1, and increased production of TNF-α (inflammation) in the colon. Further efforts are required to determine whether this condition may potentially result in the inactivation of the anticancer capacity of the TNF network and lead to apoptosis resistance.

Since TNF-α can activate the SphK1/S1Ps receptor axis (and vice versa), it is tempting to hypothesize that this mechanism provides a circuit point which may be essential for internal outcomes of the cell/tissue responses to pro-inflammatory signals. Depending on the existing balance within the sphingolipid network of cancer cells/tissue, the activation of TNF-α/TNFR axis may result in either activation of proliferation (so-called TNF-α resistance mechanism) or apoptosis (traditional death-promoting pathway). The relevant question to ask, what is the 3rd factor(s) that tips the scale of metabolism towards one or another biological process? Considering the role of obesity as a contributing factor in carcinogenesis, adipokines can serve as important contributing factors which may link obesity to advanced cancers and drug resistance. However, high cancer cell heterogeneity (genetic/inherited factors) and the impact of established anti-apoptotic effectors (proteomics and epigenetics) must not be overlooked as powerful contributors.

The role of sphingolipids in the interaction between ubiquitin-editing enzyme A20 and pro-apoptotic TNF-α signaling

Diverse A20 functions have been linked to dual deubiquitylating enzyme (DUB) and E3-ubiquiting ligase actions [242]. A20 is encoded by TNF-α-induced protein 3 (TNFAIP3) gene, a critical anti-inflammatory effector in the TNF network [243]. Anti-apoptotic and cancer stem-cell (CSC) promoting effects of A20 were reported [244]. A20 was defined as an anti-apoptotic and anti-inflammatory effector [245], although A20’s role in the regulation of cancer immune evasion remains largely unclear. For instance, liver regeneration was associated with A20 activities that promoted IL-6/STAT3 pro-inflammatory signaling and suppressor of cytokine signaling 3 (SOCS3) proteolysis [246].

Overexpression of A20 was detected in multiple solid tumors [244], including basal breast cancers with advanced metastatic properties and EMT phenotype [49, 247]. Increased A20 expression in triple-negative breast cancers (TNBC) protected from TNF-α-induced cytotoxic cell death [247]. Lee and co-authors [247] demonstrated that TNF-α induced association of A20 with HSP70, the protein involved in proteolytic removal of damaged and/or incorrectly folded proteins. The formed complex demonstrated increased stability and facilitated resistance to apoptosis in TNBCs, although the effect was not observed in estrogen receptor positive (ER+) luminal cell lines. The failure of TNF-α to trigger A20/HSP70 association in ER+ cells suggested a role for ER in this signaling network [247]. Notably, ER-linked signaling was shown to trigger the SphK1/S1PR axis in ER+ cells (such as MCF-7 cells) [167], while TNF-α was shown to induce apoptosis [153]. Complex and controversial interactions between A20 and estrogen/ER networks were observed [248]. The reported data suggested a potential mutual association between all four effectors (TNF-α, sphingolipids, estrogen, and A20), which remains to be assessed.

A20 was shown to interact with sphingolipid signaling and mediate resistance to Fas/FasL-dependent apoptosis [249]. A recent study indicated that δ-tocotrienol (δTE, a vitamin E form) can stimulate the expression of A20 and inhibit TNF-α-induced activation of NF-κB and LPS-stimulated IL-6 in a concentration- and time-dependent manner in RAW264.7 macrophages [249]. These findings were validated in A20 knockout cells. Treatment with δTE induced generation of dihydroceramides, marked by the activation of cellular stress. Supporting the role of sphingolipid metabolism in A20-dependent effects, myriocin (an inhibitor of de novo sphingolipids synthesis) partially inhibited induction of A20 and A20-induced inhibition of NF-κB by δTE in immune cells [249]. However, this pathway was not tested in cancer cells. Moreover, pro-apoptotic and growth-inhibitory effects of TNF-α were not always associated with the induction of classical NF-κB signaling [250], indicating roles of other genomic and non-genomic mechanisms. A20 was also found to be involved in the regulation of autophagy in T cells [251, 252]. However, less-differentiated (immature) T cells are resistant to TNF-α-induced apoptosis [253]. Considering that T cells express S1PR and are responsive to S1P stimulation [193], it remains to be determined whether the A20/autophagy/sphingolipids signaling mechanism is active in TNF-α-resistant cancer cells and the TME. Supporting the importance of this investigation, sphingolipids were also found to be involved in the regulation of autophagy in different cell types [147, 199].

NK signaling, TME, and sphingolipids

Human NK cells are a crucial part of the innate immune system responsible for the identification of self/non-self-CD1d (dendritic cells)-presented glycosphingolipids and cytokine-elaborating response [254]. NK cells are cytotoxic towards tumors and demonstrate anti-metastatic properties. Therefore, mutual interactions between TME and NK cells are complex and represent a promising therapeutic avenue for drug development [255]. Tumor cells develop characteristics which allow them to circumvent NK cells, and escape NK-based cytotoxicity. The process is facilitated by chronic stress (hypoxia or ROS) which forces the TME and NK cells to adjust their antitumor functions [256]. The modified TME is immunosuppressive and limits NK cell activity, thus, stimulating tumor progression and spread. NK-mediated resistance was correlated to mutations in DRs/TRAILRs [257]. Anti-apoptotic sphingolipids may contribute this process via their interactions with TNF signaling.

The list of major regulators of TME/NK responses and biological activities includes the TNF network (Fig.7). For instance, TRAF2 was shown to regulate NK responses [258]. TRAF2 is an adapter protein with E3 ligase properties which binds and activates various signaling molecules, such as membrane-bound receptors, kinases, and phosphatases [242]. TRAF2 can engage E3 ligases, including cIAP1/IAP2, and enable ubiquitination of Complex I components [259]. TRAF2 can be recruited to most proteins in the TNF receptor superfamily and transmits signals to the IKK complex and further to the NF-κB pathway [259]. Recent investigation defined the key role of the cold shock protein Y-box binding protein-1 (YB-1) in the regulation of pro-survival NF-κB p65 signaling by TNF-α via TRAF2. Higher expression of YB-1 was associated with adenocarcinoma invasiveness and expression of CerS6, which regulates cell migration [260]. However, the lower expression of CerS6 was found responsible for enhanced inflammation in a mouse colitis model [261], indicating a diverse role of sphingolipid metabolizing enzymes in the progression of pre-cancerous (pro-inflammatory) and cancerous conditions. As a mediator of TNF-α signaling, TRAF2 has been considered as a potential therapeutic target in cancers. For instance, regulatory T cell (Treg) signaling was targeted by immunotherapeutic approaches which also inhibit TRAF2 [262]. The TNFR2/TRAF2 axis is responsible for co-stimulation of CD8+ T cells, which sensitize cancer cells to cytotoxic effects [41]. TNF-α, was shown to activate Tregs via TNFR2, thus promoting Treg expansion and potential anticancer immunity [11]. However, the role of TNFR2 remains controversial, as both TNFR2 antagonists and agonists have demonstrated anticancer effects [263].

Fig. 7
figure 7

The conceptual model for the regulation of immune T cell responses by the SphK/S1P/S1PR axis during cancer progression. Sphingolipids were shown to impact cancer cell recognition and killing by immune cells at different levels. Abbreviations: PD-1, programmed death-1; PD-L1, programmed death-1 (PD-1) ligand 1; ROS, reactive oxygen species; S1P, sphingosine-1-phosphate; S1P1/S1P3, sphingosine-1-phosphate receptor 1 and 3; SphK, sphingosine kinase; WNT, wingless-related integration site

S1P was demonstrated to bind to TRAF2 as a cofactor, changing its E3 ligase biological activity [67, 264]. Blockade of the SphK1/S1P axis resulted in recovery of death-related effects provoked by DR5 knockdown [264]. Generation of S1P was found to be an essential step for TRAF2 polyubiquitination (stabilization), and subsequent promotion of cell invasion [264]. Therefore, TRAF2 is a putative SphK/S1P target during the cancer immune evasion. Binding of S1P to TRAF2, suggestively independent of S1PRs, was associated with activation of ERK1/2 and pro-metastatic cell behavior [264], although this conclusion may require further analysis. Many studies indicated regulation of cell migration by S1P receptors [21, 185]. For instance, S1P receptor 1 (S1P1) transmits S1P effects in various immune cells and regulates egress of lymphocytes into the circulation from the spleen and lymph nodes (LNs) [265]. The binding of S1P (or S1P receptor modulators/ligands) to S1P receptors leads to the receptor internalization and, thus, decreased presence of the receptors on plasma membrane and potential unresponsiveness to future stimulation. The internalization and degradation of S1P receptors may lead to the utilization of intracellular S1P too, which is translated into reduced lymphocyte egress, less circulating lymphocytes, and inhibition of inflammation-linked tissue damage [193]. Accordingly, administration of S1P modulators, such as fingolimod, provoked T cell-targeting immunomodulatory effect indicated by fast decline of the number of blood-circulating T cells [266]. S1P1 also regulates migration of osteoclast precursor cells via Fas/Rac1/NF-κB [219].

Sphingolipid signaling was linked to TME modifications and resistance of cancer cells to NK cell-based elimination [267]. For instance, S1P-stimulated lung cancer-derived monocytes secreted TNF-α and IL-6 in S1P receptor 3 (S1P3)/mTOR/K-Ras-dependent manner, while NF-κB was not implicated [268]. The authors suggested that greater presence of S1P within the TME of lung cancer may orchestrate tumorigenic immune responses [268]. However, this statement requires experimental confirmation, as the exact mechanisms (specific S1P targets) of this effect remain unclear and/or controversial [269, 270]. Furthermore, in non-Hodgkin’s lymphoma, SphK1 silencing resulted in activation of NKs, associated with increased secretion of IL-2 and IFN-γ, which are downstream of the classical NF-κB pathway [271]. Among S1P targets in immune cells, S1P4 (expressed by majority of immune cells) was indicated as a major effector of sphingolipid-dependent effects in innate immunity and lymphocyte trafficking [272, 273]. Therefore, S1P4 signaling could be another potential target to prevent cancer immune evasion. Chemokine CCL2 production by resident macrophages was regulated by S1P4 and synergized with Toll-like receptor (TLR) signaling, indicating sphingolipid receptor involvement in innate immunity responses [274]. Another sphingolipid metabolizing enzyme, S1P lyase, was found to be responsible for suppressing tumorigenicity within the TME [275]. S1P lyase was purported to be a death-promoting enzyme which eliminates S1P and its survival promoting effects [276].

Several independent research groups reported that S1P generation/S1P receptor expression profile stimulates migration of macrophages [277, 278]. Migration toward S1P was found to be mediated by expression of S1P1/S1P3, while expression of S1P2 decreased migration [278]. Interestingly, SphK2 was defined as an anti-inflammatory protein in human macrophages [279], although the role of SphK2 in inflammation remains controversial [221, 280]. Despite a growing number of relevant publications, the physiological roles of secreted and/or intracellularly generated S1P and S1P receptor (subtype-specific) expression in the regulation of macrophage/NK cell migration and activity remain largely unclear. However, the reported S1P-induced activation of human/rodent macrophages by apoptotic cells in an S1P1-dependent manner [278] opens a perspective to use sphingolipids as regulators of chemoattraction in TMEs and potentially increase effectiveness of NK cells. Accordingly, novel S1P receptor modulators and inhibitors require serious testing in vivo [281].

Decreased cytotoxicity of NK cells was associated with changes in chemoattraction and migration of myeloid-derived suppressor (MDS) cells towards tumor tissues [282]. MDS [283], Treg cells, and tumor-associated macrophages (TAMs) are common components of the TME, which can release immunosuppressive cytokines (such as TGFβ) and decrease NK cell-induced apoptosis [284]. In normal tissues, macrophages produce large amounts of TNF-α to clear bacterial and viral infections [285]. However, TAMs are unable to recognize cancer as a tissue destined for clearance, suggesting that the TNF signaling axis is reprogrammed in TAMs. Moreover, low endogenous concentrations of TNF-α derived from macrophages were found to promote metastasis via diverse downstream pro-survival mechanisms [48]. Therefore, TAMs represent a distinct phenotype of macrophages, making them a target for anticancer therapy. Further research is required to define which players within the TNF-α network may be responsible for the cancer-tolerating transformation of TAMs (often defined as de-differentiation) [286] and whether sphingolipids can be targeted in TAMs.

S1P1 and S1P3 represent other promising targets to dimmish cancer immune evasion. S1P1 receptor promoted Treg infiltration and tumor driven Treg expansion in bladder cancer [287]. S1P3 was shown to play a role in modulating the effects of TGFβ in cancer stem cells [288]. These effects were mediated by SphK1 and increased levels of S1P. Similar activation of SphK1 by TGFβ signaling was recently reported in A549 cells [289]. Other anti-apoptotic TME conditions contributed to the blunted immune response and cancer progression. For instance, hypoxia in the TME helps to reduce levels of pro-apoptotic Bax [290] and enhances levels of pro-survival proteins cIAP2 and Mcl-1 [291]. SphK/S1P receptors were shown to be involved in the regulation of this process. SphK2 promoted leukemia cell survival via Mcl-1 [292]. Mcl-1 upregulation was also mediated by S1P1 in mammary cancer cells [293]. Alternatively, fingolimod, an S1P receptor modulator, acted synergistically with TRAIL-induced apoptosis and downregulated Mcl-1 in various human cancer cells [294]. ONO-4641 (another S1P receptor modulator) stimulated the growth of CD11b + Gr-1 + (MDS) cells, decreased T cell proliferation, and lowered INF-γ secretion by CD3+ T cells (with similar characteristics to MDS cells) in the lungs of naïve mice, resulting in the lymphocytopenia [295]. In this mouse model of emphysema, the effect of ONO-4641 was desired [295], although to improve breast cancer immunotherapy depletion of MDS cells should be achieved [288].

The TME also contains non-immune cells (stroma) that promote downregulation of NK cell-mediated effects. Cancer associated fibroblasts (CAFs) are the major component of stroma [288] and confer documented inhibitory effects on NK-cytotoxicity [296, 297]. CAFs were shown to trigger NK cell exhaustion [298, 299] and secrete a range of immunosuppressive cytokines, including IL-10 [300], TGFβ [284], PGE2 [301], and indoleamine 2,3-dioxygenase (IDO) [302]. SphK2 was found to regulate CAF activation via interactions within the p53 network and facilitate the development of cancer tolerance of the TME [303]. Mesenchymal stem cells, which were also observed in stroma, can secrete PGE2/IDO and silence NK cell antitumor effects [301, 304]. S1P1 interaction with IL-22 receptor signaling was found to be involved in the promotion of metastasis to bone by mesenchymal stem cells [305]. Hypoxic conditions are linked to metastasis, inhibition of cancer growth in the initial phase, but promotion of cancer spreading at the later stages. The effects of hypoxia on the TNF-α signaling axis and its association with sphingolipid network are controversial and require further investigations [123, 306].

The controversy relies on the findings that hypoxia can enhance both secretion of pro-inflammatory cytokines (pro-apoptotic effect) and anti-apoptotic hypoxia-inducible factor-1α (HIF-1α) [307]. Hypoxia was also shown to promote resistance of cancer cells to NK cell cytotoxicity [157]. However, there are many network factors involved in the regulation of this process, including expression of HSP90 isoforms [307]. Hypoxia was shown to stimulate S1P generation in HepG2 cells [308] and in ovarian cancer cells [309]. In turn, sphingolipids may regulate hypoxia-related events at different levels. S1P/S1P1, as downstream effectors, mediated HIF-1α signaling during wound healing [212]. Downregulation of SphK1 expression reversed hypoxia-induced cell resistance to NK cell killing via blockade of the S1P/HIF-1α signaling arch [157]. Therefore, silencing or inhibition of SphK1 may be employed to strengthen NK effects. S1P signaling, as an upstream effector, also activated HIF-1α/HSP70 in normal rat pulmonary and cerebral cells [310]. Conclusively, HIF-1α activation by S1P was observed in various cells [311], thus, confirming the hindering effect of S1P in anti-cancer imunity.

The S1P/S1P1-3 axis was found to be involved in the regulation of glucose metabolism in mouse embryonic fibroblasts (S1P lyase knockdown model) via HIF-1α [312]. The S1P2 receptor was reported to be involved in preconditioning of macrophages towards a cancer-hospitable type in the TME [313]. Under hypoxia, a novel sphingosine metabolite O-cyclic phytosphingosine-1-phosphate suppressed mitochondrial dysfunction and apoptosis in mesenchymal stem cells via induction of HIF-1α signaling and calcium-dependent PKCα/ mTOR signaling pathway [247]. S1P modulator fingolimod inhibited HIF-1 and HIF-2 intratumoral levels and sensitized cancer cells to chemotherapy in vivo [314]. The crucial importance of HIF-1 in macrophages is associated with hypoxia-dependent regulation of macrophage interaction with cancer cells and angiogenic potential (interaction with endothelial tissues). Accordingly, strategies to prime macrophages towards anticancer toxicity, attract cytotoxic lymphocytes, and prevent angiogenesis/metastasis using specific inhibitors/modulators of sphingolipid axis (before or together with immune checkpoint inhibitors) could be beneficial.

Cyclooxygenase-2 (COX-2) and sphingolipids crosstalk

COX-2 is a key enzyme responsible for the production of PGE2, a multifunctional mediator of inflammation, and has been implicated in both inflammation and carcinogenesis. Crosstalk between COX-2 signaling and activation of the PI3K/Akt network has been established. It has been found that the COX-2/PGE2 axis can promote cancer cells survival via PI3K/Akt signaling [315] and Ras-MAPK cascades [316, 317]. Selective nonsteroidal anti-inflammatory drugs (NSAIDs) (such as celecoxib, valdecoxib, and rofecoxib) are widely used to control inflammation and cytokine production [318]. COX-2 is the most studied target of aspirin (the common anti-inflammatory agent), which has also demonstrated anticancer properties [319]. Different COX-2 inhibitors have been suggested as anticancer treatments [317, 320]. Selective COX-2 inhibitors NS-398 and nimesulide have been demonstrated to increase TNF-α sensitivity of TNF-α-resistant HeLa H21 cells [320]. Although nimesulide augmented TNF-α (CD95 or TRAIL receptors)-induced apoptosis, the interaction of TNF-α and COX-2 signaling pathway was not linked to the enzymatic activity of COX-2 [320], and so further analysis is required.

SphK/S1P axis may be involved in COX-2-mediated inflammation via orchestrated interactions with the TNF-α signaling pathway. The S1P receptor-based process seems to rely on both direct COX-2 activation and feedback mechanisms, as TNF-α and other cytokines can trigger SphK1, representing a loop of inflammation-enhancing interactions [213]. S1P-dependent activation of COX-2 was observed in a remarkable variety of normal and malignant cells and tissues, including endothelial [321], and various cancer cells [214, 322]. The SphK1/S1P receptor network also controls PGE2-mediated effects in various cells [81, 323, 324]. Notably, an aspirinyl-conjugated SphK inhibitor (SKI-I-Asp) containing aspirin to bolster oral bioavailability was generated and tested as a promising anticancer drug [325].

S1P effects on COX-2 expression and activity are mediated by its receptors. For instance, S1P stimulated expression of COX-2 and PGE2 production via S1P1 or S1P3 in human granulosa cells [326]. S1P3 antagonist blocked the LPS-dependent induction of COX-2 gene expression [209]. S1P2 mediated inflammation-related effects of S1P in renal cells. However, other enzymes were reported to mediate sphingolipid-induced activation of PGE2 synthesis. SphK1 knockdown decreased cytokine-induced PGE2 production via inhibition of microsomal PGE synthase-1 [322]. It is unclear whether DR expression/signaling is being altered during these effects. In conclusion, tripartite interactions between TNF-α/COX-2/sphingolipid network warrants future investigations.

DRs cross talk with PKC, STAT1, and SphK1/S1P3

Akt is a serine/threonine kinase which can be activated downstream of PI3K to provide a critical defense against apoptosis [327]. Activated Akt/PI3K can phosphorylate many mediators of DR signaling, including caspase-3 and caspase-9, Bad, MDM2, and different transcription factors [293, 328, 329]. In TNF-α-treated breast cancer cells, PKCε mediated anti-apoptotic effects via direct association with Akt [330,331,332]. Aside from anti-apoptotic ERK1/2 [98], the PKC/Akt axis also mediates sphingolipid effects [333]. For instance, PKC was found to be involved in S1P-mediated calcium fluxes and induction of insulin secretion in pancreatic β cells [334]. PKC has been reported to mediate the activation of endothelial cell migration and signaling [189, 335], and survival of malignant cells [336]. Akt activation by S1P, which mediated resistance to ischemia/reperfusion injury, was also reported in endothelial cells and cardiac myocytes [188, 337]. S1P3 was found to be involved in stabilization of Akt mRNA and stimulated Akt protein expression [338]. PKC/Akt may mediate poor response to immune checkpoint blockade therapy [339], and, thus, inhibition S1P axis may be beneficial in less responsive patients [340]. Interestingly, S1P2 was reported to mediate PKC inhibition [341].

Contrary to the S1P/S1P3 receptor network, ceramide and sphingosine may serve as negative regulators of PKC/PI3K/Akt signaling via several potential mechanisms [342,343,344]. Binding of PKCζ to 14-3-3 scaffolding proteins was found to be disrupted by ceramide, leading to PKCζ recruitment to lipid rafts [344]. Ceramide can also regulate Akt translocation to the plasma membrane and redirect (or block) its anti-apoptotic effects [343]. Ceramide-induced negative regulation of growth was marked by decreased ERK activity through PKCε-dependent effects [342]. PKCε was blocked by ceramide which prevented PKCε binding to Raf-1 and ERK in cells treated with insulin-like growth factor [342].

The PKC/Akt axis is a key regulator of autophagy [345] which can be activated by C2-ceramide in cancer cells [346,347,348]. SphK1 was also found to be activated in starved cells [349]. However, the role of SphK1 and S1P in the regulation of autophagy remains controversial and may be independent of Akt signaling [350]. Moreover, SphK1 activation during starvation may be a result of inducible cytoprotective mechanisms. This suggestion is supported by a study which indicated that SphK1 downregulation by siRNA enhanced starved cell death [350]. The involvement of the sphingolipid axis in the TNF/TRAIL-induced cell death may be also more complex than it was originally anticipated. However, the role of SphK/S1P axis in the regulation of autophagy in immune cells warrants further investigation, considering that autophagy is a promising target to overcome resistance to immunotherapy [351].

Stimulation of proliferation and anti-apoptotic effects of PKCε were mediated by a network which includes not only ERK1/2 and PI3K/Akt, but also STAT1, STAT3, and NF-κB pathways [352]. The potential role of putative STAT1 sites in the regulation of PKCε transcriptional activities was tested in MCF-7 cells [353]. The study demonstrated involvement of STAT1 and Sp1 in the upregulation of PKCε in MCF-7 cells in vitro [353]. The interaction is also a two-way mechanism, as inhibition of classical PKC isoenzymes resulted in downregulation of STAT1 in macrophages [354]. STAT1 was shown to regulate mammary tumorigenesis via multiple effectors [45, 219, 300]. SphK1 was reported to suppress activation of STAT1 in both parental and breast CSC cultures [220]. Another recent study indicated that STAT1 may bind the promoter region of S1P1 receptor [355]. It remains to be discovered how the anti-apoptotic effects of Akt/PKC can be integrated with STAT1 and sphingolipid networks in cells resistant to TNF-α/TRAIL-induced apoptosis.

Exploring the role of natural dietary and plant-based compounds as regulators of inflammation and sphingolipid metabolism

Selective anti-inflammatory molecules, including natural plant compounds and dietary components, have been shown to impact activation of pro-apoptotic cytokine signaling, suggesting their potential as safe and efficacious options for drug-resistant tumors. For instance, sulforaphane (SFN), a dietary component of broccoli, is an effective antioxidant with anticancer and anti-inflammatory characteristics [356] that has been tested for its cancer chemo-preventive properties [357,358,359]. Cytoprotective effects of SFN were associated with induction of the Nrf2 signaling pathway [357]. Alternatively, SFN-induced downregulation of Nrf2 expression was linked to increased apoptosis and elevated ROS [360]. SFN reversed ceramide-mediated apoptosis [361] in mouse hepatocytes that resulted from a high-fat diet (HFD) via the Nrf2 pathway [361]. Involvement of the Nrf2 pathway was also observed during application of siponimod (BAF312), a selective modulator of S1P1 and S1P5 receptors [362], supporting the existence of connections between Nrf2 and the sphingolipid signaling network. Siponemod demonstrated anti-inflammatory properties and microglia-protecting effects in the brain [363]. These effects provide insight into the regulation of S1P receptor signaling during inflammation, although the immune re-activating effects of these agents remains to be tested.

Natural flavonoids can regulate redox-sensitive pathways and transcription factors (such as Nrf2 and NF-κB) associated with increased release of free radicals/ROS and chronic inflammation [358, 364]. Many natural compounds were also found to target TNF-α/NF-κB and DR5 expression/pathway in cancer cells [365]. However, the effect of natural compounds on sphingolipid and TNF signaling networks during cancer immune evasion remains largely unclear. Only some of plant-derived and dietary compounds were tested and reported to influence sphingolipid metabolism and/or TNF network activity. One of the most studied agents, apigenin (4’,5,6-trihydroxyflavone), an anti-inflammatory compound isolated from parsley, oranges, and other plants, demonstrated strong anticancer properties via regulation of TNF-α, and DR4/DR5 pathways [35, 366, 367]. In conjunction with TNF-α, apigenin was shown to stimulate apoptosis and effectively decreased the survival of colon cancer cells [367]. In HepG2 cells, apigenin stimulated apoptosis via activation of pro-apoptotic TNF-α signaling [368]. Sensitization to Apo2L/TRAIL-induced apoptosis was also reported in prostate [369], HepG2 [370], Huh-7 (HCC) [371], and lung cancer cells [372] treated with apigenin. This dietary compound induces NF-κB activation [373]. Upregulation of TNF-α synthesis by apigenin was observed in J774.2 macrophages [374]. Notably, apigenin was also shown to inhibit SphK1/S1P axis in cardiac cells during endotoxemic shock [136]. However, in breast cancer cells, a dual effect of apigenin fostered some doubts about clinical application of this agent. Low doses of apigenin stimulated cancer cell growth, while high doses activated apoptosis via the TNF-α pathway [375]. Controversial findings were also reported in RAW264.7 macrophages where apigenin inhibited the effects of TNF-α [376]. Accordingly, detailed investigation is warranted to confirm the anticancer and SphK1/S1P-inbiting effects of apigenin in resistant tumors.

Other promising anti-inflammatory and SphK1-inhibiting agents (phenols and polyphenols) capable of sensitizing cancer cells to the pro-apoptotic effects of TNF-α (and/or stimulate pro-apoptotic effects of TRAIL/DR signaling) include the flavonoid epigallocatechin gallate (EGCG) [377] and the polyphenol resveratrol [378,379,380]. Protective effects of EGCG gavage were associated with increased levels of immune-enhancing substances. The agent also helped to balance regulation of the serum levels of sphingomyelin and sphingomyelin in the LPS-induced acute injury models, leading to reduced effects of harmful substances and inflammation [381]. Resveratrol was shown to impact sphingolipid metabolism in lung adenocarcinoma cells and downregulate inflammation via SphK1 inhibition [101, 380]. Another flavonoid, quercetin, also demonstrated antioxidant properties and reduced the production of S1P in HepG2 cells [382]. However, further testing is required to determine the immunomodulatory effects of dietary compounds in patients with resistant cancers.

Future perspectives of TNF-α/TRAIL therapy and clinical application of agents targeting the sphingolipid pathway

Major immunotherapies aim to increase the amount of tumor antigen-specific effector T cells in the circulation, block immunosuppressive effects of the TME [48], and stimulate cancer cell-targeted inflammation. The decision to initiate immunotherapy should be made on a per-patient basis according to the expression of predictive biomarkers (“immune response” gene signature). Several recent clinical trials have tested recombinant human TRAIL (rhTRAIL) and TRAIL receptor agonists (TRAs) against TRAIL-R1 and TRAIL-R2 [2]. DRs/TNFR1 have been the target of monoclonal antibodies (mAbs) in clinical trials over the last decade with variable levels of success [47]. Recent trials indicate high mAb specificity, longer half-life, and fewer adverse effects compared to conventional treatment [383]. TNF-α-containing fusion proteins have been designed and show effective anticancer properties [384]. However, only gene therapy with VB-111 (ofranergene obadenovec) yielded significantly improved progression-free survival in one trial [385], while another failed to confirm its efficacy in combination with bevacizumab (phase III study: NTC02511405) [386]. VB-111 was constructed using a replication-defective adenovirus serotype 5 vector attached to a modified murine pre-proendothelin promoter (PPE-1) and human Fas-chimera transgene [387]. Current data indicates that sphingolipids contribute to the development of cancer resistance to both immune surveillance and TNF/TRAIL-induced apoptosis, representing a promising target for future clinical strategies. The addition of sphingolipid modulators may increase the efficacy of this treatment, although this hypothesis is yet to be tested.

Several decades ago, the glycolytic pathway was suggested as a clinical target to sensitize tumor cells to soluble death ligands [52]. Glucose deprivation or inhibition of glucose metabolism enhanced apoptosis induced by TNF-α, CD95 agonistic antibody, and TRAIL in myeloid leukemia U937, cervical carcinoma HeLa, and breast carcinoma MCF-7 cells [52]. The effect was also observed in the human B-lymphoblastoid cell line SKW6.4, a prototype line for mitochondria-independent DR-induced apoptosis. Changes in c-FLIP(L) and cFLIPs levels were observed in some but not all studies cell lines under glucose deprivation [52, 388]. The changes were associated with activation of mitochondrial metabolism [388, 389]. Recent findings indicate a key role of sphingolipids in the regulation of cancer metabolism and anticancer immune responses [20, 390]. Dramatic changes in sphingolipid composition and processing were reported in cancer tissues [391]. Considering the involvement of sphingolipid network in TNF-α/TRAIL-activated signaling, it is reasonable to test SphK1/S1P receptor axis modulators as substances capable of strengthening anticancer therapy and increasing overall survival.

The delivery of TME-stimulating agents and reprogramming of the TME can be facilitated by nanocarriers [222, 392]. It has been shown that localized delivery of a nanoparticle-conjugated TLR7/8 agonist triggered lymph node-located DCs activation and promoted proliferation of tumor antigen-specific CD8+ T cells [392]. Cancer cell-targeted delivery of complex death-enhancing agents has demonstrated promising preclinical results. TRAIL-anchored artificial liposomes (defined as large unilamellar vesicle (LUV)) were constructed and loaded with DOX (named as LUVDOX-TRAIL). The liposome nanoparticle permitted synergistic cytotoxic potential compared to the effects of DOX or TRAIL alone. LUVDOX-TRAIL cytotoxicity was associated with faster internalization of the DOX-loaded liposomes and TRAIL-induced activation of caspase-8 [393]. Manipulation of tumor ceramide (and/or ceramide-conjugate substance) levels was explored as a potential strategy against drug resistant breast cancers [394, 395]. Some original studies have utilized the structurally modified analogs of the sphingoid backbone of d-erythro-N-octanoyl-sphingosine (Cer). The most potent anti-proliferative analog (2S,3R)-(4E,6E)-2-octanoylamidooctadecadiene-1,3-diol (4,6-diene-Cer) induced apoptosis in TNF-α-resistant MCF-7 cells, MDA-MB-231, and NCI/ADR-RES breast cancer cell lines [395]. Detected death-related mechanisms of 4,6-diene-Cer included a prolonged elevation of intracellular Cer and were mediated by the mitochondrial apoptotic pathway. Moreover, the valuable clinical characteristics of 4,6-diene-Cer include selectivity toward transformed breast cells [395]. Although the original substances turned out to be quite toxic in vivo, the search for less toxic substances continues. It has been found that 3-ketone-4,6-diene ceramide efficiently kills chemo-resistant breast cancer cells [396]. Recently, new ceramides with anticancer properties were extracted from red algae of the Red Sea [397].

Several novel methods were designed to deliver TNF-α locally as part of intratumoral vaccination [398]. The efficient nonviral gene therapy was developed to provide localized transfer of multiple genes into tumors in vivo. Gene electrotransfer (GET) was named as the most efficient method of local delivery of toxic cytokines. For instance, TNF-α and IL-12 (both can boost the primed local immune response) genes were transferred in murine melanoma cancers using GET [399]. The transfer was followed by a pronounced delay in tumor growth associated with strong antitumor immune response with extensive infiltration of immune cells in the tumor site [400]. Notably, GET was accompanied by resistance of the mice to secondary challenge with tumor cells [399]. Furthermore, phage and yeast display (bacteriophage strategy) were used for a pre-selection of non-neutralizing antibodies which were used to “piggyback” on TNF-α and enter cells through binding TNFRs [401]. This approach successfully reshaped the TME towards recruitment of antitumor immune cells (such as N1 neutrophils, M1 macrophages, and activated CD4+/CD8+ T cells) [401]. Combined testing of this bacteriophage technology with SphK1/S1P-targeting agents warrants future investigation.

Several conserved TNF-derived peptides can trigger apoptosis and/or necrosis in tumor cells [402] independent of TNFRs. Some of the necrosis-inducing TNF-derived peptides (like P1516) with strong membrane-disrupting characteristics may be released during TNF degradation [402]. The peptide’s cytolytic property was linked to its unique β-barrel/β-hairpin secondary structure [3]. Immunohistochemical analysis of tumor tissues from P1516-treated mice indicated extensive destruction to the cancer vasculature [402], which was associated with lower metastasis and better survival. The study indicated that TNF sequence contains cryptic functions that are triggered only after TNF partial and or specific degradation. This finding opens a previously unexplored perspective of TNF biology relevant to immune regulation and cancer immune surveillance. TNF-derived peptides P15 and P16 were suggested as a novel class of antitumor agents [402].

Tumor-specific cytotoxic T lymphocytes (CTLs) represent a natural and highly effective tool in cancer immunotherapy [4]. Considering the immunoregulatory role of sphingolipids (specifically the SphK1/S1P receptor axis), agents targeting sphingolipids may be employed to manipulate CTLs. However, only very few agents targeting this pathway have been approved for anticancer clinical testing. The approved agents include fingolimod (an S1P receptor antagonist; Phase I; NCT02490930 and NCT03941743); Safingol (L-threo-dihydrosphingosine; a PKC inhibitor; Phase 1; NCT01553071 and NCT00084812); sonepcizumab (ASONEP; an S1P-specific monoclonal antibody; Phase I/II; NCT00661414 and NCT01762033); ABC294640 (an SphK2 inhibitor; Phase I/II; NCT01488513, NCT02229981, NCT02757326, NCT02939807, NCT03377179, and NCT03414489). One study is currently recruiting to assess sphingolipids as predictive biomarkers in melanoma (NCT03627026). However, the potential testing will be on the way when new SphK1/S1PR modulators/inhibitors are generated and evaluated in preclinical settings. As a promising sign, the synergistic effects of DOX and SphK1 inhibition were reported in breast cancer cells [220].

A novel and less toxic strategy for advanced T cell infiltration in cancers has been suggested recently. A fusion protein Cys–Asn–Gly–Arg–Cys–Gly–TNF (called NGR-TNF) capable of targeting the cancer vasculature was constructed by Elia et al. [403] to assist intratumor infiltration by activated CTLs. It has been reported that, in a transgenic prostate adenocarcinoma mice model, combined treatment with NGR-TNF (with adoptive T cell therapy (ACT) and immune checkpoint blockade) effectively improved overall survival and delayed the disease progression. NGR-TNF promoted tumor infiltration by CTLs associated with beneficial T-effector/Treg cell ratios [403]. The authors of this study suggest that therapeutic targeting of sphingolipid pathway may contribute to this process.

Conclusions

Despite all the therapeutic impediments of TNF-α/TRAIL application, the cytotoxic cytokines remain the strongest natural defense to cancer in humans. TNF-α to be a prominent effector of immune surveillance which can kill mutated or abnormal cells, including cancer cells, under physiologic conditions [37]. Thus, to improve current therapeutic methods, it would be beneficial to preserve the pro-apoptotic capacity of TNF-α and block only its pro-survival branch. Notably, the cancer-promoting chronic inflammation which contributes oncogenic transformation, underscores a need to decipher the DR pathway and design agents that will block TNF/TRAIL/DR pro-survival signaling [401]. A wide range of substances and therapeutic methods has been developed to enhance immunotherapy effects in cancer patients [394, 396,397,398,399, 401,402,403,404], although combined application of sphingolipid-targeting agents and TNF-α pathway activating methods seems neglected. Apparently, SphK/S1P/S1PR axis plays an important role in transduction of TNF-α effects, both as a mediator and regulator of the cytokine signaling (Table 1). The generated anticancer agents which can selectively inhibit the growth-promoting effects of SphK (including dual SphK isozyme inhibitor, SKI-II (4-[4-(4-chloro-phenyl)-thiazol-2-ylamino]-phenol) represent a class of promising therapeutic substances [119, 405]. However, the production of agents that target the proper SphK isoform in cancer cells is challenging, although a large group of patented agents has been synthesized [406].

Nanoparticles represent a very promising approach for the targeted delivery of immunotherapy agents. Among the cutting-edge nanomedicine vehicles is a group of artificial liposomes with anchored sTRAIL, called LUV-TRAIL, which also improved delivery and reduced toxicity of immunotherapy [393]. Recent studies have tested the delivery and anticancer effects of TNF-α-loaded liposomes [404] or plant viral nanoparticles [407], TRAIL/paclitaxel multifunctional nanocarrier, graphene-based nanocarrier with DR4-targeting antibody/AKT siRNA, and anti-DR5-conjugated lipid-based nanocarriers [408]. Other nanoparticle-based agents displayed efficient pro-apoptotic properties via interactions with DR-signaling, including CD95 receptors [409] and the TRAIL network [400]. However, most of these studies were conducted in vitro, indicating a need for additional in vivo experiments before clinical testing may be considered. The addition of sphingolipid modulators to this regimen, specifically novel inhibitors of S1P1-S1P3 receptors, may augment the efficacy of nanoparticles in future studies. The success of personalized immunotherapy towards the re-activation and/or reformation of natural anti-cancer immunity may be defined by the de-activation of SphK/S1P/S1PR axis using novel inhibitors of sphingolipid pathway.

Availability of data and materials

Not applicable.

Abbreviations

4,6-diene-Cer:

(2S,3R)-(4E,6E)-2-octanoylamidooctadecadiene-1,3-diol

ACE:

Angiotensin converting enzyme

Apaf-1:

Apoptosis protease activating factor-1

ATF4:

Protein kinase R-like endoplasmic reticulum kinase-mediated activating transcription factor 4

Bak:

Bcl-2 antagonist killer 1

Bax:

Bcl2 associated X protein

Bcl-2:

B-cell lymphoma-2 protein

BMM:

Bone marrow-derived monocyte/macrophage

C1P:

Ceramide-1-phosphate

CAFs:

Cancer associated fibroblasts

CDase:

Ceramidase

Cer:

D-erythro-N-octanoyl-sphingosine

CERK:

Ceramide kinase

CerS:

Ceramide synthase

CERT:

Ceramide transport protein

CFTR:

Cystic fibrosis transmembrane conductance regulator

CGA:

Chlorogenic acid

cIAP:

Cellular inhibitor of apoptosis protein

COX-2:

Cyclooxygenase-2

CRDs:

Cysteine-rich domains

CREB:

Cyclic-AMP responsive element binding protein

CSC:

Cancer stem-cell

CTLs:

Cytotoxic T lymphocyte

cyt c :

Cytochrome c

DcR:

Decoy receptor

DD:

Death domain

DED:

Death effector domain

DENV:

Dengue virus

DHS:

Dihydroxysphingosine

DIABLO:

Direct Inhibitor of Apoptosis-Binding protein with Low pI

DMS:

N,N-dimethylsphingosine

DUB:

Dual deubiquitylating enzyme

EGCG:

Epigallocatechin gallate

EMT:

Epithelial-to-mesenchymal transition

eNOS:

Endothelial nitric oxide (NO) synthase

ER:

Estrogen receptor

ERK:

Extracellular-signal-regulated kinase

EZH2:

Enhancer of zeste homolog 2, a histone methyltransferase

FADD:

Fas-associated death domain

FLICE:

FADD-like IL-1β-converting enzyme

FLIP:

FLICE inhibitory protein

fMLP:

N-Formylmethionyl-leucyl-phenylalanine

GET:

Gene electrotransfer

GFAP:

Glial fibrillary acidic protein

HCC:

Hepatocellular carcinoma

HDAC:

Histone deacetylase

HFD:

High-fat diet

HIF-1α:

Hypoxia-inducible factor-1α

HMVEC-C:

Human cardiac microvascular endothelial cells

HSP:

Heat shock protein

ICIs:

Immune checkpoint inhibitors

IDO:

Indoleamine 2,3-dioxygenase

IFN-γ:

Interferon-γ

IGF:

Insulin-like growth factor

IKK:

I-kappa-B kinase

iNOS:

Inducible nitric oxide synthase

IRE1α:

Inositol-requiring transmembrane kinase/endoribonuclease 1α

JAK2:

Janus kinase 2

JIP3:

JNK-interacting protein 3

JNK:

C-Jun N-terminal kinase

LAMP-2:

Lysosomal associated membrane protein-2

LEPR:

Leptin receptor

LPS:

Lipopolysaccharide

LUV:

Large unilamellar vesicle

mAbs:

Monoclonal antibodies

MAPK:

Mitogen-activated protein kinase

MDS:

Myeloid-derived suppressor

MFBD:

Myristate-enriched milk fat-based diet

MMP:

Matrix metalloproteinase

MnSOD:

Mitochondrial superoxide dismutase

mTNF-α:

Transmembrane TNF-α

mTOR:

Akt/mammalian target of rapamycin

mtROS:

Mitochondrial reactive oxygen species

NADPH:

Nicotinamide adenine dinucleotide phosphate hydrogen

NF-κB:

Nuclear factor kappa B

NGR-TNF:

Cys–Asn–Gly–Arg–Cys–Gly–TNF fusion protein

NK:

Natural killer T cells

NSAIDs:

Nonsteroidal anti-inflammatory drugs

OPG:

Osteoprotegerin

OPGL:

Osteoprotegerin ligand

OXPHOS:

Oxidative phosphorylation

PGE2:

Prostaglandin E2

PI3K:

Phosphatidylinositol 3-kinase

PPE-1:

Pre-proendothelin promoter

RANTES:

Regulated upon activation, normal T cell expressed and secreted (CCL5)

rhTRAIL:

Recombinant human TRAIL

RIP:

Receptor interacting protein

RIPK1:

Receptor-interacting protein kinase 1

RTKs:

Receptor tyrosine kinases

S1P:

Sphingosine-1-phosphate

S1P1:

S1P3, sphingosine 1-phosphate receptor 1 and 3

S1PR:

S1P receptor

SFAs:

Saturated fatty acids

SFN:

Sulforaphane

siRNA:

Small interfering ribonucleic acid

SKI-I-Asp:

Aspirinyl-conjugated SphK inhibitor

SKI-II:

4-[4-(4-Chloro-phenyl)-thiazol-2-ylamino]-phenol

Smac:

Second mitochondria-derived activator of caspase

SOCS3:

Suppressor of cytokine signaling 3

SphK:

Sphingosine kinase

SPL:

S1P lyase

SPPase1:

Sphingosine-1-phosphate (S1P) phosphatase 1

SPT:

Serine palmitoyltransferase

STAT:

Signal transducer and activator of transcription

TAM:

Tumor-associated macrophage

TGFβ:

Transforming growth factor β

TL1A:

TNF-like cytokine 1A

TME:

Tumor microenvironment

TNFAIP3:

TNF-α-induced protein 3

TNFR:

TNF-α receptor

TNF-α:

Tumor necrosis factor-α

TRADD:

TNFR-associated death domain

TRAF2:

TNF receptor-associated factor 2

TRAIL:

TNF-related apoptosis-inducing ligand

Treg:

Regulatory T cell

VCAM-1:

Vascular cell adhesion molecule-1

VEGF:

Vascular endothelial growth factor

YB-1:

Y-box binding protein-1

δTE:

δ-Tocotrienol

References

  1. Kearney CJ, Vervoort SJ, Hogg SJ, Ramsbottom KM, Freeman AJ, Lalaoui N, Pijpers L, Michie J, Brown KK, Knight DA, Sutton V, Beavis PA, Voskoboinik I, Darcy PK, Silke J, Trapani JA, Johnstone RW, Oliaro J. Tumor immune evasion arises through loss of TNF sensitivity. Sci Immunol. 2018;3(23):eaar3451.

    Article  PubMed  Google Scholar 

  2. Josephs SF, Ichim TE, Prince SM, Kesari S, Marincola FM, Escobedo AR, Jafri A. Unleashing endogenous TNF-alpha as a cancer immunotherapeutic. J Transl Med. 2018;16(1):242.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Wajant H. CD95L/FasL and TRAIL in tumour surveillance and cancer therapy. Cancer Treat Res. 2006;130:141–65.

    Article  CAS  PubMed  Google Scholar 

  4. El Baba R, Herbein G. Immune landscape of CMV infection in cancer patients: from “canonical” diseases toward virus-elicited oncomodulation. Front Immunol. 2021;12:730765.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Parameswaran N, Patial S. Tumor necrosis factor-alpha signaling in macrophages. Crit Rev Eukaryot Gene Expr. 2010;20(2):87–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ham B, Fernandez MC, D’Costa Z, Brodt P. The diverse roles of the TNF axis in cancer progression and metastasis. Trends Cancer Res. 2016;11(1):1–27.

    PubMed  PubMed Central  Google Scholar 

  7. Popper HH. Manipulation of the immune system by non-small cell lung cancer and possible therapeutic interference. Cancer Drug Resist. 2020;3(4):710–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Poehlein CH, Hu HM, Yamada J, Assmann I, Alvord WG, Urba WJ, Fox BA. TNF plays an essential role in tumor regression after adoptive transfer of perforin/IFN-gamma double knockout effector T cells. J Immunol. 2003;170(4):2004–13.

    Article  CAS  PubMed  Google Scholar 

  9. Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci U S A. 1975;72(9):3666–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Guicciardi ME, Gores GJ. Life and death by death receptors. FASEB J. 2009;23(6):1625–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Salomon BL, Leclerc M, Tosello J, Ronin E, Piaggio E, Cohen JL. Tumor necrosis factor alpha and regulatory T cells in oncoimmunology. Front Immunol. 2018;9:444.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Lejeune FJ, Lienard D, Matter M, Ruegg C. Efficiency of recombinant human TNF in human cancer therapy. Cancer Immun. 2006;6:6.

    PubMed  Google Scholar 

  13. Xia P, Gamble JR, Rye KA, Wang L, Hii CS, Cockerill P, Khew-Goodall Y, Bert AG, Barter PJ, Vadas MA. Tumor necrosis factor-alpha induces adhesion molecule expression through the sphingosine kinase pathway. Proc Natl Acad Sci U S A. 1998;95(24):14196–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Balkwill F. Tumour necrosis factor and cancer. Nat Rev Cancer. 2009;9(5):361–71.

    Article  CAS  PubMed  Google Scholar 

  15. Yang H, Qiu B, Chen S, Xun Y, Pan Y, Chen M, Li WX, Liao W, El-Ashram S, Yang A, Liu F. Soluble CXCL16 promotes TNF-alpha-induced apoptosis in DLBCL via the AMAD10-NF-kappaB regulatory feedback loop. Cell Biol Int. 2019;43(8):863–74.

    Article  CAS  PubMed  Google Scholar 

  16. Mohr A, Buneker C, Gough RP, Zwacka RM. MnSOD protects colorectal cancer cells from TRAIL-induced apoptosis by inhibition of Smac/DIABLO release. Oncogene. 2008;27(6):763–74.

    Article  CAS  PubMed  Google Scholar 

  17. Tomiyama A, Serizawa S, Tachibana K, Sakurada K, Samejima H, Kuchino Y, Kitanaka C. Critical role for mitochondrial oxidative phosphorylation in the activation of tumor suppressors Bax and Bak. J Natl Cancer Inst. 2006;98(20):1462–73.

    Article  CAS  PubMed  Google Scholar 

  18. Neophytou CM, Trougakos IP, Erin N, Papageorgis P. Apoptosis deregulation and the development of cancer multi-drug resistance. Cancers (Basel). 2021;13(17):4363.

    Article  CAS  PubMed  Google Scholar 

  19. Legembre P, Barnhart BC, Zheng L, Vijayan S, Straus SE, Puck J, Dale JK, Lenardo M, Peter ME. Induction of apoptosis and activation of NF-kappaB by CD95 require different signalling thresholds. EMBO Rep. 2004;5(11):1084–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Giussani P, Prinetti A, Tringali C. The role of sphingolipids in cancer immunotherapy. Int J Mol Sci. 2021;22(12):6492.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sukocheva OA. Expansion of sphingosine kinase and sphingosine-1-phosphate receptor function in normal and cancer cells: from membrane restructuring to mediation of estrogen signaling and stem cell programming. Int J Mol Sci. 2018;19(2):420.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Huang CC, Tseng TT, Liu SC, Lin YY, Law YY, Hu SL, Wang SW, Tsai CH, Tang CH. S1P increases VEGF production in osteoblasts and facilitates endothelial progenitor cell angiogenesis by inhibiting miR-16-5p expression via the c-Src/FAK signaling pathway in rheumatoid arthritis. Cells. 2021;10(8):2168.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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(1):107–20.

    Article  CAS  PubMed  Google Scholar 

  24. Aoki M, Aoki H, Ramanathan R, Hait NC, Takabe K. Sphingosine-1-phosphate signaling in immune cells and inflammation: roles and therapeutic potential. Mediators Inflamm. 2016;2016:8606878.

    PubMed  PubMed Central  Google Scholar 

  25. Yuza K, Nakajima M, Nagahashi M, Tsuchida J, Hirose Y, Miura K, Tajima Y, Abe M, Sakimura K, Takabe K, Wakai T. Different roles of sphingosine kinase 1 and 2 in pancreatic cancer progression. J Surg Res. 2018;232:186–94.

    Article  CAS  PubMed  Google Scholar 

  26. Diaz Arguello OA, Haisma HJ. Apoptosis-inducing TNF superfamily ligands for cancer therapy. Cancers (Basel). 2021;13(7):1543.

    Article  PubMed  Google Scholar 

  27. Brenner D, Blaser H, Mak TW. Regulation of tumour necrosis factor signalling: live or let die. Nat Rev Immunol. 2015;15(6):362–74.

    Article  CAS  PubMed  Google Scholar 

  28. Yi F, Frazzette N, Cruz AC, Klebanoff CA, Siegel RM. Beyond cell death: new functions for TNF family cytokines in autoimmunity and tumor immunotherapy. Trends Mol Med. 2018;24(7):642–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Singh A, Ni J, Aggarwal BB. Death domain receptors and their role in cell demise. J Interferon Cytokine Res. 1998;18(7):439–50.

    Article  CAS  PubMed  Google Scholar 

  30. Wajant H, Siegmund D. TNFR1 and TNFR2 in the control of the life and death balance of macrophages. Front Cell Dev Biol. 2019;7:91.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Aggarwal BB. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol. 2003;3(9):745–56.

    Article  CAS  PubMed  Google Scholar 

  32. Varfolomeev E, Vucic D. Intracellular regulation of TNF activity in health and disease. Cytokine. 2018;101:26–32.

    Article  CAS  PubMed  Google Scholar 

  33. Ware CF. The TNF superfamily-2008. Cytokine Growth Factor Rev. 2008;19(3–4):183–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Micheau O, Tschopp J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 2003;114(2):181–90.

    Article  CAS  PubMed  Google Scholar 

  35. Chen M, Wang X, Zha D, Cai F, Zhang W, He Y, Huang Q, Zhuang H, Hua ZC. Apigenin potentiates TRAIL therapy of non-small cell lung cancer via upregulating DR4/DR5 expression in a p53-dependent manner. Sci Rep. 2016;6:35468.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mohr A, Yu R, Zwacka RM. TRAIL-receptor preferences in pancreatic cancer cells revisited: both TRAIL-R1 and TRAIL-R2 have a licence to kill. BMC Cancer. 2015;15:494.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Walczak H. Death receptor-ligand systems in cancer, cell death, and inflammation. Cold Spring Harb Perspect Biol. 2013;5(5):a008698.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Safa AR. c-FLIP, a master anti-apoptotic regulator. Exp Oncol. 2012;34(3):176–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Youle RJ, Strasser A. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008;9(1):47–59.

    Article  CAS  PubMed  Google Scholar 

  40. Borghi A, Verstrepen L, Beyaert R. TRAF2 multitasking in TNF receptor-induced signaling to NF-kappaB, MAP kinases and cell death. Biochem Pharmacol. 2016;116:1–10.

    Article  CAS  PubMed  Google Scholar 

  41. Moatti A, Cohen JL. The TNF-alpha/TNFR2 pathway: targeting a brake to release the anti-tumor immune response. Front Cell Dev Biol. 2021;9:725473.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Messmer UK, Pereda-Fernandez C, Manderscheid M, Pfeilschifter J. Dexamethasone inhibits TNF-alpha-induced apoptosis and IAP protein downregulation in MCF-7 cells. Br J Pharmacol. 2001;133(4):467–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Pei Y, Xing D, Gao X, Liu L, Chen T. Real-time monitoring full length bid interacting with Bax during TNF-alpha-induced apoptosis. Apoptosis. 2007;12(9):1681–90.

    Article  CAS  PubMed  Google Scholar 

  44. Blaser H, Dostert C, Mak TW, Brenner D. TNF and ROS crosstalk in inflammation. Trends Cell Biol. 2016;26(4):249–61.

    Article  CAS  PubMed  Google Scholar 

  45. Koromilas AE, Sexl V. The tumor suppressor function of STAT1 in breast cancer. JAKSTAT. 2013;2(2):e23353.

    PubMed  PubMed Central  Google Scholar 

  46. Medler J, Wajant H. Tumor necrosis factor receptor-2 (TNFR2): an overview of an emerging drug target. Expert Opin Ther Targets. 2019;23(4):295–307.

    Article  CAS  PubMed  Google Scholar 

  47. Cruceriu D, Baldasici O, Balacescu O, Berindan-Neagoe I. The dual role of tumor necrosis factor-alpha (TNF-alpha) in breast cancer: molecular insights and therapeutic approaches. Cell Oncol (Dordr). 2020;43(1):1–18.

    Article  CAS  PubMed  Google Scholar 

  48. Laha D, Grant R, Mishra P, Nilubol N. The role of tumor necrosis factor in manipulating the immunological response of tumor microenvironment. Front Immunol. 2021;12:656908.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Dash S, Sahu AK, Srivastava A, Chowdhury R, Mukherjee S. Exploring the extensive crosstalk between the antagonistic cytokines- TGF-beta and TNF-alpha in regulating cancer pathogenesis. Cytokine. 2021;138:155348.

    Article  CAS  PubMed  Google Scholar 

  50. Shin MS, Kim HS, Lee SH, Park WS, Kim SY, Park JY, Lee JH, Lee SK, Lee SN, Jung SS, Han JY, Kim H, Lee JY, Yoo NJ. Mutations of tumor necrosis factor-related apoptosis-inducing ligand receptor 1 (TRAIL-R1) and receptor 2 (TRAIL-R2) genes in metastatic breast cancers. Cancer Res. 2001;61(13):4942–6.

    CAS  PubMed  Google Scholar 

  51. Tuomela K, Ambrose AR, Davis DM. Escaping death: how cancer cells and infected cells resist cell-mediated cytotoxicity. Front Immunol. 2022;13:867098.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Munoz-Pinedo C, Ruiz-Ruiz C, Ruiz de Almodovar C, Palacios C, Lopez-Rivas A. Inhibition of glucose metabolism sensitizes tumor cells to death receptor-triggered apoptosis through enhancement of death-inducing signaling complex formation and apical procaspase-8 processing. J Biol Chem. 2003;278(15):12759–68.

    Article  CAS  PubMed  Google Scholar 

  53. Wang Y, Qi H, Liu Y, Duan C, Liu X, Xia T, Chen D, Piao HL, Liu HX. The double-edged roles of ROS in cancer prevention and therapy. Theranostics. 2021;11(10):4839–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sukocheva OA, Furuya H, Ng ML, Friedemann M, Menschikowski M, Tarasov VV, Chubarev VN, Klochkov SG, Neganova ME, Mangoni AA, Aliev G, Bishayee A. Sphingosine kinase and sphingosine-1-phosphate receptor signaling pathway in inflammatory gastrointestinal disease and cancers: a novel therapeutic target. Pharmacol Ther. 2020;207:107464.

    Article  CAS  PubMed  Google Scholar 

  55. Cuvillier O, Levade T. Sphingosine 1-phosphate antagonizes apoptosis of human leukemia cells by inhibiting release of cytochrome c and Smac/DIABLO from mitochondria. Blood. 2001;98(9):2828–36.

    Article  CAS  PubMed  Google Scholar 

  56. Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Coso OA, Gutkind S, Spiegel S. Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature. 1996;381(6585):800–3.

    Article  CAS  PubMed  Google Scholar 

  57. Ogretmen B. Sphingolipid metabolism in cancer signalling and therapy. Nat Rev Cancer. 2018;18(1):33–50.

    Article  CAS  PubMed  Google Scholar 

  58. Xia P, Wang L, Gamble JR, Vadas MA. Activation of sphingosine kinase by tumor necrosis factor-alpha inhibits apoptosis in human endothelial cells. J Biol Chem. 1999;274(48):34499–505.

    Article  CAS  PubMed  Google Scholar 

  59. Kozawa O, Tanabe K, Ito H, Matsuno H, Niwa M, Kato K, Uematsu T. Sphingosine 1-phosphate regulates heat shock protein 27 induction by a p38 MAP kinase-dependent mechanism in aortic smooth muscle cells. Exp Cell Res. 1999;250(2):376–80.

    Article  CAS  PubMed  Google Scholar 

  60. Kozawa O, Tokuda H, Matsuno H, Uematsu T. Sphingosine modulates interleukin-6 synthesis in osteoblasts. J Cell Biochem. 1998;70(3):338–45.

    Article  CAS  PubMed  Google Scholar 

  61. Prieschl EE, Csonga R, Novotny V, Kikuchi GE, Baumruker T. The balance between sphingosine and sphingosine-1-phosphate is decisive for mast cell activation after Fc epsilon receptor I triggering. J Exp Med. 1999;190(1):1–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Price MM, Oskeritzian CA, Falanga YT, Harikumar KB, Allegood JC, Alvarez SE, Conrad D, Ryan JJ, Milstien S, Spiegel S. A specific sphingosine kinase 1 inhibitor attenuates airway hyperresponsiveness and inflammation in a mast cell-dependent murine model of allergic asthma. J Allergy Clin Immunol. 2013;131(2):501-11.e1.

    Article  CAS  PubMed  Google Scholar 

  63. Niwa M, Kozawa O, Matsuno H, Kanamori Y, Hara A, Uematsu T. Tumor necrosis factor-alpha-mediated signal transduction in human neutrophils: involvement of sphingomyelin metabolites in the priming effect of TNF-alpha on the fMLP-stimulated superoxide production. Life Sci. 2000;66(3):245–56.

    Article  CAS  PubMed  Google Scholar 

  64. 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(16):4468–74.

    CAS  PubMed  Google Scholar 

  65. Osawa Y, Banno Y, Nagaki M, Brenner DA, Naiki T, Nozawa Y, Nakashima S, Moriwaki H. TNF-alpha-induced sphingosine 1-phosphate inhibits apoptosis through a phosphatidylinositol 3-kinase/Akt pathway in human hepatocytes. J Immunol. 2001;167(1):173–80.

    Article  CAS  PubMed  Google Scholar 

  66. Osawa Y, Hannun YA, Proia RL, Brenner DA. Roles of AKT and sphingosine kinase in the antiapoptotic effects of bile duct ligation in mouse liver. Hepatology. 2005;42(6):1320–8.

    Article  CAS  PubMed  Google Scholar 

  67. Hanna AN, Berthiaume LG, Kikuchi Y, Begg D, Bourgoin S, Brindley DN. Tumor necrosis factor-alpha induces stress fiber formation through ceramide production: role of sphingosine kinase. Mol Biol Cell. 2001;12(11):3618–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Manggau M, Kim DS, Ruwisch L, Vogler R, Korting HC, Schafer-Korting M, Kleuser B. 1Alpha,25-dihydroxyvitamin D3 protects human keratinocytes from apoptosis by the formation of sphingosine-1-phosphate. J Invest Dermatol. 2001;117(5):1241–9.

    Article  CAS  PubMed  Google Scholar 

  69. Hammer S, Sauer B, Spika I, Schraut C, Kleuser B, Schafer-Korting M. Glucocorticoids mediate differential anti-apoptotic effects in human fibroblasts and keratinocytes via sphingosine-1-phosphate formation. J Cell Biochem. 2004;91(4):840–51.

    Article  CAS  PubMed  Google Scholar 

  70. Vann LR, Payne SG, Edsall LC, Twitty S, Spiegel S, Milstien S. Involvement of sphingosine kinase in TNF-alpha-stimulated tetrahydrobiopterin biosynthesis in C6 glioma cells. J Biol Chem. 2002;277(15):12649–56.

    Article  CAS  PubMed  Google Scholar 

  71. Xia P, Wang L, Moretti PA, Albanese N, Chai F, Pitson SM, D’Andrea RJ, Gamble JR, Vadas MA. Sphingosine kinase interacts with TRAF2 and dissects tumor necrosis factor-alpha signaling. J Biol Chem. 2002;277(10):7996–8003.

    Article  CAS  PubMed  Google Scholar 

  72. Alvarez SE, Harikumar KB, Hait NC, Allegood J, Strub GM, Kim EY, Maceyka M, Jiang H, Luo C, Kordula T, Milstien S, Spiegel S. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature. 2010;465(7301):1084–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. MacKinnon AC, Buckley A, Chilvers ER, Rossi AG, Haslett C, Sethi T. Sphingosine kinase: a point of convergence in the action of diverse neutrophil priming agents. J Immunol. 2002;169(11):6394–400.

    Article  CAS  PubMed  Google Scholar 

  74. Johnson KR, Johnson KY, Becker KP, Bielawski J, Mao C, Obeid LM. Role of human sphingosine-1-phosphate phosphatase 1 in the regulation of intra- and extracellular sphingosine-1-phosphate levels and cell viability. J Biol Chem. 2003;278(36):34541–7.

    Article  CAS  PubMed  Google Scholar 

  75. Pettus BJ, Bielawski J, Porcelli AM, Reames DL, Johnson KR, Morrow J, Chalfant CE, Obeid LM, Hannun YA. The sphingosine kinase 1/sphingosine-1-phosphate pathway mediates COX-2 induction and PGE2 production in response to TNF-alpha. FASEB J. 2003;17(11):1411–21.

    Article  CAS  PubMed  Google Scholar 

  76. Sawada M, Kiyono T, Nakashima S, Shinoda J, Naganawa T, Hara S, Iwama T, Sakai N. Molecular mechanisms of TNF-alpha-induced ceramide formation in human glioma cells: P53-mediated oxidant stress-dependent and -independent pathways. Cell Death Differ. 2004;11(9):997–1008.

    Article  CAS  PubMed  Google Scholar 

  77. Chen XL, Grey JY, Thomas S, Qiu FH, Medford RM, Wasserman MA, Kunsch C. Sphingosine kinase-1 mediates TNF-alpha-induced MCP-1 gene expression in endothelial cells: upregulation by oscillatory flow. Am J Physiol Heart Circ Physiol. 2004;287(4):H1452–8.

    Article  CAS  PubMed  Google Scholar 

  78. Misasi R, Garofalo T, Di Marzio L, Mattei V, Gizzi C, Hiraiwa M, Pavan A, Grazia Cifone M, Sorice M. Prosaposin: a new player in cell death prevention of U937 monocytic cells. Exp Cell Res. 2004;298(1):38–47.

    Article  CAS  PubMed  Google Scholar 

  79. Vlasenko LP, Melendez AJ. A critical role for sphingosine kinase in anaphylatoxin-induced neutropenia, peritonitis, and cytokine production in vivo. J Immunol. 2005;174(10):6456–61.

    Article  CAS  PubMed  Google Scholar 

  80. Mastrandrea LD, Sessanna SM, Laychock SG. Sphingosine kinase activity and sphingosine-1 phosphate production in rat pancreatic islets and INS-1 cells: response to cytokines. Diabetes. 2005;54(5):1429–36.

    Article  CAS  PubMed  Google Scholar 

  81. Billich A, Bornancin F, Mechtcheriakova D, Natt F, Huesken D, Baumruker T. Basal and induced sphingosine kinase 1 activity in A549 carcinoma cells: function in cell survival and IL-1beta and TNF-alpha induced production of inflammatory mediators. Cell Signal. 2005;17(10):1203–17.

    Article  CAS  PubMed  Google Scholar 

  82. Yang J, Castle BE, Hanidu A, Stevens L, Yu Y, Li X, Stearns C, Papov V, Rajotte D, Li J. Sphingosine kinase 1 is a negative regulator of CD4+ Th1 cells. J Immunol. 2005;175(10):6580–8.

    Article  CAS  PubMed  Google Scholar 

  83. Taha TA, Kitatani K, Bielawski J, Cho W, Hannun YA, Obeid LM. Tumor necrosis factor induces the loss of sphingosine kinase-1 by a cathepsin B-dependent mechanism. J Biol Chem. 2005;280(17):17196–202.

    Article  CAS  PubMed  Google Scholar 

  84. Yadav M, Clark L, Schorey JS. Macrophage’s proinflammatory response to a mycobacterial infection is dependent on sphingosine kinase-mediated activation of phosphatidylinositol phospholipase C, protein kinase C, ERK1/2, and phosphatidylinositol 3-kinase. J Immunol. 2006;176(9):5494–503.

    Article  CAS  PubMed  Google Scholar 

  85. Prakash H, Luth A, Grinkina N, Holzer D, Wadgaonkar R, Gonzalez AP, Anes E, Kleuser B. Sphingosine kinase-1 (SphK-1) regulates Mycobacterium smegmatis infection in macrophages. PLoS One. 2010;5(5):e10657.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Kang JS, Yoon YD, Han MH, Han SB, Lee K, Lee KH, Park SK, Kim HM. Glabridin suppresses intercellular adhesion molecule-1 expression in tumor necrosis factor-alpha-stimulated human umbilical vein endothelial cells by blocking sphingosine kinase pathway: implications of Akt, extracellular signal-regulated kinase, and nuclear factor-kappaB/Rel signaling pathways. Mol Pharmacol. 2006;69(3):941–9.

    Article  CAS  PubMed  Google Scholar 

  87. Bu S, Yamanaka M, Pei H, Bielawska A, Bielawski J, Hannun YA, Obeid L, Trojanowska M. Dihydrosphingosine 1-phosphate stimulates MMP1 gene expression via activation of ERK1/2-Ets1 pathway in human fibroblasts. FASEB J. 2006;20(1):184–6.

    Article  CAS  PubMed  Google Scholar 

  88. De Palma C, Meacci E, Perrotta C, Bruni P, Clementi E. Endothelial nitric oxide synthase activation by tumor necrosis factor alpha through neutral sphingomyelinase 2, sphingosine kinase 1, and sphingosine 1 phosphate receptors: a novel pathway relevant to the pathophysiology of endothelium. Arterioscler Thromb Vasc Biol. 2006;26(1):99–105.

    Article  PubMed  Google Scholar 

  89. Maines LW, French KJ, Wolpert EB, Antonetti DA, Smith CD. Pharmacologic manipulation of sphingosine kinase in retinal endothelial cells: implications for angiogenic ocular diseases. Invest Ophthalmol Vis Sci. 2006;47(11):5022–31.

    Article  PubMed  Google Scholar 

  90. 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(4):997–1012.

    Article  CAS  PubMed  Google Scholar 

  91. Suzuki H, Riley RT, Sharma RP. Inducible nitric oxide has protective effect on fumonisin B1 hepatotoxicity in mice via modulation of sphingosine kinase. Toxicology. 2007;229(1–2):42–53.

    Article  CAS  PubMed  Google Scholar 

  92. Radeff-Huang J, Seasholtz TM, Chang JW, Smith JM, Walsh CT, Brown JH. Tumor necrosis factor-alpha-stimulated cell proliferation is mediated through sphingosine kinase-dependent Akt activation and cyclin D expression. J Biol Chem. 2007;282(2):863–70.

    Article  CAS  PubMed  Google Scholar 

  93. Hammad SM, Crellin HG, Wu BX, Melton J, Anelli V, Obeid LM. Dual and distinct roles for sphingosine kinase 1 and sphingosine 1 phosphate in the response to inflammatory stimuli in RAW macrophages. Prostaglandins Other Lipid Mediat. 2008;85(3–4):107–14.

    Article  CAS  PubMed  Google Scholar 

  94. Mechtcheriakova D, Wlachos A, Sobanov J, Kopp T, Reuschel R, Bornancin F, Cai R, Zemann B, Urtz N, Stingl G, Zlabinger G, Woisetschlager M, Baumruker T, Billich A. Sphingosine 1-phosphate phosphatase 2 is induced during inflammatory responses. Cell Signal. 2007;19(4):748–60.

    Article  CAS  PubMed  Google Scholar 

  95. Donati C, Nincheri P, Cencetti F, Rapizzi E, Farnararo M, Bruni P. Tumor necrosis factor-alpha exerts pro-myogenic action in C2C12 myoblasts via sphingosine kinase/S1P2 signaling. FEBS Lett. 2007;581(23):4384–8.

    Article  CAS  PubMed  Google Scholar 

  96. Clarke CJ, Truong TG, Hannun YA. Role for neutral sphingomyelinase-2 in tumor necrosis factor alpha-stimulated expression of vascular cell adhesion molecule-1 (VCAM) and intercellular adhesion molecule-1 (ICAM) in lung epithelial cells: p38 MAPK is an upstream regulator of nSMase2. J Biol Chem. 2007;282(2):1384–96.

    Article  CAS  PubMed  Google Scholar 

  97. Dai W, Yan J, Chen G, Hu G, Zhou X, Zeng X. AQP4-knockout alleviates the lipopolysaccharide-induced inflammatory response in astrocytes via SPHK1/MAPK/AKT signaling. Int J Mol Med. 2018;42(3):1716–22.

    CAS  PubMed  Google Scholar 

  98. Oskeritzian CA, Alvarez SE, Hait NC, Price MM, Milstien S, Spiegel S. Distinct roles of sphingosine kinases 1 and 2 in human mast-cell functions. Blood. 2008;111(8):4193–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Lai WQ, Irwan AW, Goh HH, Melendez AJ, McInnes IB, Leung BP. Distinct roles of sphingosine kinase 1 and 2 in murine collagen-induced arthritis. J Immunol. 2009;183(3):2097–103.

    Article  CAS  PubMed  Google Scholar 

  100. Wadgaonkar R, Patel V, Grinkina N, Romano C, Liu J, Zhao Y, Sammani S, Garcia JG, Natarajan V. Differential regulation of sphingosine kinases 1 and 2 in lung injury. Am J Physiol Lung Cell Mol Physiol. 2009;296(4):L603–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Issuree PD, Pushparaj PN, Pervaiz S, Melendez AJ. Resveratrol attenuates C5a-induced inflammatory responses in vitro and in vivo by inhibiting phospholipase D and sphingosine kinase activities. FASEB J. 2009;23(8):2412–24.

    Article  CAS  PubMed  Google Scholar 

  102. 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(1):143–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Limaye V, Xia P, Hahn C, Smith M, Vadas MA, Pitson SM, Gamble JR. Chronic increases in sphingosine kinase-1 activity induce a pro-inflammatory, pro-angiogenic phenotype in endothelial cells. Cell Mol Biol Lett. 2009;14(3):424–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Baker DA, Barth J, Chang R, Obeid LM, Gilkeson GS. Genetic sphingosine kinase 1 deficiency significantly decreases synovial inflammation and joint erosions in murine TNF-alpha-induced arthritis. J Immunol. 2010;185(4):2570–9.

    Article  CAS  PubMed  Google Scholar 

  105. Baker DA, Eudaly J, Smith CD, Obeid LM, Gilkeson GS. Impact of sphingosine kinase 2 deficiency on the development of TNF-alpha-induced inflammatory arthritis. Rheumatol Int. 2013;33(10):2677–81.

    Article  CAS  PubMed  Google Scholar 

  106. Nayak D, Huo Y, Kwang WX, Pushparaj PN, Kumar SD, Ling EA, Dheen ST. Sphingosine kinase 1 regulates the expression of proinflammatory cytokines and nitric oxide in activated microglia. Neuroscience. 2010;166(1):132–44.

    Article  CAS  PubMed  Google Scholar 

  107. Lan T, Bi H, Xu S, Le K, Xie Z, Liu Y, Huang H. Determination of sphingosine kinase activity in biological samples by liquid chromatography-tandem mass spectrometry. Biomed Chromatogr. 2010;24(10):1075–83.

    Article  CAS  PubMed  Google Scholar 

  108. Sun WY, Pitson SM, Bonder CS. Tumor necrosis factor-induced neutrophil adhesion occurs via sphingosine kinase-1-dependent activation of endothelial alpha5beta1 integrin. Am J Pathol. 2010;177(1):436–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Gurgui M, Broere R, Kalff JC, van Echten-Deckert G. Dual action of sphingosine 1-phosphate in eliciting proinflammatory responses in primary cultured rat intestinal smooth muscle cells. Cell Signal. 2010;22(11):1727–33.

    Article  CAS  PubMed  Google Scholar 

  110. Scherer EQ, Yang J, Canis M, Reimann K, Ivanov K, Diehl CD, Backx PH, Wier WG, Strieth S, Wangemann P, Voigtlaender-Bolz J, Lidington D, Bolz SS. Tumor necrosis factor-alpha enhances microvascular tone and reduces blood flow in the cochlea via enhanced sphingosine-1-phosphate signaling. Stroke. 2010;41(11):2618–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Yu HM, Li Q, Perelman JM, Kolosov VP, Zhou XD. Regulation of sphingosine kinase 1 in the TNF-alpha-induced expression of MUC5AC in airway epithelial cells. Zhonghua Yi Xue Za Zhi. 2011;91(6):391–5.

    CAS  PubMed  Google Scholar 

  112. Doyle T, Chen Z, Obeid LM, Salvemini D. Sphingosine-1-phosphate acting via the S1P(1) receptor is a downstream signaling pathway in ceramide-induced hyperalgesia. Neurosci Lett. 2011;499(1):4–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Wati S, Rawlinson SM, Ivanov RA, Dorstyn L, Beard MR, Jans DA, Pitson SM, Burrell CJ, Li P, Carr JM. Tumour necrosis factor alpha (TNF-alpha) stimulation of cells with established dengue virus type 2 infection induces cell death that is accompanied by a reduced ability of TNF-alpha to activate nuclear factor kappaB and reduced sphingosine kinase-1 activity. J Gen Virol. 2011;92(Pt 4):807–18.

    Article  CAS  PubMed  Google Scholar 

  114. Keul P, Lucke S, von Wnuck Lipinski K, Bode C, Graler M, Heusch G, Levkau B. Sphingosine-1-phosphate receptor 3 promotes recruitment of monocyte/macrophages in inflammation and atherosclerosis. Circ Res. 2011;108(3):314–23.

    Article  CAS  PubMed  Google Scholar 

  115. Awad AS, Rouse MD, Khutsishvili K, Huang L, Bolton WK, Lynch KR, Okusa MD. Chronic sphingosine 1-phosphate 1 receptor activation attenuates early-stage diabetic nephropathy independent of lymphocytes. Kidney Int. 2011;79(10):1090–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Sauve M, Hui SK, Dinh DD, Foltz WD, Momen A, Nedospasov SA, Offermanns S, Husain M, Kroetsch JT, Lidington D, Bolz SS. Tumor necrosis factor/sphingosine-1-phosphate signaling augments resistance artery myogenic tone in diabetes. Diabetes. 2016;65(7):1916–28.

    Article  CAS  PubMed  Google Scholar 

  117. Liu Q, Rehman H, Shi Y, Krishnasamy Y, Lemasters JJ, Smith CD, Zhong Z. Inhibition of sphingosine kinase-2 suppresses inflammation and attenuates graft injury after liver transplantation in rats. PLoS One. 2012;7(7):e41834.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Dakroub Z, Kreydiyyeh SI. Sphingosine-1-phosphate is a mediator of TNF-alpha action on the Na+/K+ ATPase in HepG2 cells. J Cell Biochem. 2012;113(6):2077–85.

    Article  CAS  PubMed  Google Scholar 

  119. Antoon JW, White MD, Burow ME, Beckman BS. Dual inhibition of sphingosine kinase isoforms ablates TNF-induced drug resistance. Oncol Rep. 2012;27(6):1779–86.

    CAS  PubMed  Google Scholar 

  120. Meissner A, Yang J, Kroetsch JT, Sauve M, Dax H, Momen A, Noyan-Ashraf MH, Heximer S, Husain M, Lidington D, Bolz SS. Tumor necrosis factor-alpha-mediated downregulation of the cystic fibrosis transmembrane conductance regulator drives pathological sphingosine-1-phosphate signaling in a mouse model of heart failure. Circulation. 2012;125(22):2739–50.

    Article  CAS  PubMed  Google Scholar 

  121. Yang J, Noyan-Ashraf MH, Meissner A, Voigtlaender-Bolz J, Kroetsch JT, Foltz W, Jaffray D, Kapoor A, Momen A, Heximer SP, Zhang H, van Eede M, Henkelman RM, Matthews SG, Lidington D, Husain M, Bolz SS. Proximal cerebral arteries develop myogenic responsiveness in heart failure via tumor necrosis factor-alpha-dependent activation of sphingosine-1-phosphate signaling. Circulation. 2012;126(2):196–206.

    Article  CAS  PubMed  Google Scholar 

  122. Wu X, Xu J, Li X, Dai J, Wang L. Inhibition of SphK1/S1P signaling pathway alleviates fibrosis and inflammation of rat myocardium after myocardial infarction. Comput Math Methods Med. 2022;2022:5985375.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Li Q, Wang C, Zhang Q, Tang C, Li N, Li J. The role of sphingosine kinase 1 in patients with severe acute pancreatitis. Ann Surg. 2012;255(5):954–62.

    Article  PubMed  Google Scholar 

  124. Barth BM, Gustafson SJ, Kuhn TB. Neutral sphingomyelinase activation precedes NADPH oxidase-dependent damage in neurons exposed to the proinflammatory cytokine tumor necrosis factor-alpha. J Neurosci Res. 2012;90(1):229–42.

    Article  CAS  PubMed  Google Scholar 

  125. Bakar AM, Park SW, Kim M, Lee HT. Isoflurane protects against human endothelial cell apoptosis by inducing sphingosine kinase-1 via ERK MAPK. Int J Mol Sci. 2012;13(1):977–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Altay O, Suzuki H, Hasegawa Y, Ostrowski RP, Tang J, Zhang JH. Isoflurane on brain inflammation. Neurobiol Dis. 2014;62:365–71.

    Article  CAS  PubMed  Google Scholar 

  127. Lufrano M, Jacob A, Zhou M, Wang P. Sphingosine kinase-1 mediates endotoxemia-induced hyperinflammation in aged animals. Mol Med Rep. 2013;8(2):645–9.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Adada MM, Orr-Gandy KA, Snider AJ, Canals D, Hannun YA, Obeid LM, Clarke CJ. Sphingosine kinase 1 regulates tumor necrosis factor-mediated RANTES induction through p38 mitogen-activated protein kinase but independently of nuclear factor kappaB activation. J Biol Chem. 2013;288(38):27667–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Zheng XD, Zhang Y, Qi XW, Wang MH, Sun P, Zhang Y, Jiang J. Role of Sphk1 in the malignant transformation of breast epithelial cells and breast cancer progression. Indian J Cancer. 2014;51(4):524–9.

    Article  CAS  PubMed  Google Scholar 

  130. Hamada Y, Nagasaki H, Fujiya A, Seino Y, Shang QL, Suzuki T, Hashimoto H, Oiso Y. Involvement of de novo ceramide synthesis in pro-inflammatory adipokine secretion and adipocyte-macrophage interaction. J Nutr Biochem. 2014;25(12):1309–16.

    Article  CAS  PubMed  Google Scholar 

  131. Oizumi A, Nakayama H, Okino N, Iwahara C, Kina K, Matsumoto R, Ogawa H, Takamori K, Ito M, Suga Y, Iwabuchi K. Pseudomonas-derived ceramidase induces production of inflammatory mediators from human keratinocytes via sphingosine-1-phosphate. PLoS One. 2014;9(2):e89402.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Moon E, Han JE, Jeon S, Ryu JH, Choi JW, Chun J. Exogenous S1P exposure potentiates ischemic stroke damage that is reduced possibly by inhibiting S1P receptor signaling. Mediators Inflamm. 2015;2015:492659.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Sun W, Ding Z, Xu S, Su Z, Li H. Crosstalk between TLR2 and Sphk1 in microglia in the cerebral ischemia/reperfusion-induced inflammatory response. Int J Mol Med. 2017;40(6):1750–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Lei YC, Yang LL, Li W, Luo P, Zheng PF. Inhibition of sphingosine kinase 1 ameliorates acute liver failure by reducing high-mobility group box 1 cytoplasmic translocation in liver cells. World J Gastroenterol. 2015;21(46):13055–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Farnoud AM, Bryan AM, Kechichian T, Luberto C, Del Poeta M. The granuloma response controlling cryptococcosis in mice depends on the sphingosine kinase 1-sphingosine 1-phosphate pathway. Infect Immun. 2015;83(7):2705–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Zhang T, Yan T, Du J, Wang S, Yang H. Apigenin attenuates heart injury in lipopolysaccharide-induced endotoxemic model by suppressing sphingosine kinase 1/sphingosine 1-phosphate signaling pathway. Chem Biol Interact. 2015;233:46–55.

    Article  CAS  PubMed  Google Scholar 

  137. Poti F, Ceglarek U, Burkhardt R, Simoni M, Nofer JR. SKI-II–a sphingosine kinase 1 inhibitor–exacerbates atherosclerosis in low-density lipoprotein receptor-deficient (LDL-R-/-) mice on high cholesterol diet. Atherosclerosis. 2015;240(1):212–5.

    Article  CAS  PubMed  Google Scholar 

  138. Yu H, Sun C, Argraves KM. Periodontal inflammation and alveolar bone loss induced by Aggregatibacter actinomycetemcomitans is attenuated in sphingosine kinase 1-deficient mice. J Periodontal Res. 2016;51(1):38–49.

    Article  CAS  PubMed  Google Scholar 

  139. Lei YC, Lu CL, Chen L, Ge K, Yang LL, Li W, Wu YH. C5a/C5aR pathway is essential for up-regulating SphK1 expression through p38-MAPK activation in acute liver failure. World J Gastroenterol. 2016;22(46):10148–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Crespo I, San-Miguel B, Sanchez DI, Gonzalez-Fernandez B, Alvarez M, Gonzalez-Gallego J, Tunon MJ. Melatonin inhibits the sphingosine kinase 1/sphingosine-1-phosphate signaling pathway in rabbits with fulminant hepatitis of viral origin. J Pineal Res. 2016;61(2):168–76.

    Article  CAS  PubMed  Google Scholar 

  141. Vasconcelos JF, Meira CS, Silva DN, Nonaka CKV, Daltro PS, Macambira SG, Domizi PD, Borges VM, Ribeiro-Dos-Santos R, de Freitas Souza BS, Soares MBP. Therapeutic effects of sphingosine kinase inhibitor N, N-dimethylsphingosine (DMS) in experimental chronic Chagas disease cardiomyopathy. Sci Rep. 2017;7(1):6171.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Maia LP, Santos PS, Alves PT, Rodrigues CM, Araujo TG, Maia YCP, Camara ATF, Santos DW, Goulart LR. Altered leukocyte sphingolipid pathway in breast cancer. Int J Mol Sci. 2017;18(12):2521.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Pulkoski-Gross MJ, Uys JD, Orr-Gandy KA, Coant N, Bialkowska AB, Szulc ZM, Bai A, Bielawska A, Townsend DM, Hannun YA, Obeid LM, Snider AJ. Novel sphingosine kinase-1 inhibitor, LCL351, reduces immune responses in murine DSS-induced colitis. Prostaglandins Other Lipid Mediat. 2017;130:47–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Crespo I, San-Miguel B, Mauriz JL, Ortiz de Urbina JJ, Almar M, Tunon MJ, Gonzalez-Gallego J. Protective effect of protocatechuic acid on TNBS-induced colitis in mice is associated with modulation of the SphK/S1P signaling pathway. Nutrients. 2017;9(3):288.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Bagalagel A, Diri R, Noor A, Almasri D, Bakhsh HT, Kutbi HI, Al-Gayyar MMH. The therapeutic effects of cycloastragenol in ulcerative colitis by modulating SphK/MIP-1alpha/miR-143 signalling. Basic Clin Pharmacol Toxicol. 2022;131(5):406–19.

    Article  CAS  PubMed  Google Scholar 

  146. Yang G, Gu M, Chen W, Liu W, Xiao Y, Wang H, Lai W, Xian G, Zhang Z, Li Z, Sheng P. SPHK-2 promotes the particle-induced inflammation of RAW264.7 by maintaining consistent expression of TNF-alpha and IL-6. Inflammation. 2018;41(4):1498–507.

    Article  CAS  PubMed  Google Scholar 

  147. Orsini M, Chateauvieux S, Rhim J, Gaigneaux A, Cheillan D, Christov C, Dicato M, Morceau F, Diederich M. Sphingolipid-mediated inflammatory signaling leading to autophagy inhibition converts erythropoiesis to myelopoiesis in human hematopoietic stem/progenitor cells. Cell Death Differ. 2019;26(9):1796–812.

    Article  CAS  PubMed  Google Scholar 

  148. Alqarni I, Bassiouni YA, Badr AM, Ali RA. Telmisartan and/or chlorogenic acid attenuates fructose-induced non-alcoholic fatty liver disease in rats: Implications of cross-talk between angiotensin, the sphingosine kinase/sphingoine-1-phosphate pathway, and TLR4 receptors. Biochem Pharmacol. 2019;164:252–62.

    Article  CAS  PubMed  Google Scholar 

  149. Qiang GH, Wang ZX, Ji AL, Wu JY, Cao Y, Zhang G, Zhang YY, Jiang CP. Sphingosine kinase 1 knockout alleviates hepatic ischemia/reperfusion injury by attenuating inflammation and oxidative stress in mice. Hepatobiliary Pancreat Dis Int. 2019;18(3):255–65.

    Article  CAS  PubMed  Google Scholar 

  150. Kwong EK, Liu R, Zhao D, Li X, Zhu W, Wang X, Gurley EC, Lai G, Liu J, Hylemon PB, Zhou H. The role of sphingosine kinase 2 in alcoholic liver disease. Dig Liver Dis. 2019;51(8):1154–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Choi S, Snider JM, Cariello CP, Lambert JM, Anderson AK, Cowart LA, Snider AJ. Sphingosine kinase 1 is required for myristate-induced TNFalpha expression in intestinal epithelial cells. Prostaglandins Other Lipid Mediat. 2020;149:106423.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Terlizzi M, Colarusso C, Ferraro G, Monti MC, Cerqua I, Roviezzo F, Pinto A, Sorrentino R. Sphingosine-1-phosphate contributes to TLR9-induced TNF-alpha release in lung tumor cells. Cell Physiol Biochem. 2021;55(2):222–34.

    Article  CAS  PubMed  Google Scholar 

  153. Sukocheva OA, Hu DG, Meech R, Bishayee A. Divergence of intracellular trafficking of sphingosine kinase 1 and sphingosine-1-phosphate receptor 3 in MCF-7 breast cancer cells and MCF-7-derived stem cell-enriched mammospheres. Int J Mol Sci. 2021;22(9):4314.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Bernacchioni C, Ghini V, Squecco R, Idrizaj E, Garella R, Puliti E, Cencetti F, Bruni P, Donati C. Role of sphingosine 1-phosphate signalling axis in muscle atrophy induced by TNFalpha in C2C12 myotubes. Int J Mol Sci. 2021;22(3):1280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Avni D, Harikumar KB, Sanyal AJ, Spiegel S. Deletion or inhibition of SphK1 mitigates fulminant hepatic failure by suppressing TNFalpha-dependent inflammation and apoptosis. FASEB J. 2021;35(3):e21415.

    Article  CAS  PubMed  Google Scholar 

  156. Fakhr Y, Koshti S, Habibyan YB, Webster K, Hemmings DG. Tumor necrosis factor-alpha induces a preeclamptic-like phenotype in placental villi via sphingosine kinase 1 activation. Int J Mol Sci. 2022;23(7):3750.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Li L, Wang D, Xin S, Ren X, Zhang J. Sphingosine kinase 1 acts as a hypoxia-upregulated oncogene to regulate cell invasion and resistance to NK cell killing in bladder carcinoma cells. Ann Clin Lab Sci. 2022;52(5):763–71.

    CAS  PubMed  Google Scholar 

  158. Powell JA, Pitman MR, Zebol JR, Moretti PAB, Neubauer HA, Davies LT, Lewis AC, Dagley LF, Webb AI, Costabile M, Pitson SM. Kelch-like protein 5-mediated ubiquitination of lysine 183 promotes proteasomal degradation of sphingosine kinase 1. Biochem J. 2019;476(21):3211–26.

    Article  CAS  PubMed  Google Scholar 

  159. 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(19):20546–54.

    Article  CAS  PubMed  Google Scholar 

  160. Hannun YA, Obeid LM. Sphingolipids and their metabolism in physiology and disease. Nat Rev Mol Cell Biol. 2018;19(3):175–91.

    Article  CAS  PubMed  Google Scholar 

  161. Iessi E, Marconi M, Manganelli V, Sorice M, Malorni W, Garofalo T, Matarrese P. On the role of sphingolipids in cell survival and death. Int Rev Cell Mol Biol. 2020;351:149–95.

    Article  CAS  PubMed  Google Scholar 

  162. Hatoum D, Haddadi N, Lin Y, Nassif NT, McGowan EM. Mammalian sphingosine kinase (SphK) isoenzymes and isoform expression: challenges for SphK as an oncotarget. Oncotarget. 2017;8(22):36898–929.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Zhang Y, Cheng L, Shi X, Song Y, Chen XY, Chen MB, Yao J, Zhang ZQ, Cai S. The sphingosine kinase inhibitor SKI-V suppresses cervical cancer cell growth. Int J Biol Sci. 2022;18(7):2994–3005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Takeshita A, Shinoda H, Nakabayashi Y, Takano A, Matsumoto K, Suetsugu M, Miyazawa K, Tanaka S, Endo H, Tanaka S, Ueyama Y, Hanzawa A, Suda Y, Kanegae H, Yasui T. Sphingosine 1-phosphate acts as a signal molecule in ceramide signal transduction of TNF-alpha-induced activator protein-1 in osteoblastic cell line MC3T3-E1 cells. J Oral Sci. 2005;47(1):43–51.

    Article  CAS  PubMed  Google Scholar 

  165. 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(44):37118–29.

    Article  CAS  PubMed  Google Scholar 

  166. Hasanifard L, Sheervalilou R, Majidinia M, Yousefi B. New insights into the roles and regulation of SphK2 as a therapeutic target in cancer chemoresistance. J Cell Physiol. 2019;234(6):8162–81.

    Article  CAS  PubMed  Google Scholar 

  167. Sukocheva O, Wadham C, Holmes A, Albanese N, Verrier E, Feng F, Bernal A, Derian CK, Ullrich A, Vadas MA, Xia P. Estrogen transactivates EGFR via the sphingosine 1-phosphate receptor Edg-3: the role of sphingosine kinase-1. J Cell Biol. 2006;173(2):301–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Bravo GA, Cedeno RR, Casadevall MP, Ramio-Torrenta L. Sphingosine-1-phosphate (S1P) and S1P signaling pathway modulators, from current insights to future perspectives. Cells. 2022;11(13):2058.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Hallisey VM, Schwab SR. Get me out of here: sphingosine 1-phosphate signaling and T cell exit from tissues during an immune response. Immunol Rev. 2023;317(1):8–19.

    Article  CAS  PubMed  Google Scholar 

  170. de Oliveira PG, Ramos MLS, Amaro AJ, Dias RA, Vieira SI. G(i/o)-protein coupled receptors in the aging brain. Front Aging Neurosci. 2019;11:89.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Adada M, Canals D, Hannun YA, Obeid LM. Sphingosine-1-phosphate receptor 2. FEBS J. 2013;280(24):6354–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Petti L, Rizzo G, Rubbino F, Elangovan S, Colombo P, Restelli S, Piontini A, Arena V, Carvello M, Romano B, Cavalleri T, Anselmo A, Ungaro F, D’Alessio S, Spinelli A, Stifter S, Grizzi F, Sgambato A, Danese S, Laghi L, Malesci A, Vetrano S. Unveiling role of sphingosine-1-phosphate receptor 2 as a brake of epithelial stem cell proliferation and a tumor suppressor in colorectal cancer. J Exp Clin Cancer Res. 2020;39(1):253.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Takashima S, Sugimoto N, Takuwa N, Okamoto Y, Yoshioka K, Takamura M, Takata S, Kaneko S, Takuwa Y. G12/13 and Gq mediate S1P2-induced inhibition of Rac and migration in vascular smooth muscle in a manner dependent on Rho but not Rho kinase. Cardiovasc Res. 2008;79(4):689–97.

    Article  CAS  PubMed  Google Scholar 

  174. Wang X, Guo W, Shi X, Chen Y, Yu Y, Du B, Tan M, Tong L, Wang A, Yin X, Guo J, Martin RC, Bai O, Li Y. S1PR1/S1PR3-YAP signaling and S1P-ALOX15 signaling contribute to an aggressive behavior in obesity-lymphoma. J Exp Clin Cancer Res. 2023;42(1):3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Graler MH, Grosse R, Kusch A, Kremmer E, Gudermann T, Lipp M. The sphingosine 1-phosphate receptor S1P4 regulates cell shape and motility via coupling to Gi and G12/13. J Cell Biochem. 2003;89(3):507–19.

    Article  CAS  PubMed  Google Scholar 

  176. Huang C, Zhu F, Zhang H, Wang N, Huang Q. Identification of S1PR4 as an immune modulator for favorable prognosis in HNSCC through machine learning. iScience. 2023;26(9):107693.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Talmont F, Mitri E, Dozier C, Besson A, Cuvillier O, Hatzoglou A. Sphingosine 1-phosphate receptor 5 (S1P5) deficiency promotes proliferation and immortalization of mouse embryonic fibroblasts. Cancers (Basel). 2022;14(7):1661.

    Article  CAS  PubMed  Google Scholar 

  178. Cuvillier O, Hatzoglou A. Sphingosine 1-phosphate signaling controls mitosis. Oncotarget. 2017;8(70):114414–5.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Murata N, Sato K, Kon J, Tomura H, Yanagita M, Kuwabara A, Ui M, Okajima F. Interaction of sphingosine 1-phosphate with plasma components, including lipoproteins, regulates the lipid receptor-mediated actions. Biochem J. 2000;352 Pt 3(Pt 3):809–15.

    Article  CAS  PubMed  Google Scholar 

  180. Kobayashi N, Kobayashi N, Yamaguchi A, Nishi T. Characterization of the ATP-dependent sphingosine 1-phosphate transporter in rat erythrocytes. J Biol Chem. 2009;284(32):21192–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Jonnalagadda D, Sunkara M, Morris AJ, Whiteheart SW. Granule-mediated release of sphingosine-1-phosphate by activated platelets. Biochim Biophys Acta. 2014;1841(11):1581–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Yatomi Y, Ozaki Y, Ohmori T, Igarashi Y. Sphingosine 1-phosphate: synthesis and release. Prostaglandins Other Lipid Mediat. 2001;64(1–4):107–22.

    Article  CAS  PubMed  Google Scholar 

  183. 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(6):669–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Obinata H, Hla T. Sphingosine 1-phosphate in coagulation and inflammation. Semin Immunopathol. 2012;34(1):73–91.

    Article  CAS  PubMed  Google Scholar 

  185. Pyne NJ, Tonelli F, Lim KG, Long JS, Edwards J, Pyne S. Sphingosine 1-phosphate signalling in cancer. Biochem Soc Trans. 2012;40(1):94–100.

    Article  CAS  PubMed  Google Scholar 

  186. Yang CC, Hsiao LD, Su MH, Yang CM. Sphingosine 1-phosphate induces cyclooxygenase-2/prostaglandin E(2) expression via PKCalpha-dependent mitogen-activated protein kinases and NF-kappaB cascade in human cardiac fibroblasts. Front Pharmacol. 2020;11:569802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Wang S, Liang Y, Chang W, Hu B, Zhang Y. Triple negative breast cancer depends on sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P)/sphingosine 1-phosphate receptor 3 (S1PR3)/notch signaling for metastasis. Med Sci Monit. 2018;24:1912–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Igarashi J, Bernier SG, Michel T. Sphingosine 1-phosphate and activation of endothelial nitric-oxide synthase: differential regulation of Akt and MAP kinase pathways by EDG and bradykinin receptors in vascular endothelial cells. J Biol Chem. 2001;276(15):12420–6.

    Article  CAS  PubMed  Google Scholar 

  189. Limaye V, Li X, Hahn C, Xia P, Berndt MC, Vadas MA, Gamble JR. Sphingosine kinase-1 enhances endothelial cell survival through a PECAM-1-dependent activation of PI-3K/Akt and regulation of Bcl-2 family members. Blood. 2005;105(8):3169–77.

    Article  CAS  PubMed  Google Scholar 

  190. Siehler S, Wang Y, Fan X, Windh RT, Manning DR. Sphingosine 1-phosphate activates nuclear factor-kappa B through Edg receptors. Activation through Edg-3 and Edg-5, but not Edg-1, in human embryonic kidney 293 cells. J Biol Chem. 2001;276(52):48733–9.

    Article  CAS  PubMed  Google Scholar 

  191. Huang LS, Sudhadevi T, Fu P, Punathil-Kannan PK, Ebenezer DL, Ramchandran R, Putherickal V, Cheresh P, Zhou G, Ha AW, Harijith A, Kamp DW, Natarajan V. Sphingosine kinase 1/S1P signaling contributes to pulmonary fibrosis by activating Hippo/YAP pathway and mitochondrial reactive oxygen species in lung fibroblasts. Int J Mol Sci. 2020;21(6):2064.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Che W, Manetsch M, Quante T, Rahman MM, Patel BS, Ge Q, Ammit AJ. Sphingosine 1-phosphate induces MKP-1 expression via p38 MAPK- and CREB-mediated pathways in airway smooth muscle cells. Biochim Biophys Acta. 2012;1823(10):1658–65.

    Article  CAS  PubMed  Google Scholar 

  193. Obinata H, Hla T. Sphingosine 1-phosphate and inflammation. Int Immunol. 2019;31(9):617–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. El-Shewy HM, Abdel-Samie SA, Al Qalam AM, Lee MH, Kitatani K, Anelli V, Jaffa AA, Obeid LM, Luttrell LM. Phospholipase C and protein kinase C-beta 2 mediate insulin-like growth factor II-dependent sphingosine kinase 1 activation. Mol Endocrinol. 2011;25(12):2144–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Etemadi N, Chopin M, Anderton H, Tanzer MC, Rickard JA, Abeysekera W, Hall C, Spall SK, Wang B, Xiong Y, Hla T, Pitson SM, Bonder CS, Wong WW, Ernst M, Smyth GK, Vaux DL, Nutt SL, Nachbur U, Silke J. TRAF2 regulates TNF and NF-kappaB signalling to suppress apoptosis and skin inflammation independently of sphingosine kinase 1. Elife. 2015;4:e10592.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, Luo C, Marmorstein R, Kordula T, Milstien S, Spiegel S. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science. 2009;325(5945):1254–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Park K, Ikushiro H, Seo HS, Shin KO, Kim YI, Kim JY, Lee YM, Yano T, Holleran WM, Elias P, Uchida Y. ER stress stimulates production of the key antimicrobial peptide, cathelicidin, by forming a previously unidentified intracellular S1P signaling complex. Proc Natl Acad Sci U S A. 2016;113(10):E1334–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Laviad EL, Albee L, Pankova-Kholmyansky I, Epstein S, Park H, Merrill AH Jr, Futerman AH. Characterization of ceramide synthase 2: tissue distribution, substrate specificity, and inhibition by sphingosine 1-phosphate. J Biol Chem. 2008;283(9):5677–84.

    Article  CAS  PubMed  Google Scholar 

  199. Young MM, Wang HG. Sphingolipids as regulators of autophagy and endocytic trafficking. Adv Cancer Res. 2018;140:27–60.

    Article  CAS  PubMed  Google Scholar 

  200. Jeffries KA, Krupenko NI. Ceramide signaling and p53 pathways. Adv Cancer Res. 2018;140:191–215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Kuribayashi K, Krigsfeld G, Wang W, Xu J, Mayes PA, Dicker DT, Wu GS, El-Deiry WS. TNFSF10 (TRAIL), a p53 target gene that mediates p53-dependent cell death. Cancer Biol Ther. 2008;7(12):2034–8.

    Article  CAS  PubMed  Google Scholar 

  202. Chen MB, Jiang Q, Liu YY, Zhang Y, He BS, Wei MX, Lu JW, Ji Y, Lu PH. C6 ceramide dramatically increases vincristine sensitivity both in vivo and in vitro, involving AMP-activated protein kinase-p53 signaling. Carcinogenesis. 2015;36(9):1061–70.

    Article  CAS  PubMed  Google Scholar 

  203. Hengst JA, Guilford JM, Fox TE, Wang X, Conroy EJ, Yun JK. Sphingosine kinase 1 localized to the plasma membrane lipid raft microdomain overcomes serum deprivation induced growth inhibition. Arch Biochem Biophys. 2009;492(1–2):62–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Sah DK, Rai Y, Chauhan A, Kumari N, Chaturvedi MM, Bhatt AN. Sphingosine kinase inhibitor, SKI-II confers protection against the ionizing radiation by maintaining redox homeostasis most likely through Nrf2 signaling. Life Sci. 2021;278:119543.

    Article  CAS  PubMed  Google Scholar 

  205. Yester JW, Bryan L, Waters MR, Mierzenski B, Biswas DD, Gupta AS, Bhardwaj R, Surace MJ, Eltit JM, Milstien S, Spiegel S, Kordula T. Sphingosine-1-phosphate inhibits IL-1-induced expression of C-C motif ligand 5 via c-Fos-dependent suppression of IFN-beta amplification loop. FASEB J. 2015;29(12):4853–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Zhao S, Adebiyi MG, Zhang Y, Couturier JP, Fan X, Zhang H, Kellems RE, Lewis DE, Xia Y. Sphingosine-1-phosphate receptor 1 mediates elevated IL-6 signaling to promote chronic inflammation and multitissue damage in sickle cell disease. FASEB J. 2018;32(5):2855–65.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Ohama T, Okada M, Murata T, Brautigan DL, Hori M, Ozaki H. Sphingosine-1-phosphate enhances IL-1beta-induced COX-2 expression in mouse intestinal subepithelial myofibroblasts. Am J Physiol Gastrointest Liver Physiol. 2008;295(4):G766–75.

    Article  CAS  PubMed  Google Scholar 

  208. Muller J, von Bernstorff W, Heidecke CD, Schulze T. Differential S1P receptor profiles on M1- and M2-polarized macrophages affect macrophage cytokine production and migration. Biomed Res Int. 2017;2017:7584621.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Heo JY, Im DS. Pro-inflammatory role of S1P(3) in macrophages. Biomol Ther (Seoul). 2019;27(4):373–80.

    Article  PubMed  PubMed Central  Google Scholar 

  210. Chakraborty P, Vaena SG, Thyagarajan K, Chatterjee S, Al-Khami A, Selvam SP, Nguyen H, Kang I, Wyatt MW, Baliga U, Hedley Z, Ngang RN, Guo B, Beeson GC, Husain S, Paulos CM, Beeson CC, Zilliox MJ, Hill EG, Mehrotra M, Yu XZ, Ogretmen B, Mehrotra S. Pro-survival lipid sphingosine-1-phosphate metabolically programs T cells to limit anti-tumor activity. Cell Rep. 2019;28(7):1879-1893.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Imbert C, Montfort A, Fraisse M, Marcheteau E, Gilhodes J, Martin E, Bertrand F, Marcellin M, Burlet-Schiltz O, Peredo AG, Garcia V, Carpentier S, Tartare-Deckert S, Brousset P, Rochaix P, Puisset F, Filleron T, Meyer N, Lamant L, Levade T, Segui B, Andrieu-Abadie N, Colacios C. Resistance of melanoma to immune checkpoint inhibitors is overcome by targeting the sphingosine kinase-1. Nat Commun. 2020;11(1):437.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Hutami IR, Izawa T, Khurel-Ochir T, Sakamaki T, Iwasa A, Tomita S, Tanaka E. HIF-1alpha controls palatal wound healing by regulating macrophage motility via S1P/S1P(1) signaling axis. Oral Dis. 2022;28(4):1157–69.

    Article  PubMed  Google Scholar 

  213. Snider AJ, Orr Gandy KA, Obeid LM. Sphingosine kinase: role in regulation of bioactive sphingolipid mediators in inflammation. Biochimie. 2010;92(6):707–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Hughes JE, Srinivasan S, Lynch KR, Proia RL, Ferdek P, Hedrick CC. Sphingosine-1-phosphate induces an antiinflammatory phenotype in macrophages. Circ Res. 2008;102(8):950–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Michaud J, Im DS, Hla T. Inhibitory role of sphingosine 1-phosphate receptor 2 in macrophage recruitment during inflammation. J Immunol. 2010;184(3):1475–83.

    Article  CAS  PubMed  Google Scholar 

  216. Skoura A, Michaud J, Im DS, Thangada S, Xiong Y, Smith JD, Hla T. Sphingosine-1-phosphate receptor-2 function in myeloid cells regulates vascular inflammation and atherosclerosis. Arterioscler Thromb Vasc Biol. 2011;31(1):81–5.

    Article  CAS  PubMed  Google Scholar 

  217. Di A, Kawamura T, Gao XP, Tang H, Berdyshev E, Vogel SM, Zhao YY, Sharma T, Bachmaier K, Xu J, Malik AB. A novel function of sphingosine kinase 1 suppression of JNK activity in preventing inflammation and injury. J Biol Chem. 2010;285(21):15848–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Lee JY, Han SH, Park MH, Baek B, Song IS, Choi MK, Takuwa Y, Ryu H, Kim SH, He X, Schuchman EH, Bae JS, Jin HK. Neuronal SphK1 acetylates COX2 and contributes to pathogenesis in a model of Alzheimer’s Disease. Nat Commun. 2018;9(1):1479.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Bailey SG, Cragg MS, Townsend PA. Role of STAT1 in the breast. JAKSTAT. 2012;1(3):197–9.

    PubMed  PubMed Central  Google Scholar 

  220. Hii LW, Chung FF, Mai CW, Yee ZY, Chan HH, Raja VJ, Dephoure NE, Pyne NJ, Pyne S, Leong CO. Sphingosine kinase 1 regulates the survival of breast cancer stem cells and non-stem breast cancer cells by suppression of STAT1. Cells. 2020;9(4):886.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Diaz Escarcega R, McCullough LD, Tsvetkov AS. The functional role of sphingosine kinase 2. Front Mol Biosci. 2021;8:683767.

    Article  PubMed  PubMed Central  Google Scholar 

  222. Sukocheva OA, Liu J, Neganova ME, Beeraka NM, Aleksandrova YR, Manogaran P, Grigorevskikh EM, Chubarev VN, Fan R. Perspectives of using microRNA-loaded nanocarriers for epigenetic reprogramming of drug resistant colorectal cancers. Semin Cancer Biol. 2022;86(Pt 2):358–75.

    Article  CAS  PubMed  Google Scholar 

  223. Sukocheva OA, Lukina E, Friedemann M, Menschikowski M, Hagelgans A, Aliev G. The crucial role of epigenetic regulation in breast cancer anti-estrogen resistance: current findings and future perspectives. Semin Cancer Biol. 2022;82:35–59.

    Article  CAS  PubMed  Google Scholar 

  224. Kang C, LeRoith D, Gallagher EJ. Diabetes, obesity, and breast cancer. Endocrinology. 2018;159(11):3801–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Terzic J, Grivennikov S, Karin E, Karin M. Inflammation and colon cancer. Gastroenterology. 2010;138(6):2101-2114.e5.

    Article  CAS  PubMed  Google Scholar 

  226. Chen K, Zhang J, Beeraka NM, Tang C, Babayeva YV, Sinelnikov MY, Zhang X, Zhang J, Liu J, Reshetov IV, Sukocheva OA, Lu P, Fan R. Advances in the prevention and treatment of obesity-driven effects in breast cancers. Front Oncol. 2022;12:820968.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Esposito K, Chiodini P, Colao A, Lenzi A, Giugliano D. Metabolic syndrome and risk of cancer: a systematic review and meta-analysis. Diabetes Care. 2012;35(11):2402–11.

    Article  PubMed  PubMed Central  Google Scholar 

  228. Kabat GC, Kim MY, Peters U, Stefanick M, Hou L, Wactawski-Wende J, Messina C, Shikany JM, Rohan TE. A longitudinal study of the metabolic syndrome and risk of colorectal cancer in postmenopausal women. Eur J Cancer Prev. 2012;21(4):326–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Tsilidis KK, Erlinger TP, Rifai N, Hoffman S, Hoffman-Bolton J, Helzlsouer KJ, Platz EA. C-reactive protein and colorectal adenoma in the CLUE II cohort. Cancer Causes Control. 2008;19(6):559–67.

    Article  PubMed  Google Scholar 

  230. Pietrzyk L, Torres A, Maciejewski R, Torres K. Obesity and obese-related chronic low-grade inflammation in promotion of colorectal cancer development. Asian Pac J Cancer Prev. 2015;16(10):4161–8.

    Article  PubMed  Google Scholar 

  231. Feldstein AE, Werneburg NW, Canbay A, Guicciardi ME, Bronk SF, Rydzewski R, Burgart LJ, Gores GJ. Free fatty acids promote hepatic lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway. Hepatology. 2004;40(1):185–94.

    Article  CAS  PubMed  Google Scholar 

  232. Colditz GA, Peterson LL. Obesity and cancer: evidence, impact, and future directions. Clin Chem. 2018;64(1):154–62.

    Article  CAS  PubMed  Google Scholar 

  233. Lin TC, Hsiao M. Leptin and cancer: updated functional roles in carcinogenesis, therapeutic niches, and developments. Int J Mol Sci. 2021;22(6):2870.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Pandit R, Beerens S, Adan RAH. Role of leptin in energy expenditure: the hypothalamic perspective. Am J Physiol Regul Integr Comp Physiol. 2017;312(6):R938–47.

    Article  CAS  PubMed  Google Scholar 

  235. Enriori PJ, Sinnayah P, Simonds SE, Garcia Rudaz C, Cowley MA. Leptin action in the dorsomedial hypothalamus increases sympathetic tone to brown adipose tissue in spite of systemic leptin resistance. J Neurosci. 2011;31(34):12189–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Howard JM, Pidgeon GP, Reynolds JV. Leptin and gastro-intestinal malignancies. Obes Rev. 2010;11(12):863–74.

    Article  CAS  PubMed  Google Scholar 

  237. Alshaker H, Krell J, Frampton AE, Waxman J, Blyuss O, Zaikin A, Winkler M, Stebbing J, Yague E, Pchejetski D. Leptin induces upregulation of sphingosine kinase 1 in oestrogen receptor-negative breast cancer via Src family kinase-mediated, janus kinase 2-independent pathway. Breast Cancer Res. 2014;16(5):426.

    Article  PubMed  PubMed Central  Google Scholar 

  238. Wang J, Badeanlou L, Bielawski J, Ciaraldi TP, Samad F. Sphingosine kinase 1 regulates adipose proinflammatory responses and insulin resistance. Am J Physiol Endocrinol Metab. 2014;306(7):E756–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Ross JS, Hu W, Rosen B, Snider AJ, Obeid LM, Cowart LA. Sphingosine kinase 1 is regulated by peroxisome proliferator-activated receptor alpha in response to free fatty acids and is essential for skeletal muscle interleukin-6 production and signaling in diet-induced obesity. J Biol Chem. 2013;288(31):22193–206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Nagahashi M, Takabe K, Liu R, Peng K, Wang X, Wang Y, Hait NC, Wang X, Allegood JC, Yamada A, Aoyagi T, Liang J, Pandak WM, Spiegel S, Hylemon PB, Zhou H. Conjugated bile acid-activated S1P receptor 2 is a key regulator of sphingosine kinase 2 and hepatic gene expression. Hepatology. 2015;61(4):1216–26.

    Article  CAS  PubMed  Google Scholar 

  241. Kim MH, Park JW, Lee EJ, Kim S, Shin SH, Ahn JH, Jung Y, Park I, Park WJ. C16-ceramide and sphingosine 1-phosphate/S1PR2 have opposite effects on cell growth through mTOR signaling pathway regulation. Oncol Rep. 2018;40(5):2977–87.

    CAS  PubMed  Google Scholar 

  242. Siegmund D, Wagner J, Wajant H. TNF receptor associated factor 2 (TRAF2) signaling in cancer. Cancers (Basel). 2022;14(16):4055.

    Article  CAS  PubMed  Google Scholar 

  243. Lee EG, Boone DL, Chai S, Libby SL, Chien M, Lodolce JP, Ma A. Failure to regulate TNF-induced NF-kappaB and cell death responses in A20-deficient mice. Science. 2000;289(5488):2350–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Hjelmeland AB, Wu Q, Wickman S, Eyler C, Heddleston J, Shi Q, Lathia JD, Macswords J, Lee J, McLendon RE, Rich JN. Targeting A20 decreases glioma stem cell survival and tumor growth. PLoS Biol. 2010;8(2):e1000319.

    Article  PubMed  PubMed Central  Google Scholar 

  245. Yuan W, Chen Y, Zhou Y, Bao K, Yu X, Xu Y, Zhang Y, Zheng J, Jiang G, Hong M. Formononetin attenuates atopic dermatitis by upregulating A20 expression via activation of G protein-coupled estrogen receptor. J Ethnopharmacol. 2021;266:113397.

    Article  CAS  PubMed  Google Scholar 

  246. Feng Y, Zhang Y, Cai Y, Liu R, Lu M, Li T, Fu Y, Guo M, Huang H, Ou Y, Chen Y. A20 targets PFKL and glycolysis to inhibit the progression of hepatocellular carcinoma. Cell Death Dis. 2020;11(2):89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Lee HJ, Jung YH, Choi GE, Kim JS, Chae CW, Lim JR, Kim SY, Lee JE, Park MC, Yoon JH, Choi MJ, Kim KS, Han HJ. O-cyclic phytosphingosine-1-phosphate stimulates HIF1alpha-dependent glycolytic reprogramming to enhance the therapeutic potential of mesenchymal stem cells. Cell Death Dis. 2019;10(8):590.

    Article  PubMed  PubMed Central  Google Scholar 

  248. Lv Q, Xie L, Cheng Y, Shi Y, Shan W, Ning C, Xie B, Yang B, Luo X, He Q, Zhu Q, Zhang Y, Zhang Z, Wang C, Chen X, Xu C. A20-mediated deubiquitination of ERalpha in the microenvironment of CD163(+) macrophages sensitizes endometrial cancer cells to estrogen. Cancer Lett. 2019;442:137–47.

    Article  CAS  PubMed  Google Scholar 

  249. Yang C, Jiang Q. Vitamin E delta-tocotrienol inhibits TNF-alpha-stimulated NF-kappaB activation by up-regulation of anti-inflammatory A20 via modulation of sphingolipid including elevation of intracellular dihydroceramides. J Nutr Biochem. 2019;64:101–9.

    Article  CAS  PubMed  Google Scholar 

  250. Dbaibo GS, Obeid LM, Hannun YA. Tumor necrosis factor-alpha (TNF-alpha) signal transduction through ceramide. Dissociation of growth inhibitory effects of TNF-alpha from activation of nuclear factor-kappa B. J Biol Chem. 1993;268(24):17762–6.

    Article  CAS  PubMed  Google Scholar 

  251. Matsuzawa Y, Oshima S, Takahara M, Maeyashiki C, Nemoto Y, Kobayashi M, Nibe Y, Nozaki K, Nagaishi T, Okamoto R, Tsuchiya K, Nakamura T, Ma A, Watanabe M. TNFAIP3 promotes survival of CD4 T cells by restricting MTOR and promoting autophagy. Autophagy. 2015;11(7):1052–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Shi CS, Kehrl JH. TRAF6 and A20 regulate lysine 63-linked ubiquitination of Beclin-1 to control TLR4-induced autophagy. Sci Signal. 2010;3(123):ra42.

    Article  PubMed  PubMed Central  Google Scholar 

  253. Riou C, Yassine-Diab B, Van grevenynghe J, Somogyi R, Greller LD, Gagnon D, Gimmig S, Wilkinson P, Shi Y, Cameron MJ, Campos-Gonzalez R, Balderas RS, Kelvin D, Sekaly RP, Haddad EK. Convergence of TCR and cytokine signaling leads to FOXO3a phosphorylation and drives the survival of CD4+ central memory T cells. J Exp Med. 2007;204(1):79–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Godfrey DI, Hammond KJ, Poulton LD, Smyth MJ, Baxter AG. NKT cells: facts, functions and fallacies. Immunol Today. 2000;21(11):573–83.

    Article  CAS  PubMed  Google Scholar 

  255. Myers JA, Miller JS. Exploring the NK cell platform for cancer immunotherapy. Nat Rev Clin Oncol. 2021;18(2):85–100.

    Article  PubMed  Google Scholar 

  256. Lorenzo-Herrero S, Sordo-Bahamonde C, Gonzalez S, Lopez-Soto A. Immunosurveillance of cancer cell stress. Cell Stress. 2019;3(9):295–309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Sordo-Bahamonde C, Lorenzo-Herrero S, Payer AR, Gonzalez S, Lopez-Soto A. Mechanisms of apoptosis resistance to NK cell-mediated cytotoxicity in cancer. Int J Mol Sci. 2020;21(10):3726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Villanueva JE, Malle EK, Gardam S, Silveira PA, Zammit NW, Walters SN, Brink R, Grey ST. TRAF2 regulates peripheral CD8(+) T-cell and NKT-cell homeostasis by modulating sensitivity to IL-15. Eur J Immunol. 2015;45(6):1820–31.

    Article  CAS  PubMed  Google Scholar 

  259. Shah A, Plaza-Sirvent C, Weinert S, Buchbinder JH, Lavrik IN, Mertens PR, Schmitz I, Lindquist JA. YB-1 mediates TNF-induced pro-survival signaling by regulating NF-kappaB activation. Cancers (Basel). 2020;12(8):2188.

    Article  CAS  PubMed  Google Scholar 

  260. Shi H, Niimi A, Takeuchi T, Shiogama K, Mizutani Y, Kajino T, Inada K, Hase T, Hatta T, Shibata H, Fukui T, Chen-Yoshikawa TF, Nagano K, Murate T, Kawamoto Y, Tomida S, Takahashi T, Suzuki M. CEBPgamma facilitates lamellipodia formation and cancer cell migration through CERS6 upregulation. Cancer Sci. 2021;112(7):2770–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  261. Helke K, Angel P, Lu P, Garrett-Mayer E, Ogretmen B, Drake R, Voelkel-Johnson C. Ceramide synthase 6 deficiency enhances inflammation in the DSS model of colitis. Sci Rep. 2018;8(1):1627.

    Article  PubMed  PubMed Central  Google Scholar 

  262. Morrish E, Brumatti G, Silke J. Future therapeutic directions for smac-mimetics. Cells. 2020;9(2):406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  263. Medler J, Kucka K, Wajant H. Tumor necrosis factor receptor 2 (TNFR2): an emerging target in cancer therapy. Cancers (Basel). 2022;14(11):2603.

    Article  CAS  PubMed  Google Scholar 

  264. Napolitano G, Karin M. Sphingolipids: the oil on the TRAFire that promotes inflammation. Sci Signal. 2010;3(141):pe34.

    Article  PubMed  Google Scholar 

  265. Blaho VA, Hla T. An update on the biology of sphingosine 1-phosphate receptors. J Lipid Res. 2014;55(8):1596–608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Calabresi PA, Radue EW, Goodin D, Jeffery D, Rammohan KW, Reder AT, Vollmer T, Agius MA, Kappos L, Stites T, Li B, Cappiello L, von Rosenstiel P, Lublin FD. Safety and efficacy of fingolimod in patients with relapsing-remitting multiple sclerosis (FREEDOMS II): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Neurol. 2014;13(6):545–56.

    Article  CAS  PubMed  Google Scholar 

  267. Ito S, Iwaki S, Kondo R, Satoh M, Iwabuchi K, Ohkawa R, Mishima Y, Yatomi Y, Furumoto T, Tsutsui H, Fujii S. TNF-alpha production in NKT cell hybridoma is regulated by sphingosine-1-phosphate: implications for inflammation in atherosclerosis. Coron Artery Dis. 2014;25(4):311–20.

    Article  PubMed  Google Scholar 

  268. Terlizzi M, Colarusso C, Somma P, De Rosa I, Panico L, Pinto A, Sorrentino R. S1P-induced TNF-alpha and IL-6 release from PBMCs exacerbates lung cancer-associated inflammation. Cells. 2022;11(16):2524.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Lai WQ, Irwan AW, Goh HH, Howe HS, Yu DT, Valle-Onate R, McInnes IB, Melendez AJ, Leung BP. Anti-inflammatory effects of sphingosine kinase modulation in inflammatory arthritis. J Immunol. 2008;181(11):8010–7.

    Article  CAS  PubMed  Google Scholar 

  270. Zhu C, Wen S, Li J, Meng H, Zhang J, Zhao K, Wang L, Zhang Y. FTY720 inhibits the development of collagen-induced arthritis in mice by suppressing the recruitment of CD4(+) T lymphocytes. Drug Des Devel Ther. 2021;15:1981–92.

    Article  PubMed  PubMed Central  Google Scholar 

  271. Lee MS, Sun W, Webb TJ. Sphingosine kinase blockade leads to increased natural killer T cell responses to mantle cell lymphoma. Cells. 2020;9(4):1030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Dillmann C, Mora J, Olesch C, Brune B, Weigert A. S1PR4 is required for plasmacytoid dendritic cell differentiation. Biol Chem. 2015;396(6–7):775–82.

    Article  CAS  PubMed  Google Scholar 

  273. Maeda Y, Matsuyuki H, Shimano K, Kataoka H, Sugahara K, Chiba K. Migration of CD4 T cells and dendritic cells toward sphingosine 1-phosphate (S1P) is mediated by different receptor subtypes: S1P regulates the functions of murine mature dendritic cells via S1P receptor type 3. J Immunol. 2007;178(6):3437–46.

    Article  CAS  PubMed  Google Scholar 

  274. Schuster C, Huard A, Sirait-Fischer E, Dillmann C, Brune B, Weigert A. S1PR4-dependent CCL2 production promotes macrophage recruitment in a murine psoriasis model. Eur J Immunol. 2020;50(6):839–45.

    Article  CAS  PubMed  Google Scholar 

  275. Schwiebs A, Herrero San Juan M, Schmidt KG, Wiercinska E, Anlauf M, Ottenlinger F, Thomas D, Elwakeel E, Weigert A, Farin HF, Bonig H, Scholich K, Geisslinger G, Pfeilschifter JM, Radeke HH. Cancer-induced inflammation and inflammation-induced cancer in colon: a role for S1P lyase. Oncogene. 2019;38(24):4788–803.

    Article  CAS  PubMed  Google Scholar 

  276. Serra M, Saba JD. Sphingosine 1-phosphate lyase, a key regulator of sphingosine 1-phosphate signaling and function. Adv Enzyme Regul. 2010;50(1):349–62.

    Article  PubMed  Google Scholar 

  277. Dillmann C, Ringel C, Ringleb J, Mora J, Olesch C, Fink AF, Roberts E, Brune B, Weigert A. S1PR4 signaling attenuates ILT 7 internalization to limit IFN-alpha production by human plasmacytoid dendritic cells. J Immunol. 2016;196(4):1579–90.

    Article  CAS  PubMed  Google Scholar 

  278. Weichand B, Weis N, Weigert A, Grossmann N, Levkau B, Brune B. Apoptotic cells enhance sphingosine-1-phosphate receptor 1 dependent macrophage migration. Eur J Immunol. 2013;43(12):3306–13.

    Article  CAS  PubMed  Google Scholar 

  279. Weigert A, von Knethen A, Thomas D, Faria I, Namgaladze D, Zezina E, Fuhrmann D, Petcherski A, Heringdorf DMZ, Radeke HH, Brune B. Sphingosine kinase 2 is a negative regulator of inflammatory macrophage activation. Biochim Biophys Acta Mol Cell Biol Lipids. 2019;1864(9):1235–46.

    Article  CAS  PubMed  Google Scholar 

  280. Ghosh M, Thangada S, Dasgupta O, Khanna KM, Yamase HT, Kashgarian M, Hla T, Shapiro LH, Ferrer FA. Cell-intrinsic sphingosine kinase 2 promotes macrophage polarization and renal inflammation in response to unilateral ureteral obstruction. PLoS One. 2018;13(3):e0194053.

    Article  PubMed  PubMed Central  Google Scholar 

  281. Zehra Okus F, Busra Azizoglu Z, Canatan H, Eken A. S1P analogues SEW2871, BAF312 and FTY720 affect human Th17 and Treg generation ex vivo. Int Immunopharmacol. 2022;107:108665.

    Article  CAS  PubMed  Google Scholar 

  282. Chiu DK, Xu IM, Lai RK, Tse AP, Wei LL, Koh HY, Li LL, Lee D, Lo RC, Wong CM, Ng IO, Wong CC. Hypoxia induces myeloid-derived suppressor cell recruitment to hepatocellular carcinoma through chemokine (C-C motif) ligand 26. Hepatology. 2016;64(3):797–813.

    Article  CAS  PubMed  Google Scholar 

  283. Liu C, Yu S, Kappes J, Wang J, Grizzle WE, Zinn KR, Zhang HG. Expansion of spleen myeloid suppressor cells represses NK cell cytotoxicity in tumor-bearing host. Blood. 2007;109(10):4336–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  284. Lazarova M, Steinle A. Impairment of NKG2D-mediated tumor immunity by TGF-beta. Front Immunol. 2019;10:2689.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  285. Monack D, Falkow S. Apoptosis as a common bacterial virulence strategy. Int J Med Microbiol. 2000;290(1):7–13.

    Article  CAS  PubMed  Google Scholar 

  286. Yang R, Yi M, Xiang B. Novel insights on lipid metabolism alterations in drug resistance in cancer. Front Cell Dev Biol. 2022;10:875318.

    Article  PubMed  PubMed Central  Google Scholar 

  287. Liu YN, Zhang H, Zhang L, Cai TT, Huang DJ, He J, Ni HH, Zhou FJ, Zhang XS, Li J. Sphingosine 1 phosphate receptor-1 (S1P1) promotes tumor-associated regulatory T cell expansion: leading to poor survival in bladder cancer. Cell Death Dis. 2019;10(2):50.

    Article  PubMed  PubMed Central  Google Scholar 

  288. Ramezani-Ali Akbari K, Khaki-Bakhtiarvand V, Mahmoudian J, Asgarian-Omran H, Shokri F, Hojjat-Farsangi M, Jeddi-Tehrani M, Shabani M. Cloning, expression and characterization of a peptibody to deplete myeloid derived suppressor cells in a murine mammary carcinoma model. Protein Expr Purif. 2022;200:106153.

    Article  CAS  PubMed  Google Scholar 

  289. Riemma MA, Cerqua I, Romano B, Irollo E, Bertolino A, Camerlingo R, Granato E, Rea G, Scala S, Terlizzi M, Spaziano G, Sorrentino R, D’Agostino B, Roviezzo F, Cirino G. Sphingosine-1-phosphate/TGF-beta axis drives epithelial mesenchymal transition in asthma-like disease. Br J Pharmacol. 2022;179(8):1753–68.

    Article  CAS  PubMed  Google Scholar 

  290. Sermeus A, Genin M, Maincent A, Fransolet M, Notte A, Leclere L, Riquier H, Arnould T, Michiels C. Hypoxia-induced modulation of apoptosis and BCL-2 family proteins in different cancer cell types. PLoS One. 2012;7(11):e47519.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Park SY, Billiar TR, Seol DW. Hypoxia inhibition of apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Biochem Biophys Res Commun. 2002;291(1):150–3.

    Article  CAS  PubMed  Google Scholar 

  292. LeBlanc FR, Pearson JM, Tan SF, Cheon H, Xing JC, Dunton W, Feith DJ, Loughran TP Jr. Sphingosine kinase-2 is overexpressed in large granular lymphocyte leukaemia and promotes survival through Mcl-1. Br J Haematol. 2020;190(3):405–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  293. Rutherford C, Childs S, Ohotski J, McGlynn L, Riddick M, MacFarlane S, Tasker D, Pyne S, Pyne NJ, Edwards J, Palmer TM. Regulation of cell survival by sphingosine-1-phosphate receptor S1P1 via reciprocal ERK-dependent suppression of Bim and PI-3-kinase/protein kinase C-mediated upregulation of Mcl-1. Cell Death Dis. 2013;4(11):e927.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  294. Woo SM, Seo BR, Min KJ, Kwon TK. FTY720 enhances TRAIL-mediated apoptosis by up-regulating DR5 and down-regulating Mcl-1 in cancer cells. Oncotarget. 2015;6(13):11614–26.

    Article  PubMed  PubMed Central  Google Scholar 

  295. Asakura T, Ishii M, Namkoong H, Suzuki S, Kagawa S, Yagi K, Komiya T, Hashimoto T, Okamori S, Kamata H, Tasaka S, Kihara A, Hegab AE, Hasegawa N, Betsuyaku T. Sphingosine 1-phosphate receptor modulator ONO-4641 stimulates CD11b(+)Gr-1(+) cell expansion and inhibits lymphocyte infiltration in the lungs to ameliorate murine pulmonary emphysema. Mucosal Immunol. 2018;11(6):1606–20.

    Article  CAS  PubMed  Google Scholar 

  296. Li T, Yi S, Liu W, Jia C, Wang G, Hua X, Tai Y, Zhang Q, Chen G. Colorectal carcinoma-derived fibroblasts modulate natural killer cell phenotype and antitumor cytotoxicity. Med Oncol. 2013;30(3):663.

    Article  PubMed  Google Scholar 

  297. Zhang R, Qi F, Zhao F, Li G, Shao S, Zhang X, Yuan L, Feng Y. Cancer-associated fibroblasts enhance tumor-associated macrophages enrichment and suppress NK cells function in colorectal cancer. Cell Death Dis. 2019;10(4):273.

    Article  PubMed  PubMed Central  Google Scholar 

  298. Ziani L, Safta-Saadoun TB, Gourbeix J, Cavalcanti A, Robert C, Favre G, Chouaib S, Thiery J. Melanoma-associated fibroblasts decrease tumor cell susceptibility to NK cell-mediated killing through matrix-metalloproteinases secretion. Oncotarget. 2017;8(12):19780–94.

    Article  PubMed  PubMed Central  Google Scholar 

  299. Lowe SW, Cepero E, Evan G. Intrinsic tumour suppression. Nature. 2004;432(7015):307–15.

    Article  CAS  PubMed  Google Scholar 

  300. Ziani L, Chouaib S, Thiery J. Alteration of the antitumor immune response by cancer-associated fibroblasts. Front Immunol. 2018;9:414.

    Article  PubMed  PubMed Central  Google Scholar 

  301. Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood. 2008;111(3):1327–33.

    Article  CAS  PubMed  Google Scholar 

  302. Cheng JT, Deng YN, Yi HM, Wang GY, Fu BS, Chen WJ, Liu W, Tai Y, Peng YW, Zhang Q. Hepatic carcinoma-associated fibroblasts induce IDO-producing regulatory dendritic cells through IL-6-mediated STAT3 activation. Oncogenesis. 2016;5(2):e198.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  303. Weigel C, Maczis MA, Palladino END, Green CD, Maceyka M, Guo C, Wang XY, Dozmorov MG, Milstien S, Spiegel S. Sphingosine kinase 2 in stromal fibroblasts creates a hospitable tumor microenvironment in breast cancer. Cancer Res. 2023;83(4):553–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  304. Galland S, Vuille J, Martin P, Letovanec I, Caignard A, Fregni G, Stamenkovic I. Tumor-derived mesenchymal stem cells use distinct mechanisms to block the activity of natural killer cell subsets. Cell Rep. 2017;20(12):2891–905.

    Article  CAS  PubMed  Google Scholar 

  305. Kim EY, Choi B, Kim JE, Park SO, Kim SM, Chang EJ. Interleukin-22 mediates the chemotactic migration of breast cancer cells and macrophage infiltration of the bone microenvironment by potentiating S1P/SIPR signaling. Cells. 2020;9(1):131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  306. Schneider G. S1P signaling in the tumor microenvironment. Adv Exp Med Biol. 2020;1223:129–53.

    Article  CAS  PubMed  Google Scholar 

  307. Heck AL, Mishra S, Prenzel T, Feulner L, Achhammer E, Sarchen V, Blagg BSJ, Schneider-Brachert W, Schutze S, Fritsch J. Selective HSP90beta inhibition results in TNF and TRAIL mediated HIF1alpha degradation. Immunobiology. 2021;226(2):152070.

    Article  CAS  PubMed  Google Scholar 

  308. Sanagawa A, Iwaki S, Asai M, Sakakibara D, Norimoto H, Sobel BE, Fujii S. Sphingosine 1-phosphate induced by hypoxia increases the expression of PAI-1 in HepG2 cells via HIF-1alpha. Mol Med Rep. 2016;14(2):1841–8.

    Article  PubMed  Google Scholar 

  309. Hart PC, Chiyoda T, Liu X, Weigert M, Curtis M, Chiang CY, Loth R, Lastra R, McGregor SM, Locasale JW, Lengyel E, Romero IL. SPHK1 is a novel target of metformin in ovarian cancer. Mol Cancer Res. 2019;17(4):870–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  310. Chawla S, Rahar B, Saxena S. S1P prophylaxis mitigates acute hypobaric hypoxia-induced molecular, biochemical, and metabolic disturbances: a preclinical report. IUBMB Life. 2016;68(5):365–75.

    Article  CAS  PubMed  Google Scholar 

  311. Cuvillier O, Ader I, Bouquerel P, Brizuela L, Gstalder C, Malavaud B. Hypoxia, therapeutic resistance, and sphingosine 1-phosphate. Adv Cancer Res. 2013;117:117–41.

    Article  CAS  PubMed  Google Scholar 

  312. Afsar SY, Alam S, Fernandez Gonzalez C, van Echten-Deckert G. Sphingosine-1-phosphate-lyase deficiency affects glucose metabolism in a way that abets oncogenesis. Mol Oncol. 2022;16(20):3642–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  313. Brune B, Weigert A, Dehne N. Macrophage polarization in the tumor microenvironment. Redox Biol. 2015;5:419.

    Article  PubMed  Google Scholar 

  314. Gstalder C, Ader I, Cuvillier O. FTY720 (Fingolimod) inhibits HIF1 and HIF2 signaling, promotes vascular remodeling, and chemosensitizes in renal cell carcinoma animal model. Mol Cancer Ther. 2016;15(10):2465–74.

    Article  CAS  PubMed  Google Scholar 

  315. Wang D, Wang H, Shi Q, Katkuri S, Walhi W, Desvergne B, Das SK, Dey SK, DuBois RN. Prostaglandin E(2) promotes colorectal adenoma growth via transactivation of the nuclear peroxisome proliferator-activated receptor delta. Cancer Cell. 2004;6(3):285–95.

    Article  CAS  PubMed  Google Scholar 

  316. Wang D, Buchanan FG, Wang H, Dey SK, DuBois RN. Prostaglandin E2 enhances intestinal adenoma growth via activation of the Ras-mitogen-activated protein kinase cascade. Cancer Res. 2005;65(5):1822–9.

    Article  CAS  PubMed  Google Scholar 

  317. Ye Y, Wang X, Jeschke U, von Schonfeldt V. COX-2-PGE(2)-EPs in gynecological cancers. Arch Gynecol Obstet. 2020;301(6):1365–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  318. Ahmadi M, Bekeschus S, Weltmann KD, von Woedtke T, Wende K. Non-steroidal anti-inflammatory drugs: recent advances in the use of synthetic COX-2 inhibitors. RSC Med Chem. 2022;13(5):471–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  319. Kirtonia A, Gala K, Fernandes SG, Pandya G, Pandey AK, Sethi G, Khattar E, Garg M. Repurposing of drugs: an attractive pharmacological strategy for cancer therapeutics. Semin Cancer Biol. 2021;68:258–78.

    Article  CAS  PubMed  Google Scholar 

  320. Totzke G, Schulze-Osthoff K, Janicke RU. Cyclooxygenase-2 (COX-2) inhibitors sensitize tumor cells specifically to death receptor-induced apoptosis independently of COX-2 inhibition. Oncogene. 2003;22(39):8021–30.

    Article  PubMed  Google Scholar 

  321. Habrukowich C, Han DK, Le A, Rezaul K, Pan W, Ghosh M, Li Z, Dodge-Kafka K, Jiang X, Bittman R, Hla T. Sphingosine interaction with acidic leucine-rich nuclear phosphoprotein-32A (ANP32A) regulates PP2A activity and cyclooxygenase (COX)-2 expression in human endothelial cells. J Biol Chem. 2010;285(35):26825–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  322. Furuya H, Tamashiro PM, Shimizu Y, Iino K, Peres R, Chen R, Sun Y, Hannun YA, Obeid LM, Kawamori T. Sphingosine Kinase 1 expression in peritoneal macrophages is required for colon carcinogenesis. Carcinogenesis. 2017;38(12):1218–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  323. 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(2):405–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  324. Pettus BJ, Kitatani K, Chalfant CE, Taha TA, Kawamori T, Bielawski J, Obeid LM, Hannun YA. The coordination of prostaglandin E2 production by sphingosine-1-phosphate and ceramide-1-phosphate. Mol Pharmacol. 2005;68(2):330–5.

    Article  CAS  PubMed  Google Scholar 

  325. Sharma AK, Sk UH, Gimbor MA, Hengst JA, Wang X, Yun J, Amin S. Synthesis and bioactivity of sphingosine kinase inhibitors and their novel aspirinyl conjugated analogs. Eur J Med Chem. 2010;45(9):4149–56.

    Article  CAS  PubMed  Google Scholar 

  326. Cheng JC, Chang HM, Liu PP, Leung PC. Sphingosine-1-phosphate induces COX-2 expression and PGE2 production in human granulosa cells through a S1P1/3-mediated YAP signaling. Cell Signal. 2016;28(6):643–51.

    Article  CAS  PubMed  Google Scholar 

  327. Hanada M, Feng J, Hemmings BA. Structure, regulation and function of PKB/AKT–a major therapeutic target. Biochim Biophys Acta. 2004;1697(1–2):3–16.

    Article  CAS  PubMed  Google Scholar 

  328. Madhunapantula SV, Mosca PJ, Robertson GP. The Akt signaling pathway: an emerging therapeutic target in malignant melanoma. Cancer Biol Ther. 2011;12(12):1032–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  329. Ruggero D, Sonenberg N. The Akt of translational control. Oncogene. 2005;24(50):7426–34.

    Article  CAS  PubMed  Google Scholar 

  330. Cao W, Ma SL, Tang J, Shi J, Lu Y. A combined treatment TNF-alpha/doxorubicin alleviates the resistance of MCF-7/Adr cells to cytotoxic treatment. Biochim Biophys Acta. 2006;1763(2):182–7.

    Article  CAS  PubMed  Google Scholar 

  331. Lu D, Huang J, Basu A. Protein kinase Cepsilon activates protein kinase B/Akt via DNA-PK to protect against tumor necrosis factor-alpha-induced cell death. J Biol Chem. 2006;281(32):22799–807.

    Article  CAS  PubMed  Google Scholar 

  332. Okhrimenko H, Lu W, Xiang C, Hamburger N, Kazimirsky G, Brodie C. Protein kinase C-epsilon regulates the apoptosis and survival of glioma cells. Cancer Res. 2005;65(16):7301–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  333. Kotelevets N, Fabbro D, Huwiler A, Zangemeister-Wittke U. Targeting sphingosine kinase 1 in carcinoma cells decreases proliferation and survival by compromising PKC activity and cytokinesis. PLoS One. 2012;7(6):e39209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  334. Liu Z, Yang H, Zhi L, Xue H, Lu Z, Zhao Y, Cui L, Liu T, Ren S, He P, Liu Y, Zhang Y. Sphingosine 1-phosphate stimulates insulin secretion and improves cell survival by blocking voltage-dependent K(+) channels in beta cells. Front Pharmacol. 2021;12:683674.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  335. Thompson B, Ancellin N, Fernandez SM, Hla T, Sha’afi RI. Protein kinase Calpha and sphingosine 1-phosphate-dependent signaling in endothelial cell. Prostaglandins Other Lipid Mediat. 2006;80(1–2):15–27.

    Article  CAS  PubMed  Google Scholar 

  336. Doll F, Pfeilschifter J, Huwiler A. The epidermal growth factor stimulates sphingosine kinase-1 expression and activity in the human mammary carcinoma cell line MCF7. Biochim Biophys Acta. 2005;1738(1–3):72–81.

    Article  PubMed  Google Scholar 

  337. Means CK, Xiao CY, Li Z, Zhang T, Omens JH, Ishii I, Chun J, Brown JH. Sphingosine 1-phosphate S1P2 and S1P3 receptor-mediated Akt activation protects against in vivo myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2007;292(6):H2944–51.

    Article  CAS  PubMed  Google Scholar 

  338. Fieber CB, Eldridge J, Taha TA, Obeid LM, Muise-Helmericks RC. Modulation of total Akt kinase by increased expression of a single isoform: requirement of the sphingosine-1-phosphate receptor, Edg3/S1P3, for the VEGF-dependent expression of Akt3 in primary endothelial cells. Exp Cell Res. 2006;312(7):1164–73.

    Article  CAS  PubMed  Google Scholar 

  339. You T, Tang H, Wu W, Gao J, Li X, Li N, Xu X, Xing J, Ge H, Xiao Y, Guo J, Wu B, Li X, Zhou L, Zhao L, Bai C, Han Q, Sun Z, Zhao RC. POSTN secretion by extracellular matrix cancer-associated fibroblasts (eCAFs) correlates with poor ICB response via macrophage chemotaxis activation of Akt signaling pathway in gastric cancer. Aging Dis. 2023;14(6):2177–92.

    Article  PubMed  PubMed Central  Google Scholar 

  340. Baudhuin LM, Jiang Y, Zaslavsky A, Ishii I, Chun J, Xu Y. S1P3-mediated Akt activation and cross-talk with platelet-derived growth factor receptor (PDGFR). FASEB J. 2004;18(2):341–3.

    Article  CAS  PubMed  Google Scholar 

  341. Sanchez T, Skoura A, Wu MT, Casserly B, Harrington EO, Hla T. Induction of vascular permeability by the sphingosine-1-phosphate receptor-2 (S1P2R) and its downstream effectors ROCK and PTEN. Arterioscler Thromb Vasc Biol. 2007;27(6):1312–8.

    Article  CAS  PubMed  Google Scholar 

  342. Bourbon NA, Yun J, Berkey D, Wang Y, Kester M. Inhibitory actions of ceramide upon PKC-epsilon/ERK interactions. Am J Physiol Cell Physiol. 2001;280(6):C1403–11.

    Article  CAS  PubMed  Google Scholar 

  343. Dobrowsky RT, Kamibayashi C, Mumby MC, Hannun YA. Ceramide activates heterotrimeric protein phosphatase 2A. J Biol Chem. 1993;268(21):15523–30.

    Article  CAS  PubMed  Google Scholar 

  344. Fox TE, Houck KL, O’Neill SM, Nagarajan M, Stover TC, Pomianowski PT, Unal O, Yun JK, Naides SJ, Kester M. Ceramide recruits and activates protein kinase C zeta (PKC zeta) within structured membrane microdomains. J Biol Chem. 2007;282(17):12450–7.

    Article  CAS  PubMed  Google Scholar 

  345. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124(3):471–84.

    Article  CAS  PubMed  Google Scholar 

  346. Daido S, Kanzawa T, Yamamoto A, Takeuchi H, Kondo Y, Kondo S. Pivotal role of the cell death factor BNIP3 in ceramide-induced autophagic cell death in malignant glioma cells. Cancer Res. 2004;64(12):4286–93.

    Article  CAS  PubMed  Google Scholar 

  347. Patschan S, Chen J, Polotskaia A, Mendelev N, Cheng J, Patschan D, Goligorsky MS. Lipid mediators of autophagy in stress-induced premature senescence of endothelial cells. Am J Physiol Heart Circ Physiol. 2008;294(3):H1119–29.

    Article  CAS  PubMed  Google Scholar 

  348. Scarlatti F, Bauvy C, Ventruti A, Sala G, Cluzeaud F, Vandewalle A, Ghidoni R, Codogno P. Ceramide-mediated macroautophagy involves inhibition of protein kinase B and up-regulation of beclin 1. J Biol Chem. 2004;279(18):18384–91.

    Article  CAS  PubMed  Google Scholar 

  349. Lavieu G, Scarlatti F, Sala G, Carpentier S, Levade T, Ghidoni R, Botti J, Codogno P. Regulation of autophagy by sphingosine kinase 1 and its role in cell survival during nutrient starvation. J Biol Chem. 2006;281(13):8518–27.

    Article  CAS  PubMed  Google Scholar 

  350. Boya P, Gonzalez-Polo RA, Casares N, Perfettini JL, Dessen P, Larochette N, Metivier D, Meley D, Souquere S, Yoshimori T, Pierron G, Codogno P, Kroemer G. Inhibition of macroautophagy triggers apoptosis. Mol Cell Biol. 2005;25(3):1025–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  351. Tang L, Zhang H, Zhou F, Wei Q, Du M, Wu J, Li C, Luo W, Zhou J, Wang X, Chen Z, Zhang Y, Huang Z, Wu Z, Wen Y, Jiang H, Liao D, Kou H, Xiong W, Mei H, Hu Y. Targeting autophagy overcomes cancer-intrinsic resistance to CAR-T immunotherapy in B-cell malignancies. Cancer Commun (Lond). 2024;44(3):408–32.

    Article  PubMed  Google Scholar 

  352. Garg R, Blando J, Perez CJ, Wang H, Benavides FJ, Kazanietz MG. Activation of nuclear factor kappaB (NF-kappaB) in prostate cancer is mediated by protein kinase C epsilon (PKCepsilon). J Biol Chem. 2012;287(44):37570–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  353. Wang H, Gutierrez-Uzquiza A, Garg R, Barrio-Real L, Abera MB, Lopez-Haber C, Rosemblit C, Lu H, Abba M, Kazanietz MG. Transcriptional regulation of oncogenic protein kinase Cϵ (PKCϵ) by STAT1 and Sp1 proteins. J Biol Chem. 2014;289(28):19823–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  354. Salonen T, Sareila O, Jalonen U, Kankaanranta H, Tuominen R, Moilanen E. Inhibition of classical PKC isoenzymes downregulates STAT1 activation and iNOS expression in LPS-treated murine J774 macrophages. Br J Pharmacol. 2006;147(7):790–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  355. Wong SHM, Kong WY, Fang CM, Loh HS, Chuah LH, Abdullah S, Ngai SC. The TRAIL to cancer therapy: hindrances and potential solutions. Crit Rev Oncol Hematol. 2019;143:81–94.

    Article  PubMed  Google Scholar 

  356. Sestili P, Fimognari C. Cytotoxic and antitumor activity of sulforaphane: the role of reactive oxygen species. Biomed Res Int. 2015;2015:402386.

    Article  PubMed  PubMed Central  Google Scholar 

  357. Yang L, Palliyaguru DL, Kensler TW. Frugal chemoprevention: targeting Nrf2 with foods rich in sulforaphane. Semin Oncol. 2016;43(1):146–53.

    Article  CAS  PubMed  Google Scholar 

  358. Neganova M, Liu J, Aleksandrova Y, Klochkov S, Fan R. Therapeutic influence on important targets associated with chronic inflammation and oxidative stress in cancer treatment. Cancers (Basel). 2021;13(23):6062.

    Article  CAS  PubMed  Google Scholar 

  359. Kaiser AE, Baniasadi M, Giansiracusa D, Giansiracusa M, Garcia M, Fryda Z, Wong TL, Bishayee A. Sulforaphane: a broccoli bioactive phytocompound with cancer preventive potential. Cancers (Basel). 2021;13(19):4796.

    Article  CAS  PubMed  Google Scholar 

  360. Wang Y, Mandal AK, Son YO, Pratheeshkumar P, Wise JTF, Wang L, Zhang Z, Shi X, Chen Z. Roles of ROS, Nrf2, and autophagy in cadmium-carcinogenesis and its prevention by sulforaphane. Toxicol Appl Pharmacol. 2018;353:23–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  361. Li J, Xie S, Teng W. Sulforaphane attenuates nonalcoholic fatty liver disease by inhibiting hepatic steatosis and apoptosis. Nutrients. 2021;14(1):76.

    Article  PubMed  PubMed Central  Google Scholar 

  362. Colombo E, Bassani C, De Angelis A, Ruffini F, Ottoboni L, Comi G, Martino G, Farina C. Siponimod (BAF312) activates Nrf2 while hampering NFkappaB in human astrocytes, and protects from astrocyte-induced neurodegeneration. Front Immunol. 2020;11:635.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  363. Spampinato SF, Costantino G, Merlo S, Canonico PL, Sortino MA. Microglia contributes to BAF-312 effects on blood-brain barrier stability. Biomolecules. 2022;12(9):1174.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  364. Jayasuriya R, Dhamodharan U, Ali D, Ganesan K, Xu B, Ramkumar KM. Targeting Nrf2/Keap1 signaling pathway by bioactive natural agents: possible therapeutic strategy to combat liver disease. Phytomedicine. 2021;92:153755.

    Article  CAS  PubMed  Google Scholar 

  365. Cuevas-Cianca SI, Romero-Castillo C, Galvez-Romero JL, Juarez ZN, Hernandez LR. Antioxidant and anti-inflammatory compounds from edible plants with anti-cancer activity and their potential use as drugs. Molecules. 2023;28(3):1488.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  366. Imran M, Aslam Gondal T, Atif M, Shahbaz M, Batool Qaisarani T, Hanif Mughal M, Salehi B, Martorell M, Sharifi-Rad J. Apigenin as an anticancer agent. Phytother Res. 2020;34(8):1812–28.

    Article  CAS  PubMed  Google Scholar 

  367. Farah M, Parhar K, Moussavi M, Eivemark S, Salh B. 5,6-Dichloro-ribifuranosylbenzimidazole- and apigenin-induced sensitization of colon cancer cells to TNF-alpha-mediated apoptosis. Am J Physiol Gastrointest Liver Physiol. 2003;285(5):G919–28.

    Article  CAS  PubMed  Google Scholar 

  368. Khan TH, Sultana S. Apigenin induces apoptosis in Hep G2 cells: possible role of TNF-alpha and IFN-gamma. Toxicology. 2006;217(2–3):206–12.

    Article  CAS  PubMed  Google Scholar 

  369. Oishi M, Iizumi Y, Taniguchi T, Goi W, Miki T, Sakai T. Apigenin sensitizes prostate cancer cells to Apo2L/TRAIL by targeting adenine nucleotide translocase-2. PLoS One. 2013;8(2):e55922.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  370. Kang CH, Molagoda IMN, Choi YH, Park C, Moon DO, Kim GY. Apigenin promotes TRAIL-mediated apoptosis regardless of ROS generation. Food Chem Toxicol. 2018;111:623–30.

    Article  CAS  PubMed  Google Scholar 

  371. Kim EY, Kim AK. Apigenin sensitizes Huh-7 human hepatocellular carcinoma cells to TRAIL-induced apoptosis. Biomol Ther (Seoul). 2012;20(1):62–7.

    Article  CAS  PubMed  Google Scholar 

  372. Voss OH, Arango D, Tossey JC, Villalona Calero MA, Doseff AI. Splicing reprogramming of TRAIL/DISC-components sensitizes lung cancer cells to TRAIL-mediated apoptosis. Cell Death Dis. 2021;12(4):287.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  373. Wu DG, Yu P, Li JW, Jiang P, Sun J, Wang HZ, Zhang LD, Wen MB, Bie P. Apigenin potentiates the growth inhibitory effects by IKK-beta-mediated NF-kappaB activation in pancreatic cancer cells. Toxicol Lett. 2014;224(1):157–64.

    Article  CAS  PubMed  Google Scholar 

  374. Kowalski J, Samojedny A, Paul M, Pietsz G, Wilczok T. Effect of apigenin, kaempferol and resveratrol on the expression of interleukin-1beta and tumor necrosis factor-alpha genes in J774.2 macrophages. Pharmacol Rep. 2005;57(3):390–4.

    CAS  PubMed  Google Scholar 

  375. Xi X, Wang J, Qin Y, You Y, Huang W, Zhan J. The biphasic effect of flavonoids on oxidative stress and cell proliferation in breast cancer cells. Antioxidants (Basel). 2022;11(4):622.

    Article  CAS  PubMed  Google Scholar 

  376. Warat M, Szliszka E, Korzonek-Szlacheta I, Krol W, Czuba ZP. Chrysin, apigenin and acacetin inhibit tumor necrosis factor-related apoptosis-inducing ligand receptor-1 (TRAIL-R1) on activated RAW264.7 macrophages. Int J Mol Sci. 2014;15(7):11510–22.

    Article  PubMed  PubMed Central  Google Scholar 

  377. Negri A, Naponelli V, Rizzi F, Bettuzzi S. Molecular targets of epigallocatechin-Gallate (EGCG): a special focus on signal transduction and cancer. Nutrients. 2018;10(12):1936.

    Article  PubMed  PubMed Central  Google Scholar 

  378. Fulda S, Debatin KM. Resveratrol-mediated sensitisation to TRAIL-induced apoptosis depends on death receptor and mitochondrial signalling. Eur J Cancer. 2005;41(5):786–98.

    Article  CAS  PubMed  Google Scholar 

  379. Ko JH, Sethi G, Um JY, Shanmugam MK, Arfuso F, Kumar AP, Bishayee A, Ahn KS. The role of resveratrol in cancer therapy. Int J Mol Sci. 2017;18(12):2589.

    Article  PubMed  PubMed Central  Google Scholar 

  380. Momchilova A, Pankov R, Staneva G, Pankov S, Krastev P, Vassileva E, Hazarosova R, Krastev N, Robev B, Nikolova B, Pinkas A. Resveratrol affects sphingolipid metabolism in A549 lung adenocarcinoma cells. Int J Mol Sci. 2022;23(18):10870.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  381. Ma Y, Liu G, Tang M, Fang J, Jiang H. Epigallocatechin gallate can protect mice from acute stress induced by LPS while stabilizing gut microbes and serum metabolites levels. Front Immunol. 2021;12:640305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  382. Momchilova A, Nikolaev G, Pankov S, Vassileva E, Krastev N, Robev B, Krastev D, Pinkas A, Pankov R. Effect of quercetin and fingolimod, alone or in combination, on the sphingolipid metabolism in HepG2 cells. Int J Mol Sci. 2022;23(22):13916.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  383. Fischer R, Kontermann RE, Pfizenmaier K. Selective targeting of TNF receptors as a novel therapeutic approach. Front Cell Dev Biol. 2020;8:401.

    Article  PubMed  PubMed Central  Google Scholar 

  384. Papadia F, Basso V, Patuzzo R, Maurichi A, Di Florio A, Zardi L, Ventura E, Gonzalez-Iglesias R, Lovato V, Giovannoni L, Tasciotti A, Neri D, Santinami M, Menssen HD, De Cian F. Isolated limb perfusion with the tumor-targeting human monoclonal antibody-cytokine fusion protein L19-TNF plus melphalan and mild hyperthermia in patients with locally advanced extremity melanoma. J Surg Oncol. 2013;107(2):173–9.

    Article  CAS  PubMed  Google Scholar 

  385. Moradi Marjaneh R, Hassanian SM, Ghobadi N, Ferns GA, Karimi A, Jazayeri MH, Nasiri M, Avan A, Khazaei M. Targeting the death receptor signaling pathway as a potential therapeutic target in the treatment of colorectal cancer. J Cell Physiol. 2018;233(10):6538–49.

    Article  CAS  PubMed  Google Scholar 

  386. Cloughesy TF, Brenner A, de Groot JF, Butowski NA, Zach L, Campian JL, Ellingson BM, Freedman LS, Cohen YC, Lowenton-Spier N, Rachmilewitz Minei T, Fain Shmueli S, Investigators GS, Wen PY. A randomized controlled phase III study of VB-111 combined with bevacizumab vs bevacizumab monotherapy in patients with recurrent glioblastoma (GLOBE). Neuro Oncol. 2020;22(5):705–17.

    Article  CAS  PubMed  Google Scholar 

  387. Brenner AJ, Peters KB, Vredenburgh J, Bokstein F, Blumenthal DT, Yust-Katz S, Peretz I, Oberman B, Freedman LS, Ellingson BM, Cloughesy TF, Sher N, Cohen YC, Lowenton-Spier N, Rachmilewitz Minei T, Yakov N, Mendel I, Breitbart E, Wen PY. Safety and efficacy of VB-111, an anticancer gene therapy, in patients with recurrent glioblastoma: results of a phase I/II study. Neuro Oncol. 2020;22(5):694–704.

    Article  CAS  PubMed  Google Scholar 

  388. Hirpara JL, Subramaniam K, Bellot G, Qu J, Seah S, Loh T, Tucker-Kellogg L, Clement MV, Pervaiz S. Superoxide induced inhibition of death receptor signaling is mediated via induced expression of apoptosis inhibitory protein cFLIP. Redox Biol. 2020;30:101403.

    Article  CAS  PubMed  Google Scholar 

  389. Foo BJ, Eu JQ, Hirpara JL, Pervaiz S. Interplay between mitochondrial metabolism and cellular redox state dictates cancer cell survival. Oxid Med Cell Longev. 2021;2021:1341604.

    Article  PubMed  PubMed Central  Google Scholar 

  390. Tian L, Ogretmen B, Chung BY, Yu XZ. Sphingolipid metabolism in T cell responses after allogeneic hematopoietic cell transplantation. Front Immunol. 2022;13:904823.

    Article  PubMed  PubMed Central  Google Scholar 

  391. Zeng J, Tan H, Huang B, Zhou Q, Ke Q, Dai Y, Tang J, Xu B, Feng J, Yu L. Lipid metabolism characterization in gastric cancer identifies signatures to predict prognostic and therapeutic responses. Front Genet. 2022;13:959170.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  392. Nuhn L, De Koker S, Van Lint S, Zhong Z, Catani JP, Combes F, Deswarte K, Li Y, Lambrecht BN, Lienenklaus S, Sanders NN, David SA, Tavernier J, De Geest BG. Nanoparticle-conjugate TLR7/8 agonist localized immunotherapy provokes safe antitumoral responses. Adv Mater. 2018;30(45):e1803397.

    Article  PubMed  Google Scholar 

  393. De Miguel D, Gallego-Lleyda A, Martinez-Ara M, Plou J, Anel A, Martinez-Lostao L. Double-edged lipid nanoparticles combining liposome-bound TRAIL and encapsulated doxorubicin showing an extraordinary synergistic pro-apoptotic potential. Cancers (Basel). 2019;11(12):1948.

    Article  PubMed  Google Scholar 

  394. Guo J, Ma S, Mai Y, Gao T, Song Z, Yang J. Combination of a cationic complexes loaded with mRNA and alpha-Galactose ceramide enhances antitumor immunity and affects the tumor immune microenvironment. Int Immunopharmacol. 2022;113(Pt A):109254.

    Article  CAS  PubMed  Google Scholar 

  395. Struckhoff AP, Bittman R, Burow ME, Clejan S, Elliott S, Hammond T, Tang Y, Beckman BS. Novel ceramide analogs as potential chemotherapeutic agents in breast cancer. J Pharmacol Exp Ther. 2004;309(2):523–32.

    Article  CAS  PubMed  Google Scholar 

  396. Ponnapakam AP, Liu J, Bhinge KN, Drew BA, Wang TL, Antoon JW, Nguyen TT, Dupart PS, Wang Y, Zhao M, Liu YY, Foroozesh M, Beckman BS. 3-Ketone-4,6-diene ceramide analogs exclusively induce apoptosis in chemo-resistant cancer cells. Bioorg Med Chem. 2014;22(4):1412–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  397. Elhady SS, Habib ES, Abdelhameed RFA, Goda MS, Hazem RM, Mehanna ET, Helal MA, Hosny KM, Diri RM, Hassanean HA, Ibrahim AK, Eltamany EE, Abdelmohsen UR, Ahmed SA. Anticancer effects of new ceramides isolated from the Red Sea red algae Hypnea musciformis in a model of ehrlich ascites carcinoma: LC-HRMS analysis profile and molecular modeling. Mar Drugs. 2022;20(1):63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  398. Kamensek U, Ursic K, Markelc B, Cemazar M, Setrajcic Dragos V, Sersa G. Mutational burden, MHC-I expression and immune infiltration as limiting factors for in situ vaccination by TNFalpha and IL-12 gene electrotransfer. Bioelectrochemistry. 2021;140:107831.

    Article  CAS  PubMed  Google Scholar 

  399. Kamensek U, Cemazar M, Lampreht Tratar U, Ursic K, Sersa G. Antitumor in situ vaccination effect of TNFalpha and IL-12 plasmid DNA electrotransfer in a murine melanoma model. Cancer Immunol Immunother. 2018;67(5):785–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  400. Zakaria AB, Picaud F, Rattier T, Pudlo M, Dufour F, Saviot L, Chassagnon R, Lherminier J, Gharbi T, Micheau O, Herlem G. Nanovectorization of TRAIL with single wall carbon nanotubes enhances tumor cell killing. Nano Lett. 2015;15(2):891–5.

    Article  CAS  PubMed  Google Scholar 

  401. Hu CW, Chang YC, Liu CH, Yu YA, Mou KY. Development of a TNF-alpha-mediated Trojan Horse for bacteria-based cancer therapy. Mol Ther. 2022;30(7):2522–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  402. Lu W, Wang Y, Zhang Q, Owen S, Green M, Ni T, Edwards M, Li Y, Zhang L, Harris A, Li JL, Jackson DG, Jiang S. TNF-derived peptides inhibit tumour growth and metastasis through cytolytic effects on tumour lymphatics. Clin Exp Immunol. 2019;198(2):198–211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  403. Elia AR, Grioni M, Basso V, Curnis F, Freschi M, Corti A, Mondino A, Bellone M. Targeting tumor vasculature with TNF leads effector T cells to the tumor and enhances therapeutic efficacy of immune checkpoint blockers in combination with adoptive cell therapy. Clin Cancer Res. 2018;24(9):2171–81.

    Article  CAS  PubMed  Google Scholar 

  404. Xia GQ, Lei TR, Yu TB, Zhou PH. Nanocarrier-based activation of necroptotic cell death potentiates cancer immunotherapy. Nanoscale. 2021;13(2):1220–30.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  406. Cao M, Ji C, Zhou Y, Huang W, Ni W, Tong X, Wei JF. Sphingosine kinase inhibitors: a patent review. Int J Mol Med. 2018;41(5):2450–60.

    CAS  PubMed  Google Scholar 

  407. Le DHT, Commandeur U, Steinmetz NF. Presentation and delivery of tumor necrosis factor-related apoptosis-inducing ligand via elongated plant viral nanoparticle enhances antitumor efficacy. ACS Nano. 2019;13(2):2501–10.

    CAS  PubMed  Google Scholar 

  408. Pindiprolu S, Krishnamurthy PT, Dev C, Chintamaneni PK. DR5 antibody conjugated lipid-based nanocarriers of gamma-secretase inhibitor for the treatment of triple negative breast cancer. Chem Phys Lipids. 2021;235:105033.

    Article  CAS  PubMed  Google Scholar 

  409. Saravanakumar K, Jeevithan E, Chelliah R, Kathiresan K, Wen-Hui W, Oh DH, Wang MH. Zinc-chitosan nanoparticles induced apoptosis in human acute T-lymphocyte leukemia through activation of tumor necrosis factor receptor CD95 and apoptosis-related genes. Int J Biol Macromol. 2018;119:1144–53.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We apologize to those authors whose works are not cited due to space constraints. This research was supported by IPAC research topic FFSN-2021-0013.

Funding

The work was supported by the Ministry of Science and Higher Education of the Russian Federation at FRC Kazan Scientific Center (grant No. 075-15-2022-1128 to Dr. Margarita Neganova and Dr. Elena Chugunova) and the National Natural Science Foundation of Henan, China (grant No. 222300420534 to Dr. Junqi Liu).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, O.S., J.L., E.T., R.F., A.B.; methodology/searching, Y.A., J.B., E.C.; software/illustrations, M.N., Y.A., J.B., E.C.; investigation and validation, J.L., M.N., Y.A., E.T., J.B., E.C.; resources, E.T., J.L., M.N., R.F.; data curation, A.B., E.T., R.F., O.S.; writing—original draft preparation, O.S., E.T., M.N.; writing—review and editing, A.B., E.T., J.B., R.F., G.S.; visualization, M.N., Y.A., E.C.; supervision, E.T., A.B., O.S., R.F.; project administration, E.T., A.B., R.F.; funding acquisition, M.N., R.F., J.L. All authors have made a substantial intellectual contribution to this work and approved submission of the manuscript.

Corresponding authors

Correspondence to Olga A. Sukocheva, Anupam Bishayee or Junqi Liu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sukocheva, O.A., Neganova, M.E., Aleksandrova, Y. et al. Signaling controversy and future therapeutical perspectives of targeting sphingolipid network in cancer immune editing and resistance to tumor necrosis factor-α immunotherapy. Cell Commun Signal 22, 251 (2024). https://doi.org/10.1186/s12964-024-01626-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-024-01626-6

Keywords