<<

Pharmacological Reports 68 (2016) 570–581

Contents lists available at ScienceDirect

Pharmacological Reports

jou rnal homepage: www.elsevier.com/locate/pharep

Review article

Regulation of

a a,b a a

Kamil Bienias , Anna Fiedorowicz , Anna Sadowska , Sławomir Prokopiuk , a,

Halina Car *

a

Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland

b

Laboratory of Tumor Molecular Immunobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław,

Poland

A R T I C L E I N F O A B S T R A C T

Article history: (SFs) represent a large class of playing diverse functions in a vast number of

Received 2 April 2015

physiological and pathological processes. Sphingomyelin (SM) is the most abundant SF in the cell, with

Received in revised form 24 November 2015

ubiquitous distribution within mammalian tissues, and particularly high levels in the Central Nervous

Accepted 28 December 2015

System (CNS). SM is an essential element of plasma membrane (PM) and its levels are crucial for the cell

Available online 11 January 2016

function. SM content in a cell is strictly regulated by the of SM metabolic pathways, which

activities create a balance between SM synthesis and degradation. The de novo synthesis via SM

Keywords:

synthases (SMSs) in the last step of the multi-stage process is the most important pathway of SM

Sphingomyelin

formation in a cell. The SM hydrolysis by sphingomyelinases (SMases) increases the concentration of

Sphingomyelin synthases

(Cer), a bioactive molecule, which is involved in cellular proliferation, growth and apoptosis. By

Acid sphingomyelinase

Neutral sphingomyelinase controlling the levels of SM and Cer, SMSs and SMases maintain cellular homeostasis. Enzymes of SM

cycle exhibit unique properties and diverse tissue distribution. Disturbances in their activities were

observed in many CNS pathologies. This review characterizes the physiological roles of SM and enzymes

controlling SM levels as well as their involvement in selected pathologies of the Central Nervous System,

such as ischemia/hypoxia, Alzheimer disease (AD), Parkinson disease (PD), depression, schizophrenia

and Niemann Pick disease (NPD).

ß 2016 Institute of Pharmacology, Polish Academy of Sciences. Published by Elsevier Sp. z o.o. All rights reserved.

Contents

Physiological roles of sphingomyelin (SM) and enzymes of SM cycle...... 571

SM – structure and function ...... 571

Sphingomyelin synthases (SMSs) ...... 572

SMS1 ...... 572

SMS2 ...... 572

SMSr...... 572

Sphingomyelinases (SMases) ...... 572

Acid Sphingomyelinase (a-SMase) ...... 572

Neutral sphingomyelinase (n-SMase) ...... 573

Alkaline SMase (alk-SMase) ...... 573

Abbreviations: a-syn, a-synuclein; Ab, amyloid b; AD, Alzheimer disease; APP, amyloid precursor protein; a-SMase, acid sphingomyelinase; alk-SMase, Alkaline SMase; Cer,

ceramide; CERT, ceramide transfer protein; CSF, cerebrospinal fluid; D609, tricyclodecan-9-yl-xanthogenate; DAG, diacyglicerol; ER, endoplasmic reticulum; FasL, Fas ligand;

FIASMA, Functional Inhibitor of Acid SphingoMyelinAse; GSH, glutathione; IR, ischemia/reperfusion; l-SMaze, sphingomyelinase; MA-nSMase, mitochondrial

sphingomyelinase; MUFA, monounsaturated fatty acid; n-SMase, neutral sphingomyelinase; NPD, Niemann–Pick disease; PC-PLC, specific phospholipase C; PD, Parkinson

disease; PM, plasma membrane; PUFA, polyunsaturated fatty acids; ROS, reactive oxygen species; SAFA, saturated fatty acids; SAM, sterile motif; SF, ; SM,

sphingomyelin; SMase, sphingomyelinase; s-SMase, secretory sphingomyelinase; SMS, sphingomyelin synthase; SPT, serine palmitoyltransferase; START, protein-related

transfer domain; TNF-a, tumor necrosis factor-a; TNFR1, tumor necrosis factor receptor 1.

* Corresponding author.

E-mail address: [email protected] (H. Car).

http://dx.doi.org/10.1016/j.pharep.2015.12.008

1734-1140/ß 2016 Institute of Pharmacology, Polish Academy of Sciences. Published by Elsevier Sp. z o.o. All rights reserved.

B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581 571

SM metabolism in selected pathologies of the CNS ...... 573

Ischemia/reperfusion ...... 573

Alzheimer disease (AD) ...... 574

Depression and schizophrenia ...... 576

Parkinson disease (PD) ...... 576

Niemann–Pick disease (NPD) ...... 576

Summary ...... 577

Conflict of interest ...... 577

Funding ...... 577

References ...... 577

Physiological roles of sphingomyelin (SM) and enzymes of SM stronger binding with cholesterol and form larger domains in

cycle comparison with racemic layers [15]. Importantly, SMs with

different fatty acid chains are not equally distributed in

SM – structure and function mammalian tissues [3], which results in unique compositions of

lipid rafts. It is worth noting that a myriad of signaling,

SM is the most abundant eukaryotic sphingolipid (SF) which cytoskeletal, mitochondrial [16] and GPI-anchored proteins have

constitutes one of the major components of the plasma membrane been discovered within lipid rafts [17].

(PM). The structural function of SM in a cell is determined by The de novo synthesis of SM is a multi-stage process which

chemical structure of a phospholipid. Specifically, the hydrophilic begins on the cytosolic side of the endoplasmic reticulum (ER) with

head and the hydrophobic tail of the phospholipid molecules the condensation of palmitoyl-CoA and serine through the action

enable them to form a double layer structure, known as a lipid of serine palmitoyltransferase (SPT). This reaction results in the

bilayer. SM (N-acyl--1-phosphorylcholine) is com- production of 3-ketosfinganine which is subsequently transformed

posed of ceramide which comprises a sphingoid backbone, an 18- into dihydrosphingosine (sphinganine) by 3-ketosphinganine

carbon aminoalcohol with one trans-double bond in position 4 [1] reductase. Next, the fatty acid chain is attached to sphinganine

with an attached hydrophobic fatty acid chain, and polar by dihydroceramide synthase to produce dihydroceramide. The

phosphocholine or, less frequently, phosphoethanolamine resi- reduction of a double bond in a molecule of dihydroceramide by

dues [2]. The naturally occurring SMs vary in their length of fatty dihydroceramide desaturase leads to the formation of ceramide

acid chain (usually from 16 to 24 carbons) and the level of their (Cer) [18]. For SM synthesis, Cer is transferred from ER to the Golgi

saturation and hydrogenation. Thus, SM can contain the saturated apparatus [19] by both vesicular and non-vesicular transport

fatty acids (SAFA), monounsaturated fatty acids (MUFA) and [20]. The latter is mediated by the ceramide transfer protein

polyunsaturated fatty acids (PUFA) [3,4]. The specific roles of the (CERT), which contains a steroidogenic acute regulatory protein-

diverse SM species in PM are poorly established, although it seems related lipid transfer domain (START), the site of Cer binding

that SM containing different fatty acids may regulate membrane [21]. Studies on the role of CERT in SM formation showed a

fluidity. There are some findings validating this statement, significant reduction in SM levels in CERT-deficient mouse

however, obtained mostly in the studies of artificial model embryos [22]. These findings point to a conclusion that the de

systems. For example, it was observed that MUFA-containing novo synthesis is the most important pathway of SM creation in the

species of SMs form more liquid domains than those with SAFA cell. Moreover, transport of Cer to the Golgi is considered as the

[5]. Furthermore, hydrogenation of the SM double bond results in rate limiting factor of the de novo SM synthesis [21]. In the Golgi

an increase in membrane raft melting temperature [6]. network, the phosphorylocholine group from phosphatidylo-

Being a major SF in PM, SM has the ability of binding cholesterol choline is transferred to Cer by sphingomyelin synthase 1

[7]. This feature of SM is particularly important in the formation of (SMS1), which eventually results in the formation of SM along

lipid rafts, which are cholesterol-enriched PM domains. The with the production of diacyglycerol (DAG) [23]. Thereafter,

specific composition of a lipid raft determines its gel ordered newly synthetized SM reaches PM in transport vesicles [24]. In

structure, which is separated from other parts of the membrane, the cell membrane, SM takes part in the lipid raft formation [5] or

thicker than the surroundings [8] and resistant to detergents undergoes hydrolysis. The common degradation process of SM

[9]. From 40% to 70% of cell SFs are gathered in lipid rafts [10], occurs in the lysosome, where SM, transported mainly from the

where SM represents approximately 2% of the total SF content. PM, is hydrolysed to Cer by the acidic SMase (a-SMase) [25]. The

Interestingly, SM accounts for 2%–15% of the total concentration of PM-localized SM can also be enzymatically degraded to Cer and

SFs in the cell [11]. Localization of a double bond in the SM fatty phosphocholine by the hydrolysis of phosphodiester bond by

acid chain influences the interactions of this lipid with cholesterol. two sphingomyelinases (SMases): secretory (s-SMase) or

13 15

Therefore, while double bonds at D or D positions do not affect neutral (n-SMase) SMase [26], which is often observed in

9

the SM binding with cholesterol, the double bond at D position pathological processes. Interestingly, Cers derived from SM

weakens this reaction. On the other hand, cholesterol does not hydrolysis can be reutilized for SM synthesis by sphingomyelin

seem to favor any SM species on the basis of their specific acyl synthase 2 (SMS2) [27].

chain length [12]. In addition, the interactions of cholesterol with Since SM is an essential modulator of PM properties which

the NH2 group of SM protect this lipid from oxidative degradation influences the membrane gathering of proteins involved in cellular

[13]. It has been reported that SM species with N-acyl branched proliferation, growth and apoptosis as well as being an important

chains form more fluid and disordered lipid rafts compared to source of ceramide, its levels are critical for cell function. The SM

domains composed of unbranched SMs. Furthermore, the interac- content in a cell is strictly regulated by the enzymes of SM metabolic

tion of cholesterol with SM is impaired when SM N-acyl chains are pathways whose activities create a balance between SM synthesis

10

branched at D position [14]. Natural SM is optically active and and degradation. This manuscript will characterize the physiological

occurs in two isomers, d-erythro SM and l-threo SM. Monolayers roles of these enzymes as well as their involvement in selected

containing only one isomer are tighter, more ordered, exhibit pathologies of the Central Nervous System.

572 B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581

Sphingomyelin synthases (SMSs) [29]. The binding of SMS2 to PM is enabled by prior palmitoylation

of its COOH-terminal tail [47]. The highest content of SMS2 was

SMSs catalyze the reaction of SM synthesis from Cer. They found in the human brain, heart, kidney, , muscle and stomach

transfer the phosphorylocholine group from phosphatidylcholine [34]. The in vivo activity of this covers 20%–40% of all SMSs

on Cer in conjunction with the production of DAG. Recent reports activity in the cell [31,32], is cell specific and depends on the

have revealed that, apart from the production of SM, SMSs are availability of substrates [48]. Importantly, the major substrates

involved in the synthesis of the SM analog, ceramide phos- for SMS2 are Cers derived from the degradation of more complex

phoethanolamine (CPE) [28]. In addition, the synthesis of CPE can SFs, which is reflected by the predominantly membrane localiza-

be catalyzed by the sphingomyelin related protein (SMSr). To date, tion of SMS2 [34]. These characteristics of SMS2 strongly indicate

two SM synthases have been described, SMS1 and SMS2. its antiapoptotic activities. In other words, the overproduction of

Structurally, both SMSs are over 50% identical. Both enzymes cytotoxic Cers by SM hydrolysis in the PM is reduced by their

also share similarities in the C-terminus, responsible for their reutilization in SM synthesis which may constitute an important

localisations [29] as well as in the D3 and D4 motifs, essential for protective mechanism in the cell. Similarly to SMS1, SMS2

their catalytic activities. One of the most striking structural catalyses the reactions of both SM and CPE synthesis. The dual

differences between the two SMSs is the presence of SAM (sterile substrate specificity of SMS2 suggests a potential regulatory

motif) domain [30] in SMS1, which is absent in SMS2. The SAM mechanism of its activity. Thus, the multi-domain of SMS2 is

motif was considered essential for binding SMS1 to the Golgi oriented toward the exoplasmic leaflet of PM, where its activity can

apparatus [7]. However, recent findings have revealed that SAM is be negatively regulated by the PE. In physiological conditions, a

responsible neither for Golgi targeting nor for SMS1 activation low level of PE on the exoplasmic leaflet stimulates a SMS2-

[30]. catalised SM synthesis. In pathology, the structure of PM is

disordered and an increased concentration of PE moves SMS2

SMS1 activity toward the production of CPE instead of SM, which may

SMS1 activity covers from 60% to 80% of all SMS activity and is result in Cer accumulation [49]. Data on SMS2 physiological

limited by the substrate availability [31,32]. Among all human functions in the literature is rather scarce. Some important

tissues, the highest levels of SMS1 were reported in the kidney, observations have been made in the nervous system, in which

lung and liver [33]. SMS1 is encoded by the SGMS 1 gene [34], SMS2 is highly expressed. Consequently, the expression of SMS2

which is translated into 413-amino acid protein. The newly only (but not SMS1) has been registered in hippocampal neurites.

synthetized SMS1 is then transported to the Golgi apparatus, The localization of the enzyme in the proximity of SM clusters

where it catalyses the reaction of the de novo synthesis of SM suggests its involvement in creating the micro-domains anchoring

[7,31,34,35]. SMS1 is essential for maintaining optimal levels of signal transduction proteins [50]. Interestingly, Zhang et al.

SM in a cell since it is one of the major enzymes required for SM reported that SMS2 may be engaged in drug transport in the

synthesis. Hence, SMS1 is implicated in many key cellular brain. They found decreased levels of the drug transporter, Pgp

processes such as proliferation, cell growth, migration or apoptosis (Mdr1/P-glycoprotein), ezrin and b-actin in some cerebral areas in

[36–38]. It was demonstrated that SMS1 is engaged in astrocyte SMS2 knockout mice [51].

proliferation induced by basic fibroblast growth factor (bFGF)

[39]. The exact mechanism of SMS1 involvement in proliferation Sphingomyelin synthase related protein (SMSr)

remains unclear. However, it is hypothesized that due to the SMSr is encoded by the SAMD8 gene and gene product is located

functional similarities of SMS1 and phosphatidylcholine specific in the endoplasmic reticulum [35]. SMSr catalyses the reaction of

phospholipase C (PC-PLC) [40], SMS1 may act by activating CPE synthesis from Cers, but its activity is very small in comparison

tyrosine kinases as it was observed for PC-PLC [41]. SMS1 has also with canonical SMSs [52]. Since SMSr is not directly involved in SM

been found to be required for maintaining cell morphology, production [28], its further description is beyond the scope of this

growth and migratory potential. This was reported in the Neuro- article.

2a cell line of murine neuroblasts, where SMS1 exerted its activity

through Akt kinase and cyclin D1 signaling [36]. The complex roles Sphingomyelinases (SMases)

which SMS1 can play in a cell have been vividly demonstrated in

studies on immune cells. Taguchi et al. showed that the IL-2- SMases (Sphingomyelin phosphodiesterases) catalyze the

mediated survival of natural killer cells depends on SMS1 reaction of SM hydrolysis. They hydrolyse the phosphodiester

activation which was suggested to occur through the action of bond of SM to generate Cer and phosphocholine. Three types of

phosphatidylinositol-3 kinase [42]. In addition, SMS1 controls the SMases can be distinguished on the basis of optimal pH values

fate of T cells. Dong et al. reported that SMS1, by involvement in required for their activation: acid (a-SMase), alkaline (alk-SMase)

lipid raft formation, provides sites for T cell activation through and neutral (n-SMase).

tyrosine phosphorylation [43]. SMS1 may also perform a

protective action toward the apoptosis of T cells evoked by Fas Acid Sphingomyelinase (a-SMase)

ligand (FasL) [38], presumably preventing Cer accumulation in the a-SMases encoded by the SMPD1 gene and translated into 629-

cells. Apart from controlling Cer levels, SMS1 regulates the aminoacid pro-protein [25], which gives rise to lysosomal (l-

concentration of the multifunctional lipid, DAG may play a SMase) and secretory (s-SMase) a-SMases [53]. Lysosomal locali-

structural function in the PM [44], but primarily it is an important zation of the enzyme requires the attachment of mannose residues

lipid second messenger, crucial in cellular proliferation and to the immature form of a-SMase, whereas lack of this modification

differentiation processes [45]. Moreover, DAG is a precursor of in the a-SMase precursor protein leads to its secretion to the

more complex lipids such as phosphatidylethanolamine and extracellular space [54]. Both l-SMase and s-SMase are equipped

phosphatidylcholine [46]. with six potential glycosylation sites which are essential for the

activities of the enzymes and their anti-proteolytic protection

SMS2 [55]. a-SMases reach their full activity in an acidic environment,

SMS2 is a product of the SGMS2 gene [34]. It is an integral however, s-SMase can also act in neutral pH [56]. Furthermore, in

protein of PM, consisting of six trans-membrane domains and the contrast to s-SMase, [57] the activation of l-SMase do not depend

C-terminal catalytic region, localized in the outer leaflet of the PM on zinc ion availability in their environment [58].

B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581 573

The biological functions of a-SMases have been extensively metabolism remains controversial. According to Sawai et al., the

studied, although the vast majority of the reports have focused on overexpression of n-SMase1 in the MCF-7 cell line did not influence

the enzyme activities in pathological conditions and only few SM content, but rather contributed to the lyso-platelet-activating

examples of physiological roles of a-SMases exist in the literature factor (lyso-PAF) of metabolism [82]. Moreover, n-SMase1-

and refer mostly to s-SMase. Active forms of s-SMase were found in deficient mice exhibited a lack of alterations in SM levels

synovial, salivary and cerebrospinal (CSF) fluids, as well as in the [83]. On the other hand, n-SMase1 was reported to mediate T

sera, tears and urine of healthy people [59,60]. It has been cell receptor signaling through the production of Cers and

demonstrated that s-SMase plays an important role in the subsequent apoptosis [84]. Furthermore, zebrafish studies

maintenance of platelet homeostasis as the enzyme is secreted revealed an engagement of n-SMase1 in apoptosis induced by

by thrombin-stimulated platelets [61,62]. Moreover, s-SMase was heat stress [85,86], which was stimulated by prior phosphorylation

reported to enhance glucose influx into platelets [63], ATP release, of the enzyme by c-Jun N-terminal kinase (JNK) [86]. Interestingly,

and thrombus formation. s-SMase, by regulating the properties of novel findings by Guo et al. indicate that n-SMase1 is partially

platelet PM, influences phosphatidylserine translocation to PM, responsible for exosome biogenesis in the hypothalamic cell line

which is the central site of thrombin production regulation [64]. In [87].

addition, a-SMase mediates translocation of phospholipase A2 to The third n-SMase, n-SMase3, is a protein of 866 amino acids,

PM in platelet cells, which is most probably facilitated by PM localized to ER and trans-Golgi compartments. The strongest

rearrangement through the a-SMase- mediated ceramide produc- expression of n-SMase3 has been noted in striated and cardiac

2+

tion [65]. muscles. Similarly to other n-SMases, n-SMase3 is a Mg -

dependent enzyme. Despite the fact that n-SMase3 shares no

Neutral sphingomyelinase (n-SMase) structural homology with other n-SMases, it exhibits activity

n-SMase are encoded by four genes, SMPD2–5 which are toward SM hydrolysis. To date, data concerning the functional

translated into four proteins, n-SMase1, n-SMase2, n-SMase3 roles of n-SMase3 have been limited, although it has been shown to

and mitochondria-associated SMase (MA-SMase) [66]. be upregulated in MCF-7 cells under TNF-a treatment [88]. In

The best characterized n-SMase is n-SMase2, which is a 655- addition, the n-SMase3 gene was overexpressed in colon cancer

amino acid protein containing the C-terminal catalytic domain and cells treated with genotoxic agents [89]. Novel findings revealed

two hydrophobic segments on its N-terminus [67]. The enzyme that n-SMase3 mediated the TNF-a-induced oxidant production in

also has two palmitoyl-binding cysteine clusters, the first located skeletal muscle [90].

in the hydrophobic region of the protein and the second in its Identified as the last n-SMase, MA-SMase, is a 483-amino acid

active center. Palmitoylation of cysteine clusters is important for protein, localized in ER and mitochondria [91,92]. MA-SMase

the stability of n-SMase2 and its binding to PM [68]. Activation of expression has been found in the testis, brain, pancreas and

2+

n-SMase2 occurs in neutral pH in the presence of Mg ions. n- epididymis. Lower levels of the enzyme have also been noted in the

SMase2 is ubiquitously expressed within mammalian tissues spleen, thymus and skin. The structure of MA-SMase is similar to

2+

with the highest content in the brain [66]. Interestingly, Crivello that of n-SMase2, and similarly to the latter enzyme, requires Mg

et al. found that n-SMase2 activity in the brain underwent ions for its full activation. There are only few reports on the role of

changes during aging. They observed an increased activity of this MA-SMase in the cell. It was demonstrated that MA-SMase can

enzyme in the PM isolated from striatum, hippocampus and the mediate apoptosis by regulating Cer levels in the mitochondria

frontal cortex of aged brains [69]. n-SMase2 plays a multitude of [93]. On the other hand, the predominant expression of MA-SMase

roles in the CNS. Stoffel et al. demonstrated that the active form of in the testis [91] may indicate its essential role in fertilization

n-SMase2 was required for embryonic and postnatal develop- processes. Sperm contains considerable amounts of SM which

ment by influencing the hypothalamic function [70]. n-SMase2 decrease significantly during fertilization [94]. MA-SMase, by SM

was also reported to mediate the trafficking of NMDA receptors to hydrolysis, increases the level of Cers which are suspected to

PM, which may suggest its function in synaptic plasticity induce membrane fusion of spermatozoa with the ovum [95].

[71]. Furthermore, n-SMase2 can augment dopamine uptake

by the PC12 cells through Cer production and increase the Alkaline SMase (alk-SMase)

intracellular calcium level [72]. The n-SMase2 mediated regula- Alk-SMase is encoded by the ENPP7 gene and translated into

tion of intracellular calcium concentration goes beyond the CNS 458-aminoacid protein which is equipped with two hydrophobic

cells. In canine cerebrovascular smooth muscle cells, n-SMase2 domains on both ends of the protein, essential for signaling and

activation led to an enhancement of intracellular influx of anchoring of the enzyme [96].

calcium required for proper contraction of the arteries [73]. In Alk-SMase differs structurally from the remaining SMases and

addition, Bautista-Pe´ rez et al. noted that the process of renal belongs to a different family of enzymes, nucleotide-pyropho-

vasoconstriction induced by angiotensin II involved Cers sphatase/phosphodiesterase [96]. It is not activated by divalent

2+

originated from the n-SMase2 catalised SM hydrolysis [74]. On ions, but inhibited by Zn . In physiological terms, localization of

the other hand, n-SMase2 was reported to stimulate the alk-SMaseis limited to the intestine [97], where it digests

production of nitric oxide by endothelial cells, which induced exogenous SM [98].

vasorelaxation of the coronary arteries [75]. The important roles

of n-SMase2 were also found in T cell co-stimulation [76] as well SM metabolism in selected pathologies of the CNS

as skeletal tissue homeostasis reviewed by [77], where n-SMase2

contributed to proper bone mineralization [78] and cartilage Ischemia/reperfusion

development [79].

In contrast to n-SMase2, the physiological functions of other n- Cerebral ischemia is a condition of decreased tissue perfusion

SMases have been poorly researched. n-SMase1 is a 423-amino resulting in an inadequate supply of oxygen, glucose and other

2+

acid protein which requires Mg ions to perform its enzymatic metabolites, and the subsequent reperfusion triggering multiple

activity [67]. n-SMase1 localizes predominantly to ER, but it has signaling cascades, which leads to brain injury. Various factors/

also been found in the Golgi apparatus [80] and the nucleus [81]. It pathways have been described in the literature as crucial for

is broadly expressed in human tissues such as the intestine, kidney, ischemia-induced neurodegeneration. Numerous changes, among

brain, liver, heart and lung [67]. Involvement of n-SMase1 in SM others, have been observed in SM metabolism, frequently noted as

574 B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581

essential for neuronal death or survival in ischemia/reperfusion accompanied by reduced ceramide utilization via glucosylcera-

(IR) processes. It was demonstrated that focal brain ischemia mide synthase [108]. Cer production, catalyzed by SMases, leads to

stimulated an early and irreversible SM hydrolysis and Cer alterations of SM profiles in a tissue. It was noted that the levels of

generation in the cerebral cortex [99]. Similarly, Nakane et al. almost all studied SM species were changed in NT-2 neuronal

described lethal forebrain ischemia in gerbil, an SM degradation precursor cells during hypoxia [112]. Likewise, the concentrations

leading to an increase in Cer levels in the hippocampus of the total SM and those SM species containing long chain fatty

[100]. Interestingly, it was shown that ischemia-induced Cers acids (C16–18) were elevated while the levels of very long chain

were localized to the astrocytic PMs in CA1 and DG regions of the (24C-) SMs were reduced in the white matter of postmortem

hippocampus, which suggested that the SMase pathway was frozen brain tissues of patients with subcortical ischemic vascular

involved in Cer production under ischemic conditions [101]. Soeda dementia D (SIVD) [113], the results being similar to those

et al. described in the pheochromocytoma cells (line PC-12) model obtained in the entorhinal cortex of AD human brains [114]. Im-

that the n-SMase activity peaked at 1 h and the increased activity portantly, the SM derangements observed in SIVD white matter

lasted thereafter for several hours. This observation was supported closely resembled the brain lipid profiles of synthase 2

by data that the difluoromethylene analogs of sphingomyelin (CerS2)-null mice [115]. n-SMase appears to be activated by

(SMAs, especially SMA-7), which inhibit of n-SMase but not multiple stimuli but the molecular mechanisms for this regulation

ceramide synthase in the serum/glucose-deprived pheochromo- are not fully understood. A study by Gu et al. suggested that

cytoma cells line PC-12, could prevent rapid ceramide production ischemia-induced n-SMase2 activation might be partially depen-

and that they significantly reduced the size of cerebral infarcts in dent on the TNF-aR signaling pathway [106]. It was described that

mice [102]. tumor necrosis factor (TNF) released during an ischemic stroke

Magnesium-dependent n-SMase2, mainly found in brain tissue, stimulated sphingomyelinase. n-SMase2 may be activated directly

is an important mediator of neuronal death induced by ischemia/ by binding to the TNF-a receptor/receptor for activated C kinase 1 –

hypoxia [102–106]. It was observed by Yoshimura et al., that the RACK1/embryonic ectoderm development – EED (TNF-aR/RACK1/

24-hour hypoxia of PC12 cells produced an increase in membrane- EED) in the early phase of ischemia [116,117]. However, the

bound n-SMase2 activities, but not in the activities of neutral inhibition of TNF-aR attenuated n-SMase2 activity to some extent,

magnesium-independent and acid SMases [104]. Comparative suggesting that the TNF-aR/RACK1/EED pathway plays a second-

analyses of n-SMase kinetics in PC12 cells cultured for 24 h under ary role in the upregulation of n-SMase2 activity in hippocampal

hypoxic conditions showed an elevated Vmax but not an altered astrocytes following ischemia.

Km of the enzyme [104]. In the in vivo model, Gu et al. observed It was observed that the activation of n-SMase was closely

early accumulation of Cers after 30-min cerebral ischemia due to linked to the initial depletion of glutathione (GSH) [118] and a high

the upregulation of n-SMase2 but not a-SMase activities [107]. The dose of vitamin E (50 or 100 mM) markedly reduced the activity of

downregulation of n-SMase2 by either a chemical inhibitor this enzyme in rat neurons during hypoxia and re-oxygenation

(GW4869) or siRNA, used prior to forebrain ischemia induction, [119]. Won and Singh stated that cellular redox potential and

reduced Cer production in rat hippocampal astrocytes and partially reactive oxygen species (ROS), which are regulated by GSH, were

reversed the neuronal damage as well as decreased mRNA levels of associated with SMase activation [120]. Similarly, Li et al. (2012)

pro-inflammatory cytokines [107]. In addition, based on the found ROS to be a factor activating a-SMase, which is in fact a

Transwell model, authors demonstrated that the effect of lysosomal enzyme important for cellular responses to ROS

activating the nSMase-2/ceramide pathway in the astrocytes can [121]. SMase activities in ischemia/hypoxia may also be regulated

be reversed by the inhibition of NF-kB signaling. Furthermore, the by the molecular pathways mediating cellular survival. In PC12

inhibition of a-SMAse with was not effective in cells over expressing Bcl-2, which exhibited strong resistance to

decreasing Cer levels. These findings suggest that at the early hypoxia, SM hydrolysis was decreased and n-SMase activation was

stages of cerebral ischemia, n-SMase2, but not a-SMase, is reduced [104]. Other studies have demonstrated that cerebral

responsible for SM hydrolysis in the rat hippocampus. In contrast, ischemia rapidly upregulated the p38 activity which in turn

other studies showed that the increase in Cer levels in ischemia is enhanced n-SMase2 activation by its phosphorylation [122]. More-

generated through the activation of both neutral and acid SMases over, p38 inhibition reversed ischemia-induced n-SMase2 activa-

[102,108]. The discrepancies among study findings presumably tion and reduced Cer levels [107]. In addition, A2B adenosine

resulted from different models of ischemia and experimental time receptor/p38 played an essential role in the activation of the n-

frames used by the authors. Thus, Ohtani et al. observed enhanced SMase2/ceramide pathway in rat hippocampus in response to

a-SMase activity 3 days after chronic cerebral ischemia induced by ischemia [107]. Under cerebral ischemia conditions, Cers are

clipping both common carotid arteries in rats [108]. The authors accumulated in the cells as a result of not only SMase over-activity

concluded that the hydrolysis of SM in the PM, catalyzed by n- but also a decreased expression of SMS1 mRNA in subcortex glia,

SMase2, was probably the first cellular response to ischemia and which is probably associated with massive necrotic cell death in

the subsequent over-activation of the lysosomal/endosomal a- the infarct area [114,123]. The return of the SMS1 mRNA level to

SMase led to cell death. Additionally, it was suggested that the baseline 72 h after occlusion, may indicate active processes of cell

deficiency of a-SMase and the dysfunction of heat shock protein proliferation.

Hsp 70.1 occurred in cerebral ischemia and it was closely related to The understanding of whether SM hydrolysis is a cause or result

lysosomal rupture and CA1 neuronal death within a 7-day period of ischemic damage is still poor. However, based on scientific

[109]. literature, it appears that SMases could be promising new targets

It appears that the mechanism of ceramide accumulation for drugs designed to reduce ischemic brain lesions, particularly as

depends on the severity of an insult to the brain. Severe and lethal the pharmacological inhibition of SMase activities has been shown

cerebral ischemia/reperfusion resulted in ceramide accumulation to protect neurons, oligodendrocytes and astrocytes against Cer-

via the activation of a-SMase and SM hydrolysis [99,100]. More- induced cell death caused by ischemia [102].

over, the extent of brain tissue damage was lower in mice lacking

a-SMase [110]. On the other hand, in mild IR, Cer accumulation Alzheimer disease (AD)

resulted from the de novo Cer biosynthesis rather from SM

hydrolysis [111]. Furthermore, in chronic cerebral ischemia, Cers AD is one of the most common age-associated dementia.

were generated due to the activation of a-SMase which was Pathomorphological features of AD involve formation of

B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581 575

neurofibrillary tangles, senile plaques with abnormal Ab symptomatic phases. In all experimental points, the levels of altered

(amyloid b) peptide accumulation and tau hyperphosphorylation SM species were reduced in the brains of APP/tau mice in

[124]. Although different aspects of AD are extensively studied, the comparison with controls, whereas in plasma of AD animals all

basic knowledge on the causes, diagnosis and treatment of this SM species were significantly elevated [137].

disease, is still lacking. SM metabolism represents a promising area An event initiating cellular changes toward AD development is

of research on AD-associated processes. The alterations in SM cycle still unknown, nevertheless it seems that one of the first steps in

were studied both in the context of seeking the potential diagnostic this process could be a structural reorganization of the PM. The

biomarkers as well as finding explanation of AD pathogenesis. alteration of PM properties could in turn lead to pathological Ab

Studying blood plasma/serum SM levels researchers obtained an processing, which occurs mainly in the lipid rafts. Ab, constitutes

incoherent results. Oresˇicˇ et al. reported that AD patients exhibited a the main component of amyloid plaques, is firstly cleaved from the

decreased content of serum SMs [125]. Mielke et al. analyzed the SM APP (amyloid precursor protein) by b-secretase. Further, Ab40 and

and Cer serum concentrations in one group of women being Ab42 peptides are produced by subsequent action of g-secretase

examined over 9 years of the study. They obtained fluctuations in the [138,139]. The highly altered lipid compositions of lipid rafts were

levels of studied SFs corresponding with the time of memory found in frontal and entorhinal cortices in non-symptomatic and

impairment onset. This suggested that both Cers and SMs could mild dementia phases, in AD brains [140]. In addition, the levels of

serve as potential biomarkers of AD development [126]. Another 9- SMs associated with the lipid rafts were significantly reduced in

year study, enrolling healthy women, showed a strong correlation frontal cortex at the early stages of AD. Authors hypothesized that

between increased Cer, but not SM, levels in serum of volunteers, such reorganization of lipid components at the beginning of the AD

and risk of AD development [127]. On the other hand, Kosicek et al. might provoke the Ab structural changes toward senile plaque

noted an augmentation of SM content in cerebrospinal fluid (CSF) of creation. Dı´az et al. did some important observations on how the

individuals with probable AD [128]. Furthermore, studies on physicochemical features of the lipid rafts in AD brains change

patients with prodromal, mild and moderate stages of AD revealed with the development of the malady [141]. Using steady-state

the increments of SMs multiple species in CSF, but only in those fluorescence polarization analyses, they reported more liquid-

participants with prodromal phase of the disease. Authors ordered and viscous structure of lipid rafts in frontal and

hypothesize that this early elevation of SMs in CSF of AD patients entorhinal cortices at the early stages of AD than in control brains.

could result from an increased level of Cers, which were Moreover, the authors stated that PM fluidity did not correlate

subsequently used to SM synthesis [129]. Similarly, Mielke et al. with the SM content, but rather with the changes in saturation

have shown in the cohort study, that SMs from CSF may participate status of the phospholipids [141]. Importantly, altered lipid raft

in early events of AD. They analyzed levels of Cers and SMs in CSF of structure can greatly influence the functional properties of

healthy individuals with parental history of AD and found the proteins anchored in the PM, such as secretase/AbPP (amyloid-

correlation between concentrations of all studied SM species and b protein precursor) and impact APP processing [142] toward

levels of Ab, total tau, and some forms of p-tau [130]. A changed SM promotion of b-amyloidosis. This may lead to the conclusion that

metabolism was also reported in AD brains. Early studies revealed a changes in total levels of SM are probably less relevant for AD

general decline of SM content in AD brains [131,132]. Further development than the species composition of this sphingolipid. On

investigations showed and an elevation of Cer levels and reduction the other hand, it was reported that Ab peptides can induce the n-

of SM concentrations in human gray matter of frontotemporal area SMase activation. Thus, treatment of rat primary astrocytes with

in AD patients [133]. These changes were associated with increased Ab25–35 peptide increased n-SMase activity [143], whereas Ab42

activity and protein expression of a-SMase in membrane, but not activated n-SMase and decreased SM levels, which was modulated

cytosolic fraction of the tissue extracts. Taking into account that any by g-secretase activity [144].

apparent changes were reported for n-SMase, it is possible that in An interesting relation between n-SMase2 activity and produc-

AD, lysosomal a-SMase translocates to the PM and becomes a tion of exosomes was found in APP/PS1 transgenic mice [145]. An

primary enzyme responsible for the Cer over-production. Cers inhibition of n-SMase-2 led to lowering the amount of exosomes in

generated from the SM hydrolysis can participate in formation of the brain and serum as well as reduction of Ab1–42 concentration

ceramide-enriched platforms, which are required for clustering of in the plaques and levels of almost all Cer species analyzed in

some specific receptors, to trigger their molecular pathway serum. Authors suggested that n-SMase2 might influence the

[134]. Interesting data was obtained from AD plaque analysis. The Ab1–42 aggregation through the exosomal signaling. In addition,

senile plaques isolated post mortem from the brains of AD patients in vitro experiments revealed that exosomes stimulated the

contained an increased concentration of predominantly Cer-C20:0 changes in Ab conformation toward reduction of toxicity of

in comparison with surrounding neuropil. On the other hand, the amyloid aggregates and enabled their engulfing by the microglia

level of SM-C20:0 was only slightly reduced. Authors suggested that cells. Moreover, exosome secretion from neurons was regulated by

elevated Cers could partially originated from the SM hydrolysis since n-SMase2 and SMS2 [146]. A new mechanism of a-SMase action in

the expressions of both a-SMase and n-SMase were found in corona, the AD was proposed by Lee et al. [147]. They noted an increment

and in some cases core, of senile plaques [135]. Changes in SM in a-SMAse activities in brains, plasma and fibroblasts obtained

concentrations in the brain were also studied in AD animal models. from patients with familiar AD or APP/PS1 (amyloid precursor

Gonza´lez-Domı´nguez et al. noted that the levels of SMs containing protein/presenilin 1) transgenic mice. The enhanced activation of

saturated acyl groups were augmented in the hippocampus, the enzyme disturbed autophagy-lysosome pathway and dimin-

whereas those with unsaturated fatty acid chains were reduced ished autophagic degradation. Suppression or even restoration of

in the cerebellum and cortex of APP/PS1 (amyloid precursor protein/ a-SMase activity in mice to the normal range reversed this

presenilin 1) mice [136]. Interestingly, decrement of SM species with processes. Furthermore, reduction of a-SMase expression lowered

very long chain fatty acid chains was detected in all studied brain also Ab content as well as improved the memory dysfunctions in

areas: hippocampus, cortex, cerebellum, striatum and olfactory mice [147]. Bandaru et al. presented an interesting observation

bulbs [136]. The lipid analysis in the brains and blood plasma of mice concerning ApoE4 gene variant, which highly predisposes its

with expression of mutated human amyloid precursor protein (APP) carriers to AD development [148]. Authors have found that ApoE4

and tau protein (Tg2576 Â JNPL3) (APP/tau mice) revealed the is not sufficient by itself to induce the SM and Cer alterations in the

changes in several SM species concentrations at different stages of brain since healthy people with ApoE4 genotype did not present

symptom development, namely pre-symptomatic and early or late any changes in the levels of these lipids. In opposite, ApoE4 AD

576 B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581

patients demonstrated multiple variations in SF concentrations in PD is caused by extensive damage to the dopaminergic neurons in

several brain regions [148]. the substantia nigra. The principal pathomorphological feature of

PD lesions is the formation of Lewy bodies and neurites, which are

Depression and schizophrenia intracellular deposits of protein aggregates containing predomi-

nantly a-synuclein (a-syn) [164]. The etiology of PD is still

Major depression is one of the most widespread mood unknown. Xilouri et al. [165] hypothesized that the disease may

disorders. Although it affects mental functions of the patients, commence with lysosomal malfunctions in a-syn degradation.

several systemic alterations have been found in the course of the This process may involve the lysosomal enzyme, a-SMAse

disease [149–151]. Interestingly, it was shown that depression [165]. Indeed, mutations in its coding sequence were found to

influences a SM metabolism, which is reflected in the changes of correlate with PD incidence [166,167]. Moreover, Gan-Or et al.

some SM-related blood parameters. A Dutch cohort study revealed suggested that mutations in the SMPD1 gene led to abnormalities

that depressed patients presented an altered pattern of plasma in the lysosome functions, which resulted in disturbed aS

SMs, wherein the SM-23:1/SM-16:0 ratio negatively correlated clearance [168]. The dysregulated SM metabolism can also

with the symptoms of the disease [152]. In addition, Kornhuber influence the process of neurotransmission in PD. It is well

et al. found over-activity of a-SMase in peripheral blood known that a-syn, by aggregation in PM of neuronal cells, causes a

mononuclear cells of patients suffering from major depression deterioration of the synaptic functions [169]. Studies by Leftin

[153]. et al. [170] on model membranes indicate that a-syn aggregate is

The pathophysiology of depression involves several systems of likely in the SM-enriched domains, which perturbs PM organiza-

neurotransmission, however, the majority of the clinical treat- tion. Disorders in the structure of PM may result in alterations of

ments are designed to regulate the activities of serotoninergic membrane fusion [170], which may have important consequences

synapses [154]. Serotonin generates the excitatory/inhibitory for the exocytosis of neurotransmitters.

signals at the synapse through binding to its receptors, 5-HT (1– In order to establish whether SM levels underwent changes

7) [155]. Sjogren et al. have shown that the inhibition of SM with PD progression, several studies on post mortem brain tissue

biosynthesis in HeLa cell line stably transfected with 5-HT(7) of PD patients have been conducted. Sidransky et al. [171] found

receptor, reduced the serotonin binding to the receptor [156]. In- that in subjects displaying mutations in the gene encoding

terestingly, a decrease in SM level changed neither the affinity of lysosomal enzyme, glucocerebrosidase, which is an important

serotonin to 5-HT(7) nor the concentration of the latter. This risk factor for PD development [171] as well as in patients with

suggests that the strength of serotonin-5-HT(7) binding may be sporadic PD no alterations in SM content were observed

regulated by the SM content in the lipid rafts of the PM [156]. On [172]. On the other hand, Fabelo et al. noted that SM levels

the other hand, serotonin can influence SM levels, which was noted were slightly decreased in patients with early and incidental PD

in mice with chemically reduced neurotransmitter [157]. The well- [173]. However, novel findings have revealed that PD-related

established medication of mood disorders is based, in a large changes in SM levels may differ depending on the brain region.

extent, on targeting the serotoninergic system. Thus, in the anterior cingulate cortex, the brain area affected by

Surprisingly, many of these drugs, particularly tricyclic antide- PD, SM content was lower in comparison with healthy controls

pressants (TCAs), turned out to exert additional activities of while in the occipital cortex, which did not display signs of

indirect a-SMase inhibitors, so-called ‘‘FIASMA’’ (Functional neurodegeneration, no alterations in SM levels were found

Inhibitor of Acid SphingoMyelinAse) [158]. A role of a-SMase [174]. In addition, the authors noted that in the cingulate cortex

regulation by the TCAs in major depression was shown in mouse of PD brains the several SM molecular species, such as SM

model by Gulbins et al. [159]. Thus, the and containing C23:0, C24:1, and C26:1 fatty acids, were reduced

fluoxetine treatments of stressed mice led to a reduction in a- versus controls, whereas the concentrations of C18:1- and C20:0-

SMase activity in the hippocampus and improved depressive-like containing SMs were augmented.

behavior of the animals, which was not observed in mice with a- Another interesting hypothesis concerning the involvement of

SMase deficiency. Interestingly, mice with a-SMase overexpression SM metabolism in PD pathogenesis connects the SM cycle and

showed stressed behavior, even without its prior induction by classical inflammatory pathways [175]. Thus, NF-kB translocation

stressors. The treatment with TCAs ameliorated a performance of to the nuclei of dopaminergic neurons in PD [176] may trigger the

the animals [159]. formation of free radicals, which in turn results in promoting SM

The disturbed SM metabolism has also been reported in other hydrolysis leading to Cer generation and apoptosis.

serious psychiatric condition, schizophrenia. In 1993, Keshavan

et al. showed an increased level of SM in the erythrocyte PM of Niemann–Pick disease (NPD)

psychotic patients [160]. Several years later, Ponizovsky et al.

found a correlation between erythrocyte SM content and negative NPD was one of the first discovered syndromes caused by the

schizophrenic symptoms [161]. On the other hand, SM levels in the altered metabolism of SM. This genetic lipid storage disorder [177]

thalamus of schizophrenic patients were demonstrated to be shed new light on a-SMase function in SM hydrolysis. There are

significantly decreased [162]. Interestingly, the altered neuronal three main types of NPD. NPD type A and B, differing in clinical

content of SM may regulate the activity of the a7 nicotinic outcomes [178], are both a result of a-SMase deficiency due to

acetylcholine receptor, a potential target for the treatment of autosomal mutation of the SMPD1 gene. A large number of

schizophrenia. Thus, Colo´ n-Sa´ez et al. observed that the enzymatic mutations found in this gene [179–184] determine the differences

hydrolysis of SM by SMase in rat primary hippocampal neurons in a-SMase levels (from an almost complete absence for type A to

changed the characteristics of the a7 nicotinic acetylcholine approximately 10% of the normal level for type B), which results in

receptor desensitization, presumably influencing its anchoring in diverse phenotypes of NPD. a-SMase deficiency leads to disturbed

the lipid rafts [163]. SM degradation and accumulation of this lipid in the cells. NPD type

C (C1 or C2) is caused by mutation in the NPC1 and 2 genes, the

Parkinson disease (PD) coding proteins taking part in cholesterol trafficking [185,186],

which has consequences in cholesterol accumulation in virtually all

PD is one of the most common neurodegenerative disorders, body organs. An increase in SM levels as well as the reduced activity

which affects predominantly the motor functions of the patients. of a-SMase are also observed in NPD type C, however, as a multi-lipid

B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581 577

References

secondary effect of cholesterol depositions [187,188]. NPD type A

produces the strongest CNS dysfunctions in comparison with other

[1] Filippov A, Ora¨dd G, Lindblom G. Sphingomyelin structure influences the

types of the disease. It is characterized by early neurodegeneration,

lateral diffusion and raft formation in lipid bilayers. Biophys J 2006;90(6):

hepatosplenomegaly, psychomotor retardation and eventually 2086–92.

[2] Stillwell W. An introduction to biological membranes from bilayers to rafts.

death, typically at the age of three [189]. Patients with NPD type B

1st ed. Elsevier; 2013 [chapter 5].

suffer from , pulmonary disorders, liver dysfunction,

[3] Karlsson AA, Miche´lsen P, Odham G. Molecular species of sphingomyelin:

growth restriction, and a reduced life span, but usually display only determination by high-performance liquid chromatography/mass spectrom-

etry with electrospray and high-performance liquid chromatography/tandem

mild or even no CNS-related symptoms [190,191]. Marathe et al.

mass spectrometry with atmospheric pressure chemical ionization. J Mass

[192] investigated the effect of the a-SMase level on the CNS

Spectrom 1998;33(12):1192–8.

functions in the mouse model of NPD type B. The animals were [4] Furland NE, Zanetti SR, Oresti GM, Maldonado EN, Aveldan˜o MI. Ceramides

designed to exhibit a moderate activity of l-SMase (up to 20% of and with high proportions of very long-chain polyunsatu-

rated fatty acids in mammalian germ cells. J Biol Chem 2007;282(25):

that from wild-type brains) and a complete absence of s-SMase.

18141–50.

The researchers did not find any severe neurological symptoms

[5] Ramstedt B, Slotte JP. Membrane properties of sphingomyelins. FEBS Lett

in the mice, up to 8 months old which indicated that a stable 2002;531(1):33–7.

[6] Kuikka M, Ramstedt B, Ohvo-Rekila¨ H, Tuuf J, Slotte JP. Membrane properties

minimal activity of l-SMase is sufficient to maintain the CNS

of D-erythro-N-acyl sphingomyelins and their corresponding dihydro spe-

function, even in the absence of s-SMase [192]. However,

cies. Biophys J 2001;80(5):2327–37.

extremely low levels of a-SMase cause abnormal SM deposition [7] Kan CC, Ruan ZS, Bittman R. Interaction of cholesterol with sphingomyelin in

bilayer membranes: evidence that the hydroxy group of sphingomyelin does

in the brain and evoke neuronal injuries. In NPD type A, SM

not modulate the rate of cholesterol exchange between vesicles. Biochemis-

accumulation affects neurons by altering lysosomal membrane

try 1991;30(31):7759–66.

permeabilisation, which impairs lysosome function in autopha- [8] Needham D, Nunn RS. Elastic deformation and failure of lipid bilayer mem-

branes containing cholesterol. Biophys J 1990;58(4):997–1009.

golysosomal degradation [193]. Neurodegeneration caused by

[9] Schroeder RJ, Ahmed SN, Zhu Y, London E, Brown DA. Cholesterol and

SM accumulation begins in axons and affects the majority of the

sphingolipid enhance the Triton X-100 insolubility of glycosylphosphatidy-

cerebellar Purkinje cells [194]. In an attempt to find a treatment linositol-anchored proteins by promoting the formation of detergent-insolu-

ble ordered membrane domains. J Biol Chem 1998;273(2):1150–7.

method for NPD, a recombinant a-SMase (RHa-SMase) has been

[10] Prinetti A, Chigorno V, Prioni S, Loberto N, Marano N, Tettamanti G, et al.

tested in animal models mimicking NBD. The intravenous

Changes in the lipid turnover, composition, and organization, as sphingoli-

administration of RHa-SMase into the NBD mouse lowered pid-enriched membrane domains, in rat cerebellar granule cells developing

peripheral levels of SM without any effect on its levels in CNS in vitro. J Biol Chem 2001;276(24):21136–45.

[11] Koval M, Pagano RE. Intracellular transport and metabolism of sphingomye-

[195]. Furthermore, RHa-SMase injected directly to the brains of

lin. Biochim Biophys Acta 1991;1082(2):113–25.

a-SMase knockout mice improved motor dysfunctions and

[12] Ramstedt B, Slotte JP. Interaction of cholesterol with sphingomyelins and

alleviated central NPD symptoms [196]. Recently, the dose- acyl-chain-matched phosphatidylcholines: a comparative study of the effect

of the chain length. Biophys J 1999;76(2):908–15.

dependent toxicity of RHa-SMase has been tested in several

[13] Bittman R, Kasireddy CR, Mattjus P, Slotte JP. Interaction of cholesterol with

animal models of NPD regarding the safety aspect of its use as a

sphingomyelin in monolayers and vesicles. Biochemistry 1994;33(39):

potential drug [197]. 11776–81.

[14] Jaikishan S, Bjo¨rkbom A, Slotte JP. Sphingomyelin analogs with branched

N-acyl chains: the position of branching dramatically affects acyl chain order

Summary and sterol interactions in bilayer membranes. Biochim Biophys Acta

2010;1798(10):1987–94.

[15] Ramstedt B, Slotte JP. Comparison of the biophysical properties of racemic

SM is the most abundant eukaryotic SF component of the

and d-erythro-N-acyl sphingomyelins. Biophys J 1999;77(3):1498–506.

plasma membrane. SM with different fatty acid chains has the [16] Bini L, Pacini S, Liberatori S, Valensin S, Pellegrini M, Raggiaschi R, et al.

Extensive temporally regulated reorganization of the lipid raft proteome

ability to create unique compositions of lipid rafts thereby it can

following T-cell antigen receptor triggering. Biochem J 2003;369(2):301–9.

strongly modify the property of the PM. The SM content in a cell is

[17] Brown DA, London E. Structure and function of sphingolipid- and cholesterol-

strictly regulated by the enzymes of SM metabolic pathways which rich membrane rafts. J Biol Chem 2000;275(23):17221–24.

[18] Merrill Jr AH, van Echten G, Wang E, Sandhoff K. Fumonisin B1 inhibits sphin-

create a balance between SM synthesis (SMSs) and degradation

gosine (sphinganine) N-acyltransferase and de novo sphingolipid biosynthesis

(SMases). Physiological roles of SMSs and SMases is not sufficiently

in cultured neurons in situ. J Biol Chem 1993;268(36):27299–306.

recognized. The activities of these enzyme undergo changes in [19] Schweizer A, Clausen H, van Meer G, Hauri HP. Localization of O-glycan

initiation, sphingomyelin synthesis, and glucosylceramide synthesis in Vero

many pathologies, also those affecting CNS, such as ischemia, AD,

cells with respect to the endoplasmic reticulum-Golgi intermediate com-

PD or depression and these alterations involve mainly SMases

partment. J Biol Chem 1994;269(6):4035–41.

degrading SM, which over-produce Cers. It is suggested that over- [20] Funato K, Riezman H. Vesicular and nonvesicular transport of ceramide from

activated enzymes could be targeted in the future therapies of CNS, ER to the Golgi apparatus in yeast. J Cell Biol 2001;155(6):949–59.

[21] Hanada K. Intracellular trafficking of ceramide by ceramide transfer protein.

which could have a protective effect toward affected brain.

Proc Jpn Acad Ser B Phys Biol Sci 2010;86(4):426–37.

[22] Wang X, Rao RP, Kosakowska-Cholody T, Masood MA, Southon E, Zhang H,

et al. Mitochondrial degeneration and not apoptosis is the primary cause of

Conflict of interest

embryonic lethality in ceramide transfer protein mutant mice. J Cell Biol

2009;184(1):143–58.

We wish to confirm that there are no known conflicts of interest [23] Futerman AH, Stieger B, Hubbard AL, Pagano RE. Sphingomyelin synthesis in

rat liver occurs predominantly at the cis and medial cisternae of the Golgi

associated with this publication and there has been no significant

apparatus. J Biol Chem 1990;265(15):8650–7.

financial support for this work that could have influenced its

[24] van Meer G, Holthuis JC. Sphingolipid transport in eukaryotic cells. Biochim

outcome. We confirm that the manuscript has been read and Biophys Acta 2000;1486(1):145–70.

[25] Schuchman EH, Suchi M, Takahashi T, Sandhoff K, Desnick RJ. Human acid

approved by all named authors and that there are no other persons

sphingomyelinase. Isolation, nucleotide sequence and expression of the full-

who satisfied the criteria for authorship but are not listed. We

length and alternatively spliced cDNAs. J Biol Chem 1991;266(13):8531–9.

further confirm that the order of authors listed in the manuscript [26] Wu BX, Clarke CJ, Hannun YA. Mammalian neutral sphingomyelinases: regu-

lation and roles in cell signaling responses. Neuromolecular Med 2010;12(4):

has been approved by all of us.

320–30.

[27] Milhas D, Clarke CJ, Hannun YA. Sphingomyelin metabolism at the plasma

Funding membrane: implications for bioactive sphingolipids. FEBS Lett 2010;584(9):

1887–94.

[28] Ding T, Kabir I, Li Y, Lou C, Yazdanyar A, Xu J, et al. All members in the

The authors declare that funding source(s) had no involvement sphingomyelin synthase gene family have ceramide phosphoethanolamine

synthase activity. J Lipid Res 2015;56(3):537–45.

in preparing this manuscript.

578 B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581

[29] Yeang C, Ding T, Chirico WJ, Jiang XC. Subcellular targeting domains of sphingomyelinase by site-directed mutagenesis. Eur J Biochem 1997;243(1–2):

sphingomyelin synthase 1 and 2. Nutr Metab (Lond) 2011;8:89. 511–7.

[30] Yeang C, Varshney S, Wang R, Zhang Y, Ye D, Jiang XC. The domain responsible [56] Schissel SL, Jiang X, Tweedie-Hardman J, Jeong T, Camejo EH, Najib J, et al.

for sphingomyelin synthase (SMS) activity. Biochim Biophys Acta Secretory sphingomyelinase, a product of the acid sphingomyelinase gene,

2008;1781(10):610–7. can hydrolyze atherogenic lipoproteins at neutral pH. Implications for ath-

[31] Li Z, Hailemariam TK, Zhou H, Li Y, Duckworth DC, Peake DA, et al. Inhibition erosclerotic lesion development. J Biol Chem 1998;273(5):2738–46.

2+

of sphingomyelin synthase (SMS) affects intracellular sphingomyelin accu- [57] Spence MW, Byers DM, Palmer FB, Cook HW. A new Zn -stimulated sphin-

mulation and plasma membrane lipid organization. Biochim Biophys Acta gomyelinase in fetal bovine serum. J Biol Chem 1989;264(10):5358–63.

2007;1771(9):1186–94. [58] Levade T, Salvayre R, Douste-Blazy L. Sphingomyelinases, and Niemann–Pick

[32] Tafesse FG, Huitema K, Hermansson M, van der Poel S, van den Dikkenberg J, disease. J Clin Chem Clin Biochem 1986;24(4):205–20.

Uphoff A, et al. Both sphingomyelin synthases SMS1 and SMS2 are required [59] Takahashi I, Takahashi T, Abe T, Watanabe W, Takada G. Distribution of acid

for sphingomyelin homeostasis and growth in human HeLa cells. J Biol Chem sphingomyelinase in human various body fluids. Tohoku J Exp Med

2007;282(24):17537–47. 2000;192(1):61–6.

[33] Sudarkina OY, Filippenkov IB, Brodsky IB, Limborska SA, Dergunova LV. [60] Mu¨ hle C, Huttner HB, Walter S, Reichel M, Canneva F, Lewczuk P, et al.

Comparative analysis of sphingomyelin synthase 1 gene expression at the Characterization of acid sphingomyelinase activity in human cerebrospinal

transcriptional and translational levels in human tissues. Mol Cell Biochem fluid. PLOS ONE 2013;8(5):e62912.

2015;406(1–2):91–9. [61] Simon Jr CG, Chatterjee S, Gear AR. Sphingomyelinase activity in human

[34] Huitema K, van den Dikkenberg J, Brouwers JF, Holthuis JC. Identification of a platelets. Thromb Res 1998;90(4):155–61.

family of animal sphingomyelin synthases. EMBO J 2004;23(1):33–44. [62] Romiti E, Vasta V, Meacci E, Farnararo M, Linke T, Ferlinz K, et al. Characteri-

[35] Tafesse FG, Ternes P, Holthuis JC. The multigenic sphingomyelin synthase zation of sphingomyelinase activity released by thrombin-stimulated plate-

family. J Biol Chem 2006;281(40):29421–25. lets. Mol Cell Biochem 2000;205(1–2):75–81.

[36] Wesley UV, Hatcher JF, Dempsey RJ. Sphingomyelin synthase 1 regulates [63] Vasta V, Meacci E, Romiti E, Farnararo M, Bruni P. Sphingomyelinase increases

neuro-2a cell proliferation and cell cycle progression through modulation of 2-deoxyglucose uptake and glucose metabolism of human platelets. Biochem

p27 expression and Akt signaling. Mol Neurobiol 2015;51(3):1530–41. Mol Biol Int 1997;43(1):217–26.

[37] Taniguchi M, Okazaki T. The role of sphingomyelin and sphingomyelin [64] Mu¨ nzer P, Borst O, Walker B, Schmid E, Feijge MA, Cosemans JM, et al. Acid

synthases in cell death, proliferation and migration-from cell and animal sphingomyelinase regulates platelet cell membrane scrambling, secretion,

models to human disorders. Biochim Biophys Acta 2014;1841(5):692–703. and thrombus formation. Arterioscler Thromb Vasc Biol 2014;34(1):61–71.

[38] Lafont E, Milhas D, Carpentier S, Garcia V, Jin ZX, Umehara H, et al. Caspase- [65] Kitatani K, Oka T, Murata T, Hayama M, Akiba S, Sato T. Acceleration by

mediated inhibition of sphingomyelin synthesis is involved in FasL-triggered ceramide of calcium-dependent translocation of phospholipase A2 from

cell death. Cell Death Differ 2010;17(4):642–54. cytosol to membranes in platelets. Arch Biochem Biophys 2000;382(2):

[39] Riboni L, Viani P, Bassi R, Giussani P, Tettamanti G. Basic fibroblast growth 296–302.

factor-induced proliferation of primary astrocytes. Evidence for the involve- [66] Hofmann K, Tomiuk S, Wolff G, Stoffel W. Cloning and characterization of the

2+

ment of sphingomyelin biosynthesis. J Biol Chem 2001;276(16):12797–804. mammalian brain-specific, Mg -dependent neutral sphingomyelinase. Proc

[40] Luberto C, Hannun YA. Sphingomyelin synthase, a potential regulator of Natl Acad Sci U S A 2000;97(11):5895–900.

intracellular levels of ceramide and diacylglycerol during SV40 transforma- [67] Tomiuk S, Hofmann K, Nix M, Zumbansen M, Stoffel W. Cloned mammalian

tion. Does sphingomyelin synthase account for the putative phosphatidyl- neutral sphingomyelinase: functions in sphingolipid signaling? Proc Natl

choline-specific phospholipase C? J Biol Chem 1998;273(23):14550–59. Acad Sci U S A 1998;95(7):3638–43.

[41] Cai Z, Bettaieb A, Mahdani NE, Legre`s LG, Stancou R, Masliah J, et al. Alteration [68] Tani M, Hannun YA. Neutral sphingomyelinase 2 is palmitoylated on multiple

of the sphingomyelin/ceramide pathway is associated with resistance of cysteine residues. Role of palmitoylation in subcellular localization. J Biol

human breast carcinoma MCF7 cells to tumor necrosis factor-alpha-mediat- Chem 2007;282(13):10047–56.

ed cytotoxicity. J Biol Chem 1997;272(11):6918–26. [69] Crivello NA, Rosenberg IH, Dallal GE, Bielinski D, Joseph JA. Age-related

[42] Taguchi Y, Kondo T, Watanabe M, Miyaji M, Umehara H, Kozutsumi Y, et al. changes in neutral sphingomyelin-specific phospholipase C activity in stria-

Interleukin-2-induced survival of natural killer (NK) cells involving phos- tum, hippocampus, and frontal cortex: implication for sensitivity to stress

phatidylinositol-3 kinase-dependent reduction of ceramide through acid and inflammation. Neurochem Int 2005;47(8):573–9.

sphingomyelinase, sphingomyelin synthase, and glucosylceramide synthase. [70] Stoffel W, Jenke B, Blo¨ck B, Zumbansen M, Koebke J. Neutral sphingomye-

Blood 2004;104(10):3285–93. linase 2 (smpd3) in the control of postnatal growth and development. Proc

[43] Dong L, Watanabe K, Itoh M, Huan CR, Tong XP, Nakamura T, et al. CD4+ T-cell Natl Acad Sci U S A 2005;102(12):4554–9.

dysfunctions through the impaired lipid rafts ameliorate concanavalin A- [71] Wheeler D, Knapp E, Bandaru VV, Wang Y, Knorr D, Poirier C, et al. Tumor

induced hepatitis in sphingomyelin synthase 1-knockout mice. Int Immunol necrosis factor-alpha-induced neutral sphingomyelinase-2 modulates syn-

2012;24(5):327–37. aptic plasticity by controlling the membrane insertion of NMDA receptors. J

[44] Gon˜i FM, Alonso A. Structure and functional properties of diacylglycerols in Neurochem 2009;109(5):1237–49.

membranes. Prog Lipid Res 1999;38(1):1–48. [72] Kim SK, Ahn KH, Ji JE, Choi JM, Jeon HJ, Jung SY, et al. Neutral sphingomye-

[45] Evangelisti C, Bortul R, Fala` F, Tabellini G, Goto K, Martelli AM. Nuclear linase 2 induces dopamine uptake through regulation of intracellular calci-

diacylglycerol kinases: emerging downstream regulators in cell signaling um. Cell Signal 2010;22(5):865–70.

networks. Histol Histopathol 2007;22(5):573–9. [73] Zheng T, Li W, Wang J, Altura BT, Altura BM. Sphingomyelinase ceramide

[46] Momchilova A, Markovska T. Phosphatidylethanolamine and phosphatidyl- analogs induce contraction and rises in [Ca(2+)](i) in canine cerebral vascular

choline are sources of diacylglycerol in ras-transformed NIH 3T3 fibroblasts. muscle. Am J Physiol Heart Circ Physiol 2000;278(5):H1421–28.

Int J Biochem Cell Biol 1999;31(2):311–8. [74] Bautista-Pe´rez R, del Valle-Mondrago´ n L, Cano-Martı´nez A, Pe´rez-Me´ndez O,

[47] Tani M, Kuge O. Sphingomyelin synthase 2 is palmitoylated at the COOH- Escalante B, Franco M. Involvement of neutral sphingomyelinase in the

terminal tail, which is involved in its localization in plasma membranes. angiotensin II signaling pathway. Am J Physiol Renal Physiol 2015;308(10):

Biochem Biophys Res Commun 2009;381(3):328–32. F1178–87.

[48] Hailemariam TK, Huan C, Liu J, Li Z, Roman C, Kalbfeisch M, et al. Sphingo- [75] Mogami K, Kishi H, Kobayashi S. Sphingomyelinase causes endothelium-

myelin synthase 2 deficiency attenuates NFkappaB activation. Arterioscler dependent vasorelaxation through endothelial nitric oxide production with-

Thromb Vasc Biol 2008;28(8):1519–26. out cytosolic Ca(2+) elevation. FEBS Lett 2005;579(2):393–7.

[49] Ternes P, Brouwers JF, van den Dikkenberg J, Holthuis JC. Sphingomyelin [76] Mueller N, Avota E, Collenburg L, Grassme´ H, Schneider-Schaulies S. Neutral

synthase SMS2 displays dual activity as ceramide phosphoethanolamine sphingomyelinase in physiological and measles virus induced T cell suppres-

synthase. J Lipid Res 2009;50(11):2270–7. sion. PLoS Pathog 2014;10(12):e1004574.

[50] Kidani Y, Ohshima K, Sakai H, Kohno T, Baba A, Hattori M. Differential [77] Khavandgar Z, Murshed M. Sphingolipid metabolism and its role in the

localization of sphingomyelin synthase isoforms in neurons regulates sphin- skeletal tissues. Cell Mol Life Sci 2015;72(5):959–69.

gomyelin cluster formation. Biochem Biophys Res Commun 2012;417(3): [78] Aubin I, Adams CP, Opsahl S, Septier D, Bishop CE, Auge N, et al. A deletion in

1014–7. the gene encoding sphingomyelin phosphodiesterase 3 (Smpd3) results in

[51] Zhang Y, Dong J, Zhu X, Wang W, Yang Q. The effect of sphingomyelin osteogenesis and dentinogenesis imperfecta in the mouse. Nat Genet

synthase 2 (SMS2) deficiency on the expression of drug transporters in 2005;37(8):803–5.

mouse brain. Biochem Pharmacol 2011;82(3):287–94. [79] Stoffel W, Jenke B, Holz B, Binczek E, Gu¨ nter RH, Knifka J, et al. Neutral

[52] Vacaru AM, Tafesse FG, Ternes P, Kondylis V, Hermansson M, Brouwers JF, sphingomyelinase (SMPD3) deficiency causes a novel form of chondrodys-

et al. Sphingomyelin synthase-related protein SMSr controls ceramide ho- plasia and dwarfism that is rescued by Col2A1-driven smpd3 transgene

meostasis in the ER. J Cell Biol 2009;185(6):1013–27. expression. Am J Pathol 2007;171(1):153–61.

[53] Marathe S, Schissel SL, Yellin MJ, Beatini N, Mintzer R, Williams KJ, et al. [80] Tomiuk S, Zumbansen M, Stoffel W. Characterization and subcellular locali-

Human vascular endothelial cells are a rich and regulatable source of zation of murine and human magnesium-dependent neutral sphingomyeli-

secretory sphingomyelinase. Implications for early atherogenesis and cer- nase. J Biol Chem 2000;275(8):5710–7.

amide-mediated cell signaling. J Biol Chem 1998;273(7):4081–8. [81] Mizutani Y, Tamiya-Koizumi K, Nakamura N, Kobayashi M, Hirabayashi Y,

2+

[54] Schissel SL, Schuchman EH, Williams KJ, Tabas I. Zn -stimulated sphingo- Yoshida S. Nuclear localization of neutral sphingomyelinase 1: biochemical

myelinase is secreted by many cell types and is a product of the acid and immunocytochemical analyses. J Cell Sci 2001;114(Pt 20):3727–36.

sphingomyelinase gene. J Biol Chem 1996;271(31):18431–36. [82] Sawai H, Domae N, Nagan N, Hannun YA. Function of the cloned putative

[55] Ferlinz K, Hurwitz R, Moczall H, Lansmann S, Schuchman EH, Sandhoff K. neutral sphingomyelinase as lyso-platelet activating factor-phospholipase C.

Functional characterization of the N-glycosylation sites of human acid J Biol Chem 1999;274(53):38131–39.

B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581 579

[83] Zumbansen M, Stoffel W. Neutral sphingomyelinase 1 deficiency in the [109] Yamashima T, Mathivanan A, Dazortsava MY, Sakai S, Kurimoto S, Zhu H, et al.

mouse causes no lipid storage disease. Mol Cell Biol 2002;22(11):3633–8. Calpain-mediated Hsp70.1 cleavage in monkey CA1 after ischemia induces

[84] Tonnetti L, Verı´ MC, Bonvini E, D’Adamio L. A role for neutral sphingomye- similar ‘lysosomal vesiculosis’ to Alzheimer neurons. J Alzheimers Dis Par-

linase-mediated ceramide production in T cell receptor-induced apoptosis kinsonism 2014;4:139.

and mitogen-activated protein kinase-mediated signal transduction. J Exp [110] Yu ZF, Nikolova-Karakashian M, Zhou D, Cheng G, Schuchman EH, Mattson

Med 1999;189(10):1581–9. MP. Pivotal role for acidic sphingomyelinase in cerebral ischemia-induced

2+

[85] Yabu T, Imamura S, Yamashita M, Okazaki T. Identification of Mg -depen- ceramide and cytokine production, and neuronal apoptosis. J Mol Neurosci

dent neutral sphingomyelinase 1 as a mediator of heat stress-induced 2000;15(2):85–97.

ceramide generation and apoptosis. J Biol Chem 2008;283(44):29971–82. [111] Yu J, Novgorodov SA, Chudakova D, Zhu H, Bielawska A, Bielawski J, et al. JNK3

[86] Yabu T, Shiba H, Shibasaki Y, Nakanishi T, Imamura S, Touhata K, et al. Stress- signaling pathway activates ceramide synthase leading to mitochondrial

induced ceramide generation and apoptosis via the phosphorylation and dysfunction. J Biol Chem 2007;282(35):25940–49.

activation of nSMase1 by JNK signaling. Cell Death Differ 2015;22(2):258–73. [112] Jin J, Hou Q, Mullen TD, Zeidan YH, Bielawski J, Kraveka JM, et al. Ceramide

[87] Guo BB, Bellingham SA, Hill AF. The neutral sphingomyelinase pathway generated by sphingomyelin hydrolysis and the salvage pathway is involved

regulates packaging of the prion protein into exosomes. J Biol Chem in hypoxia/reoxygenation-induced Bax redistribution to mitochondria in NT-

2015;290(6):3455–67. 2 cells. J Biol Chem 2008;283(39):26509–17.

[88] Krut O, Wiegmann K, Kashkar H, Yazdanpanah B, Kro¨nke M. Novel tumor [113] Lam SM, Wang Y, Duan X, Wenk MR, Kalaria RN, Chen CP, et al. Brain

necrosis factor-responsive mammalian neutral sphingomyelinase-3 is a C- lipidomes of subcortical ischemic vascular dementia and mixed dementia.

tail-anchored protein. J Biol Chem 2006;281(19):13784–93. Neurobiol Aging 2014;35(10):2369–81.

[89] Corcoran CA, He Q, Ponnusamy S, Ogretmen B, Huang Y, Sheikh MS. Neutral [114] Chan RB, Oliveira TG, Cortes EP, Honig LS, Duff KE, Small SA, et al. Compara-

sphingomyelinase-3 is a DNA damage and nongenotoxic stress-regulated tive lipidomic analysis of mouse and human brain with Alzheimer disease. J

gene that is deregulated in human malignancies. Mol Cancer Res 2008;6(5): Biol Chem 2012;287(4):2678–88.

795–807. [115] Ben-David O, Pewzner-Jung Y, Brenner O, Laviad EL, Kogot-Levin A, Weiss-

[90] Moylan JS, Smith JD, Wolf Horrell EM, McLean JB, Deevska GM, Bonnell MR, berg I, et al. Encephalopathy caused by ablation of very long acyl chain

et al. Neutral sphingomyelinase-3 mediates TNF-stimulated oxidant activity ceramide synthesis may be largely due to reduced levels.

in skeletal muscle. Redox Biol 2014;2:910–20. J Biol Chem 2011;286(34):30022–33.

[91] Wu BX, Rajagopalan V, Roddy PL, Clarke CJ, Hannun YA. Identification and [116] Adam-Klages S, Adam D, Wiegmann K, Struve S, Kolanus W, Schneider-

characterization of murine mitochondria-associated neutral sphingomyeli- Mergener J, et al. FAN, a novel WD-repeat protein, couples the p55 TNF-

nase (MA-nSMase), the mammalian sphingomyelin phosphodiesterase 5. J receptor to neutral sphingomyelinase. Cell 1996;86(6):937–47.

Biol Chem 2010;285(23):17993–8002. [117] Philipp S, Puchert M, Adam-Klages S, Tchikov V, Winoto-Morbach S, Mathieu

[92] Rajagopalan V, Canals D, Luberto C, Snider J, Voelkel-Johnson C, Obeid LM, S, et al. The Polycomb group protein EED couples TNF receptor 1 to neutral

et al. Critical determinants of mitochondria-associated neutral sphingomye- sphingomyelinase. Proc Natl Acad Sci U S A 2010;107(3):1112–7.

linase (MA-nSMase) for mitochondrial localization. Biochim Biophys Acta [118] Lavrentiadou SN, Chan C, Kawcak T, Ravid T, Tsaba A, van der Vliet A, et al.

2015;1850(4):628–39. Ceramide-mediated apoptosis in lung epithelial cells is regulated by gluta-

[93] Birbes H, Luberto C, Hsu YT, El Bawab S, Hannun YA, Obeid LM. A mitochon- thione. Am J Respir Cell Mol Biol 2001;25(6):676–84.

drial pool of sphingomyelin is involved in TNFalpha-induced Bax transloca- [119] Yamagata K, Ichinose S, Tagawa C, Tagami M. Vitamin E regulates SMase

tion to mitochondria. Biochem J 2005;386(Pt 3):445–51. activity, GSH levels, and inhibits neuronal death in stroke-prone spontane-

[94] Petcoff DW, Holland WL, Stith BJ. Lipid levels in sperm, eggs, and during ously hypertensive rats during hypoxia and reoxygenation. J Exp Stroke

fertilization in Xenopus laevis. J Lipid Res 2008;49(11):2365–78. Transl Med 2009;2(2):41–8.

[95] Furland NE, Oresti GM, Antollini SS, Venturino A, Maldonado EN, Aveldan˜o [120] Won JS, Singh I. Sphingolipid signaling and redox regulation. Free Radic Biol

MI. Very long-chain polyunsaturated fatty acids are the major acyl groups of Med 2006;40(11):1875–88.

sphingomyelins and ceramides in the head of mammalian spermatozoa. J Biol [121] Li X, Gulbins E, Zhang Y. Oxidative stress triggers Ca-dependent lysosome

Chem 2007;282(25):18151–61. trafficking and activation of acid sphingomyelinase. Cell Physiol Biochem

[96] Duan RD, Bergman T, Xu N, Wu J, Cheng Y, Duan J, et al. Identification of 2012;30(4):815–26.

human intestinal alkaline sphingomyelinase as a novel ecto-enzyme related [122] Wei W, Du C, Lv J, Zhao G, Li Z, Wu Z, et al. Blocking A2B adenosine receptor

to the nucleotide phosphodiesterase family. J Biol Chem 2003;278(40): alleviates pathogenesis of experimental autoimmune encephalomyelitis via

38528–36. inhibition of IL-6 production and Th17 differentiation. J Immunol

[97] Duan RD, Nilsson A. Purification of a newly identified alkaline sphingomye- 2013;190(1):138–46.

linase in human bile and effects of bile salts and phosphatidylcholine on [123] Dmitrieva VG, Torshina EV, Yuzhakov VV, Povarova OV, Skvortsova VI,

enzyme activity. Hepatology 1997;26(4):823–30. Limborska SA, et al. Expression of sphingomyelin synthase 1 gene in rat

[98] Nyberg L, Nilsson A, Lundgren P, Duan RD. Localization and capacity of brain focal ischemia. Brain Res 2008;1188:222–7.

sphingomyelin digestion in the rat intestinal tract. J Nutr Biochem [124] Morishima-Kawashima M, Ihara Y. Alzheimer’s disease: beta-amyloid pro-

1997;8(3):112–8. tein and tau. J Neurosci Res 2002;70:392–401.

[99] Kubota M, Narita K, Nakagomi T, Tamura A, Shimasaki H, Ueta N, et al. [125] Oresˇicˇ M, Hyo¨tyla¨inen T, Herukka SK, Sysi-Aho M, Mattila I, Seppa¨nan-Laakso

Sphingomyelin changes in rat cerebral cortex during focal ischemia. Neurol T, et al. Metabolome in progression to Alzheimer’s disease. Transl Psychiatry

Res 1996;18(4):337–41. 2011;1:e57.

[100] Nakane M, Kubota M, Nakagomi T, Tamura A, Hisaki H, Shimasaki H, et al. [126] Mielke MM, Bandaru VV, Haughey NJ, Rabins PV, Lyketsos CG, Carlson MC.

Lethal forebrain ischemia stimulates sphingomyelin hydrolysis and ceramide Serum sphingomyelins and ceramides are early predictors of memory im-

generation in the gerbil hippocampus. Neurosci Lett 2000;296(2–3):89–92. pairment. Neurobiol Aging 2010;31(1):17–24.

[101] Tian HP, Qiu TZ, Zhao J, Li LX, Guo J. Sphingomyelinase-induced ceramide [127] Mielke MM, Bandaru VV, Haughey NJ, Xia J, Fried LP, Yasar S, et al. Serum

production stimulate calcium-independent JNK and PP2A activation follow- ceramides increase the risk of Alzheimer disease: the Women’s Health and

ing cerebral ischemia. Brain Inj 2009;23(13–14):1073–80. Aging Study II. Neurology 2012;79(7):633–41.

[102] Soeda S, Tsuji Y, Ochiai T, Mishima K, Iwasaki K, Fujiwara M, et al. Inhibition of [128] Kosicek M, Kirsch S, Bene R, Trkanjec Z, Titlic M, Bindila L, et al. Nano-HPLC–

sphingomyelinase activity helps to prevent neuron death caused by ischemic MS analysis of phospholipids in cerebrospinal fluid of Alzheimer’s disease

stress. Neurochem Int 2004;45(5):619–26. patients – a pilot study. Anal Bioanal Chem 2010;398(7–8):2929–37.

[103] Liu B, Hassler DF, Smith GK, Weaver K, Hannun YA. Purification and charac- [129] Kosicek M, Zetterberg H, Andreasen N, Peter-Katalinic J, Hecimovic S. Elevat-

terization of a membrane bound neutral pH optimum magnesium-depen- ed cerebrospinal fluid sphingomyelin levels in prodromal Alzheimer’s dis-

dent and phosphatidylserine-stimulated sphingomyelinase from rat brain. J ease. Neurosci Lett 2012;516(2):302–5.

Biol Chem 1998;273(51):34472–79. [130] Mielke MM, Haughey NJ, Bandaru VV, Zetterberg H, Blennow K, Andreasson

[104] Yoshimura S, Banno Y, Nakashima S, Takenaka K, Sakai H, Nishimura Y, et al. U, et al. Cerebrospinal fluid sphingolipids, b-amyloid, and tau in adults at risk

Ceramide formation leads to caspase-3 activation during hypoxic PC12 cell for Alzheimer’s disease. Neurobiol Aging 2014;35(11):2486–94.

death. Inhibitory effects of Bcl-2 on ceramide formation and caspase-3 [131] Gottfries CG, Jungbjer B, Karlsson I, Svennerholm L. Reductions in membrane

activation. J Biol Chem 1998;273(12):6921–7. proteins and lipids in basal ganglia of classic Alzheimer disease patients.

[105] Yoshimura S, Banno Y, Nakashima S, Hayashi K, Yamakawa H, Sawada M, Alzheimer Dis Assoc Disord 1996;10(2):77–81.

et al. Inhibition of neutral sphingomyelinase activation and ceramide forma- [132] So¨derberg M, Edlund C, Alafuzoff I, Kristensson K, Dallner G. Lipid composi-

tion by glutathione in hypoxic PC12 cell death. J Neurochem 1999;73(2): tion in different regions of the brain in Alzheimer’s disease/senile dementia

675–83. of Alzheimer’s type. J Neurochem 1992;59(5):1646–53.

[106] Bernardo K, Krut O, Wiegmann K, Kreder D, Micheli M, Scha¨fer R, et al. [133] He X, Huang Y, Li B, Gong CX, Schuchman EH. Deregulation of sphingolipid

Purification and characterization of a magnesium-dependent neutral sphin- metabolism in Alzheimer’s disease. Neurobiol Aging 2010;31(3):398–408.

gomyelinase from bovine brain. J Biol Chem 2000;275(11):7641–7. [134] Gulbins E. Regulation of death receptor signaling and apoptosis by ceramide.

[107] Gu L, Huang B, Shen W, Gao L, Ding Z, Wu H, et al. Early activation of nSMase2/ Pharmacol Res 2003;47:393–9.

ceramide pathway in astrocytes is involved in ischemia-associated neuronal [135] Panchal M, Gaudin M, Lazar AN, Salvati E, Rivals I, Ayciriex S, et al. Cer-

damage via inflammation in rat hippocampi. J Neuroinflammation amides and sphingomyelinases in senile plaques. Neurobiol Dis 2014;65:

2013;10:109. 193–201.

[108] Ohtani R, Tomimoto H, Kondo T, Wakita H, Akiguchi I, Shibasaki H, et al. [136] Gonza´lez-Domı´nguez R, Garcı´a-Barrera T, Vitorica J, Go´ mez-Ariza JL. Region-

Upregulation of ceramide and its regulating mechanism in a rat model of specific metabolic alterations in the brain of the APP/PS1 transgenic mice of

chronic cerebral ischemia. Brain Res 2004;1023(1):31–40. Alzheimer’s disease. Biochim Biophys Acta 2014;1842(12 Pt A):2395–402.

580 B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581

[137] Tajima Y, Ishikawa M, Maekawa K, Murayama M, Senoo Y, Nishimaki- [164] Eriksen JL, Wszolek Z, Petrucelli L. Molecular pathogenesis of Parkinson

Mogami T, et al. Lipidomic analysis of brain tissues and plasma in a mouse disease. Arch Neurol 2005;62(3):353–7.

model expressing mutated human amyloid precursor protein/tau for Alzhei- [165] Xilouri M, Vogiatzi T, Vekrellis K, Stefanis L. Alpha-synuclein degradation by

mer’s disease. Lipids Health Dis 2013;12:68. autophagic pathways: a potential key to Parkinson’s disease pathogenesis.

[138] Cordy JM, Hooper NM, Turner AJ. The involvement of lipid rafts in Alzheimer’s Autophagy 2008;4(7):917–9.

disease. Mol Membr Biol 2006;23(1):111–22. [166] Dagan E, Schlesinger I, Ayoub M, Mory A, Nassar M, Kurolap A, et al. The

[139] Schengrund CL. Lipid rafts: keys to neurodegeneration. Brain Res Bull contribution of Niemann–Pick SMPD1 mutations to Parkinson disease in

2010;82(1–2):7–17. Ashkenazi Jews. Parkinsonism Relat Disord 2015;21(9):1067–71.

[140] Fabelo N, Martı´n V, Marı´n R, Moreno D, Ferrer I, Dı´az M. Altered lipid [167] Foo JN, Liany H, Bei JX, Yu XQ, Liu J, Au WL, et al. Rare lysosomal enzyme gene

composition in cortical lipid rafts occurs at early stages of sporadic Alzhei- SMPD1 variant (p.R591C) associates with Parkinson’s disease. Neurobiol

mer’s disease and facilitates APP/BACE1 interactions. Neurobiol Aging Aging 2013;34(12). 2890.e13-5.

2014;35(8):1801–12. [168] Gan-Or Z, Ozelius LJ, Bar-Shira A, Saunders-Pullman R, Mirelman A, Kornreich

[141] Dı´az M, Fabelo N, Martı´n V, Ferrer I, Go´ mez T, Marı´n R. Biophysical alterations R, et al. The p.L302P mutation in the lysosomal enzyme gene SMPD1 is a risk

in lipid rafts from human cerebral cortex associate with increased BACE1/ factor for Parkinson disease. Neurology 2013;80(17):1606–10.

AbPP interaction in early stages of Alzheimer’s disease. J Alzheimers Dis [169] Das AK, Pandit R, Maiti S. Effect of amyloids on the vesicular machinery:

2015;43(4):1185–98. implications for somatic neurotransmission. Philos Trans R Soc Lond B Biol

[142] Clement AB, Gimpl G, Behl C. Oxidative stress resistance in hippocampal cells Sci 2015;370(1672).

13

is associated with altered membrane fluidity and enhanced nonamyloido- [170] Leftin A, Job C, Beyer K, Brown MF. Solid-state C NMR reveals annealing of

genic cleavage of endogenous amyloid precursor protein. Free Radic Biol Med raft-like membranes containing cholesterol by the intrinsically disordered

2010;48(9):1236–41. protein a-Synuclein. J Mol Biol 2013;425(16):2973–87.

[143] Ayasolla K, Khan M, Singh AK, Singh I. Inflammatory mediator and beta- [171] Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G, Barbosa ER, et al.

amyloid (25e35)-induced ceramide generation and iNOS expression are Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease.

inhibited by vitamin E. Free Radic Biol Med 2004;37(3):325–38. N Engl J Med 2009;361(17):1651–61.

[144] Grimm MO, Grimm HS, Pa¨tzold AJ, Zinser EG, Halonen R, Duering M, et al. [172] Gegg ME, Sweet L, Wang BH, Shihabuddin LS, Sardi SP, Schapira AH. No

Regulation of cholesterol and sphingomyelin metabolism by amyloid-beta evidence for substrate accumulation in Parkinson brains with GBA muta-

and presenilin. Nat Cell Biol 2005;7(11):1118–23. tions. Mov Disord 2015 Jul;30(8):1085–9.

[145] Dinkins MB, Dasgupta S, Wang G, Zhu G, Bieberich E. Exosome reduction in [173] Fabelo N, Martı´n V, Santpere G, Marı´n R, Torrent L, Ferrer I, et al. Severe

vivo is associated with lower amyloid plaque load in the 5XFAD mouse model alterations in lipid composition of frontal cortex lipid rafts from Parkinson’s

of Alzheimer’s disease. Neurobiol Aging 2014;35(8):1792–800. disease and incidental Parkinson’s disease. Mol Med 2011;17(9–10):1107–18.

[146] Yuyama K, Sun H, Mitsutake S, Igarashi Y. Sphingolipid-modulated exosome [174] Abbott SK, Li H, Mun˜oz SS, Knoch B, Batterham M, Murphy KE, et al. Altered

secretion promotes clearance of amyloid-b by microglia. J Biol Chem ceramide acyl chain length and ceramide synthase gene expression in

2012;287(14):10977–89. Parkinson’s disease. Mov Disord 2014;29(4):518–26.

[147] Lee JK, Jin HK, Park MH, Kim BR, Lee PH, Nakauchi H, et al. Acid sphingo- [175] Wang Q, Liu Y, Zhou J. Neuroinflammation in Parkinson’s disease and its

myelinase modulates the autophagic process by controlling lysosomal bio- potential as therapeutic target. Transl Neurodegener 2015;4:19.

genesis in Alzheimer’s disease. J Exp Med 2014;211(8):1551–70. [176] Hunot S, Brugg B, Ricard D, Michel PP, Muriel MP, Ruberg M, et al. Nuclear

[148] Bandaru VV, Troncoso J, Wheeler D, Pletnikova O, Wang J, Conant K, et al. translocation of NF-kappaB is increased in dopaminergic neurons of patients

ApoE4 disrupts sterol and sphingolipid metabolism in Alzheimer’s but not with Parkinson disease. Proc Natl Acad Sci U S A 1997;94(14):7531–6.

normal brain. Neurobiol Aging 2009;30(4):591–9. [177] He X, Chen F, Gatt S, Schuchman EH. An enzymatic assay for quantifying

[149] Maes M, Smith R, Christophe A, Vandoolaeghe E, Van Gastel A, Neels H, et al. sphingomyelin in tissues and plasma from humans and mice with Niemann–

Lower serum high-density lipoprotein cholesterol (HDL-C) in major depres- Pick disease. Anal Biochem 2001;293(2):204–11.

sion and in depressed men with serious suicidal attempts: relationship with [178] Lozano J, Morales A, Cremesti A, Fuks Z, Tilly JL, Schuchman E, et al. Disease

immune-inflammatory markers. Acta Psychiatr Scand 1997;95(3):212–21. versus acid sphingomyelinase deficiency. Cell Death Differ 2001;8(1):100–3.

[150] Musselman DL, Evans DL, Nemeroff CB. The relationship of depression to [179] Levran O, Desnick RJ, Schuchman EH. A frequent missense mutation in the

cardiovascular disease: epidemiology, biology, and treatment. Arch Gen acid sphingomyelinase gene of Ashkenazi Jewish type A and B patients. Proc

Psychiatry 1998;55(7):580–92. Natl Acad Sci U S A 1991;88(9):3748–52.

[151] Cizza G, Ravn P, Chrousos GP, Gold PW. Depression: a major, unrecognized [180] Simonaro CM, Desnick RJ, McGovern MM, Wasserstein MP, Schuchman EH.

risk factor for osteoporosis? Trends Endocrinol Metab 2001;12(5):198–203. The demographics and distribution of type B Niemann–Pick disease: novel

[152] Demirkan A, Isaacs A, Ugocsai P, Liebisch G, Struchalin M, Rudan I, et al. mutations lead to new genotype/phenotype correlations. Am J Hum Genet

Plasma phosphatidylcholine and sphingomyelin concentrations are associ- 2002;71(6):1413–9.

ated with depression and anxiety symptoms in a Dutch family-based lipi- [181] To´ th B, Erdo˝ s M, Sze´kely A, Ritli L, Bagossi P, Su¨ megi J, et al. Molecular genetic

domics study. J Psychiatr Res 2013;47(3):357–62. characterization of novel sphingomyelin phosphodiesterase 1 mutations

[153] Kornhuber J, Medlin A, Bleich S, Jendrossek V, Henkel AW, Wiltfang J, et al. causing Niemann–Pick disease. JIMD Rep 2012;3:125–9.

High activity of acid sphingomyelinase in major depression. J Neural Transm [182] Zhang H, Wang Y, Gong Z, Li X, Qiu W, Han L, et al. Identification of a distinct

(Vienna) 2005;112(11):1583–90. mutation spectrum in the SMPD1 gene of Chinese patients with acid sphingo-

[154] Geddes SD, Assadzada S, Sokolovski A, Bergeron R, Haj-Dahmane S, Be´ı¨que JC. myelinase-deficient Niemann–Pick disease. Orphanet J Rare Dis 2013;8:15.

Time-dependent modulation of glutamate synapses onto 5-HT neurons by [183] Irun P, Malle´n M, Dominguez C, Rodriguez-Sureda V, Alvarez-Sala LA, Arslan

treatment. Neuropharmacology 2015;95:130–43. N, et al. Identification of seven novel SMPD1 mutations causing Niemann–

[155] Meneses A. Tianeptine: 5-HT uptake sites and 5-HT(1–7) receptors modulate Pick disease types A and B. Clin Genet 2013;84(4):356–61.

memory formation in an autoshaping Pavlovian/instrumental task. Neurosci [184] Manshadi MD, Kamalidehghan B, Keshavarzi F, Aryani O, Dadgar S, Arasteh-

Biobehav Rev 2002;26(3):309–19. kani A, et al. Four novel p.N385K, p.V36A, c.1033-1034insT and c.1417-

[156] Sjo¨gren B, Svenningsson P. Depletion of the lipid raft constituents, sphingo- 1418delCT mutations in the sphingomyelin phosphodiesterase 1 (SMPD1)

myelin and , decreases serotonin binding at human 5-HT7(a) gene in patients with types A and B Niemann–Pick disease (NPD). Int J Mol Sci

receptors in HeLa cells. Acta Physiol (Oxf) 2007;190(1):47–53. 2015;16(4):6668–76.

[157] Weng R, Shen S, Yang L, Li M, Tian Y, Bai Y, et al. Lipidomic analysis of p- [185] Kwon HJ, Abi-Mosleh L, Wang ML, Deisenhofer J, Goldstein JL, Brown MS,

chlorophenylalanine-treated mice using continuous-flow two-dimensional et al. Structure of N-terminal domain of NPC1 reveals distinct subdomains for

liquid chromatography/quadrupole time-of-flight mass spectrometry. Rapid binding and transfer of cholesterol. Cell 2009;137(7):1213–24.

Commun Mass Spectrom 2015;29(16):1491–500. [186] Vanier MT, Millat G. Structure and function of the NPC2 protein. Biochim

[158] Kornhuber J, Tripal P, Reichel M, Mu¨ hle C, Rhein C, Muehlbacher M, et al. Biophys Acta 2004;1685(1–3):14–21.

Functional Inhibitors of Acid Sphingomyelinase (FIASMAs): a novel pharma- [187] Thomas GH, Tuck-Muller CM, Miller CS, Reynolds LW. Correction of sphingo-

cological group of drugs with broad clinical applications. Cell Physiol Bio- myelinase deficiency in Niemann–Pick type C fibroblasts by removal of lipopro-

chem 2010;26(1):9–20. tein fraction from culture media. J Inherit Metab Dis 1989;12(2):139–51.

[159] Gulbins E, Palmada M, Reichel M, Lu¨ th A, Bo¨hmer C, Amato D, et al. Acid [188] Pentchev PG, Comly ME, Kruth HS, Vanier MT, Wenger DA, Patel S, et al. A

sphingomyelinase-ceramide system mediates effects of antidepressant defect in cholesterol esterification in Niemann–Pick disease (type C) patients.

drugs. Nat Med 2013;19(7):934–8. Proc Natl Acad Sci U S A 1985;82(23):8247–51.

[160] Keshavan MS, Mallinger AG, Pettegrew JW, Dippold C. Erythrocyte mem- [189] McGovern MM, Aron A, Brodie SE, Desnick RJ, Wasserstein MP. Natural

brane phospholipids in psychotic patients. Psychiatry Res 1993;49(1): history of type A Niemann–Pick disease; possible endpoints for therapeutic

89–95. trials. Neurology 2006;66(2):228–32.

[161] Ponizovsky AM, Modai I, Nechamkin Y, Barshtein G, Ritsner MS, Yedgar S, [190] Wasserstein MP, Desnick RJ, Schuchman EH, Hossain S, Wallenstein S, Lamm

et al. Phospholipid patterns of erythrocytes in schizophrenia: relationships to C, et al. The natural history of type B Niemann–Pick disease: results from a

symptomatology. Schizophr Res 2001;52(1–2):121–6. 10-year longitudinal study. Pediatrics 2004;114(6):e672–7.

[162] Schmitt A, Wilczek K, Blennow K, Maras A, Jatzko A, Petroianu G, et al. Altered [191] Wasserstein MP, Larkin AE, Glass RB, Schuchman EH, Desnick RJ, McGovern

thalamic membrane phospholipids in schizophrenia: a postmortem study. MM. Growth restriction in children with type B Niemann–Pick disease. J

Biol Psychiatry 2004;56(1):41–5. Pediatr 2003;142(4):424–8.

[163] Colo´ n-Sa´ez JO, The Yakel JL. a7 nicotinic acetylcholine receptor function in [192] Marathe S, Miranda SR, Devlin C, Johns A, Kuriakose G, Williams KJ, et al.

hippocampal neurons is regulated by the lipid composition of the plasma Creation of a mouse model for non-neurological (type B) Niemann–Pick

membrane. J Physiol 2011;589(Pt 13):3163–74. disease by stable, low level expression of lysosomal sphingomyelinase in the

B. Kamil et al. / Pharmacological Reports 68 (2016) 570–581 581

absence of secretory sphingomyelinase: relationship between brain intra- leads to visceral, but not neurological, correction of the pathophysiology.

lysosomal enzyme activity and central nervous system function. Hum Mol FASEB J 2000;14(13):1988–95.

Genet 2000;9(13):1967–76. [196] Dodge JC, Clarke J, Treleaven CM, Taksir TV, Griffiths DA, Yang W, et al.

[193] Gabande´-Rodrı´guez E, Boya P, Labrador V, Dotti CG, Ledesma MD. High Intracerebroventricular infusion of acid sphingomyelinase corrects CNS

sphingomyelin levels induce lysosomal damage and autophagy dysfunction manifestations in a mouse model of Niemann–Pick A disease. Exp Neurol

in Niemann Pick disease type A. Cell Death Differ 2014;21(6):864–75. 2009;215(2):349–57.

[194] Macauley SL, Sidman RL, Schuchman EH, Taksir T, Stewart GR. Neuropathol- [197] Murray JM, Thompson AM, Vitsky A, Hawes M, Chuang WL, Pacheco J, et al.

ogy of the acid sphingomyelinase knockout mouse model of Niemann–Pick A Nonclinical safety assessment of recombinant human acid sphingomyelinase

disease including structure–function studies associated with cerebellar Pur- (rhASM) for the treatment of acid sphingomyelinase deficiency: the utility of

kinje cell degeneration. Exp Neurol 2008;214(2):181–92. animal models of disease in the toxicological evaluation of potential thera-

[195] Miranda SR, He X, Simonaro CM, Gatt S, Dagan A, Desnick RJ, et al. Infusion of peutics. Mol Genet Metab 2015;114(2):217–25.

recombinant human acid sphingomyelinase into Niemann–Pick disease mice 本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

提供一站式文献检索和下载服务”的24 小时在线不限IP 图书馆。 图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

图书馆导航:

图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具