Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Dec 1.
Published in final edited form as: Adv Biol Regul. 2018 Sep 22;70:51–64. doi: 10.1016/j.jbior.2018.09.013

Sphingolipids in neurodegeneration (with focus on ceramide and S1P)

Guanghu Wang 1, Erhard Bieberich 1
PMCID: PMC6251739  NIHMSID: NIHMS1508650  PMID: 30287225

Abstract

For many decades, research on sphingolipids associated with neurodegenerative disease focused on alterations in glycosphingolipids, particularly glycosylceramides (cerebrosides), sulfatides, and gangliosides. This seemed quite natural since many of these glycolipids are constituents of myelin and accumulated in lipid storage diseases (sphingolipidoses) resulting from enzyme deficiencies in glycolipid metabolism. With the advent of recognizing ceramide and its derivative, sphingosine-1-phosphate (S1P), as key players in lipid cell signaling and regulation of cell death and survival, research focus shifted toward these two sphingolipids. Ceramide and S1P are invoked in a plethora of cell biological processes participating in neurodegeneration such as ER stress, autophagy, dysregulate protein and lipid transport, exosome secretion and neurotoxic protein spreading, neuroinflammation, and mitochondrial dysfunction. Hence, it is timely to discuss various functions of ceramide and S1P in neurodegenerative disease and to define sphingolipid metabolism and cell signaling pathways as potential targets for therapy.

1. Introduction to sphingolipids in neurodegeneration

Most neurodegenerative diseases, regardless of being acute or chronic, are accompanied by alterations of sphingolipid composition in a variety of cell types in the central or peripheral nervous system (CNS or PNS). This is not surprising since sphingolipids are important cell signaling molecules in cellular membranes and susceptible to enzymatic hydrolysis. However, for many of these sphingolipid alterations, profound questions remain unanswered. Firstly, because most sphingolipids are precursors as well as derivatives of other sphingolipids, it is often unclear which specific sphingolipid is associated with a disease and whether it is causative or a byproduct in the disease process. Secondly, molecular and cellular targets and mechanisms affected in the disease process are often unclear, ranging from individual proteins to alterations of the membrane structure in specific organelles, from apoptosis to sphingolipid-induced protein aggregation and oxidative stress. Thirdly, unless neurodegeneration emerges from enzyme deficiencies in sphingolipid metabolism, the immediate cause for alterations of the sphingolipid composition is often unknown. Enzymes generating ceramides such as ceramide synthases (CerSs) or sphingomyelinases (SMases) may be up-regulated, while those turning over ceramide (mainly ceramidases) may be down-regulated. Other mechanisms such as aberrant transport of sphingolipids by proteins (e.g., ceramide transport protein or CERT) or vesicles (e.g., endosomes) may also lead to dysregulated sphingolipid metabolism in neurodegenerative disease. On the other hand, since any alteration of the sphingolipid composition results from an enzymatic reaction, either of the enzyme generating or turning over a particular sphingolipid, drug targets are easier to define and offer promising therapeutic approaches.

This review will first discuss sphingolipid species that are up- or downregulated during neurodegenerative diseases. Emphasis will be on the rheostat of ceramide and sphingosine-1-phosphate (S1P) in regulation of neuronal cell survival and apoptosis. Then, we will discuss pathophysiological mechanisms and sphingolipid targets with focus on cellular transport processes and mitochondrial dysfunction. Finally, we will discuss neurodegeneration as a systemic disease involving inflammatory and immune processes regulated (and dysregulated) by sphingolipids. In all of these aspects, we approach sphingolipid metabolism and neuronal health as an integrative process and emphasize shared mechanisms. In the case of ceramide, several potential mechanisms shared between individual neurodegenerative diseases will be discussed. Therefore, we will not dedicate separate sections to individual diseases. However, in the case of S1P, common pathogenic mechanisms are less clear and we will discuss the role of S1P separately and for individual diseases. If we do not go into further detail for other sphingolipids and neurodegenerative diseases, we will provide references for excellent reviews on the topics discussed.

2. Sphingolipid levels in nervous system injuries and neurodegenerative diseases

Neurodegeneration is associated with alteration of sphingolipid metabolism and composition. In acute injuries such as stroke, concussion, spinal cord injury (SCI), or traumatic brain injury (TBI), activation of SMases and glycosidases and upregulation of ceramide and glycolipid levels are among the first responses of the injured tissue (Abe et al., 2018; Barbacci et al., 2017; Brunkhorst et al., 2015; Gu et al., 2013; Horres and Hannun, 2012; Jones and Ren, 2016; Kubota et al., 1989; Novgorodov and Gudz, 2011; Ong et al., 2015; Roux et al., 2016; Sajja et al., 2018). C18:0 and C18:1 ceramide, typical neuronal ceramides synthesized by CerS1, are dysregulated within 24 h after the insult and can be used as biomarkers for TBI (Sajja et al., 2018). Despite efforts to use S1P receptor agonists to protect neurons from injury, there is no information on actual S1P levels during TBI, SCI, or stroke available as of today. Following injury, in a delayed and prolonged response, the immune system is activated and antibodies against myelin glycolipids are generated. For example, anti-galactosylceramide antibodies are detectable for more than 40 years after the injury (Taranova et al., 1992). Despite this breadth of impressive data, it is still not known how alterations in sphingolipid composition affect the pathophysiology of injury and stroke.

Loss of function experiments using mice with deficient SMases, glycosphingolipid glycosidases, or CerS exposed to TBI, SCI, or stroke are rare. Only knockout mice for acid SMase, and SK1 and 2 were tested as critical players or drug targets in acute brain injury. For example, SK2 was shown to be critical for ischemic preconditioning to protect against stroke (Song et al., 2017; Song et al., 2018). Apart from genetic enzyme deficiencies, inhibitors specific for enzymes in sphingolipid metabolism were used to test the effect of sphingolipids on the course of acute brain or spinal cord injuries. Among those, imipramine, a tricyclic antidepressant and functional inhibitor of acid SMase showed success in TBI therapy (Han et al., 2011). However, this type of inhibitor is not specific for acid SMase and beneficial outcomes of inhibiting ceramide generation in acute brain injury are still debated. Probably, most promising are results to ameliorate SCI using inhibitors of ceramide generation, although the mechanism by which sphingolipids may prevent or cure injuries of the spinal cord is not clear. It is also unclear, which injury-related triggers activate enzymes that generate ceramide.

In contrast to acute injuries, the functional significance for dysregulated sphingolipids in chronic neurodegeneration is more thoroughly investigated. Lysosomal storage disorders or sphingolipidoses such as Gaucher (β-glucosylceramidase deficiency), Krabbe (β-galactosylceramidase deficiency), Niemann Pick type 1 (acid SMase (aSMase) deficiency), Fabry (α-galactosidase deficiency), Tay Sachs (α-hexosaminidase deficiency), and metachromatic leukodystrophy (arylsulfatase deficiency deficiency) are caused by genetically inherited enzyme deficiencies in the hydrolytic degradation of glycosphingolipids and often involve chronic neurodegeneration as a fatal outcome of a multi-symptomatic disease process (Arenz, 2017; Eckhardt, 2010; Farfel-Becker et al., 2014; Fuller and Futerman, 2018; Grassi et al., 2018; Haughey, 2010; Sandhoff, 2016; Spassieva and Bieberich, 2016). Neurotoxicity involves accumulation of glycosylated and non-glycosylated sphingolipids or lipid byproducts, which provided the first clear evidence that dysregulation of sphingolipid metabolism leads to neurodegeneration (Ariga et al., 1998; Ariga et al., 2008; Di Pardo and Maglione, 2018b; Fuller and Futerman, 2018; Grassi et al., 2018; Mencarelli and Martinez-Martinez, 2013; Mielke and Lyketsos, 2010; Schnaar, 2016; Yu et al., 2012). However, for many of these diseases the mechanism underlying neurotoxicity is not clear and hypotheses range from lysosomal dysfunction, dysregulation of mitochondria and autophagy, to accumulation of highly neurotoxic metabolic byproducts such as lysophingolipids (Audano et al., 2018; Folts et al., 2016; Piccinini et al., 2010; Seranova et al., 2017; Spassieva and Bieberich, 2016; Sural-Fehr and Bongarzone, 2016). Because impaired or loss of enzyme activity in sphingolipid degradation is the root cause of the disease, treatment approaches encompass enzyme replacement therapy or pharmacological inhibitors of enzymes in the generation of the otherwise accumulated lipids. Apart from neurodegeneration resulting from sphingolipidoses, there are diseases that are not primarily caused by mutations of enzymes in sphingolipid metabolism, but may affect sphingolipid composition and levels. Diseases with documented elevation of ceramide are Multiple Sclerosis (MS), AD, and Parkinson’s disease (PD). Alterations of S1P levels vary in these diseases and will be discussed in separate sections on S1P and neurodegenerative disease.

MS

In MS, the appearance of aberrant sphingolipid composition is not surprising since demyelination releases a variety of myelin sphingolipids and their degradation products into the brain and blood stream. However, the specific function of individual sphingolipids such as ceramide was only recently investigated and suggested new therapeutic strategies. Since MS is an autoimmune disease, a central nervous and peripheral component has to be considered. The peripheral component relies on the ability of the immune system to mobilize leukocytes that attack the CNS. It was found that in the experimental autoimmune encephalomyelitis (EAE) model, egress of leukocytes from lymph nodes and the autoimmune reaction(Eberle et al., 2014) is suppressed in CerS2 knockout mice, while it is enhanced in CerS6-deficient mice (Barthelmes et al., 2015; Schiffmann et al., 2012). CerS2 specifically catalyzes biosynthesis of very long chain (C24:0, C24:1) ceramides, while CerS6 mainly generates C16 ceramide (Levy and Futerman, 2010; Park et al., 2014; Stiban et al., 2010), suggesting a pro-inflammatory role for C24- and anti-inflammatory role for C16 ceramide. Prior to these exciting observations, it was shown that downregulation of S1P receptor 1 (S1P1) by the S1P precursor analog fingolimod (FTY720) also suppresses leukocyte egress and ameliorates MS, which led to a clinically approved MS therapy (see section on S1P). Hence, there appears to be an interplay of specific ceramides and S1P in the autoimmune response of MS, probably on the level of S1P receptor activation or turnover. In addition to these peripheral effects, knockout or inhibition of aSMase was shown to ameliorate MS by preventing apoptosis of oligodendrocytes, the cells generating myelin in the brain (Chami et al., 2017; Jana and Pahan, 2010). Hence targeting sphingolipid metabolism is a valid strategy for MS therapy.

AD and PD

Similar to MS, progression of other neurodegenerative diseases includes a CNS and a peripheral (immunological) component. In AD and PD, elevation of ceramide is often associated with neuronal cell death and often accompanied by generation of antibodies against specific sphingolipids indicative of activation of the immune system. Our group has made several discoveries showing involvement of ceramide in neuronal cell death, as well as astrocyte malfunction and immune response toward ceramide during progression of AD (Bieberich et al., 2000; Bieberich et al., 2003; Bieberich et al., 2001; Dinkins et al., 2014; Dinkins et al., 2016a; Kong et al., 2018; Wang et al., 2012; Wang et al., 2008b). More than 95% of AD cases do not rely of mutations in amyloid precursor protein (APP) or proteases processing APP to neurotoxic amyloid beta (Aβ) peptide (Sanabria-Castro et al., 2017; Tomita, 2017). These non-familial and late-onset cases of AD are of unknown etiology. In a proportion of these cases, genetic risk factors such as ApoE4 or TREM2 were identified, but exact mechanisms how these factors contribute to AD are not clear. One factor that clearly contributes to AD in any of these cases is aging. About 50 years ago, it was shown that in the gray matter of the aging brain, the level of cerebrosides continuously increases, which is further elevated in AD (Suzuki et al., 1965). Sulfatides, the derivatives of cerebrosides, are reduced in AD, suggesting that progression of the disease is associated with dysregulated sphingolipid metabolism (Han et al., 2002). While glycolipids of the aging and diseased brain were thoroughly analyzed from early on, it took several decades before ceramide became focus of aging and AD research, probably due to studies showing that it is a pro-apoptotic lipid in neurons (Brugg et al., 1996). Application of newly developed mass spectrometric methods, particularly liquid chromatography-tandem mass spectrometry (LC-MS/MS) showed that during aging and more so in AD, ceramide levels are increased in brain, cerebrospinal fluid (CSF), and serum (Kim et al., 2017; Mielke et al., 2008; Mielke et al., 2012; Mielke et al., 2010; Satoi et al., 2005). Of particular interest is the rheostat of ceramide and its derivative S1P in the regulation of neuronal cell survival and cell death in neurodegeneration.

3. Ceramide and S1P rheostat in neurodegeneration

Discovery of the cell signaling function of ceramide and S1P was intimately linked to knowledge on their metabolism. About 30 years ago, several seminal studies showed that lysosphingolipids and degradation products of sphingomyelin such as ceramide and sphingosine, can inhibit protein kinase C and are metabolically regulated by what was termed the “sphingomyelin cycle” (Hannun and Bell, 1993; Hannun et al., 1986; Okazaki et al., 1989; Okazaki et al., 1990). It was also found that acid and neutral SMase were activated by cytokines such as TNF-α, Fas-ligand, or interleukins, radiation, and intracellular cell signaling factors arresting the cell cycle or inducing apoptosis, a discovery that soon led to a wealth of studies investigating the role of ceramide in the regulation of these processes (Clarke et al., 2008; Cutler and Mattson, 2001; Devillard et al., 2010; Farooqui et al., 2010; Haimovitz-Friedman et al., 1994; Hannun, 1996; Kolesnick, 1987; Ohanian and Ohanian, 2001; Testai et al., 2004). Most of these studies were performed in non-neural cells and it took almost another 10 years until the first studies appeared on the role of the sphingomyelin cycle in the regulation of apoptosis in neurons with controversial outcomes (Brugg et al., 1996; Ito and Horigome, 1995). About 25 years ago, the regulation of cell proliferation and survival by sphingolipids was then complemented by S1P, which soon led to the idea of a “ceramide/S1P rheostat” with ceramide being pro- and S1P anti-apoptotic (Cuvillier et al., 1996; Edsall et al., 1997; Gault et al., 2010; Hait et al., 2006; Hannun and Obeid, 2008; Maceyka et al., 2002; Mao and Obeid, 2008; Mao et al., 1997; Olivera et al., 1992; Olivera et al., 1999; Osawa et al., 2005; Spiegel et al., 1998b; Taha et al., 2006). Regulation of cell signaling pathways by sphingolipids is unique in that three lipid signaling molecules, sphingomyelin, ceramide, and S1P, are tightly regulated by signaling factor and stressor response enzymes such CerS, SMases, SKs, ceramidases, and phosphatases. While the anti-apoptotic effect of S1P is mediated by a variety of S1P-specific GPCRs, which implies secretion of S1P to counteract cell death, the precise mechanism of ceramide-induced apoptosis is still debated (see also section on S1P).

More recent studies show that in addition to cell survival and death, ceramide and S1P are invoked in numerous other processes regulating neuronal and glial biology. We and others reported that ceramide is critical for establishing cell polarity and migration of neuroepithelial cells, while S1P promotes oligodendroglial differentiation (Bieberich, 2010, 2011, 2012; Coelho et al., 2010; Jung et al., 2007; Krishnamurthy et al., 2007; Pitson and Pebay, 2009; Wang et al., 2008a; Wang et al., 2018). Other studies showed that ceramide and S1P induce neurite retraction as well as outgrowth depending on the neuronal cell type investigated (Singh and Hall, 2008; Toman et al., 2004). Some of these apparently contradictory effects may emerge from extracellular signaling factors such as TNF-α activating SMases promoting generation of ceramide (e.g., as pro-apoptotic signal) as well as inducing SKs leading to metabolic turnover of ceramide into S1P (e.g., as pro-inflammatory signal) (Pettus et al., 2003; Snider, 2013; Snider et al., 2010). Most of these apparently contradictory effects, however, are likely to rely on a specific composition of target proteins specifically expressed in different types of cells, namely distinct S1P receptors or cell signaling proteins interacting with ceramide. The first proteins shown to interact with ceramide were protein phosphatases and kinases (Bieberich et al., 2000; Bourbon et al., 2000; Carlson and Hart, 1996; Dobrowsky and Hannun, 1992; Hannun, 1996; Igarashi et al., 1990; Lee et al., 1996; Lozano et al., 1994; Merrill et al., 1986; Muller et al., 1995; Wang et al., 2005; Wang et al., 1999). Particularly, ceramide activation of protein phosphatases PP2a and PP1 was reported to be involved in apoptosis induction and regulation of cell growth and cortical actin (Canals et al., 2012; Chalfant et al., 1999; Chalfant et al., 2004; Dobrowsky et al., 1993; Galadari et al., 1998; Mukhopadhyay et al., 2008; Perry et al., 2012). Our group showed that ceramide activation of atypical PKCς/λ (aPKC) promotes cell polarity, neuroprogenitor motility, and ciliogenesis, while expression of the aPKC inhibitor protein prostate apoptosis response 4 (PAR-4) renders ceramide pro-apoptotic (Bieberich, 2011, 2012; Bieberich et al., 2000; He et al., 2012; He et al., 2014; Krishnamurthy et al., 2007; Wang and Bieberich, 2010; Wang et al., 2012; Wang et al., 2009; Wang et al., 2005). More recently, we identified glycogen synthase 3β (GSK3) and tubulin as additional proteins ceramide interacts with (He et al., 2014; Kong et al., 2015; Kong et al., 2018). Our studies suggest that ceramide activation of GSK3 is critical for biogenesis and function of primary and motile cilia in the brain, while ceramide-associated tubulin (CAT) regulates voltage-dependent anion channel 1 (VDAC1), the key ADP/ATP transporter in the outer mitochondrial membrane (OMM). Activation of GSK3 is important for neurodegeneration in that it has been discussed as one of the protein kinase phosphorylating tau, the protein forming neurofibrillary tangles in AD (Drulis-Fajdasz et al., 2018; Takashima, 2006). Ceramide-activated protein kinases such as GSK3 or apoptosis-signal regulating kinase 1 (ASK1) may be critical for the etiology of neurodegenerative disease and suggest that inhibition of ceramide generation (or its interaction with the respective kinases) is a promising new strategy for therapy (Guo et al., 2017). We will devote the next sections to a more detailed discussion on the role of ceramide in neurodegeneration and therapeutic strategies. In addition to ceramide binding activating protein kinases or phosphatases and affecting mitochondrial function in neurodegeneration, ceramide is also important for autophagy and inflammation, two processes critical for neuronal cell survival by preventing intra- and extracellular accumulation of toxic proteins such as parkin or huntingtin and other polyglutamine (polyQ) proteins, and promoting clearance mechanisms such as amyloid or tau uptake and disposal by astrocytes and microglia (Audano et al., 2018; Fujikake et al., 2018; Grosch et al., 2012; Harvald et al., 2015; Jiang and Ogretmen, 2014; Karunakaran and van Echten-Deckert, 2017; Tommasino et al., 2015; van Echten-Deckert and Alam, 2018).

With regard to the regulation by sphingolipids, effects on autophagy and inflammation are critical because they operate at the intersection of several compartments including ER, mitochondria, and lysosomes. There is evidence that the different effects of ceramide on apoptosis, cell growth, polarity, autophagy, and inflammation rely on the chain length (and degree of saturation) of the fatty acid attached to sphingosine (Grosch et al., 2012; Mencarelli and Martinez-Martinez, 2013; Mullen et al., 2011; Park and Park, 2015; Pinto et al., 2011). This appears to be reasonable if associated with protein interaction or membrane structural effects depending on ceramide chain length. While it is not known if ceramide interacts directly with any protein of the inflammasome, it was recently found that ceramide-1phosphate (C1P) transport protein (CPTP) downregulates early autophagy and stimulates inflammasome assembly (Mishra et al., 2018). The exact mechanism by which ceramide regulates autophagy is still unclear, but appears to involve chain length specificity and direct interaction of specific ceramide (C18:0 ceramide) with LC3B, a key regulatory protein of autophagy (Sentelle et al., 2012). It should be noted that C18 (C18:0 and C18:1) ceramide is one of the ceramides elevated in AD and PD, and generated by CerS4, CerS5, and CerS1, a predominantly neuronal CerS (Ben-David and Futerman, 2010; Mielke et al., 2013; Savica et al., 2016; Wang et al., 2012; Xing et al., 2016). In addition to C18 ceramide, the levels of very long chain ceramides such as C24 (C24:0 and C24:1) ceramide, all of which synthesized by CerS2, are increased in AD and are also enriched in extracellular vesicles such as exosomes (Wang et al., 2012). Since CerS-deficient mice were not yet tested after cross-breeding with AD or other neurodegenerative disease models, the specific role of individual CerS or ceramide species in neurodegeneration remains elusive. Even without crossbreeding with disease models, the phenotypes of several CerS knockout mice, particularly for CerS1 and CerS2 invoke neurodegenerative processes. A recent study suggests that Purkinje cell death in CerS1 mice is rather due to elevation of long chain bases such as dihydrosphingosine or sphingosine than loss of specific ceramides (Spassieva et al., 2016). The phenotypes are at least in part consistent with the neurotoxic effect of the fungus toxin fumonsin B1, a specific inhibitor of CerS leading to defects in neural development (Gelineau-van Waes et al., 2005; Gelineau-van Waes et al., 2009; Wang et al., 1991). Of the two knockout mice deficient of CerS generating C16:0 ceramide, CerS5 and 6, only CerS6 knockout mice showed behavioral abnormalities indicating a brain phenotype, although the exact mechanism was not elucidated yet (Ebel et al., 2013; Novgorodov et al., 2011). Probably, some of the brain phenotype is associated with mitochondrial dysfunction in oligodendrocytes caused by CerS6 deficiency (Novgorodov et al., 2011). The difficulty to understand phenotypes of CerS knockout mice results from multiple effects on sphingolipid composition as the consequence of enzyme deficiencies in de novo biosynthesis of ceramide. A potential solution to test the effect of ceramide depletion are knockout mice for enzymes generating ceramide by hydrolysis of a derivative such as sphingomyelin.

Unfortunately, knockout mice for acid SMase and galactosyl- or glucosylceramidase, although clearly showing a neural developmental or neurodegenerative phenotype, are burdened by elevation of multiple sphingolipids, including that of the original substrate and neurotoxic lysosphingolipids (Eckhardt, 2010; Grassi et al., 2018; Haughey, 2010; Horres and Hannun, 2012; Ledesma et al., 2011). On the other hand, deficiency of neutral sphingomyelinase 2 (nSMase2) is a promising model to study loss of function (and dysfunction) of ceramide. We will discuss its phenotypes in the next paragraph on sphingolipids in intracellular transport and mitochondrial dysfunction. In addition to ceramide depletion, knockout mice showing ceramide elevation have been generated. These include deficiencies of enzymes hydrolyzing ceramide (ceramidases) as well as enzymes using ceramide or the product of its hydrolysis, sphingosine, as the substrate (SKs). Among these, knockout of acid ceramidase (ASAH1) causes early embryonic lethality, and cells could not survive beyond the 4 to 8 cell stage due to apoptotic cell death. This is the earliest lethal phenotype observed for a lysosomal hydrolase. Notably, treatment of 2-cell embryos from heterozygous intercrosses with S1P, rescued the ASAH1−/− embryos (Eliyahu et al., 2007). Double knockout of SKs leads to embryonic death at around E12 partly due to increased apoptosis and decrease in mitosis in the developing nervous system (Mizugishi et al., 2005).

4. Ceramide and mitochondrial dysfunction in neurodegeneration

In addition to interfacing extracellular cues and intracellular cell signaling pathways at the plasma membrane, sphingolipids are now recognized as key regulators of cellular transport and organelle function. The effects of sphingolipids on cellular transport encompasses regulation of membrane curvature, fission, and fusion in endocytosis and vesicular transport, as well as vesicular binding to transported proteins and the cytoskeleton (Adada et al., 2014; Boulgaropoulos et al., 2012; Burgert et al., 2017; Chiantia et al., 2008; Draeger and Babiychuk, 2013; Goni et al., 2005; Schneider-Schaulies and Schneider-Schaulies, 2013; Silva et al., 2012; Stoffel et al., 2016; Trajkovic et al., 2008). Mitochondrial function is another process that is regulated and dysregulated by sphingolipids (Chakrabarti et al., 2016; Hernandez-Corbacho et al., 2017; Kong et al., 2018; Law et al., 2017; Perera et al., 2016; Schwartz et al., 2018). Aberrant intra- and extracellular transport as well as mitochondrial dysfunction are gaining importance when looking for the etiology of neurodegeneration beyond protein aggregation and accumulation. There are three stages of cellular transport the dysregulation of which is critical for neurodegeneration and affected by sphingolipids: 1) processing and degradation of proteins along exo- and endocytotic transport pathways, including secretion and uptake of neuro- and gliotoxic proteins; 2) transport of organelles, including mitochondria, signalosomes, and other vesicles; and 3) extra- and intercellular transport, including that of extracellular vesicles recently gaining attention as contributing factor in neurodegeneration. Currently, it is assumed that in most of these transport processes, the lipid determines the fate of a protein, which when dysregulated becomes neurotoxic. However, we will also discuss the possibility that the protein induces dysregulation of sphingolipid metabolism as the neurotoxic trigger.

In AD, PD, and Huntington’s disease, proteins such as APP, Aβ and tau (AD), synuclein and parkin (PD), and huntingtin (HD) are proteolytically processed and secreted or accumulated inside of neurons and glia. Often, proteolytic processing and secretion requires a re-uptake process involving endo- and exocytosis. Twenty years ago, the first evidence was published that ceramide generated by nerve growth factor (NGF)/p75NTR-activated nSMase2 is required for APP processing and secretion, which was then found to involve ceramide-induced upregulation and cerebroside-mediated activation of β-secretase (BACE), the protease generating Aβ from APP in neurons (Costantini et al., 2005; Kalvodova et al., 2005; Ko and Puglielli, 2009; Puglielli et al., 2003; Rossner et al., 1998). However, all of these studies were focused on neuronal APP processing and secretion, while the contribution of sphingolipids in glia remained uninvestigated. In addition, early studies on the function of sphingolipids in neurodegeneration targeted the immediate effects of APP processing such as Aβ secretion and apoptosis induction, while other transport processes and organelles affected by Aβ and sphingolipids were not investigated until recently. Finally, genetic risk factors altering lipid metabolism and transport such as ApoE4 have not been integrated into a consistent model for the effect of sphingolipids on AD yet.

ApoE4, a lipoprotein transporting cholesterol, is a genetic risk factor for late onset AD. While homozygous carriers of ApoE4 (prevalence 2:100) have a >10-fold increased risk of developing AD at the age of 75, it is still unclear how ApoE4 contributes to AD (Liu et al., 2013; Qian et al., 2017; Ward et al., 2012). Interestingly, deficiency of NPC1, a cholesterol transport protein mutated in Niemann Pick type C disease, also increases the risk of developing AD pathology such as amyloid and tau aggregation (Borbon et al., 2012; Borbon and Erickson, 2011; Burns et al., 2003; Saito et al., 2002; Vance et al., 2005). However, NPC1 deficiencies in humans are extremely rare (prevalence 1:150,000) and lead to 50% lethality before the age of 12. Therefore, most of what we know about NPC1 stems from cell culture and animal studies. In NPC1 knockout cells, cholesterol is no longer sequestered to endosomes, but it is accumulated in lysosomes and mitochondria, where is exacerbates Aβ neurotoxicity (Colell et al., 2009; Fernandez et al., 2009). Not only transport and level of cholesterol is dysregulated, but also that of several sphingolipid classes such as sphingosine, ceramide, sphingomyelin, and glycosphingolipids (Fan et al., 2013; Sun et al., 2001). Similar to NPC1 deficiency, ApoE4 homozygosity is associated with increase of ceramide levels in AD brain, although it is not upregulated in ApoE4 normal brain (Bandaru et al., 2009; den Hoedt et al., 2017). While it is not understood how NPC1 and ApoE4 regulate sphingolipid metabolism, one is tempted to speculate that in both cases, aberrant lipid transport and accumulation, particularly of cholesterol and ceramide, leads to neurotoxicity involving Aβ and tau protein.

In many types of neurodegeneration, dysregulated protein and lipid transport in neurons and glia is likely to underlie a shared pathogenic mechanism. Particularly, aberrant traffic of accumulated or aggregated protein and lipid from endosomes to lysosomes and mitochondria has been described for AD, PD, and HD. Once at the target organelle, misdirected proteins and lipids bind to proteins critical for organelle function such as VDAC1 in mitochondria. VDAC1 binds to neurotoxic proteins such as Aβ, tau, parkin, and synuclein, which 1) impairs its transport function for ADP and ATP; 2) prevents its metabolically critical interaction with other proteins such as hexokinase; 3) promotes its interaction with pro-apoptotic proteins such as Bax; and 4) promotes assembly of multimeric VDAC1 pores that allow for the efflux of cytochrome c inducing apoptosis (Caterino et al., 2017; Chu et al., 2014; Geisler et al., 2010; Manczak and Reddy, 2012; Shoshan-Barmatz et al., 2018; Smilansky et al., 2015). As a consequence, neurodegeneration arises from mitotoxicity induced by misdirected protein and probably, lipid transport. The (dys)function of lipids may involve direct effects by binding to VDAC1 or affecting the mitochondrial membrane structure by generating ceramide pores. Lipids may also promote mitotoxic protein interaction with VDAC1 similar to that discovered by our laboratory for ceramide-induced interaction of VDAC1 with tubulin (Kong et al., 2018). The increase of ceramide levels at mitochondria and other organelles may arise from upregulation of endogenous ceramide generation or uptake of ceramide in form of exosomes. Our group has shown that generation of ceramide by nSMase2 activated by Aβ or inflammatory cytokines leads to secretion of ceramide-enriched exosomes from astrocytes (Wang et al., 2012). These “astrosomes” may transport ceramide (and neurotoxic proteins such as Aβ and tau) into other astrocytes and neurons. Our research showed that astrosomes can induce apoptosis in astrocytes, the effect on neurons is currently investigated in our laboratory. It should be noted that exosomes are not generally glio- or neurotoxic, but may also assist Aβ or tau clearance, probably depending on the cell of origin or exosome (lipid) composition (Dinkins et al., 2016b; Yuyama and Igarashi, 2017; Yuyama et al., 2012; Yuyama et al., 2015). Apart from these unknowns, it becomes increasingly clear that the classical view of pathogenic Aβ or tau accumulation in AD will be replaced by a more physiological or system biological view involving metabolism, protein and lipid transport, and interaction of different organelles and cell types. Sphingolipids such as ceramide and S1P may serve as cell signaling hubs or network nodes to regulate physiological as well as pathophysiological mechanisms in AD and other neurodegenerative diseases.

5. Metabolism and signaling pathways of S1P

S1P is another major bioactive sphingolipid that plays essential cell signaling roles in neurodegeneration (Assi et al., 2013; Blaho and Hla, 2014; Hagen et al., 2011; Proia and Hla, 2015; Pyne et al., 2018). It is a soluble lipid generated intracellularly by SK1 and SK2 (Alvarez et al., 2010; Maceyka et al., 2012; Maceyka and Spiegel, 2014; Proia and Hla, 2015). The level of intracellular S1P is orchestrated by several metabolic enzymes and transporter proteins, such as SKs, S1P lyase, S1P phosphatase, ABC transporters, Spns2, and Mfsd2b (Bradley et al., 2014; Kawahara et al., 2009; Klyachkin et al., 2014; Kobayashi et al., 2018; Kunkel et al., 2013; Long et al., 2005; Nagareddy et al., 2014; Proia and Hla, 2015; Sciorra and Morris, 2002; Spiegel and Milstien, 2011; Vu et al., 2017). Although SK1 can be secreted and generate S1P outside of cells (Sciorra and Morris, 2002), the level of extracellular S1P is regulated by exportation by ABC transporters, Spns2, and mfsd2b from red blood cells, activated platelets, and endothelial cells, which is usually found at much higher concentration than that of tissues (Fukuhara et al., 2012; Hisano et al., 2012; Kobayashi et al., 2018; Sciorra and Morris, 2002; Vu et al., 2017). In the brain, S1P regulates survival, proliferation, migration, and inflammation of all the neural cell types, including neurons, astrocytes, microglia, and oligodendrocytes (Huang et al., 2011; Kleger et al., 2007; Laurenzana et al., 2015; Marfia et al., 2014; Smith et al., 2013). However, the function of S1P is cell type- and context- dependent.

Extracellular S1P functions through five cell surface G-protein-coupled-receptors (GPCRs) S1P1–S1P5 (Huang et al., 2011). S1P stimulates various signal transduction pathways in a cell type- and context-specific manner, usually depending on the type and expression level of S1P receptor(s). Different S1P receptors are coupled to different G-proteins and regulate diverse signaling pathways. For example, S1P1 is coupled exclusively via Gi protein to activate Ras, mitogen activated protein kinase (MAPK), PI3K/Akt, and phospholipase C pathways (Huang et al., 2011). There are excellent reviews on S1P receptor signaling pathways, and the readers are encouraged to refer to those (Blaho and Hla, 2014; Kunkel et al., 2013; Proia and Hla, 2015; Spiegel et al., 1998a; Spiegel and Milstien, 2011). On the other hand, intracellular receptor-independent actions of S1P began to emerge in several cell types, including neurons. Inside of cells, S1P regulates the function of several S1P interacting proteins, including HDACs, TRAF2, Hsp90 and HRP94, in different cellular compartment of cells, such as the ER and nuclei (Alvarez et al., 2010; Blaho and Hla, 2014; Hait et al., 2009; Park et al., 2016; Proia and Hla, 2015).

6. Physiological functions of S1P in the CNS

S1P plays an essential role in neural development (Mizugishi et al., 2005). During mouse development, SK1 is highly expressed in the brain, while SK2 levels are increased in the limb buds, eyes, and branchial arches. S1P depletion in SK1/K2-double knockout mice exhibits severe defects in neurogenesis and neural cell survival, accompanied by impaired neural tube closure, increased neural cell apoptosis, and embryonic lethality (Mizugishi et al., 2005). Blockade of S1P signaling in S1P1-null mice shows a very similar neural phenotype, indicating a central function of S1P signaling in neurogenesis and brain development (Blaho and Hla, 2014; Mizugishi et al., 2005; Proia and Hla, 2015).

Besides its role in brain development, S1P is implicated in regulation of neuronal function and survival in adult brains. Convincing evidence indicates that S1P plays a role in presynaptic structure, synaptic strength, and synaptic vesicle availability in hippocampal neurons (Brailoiu et al., 2002; Camoletto et al., 2009; Darios et al., 2009; Kanno et al., 2010; Riganti et al., 2016). In addition, experimental data suggest that S1P promotes pro-survival neuronal autophagy (Moruno Manchon et al., 2015). Expression of SK1 enhances autophagic activity, while S1P lyase reduces the activity (Moruno Manchon et al., 2015; Moruno Manchon et al., 2016). Autophagy is crucial for the health and survival of neurons to get rid of damaged and aggregated proteins and organelles during basal, nutrient depletion, aging, and disease conditions, in particular, to counteract cell death induced by ceramide or other pathogens (Moruno-Manchon et al., 2018; Moruno Manchon et al., 2015; Moruno Manchon et al., 2016).

Recently, the functions of S1P in the immune system and neuroinflammation is gaining attention with respect to neurodegeneration, triggered by the approval of the Relapsing Remitting MS (RR-MS) drug Fingolimod (FTY720), a sphingosine analog and S1P1 functional antagonist. In activated microglia, SK1 expression is upregulated (Nayak et al., 2010). Lipopolysaccharide (LPS) activates the SK1/S1P axis leading to translocation of SK1 to the plasma membrane where it converts its substrate sphingosine to S1P (Fernandez-Pisonero et al., 2012; Hammad et al., 2008). Moreover, suppression of SK1 activity in activated mouse microglia inhibits the expression levels of TNF-??, IL-1??, and iNOS and release of TNF-?? and NO [59]. Exogenous addition of S1P to activated microglia enhances inflammatory responses, suggesting that S1P acts as an upstream factor to induce the production of pro-inflammatory cytokines and neurotoxins such as NO (Assi et al., 2013). A recent study shows that supplementation of S1P to primary cultured microglial cells deprived of oxygen and glucose leads to increased IL-17A expression (Lv et al., 2016). These data suggest that the SK1/S1P pathway is involved in the inflammatory response of activated microglia in an autocrine/paracrine fashion in which the secreted or intracellular S1P can regulate the release of pro-inflammatory factors by microglia.

In astrocytes, SK1 and S1P3 are functionally upregulated under pro-inflammatory conditions. Incubation with IL-1 induces the expression of SK1 and exogenous S1P induces astrogliosis (Paugh et al., 2009; Sorensen et al., 2003). In experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, a subset of astrocytes is activated in clusters that progressively expand along white matter tracts. The extent of astrocyte activation is limited by the loss of astrocytic S1P1 (Choi et al., 2011; Dusaban et al., 2017). Moreover, S1P1 modulation by FTY720 in murine and human astrocytes suppressed neurodegeneration-promoting mechanisms mediated by astrocytes, microglia, and CNS-infiltrating proinflammatory monocytes (Karunakaran and van Echten-Deckert, 2017; Rothhammer et al., 2017).

7. S1P in neurodegeneration

Since it plays a versatile roles in neural development and function, deregulation of S1P metabolism and signaling has increasingly been recognized as an essential player in various neurodegenerative diseases, including AD, PD, and HD. Although widely regarded as an autoimmune disease, MS is also a neurodegenerative disease since chronic inflammation drives severe inflammation and massive neurodegeneration (Carassiti et al., 2018; Chaudhuri, 2013; Compston and Coles, 2008; Friese et al., 2014; Stadelmann, 2011). Since the first clinical use of S1P modulators was approved in MS, we will start from MS to discuss the function of S1P and medical application of drugs targeting S1P metabolism and SIP-associated cell signaling pathways in neurodegenerative diseases.

MS

Fingolimod or FTY720, a small molecule S1P receptor modulator, is the first oral drug approved by the Food and Drug Administration to treat RR-MS in 2010 (Brinkmann et al., 2010; Kappos et al., 2006). FTY720 is phosphorylated to form FTY720-phosphate, which resembles naturally occurring S1P. FTY720-phosphate initially activates S1P1, yet subsequently causes internalization and degradation of S1P1 (Huwiler and Zangemeister-Wittke, 2018). Thus, FTY720 is a functional antagonist of S1P1, the major S1P receptor in many organs, including brain and lymphatic cells (Blaho and Hla, 2014). In RR-MS, FTY720 treatment inhibits S1P1 signaling and prevents lymphocyte egress from the lymphoid organs (Chun and Hartung, 2010; Lee et al., 2010). Although initially attributed to sequestration of lymphocytes as a mechanism of action, more recent research suggested that FTY720 directly acts on the central nervous system (Asle-Rousta et al., 2014; Hunter et al., 2016). For example, FTY720 protects against demyelination-independent of lymphocyte infiltration (Jeffery et al., 2016).

Although effective in reducing and delaying RR-MS, FTY720 has minimal effect on Primary or Secondary progressive MS (PP-MS and SP-MS) (Farez and Correale, 2016). Therefore, many emerging studies and clinical trials with various selective S1P receptor modulators are in progress to test their clinical use in PP-MS and SP-MS (Mao-Draayer et al., 2017).

AD

The function of S1P-signaling in AD conditions is elusive and largely controversial. On the one hand, there are reports showing that the ratio of S1P/sphingosine is reduced in post mortem AD patient brains and hippocampus, suggesting a protective role of S1P-signaling (Ceccom et al., 2014; Couttas et al., 2014; He et al., 2010). Accordingly, the levels of SK1 is reduced and S1P lyase increased in these brains (Ceccom et al., 2014; Couttas et al., 2014; He et al., 2010). On the other hand, there are also reports that support a disease-causative role of S1P (Hagen et al., 2011; Lei et al., 2017; Takasugi et al., 2011). S1P interacts with βsecretase BACE1 in neurons. Inhibition of SKs and overexpression of S1P lyase both decreased BACE1 activity and reduced Aβ generation. These data demonstrate that S1P production and Aβ level are directly correlated. Notably, the relative activity of SK2 is upregulated in the AD patient brains (Takasugi et al., 2011).

Another study showed that neuronal death is closely correlated with increased S1P in primary cultured neurons derived from S1P lyase-deficient mice (Hagen et al., 2011). This same group also reported that S1P lyase deficiency is associated with tau hyperphosphorylation (Hagen et al., 2011). In addition, the level of S1P is increased in the CSF of patients with mild cognitive impairment (MCI) and MCI-AD (AD developed from MCI) when compared to agematched controls; a longitudinal human study further supporting that S1P plays a major role in the initiation and/or progression of AD. These studies provide elegant molecular and mechanistic insights suggesting that S1P plays a role in Aβ generation, tau phosphorylation, and AD pathology. The function of the S1P receptor modulator FTY720 has been tested in AD models. In rat injected with Aβ peptide in the hippocampus, FTY720 attenuated Aβ-induced cognitive impairment and neuronal damage (Asle-Rousta et al., 2013). In a mouse model of AD, 5×FAD, FTY720 treatment decreased Aβ plaque density as well as soluble and insoluble Aβ (Aytan et al., 2016). However, how FTY720 functions through S1P signaling in this AD model is not clear. Furthermore, the function of S1P signaling in different neural cell types and the crosstalk between them in an AD setting needs to be systematically investigated.

HD

It was recently recognized that sphingolipid metabolism is significantly perturbed in HD. The S1P level is reduced in HD patient and animal model brain samples (Di Pardo et al., 2017; Di Pardo and Maglione, 2018a, b). Consistently, the levels of SK1 is reduced, while that of S1P lyase increased in brain tissues from HD patients and/or HD mouse models, indicating that a defect in sphingolipid metabolism potentially contributes to HD. Therefore, potential pharmacological interventions targeting S1P signaling have been pursued to treat HD. Administration of FTY720 in the R6/2 mouse model of HD improves motor function, prolongs survival, and reduces brain atrophy (Di Pardo et al., 2014). Inhibition of S1P lyase by 2-acetyl-4-tetrahydroxybutyl imidazole (THI) improves survival of neurons transfected with mutant huntingtin (Moruno Manchon et al., 2015). Furthermore, activation of SK1 by 2-hexyl-N-[2hydroxy-1-(hydroxymethyl)ethyl]-3-oxo-decanamide (K6PC-5) significantly reduces apoptosis in mouse striatal-derived HD cell lines and leads to the activation of pro-survival signaling pathways (Di Pardo et al., 2017, 2018).

PD

Recently, modulation of S1P signaling was explored in experimental models of PD. SK1 inhibition reduces survival and increases oxygen reactive species in human dopaminergic neuronal cells treated with1-methyl-4-phenylpyridinium, a PD mouse model (Pyszko and Strosznajder, 2014). Conversely, S1P supplementation significantly reduces apoptosis, suggesting that S1P expression protects these neurons (Pyszko and Strosznajder, 2014). On the other hand, SK2 inhibition results in downregulation of mitochondrial-related genes such as proliferator-activated receptor γ coactivator-1a (PGC-1a) and its downstream targets nuclear respiratory factor 1 (NRF-1) and mitochondrial transcription factor A (TFAM) in multiple PD experimental models (Sivasubramanian et al., 2015). Furthermore, treatment with S1P enhances the expression of PGC-1a and NRF-1 in a mouse model of the disease and exerts a neuroprotective effect on dopaminergic neurons via S1P1 (Sivasubramanian et al., 2015). The potential role of S1P receptor stimulation in PD has also been explored. Treatment with FTY720 attenuates motor deficit and prevents dopaminergic neuronal loss in two mouse models of PD (Zhao et al., 2017). This beneficial effect of FTY720 was abolished by W146, a S1P1-selective antagonist, indicating that FTY720 acts through S1P1 (Zhao et al., 2017).

Conclusions

Many neurodegenerative diseases are characterized by the accumulation of sphingolipids (sphingolipidoses) or proteins (AD, PD, HD). Recent research suggests that the “classical” view of the accumulated agent being neurotoxic should give way to a systems biological or physiological approach implying the molecular interaction of organelles, cell types, and tissues, namely lysosomes and mitochondria, neurons, astrocytes and microglia, and brain and immune system. At first sight, this “new” view on neurodegeneration appears to be daunting since it implies a plethora of diverse and “moving targets”. However, with the emerging role of ceramide and S1P as cell signaling hubs in the pathophysiology of several neurodegenerative diseases, unified therapeutic approaches are in reach. One of these approaches is the use of FTY720, an S1P modulator initially approved for treatment of MS, may also help in AD, PD, and HD. Enzyme inhibitors preventing generation of ceramide or its derivatives are already in use for treatment of sphingolipidoses, but may also be useful in therapy of AD and other neurodegenerative diseases. Therefore, research on sphingolipids is critical to find novel potential targets for therapy of neurodegenerative disease.

Acknowledgements

This work was the supported by the National Institutes of Health grants NIH R01AG034389 and R01NS095215. The authors also acknowledge support by the Department of Physiology (Chair Dr. Alan Daugherty), University of Kentucky, Lexington, KY.

Abbreviations

AD

Alzheimer’s disease

PKC

atypical PKC

ApoE4

apolipoprotein E4

APP

amyloid precursor protein

aSMase

acid sphingomyelinase

CerS

ceramide synthase

CERT

ceramide transport protein

CNS

central nervous system

EAD

experimental autoimmune encephalomyelitis

ER

endoplasmic reticulum

GSK

glycogen synthase kinase

HD

Huntington’s disease

MS

Multiple Sclerosis

NPC1

Niemann-Pick disease type C1 protein

nSMase2

neutral sphingomyelinase 2

OMM

outer mitochondrial membrane

PD

Parkinson’s disease

PNS

peripheral nervous system

PP1

protein phosphatase 1

PP2a

protein phosphatase 2a

SCI

spinal cord injury

S1P

sphingosine-1-phosphate

SK

sphingosine kinase

SMase

sphingomyelinase

TBI

traumatic brain injury

VDAC1

voltage-dependent anion channel 1

Footnotes

Declarations of interest: None

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Abe T, Niizuma K, Kanoke A, Saigusa D, Saito R, Uruno A, Fujimura M, Yamamoto M, Tominaga T, 2018. Metabolomic Analysis of Mouse Brain after a Transient Middle Cerebral Artery Occlusion by Mass Spectrometry Imaging. Neurologia medico-chirurgica. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Adada M, Canals D, Hannun YA, Obeid LM, 2014. Sphingolipid regulation of ezrin, radixin, and moesin proteins family: implications for cell dynamics. Biochimica et biophysica acta 1841(5), 727–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Alvarez SE, Harikumar KB, Hait NC, Allegood J, Strub GM, Kim EY, Maceyka M, Jiang H, Luo C, Kordula T, Milstien S, Spiegel S, 2010. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature 465(7301), 1084–1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Arenz C, 2017. Recent advances and novel treatments for sphingolipidoses. Future medicinal chemistry 9(14), 1687–1700. [DOI] [PubMed] [Google Scholar]
  5. Ariga T, Jarvis WD, Yu RK, 1998. Role of sphingolipid-mediated cell death in neurodegenerative diseases. Journal of lipid research 39(1), 1–16. [PubMed] [Google Scholar]
  6. Ariga T, McDonald MP, Yu RK, 2008. Role of ganglioside metabolism in the pathogenesis of Alzheimer’s disease--a review. Journal of lipid research 49(6), 1157–1175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Asle-Rousta M, Kolahdooz Z, Dargahi L, Ahmadiani A, Nasoohi S, 2014. Prominence of central sphingosine-1-phosphate receptor-1 in attenuating abeta-induced injury by fingolimod. J Mol Neurosci 54(4), 698–703. [DOI] [PubMed] [Google Scholar]
  8. Asle-Rousta M, Kolahdooz Z, Oryan S, Ahmadiani A, Dargahi L, 2013. FTY720 (fingolimod) attenuates beta-amyloid peptide (Abeta42)-induced impairment of spatial learning and memory in rats. J Mol Neurosci 50(3), 524–532. [DOI] [PubMed] [Google Scholar]
  9. Assi E, Cazzato D, De Palma C, Perrotta C, Clementi E, Cervia D, 2013. Sphingolipids and brain resident macrophages in neuroinflammation: an emerging aspect of nervous system pathology. Clin Dev Immunol 2013, 309302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Audano M, Schneider A, Mitro N, 2018. Mitochondria, lysosomes, and dysfunction: their meaning in neurodegeneration. Journal of neurochemistry. [DOI] [PubMed] [Google Scholar]
  11. Aytan N, Choi JK, Carreras I, Brinkmann V, Kowall NW, Jenkins BG, Dedeoglu A, 2016. Fingolimod modulates multiple neuroinflammatory markers in a mouse model of Alzheimer’s disease. Sci Rep 6, 24939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bandaru VV, Troncoso J, Wheeler D, Pletnikova O, Wang J, Conant K, Haughey NJ, 2009. ApoE4 disrupts sterol and sphingolipid metabolism in Alzheimer’s but not normal brain. Neurobiology of aging 30(4), 591–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Barbacci DC, Roux A, Muller L, Jackson SN, Post J, Baldwin K, Hoffer B, Balaban CD, Schultz JA, Gouty S, Cox BM, Woods AS, 2017. Mass Spectrometric Imaging of Ceramide Biomarkers Tracks Therapeutic Response in Traumatic Brain Injury. ACS chemical neuroscience 8(10), 2266–2274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Barthelmes J, de Bazo AM, Pewzner-Jung Y, Schmitz K, Mayer CA, Foerch C, Eberle M, Tafferner N, Ferreiros N, Henke M, Geisslinger G, Futerman AH, Grosch S, Schiffmann S, 2015. Lack of ceramide synthase 2 suppresses the development of experimental autoimmune encephalomyelitis by impairing the migratory capacity of neutrophils. Brain, behavior, and immunity 46, 280–292. [DOI] [PubMed] [Google Scholar]
  15. Ben-David O, Futerman AH, 2010. The role of the ceramide acyl chain length in neurodegeneration: involvement of ceramide synthases. Neuromolecular medicine 12(4), 341–350. [DOI] [PubMed] [Google Scholar]
  16. Bieberich E, 2010. There is More to a Lipid than just Being a Fat: Sphingolipid-Guided Differentiation of Oligodendroglial Lineage from Embryonic Stem Cells. Neurochemical research. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bieberich E, 2011. Ceramide in stem cell differentiation and embryo development: novel functions of a topological cell-signaling lipid and the concept of ceramide compartments. J Lipids 2011, 610306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bieberich E, 2012. It’s a lipid’s world: bioactive lipid metabolism and signaling in neural stem cell differentiation. Neurochemical research 37(6), 1208–1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bieberich E, Kawaguchi T, Yu RK, 2000. N-acylated serinol is a novel ceramide mimic inducing apoptosis in neuroblastoma cells. The Journal of biological chemistry 275(1), 177–181. [DOI] [PubMed] [Google Scholar]
  20. Bieberich E, MacKinnon S, Silva J, Noggle S, Condie BG, 2003. Regulation of cell death in mitotic neural progenitor cells by asymmetric distribution of prostate apoptosis response 4 (PAR-4) and simultaneous elevation of endogenous ceramide. The Journal of cell biology 162(3), 469–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bieberich E, MacKinnon S, Silva J, Yu RK, 2001. Regulation of apoptosis during neuronal differentiation by ceramide and b-series complex gangliosides. The Journal of biological chemistry 276(48), 44396–44404. [DOI] [PubMed] [Google Scholar]
  22. Blaho VA, Hla T, 2014. An update on the biology of sphingosine 1-phosphate receptors. J Lipid Res 55(8), 1596–1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Borbon I, Totenhagen J, Fiorenza MT, Canterini S, Ke W, Trouard T, Erickson RP, 2012. Niemann-Pick C1 mice, a model of “juvenile Alzheimer’s disease”, with normal gene expression in neurons and fibrillary astrocytes show long term survival and delayed neurodegeneration. Journal of Alzheimer’s disease : JAD 30(4), 875–887. [DOI] [PubMed] [Google Scholar]
  24. Borbon IA, Erickson RP, 2011. Interactions of Npc1 and amyloid accumulation/deposition in the APP/PS1 mouse model of Alzheimer’s. Journal of applied genetics 52(2), 213–218. [DOI] [PubMed] [Google Scholar]
  25. Boulgaropoulos B, Rappolt M, Sartori B, Amenitsch H, Pabst G, 2012. Lipid sorting by ceramide and the consequences for membrane proteins. Biophysical journal 102(9), 2031–2038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Bourbon NA, Yun J, Kester M, 2000. Ceramide directly activates protein kinase C zeta to regulate a stress-activated protein kinase signaling complex. The Journal of biological chemistry 275(45), 35617–35623. [DOI] [PubMed] [Google Scholar]
  27. Bradley E, Dasgupta S, Jiang X, Zhao X, Zhu G, He Q, Dinkins M, Bieberich E, Wang G, 2014. Critical role of spns2, a sphingosine-1-phosphate transporter, in lung cancer cell survival and migration. PLoS One 9(10), e110119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Brailoiu E, Cooper RL, Dun NJ, 2002. Sphingosine 1-phosphate enhances spontaneous transmitter release at the frog neuromuscular junction. Br J Pharmacol 136(8), 1093–1097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Brinkmann V, Billich A, Baumruker T, Heining P, Schmouder R, Francis G, Aradhye S, Burtin P, 2010. Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat Rev Drug Discov 9(11), 883–897. [DOI] [PubMed] [Google Scholar]
  30. Brugg B, Michel PP, Agid Y, Ruberg M, 1996. Ceramide induces apoptosis in cultured mesencephalic neurons. Journal of neurochemistry 66(2), 733–739. [DOI] [PubMed] [Google Scholar]
  31. Brunkhorst R, Friedlaender F, Ferreiros N, Schwalm S, Koch A, Grammatikos G, Toennes S, Foerch C, Pfeilschifter J, Pfeilschifter W, 2015. Alterations of the Ceramide Metabolism in the Peri-Infarct Cortex Are Independent of the Sphingomyelinase Pathway and Not Influenced by the Acid Sphingomyelinase Inhibitor Fluoxetine. Neural plasticity 2015, 503079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Burgert A, Schlegel J, Becam J, Doose S, Bieberich E, Schubert-Unkmeir A, Sauer M, 2017. Characterization of Plasma Membrane Ceramides by Super-Resolution Microscopy. Angewandte Chemie. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Burns M, Gaynor K, Olm V, Mercken M, LaFrancois J, Wang L, Mathews PM, Noble W, Matsuoka Y, Duff K, 2003. Presenilin redistribution associated with aberrant cholesterol transport enhances beta-amyloid production in vivo. The Journal of neuroscience : the official journal of the Society for Neuroscience 23(13), 5645–5649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Camoletto PG, Vara H, Morando L, Connell E, Marletto FP, Giustetto M, Sassoe-Pognetto M, Van Veldhoven PP, Ledesma MD, 2009. Synaptic vesicle docking: sphingosine regulates syntaxin1 interaction with Munc18. PLoS One 4(4), e5310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Canals D, Roddy P, Hannun YA, 2012. Protein phosphatase 1alpha mediates ceramideinduced ERM protein dephosphorylation: a novel mechanism independent of phosphatidylinositol 4, 5-biphosphate (PIP2) and myosin/ERM phosphatase. The Journal of biological chemistry 287(13), 10145–10155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Carassiti D, Altmann DR, Petrova N, Pakkenberg B, Scaravilli F, Schmierer K, 2018. Neuronal loss, demyelination and volume change in the multiple sclerosis neocortex. Neuropathol Appl Neurobiol 44(4), 377–390. [DOI] [PubMed] [Google Scholar]
  37. Carlson CD, Hart RP, 1996. Activation of acidic sphingomyelinase and protein kinase C zeta is required for IL-1 induction of LIF mRNA in a Schwann cell line. Glia 18(1), 49–58. [DOI] [PubMed] [Google Scholar]
  38. Caterino M, Ruoppolo M, Mandola A, Costanzo M, Orru S, Imperlini E, 2017. Protein-protein interaction networks as a new perspective to evaluate distinct functional roles of voltage-dependent anion channel isoforms. Molecular bioSystems 13(12), 2466–2476. [DOI] [PubMed] [Google Scholar]
  39. Ceccom J, Loukh N, Lauwers-Cances V, Touriol C, Nicaise Y, Gentil C, Uro-Coste E, Pitson S, Maurage CA, Duyckaerts C, Cuvillier O, Delisle MB, 2014. Reduced sphingosine kinase-1 and enhanced sphingosine 1-phosphate lyase expression demonstrate deregulated sphingosine 1-phosphate signaling in Alzheimer’s disease. Acta Neuropathol Commun 2, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Chakrabarti SS, Bir A, Poddar J, Sinha M, Ganguly A, Chakrabarti S, 2016. Ceramide and Sphingosine-1-Phosphate in Cell Death Pathways : Relevance to the Pathogenesis of Alzheimer’s Disease. Current Alzheimer research 13(11), 1232–1248. [DOI] [PubMed] [Google Scholar]
  41. Chalfant CE, Kishikawa K, Mumby MC, Kamibayashi C, Bielawska A, Hannun YA, 1999. Long chain ceramides activate protein phosphatase-1 and protein phosphatase-2A. Activation is stereospecific and regulated by phosphatidic acid. The Journal of biological chemistry 274(29), 20313–20317. [DOI] [PubMed] [Google Scholar]
  42. Chalfant CE, Szulc Z, Roddy P, Bielawska A, Hannun YA, 2004. The structural requirements for ceramide activation of serine-threonine protein phosphatases. Journal of lipid research 45(3), 496–506. [DOI] [PubMed] [Google Scholar]
  43. Chami M, Halmer R, Schnoeder L, Anne Becker K, Meier C, Fassbender K, Gulbins E, Walter S, 2017. Acid sphingomyelinase deficiency enhances myelin repair after acute and chronic demyelination. PloS one 12(6), e0178622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Chaudhuri A, 2013. Multiple sclerosis is primarily a neurodegenerative disease. J Neural Transm (Vienna) 120(10), 1463–1466. [DOI] [PubMed] [Google Scholar]
  45. Chiantia S, Ries J, Chwastek G, Carrer D, Li Z, Bittman R, Schwille P, 2008. Role of ceramide in membrane protein organization investigated by combined AFM and FCS. Biochimica et biophysica acta 1778(5), 1356–1364. [DOI] [PubMed] [Google Scholar]
  46. Choi JW, Gardell SE, Herr DR, Rivera R, Lee CW, Noguchi K, Teo ST, Yung YC, Lu M, Kennedy G, Chun J, 2011. FTY720 (fingolimod) efficacy in an animal model of multiple sclerosis requires astrocyte sphingosine 1-phosphate receptor 1 (S1P1) modulation. Proc Natl Acad Sci U S A 108(2), 751–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Chu Y, Goldman JG, Kelly L, He Y, Waliczek T, Kordower JH, 2014. Abnormal alphasynuclein reduces nigral voltage-dependent anion channel 1 in sporadic and experimental Parkinson’s disease. Neurobiology of disease 69, 1–14. [DOI] [PubMed] [Google Scholar]
  48. Chun J, Hartung HP, 2010. Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis. Clin Neuropharmacol 33(2), 91–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Clarke CJ, Guthrie JM, Hannun YA, 2008. Regulation of neutral sphingomyelinase-2 (nSMase2) by tumor necrosis factor-alpha involves protein kinase C-delta in lung epithelial cells. Mol Pharmacol 74(4), 1022–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Coelho RP, Saini HS, Sato-Bigbee C, 2010. Sphingosine-1-phosphate and oligodendrocytes: from cell development to the treatment of multiple sclerosis. Prostaglandins Other Lipid Mediat 91(3–4), 139–144. [DOI] [PubMed] [Google Scholar]
  51. Colell A, Fernandez A, Fernandez-Checa JC, 2009. Mitochondria, cholesterol and amyloid beta peptide: a dangerous trio in Alzheimer disease. Journal of bioenergetics and biomembranes 41(5), 417–423. [DOI] [PubMed] [Google Scholar]
  52. Compston A, Coles A, 2008. Multiple sclerosis. Lancet 372(9648), 1502–1517. [DOI] [PubMed] [Google Scholar]
  53. Costantini C, Weindruch R, Della Valle G, Puglielli L, 2005. A TrkA-to-p75NTR molecular switch activates amyloid beta-peptide generation during aging. The Biochemical journal 391(Pt 1), 59–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Couttas TA, Kain N, Daniels B, Lim XY, Shepherd C, Kril J, Pickford R, Li H, Garner B, Don AS, 2014. Loss of the neuroprotective factor Sphingosine 1-phosphate early in Alzheimer’s disease pathogenesis. Acta Neuropathol Commun 2, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Cutler RG, Mattson MP, 2001. Sphingomyelin and ceramide as regulators of development and lifespan. Mech Ageing Dev 122(9), 895–908. [DOI] [PubMed] [Google Scholar]
  56. Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Coso OA, Gutkind S, Spiegel S, 1996. Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature 381(6585), 800–803. [DOI] [PubMed] [Google Scholar]
  57. Darios F, Wasser C, Shakirzyanova A, Giniatullin A, Goodman K, Munoz-Bravo JL, Raingo J, Jorgacevski J, Kreft M, Zorec R, Rosa JM, Gandia L, Gutierrez LM, Binz T, Giniatullin R, Kavalali ET, Davletov B, 2009. Sphingosine facilitates SNARE complex assembly and activates synaptic vesicle exocytosis. Neuron 62(5), 683–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. den Hoedt S, Janssen CIF, Astarita G, Piomelli D, Leijten FPJ, Crivelli SM, Verhoeven AJM, de Vries HE, Walter J, Martinez-Martinez P, Sijbrands EJG, Kiliaan AJ, Mulder MT, 2017. Pleiotropic Effect of Human ApoE4 on Cerebral Ceramide and Saturated Fatty Acid Levels. Journal of Alzheimer’s disease : JAD 60(3), 769–781. [DOI] [PubMed] [Google Scholar]
  59. Devillard R, Galvani S, Thiers JC, Guenet JL, Hannun Y, Bielawski J, Negre-Salvayre A, Salvayre R, Auge N, 2010. Stress-induced sphingolipid signaling: role of type-2 neutral sphingomyelinase in murine cell apoptosis and proliferation. PloS one 5(3), e9826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Di Pardo A, Amico E, Basit A, Armirotti A, Joshi P, Neely MD, Vuono R, Castaldo S, Digilio AF, Scalabri F, Pepe G, Elifani F, Madonna M, Jeong SK, Park BM, D’Esposito M, Bowman AB, Barker RA, Maglione V, 2017. Defective Sphingosine-1-phosphate metabolism is a druggable target in Huntington’s disease. Scientific reports 7(1), 5280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Di Pardo A, Amico E, Basit A, Armirotti A, Joshi P, Neely MD, Vuono R, Castaldo S, Digilio AF, Scalabri F, Pepe G, Elifani F, Madonna M, Jeong SK, Park BM, D’Esposito M, Bowman AB, Barker RA, Maglione V, 2018. Author Correction: Defective Sphingosine-1-phosphate metabolism is a druggable target in Huntington’s disease. Sci Rep 8(1), 8266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Di Pardo A, Amico E, Favellato M, Castrataro R, Fucile S, Squitieri F, Maglione V, 2014. FTY720 (fingolimod) is a neuroprotective and disease-modifying agent in cellular and mouse models of Huntington disease. Hum Mol Genet 23(9), 2251–2265. [DOI] [PubMed] [Google Scholar]
  63. Di Pardo A, Maglione V, 2018a. The S1P Axis: New Exciting Route for Treating Huntington’s Disease. Trends in pharmacological sciences 39(5), 468–480. [DOI] [PubMed] [Google Scholar]
  64. Di Pardo A, Maglione V, 2018b. Sphingolipid Metabolism: A New Therapeutic Opportunity for Brain Degenerative Disorders. Frontiers in neuroscience 12, 249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Dinkins MB, Dasgupta S, Wang G, Zhu G, Bieberich E, 2014. Exosome reduction in vivo is associated with lower amyloid plaque load in the 5XFAD mouse model of Alzheimer’s disease. Neurobiology of aging. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Dinkins MB, Enasko J, Hernandez C, Wang G, Kong J, Helwa I, Liu Y, Terry AV Jr., Bieberich E, 2016a. Neutral Sphingomyelinase-2 Deficiency Ameliorates Alzheimer’s Disease Pathology and Improves Cognition in the 5XFAD Mouse. The Journal of neuroscience : the official journal of the Society for Neuroscience 36(33), 8653–8667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Dinkins MB, Wang G, Bieberich E, 2016b. Sphingolipid-Enriched Extracellular Vesicles and Alzheimer’s Disease: A Decade of Research. Journal of Alzheimer’s disease : JAD. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Dobrowsky RT, Hannun YA, 1992. Ceramide stimulates a cytosolic protein phosphatase. The Journal of biological chemistry 267(8), 5048–5051. [PubMed] [Google Scholar]
  69. Dobrowsky RT, Kamibayashi C, Mumby MC, Hannun YA, 1993. Ceramide activates heterotrimeric protein phosphatase 2A. The Journal of biological chemistry 268(21), 15523–15530. [PubMed] [Google Scholar]
  70. Draeger A, Babiychuk EB, 2013. Ceramide in plasma membrane repair. Handb Exp Pharmacol(216), 341–353. [DOI] [PubMed] [Google Scholar]
  71. Drulis-Fajdasz D, Rakus D, Wisniewski JR, McCubrey JA, Gizak A, 2018. Systematic analysis of GSK-3 signaling pathways in aging of cerebral tissue. Advances in biological regulation 69, 35–42. [DOI] [PubMed] [Google Scholar]
  72. Dusaban SS, Chun J, Rosen H, Purcell NH, Brown JH, 2017. Sphingosine 1-phosphate receptor 3 and RhoA signaling mediate inflammatory gene expression in astrocytes. J Neuroinflammation 14(1), 111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Ebel P, Vom Dorp K, Petrasch-Parwez E, Zlomuzica A, Kinugawa K, Mariani J, Minich D, Ginkel C, Welcker J, Degen J, Eckhardt M, Dere E, Dormann P, Willecke K, 2013. Inactivation of ceramide synthase 6 in mice results in an altered sphingolipid metabolism and behavioral abnormalities. The Journal of biological chemistry 288(29), 21433–21447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Eberle M, Ebel P, Wegner MS, Mannich J, Tafferner N, Ferreiros N, Birod K, Schreiber Y, Krishnamoorthy G, Willecke K, Geisslinger G, Grosch S, Schiffmann S, 2014. Regulation of ceramide synthase 6 in a spontaneous experimental autoimmune encephalomyelitis model is sex dependent. Biochemical pharmacology 92(2), 326–335. [DOI] [PubMed] [Google Scholar]
  75. Eckhardt M, 2010. Pathology and current treatment of neurodegenerative sphingolipidoses. Neuromolecular medicine 12(4), 362–382. [DOI] [PubMed] [Google Scholar]
  76. Edsall LC, Pirianov GG, Spiegel S, 1997. Involvement of sphingosine 1-phosphate in nerve growth factor-mediated neuronal survival and differentiation. The Journal of neuroscience : the official journal of the Society for Neuroscience 17(18), 6952–6960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Eliyahu E, Park JH, Shtraizent N, He X, Schuchman EH, 2007. Acid ceramidase is a novel factor required for early embryo survival. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 21(7), 1403–1409. [DOI] [PubMed] [Google Scholar]
  78. Fan M, Sidhu R, Fujiwara H, Tortelli B, Zhang J, Davidson C, Walkley SU, Bagel JH, Vite C, Yanjanin NM, Porter FD, Schaffer JE, Ory DS, 2013. Identification of Niemann-Pick C1 disease biomarkers through sphingolipid profiling. Journal of lipid research 54(10), 2800–2814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Farez MF, Correale J, 2016. Sphingosine 1-phosphate signaling in astrocytes: Implications for progressive multiple sclerosis. J Neurol Sci 361, 60–65. [DOI] [PubMed] [Google Scholar]
  80. Farfel-Becker T, Vitner EB, Kelly SL, Bame JR, Duan J, Shinder V, Merrill AH Jr., Dobrenis K, Futerman AH, 2014. Neuronal accumulation of glucosylceramide in a mouse model of neuronopathic Gaucher disease leads to neurodegeneration. Human molecular genetics 23(4), 843–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Farooqui AA, Ong WY, Farooqui T, 2010. Lipid mediators in the nucleus: Their potential contribution to Alzheimer’s disease. Biochimica et biophysica acta 1801(8), 906–916. [DOI] [PubMed] [Google Scholar]
  82. Fernandez-Pisonero I, Duenas AI, Barreiro O, Montero O, Sanchez-Madrid F, GarciaRodriguez C, 2012. Lipopolysaccharide and sphingosine-1-phosphate cooperate to induce inflammatory molecules and leukocyte adhesion in endothelial cells. J Immunol 189(11), 5402–5410. [DOI] [PubMed] [Google Scholar]
  83. Fernandez A, Llacuna L, Fernandez-Checa JC, Colell A, 2009. Mitochondrial cholesterol loading exacerbates amyloid beta peptide-induced inflammation and neurotoxicity. The Journal of neuroscience : the official journal of the Society for Neuroscience 29(20), 6394–6405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Folts CJ, Scott-Hewitt N, Proschel C, Mayer-Proschel M, Noble M, 2016. Lysosomal Reacidification Prevents Lysosphingolipid-Induced Lysosomal Impairment and Cellular Toxicity. PLoS biology 14(12), e1002583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Friese MA, Schattling B, Fugger L, 2014. Mechanisms of neurodegeneration and axonal dysfunction in multiple sclerosis. Nat Rev Neurol 10(4), 225–238. [DOI] [PubMed] [Google Scholar]
  86. Fujikake N, Shin M, Shimizu S, 2018. Association Between Autophagy and Neurodegenerative Diseases. Frontiers in neuroscience 12, 255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Fukuhara S, Simmons S, Kawamura S, Inoue A, Orba Y, Tokudome T, Sunden Y, Arai Y, Moriwaki K, Ishida J, Uemura A, Kiyonari H, Abe T, Fukamizu A, Hirashima M, Sawa H, Aoki J, Ishii M, Mochizuki N, 2012. The sphingosine-1-phosphate transporter Spns2 expressed on endothelial cells regulates lymphocyte trafficking in mice. J Clin Invest 122(4), 1416–1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Fuller M, Futerman AH, 2018. The brain lipidome in neurodegenerative lysosomal storage disorders. Biochemical and biophysical research communications. [DOI] [PubMed] [Google Scholar]
  89. Galadari S, Kishikawa K, Kamibayashi C, Mumby MC, Hannun YA, 1998. Purification and characterization of ceramide-activated protein phosphatases. Biochemistry 37(32), 11232–11238. [DOI] [PubMed] [Google Scholar]
  90. Gault CR, Obeid LM, Hannun YA, 2010. An overview of sphingolipid metabolism: from synthesis to breakdown. Advances in experimental medicine and biology 688, 1–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Geisler S, Holmstrom KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ, Springer W, 2010. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nature cell biology 12(2), 119–131. [DOI] [PubMed] [Google Scholar]
  92. Gelineau-van Waes J, Starr L, Maddox J, Aleman F, Voss KA, Wilberding J, Riley RT, 2005. Maternal fumonisin exposure and risk for neural tube defects: mechanisms in an in vivo mouse model. Birth Defects Res A Clin Mol Teratol 73(7), 487–497. [DOI] [PubMed] [Google Scholar]
  93. Gelineau-van Waes J, Voss KA, Stevens VL, Speer MC, Riley RT, 2009. Maternal fumonisin exposure as a risk factor for neural tube defects. Adv Food Nutr Res 56, 145–181. [DOI] [PubMed] [Google Scholar]
  94. Goni FM, Contreras FX, Montes LR, Sot J, Alonso A, 2005. Biophysics (and sociology) of ceramides. Biochem Soc Symp(72), 177–188. [DOI] [PubMed] [Google Scholar]
  95. Grassi S, Chiricozzi E, Mauri L, Sonnino S, Prinetti A, 2018. Sphingolipids and neuronal degeneration in lysosomal storage disorders. Journal of neurochemistry. [DOI] [PubMed] [Google Scholar]
  96. Grosch S, Schiffmann S, Geisslinger G, 2012. Chain length-specific properties of ceramides. Progress in lipid research 51(1), 50–62. [DOI] [PubMed] [Google Scholar]
  97. Gu L, Huang B, Shen W, Gao L, Ding Z, Wu H, Guo J, 2013. Early activation of nSMase2/ceramide pathway in astrocytes is involved in ischemia-associated neuronal damage via inflammation in rat hippocampi. Journal of neuroinflammation 10, 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Guo X, Namekata K, Kimura A, Harada C, Harada T, 2017. ASK1 in neurodegeneration. Advances in biological regulation 66, 63–71. [DOI] [PubMed] [Google Scholar]
  99. Hagen N, Hans M, Hartmann D, Swandulla D, van Echten-Deckert G, 2011. Sphingosine1-phosphate links glycosphingolipid metabolism to neurodegeneration via a calpain-mediated mechanism. Cell Death Differ 18(8), 1356–1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Haimovitz-Friedman A, Kan CC, Ehleiter D, Persaud RS, McLoughlin M, Fuks Z, Kolesnick RN, 1994. Ionizing radiation acts on cellular membranes to generate ceramide and initiate apoptosis. The Journal of experimental medicine 180(2), 525–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, Luo C, Marmorstein R, Kordula T, Milstien S, Spiegel S, 2009. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science 325(5945), 1254–1257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Hait NC, Oskeritzian CA, Paugh SW, Milstien S, Spiegel S, 2006. Sphingosine kinases, sphingosine 1-phosphate, apoptosis and diseases. Biochimica et biophysica acta 1758(12), 2016–2026. [DOI] [PubMed] [Google Scholar]
  103. Hammad SM, Crellin HG, Wu BX, Melton J, Anelli V, Obeid LM, 2008. 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 85(3–4), 107–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Han X, D, MH, McKeel DW Jr., Kelley J, Morris JC, 2002. Substantial sulfatide deficiency and ceramide elevation in very early Alzheimer’s disease: potential role in disease pathogenesis. Journal of neurochemistry 82(4), 809–818. [DOI] [PubMed] [Google Scholar]
  105. Han X, Tong J, Zhang J, Farahvar A, Wang E, Yang J, Samadani U, Smith DH, Huang JH, 2011. Imipramine treatment improves cognitive outcome associated with enhanced hippocampal neurogenesis after traumatic brain injury in mice. Journal of neurotrauma 28(6), 995–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Hannun YA, 1996. Functions of ceramide in coordinating cellular responses to stress. Science 274(5294), 1855–1859. [DOI] [PubMed] [Google Scholar]
  107. Hannun YA, Bell RM, 1993. The sphingomyelin cycle: a prototypic sphingolipid signaling pathway. Adv Lipid Res 25, 27–41. [PubMed] [Google Scholar]
  108. Hannun YA, Loomis CR, Merrill AH Jr., Bell RM, 1986. Sphingosine inhibition of protein kinase C activity and of phorbol dibutyrate binding in vitro and in human platelets. The Journal of biological chemistry 261(27), 12604–12609. [PubMed] [Google Scholar]
  109. Hannun YA, Obeid LM, 2008. Principles of bioactive lipid signalling: lessons from sphingolipids. Nature reviews. Molecular cell biology 9(2), 139–150. [DOI] [PubMed] [Google Scholar]
  110. Harvald EB, Olsen AS, Faergeman NJ, 2015. Autophagy in the light of sphingolipid metabolism. Apoptosis : an international journal on programmed cell death 20(5), 658–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Haughey NJ, 2010. Sphingolipids in neurodegeneration. Neuromolecular medicine 12(4), 301–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. He Q, Wang G, Dasgupta S, Dinkins M, Zhu G, Bieberich E, 2012. Characterization of an apical ceramide-enriched compartment regulating ciliogenesis. Molecular biology of the cell 23(16), 3156–3166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. He Q, Wang G, Wakade S, Dasgupta S, Dinkins M, Kong JN, Spassieva SD, Bieberich E, 2014. Primary cilia in stem cells and neural progenitors are regulated by neutral sphingomyelinase 2 and ceramide. Molecular biology of the cell 25(11), 1715–1729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. He X, Huang Y, Li B, Gong CX, Schuchman EH, 2010. Deregulation of sphingolipid metabolism in Alzheimer’s disease. Neurobiol Aging 31(3), 398–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Hernandez-Corbacho MJ, Salama MF, Canals D, Senkal CE, Obeid LM, 2017. Sphingolipids in mitochondria. Biochimica et biophysica acta 1862(1), 56–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Hisano Y, Kobayashi N, Yamaguchi A, Nishi T, 2012. Mouse SPNS2 functions as a sphingosine-1-phosphate transporter in vascular endothelial cells. PLoS One 7(6), e38941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Horres CR, Hannun YA, 2012. The roles of neutral sphingomyelinases in neurological pathologies. Neurochemical research 37(6), 1137–1149. [DOI] [PubMed] [Google Scholar]
  118. Huang YL, Huang WP, Lee H, 2011. Roles of sphingosine 1-phosphate on tumorigenesis. World J Biol Chem 2(2), 25–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Hunter SF, Bowen JD, Reder AT, 2016. The Direct Effects of Fingolimod in the Central Nervous System: Implications for Relapsing Multiple Sclerosis. CNS Drugs 30(2), 135–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Huwiler A, Zangemeister-Wittke U, 2018. The sphingosine 1-phosphate receptor modulator fingolimod as a therapeutic agent: Recent findings and new perspectives. Pharmacology & therapeutics 185, 34–49. [DOI] [PubMed] [Google Scholar]
  121. Igarashi Y, Kitamura K, Toyokuni T, Dean B, Fenderson B, Ogawass T, Hakomori S, 1990. A specific enhancing effect of N,N-dimethylsphingosine on epidermal growth factor receptor autophosphorylation. Demonstration of its endogenous occurrence (and the virtual absence of unsubstituted sphingosine) in human epidermoid carcinoma A431 cells. The Journal of biological chemistry 265(10), 5385–5389. [PubMed] [Google Scholar]
  122. Ito A, Horigome K, 1995. Ceramide prevents neuronal programmed cell death induced by nerve growth factor deprivation. Journal of neurochemistry 65(1), 463–466. [DOI] [PubMed] [Google Scholar]
  123. Jana A, Pahan K, 2010. Sphingolipids in multiple sclerosis. Neuromolecular medicine 12(4), 351–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Jeffery DR, Rammohan KW, Hawker K, Fox E, 2016. Fingolimod: a review of its mode of action in the context of its efficacy and safety profile in relapsing forms of multiple sclerosis. Expert Rev Neurother 16(1), 31–44. [DOI] [PubMed] [Google Scholar]
  125. Jiang W, Ogretmen B, 2014. Autophagy paradox and ceramide. Biochimica et biophysica acta 1841(5), 783–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Jones ZB, Ren Y, 2016. Sphingolipids in spinal cord injury. International journal of physiology, pathophysiology and pharmacology 8(2), 52–69. [PMC free article] [PubMed] [Google Scholar]
  127. Jung CG, Kim HJ, Miron VE, Cook S, Kennedy TE, Foster CA, Antel JP, Soliven B, 2007. Functional consequences of S1P receptor modulation in rat oligodendroglial lineage cells. Glia 55(16), 1656–1667. [DOI] [PubMed] [Google Scholar]
  128. Kalvodova L, Kahya N, Schwille P, Ehehalt R, Verkade P, Drechsel D, Simons K, 2005. Lipids as modulators of proteolytic activity of BACE: involvement of cholesterol, glycosphingolipids, and anionic phospholipids in vitro. The Journal of biological chemistry 280(44), 36815–36823. [DOI] [PubMed] [Google Scholar]
  129. Kanno T, Nishizaki T, Proia RL, Kajimoto T, Jahangeer S, Okada T, Nakamura S, 2010. Regulation of synaptic strength by sphingosine 1-phosphate in the hippocampus. Neuroscience 171(4), 973–980. [DOI] [PubMed] [Google Scholar]
  130. Kappos L, Antel J, Comi G, Montalban X, O’Connor P, Polman CH, Haas T, Korn AA, Karlsson G, Radue EW, Group FDS, 2006. Oral fingolimod (FTY720) for relapsing multiple sclerosis. N Engl J Med 355(11), 1124–1140. [DOI] [PubMed] [Google Scholar]
  131. Karunakaran I, van Echten-Deckert G, 2017. Sphingosine 1-phosphate - A double edged sword in the brain. Biochimica et biophysica acta 1859(9 Pt B), 1573–1582. [DOI] [PubMed] [Google Scholar]
  132. Kawahara A, Nishi T, Hisano Y, Fukui H, Yamaguchi A, Mochizuki N, 2009. The sphingolipid transporter spns2 functions in migration of zebrafish myocardial precursors. Science 323(5913), 524–527. [DOI] [PubMed] [Google Scholar]
  133. Kim M, Nevado-Holgado A, Whiley L, Snowden SG, Soininen H, Kloszewska I, Mecocci P, Tsolaki M, Vellas B, Thambisetty M, Dobson RJB, Powell JF, Lupton MK, Simmons A, Velayudhan L, Lovestone S, Proitsi P, Legido-Quigley C, 2017. Association between Plasma Ceramides and Phosphatidylcholines and Hippocampal Brain Volume in Late Onset Alzheimer’s Disease. Journal of Alzheimer’s disease : JAD 60(3), 809–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Kleger A, Busch T, Liebau S, Prelle K, Paschke S, Beil M, Rolletschek A, Wobus A, Wolf E, Adler G, Seufferlein T, 2007. The bioactive lipid sphingosylphosphorylcholine induces differentiation of mouse embryonic stem cells and human promyelocytic leukaemia cells. Cell Signal 19(2), 367–377. [DOI] [PubMed] [Google Scholar]
  135. Klyachkin YM, Karapetyan AV, Ratajczak MZ, Abdel-Latif A, 2014. The role of bioactive lipids in stem cell mobilization and homing: novel therapeutics for myocardial ischemia. Biomed Res Int 2014, 653543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Ko MH, Puglielli L, 2009. Two endoplasmic reticulum (ER)/ER Golgi intermediate compartment-based lysine acetyltransferases post-translationally regulate BACE1 levels. The Journal of biological chemistry 284(4), 2482–2492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Kobayashi N, Kawasaki-Nishi S, Otsuka M, Hisano Y, Yamaguchi A, Nishi T, 2018. MFSD2B is a sphingosine 1-phosphate transporter in erythroid cells. Sci Rep 8(1), 4969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Kolesnick RN, 1987. 1,2-Diacylglycerols but not phorbol esters stimulate sphingomyelin hydrolysis in GH3 pituitary cells. The Journal of biological chemistry 262(35), 16759–16762. [PubMed] [Google Scholar]
  139. Kong JN, Hardin K, Dinkins M, Wang G, He Q, Mujadzic T, Zhu G, Bielawski J, Spassieva S, Bieberich E, 2015. Regulation of Chlamydomonas flagella and ependymal cell motile cilia by ceramide-mediated translocation of GSK3. Molecular biology of the cell 26(24), 4451–4465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Kong JN, Zhu Z, Itokazu Y, Wang G, Dinkins MB, Zhong L, Lin HP, Elsherbini A, Leanhart S, Jiang X, Qin H, Zhi W, Spassieva SD, Bieberich E, 2018. Novel function of ceramide for regulation of mitochondrial ATP release in astrocytes. Journal of lipid research. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Krishnamurthy K, Wang G, Silva J, Condie BG, Bieberich E, 2007. Ceramide Regulates Atypical PKC{zeta}/{lambda}-mediated Cell Polarity in Primitive Ectoderm Cells: A NOVEL FUNCTION OF SPHINGOLIPIDS IN MORPHOGENESIS. The Journal of biological chemistry 282(5), 3379–3390. [DOI] [PubMed] [Google Scholar]
  142. Kubota M, Kitahara S, Shimasaki H, Ueta N, 1989. Accumulation of ceramide in ischemic human brain of an acute case of cerebral occlusion. The Japanese journal of experimental medicine 59(2), 59–64. [PubMed] [Google Scholar]
  143. Kunkel GT, Maceyka M, Milstien S, Spiegel S, 2013. Targeting the sphingosine-1-phosphate axis in cancer, inflammation and beyond. Nat Rev Drug Discov 12(9), 688–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Laurenzana A, Cencetti F, Serrati S, Bruno G, Japtok L, Bianchini F, Torre E, Fibbi G, Del Rosso M, Bruni P, Donati C, 2015. Endothelial sphingosine kinase/SPNS2 axis is critical for vessel-like formation by human mesoangioblasts. J Mol Med (Berl) 93(10), 1145–1157. [DOI] [PubMed] [Google Scholar]
  145. Law BA, Liao X, Moore KS, Southard A, Roddy P, Ji R, Sculz Z, Bielawska A, Schulze PC, Cowart LA, 2017. Lipotoxic very-long-chain ceramides cause mitochondrial dysfunction, oxidative stress, and cell death in cardiomyocytes. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Ledesma MD, Prinetti A, Sonnino S, Schuchman EH, 2011. Brain pathology in Niemann Pick disease type A: insights from the acid sphingomyelinase knockout mice. Journal of neurochemistry 116(5), 779–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Lee CW, Choi JW, Chun J, 2010. Neurological S1P signaling as an emerging mechanism of action of oral FTY720 (Fingolimod) in multiple sclerosis. Arch Pharm Res 33(10), 1567–1574. [DOI] [PubMed] [Google Scholar]
  148. Lee JY, Hannun YA, Obeid LM, 1996. Ceramide inactivates cellular protein kinase Calpha. The Journal of biological chemistry 271(22), 13169–13174. [DOI] [PubMed] [Google Scholar]
  149. Lei M, Shafique A, Shang K, Couttas TA, Zhao H, Don AS, Karl T, 2017. Contextual fear conditioning is enhanced in mice lacking functional sphingosine kinase 2. Behav Brain Res 333, 9–16. [DOI] [PubMed] [Google Scholar]
  150. Levy M, Futerman AH, 2010. Mammalian ceramide synthases. IUBMB life 62(5), 347–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Liu CC, Liu CC, Kanekiyo T, Xu H, Bu G, 2013. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nature reviews. Neurology 9(2), 106–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Long J, Darroch P, Wan KF, Kong KC, Ktistakis N, Pyne NJ, Pyne S, 2005. Regulation of cell survival by lipid phosphate phosphatases involves the modulation of intracellular phosphatidic acid and sphingosine 1-phosphate pools. Biochem J 391(Pt 1), 25–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Lozano J, Berra E, Municio MM, Diaz-Meco MT, Dominguez I, Sanz L, Moscat J, 1994. Protein kinase C zeta isoform is critical for kappa B-dependent promoter activation by sphingomyelinase. The Journal of biological chemistry 269(30), 19200–19202. [PubMed] [Google Scholar]
  154. Lv M, Zhang D, Dai D, Zhang W, Zhang L, 2016. Sphingosine kinase 1/sphingosine-1phosphate regulates the expression of interleukin-17A in activated microglia in cerebral ischemia/reperfusion. Inflamm Res 65(7), 551–562. [DOI] [PubMed] [Google Scholar]
  155. Maceyka M, Harikumar KB, Milstien S, Spiegel S, 2012. Sphingosine-1-phosphate signaling and its role in disease. Trends Cell Biol 22(1), 50–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Maceyka M, Payne SG, Milstien S, Spiegel S, 2002. Sphingosine kinase, sphingosine-1-phosphate, and apoptosis. Biochimica et biophysica acta 1585(2–3), 193–201. [DOI] [PubMed] [Google Scholar]
  157. Maceyka M, Spiegel S, 2014. Sphingolipid metabolites in inflammatory disease. Nature 510(7503), 58–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Manczak M, Reddy PH, 2012. Abnormal interaction of VDAC1 with amyloid beta and phosphorylated tau causes mitochondrial dysfunction in Alzheimer’s disease. Human molecular genetics 21(23), 5131–5146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Mao-Draayer Y, Sarazin J, Fox D, Schiopu E, 2017. The sphingosine-1-phosphate receptor: A novel therapeutic target for multiple sclerosis and other autoimmune diseases. Clin Immunol 175, 10–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Mao C, Obeid LM, 2008. Ceramidases: regulators of cellular responses mediated by ceramide, sphingosine, and sphingosine-1-phosphate. Biochimica et biophysica acta 1781(9), 424–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Mao C, Wadleigh M, Jenkins GM, Hannun YA, Obeid LM, 1997. Identification and characterization of Saccharomyces cerevisiae dihydrosphingosine-1-phosphate phosphatase. The Journal of biological chemistry 272(45), 28690–28694. [DOI] [PubMed] [Google Scholar]
  162. Marfia G, Campanella R, Navone SE, Di Vito C, Riccitelli E, Hadi LA, Bornati A, de Rezende G, Giussani P, Tringali C, Viani P, Rampini P, Alessandri G, Parati E, Riboni L, 2014. Autocrine/paracrine sphingosine-1-phosphate fuels proliferative and stemness qualities of glioblastoma stem cells. Glia 62(12), 1968–1981. [DOI] [PubMed] [Google Scholar]
  163. Mencarelli C, Martinez-Martinez P, 2013. Ceramide function in the brain: when a slight tilt is enough. Cellular and molecular life sciences : CMLS 70(2), 181–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Merrill AH Jr., Sereni AM, Stevens VL, Hannun YA, Bell RM, Kinkade JM Jr., 1986. Inhibition of phorbol ester-dependent differentiation of human promyelocytic leukemic (HL-60) cells by sphinganine and other long-chain bases. The Journal of biological chemistry 261(27), 12610–12615. [PubMed] [Google Scholar]
  165. Mielke MM, Bandaru VV, Haughey NJ, Rabins PV, Lyketsos CG, Carlson MC, 2008. Serum sphingomyelins and ceramides are early predictors of memory impairment. Neurobiology of aging. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Mielke MM, Bandaru VV, Haughey NJ, Xia J, Fried LP, Yasar S, Albert M, Varma V, Harris G, Schneider EB, Rabins PV, Bandeen-Roche K, Lyketsos CG, Carlson MC, 2012. Serum ceramides increase the risk of Alzheimer disease: the Women’s Health and Aging Study II. Neurology 79(7), 633–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Mielke MM, Haughey NJ, Bandaru VV, Schech S, Carrick R, Carlson MC, Mori S, Miller MI, Ceritoglu C, Brown T, Albert M, Lyketsos CG, 2010. Plasma ceramides are altered in mild cognitive impairment and predict cognitive decline and hippocampal volume loss. Alzheimer’s & dementia : the journal of the Alzheimer’s Association 6(5), 378–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Mielke MM, Lyketsos CG, 2010. Alterations of the sphingolipid pathway in Alzheimer’s disease: new biomarkers and treatment targets? Neuromolecular medicine 12(4), 331–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Mielke MM, Maetzler W, Haughey NJ, Bandaru VV, Savica R, Deuschle C, Gasser T, Hauser AK, Graber-Sultan S, Schleicher E, Berg D, Liepelt-Scarfone I, 2013. Plasma ceramide and glucosylceramide metabolism is altered in sporadic Parkinson’s disease and associated with cognitive impairment: a pilot study. PloS one 8(9), e73094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Mishra SK, Gao YG, Deng Y, Chalfant CE, Hinchcliffe EH, Brown RE, 2018. CPTP: A sphingolipid transfer protein that regulates autophagy and inflammasome activation. Autophagy 14(5), 862–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Mizugishi K, Yamashita T, Olivera A, Miller GF, Spiegel S, Proia RL, 2005. Essential role for sphingosine kinases in neural and vascular development. Molecular and cellular biology 25(24), 11113–11121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Moruno-Manchon JF, Uzor NE, Ambati CR, Shetty V, Putluri N, Jagannath C, McCullough LD, Tsvetkov AS, 2018. Sphingosine kinase 1-associated autophagy differs between neurons and astrocytes. Cell Death Dis 9(5), 521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Moruno Manchon JF, Uzor NE, Dabaghian Y, Furr-Stimming EE, Finkbeiner S, Tsvetkov AS, 2015. Cytoplasmic sphingosine-1-phosphate pathway modulates neuronal autophagy. Sci Rep 5, 15213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Moruno Manchon JF, Uzor NE, Finkbeiner S, Tsvetkov AS, 2016. SPHK1/sphingosine kinase 1-mediated autophagy differs between neurons and SH-SY5Y neuroblastoma cells. Autophagy 12(8), 1418–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Mukhopadhyay A, Saddoughi SA, Song P, Sultan I, Ponnusamy S, Senkal CE, Snook CF, Arnold HK, Sears RC, Hannun YA, Ogretmen B, 2008. Direct interaction between the inhibitor 2 and ceramide via sphingolipid-protein binding is involved in the regulation of protein phosphatase 2A activity and signaling. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Mullen TD, Spassieva S, Jenkins RW, Kitatani K, Bielawski J, Hannun YA, Obeid LM, 2011. Selective knockdown of ceramide synthases reveals complex interregulation of sphingolipid metabolism. Journal of lipid research 52(1), 68–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Muller G, Ayoub M, Storz P, Rennecke J, Fabbro D, Pfizenmaier K, 1995. PKC zeta is a molecular switch in signal transduction of TNF-alpha, bifunctionally regulated by ceramide and arachidonic acid. The EMBO journal 14(9), 1961–1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Nagareddy PR, Asfour A, Klyachkin YM, Abdel-Latif A, 2014. A novel role for bioactive lipids in stem cell mobilization during cardiac ischemia: new paradigms in thrombosis: novel mediators and biomarkers. J Thromb Thrombolysis 37(1), 24–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Nayak D, Huo Y, Kwang WX, Pushparaj PN, Kumar SD, Ling EA, Dheen ST, 2010. Sphingosine kinase 1 regulates the expression of proinflammatory cytokines and nitric oxide in activated microglia. Neuroscience 166(1), 132–144. [DOI] [PubMed] [Google Scholar]
  180. Novgorodov SA, Chudakova DA, Wheeler BW, Bielawski J, Kindy MS, Obeid LM, Gudz TI, 2011. Developmentally regulated ceramide synthase 6 increases mitochondrial Ca2+ loading capacity and promotes apoptosis. The Journal of biological chemistry 286(6), 4644–4658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Novgorodov SA, Gudz TI, 2011. Ceramide and mitochondria in ischemic brain injury. Int J Biochem Mol Biol 2(4), 347–361. [PMC free article] [PubMed] [Google Scholar]
  182. Ohanian J, Ohanian V, 2001. Sphingolipids in mammalian cell signalling. Cellular and molecular life sciences : CMLS 58(14), 2053–2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Okazaki T, Bell RM, Hannun YA, 1989. Sphingomyelin turnover induced by vitamin D3 in HL-60 cells. Role in cell differentiation. The Journal of biological chemistry 264(32), 19076–19080. [PubMed] [Google Scholar]
  184. Okazaki T, Bielawska A, Bell RM, Hannun YA, 1990. Role of ceramide as a lipid mediator of 1 alpha,25-dihydroxyvitamin D3-induced HL-60 cell differentiation. The Journal of biological chemistry 265(26), 15823–15831. [PubMed] [Google Scholar]
  185. Olivera A, Buckley NE, Spiegel S, 1992. Sphingomyelinase and cell-permeable ceramide analogs stimulate cellular proliferation in quiescent Swiss 3T3 fibroblasts. The Journal of biological chemistry 267(36), 26121–26127. [PubMed] [Google Scholar]
  186. Olivera A, Kohama T, Edsall L, Nava V, Cuvillier O, Poulton S, Spiegel S, 1999. Sphingosine kinase expression increases intracellular sphingosine-1-phosphate and promotes cell growth and survival. The Journal of cell biology 147(3), 545–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Ong WY, Herr DR, Farooqui T, Ling EA, Farooqui AA, 2015. Role of sphingomyelinases in neurological disorders. Expert Opin Ther Targets 19(12), 1725–1742. [DOI] [PubMed] [Google Scholar]
  188. Osawa Y, Uchinami H, Bielawski J, Schwabe RF, Hannun YA, Brenner DA, 2005. Roles for C16-ceramide and sphingosine 1-phosphate in regulating hepatocyte apoptosis in response to tumor necrosis factor-alpha. The Journal of biological chemistry 280(30), 27879–27887. [DOI] [PubMed] [Google Scholar]
  189. Park JW, Park WJ, Futerman AH, 2014. Ceramide synthases as potential targets for therapeutic intervention in human diseases. Biochimica et biophysica acta 1841(5), 671–681. [DOI] [PubMed] [Google Scholar]
  190. Park K, Ikushiro H, Seo HS, Shin KO, Kim YI, Kim JY, Lee YM, Yano T, Holleran WM, Elias P, Uchida Y, 2016. 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 113(10), E1334–1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Park WJ, Park JW, 2015. The effect of altered sphingolipid acyl chain length on various disease models. Biological chemistry 396(6–7), 693–705. [DOI] [PubMed] [Google Scholar]
  192. Paugh BS, Bryan L, Paugh SW, Wilczynska KM, Alvarez SM, Singh SK, Kapitonov D, Rokita H, Wright S, Griswold-Prenner I, Milstien S, Spiegel S, Kordula T, 2009. Interleukin-1 regulates the expression of sphingosine kinase 1 in glioblastoma cells. J Biol Chem 284(6), 3408–3417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Perera MN, Ganesan V, Siskind LJ, Szulc ZM, Bielawska A, Bittman R, Colombini M, 2016. Ceramide channel: Structural basis for selective membrane targeting. Chemistry and physics of lipids 194, 110–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Perry DM, Kitatani K, Roddy P, El-Osta M, Hannun YA, 2012. Identification and characterization of protein phosphatase 2C activation by ceramide. Journal of lipid research 53(8), 1513–1521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Pettus BJ, Bielawski J, Porcelli AM, Reames DL, Johnson KR, Morrow J, Chalfant CE, Obeid LM, Hannun YA, 2003. The sphingosine kinase 1/sphingosine-1-phosphate pathway mediates COX-2 induction and PGE2 production in response to TNF-alpha. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 17(11), 1411–1421. [DOI] [PubMed] [Google Scholar]
  196. Piccinini M, Scandroglio F, Prioni S, Buccinna B, Loberto N, Aureli M, Chigorno V, Lupino E, DeMarco G, Lomartire A, Rinaudo MT, Sonnino S, Prinetti A, 2010. Deregulated sphingolipid metabolism and membrane organization in neurodegenerative disorders. Molecular neurobiology 41(2–3), 314–340. [DOI] [PubMed] [Google Scholar]
  197. Pinto SN, Silva LC, Futerman AH, Prieto M, 2011. Effect of ceramide structure on membrane biophysical properties: the role of acyl chain length and unsaturation. Biochimica et biophysica acta 1808(11), 2753–2760. [DOI] [PubMed] [Google Scholar]
  198. Pitson SM, Pebay A, 2009. Regulation of stem cell pluripotency and neural differentiation by lysophospholipids. Neuro-Signals 17(4), 242–254. [DOI] [PubMed] [Google Scholar]
  199. Proia RL, Hla T, 2015. Emerging biology of sphingosine-1-phosphate: its role in pathogenesis and therapy. The Journal of clinical investigation 125(4), 1379–1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Puglielli L, Ellis BC, Saunders AJ, Kovacs DM, 2003. Ceramide stabilizes beta-site amyloid precursor protein-cleaving enzyme 1 and promotes amyloid beta-peptide biogenesis. The Journal of biological chemistry 278(22), 19777–19783. [DOI] [PubMed] [Google Scholar]
  201. Pyne S, Adams DR, Pyne NJ, 2018. Sphingosine Kinases as Druggable Targets. Handb Exp Pharmacol. [DOI] [PubMed] [Google Scholar]
  202. Pyszko JA, Strosznajder JB, 2014. The key role of sphingosine kinases in the molecular mechanism of neuronal cell survival and death in an experimental model of Parkinson’s disease. Folia Neuropathol 52(3), 260–269. [DOI] [PubMed] [Google Scholar]
  203. Qian J, Wolters FJ, Beiser A, Haan M, Ikram MA, Karlawish J, Langbaum JB, Neuhaus JM, Reiman EM, Roberts JS, Seshadri S, Tariot PN, Woods BM, Betensky RA, Blacker D, 2017. APOE-related risk of mild cognitive impairment and dementia for prevention trials: An analysis of four cohorts. PLoS medicine 14(3), e1002254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Riganti L, Antonucci F, Gabrielli M, Prada I, Giussani P, Viani P, Valtorta F, Menna E, Matteoli M, Verderio C, 2016. Sphingosine-1-Phosphate (S1P) Impacts Presynaptic Functions by Regulating Synapsin I Localization in the Presynaptic Compartment. J Neurosci 36(16), 4624–4634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Rossner S, Ueberham U, Schliebs R, Perez-Polo JR, Bigl V, 1998. p75 and TrkA receptor signaling independently regulate amyloid precursor protein mRNA expression, isoform composition, and protein secretion in PC12 cells. Journal of neurochemistry 71(2), 757–766. [DOI] [PubMed] [Google Scholar]
  206. Rothhammer V, Kenison JE, Tjon E, Takenaka MC, de Lima KA, Borucki DM, Chao CC, Wilz A, Blain M, Healy L, Antel J, Quintana FJ, 2017. Sphingosine 1-phosphate receptor modulation suppresses pathogenic astrocyte activation and chronic progressive CNS inflammation. Proc Natl Acad Sci U S A 114(8), 2012–2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Roux A, Muller L, Jackson SN, Post J, Baldwin K, Hoffer B, Balaban CD, Barbacci D, Schultz JA, Gouty S, Cox BM, Woods AS, 2016. Mass spectrometry imaging of rat brain lipid profile changes over time following traumatic brain injury. J Neurosci Methods. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Saito Y, Suzuki K, Nanba E, Yamamoto T, Ohno K, Murayama S, 2002. Niemann-Pick type C disease: accelerated neurofibrillary tangle formation and amyloid beta deposition associated with apolipoprotein E epsilon 4 homozygosity. Annals of neurology 52(3), 351–355. [DOI] [PubMed] [Google Scholar]
  209. Sajja V, Jablonska A, Haughey N, Bulte JWM, Stevens RD, Long JB, Walczak P, Janowski M, 2018. Sphingolipids and microRNA Changes in Blood following Blast Traumatic Brain Injury: An Exploratory Study. Journal of neurotrauma 35(2), 353–361. [DOI] [PubMed] [Google Scholar]
  210. Sanabria-Castro A, Alvarado-Echeverria I, Monge-Bonilla C, 2017. Molecular Pathogenesis of Alzheimer’s Disease: An Update. Annals of neurosciences 24(1), 46–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Sandhoff K, 2016. Neuronal sphingolipidoses: Membrane lipids and sphingolipid activator proteins regulate lysosomal sphingolipid catabolism. Biochimie 130, 146–151. [DOI] [PubMed] [Google Scholar]
  212. Satoi H, Tomimoto H, Ohtani R, Kitano T, Kondo T, Watanabe M, Oka N, Akiguchi I, Furuya S, Hirabayashi Y, Okazaki T, 2005. Astroglial expression of ceramide in Alzheimer’s disease brains: a role during neuronal apoptosis. Neuroscience 130(3), 657–666. [DOI] [PubMed] [Google Scholar]
  213. Savica R, Murray ME, Persson XM, Kantarci K, Parisi JE, Dickson DW, Petersen RC, Ferman TJ, Boeve BF, Mielke MM, 2016. Plasma sphingolipid changes with autopsy-confirmed Lewy Body or Alzheimer’s pathology. Alzheimer’s & dementia 3, 43–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Schiffmann S, Ferreiros N, Birod K, Eberle M, Schreiber Y, Pfeilschifter W, Ziemann U, Pierre S, Scholich K, Grosch S, Geisslinger G, 2012. Ceramide synthase 6 plays a critical role in the development of experimental autoimmune encephalomyelitis. Journal of immunology 188(11), 5723–5733. [DOI] [PubMed] [Google Scholar]
  215. Schnaar RL, 2016. Gangliosides of the Vertebrate Nervous System. Journal of molecular biology 428(16), 3325–3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Schneider-Schaulies J, Schneider-Schaulies S, 2013. Viral infections and sphingolipids. Handb Exp Pharmacol(216), 321–340. [DOI] [PubMed] [Google Scholar]
  217. Schwartz NU, Linzer RW, Truman JP, Gurevich M, Hannun YA, Senkal CE, Obeid LM, 2018. Decreased ceramide underlies mitochondrial dysfunction in Charcot-Marie-Tooth 2F. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 32(3), 1716–1728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Sciorra VA, Morris AJ, 2002. Roles for lipid phosphate phosphatases in regulation of cellular signaling. Biochim Biophys Acta 1582(1–3), 45–51. [DOI] [PubMed] [Google Scholar]
  219. Sentelle RD, Senkal CE, Jiang W, Ponnusamy S, Gencer S, Selvam SP, Ramshesh VK, Peterson YK, Lemasters JJ, Szulc ZM, Bielawski J, Ogretmen B, 2012. Ceramide targets autophagosomes to mitochondria and induces lethal mitophagy. Nature chemical biology 8(10), 831–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Seranova E, Connolly KJ, Zatyka M, Rosenstock TR, Barrett T, Tuxworth RI, Sarkar S, 2017. Dysregulation of autophagy as a common mechanism in lysosomal storage diseases. Essays in biochemistry 61(6), 733–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Shoshan-Barmatz V, Nahon-Crystal E, Shteinfer-Kuzmine A, Gupta R, 2018. VDAC1, mitochondrial dysfunction, and Alzheimer’s disease. Pharmacological research 131, 87–101. [DOI] [PubMed] [Google Scholar]
  222. Silva LC, Ben David O, Pewzner-Jung Y, Laviad EL, Stiban J, Bandyopadhyay S, Merrill AH Jr., Prieto M, Futerman AH, 2012. Ablation of ceramide synthase 2 strongly affects biophysical properties of membranes. Journal of lipid research 53(3), 430–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Singh IN, Hall ED, 2008. Multifaceted roles of sphingosine-1-phosphate: how does this bioactive sphingolipid fit with acute neurological injury? Journal of neuroscience research 86(7), 1419–1433. [DOI] [PubMed] [Google Scholar]
  224. Sivasubramanian M, Kanagaraj N, Dheen ST, Tay SS, 2015. Sphingosine kinase 2 and sphingosine-1-phosphate promotes mitochondrial function in dopaminergic neurons of mouse model of Parkinson’s disease and in MPP+ -treated MN9D cells in vitro. Neuroscience 290, 636–648. [DOI] [PubMed] [Google Scholar]
  225. Smilansky A, Dangoor L, Nakdimon I, Ben-Hail D, Mizrachi D, Shoshan-Barmatz V, 2015. The Voltage-dependent Anion Channel 1 Mediates Amyloid beta Toxicity and Represents a Potential Target for Alzheimer Disease Therapy. The Journal of biological chemistry 290(52), 30670–30683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Smith GS, Kumar A, Saba JD, 2013. Sphingosine Phosphate Lyase Regulates Murine Embryonic Stem Cell Proliferation and Pluripotency through an S1P/STAT3 Signaling Pathway. Biomolecules 3(3), 351–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Snider AJ, 2013. Sphingosine kinase and sphingosine-1-phosphate: regulators in autoimmune and inflammatory disease. Int J Clin Rheumtol 8(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Snider AJ, Orr Gandy KA, Obeid LM, 2010. Sphingosine kinase: Role in regulation of bioactive sphingolipid mediators in inflammation. Biochimie 92(6), 707–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Song DD, Zhang TT, Chen JL, Xia YF, Qin ZH, Waeber C, Sheng R, 2017. Sphingosine kinase 2 activates autophagy and protects neurons against ischemic injury through interaction with Bcl-2 via its putative BH3 domain. Cell death & disease 8(7), e2912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Song DD, Zhou JH, Sheng R, 2018. Regulation and function of sphingosine kinase 2 in diseases. Histology and histopathology 33(5), 433–445. [DOI] [PubMed] [Google Scholar]
  231. Sorensen SD, Nicole O, Peavy RD, Montoya LM, Lee CJ, Murphy TJ, Traynelis SF, Hepler JR, 2003. Common signaling pathways link activation of murine PAR-1, LPA, and S1P receptors to proliferation of astrocytes. Mol Pharmacol 64(5), 1199–1209. [DOI] [PubMed] [Google Scholar]
  232. Spassieva S, Bieberich E, 2016. Lysosphingolipids and sphingolipidoses: Psychosine in Krabbe’s disease. Journal of neuroscience research 94(11), 974–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Spassieva SD, Ji X, Liu Y, Gable K, Bielawski J, Dunn TM, Bieberich E, Zhao L, 2016. Ectopic expression of ceramide synthase 2 in neurons suppresses neurodegeneration induced by ceramide synthase 1 deficiency. Proceedings of the National Academy of Sciences of the United States of America 113(21), 5928–5933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Spiegel S, Cuvillier O, Edsall L, Kohama T, Menzeleev R, Olivera A, Thomas D, Tu Z, Van Brocklyn J, Wang F, 1998a. Roles of sphingosine-1-phosphate in cell growth, differentiation, and death. Biochemistry (Mosc) 63(1), 69–73. [PubMed] [Google Scholar]
  235. Spiegel S, Cuvillier O, Edsall LC, Kohama T, Menzeleev R, Olah Z, Olivera A, Pirianov G, Thomas DM, Tu Z, Van Brocklyn JR, Wang F, 1998b. Sphingosine-1phosphate in cell growth and cell death. Annals of the New York Academy of Sciences 845, 11–18. [DOI] [PubMed] [Google Scholar]
  236. Spiegel S, Milstien S, 2011. The outs and the ins of sphingosine-1-phosphate in immunity. Nat Rev Immunol 11(6), 403–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Stadelmann C, 2011. Multiple sclerosis as a neurodegenerative disease: pathology, mechanisms and therapeutic implications. Curr Opin Neurol 24(3), 224–229. [DOI] [PubMed] [Google Scholar]
  238. Stiban J, Tidhar R, Futerman AH, 2010. Ceramide synthases: roles in cell physiology and signaling. Advances in experimental medicine and biology 688, 60–71. [DOI] [PubMed] [Google Scholar]
  239. Stoffel W, Hammels I, Jenke B, Binczek E, Schmidt-Soltau I, Brodesser S, Schauss A, Etich J, Heilig J, Zaucke F, 2016. Neutral sphingomyelinase (SMPD3) deficiency disrupts the Golgi secretory pathway and causes growth inhibition. Cell death & disease 7(11), e2488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Sun X, Marks DL, Park WD, Wheatley CL, Puri V, O’Brien JF, Kraft DL, Lundquist PA, Patterson MC, Pagano RE, Snow K, 2001. Niemann-Pick C variant detection by altered sphingolipid trafficking and correlation with mutations within a specific domain of NPC1. American journal of human genetics 68(6), 1361–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Sural-Fehr T, Bongarzone ER, 2016. How membrane dysfunction influences neuronal survival pathways in sphingolipid storage disorders. Journal of neuroscience research 94(11), 1042–1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Suzuki K, Katzman R, Korey SR, 1965. Chemical Studies on Alzheimer’s Disease. Journal of neuropathology and experimental neurology 24, 211–224. [DOI] [PubMed] [Google Scholar]
  243. Taha TA, Mullen TD, Obeid LM, 2006. A house divided: ceramide, sphingosine, and sphingosine-1-phosphate in programmed cell death. Biochimica et biophysica acta 1758(12), 2027–2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Takashima A, 2006. GSK-3 is essential in the pathogenesis of Alzheimer’s disease. Journal of Alzheimer’s disease : JAD 9(3 Suppl), 309–317. [DOI] [PubMed] [Google Scholar]
  245. Takasugi N, Sasaki T, Suzuki K, Osawa S, Isshiki H, Hori Y, Shimada N, Higo T, Yokoshima S, Fukuyama T, Lee VM, Trojanowski JQ, Tomita T, Iwatsubo T, 2011. BACE1 activity is modulated by cell-associated sphingosine-1-phosphate. J Neurosci 31(18), 6850–6857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Taranova NP, Makarov A, Amelina OA, Luchakova OS, Loboda EB, Leikin IB, 1992. [The production of autoantibodies to nerve tissue glycolipid antigens in patients with traumatic spinal cord injuries]. Zhurnal voprosy neirokhirurgii imeni N. N. Burdenko(4–5), 21–24. [PubMed] [Google Scholar]
  247. Testai FD, Landek MA, Dawson G, 2004. Regulation of sphingomyelinases in cells of the oligodendrocyte lineage. Journal of neuroscience research 75(1), 66–74. [DOI] [PubMed] [Google Scholar]
  248. Toman RE, Payne SG, Watterson KR, Maceyka M, Lee NH, Milstien S, Bigbee JW, Spiegel S, 2004. Differential transactivation of sphingosine-1-phosphate receptors modulates NGF-induced neurite extension. The Journal of cell biology 166(3), 381–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Tomita T, 2017. Aberrant proteolytic processing and therapeutic strategies in Alzheimer disease. Advances in biological regulation 64, 33–38. [DOI] [PubMed] [Google Scholar]
  250. Tommasino C, Marconi M, Ciarlo L, Matarrese P, Malorni W, 2015. Autophagic flux and autophagosome morphogenesis require the participation of sphingolipids. Apoptosis : an international journal on programmed cell death 20(5), 645–657. [DOI] [PubMed] [Google Scholar]
  251. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, Schwille P, Brugger B, Simons M, 2008. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 319(5867), 1244–1247. [DOI] [PubMed] [Google Scholar]
  252. van Echten-Deckert G, Alam S, 2018. Sphingolipid metabolism - an ambiguous regulator of autophagy in the brain. Biological chemistry 399(8), 837–850. [DOI] [PubMed] [Google Scholar]
  253. Vance JE, Hayashi H, Karten B, 2005. Cholesterol homeostasis in neurons and glial cells. Seminars in cell & developmental biology 16(2), 193–212. [DOI] [PubMed] [Google Scholar]
  254. Vu TM, Ishizu AN, Foo JC, Toh XR, Zhang F, Whee DM, Torta F, Cazenave-Gassiot A, Matsumura T, Kim S, Toh SES, Suda T, Silver DL, Wenk MR, Nguyen LN, 2017. Mfsd2b is essential for the sphingosine-1-phosphate export in erythrocytes and platelets. Nature 550(7677), 524–528. [DOI] [PubMed] [Google Scholar]
  255. Wang E, Norred WP, Bacon CW, Riley RT, Merrill AH Jr., 1991. Inhibition of sphingolipid biosynthesis by fumonisins. Implications for diseases associated with Fusarium moniliforme. The Journal of biological chemistry 266(22), 14486–14490. [PubMed] [Google Scholar]
  256. Wang G, Bieberich E, 2010. Prenatal alcohol exposure triggers ceramide-induced apoptosis in neural crest-derived tissues concurrent with defective cranial development. Cell death & disease 1(5), e46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Wang G, Dinkins M, He Q, Zhu G, Poirier C, Campbell A, Mayer-Proschel M, Bieberich E, 2012. Astrocytes secrete exosomes enriched with proapoptotic ceramide and prostate apoptosis response 4 (PAR-4): potential mechanism of apoptosis induction in Alzheimer disease (AD). The Journal of biological chemistry 287(25), 21384–21395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Wang G, Krishnamurthy K, Bieberich E, 2009. Regulation of primary cilia formation by ceramide. Journal of lipid research. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Wang G, Krishnamurthy K, Chiang YW, Dasgupta S, Bieberich E, 2008a. Regulation of neural progenitor cell motility by ceramide and potential implications for mouse brain development. Journal of neurochemistry 106(2), 718–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Wang G, Silva J, Dasgupta S, Bieberich E, 2008b. Long-chain ceramide is elevated in presenilin 1 (PS1M146V) mouse brain and induces apoptosis in PS1 astrocytes. Glia 56(4), 449–456. [DOI] [PubMed] [Google Scholar]
  261. Wang G, Silva J, Krishnamurthy K, Tran E, Condie BG, Bieberich E, 2005. Direct binding to ceramide activates protein kinase Czeta before the formation of a pro-apoptotic complex with PAR-4 in differentiating stem cells. The Journal of biological chemistry 280(28), 26415–26424. [DOI] [PubMed] [Google Scholar]
  262. Wang G, Spassieva SD, Bieberich E, 2018. Ceramide and S1P Signaling in Embryonic Stem Cell Differentiation. Methods in molecular biology 1697, 153–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Wang YM, Seibenhener ML, Vandenplas ML, Wooten MW, 1999. Atypical PKC zeta is activated by ceramide, resulting in coactivation of NF-kappaB/JNK kinase and cell survival. Journal of neuroscience research 55(3), 293–302. [DOI] [PubMed] [Google Scholar]
  264. Ward A, Crean S, Mercaldi CJ, Collins JM, Boyd D, Cook MN, Arrighi HM, 2012. Prevalence of apolipoprotein E4 genotype and homozygotes (APOE e4/4) among patients diagnosed with Alzheimer’s disease: a systematic review and meta-analysis. Neuroepidemiology 38(1), 1–17. [DOI] [PubMed] [Google Scholar]
  265. Xing Y, Tang Y, Zhao L, Wang Q, Qin W, Ji X, Zhang J, Jia J, 2016. Associations between plasma ceramides and cognitive and neuropsychiatric manifestations in Parkinson’s disease dementia. Journal of the neurological sciences 370, 82–87. [DOI] [PubMed] [Google Scholar]
  266. Yu RK, Tsai YT, Ariga T, 2012. Functional roles of gangliosides in neurodevelopment: an overview of recent advances. Neurochemical research 37(6), 1230–1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Yuyama K, Igarashi Y, 2017. Exosomes as Carriers of Alzheimer’s Amyloid-ss. Frontiers in neuroscience 11, 229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Yuyama K, Sun H, Mitsutake S, Igarashi Y, 2012. Sphingolipid-modulated exosome secretion promotes the clearance of amyloid-beta by microglia. The Journal of biological chemistry. [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Yuyama K, Sun H, Usuki S, Sakai S, Hanamatsu H, Mioka T, Kimura N, Okada M, Tahara H, Furukawa J, Fujitani N, Shinohara Y, Igarashi Y, 2015. A potential function for neuronal exosomes: sequestering intracerebral amyloid-beta peptide. FEBS letters 589(1), 84–88. [DOI] [PubMed] [Google Scholar]
  270. Zhao P, Yang X, Yang L, Li M, Wood K, Liu Q, Zhu X, 2017. Neuroprotective effects of fingolimod in mouse models of Parkinson’s disease. FASEB J 31(1), 172–179. [DOI] [PubMed] [Google Scholar]

RESOURCES