You are on page 1of 20

Progress in Lipid Research 63 (2016) 50–69

Contents lists available at ScienceDirect

Progress in Lipid Research

journal homepage: www.elsevier.com/locate/plipres

Synthesis and degradation pathways, functions, and pathology of


ceramides and epidermal acylceramides
Akio Kihara
Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo, Nishi 6-choume, Kita-ku, Sapporo 060-0812, Japan

a r t i c l e i n f o a b s t r a c t

Article history: Ceramide (Cer) is a structural backbone of sphingolipids and is composed of a long-chain base and a fatty acid.
Received 22 February 2016 Existence of a variety of Cer species, which differ in chain-length, hydroxylation status, and/or double bond num-
Received in revised form 8 April 2016 ber of either of their hydrophobic chains, has been reported. Ceramide is produced by Cer synthases. Mammals
Accepted 20 April 2016
have six Cer synthases (CERS1–6), each of which exhibits characteristic substrate specificity toward acyl-CoAs
Available online 21 April 2016
with different chain-lengths. Knockout mice for each Cer synthase show corresponding, isozyme-specific pheno-
Keywords:
types, revealing the functional differences of Cers with different chain-lengths. Cer diversity is especially promi-
Acylceramide nent in epidermis. Changes in Cer levels, composition, and chain-lengths are associated with atopic dermatitis.
Ceramide Acylceramide (acyl-Cer) specifically exists in epidermis and plays an essential role in skin permeability barrier
Fatty acid formation. Accordingly, defects in acyl-Cer synthesis cause the cutaneous disorder ichthyosis with accompanying
Long-chain base severe skin barrier defects. Although the molecular mechanism by which acyl-Cer is generated was long unclear,
Skin barrier most genes involved in its synthesis have been identified recently. In Cer degradation pathways, the long-chain
Sphingolipid base moiety of Cer is converted to acyl-CoA, which is then incorporated mainly into glycerophospholipids. This
pathway generates the lipid mediator sphingosine 1-phosphate. This review will focus on recent advances in
our understanding of the synthesis and degradation pathways, physiological functions, and pathology of Cers/
acyl-Cers.
© 2016 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
2. Structural diversity of ceramides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
2.1. Long-chain bases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
2.2. Fatty acids in ceramides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
2.3. Diversity of ceramides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
3. Synthesis of ceramides. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
3.1. Serine palmitoyltransferases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
3.2. 3-Ketodihydrosphingosine reductase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
3.3. Ceramide production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
3.4. Ceramide synthases. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
3.4.1. Ceramide synthases in yeast . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
3.4.2. CERS1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55

Abbreviations: ABC, ATP-binding cassette; acyl-Cer, acylceramide; ACS, acyl-CoA synthetase; ACSBG, acyl-CoA synthetase bubblegum; ACSF, acyl-CoA synthetase family; ACSL, acyl-
CoA synthetase long-chain; ACSM, acyl-CoA synthetase medium-chain; ACSS, acyl-CoA synthetase short-chain; ACSVL, acyl-CoA synthetase very long-chain; ALDH, aldehyde dehydroge-
nase; ARCI, autosomal recessive congenital ichthyosis; BAT, brown adipose tissue; CE, cornified envelope; Cer, ceramide; Cer (Sph), ceramide containing sphingosine; CHO, aldehyde; CLE,
corneocyte lipid envelope; COOH, carboxylic acid; dihydro-Cer, dihydroceramide; dihydro-Sph, dihydrosphingosine; dihydro-S1P, dihydrosphingosine 1-phosphate; ER, endoplasmic re-
ticulum; FA, fatty acid; FAS, fatty acid synthase; GalCer, galactosylceramide; GlcCer, glucosylceramide; KDS, 3-ketodihydrosphingosine; KO, knockout; LacCer, lactosylceramide; LC, long-
chain; LCB, long-chain base; LCBP, long-chain base 1-phosphate; LCFA, long-chain fatty acid; MC, medium-chain; 2-OH, 2-hydroxy; ω-OH, ω-hydroxy; 6-OH Sph, 6-hydroxysphingosine;
PC, phosphatidylcholine; phyto-Cer, phytoceramide; phyto-Sph, phytosphingosine; phyto-S1P, phytosphingosine 1-phosphate; PUFA, polyunsaturated fatty acid; SLS, Sjögren-Larsson
syndrome; SM, sphingomyelin; Sph, sphingosine; S1P, sphingosine 1-phosphate; SPT, serine palmitoyltransferase; STGD3, Stargardt disease type 3; tC16:1-CHO, trans-2-hexadecenal;
tC16:1-COOH, trans-2-hexadecenoic acid; tC16:1-CoA, trans-2-hexadecenoyl-CoA; ULC, ultra-long-chain; ULCFA, ultra-long-chain fatty acid; VLC, very-long-chain; VLCFA, very-long-
chain fatty acid; X-ALD, X-linked adrenoleukodystrophy.
E-mail address: kihara@pharm.hokudai.ac.jp.

http://dx.doi.org/10.1016/j.plipres.2016.04.001
0163-7827/© 2016 Elsevier Ltd. All rights reserved.
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 51

3.4.3. CERS2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
3.4.4. CERS3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
3.4.5. CERS4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
3.4.6. CERS5 and CERS6 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
3.4.7. Regulation of ceramide synthases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
4. Very-long-chain fatty acids in ceramides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
4.1. Roles of very-long-chain fatty acids in ceramides/sphingolipids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
4.2. Generation of very-long-chain fatty acids via fatty acid elongation cycles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
4.3. Different substrate specificities and physiological functions of fatty acid elongases . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
4.3.1. ELOVL1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
4.3.2. ELOVL2 and ELOVL5 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
4.3.3. ELOVL3 and ELOVL7 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
4.3.4. ELOVL4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
4.3.5. ELOVL6 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
4.3.6. Fatty acid elongases in yeast . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
4.4. 3-Ketoacyl-CoA reductase and trans-2-enoyl-CoA reductase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
4.5. 3-Hydroxyacyl-CoA dehydratases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
5. Acylceramide and its importance in skin barrier formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
5.1. Skin barrier and structure of epidermis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
5.2. Formation of lipid lamellae in stratum corneum . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
5.3. Ceramides in epidermis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
5.4. Acylceramide synthetic pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
5.5. Protein-bound ceramide synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
6. Ceramide degradation pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
6.1. Ceramide degradation pathways. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
6.2. Ceramide degradation by ceramidases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
6.3. Long-chain base 1-phosphate metabolic enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
6.3.1. Sphingosine kinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
6.3.2. Long-chain base 1-phosphate-degrading enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 62
6.4. The aldehyde dehydrogenase ALDH3A2 and Sjögren-Larsson syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
6.4.1. Aldehyde dehydrogenases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
6.4.2. Sjögren-Larsson syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
6.5. Acyl-CoA synthetases involved in ceramide degradation pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
6.6. Dual functions of the trans-2-enoyl-CoA reductase TECR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
6.7. Metabolism of phytosphingosine to odd-numbered fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
6.8. Generation of 2-hydroxy fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
7. Concluding remarks. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64

1. Introduction Intracellular sphingolipids constitute ~ 10% of total lipids in mam-


mals [9], although their levels vary among tissues. Sphingolipids are
Ceramide (Cer) is a hydrophobic backbone of sphingolipids. enriched in the plasma membrane, especially in the outer leaflet [10].
Sphingolipids exist in all eukaryotes, but not in prokaryotes, except for Sphingolipids account for 20–30% of total plasma membrane lipids [9].
a limited number of bacteria species such as Sphingomonas [1]. Cer con- SM is the most abundant sphingolipid species in mammals; SM levels
tains two hydrophobic chains, a long-chain base (LCB) and a fatty acid are higher than even the sum of all glycosphingolipid levels in most tis-
(FA), which are connected via an amide bond (Fig. 1A). Addition of a sues. In erythrocytes, in which the only cellular membrane is the plasma
polar head group at the C1 hydroxyl group of the LCB portion of Cer en- membrane, SM and glycosphingolipids account for 18% and 10% of total
dows the resulting sphingolipid with amphipathic properties. Charac- lipids, respectively [11].
teristic in vivo head group types differ among organisms [2–5]. In Levels of glycerophospholipids, another lipid class constituting
mammals, it is either phosphocholine [in sphingomyelin (SM)] or membranes, are much higher than those of sphingolipids [9]. Therefore,
sugar chains (in glycosphingolipids) [5,6] (Fig. 1A). Hundreds of only glycerophospholipids may be sufficient to build biological mem-
glycosphingolipids differing in sugar classes and/or their linking branes, as is the case for prokaryotes. Rather, the importance of
modes exist. The sugar residues found in mammalian sphingolipids sphingolipids may lie in their conferring specialized functions and/or
are glucose, galactose, N-acetylglucosamine, N-acetylgalactosamine, fu- properties to the membranes. During the course of evolution, eukary-
cose, and sialic acid [5,7,8] (Fig. 1B). The simplest glycosphingolipids are otes produced a variety of cell types differing in functions: creation of
glucosylceramide (GlcCer) and galactosylceramide (GalCer), which re- a multiplicity of sphingolipids with individualized functions was likely
spectively contain a glucose or galactose residue linked to Cer via β- necessary to fulfill this wide range of roles. Loss of sphingolipids by mu-
linkage (Fig. 1C). GlcCer and GalCer are collectively referred as cerebro- tations in sphingolipid biosynthetic genes is lethal in all organisms ex-
side or mono-hexosylceramide. Sulfatide is a sulfated derivative of amined to date [12–15], indicating that sphingolipids indeed possess
GalCer (Fig. 1B and C). Gangliosides refer to glycosphingolipids contain- specialized functions and cannot be substituted by
ing sialic acid(s). Other derivatives of GalCer include the ganglioside glycerophospholipids.
GM4 and the gala-series of glycosphingolipids (Fig. 1C). Addition of a Sphingolipids are involved in a variety of physiological functions in-
galactose residue to GlcCer produces lactosylceramide (LacCer), which cluding skin barrier formation, myelin maintenance, immunity, blood
is a precursor of SM4, a sulfated derivative of LacCer, and numerous vessel stabilization, recognition of bacteria, bacterial toxins, and viruses,
glycosphingolipid series (the ganglio-series, asialoganglio-series, insulin resistance, spermatogenesis, and auditory sense formation [5,7,
isoganglio-series, globo-series, isoglobo-series, muco-series, lacto- 16–22]. They fulfill these roles through modulating cellular events
series, and neolacto-series) (Fig. 1C). such as lipid microdomain formation, apoptosis, organelle and
52 A. Kihara / Progress in Lipid Research 63 (2016) 50–69

Fig. 1. Structures of sphingolipids and series of glycosphingolipids. (A) Structures of sphingolipids. R represents a polar head group: phosphocholine in SM or sugar chains in
glycosphingolipids. (B) Sugars used in mammalian glycosphingolipids. The simplest glycosphingolipids are GlcCer and GalCer, which contain a glucose and a galactose residue,
respectively. Sulfatide is a sulfated derivative of GalCer. (C) Glycosphingolipid series in mammals. Each number denotes the position of a bond: the C1 hydroxy group of the numbered
sugar residue links to the numbered position of the sugar residue to its left via an α-linkage (underlined) or a β-linkage (non-underlined).

membrane structure organization, survival, migration, signaling, intra- major mammalian LCBs are dihydrosphingosine (dihydro-Sph;
cellular protein trafficking, autophagy, adhesion, stress response, and sphinganine), sphingosine (Sph), phytosphingosine (phyto-Sph), and
metabolic regulation [8,16,19,20,23–29]. Among them, Cer plays impor- 6-hydroxysphingosine (6-OH Sph) (Fig. 2A). The existence of
tant roles in skin barrier formation by forming multi-lamellar lipids sphingadienine having two double bonds and dihydroxy-
(lipid lamella) in the stratum corneum of epidermis [17,30,31]. GalCer dihydrosphingosine having four hydroxyl groups has also been report-
and sulfatide are enriched in myelin and are important for myelin func- ed [37–39]. Since the saturated LCB dihydro-Sph is synthesized in the
tion and maintenance [22]. The Cer metabolite sphingosine 1- de novo sphingolipid biosynthetic pathway, it is a common precursor
phosphate (S1P) is a lipid mediator and functions in immunity and of all sphingolipids. Accordingly, dihydro-Sph exists in all tissues, al-
blood vessel stabilization among other capacities [16,32,33]. S1P binds though its levels are low. Sph, which contains a trans double bond be-
to cell surface receptors (S1P1–S1P5) and induces several cellular re- tween C4 and C5 positions, is the major LCB in mammals and exists
sponses. In the immune system, S1P plays a central role in the egress ubiquitously among tissues. Phyto-Sph contains an extra hydroxyl
of T lymphocytes from thymus and secondary lymphoid tissues [16, group at the C4 position and exists in restricted tissues such as epider-
32,33]. This function of S1P has already been applied clinically, with mis, intestine, and kidney [38,40–42]. 6-OH Sph, which contains a
the immunomodulator fingolimod (FTY720) being used as a therapeu- trans double bond between the C4 and C5 positions and an extra hy-
tic agent for multiple sclerosis [34,35]. droxyl group at C6, exists only in epidermis [38,42]. Epidermis is unique
in that it contains all of these four types of LCBs. The chain-length of
2. Structural diversity of ceramides LCBs is mostly C18 in mammals, although LCBs with other chain-
lengths also exist [36].
2.1. Long-chain bases
2.2. Fatty acids in ceramides
Cer is composed of an LCB and a FA. The structures of LCBs varied
among organisms [36]; however, all LCBs contain two hydroxyl groups FAs are classified into short-chain FAs, medium-chain (MC) FAs,
at the C1 and C3 positions and one amino group at the C2 position. The long-chain (LC) FAs (LCFAs), and very-long-chain (VLC) FAs (VLCFAs),
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 53

Fig. 2. Components and structures of mammalian Cers. (A and B) Structures of LCBs (A) and FAs (B) and their tissue distributions. (C) Nomenclature of 12 ceramide classes. Each class is
composed of a specific FA and LCB, and its name is a combination of their corresponding abbreviations. (D) Of the 12 ceramide classes, the structures of NS [combination of non-hydroxy FA
(N) and Sph (S)] and EOS [combination of esterified ω-hydroxy FA (EO) and Sph(S)] are illustrated.
54 A. Kihara / Progress in Lipid Research 63 (2016) 50–69

depending on their carbon chain-lengths. Agreement on which ranges 3. Synthesis of ceramides


of chain-lengths correspond to which category has not been completely
reached among researchers. In this review, the most widely accepted 3.1. Serine palmitoyltransferases
classification is used: short-chain FAs (C2–C4), medium-chain FAs
(C5–C10), LCFAs (C11–C20), and VLCFAs (≥ C21) [43,44] (Fig. 2B). De novo sphingolipid biosynthesis occurs in the endoplasmic reticu-
Moreover, VLCFAs with ≥ C26 are sometimes referred as ultra-long- lum (ER) from the first reaction, catalyzed by serine
chain (ULC) FAs (ULCFAs). The FAs found in glycerophospholipids are palmitoyltransferase (SPT), to Cer synthesis. SPT catalyzes condensation
mostly LCFAs [45]. On the other hand, VLCFAs, especially lignoceric of L-serine with a FA using an acyl-CoA as a donor, generating 3-
acid (C24:0-COOH) and nervonic acid (C24:1-COOH), are often found ketodihydrosphingosine (KDS). This step is a rate-limiting step of
in sphingolipids (Fig. 2B). For example, SMs containing C24 VLCFAs ac- sphingolipid synthesis [53] (Fig. 3A). In mammals, SPTLC1, SPTLC2
count for 20–50% of total SMs in mouse tissues, with the highest in liver (SPTLC2a), and SPTLC3 (SPTLC2b) encode large subunits of SPTs, whereas
and the lowest in testis and skeletal muscle among 13 tissues examined SPTSSA (ssSPTa) and SPTSSB (ssSPTb) encode small subunits of SPTs
[44]. Another major chain-length found in the FA moiety of [53–55]. SPTs are composed of SPTLC1, either of SPTLC2 or SPTLC3, and ei-
sphingolipids is C16 (Fig. 2B). C16 SM comprises 20–60% of total ther of ssSPTa or ssSPTb. These combinations generate four types of SPTs
SMs in most tissues, whereas its levels are low in brain and skeletal (SPTLC1/2/ssSPTa, SPTLC1/3/ssSPTa, SPTLC1/2/ssSPTb, and SPTLC1/3/
muscle (10–15%). In these tissues, C18 SMs abundantly exist instead ssSPTb) (Fig. 3A), in which SPTLC2 and 3 are catalytic subunits. SPTs
(~ 40%). C22 SMs account for 5–20% of total SMs [44]. The amounts need a pyridoxal 5′-phosphate as a coenzyme [53]. Although SPTLC1, 2,
of C20 SMs are low in most tissues, usually 3–10%, but are exceptionally and 3 share high sequence similarity, SPTLC1 lacks the active site lysine
high in heart (20%) [44]. Most FAs found in sphingolipids are satu- residue that binds to pyridoxal 5′-phosphate. However, SPTLC1 is still im-
rated or monounsaturated, in contrast to glycerophospholipids, portant because it confers stability to SPTLC2 [56] and maybe also to
which often contain polyunsaturated FAs (PUFAs) at the sn-2 position SPTLC3. The activities of SPTs are regulated to control cellular Cer/
[44,45]. sphingolipid levels. Orm family proteins (Orm1 and 2 in yeast and
Sphingolipids with ULCFAs are low-level and barely detectable in ORMDL1–3 in mammals) are involved in this regulation, specifically by
most tissues; however, epidermis and spermatogenic cells in testis spe- negative regulation of SPTs through physical interactions [57,58].
cifically contain ULC-sphingolipids [21,38,42]. More specifically, Cers The substrate specificities of SPTs toward acyl-CoAs are different,
containing C26–C36 ULCFAs exist in epidermis [46]. Among them, leading to diversity in LCB chain-lengths. SPTLC1/2/ssSPTa prefers
≥ C30 Cers are often ω-hydroxylated (Fig. 2B) and esterified with C16:0-CoA and generates C18 KDS, the precursor of the most abundant
linoleic acid (C18:2-COOH), generating ω-O-acylceramides [or simply LCB class, C18 LCBs [55]. On the other hand, SPTLC1/2/ssSPTb and
acylceramides (acyl-Cers)]. The importance of acyl-Cers in skin barrier SPTLC1/3/ssSPTa exhibit high activity toward C18:0-CoA and C14:0-
formation is described below in detail. Although most of the FAs found CoA, producing C20 and C16 KDSs, respectively. SPTLC1/3/ssSPTb ex-
in sphingolipids are saturated or monounsaturated, ULC-sphingolipids hibits broad substrate specificity from C14- to C20-CoAs.
in testis are exceptionally polyunsaturated [43,44,47] (Fig. 2B). They SPTLC1 and SPTLC2 are the causative genes of hereditary sensory
are fucose-containing glycosphingolipids having C26–C32 ULCFAs neuropathy type I, which is an autosomal dominant peripheral neurop-
with four to six double bonds [21,47]. These specialized sphingolipids athy [59,60]. Although SPTs normally use serine as a substrate, specific
are required for spermatogenesis [21]. point mutations in SPTLC1 or SPTLC2 cause the substrate specificities of
Most cellular FAs are non-hydroxy FAs. However, in addition to the their gene products to become promiscuous and accept alanine and gly-
ω-hydroxy (ω-OH) FAs present in epidermis, some tissues such as epi- cine. Atypical LCBs are produced as a result: 1-deoxysphinganine (from
dermis, intestine, and brain contain 2-hydroxy (α-hydroxy; 2-OH) FAs alanine) and 1-deoxymethylsphinganine (from glycine) [61]. Since
[48–52] (Fig. 2B). These 2-OH FAs are exclusively used for sphingolipids these deoxy-LCBs lack the hydroxyl group at C1, which is required for
and are not found in glycerophospholipids. the attachment of polar head groups, they cannot be converted to com-
plex sphingolipids. The accumulated deoxy-LCBs and/or deoxy-Cers
show neurotoxicity.
2.3. Diversity of ceramides
3.2. 3-Ketodihydrosphingosine reductase
In a narrow definition, the term Cer represents only Sph-containing
Cer [Cer (Sph)] and is distinguished from dihydroceramide (dihydro- The second step of sphingolipid biosynthesis is reduction of KDS,
Cer) and phytoceramide (phyto-Cer), which contains dihydro-Sph and generating the LCB dihydro-Sph (Fig. 3A). This step is catalyzed by
phyto-Sph, respectively. However, in a wider definition, Cer represents KDS reductase, which is encoded by KDSR (FVT-1) in mammals [62].
all combinations of the above-mentioned classes of LCBs and FAs. This KDSR was reported as a candidate gene for spinal muscular atrophy in
review uses Cer in this wider sense, and Cer (Sph) for the narrow bovines [63].
definition.
Cer diversity is especially prominent in epidermis. To discriminate 3.3. Ceramide production
the epidermal Cer classes, a nomenclature is used in which abbrevia-
tions for types of LCBs (DS, dihydro-Sph; S, Sph; P, phyto-Sph; H, 6- Dihydro-Sph is converted to dihydro-Cer by Cer synthases using
OH Sph) and FAs (N, non-hydroxy; A, α-hydroxy; EO, esterified ω- acyl-CoAs as FA donors. Subsequently, dihydro-Cer is desaturated to
hydroxy) are combined (Fig. 2C). For example, NS (which is the most Cer (Sph) or hydroxylated to phyto-Cer in mammals (Fig. 3A). The
abundant Cer class in mammals) represents the combination of Sph dihydro-Cer desaturase DEGS1 (DES1) catalyzes Cer (Sph) generation
and a non-hydroxy FA (Fig. 2D). EOS, EOP, EOH, and EODS represent [64]. On the other hand, DEGS2 (DES2) is responsible for phyto-Cer pro-
acyl-Cers (Fig. 2C and D). Existence of 11 Cer classes other than EODS duction [64]. In addition to hydroxylase activity, DEGS2 also has weak
in the stratum corneum of human epidermis was reported in 2008; desaturase activity that can create Cer (Sph). In Degs1 knockout (KO)
when differences in chain-length were taken into account, the number mice, Cer (Sph) levels are largely reduced (~15% of wild type levels),
of detected Cer species reached 342 [42]. Furthermore, more recent, but the levels of dihydro-Cer, the substrate of Degs1, are concomitantly
high-resolution mass spectrometric analysis detected EODS and anoth- increased [65]. Disruption of Degs1 causes pleiotropic effects, including
er class of Cers with LCBs containing four hydroxyl groups (dihydroxy- scaly skin, sparse hair, tremors, hematological abnormalities, growth re-
dihydrosphingosine) in human stratum corneum, with an estimated tardation, decreased liver function, and reduced life span (dying within
Cer diversity of N1000 species [38]. 8 to 10 weeks of birth) [65], indicating the physiological importance of
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 55

Fig. 3. Ceramide synthetic pathways and Cer synthases. (A) Synthetic pathways of Cers, the catalytic enzymes of each reaction, and their related hereditary disorders. (B) Substrate
specificity (1), tissue distribution (2), and phenotypes of respective Cer synthase KO mice (3) are depicted for each CERS family member. WAT, white adipose tissue; HFD, high fat diet.

Cer (Sph). Expression of Degs2 mRNA is confined to skin, kidney, intes- share high sequence similarity [67,68]. Lip1 is a small subunit of Lag1
tine, and lung [66], consistent with the tissue-distribution pattern of and Lac1 that is required for their enzyme activities [69]. Deletion of
phyto-Cers. The molecular mechanism of 6-OH Cer creation remains LAG1 or LAC1 does not affect growth, whereas double deletion of LAG1
unclear. and LAC1 (lag1Δ lac1Δ) or single deletion of LIP1 (lip1Δ) cause severe
Mammals cannot directly convert dihydro-Sph to Sph. Therefore, Sph growth defects [69,70]. In addition, lag1Δ lac1Δ cells contain only
is generated only through breakdown of Cer (Sph). In the de novo trace amounts of Cers, produced via reverse reaction by ceramidases
sphingolipid synthetic pathway, Cer synthases catalyze production of [67]. Six mammalian Cer synthases (CERS1–6/LASS1–6) that differ in
dihydro-Cer from dihydro-Sph and acyl-CoA as described above. Cer substrate specificity toward acyl-CoAs have been identified based on
synthases can also use other LCBs generated through salvage pathways, their sequence similarity to yeast Lag1 and Lac1 [17]. However, mam-
i.e., those generated by ceramidases in sphingolipid degradation path- mals lack a Lip1 homolog.
ways. When Cer synthases use Sph as a substrate, Cer (Sph) is directly
produced. 3.4.2. CERS1
CERS1 (LASS1/UOG1) was the first Cer synthase to be identified in
3.4. Ceramide synthases mammals [71]. CERS1 exhibits high activity toward C18-CoA and is
expressed in brain, especially in neurons [25,71–75] (Figs. 3B and 4).
3.4.1. Ceramide synthases in yeast Cers1-deficient mice, whether the loss of function is spontaneously gen-
Cer synthases were firstly identified in yeast [67,68]. Yeast Saccharo- erated or constructed by gene targeting, exhibit defects in cerebellar de-
myces cerevisiae contains the two Cer synthases Lag1 and Lac1, which velopment, locomotion, and motor coordination [73,76].
56 A. Kihara / Progress in Lipid Research 63 (2016) 50–69

3.4.3. CERS2 glands [91]. Cers4 KO mice exhibit progressive hair loss due to physical
CERS2 (LASS2/TRH3) prefers C22- and C24-CoAs as substrates [72, blocking of the hair canal, with concomitant diminished levels of C18:0
75,77,78] (Figs. 3B and 4). CERS2 mRNA is widely expressed among tis- and C20:0 Cers in the epidermis [91] (Fig. 3B).
sues, with the highest expression in liver and kidney [25,72,78]. This ex-
pression pattern is well-correlated with the distribution patterns of C22 3.4.6. CERS5 and CERS6
and C24 VLC-sphingolipids [44]. In brain, CERS2 is highly expressed in CERS5 (LASS5/TRH4) and CERS6 (LASS6) are redundant Cer
oligodendrocytes, which form myelin [74,79]. Myelin is a multilamellar synthases exhibiting activity toward C16-CoA [25,72,75,90,92]
membrane enriched in lipids, and their electrical insulation properties (Figs. 3B and 4). CERS5 and CERS6 are expressed ubiquitously with
are important for neural functions [22]. C24 sphingolipids such as high expressions observed for CERS5 in white adipose tissue, testis,
GalCer, sulfatide, and sphingomyelin are abundant in myelin [22,74, and lung and for CERS6 in intestine and kidney [25,72,90,93,94]. Cers5
79,80]. In accordance with the preponderance of VLC-sphingolipids in KO mice are resistant to high fat diet-induced obesity and display im-
liver and myelin, disruption of Cers2 in mice causes liver dysfunction proved glucose tolerance and insulin sensitivity [94] (Fig. 3B). Cers6
such as hepatopathy and hepatocarcinomas and myelin disorders such KO mice exhibit defective hindlimb grasping and habituation in the
as myelin sheath defects and cerebellar degeneration [79,81,82] open field test [93]. In brain, Cers6 is expressed in hippocampus, cortex,
(Fig. 3B). The myelin phenotype of Cers2 KO mice seems to be linked and Purkinje cells [93]. Cers6 KO mice also exhibit exacerbation of ex-
to a decrease in C24 GalCer levels [79], since GalCer and its derivative perimental autoimmune encephalomyelitis [95]. The Cers6 product
sulfatide are important for myelin function and stability [22,83–85]. C16 Cer mediates anti-inflammatory effects, probably by suppressing
the migration and activation of neutrophils [95].
3.4.4. CERS3
Although most Cer synthases exhibit relatively strict substrate spec- 3.4.7. Regulation of ceramide synthases
ificity toward acyl-CoAs, CERS3 (LASS3) has broad substrate specificity, Cer synthases are regulated by phosphorylation. Yeast Cer synthases
exhibiting activity toward ≥ C18-CoAs [75,86] (Figs. 3B and 4). This Lag1 and Lac1 are phosphorylated at the N-terminal regions by Ypk1,
broad substrate specificity enables CERS3 to produce a wide range of the downstream kinase of target of rapamycin complex 2 (TORC2),
Cers including the ULC-Cers found in epidermis (saturated and mono- and at the C-terminal regions by the casein kinase 2 Cka2 [96,97]. The
unsaturated ULC-Cers) and in testis (polyunsaturated ULC-Cers) [21, Ypk1-dependent phosphorylation activates Cer synthase activity and
87]. CERS3 mRNA expression is restricted to these tissues [25,86,88]. is increased upon sphingolipid depletion or heat shock [96]. The phos-
Mutations in human CERS3 gene cause autosomal recessive congenital phorylation by Cka2 is also important for efficient ceramide biosynthe-
ichthyosis (ARCI) [89]. The skin disorder ichthyosis is caused by skin sis [97].
barrier defects and is characterized by dry, thickened, and scaly skin. Cer synthases are ER membrane proteins. Yeast Lag1 and Lac1 con-
Disruption of Cers3 in mice also causes severe skin barrier defects and tain eight transmembrane helixes, and their N-termini and C-termini
neonatal lethality [87] (Fig. 3B). In the keratinocytes of ARCI patients face the cytosol [98]. On the other hand, mammalian CERS1–6 lack the
and in the epidermis of Cers3 KO mice, levels of ≥ C24 Cers, including N-terminal cytosolic domain that includes the Ypk1 phosphorylation
acyl-Cers, are reduced [87,89]. These observations indicate that ULC- sites and the first transmembrane helix present in yeast Cer synthases.
Cers and/or acyl-Cers are essential for skin barrier formation. In addi- Accordingly, CERS1–6 are not phosphorylated at their N-termini. How-
tion, the polyunsaturated ULC-Cers synthesized by CERS3 are important ever, mammalian Cer synthases except CERS1 do contain the C-terminal
in spermatogenesis: germ cell-specific Cers3 KO mice exhibit male infer- phosphorylation sites and are phosphorylated accordingly [75]. CERS2,
tility [21] (Fig. 3B). CERS4, CERS5, and CERS6 are phosphorylated by casein kinase 2, where-
as another kinase seems to be responsible for the phosphorylation of
3.4.5. CERS4 CERS3. This phosphorylation is especially important for CERS2 activity,
CERS4 (LASS4/TRH1) exhibits activity toward C18- to C22-CoAs and whereas it modestly increases the activities of CERS4 and CERS5 and
is expressed in skin and lung [72,75,77,90] (Figs. 3B and 4). In mouse mildly increases those of CERS3 and CERS6. Although CERS1 is not phos-
skin, Cers4 is expressed in suprabasal epidermal layers and sebaceous phorylated at the C-terminal region, it is phosphorylated at another site

Fig. 4. Interplay between Cer synthases and FA elongases in the production of Cers with different chain-lengths. Pathways and involved enzymes (Cer synthases and FA elongases) for
mammalian FA and Cer syntheses are illustrated. C and E represent CERS and ELOVL isozymes, respectively, and their sizes reflect the strength of their enzyme activities. SFA, saturated
FA; MUFA, monounsaturated FAs.
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 57

upon activation of protein kinase C [99]. This phosphorylation protects elongated up to C26 in yeast. The third, dehydration step of the FA elon-
CERS1 from stress-induced degradation. gation cycle is also catalyzed by multiple isozymes (HACD1–4) in mam-
mals (Fig. 5A), but by only one enzyme (Phs1) in yeast [107,113]. The
4. Very-long-chain fatty acids in ceramides second and fourth steps of the FA elongation cycle are each catalyzed
by a single gene product in mammals (second step, KAR/HSD17B12;
4.1. Roles of very-long-chain fatty acids in ceramides/sphingolipids fourth step, TECR/TER) [114]. In yeast, the second step is catalyzed pri-
marily by Ifa38/Ybr159w but also by the minor isozyme Ary1 [115],
Sphingolipids contain more hydrogen donors and acceptors com- and the fourth step is catalyzed by Tsc13 [116].
pared to glycerophospholipids. Furthermore, the hydrophobic chains Each step of the FA elongation cycle seems not to occur indepen-
in sphingolipids usually do not contain a cis-double bond, which pre- dently but, rather, to be regulated cooperatively with the other steps.
vents tight packaging with neighborhood lipids, in contrast to For example, the activity of ELOVL6, which catalyzes the first step of
glycerophospholipids, which usually contain an unsaturated FA with the FA elongation cycle, is enhanced by KAR, which catalyzes the second
cis-double bonds at the sn-2 position [45]. These structural properties step [117]. Furthermore, decreased TECR, which is responsible for the
enable sphingolipids to form lipid microdomains or lipid rafts in biolog- fourth step, by mutation or gene knockdown causes a decrease in the
ical membranes (especially in plasma membrane) together with choles- enzyme activity of HACDs, which catalyze the third step [118]. These
terol [23]. Increases in chain-length and degree of saturation cause a regulations may be carried out through interactions among the protein
decrease in membrane fluidity and an increase in membrane order, re- components of the FA elongation machinery [113,119,120].
spectively, stimulating stability of membrane microdomains [100,101]. Production of VLCFAs is essential for embryogenesis and survival in
Thus, VLC-sphingolipids are important for lipid microdomain formation mammals. Disruption of KAR in mice causes lethality at embryonic day
and stabilization. Lipid microdomains act as a platform for signaling 9.5 [121]. VLCFAs are also essential for yeast growth. Mutations that
proteins and facilitate efficient signaling [102]. It has been shown that cause complete abrogation of VLCFA production, such as fen1Δ sur4Δ,
C24 sphingolipids are required for the activation of the Src family kinase ifa38Δ ary1Δ, phs1Δ, and tsc13Δ mutations, are all lethal [107,115,116,
Lyn [103,104]. Apoptosis signaling is also modulated by VLC- 122]. The lethality of such mutations in yeast is closely related to the
sphingolipids. Altering total sphingolipid composition to contain fewer organism's inability to produce sphingolipids [123–125]. The yeast Cer
VLC- and more LC-sphingolipids increases susceptibility to apoptosis synthase Lag1/Lac1 seems unable to use LC acyl-CoAs as substrates.
[25,105]. VLC-sphingolipids seem to be also important for generation When the mammalian CERS5, which produces C16 and C18 Cers, is
and stabilization of highly curved membrane structures. Myelin is one expressed in tsc13Δ cells, TSC13 disruption is no longer lethal [125].
such structure: the Cers2 KO mice studies described above demonstrate
the importance in VLC-sphingolipids in myelin function and stability 4.3. Different substrate specificities and physiological functions of fatty acid
[79,81]. VLCFAs have also been reported to be important in vesicular elongases
trafficking (especially at the late endosome/multivesicular body),
where a highly curved membrane is generated during vesicle budding 4.3.1. ELOVL1
and fusion, and in formation of the nuclear pore complex, around Just like Cer synthases, mammalian ELOVLs exhibit different sub-
which highly curved membranes also exist [27,106]. strate specificities toward acyl-CoAs [43,44,104,126]. ELOVL1 is
expressed ubiquitously among tissues and is important for production
4.2. Generation of very-long-chain fatty acids via fatty acid elongation of C22–C26 saturated and monounsaturated VLCFAs [104,127] (Figs. 4
cycles and 5B). In most tissues, ELOVL1 mainly generates C24 VLCFAs, which
are converted to C24 Cers by the Cer synthase CERS2 [127]. Knockdown
Cellular FAs are derived either from palmitic acid (C16:0-COOH) of CERS2 causes a reduction in this ELOVL1 activity, indicating that
produced by FA synthase (FAS) or from LCFAs taken from food. After ac- CERS2 positively regulates ELOVL1 [104]. On the other hand, ELOVL1
tivation to acyl-CoAs, some of these FAs are converted to VLCFAs via FA is regulated by another Cer synthase, CERS3, in the epidermal stratum
elongation cycles occurring on the cytosolic side of the ER membrane granulosum, where CERS3 but not CERS2 is expressed [127,128].
[43,44,107,108] (Fig. 4). The FA elongation cycle is composed of four CERS3 enables ELOVL1 to produce C26-CoAs beyond C24-CoAs in stra-
steps: condensation, reduction, dehydration, and reduction again [43, tum granulosum [127]. The produced C26-CoAs are either converted
44] (Fig. 5A). The chain-length of acyl-CoA is increased by two in each to C26 Cers or further elongated to ≥ C28-CoAs by ELOVL4. Elovl1 KO
cycle. The first, condensation step is the rate-limiting step of the FA mice have greatly decreased levels of ≥ C26 Cers including acyl-Cers,
elongation cycle, and is catalyzed by FA elongases using a malonyl- and as a result die within one day of birth due to transepidermal
CoA as a two-carbon unit donor [109]. Acyl-CoA is converted to 3- water loss [127] (Fig. 5B).
ketoacyl-CoA by this reaction. The second step is reduction of 3- Although LCFAs are degraded in mitochondria by β-oxidation,
ketoacyl-CoA to 3-hydroxyacyl-CoA, which is catalyzed by 3-ketoacyl- VLCFAs are degraded in peroxisomes [129,130]. The ATP-binding cas-
CoA reductase. The subsequent third step is dehydration of 3- sette (ABC) transporter ABCD1 transports VLC acyl-CoAs to peroxi-
hydroxyacyl-CoA to trans-2-enoyl-CoA by 3-hydroxyacyl-CoA somes for degradation [131]. Mutations in ABCD1 cause X-linked
dehydratases. The final step is reduction/saturation of trans-2-enoyl- adrenoleukodystrophy (X-ALD) accompanied by accumulation of
CoA to an acyl-CoA having two more carbons than the original acyl- lignoceric acid (C24:0-COOH) and cerotic acid (C26:0-COOH) [131,
CoA, and is catalyzed by trans-2-enoyl-CoA reductase. 132]. To date, no therapeutic agent for X-ALD has been successfully de-
The FA elongation cycle and the involved enzymes are conserved veloped. ELOVL1 is one potential pharmacological target, since it is re-
among eukaryotes [43,110,111]. S. cerevisiae and mammals respectively sponsible for the production of these VLCFAs. Indeed, knockdown of
possess three and seven FA elongase genes (yeast, ELO1, FEN1/ELO2, and ELOVL1 in X-ALD fibroblasts reduces the levels of C26:0-COOH [133].
SUR4/ELO3; mammals, ELOVL1–7) (Fig. 5A). The difference in the num- Furthermore, Lorenzo's oil, a 4:1 mixture of glyceryl trioleate and glyc-
ber of the FA elongases seems to reflect the complex FA elongation path- eryl trierucate, inhibits ELOVL1 activity [134] and has been used in an
ways in mammals with respect to the simpler yeast pathways. In experimental treatment of X-ALD [135].
mammals, saturated, monounsaturated, and polyunsaturated (n-3 and
n-6) FAs are all elongated up to C36 [44] (Fig. 4). On the other hand, 4.3.2. ELOVL2 and ELOVL5
yeast contains ≤C26 saturated FAs and essentially two monounsaturat- ELOVL2 and ELOVL5 exhibit activity toward polyunsaturated acyl-
ed FAs—palmitoleic acid (C16:1-COOH) and oleic acid (C18:1- CoAs (ELOVL2, C20- and C22-CoAs; ELOVL5, 18- and 20-CoAs) [42,
COOH)—but no PUFAs [112]. Therefore, only saturated FAs are 104,126,136–139] (Figs. 4 and 5B). The expression of ELOVL2 mRNA is
58 A. Kihara / Progress in Lipid Research 63 (2016) 50–69

Fig. 5. FA elongation cycle and FA elongases. (A) The FA elongation cycle consists of four reactions (condensation, reduction, dehydration, and reduction); mammalian enzymes and related
hereditary disorders are indicated for each reaction. (B) Substrate specificity (1), tissue distribution (2), and phenotypes of the respective FA elongase KO mice (3) are depicted for each
ELOVL family member. S/MU, saturated and monounsaturated; PU, polyunsaturated.

tissue-specific: testis and liver have the highest expression, followed by expression in prostate [104,144]. The involvement of ELOVL7 in prostate
pancreas and placenta [104]. Elovl2 KO mice exhibit male infertility due cancer growth has been reported [144].
to decrease in ULC-PUFAs [140], an essential component of spermato-
genesis as described above. ELOVL5 is expressed ubiquitously among
tissues [104]. Levels of arachidonic acid (C20:4-COOH) and 4.3.4. ELOVL4
docosahexaenoic acid (C22:6-COOH) are reduced in the liver of Elovl5 ELOVL4 is responsible for saturated and unsaturated ULCFA produc-
KO mice, and their reductions cause enhanced expressions of several tion [104,120,126,146–148] (Figs. 4 and 5B). Two types of disorders
lipid-related genes through activation of sterol regulatory element- have been reported to arise from ELOVL4 mutations. One is dominantly
binding protein SREBP-1c, leading to hepatic steatosis [141] (Fig. 5B). inherited macular dystrophy, termed Stargardt disease type 3 (STGD3)
[149], and the other is a recessively inherited neurocutaneous disorder
characterized by ichthyosis, intellectual disability, and spastic quadri-
4.3.3. ELOVL3 and ELOVL7 plegia [150]. Phosphatidylcholines (PCs) containing ULC-PUFAs at the
ELOVL3 and ELOVL7 are active toward saturated and unsaturated sn-1 position exist in retina and brain, whereas saturated and monoun-
C16- to C20-CoAs, with the highest activity toward C18-CoAs [104, saturated ULC-Cers including acyl-Cers exist in epidermis [43,44]. Since
126,142–145] (Figs. 4 and 5B). ELOVL3 mRNA is expressed in restricted these unique ULC lipids play important functions in their respective tis-
tissues/cells such as hair follicles and sebaceous glands in skin, brown sues, their decreases due to ELOVL4 mutation lead to the corresponding,
adipose tissue (BAT), and testis [104,142]. Their expression in BAT is tissue-related disorders. As are other proteins involved in the FA elonga-
greatly heightened upon cold stress [142]. Elovl3 KO mice display a tion cycle, ELOVL4 is localized in the ER, and its localization requires a C-
sparse hair coat and mild skin barrier defects [142] (Fig. 5B). ELOVL7 terminal ER retention signal [151]. STGD3 mutations cause loss of this sig-
mRNA is expressed in a broad range of tissues, with the highest nal, resulting in mislocalization of the truncated ELOVL4 protein
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 59

(ELOVL4ΔC). Moreover, ELOVL4ΔC interacts with wild type ELOVL4, caus- Yeast contains only one 3-hydroxyacyl-CoA dehydratase gene, PHS1
ing its mislocalization as well. This cascading dysfunction is the basis of the [107]. Phs1 is an ER membrane protein with six transmembrane helixes,
dominant transmission pattern of STGD3 [151]. Elovl4 KO mice exhibit and the active site residues (Tyr-149 and Glu-156) reside in transmem-
neonatal lethality due to skin barrier defects [147,152] (Fig. 5B). These brane helix 5 [108,164].
mice lack most acyl-Cers and ≥C28 non-acyl Cers [147,152].
5. Acylceramide and its importance in skin barrier formation
4.3.5. ELOVL6
ELOVL6 exhibits activity toward saturated and monounsaturated 5.1. Skin barrier and structure of epidermis
C12–C16 acyl-CoAs in vitro [104,126,137,138,153] (Figs. 4 and 5B). In
Elovl6 KO mice, C16:0-COOH and C16:1-COOH levels are increased, The skin permeability barrier serves a vital function in land animals
whereas stearic acid (C18:0-COOH) and C18:1-COOH levels are de- by protecting against internal water loss. The skin barrier also impor-
creased [154]. These changes in FA profile cause amelioration of tantly prevents the invasion of pathogens, allergens, and toxic com-
obesity-induced insulin resistance and nonalcoholic steatohepatitis pounds. Accordingly, decreases in skin barrier formation lead to
[154,155] (Fig. 5B). several cutaneous disorders (e.g., ichthyosis, atopic dermatitis, and pso-
riasis), infectious disease, and dry skin [165–168]. Skin is composed of
4.3.6. Fatty acid elongases in yeast the outer epidermis and inner dermis. The skin barrier is primarily
Yeast contains three FA elongases: ELO1, FEN1/ELO2, and SUR4/ELO3. formed in the outermost cell layer of the epidermis stratum corneum,
Elo1 is involved in the elongation of LCFAs [156], whereas Fen1 and Sur4 where intercellular spaces are filled with multilayered lipids (lipid la-
redundantly function in producing VLCFAs [122]. Double deletion of mellae) [165,167,168] (Fig. 6). The high hydrophobicity of the lipid la-
FEN1 and SUR4 (fen1Δ sur4Δ) genes causes lethality [122]. In sur4Δ mellae inhibits internal water loss and invasion of external materials.
cells, C26 VLCFAs are diminished and C24 VLCFAs are concomitantly ac- Mammalian epidermis consists of four cell layers: stratum basale,
cumulated [157]. On the other hand, in fen1Δ cells, overall VLCFA stratum spinosum, stratum granulosum, and stratum corneum [17,
amounts are decreased to ~ 1/3 of levels in wild type cells [157]. Fen1 165] (Fig. 6). Approximately 95% of epidermal cells are keratinocytes.
and Sur4 are ER-resident, integral membrane proteins with seven trans- Keratinocytes proliferate in stratum basale and migrate outward while
membrane helixes [107]. The positions of the Lys residues in transmem- differentiating into cell layer-specific cells. Stratum granulosum is char-
brane helix 6 of Fen1 and Sur4 determine the chain-lengths of their acterized by the presence of keratohyalin granules containing
products (C24 for Fen1 and C26 for Sur4) [107]. profilaggrin, the precursor of filaggrin. Mutations in the filaggrin gene
(FLG) cause ichthyosis vulgaris [166,169], and they are also frequently
4.4. 3-Ketoacyl-CoA reductase and trans-2-enoyl-CoA reductase found in atopic dermatitis patients [170,171]. Filaggrin is a keratin inter-
mediate filament-associated protein, and its degradation product acts as
Two reductases, the 3-ketoacyl-CoA reductase KAR and trans-2- a natural moisturizing factor [171].
enoyl-CoA reductase TECR, catalyze the second and fourth steps of the In the upper layers of epidermis, cornified envelope (CE) forms on
mammalian FA elongation cycle, respectively, using NADPH as a reduc- the periphery of keratinocytes (Fig. 6). CE is important for mechanical
ing agent [114] (Fig. 5A). A point mutation in TECR where Pro-182 is and permeability barriers, and is produced through crosslinking of pro-
substituted with Leu in the gene product (TECR P182L) causes nonsyn- teins such as involucrin, envoplakin, periplakin, loricrin, and small
dromic mental retardation [158]. This mutation reduces the activity and proline-rich proteins by transglutaminases beneath the plasma mem-
stability of TECR, thereby impairing VLCFA synthesis and, in turn, de- brane [172,173]. During the cornification process, the lipid bilayer of
creasing VLC-sphingolipid levels [118]. However, TECR P182L is a rela- the plasma membrane is replaced with a monolayer of protein-bound
tively weak mutation, and the corresponding decrease in VLC- Cers, in which ω-OH Cers are crosslinked with CE proteins such as
sphingolipid levels is small [118]. Since TECR is solely responsible for involucrin, envoplakin, and periplakin [172,174,175] (Fig. 6). The mem-
the fourth step of the FA elongation cycle, its complete functional distur- brane structure containing protein-bound Cers is called the corneocyte
bance should cause complete loss of VLCFAs and embryonic lethality. lipid envelope (CLE), and functions to connect corneocytes with lipid
Considering that only neural function is affected by the weak mutation lamellae.
TECR P182L, the nervous system may require VLCFAs more dearly than
any other tissues. The importance of VLC-sphingolipids in myelin main- 5.2. Formation of lipid lamellae in stratum corneum
tenance and function described above may be related to this
requirement. Lipid lamellae are mainly composed of Cers (~ 50%), cholesterol
(~25%), and free FAs (~15%) [31,168]. The major free FAs of human ab-
4.5. 3-Hydroxyacyl-CoA dehydratases domen stratum corneum are C16:0-COOH (36.8%), followed by C18:1-
COOH (33.1%) and C18:2-COOH (12.5%) [176]. The precursor lipids of
Mammals contain four 3-hydroxyacyl-CoA dehydratase genes: lipid lamellae are stored in intracellular lamellar bodies, which exist in
HACD1/PTPLA, HACD2/PTPLB, HACD3/PTPLAD1, and HACD4/PTPLAD2 the upper layers of stratum spinosum and in stratum granulosum. The
[113] (Fig. 5A). Among them, HACD2 mRNA is expressed ubiquitously ABC transporter ABCA12 transports the lipids into lamellar bodies
and seems to be the major 3-hydroxyacyl-CoA dehydratase in mammals [177]; its gene, ABCA12, is an ARCI-causative gene [166,167,178]. ARCI
[159]. HACD1 mRNA is expressed only in heart and skeletal muscle is classified into harlequin ichthyosis, lamellar ichthyosis, and congeni-
[160]. Mutations in HACD1 are associated with recessive congenital my- tal ichthyosiform erythroderma [179]. Mutations in the ABCA12 gene
opathy characterized by early-onset muscle weakness in human and can cause harlequin ichthyosis, which is accompanied by the most se-
dogs [161,162]. Hacd1 KO mice also display a myopathic phenotype vere barrier defect among ARCIs [178].
with reduced muscle weight and muscle fiber growth [163]. HACD1 At the interface of stratum granulosum and stratum corneum, lamel-
functions in myoblast fusion during muscle development and regenera- lar bodies are fused with the plasma membrane [165] (Fig. 6). The
tion [163]. HACD3 mRNA is expressed in a broad range of tissues with contained lipids are released to form lipid lamellae. The major precur-
high expression in brain, liver, kidney, and placenta [113]. The expres- sors for Cers in lipid lamellae are GlcCers; the minor precursors are
sion of HACD4 mRNA is restricted to leukocytes [113]. To date, substrate SMs. These are respectively converted to Cers by β-glucocerebrosidase
specificities of HACDs toward 3-hydroxyacyl-CoA substrates with dif- and sphingomyelinase upon release into the extracellular spaces [30,
ferent chain-lengths and double bond numbers have not yet been eluci- 31,168]. Mutations in the β-glucocerebrosidase gene (GBA) cause
dated. Furthermore, the functions of HACD3 and HACD4 are unclear. Gaucher disease, one of the sphingolipid storage diseases
60 A. Kihara / Progress in Lipid Research 63 (2016) 50–69

Fig. 6. Generation of acyl-Cer and protein-bound Cer in epidermis. The structure of epidermis is illustrated as well as the enzymes and reactions responsible for creating acyl-Cer and
protein-bound Cer. SB, stratum basale; SS, stratum spinosum; SG, stratum granulosum; SC, stratum corneum; G, glucose.

(sphingolipidoses) [180]. Gaucher patients and GBA KO mice alike dis- stratum corneum Cer classes [187], as well as shortened Cer chain-
play ichthyosis phenotype due to failure of GlcCer-to-Cer conversion lengths [167,187,188]. Changes in Cer composition are also observed
in stratum corneum [181,182]. in patients afflicted with psoriasis [189], which is an immuno-
inflammatory disease characterized by red, itchy, and scaly patches
5.3. Ceramides in epidermis and dryness. EOS, NP, and AP levels are decreased in psoriatic scales
[189].
Because of the tissue's specialized role in skin barrier formation, Cers
in epidermis are much more abundant and diverse than in other tissues 5.4. Acylceramide synthetic pathway
[17,30,31,167]. The composition of Cer classes in human stratum
corneum is NP (22.1%), NH (14.5%), AH (10.8%), NDS (9.8%), AS Acyl-Cers are unique in their structures. Although normal Cers con-
(9.6%), AP (8.8%), NS (7.4%), EOS (6.5%), EOH (4.3%), ADS (1.6%), EOP tain two hydrophobic chains (an LCB and a FA), acyl-Cers have three hy-
(1.1%), and EODS (0.4%) [31,38]. Of these, acyl-Cers (EOS, EOH, EOP, drophobic chains (an LCB, an ω-OH ULCFA with C30–36, and C18:2-
and EODS) account for ~ 12% of stratum corneum Cers. The chain- COOH) (Fig. 6). Therefore, production of acyl-Cers requires more en-
lengths of the FA portions of non-acyl-Cers in human stratum corneum zymes than does that of normal Cers. ULC acyl-CoAs are generated by
are mostly C24–C30, whereas those of acyl-Cers are C30–C36 [46]. Inter- sequential actions of the FA elongases ELOVL1 (up to C26) and
estingly, levels of Cers containing odd-numbered FAs are especially high ELOVL4 (≥ C28) [104,127,147] (Fig. 6). After removal of CoAs, the
(~30% of total Cers) in stratum corneum [46]. resulting ULCFAs are ω-hydroxylated by the cytochrome P450 member
Acyl-Cers are epidermis-specific and are quite important for skin CYP4F22, generating ω-OH ULCFAs [190]. CYP4F22 is a type I ER mem-
barrier formation [17,30,31,167]. Accordingly, mutations in the genes brane protein, and its active site is exposed to the cytosolic side of the ER
involved in acyl-Cer production such as CERS3, ELOVL4, and CYP4F22 membrane [190]. CYP4F22 is an ARCI-causative gene [183,184]: al-
cause ichthyosis symptoms with severe barrier defects in human pa- though such patients are rare, acyl-Cers were almost absent in the one
tients and mouse models [87,89,147,150,152,183,184]. Atopic dermati- CYP4F22-mutant ARCI patient examined to date [190]. After conversion
tis is also accompanied by mild skin barrier defects. In atopic dermatitis of ω-OH ULCFAs to ω-OH ULC acyl-CoAs by acyl-CoA synthetases
patients, total Cer levels in skin are reduced to ~2/3 of those in healthy (ACSs), the generated ω-OH ULC acyl-CoAs are used for ω-OH Cer syn-
controls [185], and Cer compositions are altered [185–187]. Atopic der- thesis by the Cer synthase CERS3 [87]. The de novo synthesized dihydro-
matitis patients exhibit decreased EOS, EOH, EOP NH, and NP among the Sph appears to be the major LCB donor for ω-OH Cer synthesis, but other
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 61

LCBs generated from the salvage pathways can be used. The LCB part 6. Ceramide degradation pathways
of ω-OH Cers containing dihydro-Sph is converted to other types of
LCBs (Sph by DEGS1, phyto-Sph by DEGS2, or to 6-OH Sph by an un- 6.1. Ceramide degradation pathways
known enzyme). A scaly skin phenotype is observed in Degs1 KO
mice [65], indicating the importance of Sph-based Cers/acyl-Cers in In sphingolipid biosynthetic pathways, Cer is converted to the com-
skin barrier formation. Finally, an ester linkage is formed between plex sphingolipids SM and glycosphingolipids. Alternatively, Cer can be
C18:2-COOH and the ω-OH group of ω-OH Cer to create an acyl- converted to Cer 1-phosphate by Cer kinase [206]. Cer 1-phosphate is
Cer; the acyltransferase or transacylase responsible has yet to be involved in several cellular processes including phagolysosome forma-
identified. C18:2-COOH seems to be supplied from triglycerides tion, mast cell degranulation, and inflammation [6,207].
[191], since KO mice for the diacylglycerol acyltransferase Dgat2, In their degradation pathways, Cers are first converted to LCBs and
which is involved in triglyceride synthesis, exhibit severe skin barri- FAs by ceramidases [6,208]. The released FAs can be used for lipid syn-
er defects [192]. thesis or energy production via β-oxidation. On the other hand, the re-
All of the enzymes involved in acyl-Cer production (except for the leased LCBs are exclusively recycled back to sphingolipids, since they
unidentified acyltransferase/transacylase) are localized in the ER, indi- are used only as components of sphingolipids. Therefore, LCBs must be
cating that acyl-Cer production takes place there [190]. Produced acyl- converted to other metabolizable compounds in order for homeostasis
Cers are then converted to acyl-GlcCers in the cis-Golgi by the UDP- to be maintained in terms of the concentrations of LCBs and
glucose Cer glucosyltransferase UGCG [193,194], followed by transport sphingolipids. Eukaryotic cells share metabolic pathways for converting
into lamellar bodies by the ABC transporter ABCA12 [31,177]. LCBs to acyl-CoAs via LCB 1-phosphates (LCBPs), fatty aldehydes, and
Epidermis-specific Ugcg KO mice display severe skin barrier defects FAs [5,209,210] (Fig. 7A). The generated acyl-CoAs are then used as
[195]. In these mice, levels of GlcCers/acyl-GlcCers are reduced, but in- the FA components of lipids (mainly ester-linked glycerophospholipids
stead of the acyl-GlcCers disappearing, unusual acyl-SMs are produced. and partly ether-linked glycerophospholipids, triglycerides, and
The amounts of acyl-Cer EOS and protein-bound Cers are unchanged. sphingolipids) or degraded to CO2 via β-oxidation [5,125,209–212]
Acyl-SMs cannot substitute for acyl-GlcCers in lamellar body formation: (Fig. 7B). C16:0-CoA is generated from Sph and dihydro-Sph, whereas
abnormal arrangements of lamellae were observed in the lamellar bod- odd-numbered C15:0-CoA is created from phyto-Sph through Cer deg-
ies of Ugcg KO mice [195]. Finally, acyl-GlcCers in lamellar bodies are re- radation pathways [209,213,214] (Fig. 7A). Some of the produced acyl-
leased into extracellular spaces at the interface of stratum granulosum CoAs are elongated or desaturated before being incorporating into lipids
and stratum corneum and are reverted to acyl-Cers by β- [211–214].
glucocerebrosidase [30,168] (Fig. 6). As described above, most LCBs are either recycled to sphingolipids or
metabolized to glycerophospholipids. What percentages of LCBs under-
go the latter pathway varies among tissues. HeLa cells, PC12 (neuronal)
5.5. Protein-bound ceramide synthesis cells, HepG2 hepatocytes, and IEC-6 intestinal epithelial cells were la-
beled with [3H]Sph, and 23–42% of it was found to be metabolized to
Protein-bound Cers, components of the CLE, are produced from acyl- ester-linked glycerophospholipids (HeLa, 23%; PC12, 32%; HepG2, 21%;
Cers [175,196]. Thus, acyl-Cers are important not only as lipid lamella and IEC-6, 42%) [125]. In addition, a trace amount was metabolized to
components but also as precursors of protein-bound Cers for skin barri- ether-linked glycerophospholipids in PC12 and HepG2 cells [125].
er formation. The Cer moieties of protein-bound Cers are OS (ω-OH
acyl-Sph derived from EOS), OH (ω-OH acyl-6-OH Sph derived from
6.2. Ceramide degradation by ceramidases
EOH), or OP (ω-OH acyl-phyto-Sph derived from EOP); their respective
percentages in stratum corneum are 54%, 31%, and 15% [31,38].
The first step of Cer degradation pathways—i.e., cleavage of Cer to a
In the course of protein-bound Cer production, the C18:2-COOH por-
FA and an LCB—is catalyzed by ceramidases (Fig. 7A). Mammals contain
tions of acyl-Cers (or acyl-GlcCers) are subjected to peroxidation and
five ceramidases having different optimal pH: the acid ceramidase
subsequent epoxyalcohol derivatization by successive actions of two
ASAH1/AC, the neutral ceramidase ASAH2/NC, and the alkaline
lipoxygenases (the 12R-lipoxygenase ALOX12B and the epidermal
ceramidases 1–3 (ACER1, ACER2, and ACER3) [208]. Among them, the
lipoxygenase-3 ALOXE3) [196,197] (Fig. 6). Both ALOX12B and ALOXE3
lysosome-localized ASAH1 is the physiologically most important. Muta-
are ARCI-causative genes [198]. Furthermore, severe skin barrier defect
tions in ASAH1 cause the sphingolipid storage disease Farber disease,
phenotypes are observed in Alox12b and Aloxe3 KO mice [199–201].
characterized by deformed joints, progressive hoarseness, subcutane-
These results indicate the pivotal role of the CLE in skin barrier forma-
ous nodules, and neurologic deterioration [215]. Disruption of mouse
tion. The oxygenation by the two lipoxygenases seems to act as a signal
Asah1 causes early embryonic lethality [216].
for subsequent esterase-catalyzed hydrolysis of the oxidized C18:2-
COOH, generating ω-OH Cers. The exposed ω-OH group may be then
crosslinked with CE proteins such as involucrin, envoplakin, and 6.3. Long-chain base 1-phosphate metabolic enzymes
periplakin, producing the CLE [174,175]. In vitro experiments initially
suggested that this reaction is catalyzed by the transglutaminase 6.3.1. Sphingosine kinases
transglutaminase 1 (TGM1), which is known to function in forming In Cer degradation pathways, LCBs are converted to LCBPs: S1P from
the CE (via isopeptide bond formation among CE proteins) [202]. How- Sph, dihydro-Sph 1-phosphate (dihydro-S1P) from dihydro-Sph, and
ever, later study showed that ARCI (lamellar ichthyosis) patients with phyto-Sph 1-phosphate (phyto-S1P) from phyto-Sph (Fig. 7A). The
absent or low TGM1 activity/protein levels had normal levels of two redundant sphingosine kinases SPHK1 and SPHK2 catalyze the pro-
protein-bound Cers [203]. Therefore, protein-bound Cers may be pro- duction of LCBPs in mammals [217,218]. Although KO mice having sin-
duced by other enzymes or via another mechanism. The removal of glu- gle gene deletion of Sphk exhibit no obvious phenotype, Sphk1 Sphk2
cose from acyl-GlcCers by β-glucocerebrosidase may occur after the double KO mice are embryonic lethal due to impaired neurogenesis
creation of protein-bound GlcCers, since protein-bound GlcCers are in- and angiogenesis [219]. SPHK1 has higher substrate specificity than
creased in β-glucocerebrosidase-deficient type 2 Gaucher mice and in SPHK2; it exhibits activity toward Sph and dihydro-Sph, whereas
KO mice lacking the gene encoding the activator protein for β- SPHK2 is active toward not only all LCBs but also the LCB analog
glucocerebrosidase (saposin-C) [204,205]. However, it is entirely possi- fingolimod/FTY720 [217,218,220], which is therapeutic for multiple
ble that the order of the crosslinking with respect to the glucose remov- sclerosis [34,35]. Fingolimod is activated by phosphorylation, then acts
al is not strict. by binding to S1P receptors, mainly S1P1 [16,34,35].
62 A. Kihara / Progress in Lipid Research 63 (2016) 50–69

Fig. 7. Cer degradation pathways. (A) Pathways, reactions, and enzymes involved in the degradation of dihydro-Cer, Cer (Sph), and phyto-Cer in yeast and mammals. (B) Overall flow of
Cer degradation pathways leading to other classes of lipids. In Cer degradation pathways, LCBs are metabolized mainly to ester-linked glycerophospholipids but partly to other lipids such
as triglyceride (TG), sphingolipids, and ether-linked glycerophospholipids or CO2. In SLS patients, the conversion of the fatty aldehydes to FAs is impaired. Accordingly, metabolism of LCBs
to ester-linked glycerophospholipids is abrogated, and instead that to ether-linked glycerophospholipids becomes a major metabolic route. F-Ald, fatty aldehyde; F-Alc, fatty alcohol.

S1P is a well-known lipid mediator in extracellular fluid (such as generated Sph is transported to the plasma membrane or other mem-
plasma), whereas in cells it is a metabolic intermediate of sphingolipids branes, whereupon SPHKs phosphorylate it.
[6,209,210]. Only limited cell types secrete S1P extracellularly: the
major sources of plasma S1P are erythrocytes and vascular endothelial
cells [16,221–223]. Fingolimod phosphate is mainly generated by 6.3.2. Long-chain base 1-phosphate-degrading enzymes
SPHK2 in platelets before being secreted into plasma [16,224]. On the LCBPs are either dephosphorylated to LCBs by S1P phosphatases or
other hand, sphingolipid metabolism via LCBPs occurs in all tissues ex- cleaved to fatty aldehydes [trans-2-hexadecenal (tC16:1-CHO) from
cept for platelets and erythrocytes, which lack S1P lyase [210,221,225, S1P, hexadecanal (C16:0-CHO) from dihydro-S1P, and 2-
226]. Accordingly, SPHKs are expressed in all tissues [227]. hydroxyhexadecanal (2-OH C16:0-CHO) from phyto-S1P] and
SPHK1 is localized intracellularly mainly in the cytosol and partly in phosphoethanolamine by S1P lyase [6,210] (Fig. 7A).
the plasma membrane [228–232]. SPHK2 is also primarily localized in Phosphoethanolamine is metabolized to phosphatidylethanolamine
cytosol, but incidence in the plasma membrane, internal membranes, via CDP-ethanolamine [211]. Two S1P phosphatases (SGPP1/SPP1 and
and nucleus has also been reported [230,231]. Although SPHK1 and SGPP2/SPP2) and one S1P lyase (SGPL/SPL) have been identified in
SPHK2 are mostly located in the cytosol, it is membranes where they mammals [233–235]. All of these S1P-degrading enzymes are localized
must function, given that their substrate LCBs are too hydrophobic to in the ER [225,235,236]. SGPP1 and SPL are expressed ubiquitously,
freely diffuse into the cytosol. Since the lysosomal acid ceramidase whereas expression of SGPP2 is limited to certain tissues such as kidney
Asah1 is the major ceramidase as described above, Sph seems to be and heart [225,235,237]. Sgpp1 KO mice exhibit focal desquamation and
mainly generated in lysosomes. However, the site of S1P production morphological aberration in epidermis, and most die before weaning
still remains unclear. It is possible that cytosolic SPHKs are transiently [238]. Moreover, S1P levels are increased in their keratinocytes, and
recruited to lysosomes for S1P production, or that lysosomally their keratinocyte differentiation is accelerated [238].
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 63

The S1P lyase-catalyzed reaction is irreversible, and is the rate- 6.4.2. Sjögren-Larsson syndrome
limiting step of Cer degradation pathways. Generally, biomolecules ALDH3A2 is a causative gene of the neurocutaneous disorder
must be kept in narrow ranges of concentrations; otherwise cellular Sjögren-Larsson syndrome (SLS), which is characterized by mental re-
functions can become impaired, leading to disorders. Therefore, the bal- tardation, spastic di- or tetraplegia, and ichthyosis [253]. Aldehyde mol-
ance between synthesis and degradation must always be properly ecules are reactive, such as by forming Schiff bases with primary amines,
maintained. This is no less true of Cer degradation pathways. Their and thus toxic to cells. Therefore, the SLS pathology is considered to be
blockage by the disruption of Sgpl1 in mice causes accumulation of caused by accumulated fatty aldehydes reacting with certain important
S1P as well as increases in Cer and SM levels [239]. Sgpl1 KO mice exhibit proteins in epidermis and the nervous system. Several metabolic path-
several phenotypes, such as lymphopenia, thymus atrophy, myeloid cell ways can also generate the substrates of ALDH3A2, fatty aldehydes,
hyperplasia, disturbance of lipid homeostasis in liver, lesions in lung, other than Cer degradation pathways, such as those of leukotriene B4,
heart, urinary tract, and bone, and early mortality (20–50 days after diet-derived phytol, plasmalogens, and fatty alcohols [254]. It is still un-
birth) [239–242]. clear which fatty aldehyde or which metabolic pathway is the main con-
tributor to the pathogenesis of SLS. However, Cer degradation pathways
are a likely candidate. Cer degradation is ubiquitous and occurs actively,
6.4. The aldehyde dehydrogenase ALDH3A2 and Sjögren-Larsson syndrome suggesting that tC16:1-CHO is generated constantly at a relatively high
level. Furthermore, the site of tC16:1-CHO generation via Cer degrada-
6.4.1. Aldehyde dehydrogenases tion and that of ALDH3A2 localization are both the ER [225,248]. The
Fatty aldehydes produced by S1P lyase are converted to FAs [C16:0- Sph metabolite tC16:1-CHO is an α,β-unsaturated aldehyde and hence
COOH from C16:0-CHO, trans-2-hexadecenoic acid (tC16:1-COOH) a particularly reactive fatty aldehyde. Although normal aldehydes can
from tC16:1-CHO, and 2-hydroxypalmitic acid (2-OH C16:0-COOH) only react with primary amines to form Schiff bases, α,β-unsaturated al-
from 2-OH C16:0-CHO] by aldehyde dehydrogenases (ALDHs). Multiple dehydes can react with general nucleophiles such as lysine, cysteine,
ALDHs exist in mammals (19 in human and 21 in mouse), divided into and histidine side-chains via 1,4-Michael addition [255].
eleven subfamilies (ALDH1–9, −16, and −18) [243,244]. Of these, the
ALDH3 subfamily is responsible for the oxidation of fatty aldehydes. 6.5. Acyl-CoA synthetases involved in ceramide degradation pathways
Humans possess four ALDH3 subfamily genes [ALDH3A1, ALDH3A2,
ALDH3B1, and (the pseudogene) ALDH3B2], whereas the mouse genome Conversion to acyl-CoAs by ACSs is necessary for FAs to be incorpo-
has five Aldh3 genes (Aldh3a1, Aldh3a2, Aldh3b1, Aldh3b2, and Aldh3b3) rated into lipids, degraded via β-oxidation, or elongated via the FA elon-
[243,244]. ALDH3A1/Aldh3a1 is a cytosolic protein highly expressed in gation cycle [256]. Therefore, the FAs generated in Cer degradation
cornea and exhibits high activity toward MC aldehydes [245,246]. pathways must also be activated for further metabolism. ACSs are divid-
ALDH3A2/Aldh3a2, ALDH3B1/Aldh3b1, Aldh3b2, and Aldh3b3 prefer- ed into six subfamilies [ACS short-chain (ACSS), ACS medium-chain
entially oxidize LC aldehydes, although they show weak activities to- (ACSM), ACS long-chain (ACSL), ACS very-long-chain (ACSVL), ACS
ward MC aldehydes [246,247]. Among them, only ALDH3A2/Aldh3a2 bubblegum (ACSBG), and ACS family (ACSF)], depending on their sub-
is localized in the ER [248], where LCBP-to-glycerophospholipid conver- strate specificities and sequence similarities [257]. The human genome
sion occurs. In contrast, ALDH3B1/Aldh3b1 and Aldh3b3 are localized in contains 26 ACS genes (ACSS1–3, ACSM1–5, ACSL1, 3–6, ACSVL1–6,
the plasma membrane, and Aldh3b2 in lipid droplets [246,247]. Accord- ACSBG1, 2, and ACSF1–4) [257]. Among them, eight ACSs (ACSL1, 3, 4,
ingly, ALDH3A2/Aldh3a2 is the major ALDH involved in Cer degradation 5, 6, ACSVL1, 4, and ACSBG1), which exhibit activity toward LCFAs
pathways [213]. In one of our previous studies, Aldh3a2-null CHO-K1 [256], are involved in Cer degradation pathways [213,258].
cells were labeled with [3H]Sph or [3H]dihydro-Sph, and the metabo-
lism of Sph/dihydro-Sph to ester-linked glycerophospholipids was 6.6. Dual functions of the trans-2-enoyl-CoA reductase TECR
largely impaired [213]. Instead, a fraction of unmetabolized fatty alde-
hydes were reduced to fatty alcohols and incorporated into ether- Dihydro-Sph is metabolized to C16:0-CoA via four reactions (phos-
linked glycerophospholipids. phorylation, cleavage, oxidation, and CoA addition) as described above
Ether-linked glycerophospholipids include plasmanyl (1-alkyl-2- (Fig. 7A). Since Sph contains a trans-double bond, it needs an additional
acyl) and plasmenyl (1-alkenyl-2-acyl) types, the latter of which are saturation step for conversion to C16:0-CoA. After creation of trans-2-
collectively called plasmalogens (Fig. 7B). The polar head groups of hexadecenoyl-CoA (tC16:1-CoA) by ACSs from the ALDH product
ether-linked glycerophospholipids are exclusively tC16:1-COOH, the resulting tC16:1-CoA is saturated to C16:0-CoA by
phosphoethanolamine (plasmanyl/plasmenylethanolamine) and the trans-2-enoyl-CoA reductase TECR [125] (Fig. 7A). As mentioned
phosphocholine (plasmanyl/plasmenylcholine) [249]. Plasmalogens ac- above, TECR is also involved in the fourth step of the FA elongation
count for ~18% of total human phospholipids and are highly abundant in cycle. Thus, TECR functions in two lipid metabolic pathways: Cer degra-
erythrocytes, kidney, lung, testis, skeletal muscle, and myelin [249]. dation pathways and FA elongation cycle.
They play several physiological functions including removal of oxidants,
cell–cell or cell-extracellular matrix interactions, lipid microdomain for- 6.7. Metabolism of phytosphingosine to odd-numbered fatty acids
mation, intracellular cholesterol homeostasis, spermatogenesis, and
myelination [250]. In the ether-linked glycerophospholipid biosynthetic Phyto-Sph has an extra hydroxyl bond at the C4 position compared
pathway, 1-acyl-dihydroxyacetone phosphate is first created from acyl- to dihydro-Sph. The hydroxyl bond at C4 in phyto-Sph is shifted to the
CoA and dihydroxyacetone phosphate, and then the acyl-chain of 1- C2 position by removal of a two-carbon unit by S1P lyase: the product
acyl-dihydroxyacetone phosphate is replaced with a fatty alcohol by is 2-OH C16:0-CHO [214] (Fig. 7A). Existence of this hydroxyl bond
alkyl dihydroxyacetone phosphate synthase [249]. Fatty alcohols are makes the metabolic pathway of phyto-Sph different from those of
mainly generated from acyl-CoAs by fatty acyl-CoA reductases [249, dihydro-Sph and Sph. Although dihydro-Sph and Sph are metabolized
251]. Although most fatty aldehydes generated through ceramide deg- to glycerophospholipids as C16:0-COOH or its derivatives (C18:0-
radation pathways are converted to FAs by ALDHs, a small amount of COOH and C16:1-COOH), phyto-Sph is metabolized to
them is converted to fatty alcohols and incorporated into ether-linked glycerophospholipids as pentadecanoic acid (C15:0-COOH) or its deriv-
glycerophospholipids, even in ALDH3A2-active cells [125,209,252] atives [heptadecanoic acid (C17:0-COOH) and pentadecenoic acid
(Fig. 7B). However, the contribution of ceramide degradation pathways (C15:1-COOH)] [209,214]. After phyto-S1P is cleaved to 2-OH C16:0-
to the total fatty alcohol generation for ether-linked CHO by S1P lyase, it is oxidized to 2-OH C16:0-COOH by ALDHs. The
glycerophospholipid synthesis seems to be low. resulting 2-OH C16:0-COOH [or its acyl-CoA derivative 2-
64 A. Kihara / Progress in Lipid Research 63 (2016) 50–69

hexadecanoyl-CoA (2-OH C16:0-CoA)] is subjected to α-oxidation, cre- the levels of cerebrosides and sulfatides containing C23:0-COOH are es-
ating C15:0-COOH [214]. pecially high, reflecting the abundance of sphingolipids containing
Cer degradation pathways are conserved among eukaryotes includ- cerebronic acid (2-OH C24:0-COOH) in myelin [49].
ing yeast, in which dihydro-Sph and phyto-Sph are two major LCBs. The The phyto-Sph metabolic pathway is not the sole pathway that gen-
yeast S. cerevisiae contains two alkaline ceramidases, Ydc1 and Ypc1 erates 2-OH FAs. Rather, most 2-OH FAs are generated via direct hydrox-
[259,260]. Ypc1 exhibits high activity toward dihydro-Cer and weak ac- ylation of FAs by the FA 2-hydroxylase FA2H [263,268]. A large
tivity toward phyto-Cer [259,260]. On the other hand, Ydc1 is highly proportion of GalCers and sulfatides contains 2-OH FAs (i.e., contains
specific to dihydro-Cer [260]. As in mammals, in yeast the produced AS type Cers) in brain [49,50,52]. The 2-OH group seems important for
dihydro-Sph is sequentially metabolized to dihydro-S1P (by LCB kinase GalCers and sulfatides to exert their functions in myelin, perhaps en-
Lcb4), C16:0-CHO (by LCBP lyase Dpl1), C16:0-COOH (by ALDH Hfd1), hancing lipid-lipid interactions via hydrogen bonds and thereby stabi-
and C16:0-CoA (by ACSs Faa1 and Faa4), followed by incorporation lizing the myelin sheath. Accordingly, FA2H mutations in humans
into glycerophospholipids [213] (Fig. 7A). Although Sph is not a natural cause hereditary spastic paraplegia [269,270], and Fa2h KO mice exhibit
LCB in yeast, exogenously added Sph can be metabolized to both late-onset axon and myelin sheath degeneration [52,271].
sphingolipids and glycerophospholipids [261]. In the latter metabolic Epidermis contains high amounts of 2-OH FA-containing Cers:
pathway, again as in mammals, Sph is metabolized to C16:0-CoA by i.e., AS, AP, AH, and ADS types of Cers. Disruption of Fa2h in mice, how-
the above enzymes plus the trans-2-enoyl-CoA reductase Tsc13 (i.e., a ever, does not cause changes in levels of 2-OH FA-containing Cers in epi-
homolog of TECR), followed by incorporation into glycerophospholipids dermis [272]. Expression of Fa2h in skin is restricted to the sebaceous
[125,213]. glands, and Fa2h KO mice exhibit delayed fur development and cycle al-
Phyto-Sph is metabolized to C15:0-COOH (or its derivative C17:0- opecia [272]. Therefore, another, unknown FA 2-hydroxylase must exist
COOH) in yeast as in mammals [214]. In yeast mutants defective in in epidermis.
phyto-Sph production (i.e., by deletion of the dihydro-Sph C4-
hydroxylase gene SUR2) or metabolism (i.e., by deletion of the LCBP 7. Concluding remarks
lyase gene DPL1), the levels of odd-numbered PCs are reduced to
~ 40% of those in wild type cells [214]. Thus, the phyto-Sph metabolic Cers are highly diverse molecules, with recently developed high-
pathway is the major source of cellular odd-numbered FAs in yeast. Al- sensitivity mass spectrometry detecting over 1000 species [38]. Most
though the details of the α-oxidation reaction remain unclear, the novel of the genes involved in the synthesis, degradation, and modification
yeast gene MPO1 has been implicated in the α-oxidation step of the of Cers have been identified in the last 15 years. Through analyses of
phyto-Sph metabolic pathway [214]. Mpo1 is an ER protein [214], indi- these genes, such as screenings for mutations in hereditary diseases, ex-
cating that the α-oxidation step takes place in the ER in yeast. Mpo1 be- aminations of changes in their expression or activity patterns in meta-
longs to the large protein family DUF962 (DUF: domain of unknown bolic diseases and cancer, and phenotypic analyses using KO mice,
function), whose function had been unknown until MPO1 was identi- vast knowledge about the physiological and pathological importance
fied. DUF962 family members exist in bacteria, fungi, plants, and pro- of Cers and sphingolipids has been accumulated. We now realize that
tists, whereas mammals contain none, suggesting that α-oxidation even small differences between Cer species—i.e., in chain-length, hy-
pathways differ in yeast and mammals. In mammals, the 2- droxylation status, and double bond number—bear great significance
hydroxyacyl-CoA lyase HACL1, which is localized to the peroxisomes, on how they fulfill their species-specific functions. These findings can
was shown to exhibit activity to convert 2-OH octadecanoyl-CoA (2- lead to clinical applications in the future, through the development of
OH C18:0-CoA) to heptadecanol (C17:0-CHO) in vitro [262]. Therefore, inhibitors or activators of these Cer-related gene products.
it is possible that 2-OH C16:0-COOH derived from phyto-Sph is sequen-
tially metabolized to 2-OH C16:0-CoA by ACSs, to C15:0-CHO by HACL1,
Acknowledgments
and to C15:0-COOH by ALDHs in mammals.
The purpose of phyto-Sph metabolism to odd-numbered FAs may
This work was supported by funding from Advanced Research and
not be to generate odd-numbered FAs per se, but to convert 2-OH FAs,
Development Programs for Medical Innovation (AMED-CREST) from
which are difficult to metabolize, to easily metabolizable non-hydroxy
the Japan Agency for Medical Research and Development (AMED), by
FAs. Only sphingolipids contain 2-OH FAs, and 2-OH FA-containing
funding from Creation of Innovation Centers for Advanced Interdisci-
sphingolipids exist in restricted tissues such as stomach, intestine,
plinary Research Areas Program from the Ministry of Education, Culture,
brain, kidney, and epidermis [38,48,263]. Therefore, unless they are
Sports, Science and Technology of Japan, and by a Grant-in-Aid for Sci-
first converted to other metabolizable compounds, the metabolic fate
entific Research (A) 26251010 from the Japan Society for the Promotion
of 2-OH FAs is limited to conversion to sphingolipids. On the other
of Science (JSPS).
hand, once 2-OH FAs are metabolized to odd-numbered FAs, they can
be metabolized to several lipids and be degraded via β-oxidation as
even-numbered FAs. Although the β-oxidation products of even- References
numbered FAs are always acetyl-CoAs, β-oxidation of odd-numbered [1] Yamamoto A, Yano I, Masui M, Yabuuchi E. Isolation of a novel sphingoglycolipid
FAs generates acetyl-CoAs and propionyl-CoA, the latter of which can containing glucuronic acid and 2-hydroxy fatty acid from Flavobacterium devorans
be further metabolized to succinyl-CoA for breakdown in the citric ATCC 10829. J Biochem 1978;83:1213–6.
[2] Dickson RC, Sumanasekera C, Lester RL. Functions and metabolism of sphingolipids
acid cycle [264]. in Saccharomyces cerevisiae. Prog Lipid Res 2006;45:447–65.
[3] Vacaru AM, van den Dikkenberg J, Ternes P, Holthuis JC. Ceramide
6.8. Generation of 2-hydroxy fatty acids phosphoethanolamine biosynthesis in Drosophila is mediated by a unique ethanol-
amine phosphotransferase in the Golgi lumen. J Biol Chem 2013;288:11520–30.
[4] Markham JE, Lynch DV, Napier JA, Dunn TM, Cahoon EB. Plant sphingolipids: func-
Although most cellular FAs are even-numbered FAs, the existence of tion follows form. Curr Opin Plant Biol 2013;16:350–7.
odd-numbered FAs has long been known. Their levels are quite low in [5] Kihara A. Sphingolipid metabolism via sphingosine 1-phosphate. In: Yokomizo T,
Murakami M, editors. Bioactive lipid mediators: current reviews and protocols.
most tissues (b1% of those of even-numbered FAs) [265]. However, in
Tokyo: Springer Japan; 2015. p. 127–38.
specific tissues such as brain and epidermis, odd-numbered FA levels [6] Kihara A, Mitsutake S, Mizutani Y, Igarashi Y. Metabolism and biological functions
are unusually high [46,49]. At first, a de novo FA synthetic pathway of two phosphorylated sphingolipids, sphingosine 1-phosphate and ceramide 1-
that used propionate as a starting material was proposed to be the phosphate. Prog Lipid Res 2007;46:126–44.
[7] Wennekes T, van den Berg RJ, Boot RG, van der Marel GA, Overkleeft HS, Aerts JM.
major source of odd-numbered FAs [266]. However, later it turned out Glycosphingolipids—nature, function, and pharmacological modulation. Angew
that 2-OH FAs are major sources of odd-numbered FAs [267]. In brain, Chem Int Ed Engl 2009;48:8848–69.
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 65

[8] Pontier SM, Schweisguth F. Glycosphingolipids in signaling and development: from [44] Sassa T, Kihara A. Metabolism of very long-chain fatty acids: genes and pathophys-
liposomes to model organisms. Dev Dyn 2012;241:92–106. iology. Biomol Ther 2014;22:83–92.
[9] Vance DE, Vance JE. Biochemistry of lipids, lipoproteins and membranes. fifth ed. [45] Yamashita A, Sugiura T, Waku K. Acyltransferases and transacylases involved in
Netherlands: Springer-Verlag; 2008. fatty acid remodeling of phospholipids and metabolism of bioactive lipids in mam-
[10] Ikeda M, Kihara A, Igarashi Y. Lipid asymmetry of the eukaryotic plasma mem- malian cells. J Biochem 1997;122:1–16.
brane: functions and related enzymes. Biol Pharm Bull 2006;29:1542–6. [46] Farwanah H, Wohlrab J, Neubert RH, Raith K. Profiling of human stratum corneum
[11] Tanford C. The hydrophobic effect: formation of micelles and biological mem- ceramides by means of normal phase LC/APCI-MS. Anal Bioanal Chem 2005;383:
branes. second ed. New York: Wiley; 1980. 632–7.
[12] Pinto WJ, Srinivasan B, Shepherd S, Schmidt A, Dickson RC, Lester RL. Sphingolipid [47] Sandhoff R. Very long chain sphingolipids: tissue expression, function and synthe-
long-chain-base auxotrophs of Saccharomyces cerevisiae: genetics, physiology, and sis. FEBS Lett 2010;584:1907-13.
a method for their selection. J Bacteriol 1992;174:2565–74. [48] Kishimoto Y, Radin NS. Occurrence of 2-hydroxy fatty acids in animal tissues. J Lipid
[13] Adachi-Yamada T, Gotoh T, Sugimura I, Tateno M, Nishida Y, Onuki T, et al. De novo Res 1963;4:139-43.
synthesis of sphingolipids is required for cell survival by down-regulating c-Jun N- [49] Svennerholm L, Ställberg-Stenhagen S. Changes in the fatty acid composition of ce-
terminal kinase in Drosophila imaginal discs. Mol Cell Biol 1999;19:7276–86. rebrosides and sulfatides of human nervous tissue with age. J Lipid Res 1968;9:
[14] Hojjati MR, Li Z, Jiang XC. Serine palmitoyl-CoA transferase (SPT) deficiency and 215–25.
sphingolipid levels in mice. Biochim Biophys Acta 1737;2005:44–51. [50] Bowen DM, Radin NS. Hydroxy fatty acid metabolism in brain. Adv Lipid Res 1968;
[15] Chen M, Han G, Dietrich CR, Dunn TM, Cahoon EB. The essential nature of 6:255–72.
sphingolipids in plants as revealed by the functional identification and characteri- [51] Umesaki Y, Suzuki A, Kasama T, Tohyama K, Mutai M, Yamakawa T. Presence of
zation of the Arabidopsis LCB1 subunit of serine palmitoyltransferase. Plant Cell asialo GM1 and glucosylceramide in the intestinal mucosa of mice and induction
2006;18:3576–93. of fucosyl asialo GM1 by conventionalization of germ-free mice. J Biochem 1981;
[16] Kihara A, Igarashi Y. Production and release of sphingosine 1-phosphate and the 90:1731–8.
phosphorylated form of the immunomodulator FTY720. Biochim Biophys Acta [52] Zöller I, Meixner M, Hartmann D, Büssow H, Meyer R, Gieselmann V, et al. Absence of
1781;2008:496–502. 2-hydroxylated sphingolipids is compatible with normal neural development but
[17] Mizutani Y, Mitsutake S, Tsuji K, Kihara A, Igarashi Y. Ceramide biosynthesis in causes late-onset axon and myelin sheath degeneration. J Neurosci 2008;28:9741–54.
keratinocyte and its role in skin function. Biochimie 2009;91:784–90. [53] Hanada K. Serine palmitoyltransferase, a key enzyme of sphingolipid metabolism.
[18] Kulkarni AA, Weiss AA, Iyer SS. Glycan-based high-affinity ligands for toxins and Biochim Biophys Acta 1632;2003:16–30.
pathogen receptors. Med Res Rev 2010;30:327–93. [54] Hornemann T, Richard S, Rutti MF, Wei Y, von Eckardstein A. Cloning and initial
[19] Inokuchi J. Physiopathological function of hematoside (GM3 ganglioside). Proc Jpn characterization of a new subunit for mammalian serine-palmitoyltransferase. J
Acad Ser B Phys Biol Sci 2011;87:179–98. Biol Chem 2006;281:37275–81.
[20] Maceyka M, Spiegel S. Sphingolipid metabolites in inflammatory disease. Nature [55] Han G, Gupta SD, Gable K, Niranjanakumari S, Moitra P, Eichler F, et al. Identifica-
2014;510:58–67. tion of small subunits of mammalian serine palmitoyltransferase that confer dis-
[21] Rabionet M, Bayerle A, Jennemann R, Heid H, Fuchser J, Marsching C, et al. Male tinct acyl-CoA substrate specificities. Proc Natl Acad Sci U S A 2009;106:8186–91.
meiotic cytokinesis requires ceramide synthase 3-dependent sphingolipids with [56] Yasuda S, Nishijima M, Hanada K. Localization, topology, and function of the LCB1
unique membrane anchors. Hum Mol Genet 2015;24:4792–808. subunit of serine palmitoyltransferase in mammalian cells. J Biol Chem 2003;278:
[22] Schmitt S, Castelvetri LC, Simons M. Metabolism and functions of lipids in myelin. 4176–83.
Biochim Biophys Acta 1851;2015:999–1005. [57] Breslow DK, Collins SR, Bodenmiller B, Aebersold R, Simons K, Shevchenko A, et al.
[23] Ramstedt B, Slotte JP. Sphingolipids and the formation of sterol-enriched ordered Orm family proteins mediate sphingolipid homeostasis. Nature 2010;463:1048–53.
membrane domains. Biochim Biophys Acta 1758;2006:1945–56. [58] Siow DL, Wattenberg BW. Mammalian ORMDL proteins mediate the feedback re-
[24] Yamagata M, Obara K, Kihara A. Sphingolipid synthesis is involved in autophagy in sponse in ceramide biosynthesis. J Biol Chem 2012;287:40198–204.
Saccharomyces cerevisiae. Biochem Biophys Res Commun 2011;410:786–91. [59] Verhoeven K, Timmerman V, Mauko B, Pieber TR, De Jonghe P, Auer-Grumbach M.
[25] Grösch S, Schiffmann S, Geisslinger G. Chain length-specific properties of Recent advances in hereditary sensory and autonomic neuropathies. Curr Opin
ceramides. Prog Lipid Res 2012;51:50–62. Neurol 2006;19:474–80.
[26] Yamagata M, Obara K, Kihara A. Unperverted synthesis of complex sphingolipids is [60] Rotthier A, Auer-Grumbach M, Janssens K, Baets J, Penno A, Almeida-Souza L, et al.
essential for cell survival under nitrogen starvation. Genes Cells 2013;18:650–9. Mutations in the SPTLC2 subunit of serine palmitoyltransferase cause hereditary
[27] Obara K, Kojima R, Kihara A. Effects on vesicular transport pathways at the late en- sensory and autonomic neuropathy type I. Am J Hum Genet 2010;87:513–22.
dosome in cells with limited very long-chain fatty acids. J Lipid Res 2013;54: [61] Penno A, Reilly MM, Houlden H, Laura M, Rentsch K, Niederkofler V, et al. Heredi-
831–42. tary sensory neuropathy type 1 is caused by the accumulation of two neurotoxic
[28] Mitsutake S, Igarashi Y. Sphingolipids in lipid microdomains and obesity. Vitam sphingolipids. J Biol Chem 2010;285:11178–87.
Horm 2013;91:271–84. [62] Kihara A, Igarashi Y. FVT-1 is a mammalian 3-ketodihydrosphingosine reductase
[29] Harvald EB, Olsen AS, Færgeman NJ. Autophagy in the light of sphingolipid metab- with an active site that faces the cytosolic side of the endoplasmic reticulum mem-
olism. Apoptosis 2015;20:658–70. brane. J Biol Chem 2004;279:49243–50.
[30] Uchida Y, Holleran WM. Omega-O-acylceramide, a lipid essential for mammalian [63] Krebs S, Medugorac I, Rother S, Strasser K, Förster M. A missense mutation in the 3-
survival. J Dermatol Sci 2008;51:77–87. ketodihydrosphingosine reductase FVT1 as candidate causal mutation for bovine
[31] Breiden B, Sandhoff K. The role of sphingolipid metabolism in cutaneous perme- spinal muscular atrophy. Proc Natl Acad Sci U S A 2007;104:6746–51.
ability barrier formation. Biochim Biophys Acta 1841;2014:441–52. [64] Ternes P, Franke S, Zahringer U, Sperling P, Heinz E. Identification and characteriza-
[32] Rosen H, Stevens RC, Hanson M, Roberts E, Oldstone MB. Sphingosine-1-phosphate tion of a sphingolipid Δ4-desaturase family. J Biol Chem 2002;277:25512–8.
and its receptors: structure, signaling, and influence. Annu Rev Biochem 2013;82: [65] Holland WL, Brozinick JT, Wang LP, Hawkins ED, Sargent KM, Liu Y, et al. Inhibition
637–62. of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-
[33] Blaho VA, Hla T. An update on the biology of sphingosine 1-phosphate receptors. J induced insulin resistance. Cell Metab 2007;5:167–79.
Lipid Res 2014;55:1596–608. [66] Mizutani Y, Kihara A, Igarashi Y. Identification of the human sphingolipid C4-
[34] Kappos L, Antel J, Comi G, Montalban X, O'Connor P, Polman CH, et al. Oral hydroxylase, hDES2, and its up-regulation during keratinocyte differentiation.
fingolimod (FTY720) for relapsing multiple sclerosis. N Engl J Med 2006;355: FEBS Lett 2004;563:93–7.
1124-40. [67] Schorling S, Vallée B, Barz WP, Riezman H, Oesterhelt D. Lag1p and Lac1p are es-
[35] Bigaud M, Guerini D, Billich A, Bassilana F, Brinkmann V. Second generation S1P sential for the acyl-CoA-dependent ceramide synthase reaction in Saccharomyces
pathway modulators: research strategies and clinical developments. Biochim cerevisae. Mol Biol Cell 2001;12:3417–27.
Biophys Acta 1841;2014:745-58. [68] Guillas I, Kirchman PA, Chuard R, Pfefferli M, Jiang JC, Jazwinski SM, et al. C26-CoA-
[36] Pruett ST, Bushnev A, Hagedorn K, Adiga M, Haynes CA, Sullards MC, et al. Biodiver- dependent ceramide synthesis of Saccharomyces cerevisiae is operated by Lag1p
sity of sphingoid bases (“sphingosines”) and related amino alcohols. J Lipid Res and Lac1p. EMBO J 2001;20:2655–65.
2008;49:1621-39. [69] Vallée B, Riezman H. Lip1p: a novel subunit of acyl-CoA ceramide synthase. EMBO J
[37] Renkonen O, Hirvisalo EL. Structure of plasma sphingadienine. J Lipid Res 1969;10: 2005;24:730–41.
687-93. [70] Barz WP, Walter P. Two endoplasmic reticulum (ER) membrane proteins that facil-
[38] t'Kindt R, Jorge L, Dumont E, Couturon P, David F, Sandra P, et al. Profiling and char- itate ER-to-Golgi transport of glycosylphosphatidylinositol-anchored proteins. Mol
acterizing skin ceramides using reversed-phase liquid chromatography- Biol Cell 1999;10:1043–59.
quadrupole time-of-flight mass spectrometry. Anal Chem 2012;84:403–11. [71] Venkataraman K, Riebeling C, Bodennec J, Riezman H, Allegood JC, Sullards MC,
[39] Narayanaswamy P, Shinde S, Sulc R, Kraut R, Staples G, Thiam CH, et al. Lipidomic et al. Upstream of growth and differentiation factor 1 (uog1), a mammalian homo-
“deep profiling”: an enhanced workflow to reveal new molecular species of signal- log of the yeast longevity assurance gene 1 (LAG1), regulates N-stearoyl-
ing lipids. Anal Chem 2014;86:3043-7. sphinganine (C18-(dihydro)ceramide) synthesis in a fumonisin B1-independent
[40] Iwamori M, Costello C, Moser HW. Analysis and quantitation of free ceramide con- manner in mammalian cells. J Biol Chem 2002;277:35642–9.
taining nonhydroxy and 2-hydroxy fatty acids, and phytosphingosine by high- [72] Mizutani Y, Kihara A, Igarashi Y. Mammalian Lass6 and its related family members
performance liquid chromatography. J Lipid Res 1979;20:86–96. regulate synthesis of specific ceramides. Biochem J 2005;390:263–71.
[41] Nishimura K. Phytosphingosine is a characteristic component of the glycolipids in [73] Ginkel C, Hartmann D, vom Dorp K, Zlomuzica A, Farwanah H, Eckhardt M, et al.
the vertebrate intestine. Comp Biochem Physiol B 1987;86:149–54. Ablation of neuronal ceramide synthase 1 in mice decreases ganglioside levels
[42] Masukawa Y, Narita H, Shimizu E, Kondo N, Sugai Y, Oba T, et al. Characterization of and expression of myelin-associated glycoprotein in oligodendrocytes. J Biol
overall ceramide species in human stratum corneum. J Lipid Res 2008;49:1466-76. Chem 2012;287:41888-902.
[43] Kihara A. Very long-chain fatty acids: elongation, physiology and related disorders. [74] Sugimoto M, Shimizu Y, Yoshioka T, Wakabayashi M, Tanaka Y, Higashino K, et al.
J Biochem 2012;152:387-95. Histological analyses by matrix-assisted laser desorption/ionization-imaging mass
66 A. Kihara / Progress in Lipid Research 63 (2016) 50–69

spectrometry reveal differential localization of sphingomyelin molecular species [104] Ohno Y, Suto S, Yamanaka M, Mizutani Y, Mitsutake S, Igarashi Y, et al. ELOVL1 pro-
regulated by particular ceramide synthase in mouse brains. Biochim Biophys Acta duction of C24 acyl-CoAs is linked to C24 sphingolipid synthesis. Proc Natl Acad Sci
1851;2015:1554–65. U S A 2010;107:18439–44.
[75] Sassa T, Hirayama T, Kihara A. Enzyme activities of the ceramide synthases [105] Sassa T, Suto S, Okayasu Y, Kihara A. A shift in sphingolipid composition from C24
CERS2–6 are regulated by phosphorylation in the C-terminal region. J Biol Chem to C16 increases susceptibility to apoptosis in HeLa cells. Biochim Biophys Acta
2016;291:7477-87. 1821;2012:1031–7.
[76] Zhao L, Spassieva SD, Jucius TJ, Shultz LD, Shick HE, Macklin WB, et al. A deficiency [106] Schneiter R, Hitomi M, Ivessa AS, Fasch EV, Kohlwein SD, Tartakoff AM. A yeast ace-
of ceramide biosynthesis causes cerebellar purkinje cell neurodegeneration and tyl coenzyme a carboxylase mutant links very-long-chain fatty acid synthesis to the
lipofuscin accumulation. PLoS Genet 2011;7, e1002063. structure and function of the nuclear membrane-pore complex. Mol Cell Biol 1996;
[77] Guillas I, Jiang JC, Vionnet C, Roubaty C, Uldry D, Chuard R, et al. Human homo- 16:7161–72.
logues of LAG1 reconstitute acyl-CoA-dependent ceramide synthesis in yeast. J [107] Denic V, Weissman JS. A molecular caliper mechanism for determining very long-
Biol Chem 2003;278:37083–91. chain fatty acid length. Cell 2007;130:663–77.
[78] Laviad EL, Albee L, Pankova-Kholmyansky I, Epstein S, Park H, Merrill Jr AH, et al. [108] Kihara A, Sakuraba H, Ikeda M, Denpoh A, Igarashi Y. Membrane topology and es-
Characterization of ceramide synthase 2: tissue distribution, substrate specificity, sential amino acid residues of Phs1, a 3-hydroxyacyl-CoA dehydratase involved in
and inhibition by sphingosine 1-phosphate. J Biol Chem 2008;283:5677–84. very long-chain fatty acid elongation. J Biol Chem 2008;283:11199–209.
[79] Ben-David O, Pewzner-Jung Y, Brenner O, Laviad EL, Kogot-Levin A, Weissberg I, [109] Nugteren DH. The enzymic chain elongation of fatty acids by rat-liver microsomes.
et al. Encephalopathy caused by ablation of very long acyl chain ceramide synthesis Biochim Biophys Acta 1965;106:280–90.
may be largely due to reduced galactosylceramide levels. J Biol Chem 2011;286: [110] Haslam TM, Kunst L. Extending the story of very-long-chain fatty acid elongation.
30022–33. Plant Sci 2013;210:93–107.
[80] Honke K. Biosynthesis and biological function of sulfoglycolipids. Proc Jpn Acad Ser [111] Ianiri G, Abhyankar R, Kihara A, Idnurm A. Phs1 and the synthesis of very long chain
B Phys Biol Sci 2013;89:129–38. fatty acids are required for ballistospore formation. PLoS ONE 2014;9, e105147.
[81] Imgrund S, Hartmann D, Farwanah H, Eckhardt M, Sandhoff R, Degen J, et al. Adult [112] Ejsing CS, Sampaio JL, Surendranath V, Duchoslav E, Ekroos K, Klemm RW, et al.
ceramide synthase 2 (CERS2)-deficient mice exhibit myelin sheath defects, cere- Global analysis of the yeast lipidome by quantitative shotgun mass spectrometry.
bellar degeneration, and hepatocarcinomas. J Biol Chem 2009;284:33549–60. Proc Natl Acad Sci U S A 2009;106:2136–41.
[82] Pewzner-Jung Y, Brenner O, Braun S, Laviad EL, Ben-Dor S, Feldmesser E, et al. A [113] Ikeda M, Kanao Y, Yamanaka M, Sakuraba H, Mizutani Y, Igarashi Y, et al. Character-
critical role for ceramide synthase 2 in liver homeostasis: II. Insights into molecular ization of four mammalian 3-hydroxyacyl-CoA dehydratases involved in very long-
changes leading to hepatopathy. J Biol Chem 2010;285:10911–23. chain fatty acid synthesis. FEBS Lett 2008;582:2435–40.
[83] Bosio A, Binczek E, Stoffel W. Functional breakdown of the lipid bilayer of the my- [114] Moon YA, Horton JD. Identification of two mammalian reductases involved in the
elin membrane in central and peripheral nervous system by disrupted two-carbon fatty acyl elongation cascade. J Biol Chem 2003;278:7335–43.
galactocerebroside synthesis. Proc Natl Acad Sci U S A 1996;93:13280–5. [115] Han G, Gable K, Kohlwein SD, Beaudoin F, Napier JA, Dunn TM. The Saccharomyces
[84] Coetzee T, Fujita N, Dupree J, Shi R, Blight A, Suzuki K, et al. Myelination in the ab- cerevisiae YBR159w gene encodes the 3-ketoreductase of the microsomal fatty acid
sence of galactocerebroside and sulfatide: normal structure with abnormal func- elongase. J Biol Chem 2002;277:35440–9.
tion and regional instability. Cell 1996;86:209–19. [116] Kohlwein SD, Eder S, Oh CS, Martin CE, Gable K, Bacikova D, et al. Tsc13p is required
[85] Honke K, Hirahara Y, Dupree J, Suzuki K, Popko B, Fukushima K, et al. Paranodal for fatty acid elongation and localizes to a novel structure at the nuclear-vacuolar
junction formation and spermatogenesis require sulfoglycolipids. Proc Natl Acad interface in Saccharomyces cerevisiae. Mol Cell Biol 2001;21:109–25.
Sci U S A 2002;99:4227–32. [117] Naganuma T, Kihara A. Two modes of regulation of the fatty acid elongase ELOVL6
[86] Mizutani Y, Kihara A, Igarashi Y. LASS3 (longevity assurance homologue 3) is a by the 3-ketoacyl-CoA reductase KAR in the fatty acid elongation cycle. PLoS ONE
mainly testis-specific (dihydro)ceramide synthase with relatively broad substrate 2014;9, e101823.
specificity. Biochem J 2006;398:531–8. [118] Abe K, Ohno Y, Sassa T, Taguchi R, Caliskan M, Ober C, et al. Mutation for nonsyn-
[87] Jennemann R, Rabionet M, Gorgas K, Epstein S, Dalpke A, Rothermel U, et al. Loss of dromic mental retardation in the trans-2-enoyl-CoA reductase TER gene involved
ceramide synthase 3 causes lethal skin barrier disruption. Hum Mol Genet 2012; in fatty acid elongation impairs the enzyme activity and stability, leading to change
21:586–608. in sphingolipid profile. J Biol Chem 2013;288:36741–9.
[88] Mizutani Y, Kihara A, Chiba H, Tojo H, Igarashi Y. 2-hydroxy-ceramide synthesis by [119] Konishi H, Okuda A, Ohno Y, Kihara A. Characterization of HACD1 K64Q mutant
ceramide synthase family: enzymatic basis for the preference of FA chain length. J found in arrhythmogenic right ventricular dysplasia patients. J Biochem 2010;
Lipid Res 2008;49:2356–64. 148:617–22.
[89] Eckl KM, Tidhar R, Thiele H, Oji V, Hausser I, Brodesser S, et al. Impaired epidermal cer- [120] Okuda A, Naganuma T, Ohno Y, Abe K, Yamagata M, Igarashi Y, et al. Hetero-
amide synthesis causes autosomal recessive congenital ichthyosis and reveals the im- oligomeric interactions of an ELOVL4 mutant protein: implications in the molecu-
portance of ceramide acyl chain length. J Invest Dermatol 2013;133:2202–11. lar mechanism of Stargardt-3 macular dystrophy. Mol Vis 2010;16:2438–45.
[90] Riebeling C, Allegood JC, Wang E, Merrill Jr AH, Futerman AH. Two mammalian lon- [121] Rantakari P, Lagerbohm H, Kaimainen M, Suomela JP, Strauss L, Sainio K, et al. Hy-
gevity assurance gene (LAG1) family members, trh1 and trh4, regulate droxysteroid (17β) dehydrogenase 12 is essential for mouse organogenesis and
dihydroceramide synthesis using different fatty acyl-CoA donors. J Biol Chem embryonic survival. Endocrinology 2010;151:1893–901.
2003;278:43452–9. [122] Revardel E, Bonneau M, Durrens P, Aigle M. Characterization of a new gene family
[91] Ebel P, Imgrund S, Vom Dorp K, Hofmann K, Maier H, Drake H, et al. Ceramide syn- developing pleiotropic phenotypes upon mutation in Saccharomyces cerevisiae.
thase 4 deficiency in mice causes lipid alterations in sebum and results in alopecia. Biochim Biophys Acta 1995;1263:261–5.
Biochem J 2014;461:147–58. [123] Cerantola V, Vionnet C, Aebischer OF, Jenny T, Knudsen J, Conzelmann A. Yeast
[92] Lahiri S, Futerman AH. LASS5 is a bona fide dihydroceramide synthase that selec- sphingolipids do not need to contain very long chain fatty acids. Biochem J 2007;
tively utilizes palmitoyl-CoA as acyl donor. J Biol Chem 2005;280:33735–8. 401:205–16.
[93] Ebel P, Vom Dorp K, Petrasch-Parwez E, Zlomuzica A, Kinugawa K, Mariani J, et al. [124] Epstein S, Castillon GA, Qin Y, Riezman H. An essential function of sphingolipids in
Inactivation of ceramide synthase 6 in mice results in an altered sphingolipid me- yeast cell division. Mol Microbiol 2012;84:1018–32.
tabolism and behavioral abnormalities. J Biol Chem 2013;288:21433–47. [125] Wakashima T, Abe K, Kihara A. Dual functions of the trans-2-enoyl-CoA reductase
[94] Gosejacob D, Jäger PS, Vom Dorp K, Frejno M, Carstensen AC, Köhnke M, et al. Cer- TER in the sphingosine 1-phosphate metabolic pathway and in fatty acid elonga-
amide synthase 5 is essential to maintain C16:0 ceramide pools and contributes to tion. J Biol Chem 2014;289:24736–48.
the development of diet induced obesity. J Biol Chem 2016;291:6989–7003. [126] Kitazawa H, Miyamoto Y, Shimamura K, Nagumo A, Tokita S. Development of a
[95] Schiffmann S, Ferreiros N, Birod K, Eberle M, Schreiber Y, Pfeilschifter W, et al. Cer- high-density assay for long-chain fatty acyl-CoA elongases. Lipids 2009;44:765–73.
amide synthase 6 plays a critical role in the development of experimental autoim- [127] Sassa T, Ohno Y, Suzuki S, Nomura T, Nishioka C, Kashiwagi T, et al. Impaired epi-
mune encephalomyelitis. J Immunol 2012;188:5723–33. dermal permeability barrier in mice lacking Elovl1, the gene responsible for very-
[96] Muir A, Ramachandran S, Roelants FM, Timmons G, Thorner J. TORC2-dependent long-chain fatty acid production. Mol Cell Biol 2013;33:2787–96.
protein kinase Ypk1 phosphorylates ceramide synthase to stimulate synthesis of [128] Mizutani Y, Sun H, Ohno Y, Sassa T, Wakashima T, Obara M, et al. Cooperative syn-
complex sphingolipids. eLife 2014;3. thesis of ultra long-chain fatty acid and ceramide during keratinocyte differentia-
[97] Fresques T, Niles B, Aronova S, Mogri H, Rakhshandehroo T, Powers T. Regulation of tion. PLoS ONE 2013;8, e67317.
ceramide synthase by casein kinase 2-dependent phosphorylation in Saccharomy- [129] Eaton S, Bartlett K, Pourfarzam M. Mammalian mitochondrial β-oxidation. Biochem
ces cerevisiae. J Biol Chem 2015;290:1395–403. J 1996;320:345–57.
[98] Kageyama-Yahara N, Riezman H. Transmembrane topology of ceramide synthase [130] Wanders RJ. Peroxisomes, lipid metabolism, and peroxisomal disorders. Mol Genet
in yeast. Biochem J 2006;398:585–93. Metab 2004;83:16–27.
[99] Sridevi P, Alexander H, Laviad EL, Pewzner-Jung Y, Hannink M, Futerman AH, et al. [131] Kemp S, Berger J, Aubourg P. X-linked adrenoleukodystrophy: clinical, metabolic,
Ceramide synthase 1 is regulated by proteasomal mediated turnover. Biochim genetic and pathophysiological aspects. Biochim Biophys Acta 1822;2012:
Biophys Acta 1793;2009:1218–27. 1465–74.
[100] Niemelä PS, Hyvonen MT, Vattulainen I. Influence of chain length and unsaturation [132] Mosser J, Douar AM, Sarde CO, Kioschis P, Feil R, Moser H, et al. Putative X-linked
on sphingomyelin bilayers. Biophys J 2006;90:851–63. adrenoleukodystrophy gene shares unexpected homology with ABC transporters.
[101] Björkqvist YJ, Brewer J, Bagatolli LA, Slotte JP, Westerlund B. Thermotropic behavior Nature 1993;361:726–30.
and lateral distribution of very long chain sphingolipids. Biochim Biophys Acta [133] Ofman R, Dijkstra IM, van Roermund CW, Burger N, Turkenburg M, van Cruchten A,
1788;2009:1310–20. et al. The role of ELOVL1 in very long-chain fatty acid homeostasis and X-linked ad-
[102] Simons K, Ikonen E. Functional rafts in cell membranes. Nature 1997;387:569–72. renoleukodystrophy. EMBO Mol Med 2010;2:90–7.
[103] Iwabuchi K, Prinetti A, Sonnino S, Mauri L, Kobayashi T, Ishii K, et al. Involvement of [134] Sassa T, Wakashima T, Ohno Y, Kihara A. Lorenzo's oil inhibits ELOVL1 and lowers
very long fatty acid-containing lactosylceramide in lactosylceramide-mediated su- the level of sphingomyelin with a saturated very long-chain fatty acid. J Lipid Res
peroxide generation and migration in neutrophils. Glycoconj J 2008;25:357–74. 2014;55:524–30.
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 67

[135] Moser HW, Raymond GV, Lu SE, Muenz LR, Moser AB, Xu J, et al. Follow-up of 89 [165] Proksch E, Brandner JM, Jensen JM. The skin: an indispensable barrier. Exp
asymptomatic patients with adrenoleukodystrophy treated with Lorenzo's oil. Dermatol 2008;17:1063–72.
Arch Neurol 2005;62:1073–80. [166] Traupe H, Fischer J, Oji V. Nonsyndromic types of ichthyoses—an update. J Dtsch
[136] Leonard AE, Bobik EG, Dorado J, Kroeger PE, Chuang LT, Thurmond JM, et al. Cloning Dermatol Ges 2014;12:109–21.
of a human cDNA encoding a novel enzyme involved in the elongation of long- [167] van Smeden J, Janssens M, Gooris GS, Bouwstra JA. The important role of stratum
chain polyunsaturated fatty acids. Biochem J 2000;350:765–70. corneum lipids for the cutaneous barrier function. Biochim Biophys Acta 1841;
[137] Moon YA, Shah NA, Mohapatra S, Warrington JA, Horton JD. Identification of a 2014:295–313.
mammalian long chain fatty acyl elongase regulated by sterol regulatory [168] Feingold KR, Elias PM. Role of lipids in the formation and maintenance of the cuta-
element-binding proteins. J Biol Chem 2001;276:45358–66. neous permeability barrier. Biochim Biophys Acta 1841;2014:280–94.
[138] Inagaki K, Aki T, Fukuda Y, Kawamoto S, Shigeta S, Ono K, et al. Identification and [169] Thyssen JP, Godoy-Gijon E, Elias PM. Ichthyosis vulgaris: the filaggrin mutation dis-
expression of a rat fatty acid elongase involved in the biosynthesis of C18 fatty ease. Br J Dermatol 2013;168:1155–66.
acids. Biosci Biotechnol Biochem 2002;66:613–21. [170] Palmer CN, Irvine AD, Terron-Kwiatkowski A, Zhao Y, Liao H, Lee SP, et al. Common
[139] Leonard AE, Kelder B, Bobik EG, Chuang LT, Lewis CJ, Kopchick JJ, et al. Identification loss-of-function variants of the epidermal barrier protein filaggrin are a major pre-
and expression of mammalian long-chain PUFA elongation enzymes. Lipids 2002; disposing factor for atopic dermatitis. Nat Genet 2006;38:441–6.
37:733–40. [171] O'Regan GM, Sandilands A, McLean WH, Irvine AD. Filaggrin in atopic dermatitis. J
[140] Zadravec D, Tvrdik P, Guillou H, Haslam R, Kobayashi T, Napier JA, et al. ELOVL2 Allergy Clin Immunol 2008;122:689–93.
controls the level of n-6 28:5 and 30:5 fatty acids in testis, a prerequisite for [172] Candi E, Schmidt R, Melino G. The cornified envelope: a model of cell death in the
male fertility and sperm maturation in mice. J Lipid Res 2011;52:245–55. skin. Nat Rev Mol Cell Biol 2005;6:328–40.
[141] Moon YA, Hammer RE, Horton JD. Deletion of ELOVL5 leads to fatty liver through [173] Kypriotou M, Huber M, Hohl D. The human epidermal differentiation complex:
activation of SREBP-1c in mice. J Lipid Res 2009;50:412–23. cornified envelope precursors, S100 proteins and the ‘fused genes’ family. Exp
[142] Westerberg R, Tvrdik P, Undén AB, Mansson JE, Norlén L, Jakobsson A, et al. Role for Dermatol 2012;21:643–9.
ELOVL3 and fatty acid chain length in development of hair and skin function. J Biol [174] Marekov LN, Steinert PM. Ceramides are bound to structural proteins of the human
Chem 2004;279:5621–9. foreskin epidermal cornified cell envelope. J Biol Chem 1998;273:17763–70.
[143] Westerberg R, Mansson JE, Golozoubova V, Shabalina IG, Backlund EC, Tvrdik P, [175] Elias PM, Gruber R, Crumrine D, Menon G, Williams ML, Wakefield JS, et al. Forma-
et al. ELOVL3 is an important component for early onset of lipid recruitment in tion and functions of the corneocyte lipid envelope (CLE). Biochim Biophys Acta
brown adipose tissue. J Biol Chem 2006;281:4958–68. 1841;2014:314–8.
[144] Tamura K, Makino A, Hullin-Matsuda F, Kobayashi T, Furihata M, Chung S, et al. [176] Lampe MA, Burlingame AL, Whitney J, Williams ML, Brown BE, Roitman E, et al.
Novel lipogenic enzyme ELOVL7 is involved in prostate cancer growth through sat- Human stratum corneum lipids: characterization and regional variations. J Lipid
urated long-chain fatty acid metabolism. Cancer Res 2009;69:8133–40. Res 1983;24:120–30.
[145] Naganuma T, Sato Y, Sassa T, Ohno Y, Kihara A. Biochemical characterization of the [177] Akiyama M. The roles of ABCA12 in epidermal lipid barrier formation and
very long-chain fatty acid elongase ELOVL7. FEBS Lett 2011;585:3337–41. keratinocyte differentiation. Biochim Biophys Acta 1841;2014:435–40.
[146] McMahon A, Butovich IA, Mata NL, Klein M, Ritter 3rd R, Richardson J, et al. Retinal [178] Akiyama M, Sugiyama-Nakagiri Y, Sakai K, McMillan JR, Goto M, Arita K, et al. Mu-
pathology and skin barrier defect in mice carrying a Stargardt disease-3 mutation tations in lipid transporter ABCA12 in harlequin ichthyosis and functional recovery
in elongase of very long chain fatty acids-4. Mol Vis 2007;13:258–72. by corrective gene transfer. J Clin Invest 2005;115:1777–84.
[147] Vasireddy V, Uchida Y, Salem Jr N, Kim SY, Mandal MN, Reddy GB, et al. Loss of func- [179] Oji V, Tadini G, Akiyama M, Blanchet Bardon C, Bodemer C, Bourrat E, et al. Revised
tional ELOVL4 depletes very long-chain fatty acids (≥C28) and the unique ω-O- nomenclature and classification of inherited ichthyoses: results of the first
acylceramides in skin leading to neonatal death. Hum Mol Genet 2007;16:471–82. ichthyosis consensus conference in Soreze 2009. J Am Acad Dermatol 2010;63:
[148] Agbaga MP, Brush RS, Mandal MN, Henry K, Elliott MH, Anderson RE. Role of 607–41.
Stargardt-3 macular dystrophy protein (ELOVL4) in the biosynthesis of very long [180] Sillence DJ, Platt FM. Storage diseases: new insights into sphingolipid functions.
chain fatty acids. Proc Natl Acad Sci U S A 2008;105:12843–8. Trends Cell Biol 2003;13:195–203.
[149] Zhang K, Kniazeva M, Han M, Li W, Yu Z, Yang Z, et al. A 5-bp deletion in ELOVL4 is [181] Sidransky E, Sherer DM, Ginns EI. Gaucher disease in the neonate: a distinct Gauch-
associated with two related forms of autosomal dominant macular dystrophy. Nat er phenotype is analogous to a mouse model created by targeted disruption of the
Genet 2001;27:89–93. glucocerebrosidase gene. Pediatr Res 1992;32:494–8.
[150] Aldahmesh MA, Mohamed JY, Alkuraya HS, Verma IC, Puri RD, Alaiya AA, et al. Re- [182] Holleran WM, Ginns EI, Menon GK, Grundmann JU, Fartasch M, McKinney CE, et al.
cessive mutations in ELOVL4 cause ichthyosis, intellectual disability, and spastic Consequences of β-glucocerebrosidase deficiency in epidermis. Ultrastructure and
quadriplegia. Am J Hum Genet 2011;89:745–50. permeability barrier alterations in Gaucher disease. J Clin Invest 1994;93:1756–64.
[151] Vasireddy V, Wong P, Ayyagari R. Genetics and molecular pathology of Stargardt- [183] Lefèvre C, Bouadjar B, Ferrand V, Tadini G, Mégarbané A, Lathrop M, et al. Mutations
like macular degeneration. Prog Retin Eye Res 2010;29:191–207. in a new cytochrome P450 gene in lamellar ichthyosis type 3. Hum Mol Genet
[152] Li W, Sandhoff R, Kono M, Zerfas P, Hoffmann V, Ding BC, et al. Depletion of 2006;15:767–76.
ceramides with very long chain fatty acids causes defective skin permeability bar- [184] Sugiura K, Takeichi T, Tanahashi K, Ito Y, Kosho T, Saida K, et al. Lamellar ichthyosis
rier function, and neonatal lethality in ELOVL4 deficient mice. Int J Biol Sci 2007;3: in a collodion baby caused by CYP4F22 mutations in a non-consanguineous family
120–8. outside the Mediterranean. J Dermatol Sci 2013;72:193–5.
[153] Matsuzaka T, Shimano H, Yahagi N, Yoshikawa T, Amemiya-Kudo M, Hasty AH, [185] Imokawa G, Abe A, Jin K, Higaki Y, Kawashima M, Hidano A. Decreased level of
et al. Cloning and characterization of a mammalian fatty acyl-CoA elongase as a ceramides in stratum corneum of atopic dermatitis: an etiologic factor in atopic
lipogenic enzyme regulated by SREBPs. J Lipid Res 2002;43:911–20. dry skin? J Invest Dermatol 1991;96:523–6.
[154] Matsuzaka T, Shimano H, Yahagi N, Kato T, Atsumi A, Yamamoto T, et al. Crucial [186] Di Nardo A, Wertz P, Giannetti A, Seidenari S. Ceramide and cholesterol composi-
role of a long-chain fatty acid elongase, Elovl6, in obesity-induced insulin resis- tion of the skin of patients with atopic dermatitis. Acta Derm Venereol 1998;78:
tance. Nat Med 2007;13:1193–202. 27–30.
[155] Matsuzaka T, Atsumi A, Matsumori R, Nie T, Shinozaki H, Suzuki-Kemuriyama N, [187] Ishikawa J, Narita H, Kondo N, Hotta M, Takagi Y, Masukawa Y, et al. Changes in the
et al. Elovl6 promotes nonalcoholic steatohepatitis. Hepatology 2012;56:2199–208. ceramide profile of atopic dermatitis patients. J Invest Dermatol 2010;130:2511–4.
[156] Toke DA, Martin CE. Isolation and characterization of a gene affecting fatty acid [188] Janssens M, van Smeden J, Gooris GS, Bras W, Portale G, Caspers PJ, et al. Increase in
elongation in Saccharomyces cerevisiae. J Biol Chem 1996;271:18413–22. short-chain ceramides correlates with an altered lipid organization and decreased
[157] Oh CS, Toke DA, Mandala S, Martin CE. ELO2 and ELO3, homologues of the Saccha- barrier function in atopic eczema patients. J Lipid Res 2012;53:2755–66.
romyces cerevisiae ELO1 gene, function in fatty acid elongation and are required for [189] Motta S, Monti M, Sesana S, Caputo R, Carelli S, Ghidoni R. Ceramide composition of
sphingolipid formation. J Biol Chem 1997;272:17376–84. the psoriatic scale. Biochim Biophys Acta 1993;1182:147–51.
[158] Caliskan M, Chong JX, Uricchio L, Anderson R, Chen P, Sougnez C, et al. Exome se- [190] Ohno Y, Nakamichi S, Ohkuni A, Kamiyama N, Naoe A, Tsujimura H, et al. Essential
quencing reveals a novel mutation for autosomal recessive non-syndromic mental re- role of the cytochrome P450 CYP4F22 in the production of acylceramide, the key
tardation in the TECR gene on chromosome 19p13. Hum Mol Genet 2011;20:1285–9. lipid for skin permeability barrier formation. Proc Natl Acad Sci U S A 2015;112:
[159] Wang B, Pelletier J, Massaad MJ, Herscovics A, Shore GC. The yeast split-ubiquitin 7707–12.
membrane protein two-hybrid screen identifies BAP31 as a regulator of the turn- [191] Radner FP, Fischer J. The important role of epidermal triacylglycerol metabolism for
over of endoplasmic reticulum-associated protein tyrosine phosphatase-like B. maintenance of the skin permeability barrier function. Biochim Biophys Acta 1841;
Mol Cell Biol 2004;24:2767–78. 2014:409–15.
[160] Li D, Gonzalez O, Bachinski LL, Roberts R. Human protein tyrosine phosphatase-like [192] Stone SJ, Myers HM, Watkins SM, Brown BE, Feingold KR, Elias PM, et al. Lipopenia
gene: expression profile, genomic structure, and mutation analysis in families with and skin barrier abnormalities in DGAT2-deficient mice. J Biol Chem 2004;279:
ARVD. Gene 2000;256:237–43. 11767–76.
[161] Péle M, Tiret L, Kessler JL, Blot S, Panthier JJ. SINE exonic insertion in the PTPLA gene [193] Futerman AH, Pagano RE. Determination of the intracellular sites and topology of
leads to multiple splicing defects and segregates with the autosomal recessive glucosylceramide synthesis in rat liver. Biochem J 1991;280:295–302.
centronuclear myopathy in dogs. Hum Mol Genet 2005;14:1417–27. [194] D'Angelo G, Polishchuk E, Di Tullio G, Santoro M, Di Campli A, Godi A, et al.
[162] Muhammad E, Reish O, Ohno Y, Scheetz T, Deluca A, Searby C, et al. Congenital my- Glycosphingolipid synthesis requires FAPP2 transfer of glucosylceramide. Nature
opathy is caused by mutation of HACD1. Hum Mol Genet 2013;22:5229–36. 2007;449:62–7.
[163] Blondelle J, Ohno Y, Gache V, Guyot S, Storck S, Blanchard-Gutton N, et al. HACD1, a [195] Jennemann R, Sandhoff R, Langbein L, Kaden S, Rothermel U, Gallala H, et al. Integ-
regulator of membrane composition and fluidity, promotes myoblast fusion and rity and barrier function of the epidermis critically depend on glucosylceramide
skeletal muscle growth. J Mol Cell Biol 2015;7:429–40. synthesis. J Biol Chem 2007;282:3083–94.
[164] Yazawa T, Naganuma T, Yamagata M, Kihara A. Identification of residues important [196] Muñoz-Garcia A, Thomas CP, Keeney DS, Zheng Y, Brash AR. The importance of the
for the catalysis, structure maintenance, and substrate specificity of yeast 3- lipoxygenase-hepoxilin pathway in the mammalian epidermal barrier. Biochim
hydroxyacyl-CoA dehydratase Phs1. FEBS Lett 2013;587:804–9. Biophys Acta 1841;2014:401–8.
68 A. Kihara / Progress in Lipid Research 63 (2016) 50–69

[197] Zheng Y, Yin H, Boeglin WE, Elias PM, Crumrine D, Beier DR, et al. Lipoxygenases phosphate binding domain exposed to the cytosol. Biochem Biophys Res Commun
mediate the effect of essential fatty acid in skin barrier formation: a proposed 2004;325:338–43.
role in releasing omega-hydroxyceramide for construction of the corneocyte lipid [226] Yatomi Y, Yamamura S, Ruan F, Igarashi Y. Sphingosine 1-phosphate induces plate-
envelope. J Biol Chem 2011;286:24046–56. let activation through an extracellular action and shares a platelet surface receptor
[198] Jobard F, Lefevre C, Karaduman A, Blanchet-Bardon C, Emre S, Weissenbach J, et al. with lysophosphatidic acid. J Biol Chem 1997;272:5291–7.
Lipoxygenase-3 (ALOXE3) and 12(R)-lipoxygenase (ALOX12B) are mutated in non- [227] Fukuda Y, Kihara A, Igarashi Y. Distribution of sphingosine kinase activity in mouse
bullous congenital ichthyosiform erythroderma (NCIE) linked to chromosome tissues: contribution of SPHK1. Biochem Biophys Res Commun 2003;309:155–60.
17p13.1. Hum Mol Genet 2002;11:107-13. [228] Johnson KR, Becker KP, Facchinetti MM, Hannun YA, Obeid LM. PKC-dependent ac-
[199] Epp N, Furstenberger G, Muller K, de Juanes S, Leitges M, Hausser I, et al. 12R- tivation of sphingosine kinase 1 and translocation to the plasma membrane. Extra-
lipoxygenase deficiency disrupts epidermal barrier function. J Cell Biol 2007;177: cellular release of sphingosine-1-phosphate induced by phorbol 12-myristate 13-
173–82. acetate (PMA). J Biol Chem 2002;277:35257–62.
[200] Moran JL, Qiu H, Turbe-Doan A, Yun Y, Boeglin WE, Brash AR, et al. A mouse muta- [229] Pitson SM, Moretti PA, Zebol JR, Lynn HE, Xia P, Vadas MA, et al. Activation of sphin-
tion in the 12R-lipoxygenase, Alox12b, disrupts formation of the epidermal perme- gosine kinase 1 by ERK1/2-mediated phosphorylation. EMBO J 2003;22:5491–500.
ability barrier. J Invest Dermatol 2007;127:1893–7. [230] Igarashi N, Okada T, Hayashi S, Fujita T, Jahangeer S, Nakamura S. Sphingosine ki-
[201] Krieg P, Rosenberger S, de Juanes S, Latzko S, Hou J, Dick A, et al. Aloxe3 knockout nase 2 is a nuclear protein and inhibits DNA synthesis. J Biol Chem 2003;278:
mice reveal a function of epidermal lipoxygenase-3 as hepoxilin synthase and its 46832–9.
pivotal role in barrier formation. J Invest Dermatol 2013;133:172–80. [231] Maceyka M, Sankala H, Hait NC, Le Stunff H, Liu H, Toman R, et al. SphK1 and
[202] Nemes Z, Marekov LN, Fesus L, Steinert PM. A novel function for transglutaminase SphK2, sphingosine kinase isoenzymes with opposing functions in sphingolipid
1: attachment of long-chain ω-hydroxyceramides to involucrin by ester bond for- metabolism. J Biol Chem 2005;280:37118–29.
mation. Proc Natl Acad Sci U S A 1999;96:8402–7. [232] Kihara A, Anada Y, Igarashi Y. Mouse sphingosine kinase isoforms SPHK1a and
[203] Elias PM, Schmuth M, Uchida Y, Rice RH, Behne M, Crumrine D, et al. Basis for the SPHK1b differ in enzymatic traits including stability, localization, modification,
permeability barrier abnormality in lamellar ichthyosis. Exp Dermatol 2002;11: and oligomerization. J Biol Chem 2006;281:4532–9.
248–56. [233] Zhou J, Saba JD. Identification of the first mammalian sphingosine phosphate lyase
[204] Doering T, Holleran WM, Potratz A, Vielhaber G, Elias PM, Suzuki K, et al. gene and its functional expression in yeast. Biochem Biophys Res Commun 1998;
Sphingolipid activator proteins are required for epidermal permeability barrier for- 242:502–7.
mation. J Biol Chem 1999;274:11038–45. [234] Mandala SM, Thornton R, Galve-Roperh I, Poulton S, Peterson C, Olivera A, et al.
[205] Doering T, Proia RL, Sandhoff K. Accumulation of protein-bound epidermal Molecular cloning and characterization of a lipid phosphohydrolase that degrades
glucosylceramides in β-glucocerebrosidase deficient type 2 Gaucher mice. FEBS sphingosine-1- phosphate and induces cell death. Proc Natl Acad Sci U S A 2000;
Lett 1999;447:167–70. 97:7859–64.
[206] Sugiura M, Kono K, Liu H, Shimizugawa T, Minekura H, Spiegel S, et al. Ceramide [235] Ogawa C, Kihara A, Gokoh M, Igarashi Y. Identification and characterization of a
kinase, a novel lipid kinase. Molecular cloning and functional characterization. J novel human sphingosine-1-phosphate phosphohydrolase, hSPP2. J Biol Chem
Biol Chem 2002;277:23294–300. 2003;278:1268–72.
[207] Gomez-Muñoz A, Presa N, Gomez-Larrauri A, Rivera IG, Trueba M, Ordoñez M. Con- [236] Kihara A, Sano T, Iwaki S, Igarashi Y. Transmembrane topology of sphingoid long-
trol of inflammatory responses by ceramide, sphingosine 1-phosphate and cer- chain base-1-phosphate phosphatase, Lcb3p. Genes Cells 2003;8:525–35.
amide 1-phosphate. Prog Lipid Res 2015;61:51–62. [237] Borowsky AD, Bandhuvula P, Kumar A, Yoshinaga Y, Nefedov M, Fong LG, et al.
[208] Mao C, Obeid LM. Ceramidases: regulators of cellular responses mediated by cer- Sphingosine-1-phosphate lyase expression in embryonic and adult murine tissues.
amide, sphingosine, and sphingosine-1-phosphate. Biochim Biophys Acta 1781; J Lipid Res 2012;53:1920–31.
2008:424–34. [238] Allende ML, Sipe LM, Tuymetova G, Wilson-Henjum KL, Chen W, Proia RL. Sphingo-
[209] Stoffel W. Sphingosine metabolism and its link to phospholipid biosynthesis. Mol sine-1-phosphate phosphatase 1 regulates keratinocyte differentiation and epider-
Cell Biochem 1973;1:147–55. mal homeostasis. J Biol Chem 2013;288:18381–91.
[210] Kihara A. Sphingosine 1-phosphate is a key metabolite linking sphingolipids to [239] Bektas M, Allende ML, Lee BG, Chen W, Amar MJ, Remaley AT, et al. Sphingosine 1-
glycerophospholipids. Biochim Biophys Acta 1841;2014:766–72. phosphate lyase deficiency disrupts lipid homeostasis in liver. J Biol Chem 2010;
[211] Stoffel W, Sticht G. Metabolism of sphingosine bases, II. Studies on the degradation 285:10880–9.
and transformation of [3-14C]erythro-DL-dihydrosphingosine, [7-3H]erythro-DL- [240] Vogel P, Donoviel MS, Read R, Hansen GM, Hazlewood J, Anderson SJ, et al. Incom-
sphingosine, [5-3H]threo-L-dihydrosphingosine and [3-14C;1-3H]erythro-DL- plete inhibition of sphingosine 1-phosphate lyase modulates immune system func-
dihydrosphingosine in rat liver. Hoppe Seylers Z Physiol Chem 1967;348:1345–51. tion yet prevents early lethality and non-lymphoid lesions. PLoS ONE 2009;4,
[212] Stoffel W, Sticht G. Metabolism of sphingosine bases, I. Degradation and incorpora- e4112.
tion of [3-14C]erythro-DL-dihydrosphingosine and [7-3H2]erythro-DL-sphingosine [241] Weber C, Krueger A, Munk A, Bode C, Van Veldhoven PP, Graler MH. Discontinued
into sphingolipids of rat liver. Hoppe Seylers Z Physiol Chem 1967;348:941–3. postnatal thymocyte development in sphingosine 1-phosphate-lyase-deficient
[213] Nakahara K, Ohkuni A, Kitamura T, Abe K, Naganuma T, Ohno Y, et al. The Sjögren- mice. J Immunol 2009;183:4292–301.
Larsson syndrome gene encodes a hexadecenal dehydrogenase of the sphingosine [242] Allende ML, Bektas M, Lee BG, Bonifacino E, Kang J, Tuymetova G, et al. Sphingo-
1-phosphate degradation pathway. Mol Cell 2012;46:461–71. sine-1-phosphate lyase deficiency produces a pro-inflammatory response while
[214] Kondo N, Ohno Y, Yamagata M, Obara T, Seki N, Kitamura T, et al. Identification of impairing neutrophil trafficking. J Biol Chem 2011;286:7348–58.
the phytosphingosine metabolic pathway leading to odd-numbered fatty acids. Nat [243] Marchitti SA, Brocker C, Stagos D, Vasiliou V. Non-P450 aldehyde oxidizing en-
Commun 2014;5:5338. zymes: the aldehyde dehydrogenase superfamily. Expert Opin Drug Metab Toxicol
[215] Park JH, Schuchman EH. Acid ceramidase and human disease. Biochim Biophys 2008;4:697–720.
Acta 1758;2006:2133–8. [244] Jackson B, Brocker C, Thompson DC, Black W, Vasiliou K, Nebert DW, et al. Update
[216] Li CM, Park JH, Simonaro CM, He X, Gordon RE, Friedman AH, et al. Insertional mu- on the aldehyde dehydrogenase gene (ALDH) superfamily. Hum Genomics 2011;5:
tagenesis of the mouse acid ceramidase gene leads to early embryonic lethality in 283–303.
homozygotes and progressive lipid storage disease in heterozygotes. Genomics [245] Estey T, Piatigorsky J, Lassen N, Vasiliou V. ALDH3A1: a corneal crystallin with di-
2002;79:218–24. verse functions. Exp Eye Res 2007;84:3–12.
[217] Kohama T, Olivera A, Edsall L, Nagiec MM, Dickson R, Spiegel S. Molecular cloning [246] Kitamura T, Takagi S, Naganuma T, Kihara A. Mouse aldehyde dehydrogenase
and functional characterization of murine sphingosine kinase. J Biol Chem 1998; ALDH3B2 is localized to lipid droplets via two C-terminal tryptophan residues
273:23722–8. and lipid modification. Biochem J 2015;465:79–87.
[218] Liu H, Sugiura M, Nava VE, Edsall LC, Kono K, Poulton S, et al. Molecular cloning and [247] Kitamura T, Naganuma T, Abe K, Nakahara K, Ohno Y, Kihara A. Substrate specific-
functional characterization of a novel mammalian sphingosine kinase type 2 iso- ity, plasma membrane localization, and lipid modification of the aldehyde dehy-
form. J Biol Chem 2000;275:19513–20. drogenase ALDH3B1. Biochim Biophys Acta 1831;2013:1395–401.
[219] Mizugishi K, Yamashita T, Olivera A, Miller GF, Spiegel S, Proia RL. Essential role for [248] Ashibe B, Hirai T, Higashi K, Sekimizu K, Motojima K. Dual subcellular localization
sphingosine kinases in neural and vascular development. Mol Cell Biol 2005;25: in the endoplasmic reticulum and peroxisomes and a vital role in protecting
11113–21. against oxidative stress of fatty aldehyde dehydrogenase are achieved by alterna-
[220] Billich A, Bornancin F, Devay P, Mechtcheriakova D, Urtz N, Baumruker T. Phos- tive splicing. J Biol Chem 2007;282:20763–73.
phorylation of the immunomodulatory drug FTY720 by sphingosine kinases. J [249] Nagan N, Zoeller RA. Plasmalogens: biosynthesis and functions. Prog Lipid Res
Biol Chem 2003;278:47408–15. 2001;40:199–229.
[221] Ito K, Anada Y, Tani M, Ikeda M, Sano T, Kihara A, et al. Lack of sphingosine 1- [250] Gorgas K, Teigler A, Komljenovic D, Just WW. The ether lipid-deficient mouse:
phosphate-degrading enzymes in erythrocytes. Biochem Biophys Res Commun tracking down plasmalogen functions. Biochim Biophys Acta 1763;2006:1511–26.
2007;357:212–7. [251] Honsho M, Asaoku S, Fujiki Y. Posttranslational regulation of fatty acyl-CoA reduc-
[222] Pappu R, Schwab SR, Cornelissen I, Pereira JP, Regard JB, Xu Y, et al. Promotion of tase 1, Far1, controls ether glycerophospholipid synthesis. J Biol Chem 2010;285:
lymphocyte egress into blood and lymph by distinct sources of sphingosine-1- 8537–42.
phosphate. Science 2007;316:295–8. [252] Naganuma T, Takagi S, Kanetake T, Kitamura T, Sassa T, Kihara A. Disruption of the
[223] Nishi T, Kobayashi N, Hisano Y, Kawahara A, Yamaguchi A. Molecular and physio- Sjögren-Larsson syndrome gene Aldh3a2 in mice increases keratinocyte growth
logical functions of sphingosine 1-phosphate transporters. Biochim Biophys Acta and retards skin barrier recovery. J. Biol. Chem. 2016; (in press).
1841;2014:759–65. [253] Rizzo WB. Sjögren-Larsson syndrome: molecular genetics and biochemical patho-
[224] Anada Y, Igarashi Y, Kihara A. The immunomodulator FTY720 is phosphorylated genesis of fatty aldehyde dehydrogenase deficiency. Mol Genet Metab 2007;90:
and released from platelets. Eur J Pharmacol 2007;568:106–11. 1–9.
[225] Ikeda M, Kihara A, Igarashi Y. Sphingosine-1-phosphate lyase SPL is an endoplas- [254] Rizzo WB. Fatty aldehyde and fatty alcohol metabolism: review and importance for
mic reticulum-resident, integral membrane protein with the pyridoxal 5′- epidermal structure and function. Biochim Biophys Acta 1841;2014:377–89.
A. Kihara / Progress in Lipid Research 63 (2016) 50–69 69

[255] Catalá A. Lipid peroxidation of membrane phospholipids generates hydroxy- [264] Metz J. Cobalamin deficiency and the pathogenesis of nervous system disease.
alkenals and oxidized phospholipids active in physiological and/or pathological Annu Rev Nutr 1992;12:59–79.
conditions. Chem Phys Lipids 2009;157:1–11. [265] Gotoh N, Moroda K, Watanabe H, Yoshinaga K, Tanaka M, Mizobe H, et al. Metab-
[256] Grevengoed TJ, Klett EL, Coleman RA. Acyl-CoA metabolism and partitioning. Annu olism of odd-numbered fatty acids and even-numbered fatty acids in mouse. J Oleo
Rev Nutr 2014;34:1–30. Sci 2008;57:293–9.
[257] Watkins PA, Maiguel D, Jia Z, Pevsner J. Evidence for 26 distinct acyl-coenzyme a [266] Hajra AK, Radin NS. Biosynthesis of the cerebroside odd-numbered fatty acids. J
synthetase genes in the human genome. J Lipid Res 2007;48:2736–50. Lipid Res 1962;3:327–32.
[258] Ohkuni A, Ohno Y, Kihara A. Identification of acyl-CoA synthetases involved in the [267] Mead JF, Levis GM. A 1 carbon degradation of the long chain fatty acids of brain
mammalian sphingosine 1-phosphate metabolic pathway. Biochem Biophys Res sphingolipids. J Biol Chem 1963;238:1634–6.
Commun 2013;442:195–201. [268] Alderson NL, Rembiesa BM, Walla MD, Bielawska A, Bielawski J, Hama H. The
[259] Mao C, Xu R, Bielawska A, Obeid LM. Cloning of an alkaline ceramidase from Sac- human FA2H gene encodes a fatty acid 2-hydroxylase. J Biol Chem 2004;279:
charomyces cerevisiae. An enzyme with reverse (CoA-independent) ceramide syn- 48562–8.
thase activity. J Biol Chem 2000;275:6876–84. [269] Edvardson S, Hama H, Shaag A, Gomori JM, Berger I, Soffer D, et al. Mutations in the
[260] Mao C, Xu R, Bielawska A, Szulc ZM, Obeid LM. Cloning and characterization of a fatty acid 2-hydroxylase gene are associated with leukodystrophy with spastic
Saccharomyces cerevisiae alkaline ceramidase with specificity for dihydroceramide. paraparesis and dystonia. Am J Hum Genet 2008;83:643–8.
J Biol Chem 2000;275:31369–78. [270] Dick KJ, Eckhardt M, Paisan-Ruiz C, Alshehhi AA, Proukakis C, Sibtain NA, et al. Mu-
[261] Tani M, Kihara A, Igarashi Y. Rescue of cell growth by sphingosine with disruption tation of FA2H underlies a complicated form of hereditary spastic paraplegia
of lipid microdomain formation in Saccharomyces cerevisiae deficient in (SPG35). Hum Mutat 2010;31:E1251–60.
sphingolipid biosynthesis. Biochem J 2006;394:237–42. [271] Potter KA, Kern MJ, Fullbright G, Bielawski J, Scherer SS, Yum SW, et al. Central ner-
[262] Foulon V, Sniekers M, Huysmans E, Asselberghs S, Mahieu V, Mannaerts GP, et al. vous system dysfunction in a mouse model of Fa2h deficiency. Glia 2011;59:
Breakdown of 2-hydroxylated straight chain fatty acids via peroxisomal 2- 1009–21.
hydroxyphytanoyl-CoA lyase: a revised pathway for the α-oxidation of straight [272] Maier H, Meixner M, Hartmann D, Sandhoff R, Wang-Eckhardt L, Zoller I, et al. Nor-
chain fatty acids. J Biol Chem 2005;280:9802–12. mal fur development and sebum production depends on fatty acid 2-hydroxylase
[263] Hama H. Fatty acid 2-hydroxylation in mammalian sphingolipid biology. Biochim expression in sebaceous glands. J Biol Chem 2011;286:25922–34.
Biophys Acta 1801;2010:405–14.

You might also like