ArticlePDF Available

Elimination of a Hydroxyl Group in FTY720 Dramatically Improves the Phosphorylation Rate

Authors:

Abstract and Figures

The new immunosuppressant FTY720 (fingolimod), an analog of the endogenous lipid sphingosine, induces transient lymphopenia through the sequestration of lymphocytes in secondary lymphoid organs. Phosphorylation of FTY720 by sphingosine kinase 2 (SphK2) yields the active metabolite FTY720-phosphate (FTY-P), which induces lymphopenia through agonism of the sphingosine 1-phosphate receptor S1P(1) on endothelial cells and lymphocytes. Dephosphorylation of circulating FTY-P creates an equilibrium between FTY720 and its phosphate, and results with human patients indicate that phosphorylation of FTY720 could be rate limiting for efficacy. We report that the FTY720 derivative 2-amino-4-(4-heptyloxyphenyl)-2-methylbutanol [AAL(R)] is phosphorylated much more rapidly than FTY720 in cultured human cells and whole blood. The K(cat) for AAL(R) with recombinant SphK2 is 8-fold higher than for FTY720, whereas the K(m) for the two substrates is very similar, indicating that the increased rate of phosphorylation results from faster turnover by SphK2 rather than a higher binding affinity. Consequently, treating cells with AAL(R), but not FTY720, triggers an apoptotic pathway that is dependent on excessive intracellular accumulation of long-chain base phosphates. In agreement with the in vitro results, phosphorylation of AAL(R) is more complete than that of FTY720 in vivo (mice), and AAL(R) is a more potent inducer of lymphopenia. These differences may be magnified in humans, because phosphorylation of FTY720 is much less efficient in humans compared with rodents. Our results suggest that AAL(R) is a better tool than FTY720 for in vivo studies with S1P analogs and would probably be a more effective immunosuppressant than FTY720.
Content may be subject to copyright.
Elimination of a Hydroxyl Group in FTY720 Dramatically
Improves the Phosphorylation Rate
Eve Jary, Thomas Bee, Scott R. Walker, Sung-Kee Chung, Kyung-Chang Seo,
Jonathan C. Morris, and Anthony S. Don
Lowy Cancer Research Centre and Prince of Wales Clinical School, Faculty of Medicine (E.J., T.B., A.S.D.), and Department of
Chemistry (S.R.W., J.C.M.), University of New South Wales, Sydney, Australia; and Department of Chemistry, Pohang
University of Science & Technology, Pohang, Korea (S.K.C., K.C.S.)
Received March 18, 2010; accepted July 7, 2010
ABSTRACT
The new immunosuppressant FTY720 (fingolimod), an analog
of the endogenous lipid sphingosine, induces transient
lymphopenia through the sequestration of lymphocytes in sec-
ondary lymphoid organs. Phosphorylation of FTY720 by sphin-
gosine kinase 2 (SphK2) yields the active metabolite FTY720-
phosphate (FTY-P), which induces lymphopenia through
agonism of the sphingosine 1-phosphate receptor S1P
1
on
endothelial cells and lymphocytes. Dephosphorylation of circu-
lating FTY-P creates an equilibrium between FTY720 and its
phosphate, and results with human patients indicate that phos-
phorylation of FTY720 could be rate limiting for efficacy. We
report that the FTY720 derivative 2-amino-4-(4-heptyloxyphe-
nyl)-2-methylbutanol [AAL(R)] is phosphorylated much more
rapidly than FTY720 in cultured human cells and whole blood.
The K
cat
for AAL(R) with recombinant SphK2 is 8-fold higher
than for FTY720, whereas the K
m
for the two substrates is very
similar, indicating that the increased rate of phosphorylation
results from faster turnover by SphK2 rather than a higher
binding affinity. Consequently, treating cells with AAL(R), but
not FTY720, triggers an apoptotic pathway that is dependent
on excessive intracellular accumulation of long-chain base
phosphates. In agreement with the in vitro results, phosphory-
lation of AAL(R) is more complete than that of FTY720 in vivo
(mice), and AAL(R) is a more potent inducer of lymphopenia.
These differences may be magnified in humans, because phos-
phorylation of FTY720 is much less efficient in humans com-
pared with rodents. Our results suggest that AAL(R) is a better
tool than FTY720 for in vivo studies with S1P analogs and
would probably be a more effective immunosuppressant than
FTY720.
Introduction
FTY720 (fingolimod) is a new type of immunosuppressant
that induces a transient, reversible lymphopenia by trapping
lymphocytes in the secondary lymphoid organs and thereby
keeping them out of the circulation. This mode of immuno-
suppression is unique among pharmacological immunosup-
pressants and has made FTY720 the subject of intense inter-
est, from both a therapeutic and a mechanistic/physiological
perspective. Lymphopenia induced by FTY720 is dependent
on the phosphorylation of the compound by sphingosine ki-
nase 2 (SphK2) (Billich et al., 2003; Zemann et al., 2006). The
phosphorylated compound acts as an agonist at four of the
five sphingosine 1-phosphate (S1P) receptors, a family of
G-protein-coupled receptors that respond to extracellular
S1P (Brinkmann et al., 2002; Mandala et al., 2002). Activa-
tion (agonism) of the S1P
1
receptor is responsible for seques-
tration of T cells in the peripheral lymphoid organs, demon-
strated with the observations that a range of S1P
1
-selective
agonists can induce lymphopenia, that this is reversible with
an S1P
1
antagonist, and that S1P
1
-deficient lymphocytes are
resistant to the effects of FTY720 (Matloubian et al., 2004;
Pan et al., 2006; Sanna et al., 2006). Two models have been
put forward to explain exactly how S1P
1
agonists induce
lymphopenia (Brinkmann, 2007; Rosen et al., 2008): in one
model activation of S1P
1
receptors on endothelial cells ex-
This project was supported by the Cancer Institute New South Wales
[Grant 08/ECF/1-03]; the Australian Research Council [Grant DP0770653];
and the National Health and Medical Research Council, Australia [Fellowship
300606]. Work at POSTECH, Korea, was supported by the BK21 program and
the Korea Science and Engineering Foundation BT-Glycobiology Program
[Grant 200402087].
Article, publication date, and citation information can be found at
http://molpharm.aspetjournals.org.
doi:10.1124/mol.110.064873.
ABBREVIATIONS: FTY720, fingolimod; SphK, sphingosine kinase; S1P, sphingosine 1-phosphate; FTY-P, FTY720-phosphate; AAL(R), (R)-2-
amino-4-(4-heptyloxyphenyl)-2-methylbutanol; AAL(S), (S)-2-amino-4-(4-heptyloxyphenyl)-2-methylbutanol; AAL-P, phosphorylated (R)-2-amino-
4-(4-heptyloxyphenyl)-2-methylbutanol; dhSph, dihydrosphingosine; FBS, fetal bovine serum; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetra
zolium; PI, propidium iodide; PCR, polymerase chain reaction; siRNA, small interfering RNA; LC, liquid chromatography; TLC, thin-layer
chromatography; HMEC, human microvascular endothelial cell.
0026-895X/10/7804-685–692$20.00
MOLECULAR PHARMACOLOGY Vol. 78, No. 4
Copyright © 2010 The American Society for Pharmacology and Experimental Therapeutics 64873/3624060
Mol Pharmacol 78:685–692, 2010 Printed in U.S.A.
685
at ASPET Journals on December 31, 2015molpharm.aspetjournals.orgDownloaded from
posed to the blood or lymph results in the closure of endothe-
lial gates through which lymphocytes exit the lymph nodes;
the other model invokes lymphocyte migration from the low
S1P environment of the lymph nodes toward the higher S1P
concentration of the blood or lymph, requiring stimulation of
their S1P
1
receptors. In a simple interpretation of this model,
the presence of FTY720-phosphate (FTY-P) in lymph nodes
disrupts this gradient. However, another interpretation is
derived from the observation that FTY-P acts as a superago-
nist of S1P
1
, promoting internalization and degradation of
the receptor. This impairs the ability of lymphocytes to re-
spond to the proposed S1P gradient (Gonzalez-Cabrera et al.,
2007; Oo et al., 2007).
S1P receptor modulating compounds such as FTY-P have
found application in a wide variety of experimental settings,
which include immunosuppression during organ transplant
(Pan et al., 2006; Brinkmann, 2007), treatment of autoim-
mune conditions (Fujino et al., 2003; Maki et al., 2005),
recovery after ischemia/reperfusion injury (Hofmann et al.,
2009), and as a means of increasing endothelial barrier func-
tion (Sanna et al., 2006). In the clinic, there have been trials
of FTY720 in patients undergoing kidney transplant and
patients with multiple sclerosis (Brinkmann, 2007). The tri-
als in patients undergoing transplant failed to show any
improvement in efficacy over the current standard of care,
but the compound has showed great promise in phase III
trials in patients with relapsing-remitting multiple sclerosis
(Cohen et al., 2010; Kappos et al., 2010). Fewer relapses were
reported with FTY720 than with the current treatment, in-
tramuscular interferon
(Cohen et al., 2010).
The FTY720 analog AAL(R) has been used in a number of
studies, because its chiral enantiomer AAL(S) is not a sub-
strate for SphK2 and therefore acts as a useful control for
effects of the compound that are not attributed to its phos-
phorylation (Kiuchi et al., 2000; Brinkmann et al., 2002; Don
et al., 2007). Because FTY720 and AAL(R) are very similar
compounds, they have been used interchangeably. In this
study, we showed that AAL(R) is a much better substrate for
SphK2 than FTY720, which translates into a faster rate of
phosphorylation by cultured cells and in whole blood and
almost complete phosphorylation in living mice. Phosphory-
lation of FTY720 occurs much more rapidly in rodent than in
human blood, suggesting that AAL(R) would prove signifi-
cantly more effective than FTY720 as a sphingosine 1-phos-
phate receptor agonist in humans.
Materials and Methods
Materials. FTY720 was purchased from Millipore Bioscience Re-
search Reagents (Temecula, CA), whereas AAL(R) was a gift from
Professor Hugh Rosen (The Scripps Research Institute, La Jolla,
CA). AAL-P to use as a standard for mass spectrometry was prepared
by chemical phosphorylation: The amino group of AAL was protected
(Boc
2
O, NaHCO
3
, 56%), then reaction with N,N-diisopropyl phos-
phoramidite dichloride and 5-ethylthio-1H-tetrazole, followed by ox-
idation with hydrogen peroxide, gave protected AAL-P in 19% yield.
The compound was deprotected with trifluoroacetic acid. Dihy-
drosphingosine (dhSph) was purchased from Avanti Polar Lipids
(Alabaster, AL). Synthesis of 3-deoxy-dhSph has been reported pre-
viously (Lim et al., 2004).
Cell Culture and Viability Assays. Jurkat cells and primary
splenocytes were cultured in RPMI 1640 medium supplemented with
10% fetal bovine serum (FBS), 2 mM L-glutamine, and penicillin/
streptomycin solution. The human microvascular endothelial cell
line HMEC-1 (Ades et al., 1992) was cultured in MCDB131 medium
(Invitrogen) supplemented with 10% FBS, glutamine, and antibiot-
ics. Rat splenocytes were isolated by crushing the spleen between
frosted glass slides and filtering through a 70
M filter, followed by
two rounds of red cell lysis in 0.17 M NH
4
Cl, 10 mM NaHCO
3
, and
0.1 mM EDTA for 5 min on ice. Isolated splenocytes were resus-
pended at a density of 1.5 10
6
viable cells/ml in complete RPMI
medium, cultured in the presence of AAL(R), AAL(S), or FTY720 for
20 h, then stained with propidium iodide (PI) for flow cytometry. For
MTT assays, cells were cultured in 96-well plates, using 0.1 ml of
medium per well. Ten microliters of 0.5% (w/v) MTT reagent (Sigma,
St. Louis, MO) in PBS was added to each well, and cells were
returned to the incubator for 2 h. MTT was solubilized by adding 0.1
ml of 10% SDS/10 mM HCl to each well and shaking overnight, and
absorbance was read at 650 nM. Alternatively, viability was assessed
by flow cytometry: cells were resuspended in 100
lof20mM
HEPES, pH 7.4, 150 mM NaCl, and 2.5 mM CaCl
2
and incubated for
15 min on ice with 2
l of Annexin V-allophycocyanin and 1
g/ml PI,
then subjected to flow cytometry.
siRNA Treatment of HeLa Cells and Real-Time Quantita-
tive PCR. Cells were transfected in six-well plates, in 2 ml of
OptiMEM I medium, using siRNA molecules purchased from
QIAGEN at a final concentration of 100 nM. siRNAs were premixed
in 200
l of OptiMEM with 4
l of lipofectamine 2000 (Invitrogen) for
30 min, then added to the cells for 6 h, after which the medium was
replaced with standard growth medium. On the day after transfec-
tion, the cells were detached and reseeded into a 96-well plate at a
density of 10
4
cells/well for MTT assay. Cells were treated with
FTY720 or AAL(R) at 48 h after transfection, and viability was
assayed with MTT reagent at 72 h after transfection. Real-time PCR
was used to measure transcript levels, using the following primers
taken from PrimerBank (Spandidos et al., 2010): SphK1: fwd, AG-
GCTGAAATCTCCTTCACGC; rev, GTCTCCAGACATGACCAC-
CAG; SphK2: fwd: GCTGCTGCGCCTTTTCTTG; rev, CCTGTAGCG-
GCCCATACTC; and glyceraldehyde 3-phosphate dehydrogenase:
fwd, TGTTGCCATCAATGACCCCTT; rev, CTCCACGACGTACT-
CAGCG. RNA was prepared with an RNEasy Mini Kit (QIAGEN);
cDNA was prepared with Moloney murine leukemia virus reverse
transcriptase (Invitrogen); and a SYBR Green with ROX Kit (Invitro-
gen) was used for quantitative PCR, on an Mx3000 cycler (Strata-
gene, La Jolla, CA).
Assays of Compound Phosphorylation In Vitro. Jurkat cells
were cultured for2hinmedium containing 5
M FTY720 or AAL(R),
in triplicate, at a density of 4 10
5
cells/ml. The cells were then
pelleted and resuspended at the same density in fresh medium.
Samples (0.4 ml) were removed from the culture at the times indi-
cated in Fig. 3, snap-frozen, and stored at 80°C. Samples were
extracted with ethyl acetate/isopropanol (Bielawski et al., 2006). In
total, the culture medium was extracted four times with ethyl ace-
tate/isopropanol, twice under acidic conditions. The four organic
extracts were combined, dried under vacuum, and resuspended in
100
l of 80% methanol/20% water (mobile phase for LC). Lipids
were quantified by LC-tandem mass spectrometry, using a C8 col-
umn coupled to a Thermo Quantum TSQ mass spectrometer
(Thermo Fisher Scientific, Waltham, MA) operating in positive ion
multiple reaction monitoring mode. The compounds were separated
with a gradient of 80% methanol/20% water increasing to 85.5%
methanol over 5 min. Precursor and product ion m/zvalues were as
follows: FTY720, 308.3 and 255.1; FTY720-P, 388.0 and 255.1;
AAL(R), 394.0 and 161.1; AAL-P, 374.1 and 161.1.
To assay phosphorylation of compounds in whole blood, human or
rat blood was collected into heparin-coated tubes, then mixed 1:1
with RPMI 1640 medium. One nanomole of FTY720 or AAL(R) was
added directly to 250
l of blood/RPMI 1640 mix and incubated at
35°C for the indicated times. Reactions were stopped with the addi-
tion of 1 ml of ice-cold methanol, and the mixture was cleared by
centrifuging at 21,000gfor 15 min. The insoluble pellets were re-
686 Jary et al.
at ASPET Journals on December 31, 2015molpharm.aspetjournals.orgDownloaded from
extracted by sonicating in 1 ml of methanol. The supernatants from
both steps were combined in 4 ml of glass tubes, dried down in a
SpeedVac SC210 (Thermo Fisher Scientific), and the extracts were
resuspended in 200
l of 80% methanol/20% water (LC mobile
phase). Extraction efficiency was determined by spiking blood with
compounds, then immediately extracting.
Sphingosine Kinase Assays. The radioactive kinase assays
were based on published methods (Olivera et al., 2000; Siow and
Wattenberg, 2007). Kinase assays were set up in 50 mM Tris, pH 7.4,
150 mM NaCl, 10 mM MgCl
2
, 1 mM DTT, 2 mM ATP, 0.1% fatty
acid-free BSA, and 5
Ci/reaction radiolabeled [
32
P]ATP (PerkinElmer
Life and Analytical Sciences). Reactions (0.1 ml) were started with
the addition of recombinant human SphK2, produced in insect cells
(BIOMOL Research Laboratories, Plymouth Meeting, PA). The final
enzyme concentrations were 0.2
g/ml for dhSph and 3-deoxy-dhSph
and 1
g/ml for AAL(R) and FTY720. Reactions were run for 30 min
at 35°C for dhSph, 3-deoxy-dhSph, and AAL(R) and for 150 min for
FTY720. Note that the different enzyme concentrations and times
were used to ensure that enzyme, and not available substrate, was
rate limiting. Reactions were stopped with the addition of 350
lof
methanol/HCl (150:1), followed by 250
l of 2M KCl, and 350
lof
chloroform. Tubes were vortexed, then spun in a refrigerated Mi-
crofuge (Beckman Coulter, Fullerton, CA) at 14,000 rpm to resolve
the phases. The upper aqueous phase was discarded, and 4
lofthe
(lower) organic phase was spotted onto Silica Gel 60 TLC plates
(Fluka, Buchs, Switzerland). TLC plates were resolved in butanol/
acetic acid/water (3:1:1), then exposed to Fuji Imaging Plates and
imaged by filmless autoradiographic analysis with a Fuji FLA7000
(Fujifilm, Tokyo, Japan). The concentration of product in each spot
was derived from a standard curve constructed with the [
32
P]ATP
reaction mix.
Lipid Phosphatase Assay. To prepare radiolabeled FTY-P and
AAL-P, solutions of 50
M FTY720 or AAL(R) were phosphorylated
in kinase assay buffer containing 10
Ci/400
l of reaction [
32
P]ATP,
for4hat35°C, using 3.75
g/ml (for FTY720) or 0.75
g/ml [for
AAL(R)] recombinant SphK2. Reactions were stopped and extracted
with addition of 400
l of methanol, 40
l of 3 M NaOH, and 400
l
of chloroform. Tubes were vortexed, phases were separated by cen-
trifugation, and the upper aqueous phase, containing the radiola-
beled phosphates, was transferred to a new tube. This aqueous
extract was re-extracted by adding 80
l of concentrated HCl and 400
l of chloroform, this time discarding the aqueous phase and retain-
ing the lower organic phase. This method effectively separates the
sphingoid bases from their phosphates (Maceyka et al., 2007). The
organic extract was dried down and resuspended in 400
lof50mM
Tris, pH 7.4, 150 mM NaCl, and 0.1% fatty acid free BSA, and the
concentration of the radiolabeled phosphate was measured by resolv-
ing the resuspended compound on TLC and quantification of FTY-P
or AAL-P spots by filmless autoradiographic analysis.
To assay dephosphorylation, HMEC-1 cells were seeded in a 24-
well plate at a density of 2 10
5
cells/well. On the following day, the
medium was replaced with 0.3 ml of fresh growth medium containing
100 nM radiolabeled FTY-P or AAL-P. Samples (2.5
l) were re-
moved at indicated times and spotted onto a TLC plate, then resolved
and imaged as described above.
In Vivo Measurement of Compounds and Circulating Lym-
phocytes. AAL(R) or FTY720 were administered by intraperitoneal
injection, in 0.1 ml of sterile water, to groups of four C57BL6 mice
(per treatment). Mice were euthanized, and blood was drawn by
cardiac puncture 18 h after dosing. A 0.1-ml aliquot of blood from
each mouse receiving 0.3 mg/kg AAL(R) or FTY720 was immediately
mixed with 0.4 ml of ice-cold methanol, and the samples were pro-
cessed for mass spectrometry as described above. To assay the pro-
portion of T cells in the blood, 0.3-ml blood samples were first
subjected to three rounds of red cell lysis (each 5 min at room
temperature) in 0.17 M NH
4
Cl, 10 mM NaHCO
3
, and 0.1 mM EDTA.
The resulting leukocytes were then incubated for 30 min with a 1:100
dilution of both anti-mouse CD4-PE and anti-mouse CD8-eFluor450
(eBioscience, San Diego, CA) in PBS/2% FBS. Cells were washed,
then fixed for 10 min at room temperature with 1% paraformalde-
hyde in PBS, washed once more, and analyzed the following day
using a FACSCanto II flow cytometer (BD Biosciences, San Jose, CA)
and FlowJo software (TreeStar Inc., Ashland, OR). Total T cells
shown are the sum of CD4- and CD8-positive cells. These experi-
ments were approved by the Animal Care and Ethics Committee of
the University of New South Wales.
Results
AAL(R) but Not FTY720 Treatment Triggers SphK2-
Dependent Cell Death. We have shown previously that
phosphorylation of AAL(R) by SphK2 is required for this
compound to induce a loss of viability in cultured murine
splenocytes, based on two observations: first, AAL(R) was
much more efficient than its nonphosphorylatable enantio-
mer, AAL(S), at inducing loss of viability; second, splenocytes
derived from SphK2 knockout mice were resistant to AAL(R)
(Don et al., 2007). These findings led us to propose that a
specific apoptotic response is triggered by excessive intracel-
lular accumulation of AAL-P. To our surprise, we have found
Fig. 1. AAL(R) but not FTY720 triggers a SphK2-dependent apoptosis
pathway in lymphocytes. A, viability of mouse splenocytes incubated for
24 h with FTY720 (), AAL(R) (f), or AAL(S) (E) was assessed by PI
exclusion. Proportion of viable cells was normalized relative to vehicle-
treated. B, viability of Jurkat cells treated for 24 h with FTY720 (),
AAL(R) (f), or AAL(S) (E) was assessed by annexin V/PI staining. Non-
viable cells are those that were positive for annexin V, PI, or both. C, MTT
assay was used to asses viability of Jurkat cells (closed symbols) or the
SphK2-deficient Jurkat derivative cell line SBR1 (open symbols) (Don et
al., 2007), after a 20-h treatment with FTY720 (circles) or AAL(R)
(squares). D, Jurkat or SBR1 cells were treated for 24 h with 0 or 8
M
AAL(R) or FTY720. Viability was assessed by annexin V/PI staining. All
results shown are the combined results of three separate experiments,
each consisting of triplicate treatments (i.e., n9 per data point).
Two-way analysis of variance with Bonferroni post test was used to
determine the statistical significance of differences between AAL(R) and
both FTY720 and AAL(S) (in A and B) and between AAL(R)-treated
Jurkat and SBR1 cells (in C and D); ,P0.05; ⴱⴱ,P0.01; ⴱⴱⴱ,P
0.0001.
Deoxy FTY720 Derivative Is Much More Rapidly Phosphorylated 687
at ASPET Journals on December 31, 2015molpharm.aspetjournals.orgDownloaded from
that FTY720 is much less potent than AAL(R), and equipo-
tent with AAL(S), at inducing loss of viability in cultured
mouse splenocytes (Fig. 1A). FTY720 was also less efficient
than AAL(R) at inducing apoptosis in the Jurkat T-lympho-
blast cell line, at concentrations below 10
M (Fig. 1, B and
C). As observed previously (Don et al., 2007), the SphK2-
deficient Jurkat cell line SBR1 was resistant to apoptosis
induced with AAL(R). However, these cells were not resistant
to FTY720 (Fig. 1, C and D). These results indicated that a
SphK2-dependent apoptotic pathway is triggered by treating
cells with AAL(R), but not FTY720. At higher concentrations,
apoptosis induced with AAL(R) becomes SphK2-indepen-
dent, suggesting activation of a second apoptotic pathway,
which is the same as that triggered by treating cells with
FTY720 or AAL(S). Similar results were seen in HeLa cells
pretreated with siRNA to SphK2, then treated for 24 h with
AAL(R) or FTY720 (Fig. 2). As a potential explanation for
why AAL(R) but not FTY720 triggers a SphK2-dependent
apoptotic response, we investigated whether AAL-P accumu-
lates inside cells to a greater extent than FTY-P.
AAL(R) Is More Rapidly Phosphorylated than
FTY720. We found that AAL(R) is phosphorylated much
more rapidly than FTY720 by cultured Jurkat cells, using
LC-tandem mass spectrometry (Fig. 3A). We therefore com-
pared the phosphorylation rate for these compounds in whole
blood, which is rich in sphingosine kinase 2 activity (Billich
et al., 2003). The rate of phosphorylation in human blood was
8.9-fold faster with AAL(R) than with FTY720 as substrate
(Fig. 3B and Table 1). Both compounds were phosphorylated
much more rapidly in rat blood, compared with human blood:
the rate of phosphorylation was 35-fold higher for FTY720
and 27-fold higher for AAL(R), in rat versus human blood.
The more rapid conversion of FTY720 by mouse or rat blood,
compared with human blood, has been reported previously,
although without quantification of the difference in rate (Bil-
lich et al., 2003). The difference was attributed to the higher
SphK2 activity of rodent blood compared with human blood,
rather than any difference in the rate of FTY720 phosphor-
ylation by rodent versus human SphK2.
We next investigated whether AAL(R) is a better substrate
for SphK2 than FTY720, using an in vitro reaction with
recombinant human SphK2 (Fig. 4A). The enzyme turnover
rate was 7.9 times higher with AAL(R) as the substrate,
whereas the ability of the enzyme to bind the substrate
(measured as K
m
) was very similar (Table 2). This difference
in phosphorylation rate is very similar to that observed with
whole human blood [8.9-fold higher with AAL(R) as sub-
strate]. Similar results were seen when lysates of human
embryonic kidney 293 cells overexpressing human SphK2
were used as the source of SphK2 activity: the turnover rate
was 14 times higher with AAL(R) than with FTY720 as the
substrate, whereas the K
m
was similar [7.4
M for AAL(R);
13.2
M for FTY720]. These results indicate that although
there seems to be no difference in the affinity of SphK2 for
the two substrates, the active site is better able to turn over
AAL(R) than FTY720.
The key structural difference between FTY720 and AAL(R)
is a hydroxymethyl to methyl substitution on the quaternary
Fig. 2. AAL(R) but not FTY720 triggers a SphK2-dependent apoptosis pathway in HeLa cells. HeLa cells were pretreated for 48 h with two different
siRNA molecules targeting SphK2 (or , universal negative control siRNA (E), or lipofectamine only (f), then incubated for 24 h in the presence
of AAL(R) (A) or FTY720 (B). Viability was determined by MTT assay and normalized to vehicle control-treated cells. Results shown are the combined
results of two separate experiments, each consisting of triplicate treatments (i.e., n6 per data point). C, expression of SphK2 (closed bars) and, as
a control, SphK1 (open bars) was measured 48 h after siRNA treatment, using real-time PCR. Expression was normalized relative to glyceraldehyde
3-phosphate dehydrogenase (G3PDH) and is expressed proportional to the lipofectamine only control (Mock). Results shown are mean and S.E. of four
data points, derived from two separate experiments for each siRNA. Two-way analysis of variance with Bonferroni post test was used to determine
the statistical significance of differences between negative control and both SK2-specific siRNAs (A and B) or all siRNAs compared with mock
transfected (C); ,P0.05; ⴱⴱ,P0.01; ⴱⴱⴱ,P0.0001.
Fig. 3. AAL(R) is more rapidly phosphorylated than
FTY720. A, phosphorylation of AAL(R) or FTY720 by cul-
tured Jurkat cells was measured over time, by quantifying
the amount of AAL-P (f) or FTY-P (F) in both cells and
culture medium. Results are mean and S.E. derived from
triplicate cell treatments and representative of two inde-
pendent experiments. B, formation of AAL-P (squares) or
FTY-P (circles) in human (solid symbols) or rat (open sym-
bols) blood was measured as a function of time after addi-
tion of 1 nmol of AAL(R) or FTY720 to 125
l of whole
blood, as described under Materials and Methods. Results
shown are combined data from two separate experiments
(n5, rat blood; n6, human blood).
688 Jary et al.
at ASPET Journals on December 31, 2015molpharm.aspetjournals.orgDownloaded from
(second) carbon of the headgroup (Fig. 4C), suggesting that
the presence of this second hydroxyl group interferes with
catalysis or release of the product. In the natural substrates
sphingosine and dhSph, a second hydroxyl group located on
the third carbon of the acyl chain is not accessible for phos-
phorylation by sphingosine kinases. To gain some insight
into whether this 3-OH group influences the phosphorylation
rate or substrate affinity, we determined the Michaelis-Men-
ten kinetics for phosphorylation of dhSph and 3-deoxy-dhSph
(Fig. 4D) by SphK2 (Fig. 4B and table 2). Removal of the
hydroxyl group reduced the enzyme turnover rate and
slightly increased the K
m
, but the effects were not dramatic,
indicating that the 3-OH group plays a minor role in sub-
strate recognition and turnover by SphK2.
FTY720 Is Dephosphorylated Faster than AAL(R).
The steady-state level of FTY-P achieved in living organisms
is a function not only of phosphorylation but also of dephos-
phorylation. A likely candidate organ for dephosphorylation
of circulating FTY-P is the endothelium, which on the other
hand has very little SphK2 activity and is therefore unlikely
to contribute significantly to the compound’s phosphorylation
(Anada et al., 2007). FTY-P is a membrane-impermeant com-
pound, and recent evidence indicates that it may be dephos-
phorylated extracellularly by endothelial lipid phosphate
phosphatases, specifically subtypes 1a and 3 (Mechtcheri-
akova et al., 2007; Yamanaka et al., 2008). To determine
whether there are any differences in the ability of lipid phos-
phatases to dephosphorylate the two compounds, we tested
the ectophosphatase activity of cultured human endothelial
cells toward both FTY-P and AAL-P (Fig. 5). In direct con-
trast to the rate of phosphorylation by SphK2, the rate of
dephosphorylation was faster with FTY-P. Using a one-phase
exponential decay model to fit the data, the difference in
dephosphorylation rate was 1.5, 1.6, and 2.9-fold (faster in
the case of FTY-P) in three separate experiments, and was
statistically significant (P0.001, sum-of-squares test).
AAL(R) Is More Fully Phosphorylated In Vivo. Our in
vitro results indicated that AAL(R) should be more com-
pletely phosphorylated than FTY720, at steady state, in vivo.
To test this, we administered a single 0.3 mg/kg dose of
AAL(R) or FTY720 to mice, and measured the amount of
AAL(R) and AAL-P, or FTY720 and FTY-P, in the blood 18 h
later (Fig. 6). For AAL(R), 3.7 1.3% of the compound
remained unphosphorylated (i.e., 96% phosphorylated),
whereas for FTY720, this was 19.1 3.1% (81% phosphory-
lated), a statistically significant difference (P0.001, un-
paired ttest). Measurements of FTY720 phosphorylation in
mice at 2 h (82%) and 6 h (83%) indicated that a steady-state
balance was rapidly achieved, and although the total amount
of compound in blood declined over time, the proportion of
phosphorylated compound remained steady.
Discussion
Induction of lymphopenia with FTY720 is dependent on
stimulation of the S1P
1
receptor by FTY-P. The potency
(EC
50
) and efficacy (E
max
) for AAL-P, FTY-P, and S1P on the
human S1P
1
receptor are essentially identical (Brinkmann et
al., 2002). Despite this, two publications have reported that
the EC
50
for induction of lymphopenia in rats is three times
lower with AAL(R) than with FTY720 (Kiuchi et al., 2000;
Ho¨ genauer et al., 2008). Our own measurements of blood T
cells in mice confirm the greater potency of AAL(R) as an
inducer of lymphopenia: the EC
50
for depletion of blood T
cells after 18 h was 27
g/kg with AAL(R) and 51
g/kg with
FTY720 (n4, P0.036, by sum-of-squares F test). These
results support the conclusion that the more rapid phosphor-
ylation of AAL(R) in vitro translates into a greater proportion
of phosphorylated compound in vivo, and a consequent in-
crease in potency. Although AAL(R) is a better substrate
than FTY720 for phosphorylation, dephosphorylation of
FTY-P by human endothelial cells was faster than for AAL-P
Fig. 4. Turnover rate by SphK2 is higher with AAL(R) than
with FTY720. A, phosphorylation of AAL(R) (f) or FTY720
(E) by recombinant human SphK2, as a function of sub-
strate concentration. B, phosphorylation of dhSph (f)or
3-deoxy-dhSph (E) by recombinant human SphK2 as a
function of substrate concentration. Michaelis-Menten
curves were fitted to 12 data points with Prism (GraphPad
Software, San Diego, CA), and V
max
and K
m
values are
shown in Table 2. C, structures for FTY720 and AAL(R). D,
structures for dhSph and 3-deoxy-dhSph.
TABLE 1
Rate of FTY720 and AAL(R) phosphorylation in whole blood
Phosphorylation rates were calculated using only the linear portion of the phosphor-
ylation curves shown in Fig. 3B. Data are presented as mean S.E.
Blood FTY720 AAL(R)
nmol product/h/ml of blood
Human 0.056 0.001 0.496 0.014
Rat 1.93 0.040 13.3 1.13
Deoxy FTY720 Derivative Is Much More Rapidly Phosphorylated 689
at ASPET Journals on December 31, 2015molpharm.aspetjournals.orgDownloaded from
(Fig. 5), suggesting that FTY-P is a better substrate for lipid
phosphate phosphatases. This would further exacerbate the
difference between the two compounds in terms of steady-
state phosphorylation.
The more rapid phosphorylation of AAL(R) by SphK2 is
supported by our initial observation that AAL(R) induces a
SphK2-dependent apoptosis pathway, whereas FTY720 does
not. The apoptotic response seems to be triggered by exces-
sive intracellular accumulation of long-chain base phos-
phates such as AAL-P (Don et al., 2007), cis-4-methylsphin-
gosine 1-phosphate (van Echten-Deckert et al., 1997), or, in
yeast, phytosphingosine 1-phosphate (Zhang et al., 2001).
Further experiments have shown that apoptosis triggered by
AAL(R) proceeds through mitochondrial depolarization (not
shown), but the precise nature of the intracellular target for
AAL-P that triggers apoptosis is currently unknown. AAL(R)
and FTY720 both trigger a SphK2-independent apoptotic
pathway at concentrations of approximately 10
M. Apopto-
sis induced with FTY720 forms the basis for its anticancer
properties, and is believed to occur through activation of the
broad-spectrum serine/threonine protein phosphatase 2A, at
least in leukemia cells (Matsuoka et al., 2003; Neviani et al.,
2007; Liu et al., 2008). Our results are in agreement with
those of others, who have shown that FTY720 does not need
to be phosphorylated to induce apoptosis in leukemia cells
(Neviani et al., 2007; Liu et al., 2008). Neither the SphK2-
dependent nor the SphK2-independent apoptotic pathway
is relevant to immunosuppression, because these path-
ways are activated at concentrations of the drug that are 1
to 2 orders of magnitude higher than the concentration
required to achieve effective immunosuppression in hu-
mans or rodents.
There are two structural differences between AAL(R) and
FTY720 (Fig. 4C): the introduction of an ether linkage (re-
placing a carbon) between the lipid tail and the aromatic ring
in AAL(R), and elimination of one of the FTY720 hydroxyl
headgroups. It has been shown previously (Kiuchi et al.,
2000) that introduction of the ether linkage into FTY720 does
not improve, or significantly alter, its potency as an inducer
of lymphopenia. We therefore conclude that elimination of
one of the hydroxyl groups improves catalysis by SphK2. The
hydroxyl group on the third carbon of dhSph does not slow
down its phosphorylation by SphK2 relative to 3-deoxy-dh-
Sph (Fig. 4B), indicating that it is the position of the second
hydroxyl group in FTY720 that interferes with catalysis. It is
likely that the presence of two hydroxyl groups in FTY720,
both accessible to the SphK2 catalytic site, interferes with
release of the product or the transfer of phosphate from ATP.
In rodents, a cycle of phosphorylation and dephosphoryla-
tion maintains an equilibrium between FTY720 and FTY-P
in the blood, with 20 to 30% of the compound in the nonphos-
phorylated form (Brinkmann et al., 2002; Mandala et al.,
2002). In the current study, we show that when AAL(R) is
used, the equilibrium is shifted in favor of the phosphate
(Fig. 6), and this gives rise to an increase in potency. For
this reason, AAL(R) is probably superior to FTY720 as a
research tool for determining the effects the sphingosine
1-phosphate receptor agonists on animal physiology and
pathophysiology, especially given the availability of a
chemically identical, nonphosphorylatable control com-
pound in the form of the S-enantiomer. We note that
asymmetric synthesis of AAL(R) or AAL(S) is not difficult
and can be achieved by starting with the chiral headgroup
and adding the lipophilic portion of the molecule to this.
This approach circumvents the need for any chiral separa-
tion (Hinterding et al., 2003).
In human patients, FTY720 phosphorylation seems to be rate
limiting for efficacy. Human blood possesses a much lower in-
trinsic SphK2 activity than rodent blood (Billich et al., 2003),
resulting in a much slower rate of phosphorylation for both
FTY720 and AAL(R) (Fig. 3B). Results with human patients
indicate that the equilibrium between FTY720 and its phos-
phate rests more heavily in favor of dephosphorylation: the
plasma concentration of FTY-P drops below that of FTY720 12
to 24 h after a single 5-mg dose of FTY720; thereafter, FTY-P
declines as a proportion of the FTY720 concentration (Kovarik
et al., 2008, 2009). On this basis, one would predict that AAL(R)
would achieve effective immunosuppression at a significantly
lower dose in humans than FTY720. Lymphopenia in humans
is achieved with doses of FTY720 at or above 1 mg/day. At this
dose, the steady-state FTY720 concentration in plasma reaches
5.7 ng/ml (18.6 nM). More effective lymphopenia is achieved at
2.5 mg/day (steady state FTY720 concentration of 36.5 nM in
plasma) (Kahan et al., 2003; Brinkmann, 2007). At this concen-
tration FTY720 may have effects that are not dependent on its
phosphorylation, such as inhibition of cytosolic phospholipase
A
2
, with consequent inhibition of prostaglandin and prostacy-
clin synthesis (Payne et al., 2007), or inhibition of protein ki-
nase C isoforms (Sensken and Gra¨ ler, 2010).
In summary, in this article we report that the R-enantio-
mer of the FTY720 derivative 2-amino-1,3-propanediol is a
Fig. 5. Dephosphorylation of FTY-P and AAL-P. Radiola-
beled AAL-P (f) or FTY-P (E) was added at 100 nM to the
medium of cultured HMEC-1 cells. The medium was sam-
pled at indicated times and loss of the phosphorylated
substrate from the growth medium was assessed by TLC
and filmless autoradiographic analysis. Image from the
48-h incubation, plus no-cell control, is shown on the right,
and graph shows loss of radiolabeled substrate over time.
Results shown are mean of triplicate incubations and are
representative of three independent experiments. Data
points were fitted to a one phase decay model using Graph-
Pad Prism.
TABLE 2
Michaelis-Menten kinetics for phosphorylation of dhSph, 3-deoxy-
dhSph, FTY720, and AAL(R) by purified recombinant human SphK2
Michaelis-Menten curves were fitted to 12 data points for each compound with the
use of GraphPad Prism. Values shown are best fit values S.E.
K
m
V
max
K
cat
M pmol product/min/
g of enzyme
dhSph 28.8 7.2 1281 131 89.6
3-Deoxy-dhSph 38.9 6.8 781 62.8 54.6
AAL(R) 15.6 2.7 86.8 5.0 6.07
FTY720 13.1 2.3 11.0 0.61 0.77
690 Jary et al.
at ASPET Journals on December 31, 2015molpharm.aspetjournals.orgDownloaded from
much better substrate for SphK2 than FTY720 itself. This
results from a faster enzyme turnover rather than higher
affinity and leads to a significantly more rapid rate of phos-
phorylation in human blood. These results have therapeutic
importance, because the efficacy of FTY720 as an immuno-
suppressant is dependent on the phosphate rather than the
parent compound. First, effective immunosuppression could
be achieved with lower doses of AAL(R) than are needed
with FTY720. Second, use of AAL(R) would reduce the
amount of nonphosphorylated compound in circulation,
thus obviating any effects associated with the nonphosphor-
ylated compound.
Acknowledgments
We thank Russell Pickford, Sonia Bustamante, and Lewis Adler of
the Biomedical Mass Spectrometry Facility at the University of New
South Wales for their assistance with mass spectrometry and Sandra
Spathos for assistance with obtaining rat blood.
References
Ades EW, Candal FJ, Swerlick RA, George VG, Summers S, Bosse DC, and Lawley
TJ (1992) HMEC-1: establishment of an immortalized human microvascular en-
dothelial cell line. J Invest Dermatol 99:683–690.
Anada Y, Igarashi Y, and Kihara A (2007) The immunomodulator FTY720 is phos-
phorylated and released from platelets. Eur J Pharmacol 568:106–111.
Bielawski J, Szulc ZM, Hannun YA, and Bielawska A (2006) Simultaneous quanti-
tative analysis of bioactive sphingolipids by high-performance liquid chromatog-
raphy-tandem mass spectrometry. Methods 39:82–91.
Billich A, Bornancin F, De´ vay P, Mechtcheriakova D, Urtz N, and Baumruker T
(2003) Phosphorylation of the immunomodulatory drug FTY720 by sphingosine
kinases. J Biol Chem 278:47408–47415.
Brinkmann V (2007) Sphingosine 1-phosphate receptors in health and disease:
mechanistic insights from gene deletion studies and reverse pharmacology. Phar-
macol Ther 115:84–105.
Brinkmann V, Davis MD, Heise CE, Albert R, Cottens S, Hof R, Bruns C, Prieschl E,
Baumruker T, Hiestand P, et al. (2002) The immune modulator FTY720 targets
sphingosine 1-phosphate receptors. J Biol Chem 277:21453–21457.
Cohen JA, Barkhof F, Comi G, Hartung HP, Khatri BO, Montalban X, Pelletier J,
Capra R, Gallo P, Izquierdo G, et al. (2010) Oral fingolimod or intramuscular
interferon for relapsing multiple sclerosis. N Engl J Med 362:402–415.
Don AS, Martinez-Lamenca C, Webb WR, Proia RL, Roberts E, and Rosen H (2007)
Essential requirement for sphingosine kinase 2 in a sphingolipid apoptosis path-
way activated by FTY720 analogues. J Biol Chem 282:15833–15842.
Fujino M, Funeshima N, Kitazawa Y, Kimura H, Amemiya H, Suzuki S, and Li XK
(2003) Amelioration of experimental autoimmune encephalomyelitis in Lewis rats
by FTY720 treatment. J Pharmacol Exp Ther 305:70–77.
Gonzalez-Cabrera PJ, Hla T, and Rosen H (2007) Mapping pathways downstream of
sphingosine 1-phosphate subtype 1 by differential chemical perturbation and
proteomics. J Biol Chem 282:7254–7264.
Hinterding K, Cottens S, Albert R, Zecri F, Buehlmayer PB, Spanka C, Brinkmann
V, Nussbaumer P, Ettmayer P, Hoegenauer K, et al. (2003) Synthesis of chiral
analogues of FTY720 and its phosphate. Synthesis-Stuttgart 2003:1667–1670.
Hofmann U, Burkard N, Vogt C, Thoma A, Frantz S, Ertl G, Ritter O, and Bonz A
(2009) Protective effects of sphingosine-1-phosphate receptor agonist treatment
after myocardial ischaemia-reperfusion. Cardiovasc Res 83:285–293.
Ho¨ genauer K, Billich A, Pally C, Streiff M, Wagner T, Welzenbach K, and Nuss-
baumer P (2008) Phosphorylation by sphingosine kinase 2 is essential for in vivo
potency of FTY720 analogues. Chem Med Chem 3:1027–1029.
Kahan BD, Karlix JL, Ferguson RM, Leichtman AB, Mulgaonkar S, Gonwa TA,
Skerjanec A, Schmouder RL, and Chodoff L (2003) Pharmacodynamics, pharma-
cokinetics, and safety of multiple doses of FTY720 in stable renal transplant
patients: a multicenter, randomized, placebo-controlled, phase I study. Transplan-
tation 76:1079–1084.
Kappos L, Radue EW, O’Connor P, Polman C, Hohlfeld R, Calabresi P, Selmaj K,
Agoropoulou C, Leyk M, Zhang-Auberson L, et al. (2010) A placebo-controlled trial
of oral fingolimod in relapsing multiple sclerosis. N Engl J Med 362:387–401.
Kiuchi M, Adachi K, Kohara T, Minoguchi M, Hanano T, Aoki Y, Mishina T, Arita M,
Nakao N, Ohtsuki M, et al. (2000) Synthesis and immunosuppressive activity of
2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols. J Med Chem 43:
2946–2961.
Kovarik JM, Dole K, Riviere GJ, Pommier F, Maton S, Jin Y, Lasseter KC, and
Schmouder RL (2009) Ketoconazole increases fingolimod blood levels in a drug
interaction via CYP4F2 inhibition. J Clin Pharmacol 49:212–218.
Kovarik JM, Slade A, Riviere GJ, Neddermann D, Maton S, Hunt TL, and
Schmouder RL (2008) The ability of atropine to prevent and reverse the negative
chronotropic effect of fingolimod in healthy subjects. Br J Clin Pharmacol 66:199
206.
Lim HS, Park JJ, Ko K, Lee MH, and Chung SK (2004) Syntheses of sphingosine-
1-phosphate analogues and their interaction with EDG/S1P receptors. Bioorg Med
Chem Lett 14:2499–2503.
Liu Q, Zhao X, Frissora F, Ma Y, Santhanam R, Jarjoura D, Lehman A, Perrotti D,
Chen CS, Dalton JT, et al. (2008) FTY720 demonstrates promising preclinical
activity for chronic lymphocytic leukemia and lymphoblastic leukemia/lymphoma.
Blood 111:275–284.
Maceyka M, Milstien S, and Spiegel S (2007) Measurement of mammalian sphin-
gosine-1-phosphate phosphohydrolase activity in vitro and in vivo. Methods Enzy-
mol 434:243–256.
Maki T, Gottschalk R, Ogawa N, and Monaco AP (2005) Prevention and cure of
autoimmune diabetes in nonobese diabetic mice by continuous administration of
FTY720. Transplantation 79:1051–1055.
Mandala S, Hajdu R, Bergstrom J, Quackenbush E, Xie J, Milligan J, Thornton R,
Shei GJ, Card D, Keohane C, et al. (2002) Alteration of lymphocyte trafficking by
sphingosine-1-phosphate receptor agonists. Science 296:346–349.
Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brinkmann V, Allende ML,
Proia RL, and Cyster JG (2004) Lymphocyte egress from thymus and peripheral
lymphoid organs is dependent on S1P receptor 1. Nature 427:355–360.
Matsuoka Y, Nagahara Y, Ikekita M, and Shinomiya T (2003) A novel immunosup-
pressive agent FTY720 induced Akt dephosphorylation in leukemia cells. Br J
Pharmacol 138:1303–1312.
Mechtcheriakova D, Wlachos A, Sobanov J, Bornancin F, Zlabinger G, Baumruker T,
and Billich A (2007) FTY720-phosphate is dephosphorylated by lipid phosphate
phosphatase 3. FEBS Lett 581:3063–3068.
Neviani P, Santhanam R, Oaks JJ, Eiring AM, Notari M, Blaser BW, Liu S, Trotta
R, Muthusamy N, Gambacorti-Passerini C, et al. (2007) FTY720, a new alternative
for treating blast crisis chronic myelogenous leukemia and Philadelphia chromo-
some-positive acute lymphocytic leukemia. J Clin Invest 117:2408–2421.
Olivera A, Barlow KD, and Spiegel S (2000) Assaying sphingosine kinase activity.
Methods Enzymol 311:215–223.
Oo ML, Thangada S, Wu MT, Liu CH, Macdonald TL, Lynch KR, Lin CY, and Hla T
(2007) Immunosuppressive and anti-angiogenic sphingosine 1-phosphate recep-
tor-1 agonists induce ubiquitinylation and proteasomal degradation of the recep-
tor. J Biol Chem 282:9082–9089.
Pan S, Mi Y, Pally C, Beerli C, Chen A, Guerini D, Hinterding K, Nuesslein-
Hildesheim B, Tuntland T, Lefebvre S, et al. (2006) A monoselective sphingosine-
1-phosphate receptor-1 agonist prevents allograft rejection in a stringent rat heart
transplantation model. Chem Biol 13:1227–1234.
Payne SG, Oskeritzian CA, Griffiths R, Subramanian P, Barbour SE, Chalfant CE,
Milstien S, and Spiegel S (2007) The immunosuppressant drug FTY720 inhibits
cytosolic phospholipase A2 independently of sphingosine-1-phosphate receptors.
Blood 109:1077–1085.
Rosen H, Gonzalez-Cabrera P, Marsolais D, Cahalan S, Don AS, and Sanna MG
(2008) Modulating tone: the overture of S1P receptor immunotherapeutics. Immu-
nol Rev 223:221–235.
Sanna MG, Wang SK, Gonzalez-Cabrera PJ, Don A, Marsolais D, Matheu MP, Wei
SH, Parker I, Jo E, Cheng WC, et al. (2006) Enhancement of capillary leakage and
restoration of lymphocyte egress by a chiral S1P1 antagonist in vivo. Nat Chem
Biol 2:434–441.
Sensken SC and Gra¨ ler MH (2010) Down-regulation of S1P1 receptor surface ex-
pression by protein kinase C inhibition. J Biol Chem 285:6298–6307.
Siow DL and Wattenberg BW (2007) An assay system for measuring the acute
production of sphingosine 1-phosphate in intact monolayers. Anal Biochem 371:
184–193.
Spandidos A, Wang X, Wang H, and Seed B (2010) PrimerBank: a resource of human
and mouse PCR primer pairs for gene expression detection and quantification.
Nucleic Acids Res 38:D792–D799.
van Echten-Deckert G, Zschoche A, Ba¨ r T, Schmidt RR, Raths A, Heinemann T, and
Sandhoff K (1997) cis-4-Methylsphingosine decreases sphingolipid biosynthesis by
Fig. 6. AAL(R) is more completely phosphorylated in vivo. Amount of
AAL(R) and AAL-P, or FTY720 and FTY-P, in the blood, 18 h after
intraperitoneal administration of 0.3 mg/kg AAL(R) or FTY720 to
C57BL6 mice (n4). Free base (shaded bars) refers to AAL(R) and
FTY720, whereas base phosphate (clear bars) refers to AAL-P and
FTY-P.
Deoxy FTY720 Derivative Is Much More Rapidly Phosphorylated 691
at ASPET Journals on December 31, 2015molpharm.aspetjournals.orgDownloaded from
specifically interfering with serine palmitoyltransferase activity in primary cul-
tured neurons. J Biol Chem 272:15825–15833.
Yamanaka M, Anada Y, Igarashi Y, and Kihara A (2008) A splicing isoform of LPP1,
LPP1a, exhibits high phosphatase activity toward FTY720 phosphate. Biochem
Biophys Res Commun 375:675–679.
Zemann B, Kinzel B, Mu¨ ller M, Reuschel R, Mechtcheriakova D, Urtz N, Bornancin F,
Baumruker T, and Billich A (2006) Sphingosine kinase type 2 is essential for lym-
phopenia induced by the immunomodulatory drug FTY720. Blood 107:1454–1458.
Zhang X, Skrzypek MS, Lester RL, and Dickson RC (2001) Elevation of endogenous
sphingolipid long-chain base phosphates kills Saccharomyces cerevisiae cells. Curr
Genet 40:221–233.
Address correspondence to: Dr. Anthony Don, Level 2, C25 Lowy Cancer
Institute, Faculty of Medicine, University of New South Wales, Sydney, 2052,
NSW, Australia. E-mail: anthonyd@unsw.edu.au
692 Jary et al.
at ASPET Journals on December 31, 2015molpharm.aspetjournals.orgDownloaded from
... In addition, intracellular S1P can cause autophagy (Liao et al., 2012) and inhibit proliferation (Liao et al., 2007), which in turn, could counteract ASM thickening (Stamatiou et al., 2009). Importantly, in vitro data show that sphingosine analogs can counteract cell amplification and/or survival via SphK2-dependent intracellular accumulation of phosphorylated sphingoid bases (Don et al., 2007;Jary et al., 2010). In agreement with this growing body of evidence, we tested the concept that this class of agents could reverse ASM thickening. ...
... Human primary ASM cells (CC-2576; Lonza, Walkersville, MD, USA) were plated at a density of 10 4 cells per cm 2 in Dulbecco's Modified Eagle Medium containing 10% fetal bovine serum (complete medium) for 24 h, and then incubated with sphingosine analogs, the S1P lyase inhibitor (SM4; Zhao et al., 2015) or the dual S1P receptor (S1P 1−3 ) antagonist (Collison et al., 2013). The cell-impermeant molecule AFD-R can either accumulate intracellularly to exert biological effects via unknown targets (Jary et al., 2010), or be transported into the extracellular space where it can modulate cell surface S1P receptors. ...
... For instance, AAL-R is mainly phosphorylated by SphK2 and intracellular AFD-R accumulation is much faster than the accumulation of other sphingosine analogs (like FTY720). Importantly, this faster conversion relates to the preferential ability of AAL-R to interfere with Jurkat T cell accumulation in vitro, when compared with FTY720 (Jary et al., 2010). In agreement with these observations, we determined that ASM cells express SphK2 and display a nearly 2:1 AFD-R to AAL-R ratio. ...
Article
Full-text available
In asthma, excessive bronchial narrowing associated with thickening of the airway smooth muscle (ASM) causes respiratory distress. Numerous pharmacological agents prevent experimental airway hyperresponsiveness (AHR) when delivered prophylactically. However, most fail to resolve this feature after disease is instated. Although sphingosine analogs are primarily perceived as immune modulators with the ability to prevent experimental asthma, they also influence processes associated with tissue atrophy, supporting the hypothesis that they could interfere with mechanisms sustaining pre-established AHR. We thus assessed the ability of a sphingosine analog (AAL-R) to reverse AHR in a chronic model of asthma. We dissected the pharmacological mechanism of this class of agents using the non-phosphorylatable chiral isomer AAL-S and the pre-phosphorylated form of AAL-R (AFD-R) in vivo and in human ASM cells. We found that a therapeutic course of AAL-R reversed experimental AHR in the methacholine challenge test, which was not replicated by dexamethasone or the non-phosphorylatable isomer AAL-S. AAL-R efficiently interfered with ASM cell proliferation in vitro, supporting the concept that immunomodulation is not necessary to interfere with cellular mechanisms sustaining AHR. Moreover, the sphingosine-1-phosphate lyase inhibitor SM4 and the sphingosine-1-phosphate receptor antagonist VPC23019 failed to inhibit proliferation, indicating that intracellular accumulation of sphingosine-1-phosphate or interference with cell surface S1P1/S1P3 activation, are not sufficient to induce cytostasis. Potent AAL-R-induced cytostasis specifically related to its ability to induce intracellular AFD-R accumulation. Thus, a sphingosine analog that possesses the ability to be phosphorylated in situ interferes with cellular mechanisms that beget AHR.
... For instance, the drug FTY720 (Fingolimod, Gilenya) is currently used for patients with multiple sclerosis [5]. The compound AAL-R is an analog of FTY720 that has been broadly used to study the mechanisms of action of this class of compounds [6][7][8][9]. Sphingosine analogs such as FTY720 and AAL-R are cell-permeant and become phosphorylated intracellularly to become a sphingosine-1-phosphate (S1P) analog. They are then actively exported from the cells. ...
... Accordingly, we show that AAL-R, but not AFD-R, potently induces lymphocyte apoptosis/necrosis in the lung. This is consistent with AAL-R's rapid phosphorylation, low turnover, and potent induction of apoptosis in murine splenocytes through intracellular accumulation of AFD-R [7]. Interestingly, AAL-R-induced apoptosis/necrosis appears to affect CD4 + T cells and B cells, which are crucially involved in allergic airway disease, while having no effect on CD8 + T cells, DCs and the CD90 − CD19 − AF − SSC med-hi granulocyte-enriched fraction. ...
Article
Full-text available
Background In vivo phosphorylation of sphingosine analogs with their ensuing binding and activation of their cell-surface sphingosine-1-phosphate receptors is regarded as the main immunomodulatory mechanism of this new class of drugs. Prophylactic treatment with sphingosine analogs interferes with experimental asthma by impeding the migration of dendritic cells to draining lymph nodes. However, whether these drugs can also alleviate allergic airway inflammation after its onset remains to be determined. Herein, we investigated to which extent and by which mechanisms the sphingosine analog AAL-R interferes with key features of asthma in a murine model during ongoing allergic inflammation induced by Dermatophagoides pteronyssinus.MethodsBALB/c mice were exposed to either D. pteronyssinus or saline, intranasally, once-daily for 10 consecutive days. Mice were treated intratracheally with either AAL-R, its pre-phosphorylated form AFD-R, or the vehicle before every allergen challenge over the last four days, i.e. after the onset of allergic airway inflammation. On day 11, airway responsiveness to methacholine was measured; inflammatory cells and cytokines were quantified in the airways; and the numbers and/or viability of T cells, B cells and dendritic cells were assessed in the lungs and draining lymph nodes.ResultsAAL-R decreased airway hyperresponsiveness induced by D. pteronyssinus by nearly 70%. This was associated with a strong reduction of IL-5 and IL-13 levels in the airways and with a decreased eosinophilic response. Notably, the lung CD4+ T cells were almost entirely eliminated by AAL-R, which concurred with enhanced apoptosis/necrosis in that cell population. This inhibition occurred in the absence of dendritic cell number modulation in draining lymph nodes. On the other hand, the pre-phosphorylated form AFD-R, which preferentially acts on cell-surface sphingosine-1-phosphate receptors, was relatively impotent at enhancing cell death, which led to a less efficient control of T cell and eosinophil responses in the lungs.Conclusion Airway delivery of the non-phosphorylated sphingosine analog, but not its pre-phosphorylated counterpart, is highly efficient at controlling the local T cell response after the onset of allergic airway inflammation. The mechanism appears to involve local induction of lymphocyte apoptosis/necrosis, while mildly affecting dendritic cell and T cell accumulation in draining lymph nodes.
... Although Sphk2 −/− MEFs were not resistant to FTY720, FTY720-P may contribute to FTY720's anti-cancer activity as sensitivity is somewhat correlated to SPHK2 expression (Figure 5a, right). The efficient phosphorylation of SH-RF-177 in vitro (Figure 3c), in cells (Figures 3a and b), and in mice (Figures 3d-f) is consistent with previous reports that (R) stereoisomers of FTY720 analogs are better SPHK2 substrates 20 and with observations that removing the non-phosphorylated hydroxyl group from the aminodiol portion of FTY720 enhances phosphorylation 43 . In sum, our findings suggest that SH-RF-177 represents a novel class of anti-leukemic FTY720 analogs with increased therapeutic potential: compounds that are efficiently phosphorylated but fail to activate S1PRs. ...
Article
The frequency of poor outcomes in relapsed leukemia patients underscores the need for novel therapeutic approaches. The FDA-approved immunosuppressant FTY720 limits leukemia progression by activating protein phosphatase 2A and restricting nutrient access. Unfortunately, FTY720 cannot be re-purposed for use in cancer patients due to on-target toxicity associated with S1P receptor activation at the elevated, anti-neoplastic dose. Here we show that the constrained azacyclic FTY720 analog SH-RF-177 lacks S1P receptor activity but maintains anti-leukemic activity in vitro and in vivo. SH-RF-177 was not only more potent than FTY720, but killed via a distinct mechanism. Phosphorylation is dispensable for FTY720's anti-leukemic actions. However, chemical biology and genetic approaches demonstrated that the sphingosine kinase 2- (SPHK2) mediated phosphorylation of SH-RF-177 led to engagement of a pro-apoptotic target and increased potency. The cytotoxicity of membrane-permeant FTY720 phosphonate esters suggests that the enhanced potency of SH-RF-177 stems from its more efficient phosphorylation. The tight inverse correlation between SH-RF-177 IC50 and SPHK2 mRNA expression suggests a useful biomarker for SH-RF-177 sensitivity. In summary, these studies indicate that FTY720 analogs that are efficiently phosphorylated but fail to activate S1P receptors may be superior anti-leukemic agents compared to compounds that avoid cardiotoxicity by eliminating phosphorylation.Leukemia accepted article preview online, 30 August 2016. doi:10.1038/leu.2016.244.
... This is accompanied by reduction in proinflammatory cytokine production, including type I interferons and macrophage chemokines (55). AAL(R) is phosphorylated in vivo by sphingosine kinase 2 to AFD(R) ([(2R)-2-amino-4-(4-heptoxyphenyl)-2-methylbutyl] dihydrogen phosphate), which stimulates S1P receptors (S1PR) 1, 3, 4, and 5 (27,42). A more selective S1PR1 agonist, CYM5442, also reduces cytokine production by acting on endothelial cells rather than lymphocytes (52), although its effect on mortality has not, to our knowledge, been reported. ...
Article
In this Perspective, we discuss some recent developments in the pathogenesis of acute lung injury following influenza infection, with an emphasis on promising therapeutic leads. Damage to the alveolar-capillary barrier has been quantified in mice, and agents have been identified that can help to preserve barrier integrity, such as Vasculotide, angiopoietin-like 4 neutralisation, and sphingosine 1-phosphate mimics. The roles of Fatty Acid Binding Protein 5, prostaglandin E2 and the interplay between IFNγ and STAT1 in epithelial signalling during infection have been addressed in vitro. Finally, we discuss the role of autophagy in inflammatory cytokine production and the viral lifecycle, and the opportunities this presents for intervention.
... FTY720 is an analog of sphingosine and is phosphorylated in vivo exclusively by Sphk2 to generate FTY720-P (Paugh et al. 2003;Kharel et al. 2005), which can be exported into the extracellular environment by Spns2 (Hisano et al. 2011). AAL-R, a close relative of FTY720 differing only in the lack of a hydroxyl group, is also efficiently phosphorylated by Sphk2 to generate AFD-R, a nonselective agonist of S1P receptors (Jary et al. 2010). Treatment with nonselective S1P receptor agonists leads to rapid, sustained sequestration of both T and B cells from the blood and the lymph and causes short-lasting bradycardia in both mice and humans (Luo et al. 1999;Budde et al. 2002;Sanna et al. 2004). ...
Article
The zwitterionic lysophospholipid Sphingosine 1-Phosphate (S1P) is a pleiotropic mediator of physiology and pathology. The synthesis, transport, and degradation of S1P are tightly regulated to ensure that S1P is present in the proper concentrations in the proper location. The binding of S1P to five G protein-coupled S1P receptors regulates many physiological systems, particularly the immune and vascular systems. Our understanding of the functions of S1P has been aided by the tractability of the system to both chemical and genetic manipulation. Chemical modulators have been generated to affect most of the known components of S1P biology, including agonists of S1P receptors and inhibitors of enzymes regulating S1P production and degradation. Genetic knockouts and manipulations have been similarly engineered to disrupt the functions of individual S1P receptors or enzymes involved in S1P metabolism. This chapter will focus on the development and utilization of these chemical and genetic tools to explore the complex biology surrounding S1P and its receptors, with particular attention paid to the in vivo findings that these tools have allowed for.
... For GNS cells, culture plates were coated with Matrigel (BD Biosciences) diluted 1:100 in PBS. Cells were seeded at a density of 10 6 cells/ 10-cm dish to quantify the effect of inhibitors on sphingolipid levels and a density of 500 cells/well in 96-well plates for MTT assays (29). Cell proliferation over time was assayed using an xCELLigence MP system (Roche Diagnostics). ...
Article
Full-text available
Studies in cell culture and mouse models of cancer have indicated that the soluble sphingolipid metabolite sphingosine 1-phosphate (S1P) promotes cancer cell proliferation, survival, invasiveness, and tumour angiogenesis. In contrast its metabolic precursor ceramide is pro-differentiative and pro-apoptotic. To determine whether sphingolipid balance plays a significant role in glioma malignancy, we undertook a comprehensive analysis of sphingolipid metabolites in human glioma and normal grey matter (NGM) tissue specimens. We demonstrate, for the first time, a systematic shift in sphingolipid metabolism favouring S1P over ceramide, which increases with increasing cancer grade. S1P content was on average 9-fold higher in glioblastoma (GBM) tissues compared to NGM; whilst the most abundant form of ceramide in the brain, C18 ceramide, was on average 5-fold lower. Increased S1P content in the tumours was significantly correlated with increased Sphingosine Kinase 1 (SPHK1) and decreased Sphingosine Phosphate Phosphatase 2 (SGPP2) expression. Inhibition of S1P production by cultured GBM cells, using a highly potent and selective SPHK1 inhibitor, blocked angiogenesis in co-cultured endothelial cells without affecting VEGF secretion. Our findings validate the hypothesis that altered ceramide/S1P balance is an important feature of human cancers and support the development of SPHK1 inhibitors as anti-angiogenic agents for cancer therapy.
Article
The sphingoid base derived class of lipids (sphingolipids) is a family of interconverting molecules that play key roles in numerous structural and signaling processes. The biosynthetic pathway of the sphingolipids affords many opportunities for therapeutic intervention: targeting the ligands directly, targeting the various proteins involved in the interconversion of the ligands, or targeting the receptors that respond to the ligands. The focus of this article is on the most advanced of the sphingosine-related therapeutics, agonists of sphingosine-1-phosphate receptor 1 (S1P1). The diverse structural classes of S1P1 agonists will be discussed and the status of compounds of clinical relevance will be detailed. An examination of how potential safety concerns are being navigated with compounds currently under clinical evaluation is followed by a discussion of the novel methods being explored to identify next-generation S1P1 agonists with improved safety profiles. Finally, therapeutic opportunities for sphingosine-related targets outside of S1P1 are touched upon.
Article
Full-text available
AAL(S), the chiral deoxy analog of the FDA approved drug FTY720, has been shown to inhibit proliferation and apoptosis in several cancer cell lines. It has been suggested that it does this by activating protein phosphatase 2A (PP2A). Here we report the synthesis of new cytotoxic analogs of AAL(S) and the evaluation of their cytotoxicity in two myeloid cell lines, one of which is sensitive to PP2A activation. We show that these analogs activate PP2A in these cells supporting the suggested mechanism for their cytotoxic properties. Our findings identify key structural motifs required for anti-cancer effects.
Article
A convergent synthesis to access hydrophobic tail analogs and head group modifications of AAL(S) is described. The analogs synthesised were evaluated for their ability to inhibit ceramide synthase 1 and for their cytotoxicity in K562 cells. Our results have identified inhibitors which are non-cytotoxic yet maintain CerS1 inhibition.
Article
Full-text available
The role of sphingosine-1-phosphate (S1P) in regulation of cellular functions and cell protection is reviewed. S1P, along with other sphingolipid metabolites, is believed to act as an intracellular second messenger and as an extracellular mediator molecule. S1P chemistry, production and metabolism are described. Cellular receptors for S1P and their tissue specificity are described. Platelets and erythrocytes have a crucial significance in blood transport of S1P. Hypoxic conditions induce an increase in S1P, which initiates a set of cytoprotective events via its cellular receptors. S1P involvement in regulation of cell migration, myogenesis, control of skeletal muscle function is described. It is shown that S1P balance disturbances may mediate pathological state. S1P system implication in regulation of the most important cellular functions allows considering it as a prospective remedial target.
Article
Full-text available
Fingolimod (FTY720), a sphingosine-1-phosphate-receptor modulator that prevents lymphocyte egress from lymph nodes, showed clinical efficacy and improvement on imaging in a phase 2 study involving patients with multiple sclerosis. In this 12-month, double-blind, double-dummy study, we randomly assigned 1292 patients with relapsing-remitting multiple sclerosis who had a recent history of at least one relapse to receive either oral fingolimod at a daily dose of either 1.25 or 0.5 mg or intramuscular interferon beta-1a (an established therapy for multiple sclerosis) at a weekly dose of 30 microg. The primary end point was the annualized relapse rate. Key secondary end points were the number of new or enlarged lesions on T(2)-weighted magnetic resonance imaging (MRI) scans at 12 months and progression of disability that was sustained for at least 3 months. A total of 1153 patients (89%) completed the study. The annualized relapse rate was significantly lower in both groups receiving fingolimod--0.20 (95% confidence interval [CI], 0.16 to 0.26) in the 1.25-mg group and 0.16 (95% CI, 0.12 to 0.21) in the 0.5-mg group--than in the interferon group (0.33; 95% CI, 0.26 to 0.42; P<0.001 for both comparisons). MRI findings supported the primary results. No significant differences were seen among the study groups with respect to progression of disability. Two fatal infections occurred in the group that received the 1.25-mg dose of fingolimod: disseminated primary varicella zoster and herpes simplex encephalitis. Other adverse events among patients receiving fingolimod were nonfatal herpesvirus infections, bradycardia and atrioventricular block, hypertension, macular edema, skin cancer, and elevated liver-enzyme levels. This trial showed the superior efficacy of oral fingolimod with respect to relapse rates and MRI outcomes in patients with multiple sclerosis, as compared with intramuscular interferon beta-1a. Longer studies are needed to assess the safety and efficacy of treatment beyond 1 year. (ClinicalTrials.gov number, NCT00340834.)
Article
Full-text available
Oral fingolimod, a sphingosine-1-phosphate-receptor modulator that prevents the egress of lymphocytes from lymph nodes, significantly improved relapse rates and end points measured on magnetic resonance imaging (MRI), as compared with either placebo or intramuscular interferon beta-1a, in phase 2 and 3 studies of multiple sclerosis. In our 24-month, double-blind, randomized study, we enrolled patients who had relapsing-remitting multiple sclerosis, were 18 to 55 years of age, had a score of 0 to 5.5 on the Expanded Disability Status Scale (which ranges from 0 to 10, with higher scores indicating greater disability), and had had one or more relapses in the previous year or two or more in the previous 2 years. Patients received oral fingolimod at a dose of 0.5 mg or 1.25 mg daily or placebo. End points included the annualized relapse rate (the primary end point) and the time to disability progression (a secondary end point). A total of 1033 of the 1272 patients (81.2%) completed the study. The annualized relapse rate was 0.18 with 0.5 mg of fingolimod, 0.16 with 1.25 mg of fingolimod, and 0.40 with placebo (P<0.001 for either dose vs. placebo). Fingolimod at doses of 0.5 mg and 1.25 mg significantly reduced the risk of disability progression over the 24-month period (hazard ratio, 0.70 and 0.68, respectively; P=0.02 vs. placebo, for both comparisons). The cumulative probability of disability progression (confirmed after 3 months) was 17.7% with 0.5 mg of fingolimod, 16.6% with 1.25 mg of fingolimod, and 24.1% with placebo. Both fingolimod doses were superior to placebo with regard to MRI-related measures (number of new or enlarged lesions on T(2)-weighted images, gadolinium-enhancing lesions, and brain-volume loss; P<0.001 for all comparisons at 24 months). Causes of study discontinuation and adverse events related to fingolimod included bradycardia and atrioventricular conduction block at the time of fingolimod initiation, macular edema, elevated liver-enzyme levels, and mild hypertension. As compared with placebo, both doses of oral fingolimod improved the relapse rate, the risk of disability progression, and end points on MRI. These benefits will need to be weighed against possible long-term risks. (ClinicalTrials.gov number, NCT00289978.)
Article
Full-text available
The sphingosine 1-phosphate receptor type 1 (S1P1) is important for the maintenance of lymphocyte circulation. S1P1 receptor surface expression on lymphocytes is critical for their egress from thymus and lymph nodes. Premature activation-induced internalization of the S1P1 receptor in lymphoid organs, mediated either by pharmacological agonists or by inhibition of the S1P degrading enzyme S1P-lyase, blocks lymphocyte egress and induces lymphopenia in blood and lymph. Regulation of S1P1 receptor surface expression is therefore a promising way to control adaptive immunity. Hence, we analyzed potential cellular targets for their ability to alter S1P1 receptor surface expression without stimulation. The initial observation that preincubation of mouse splenocytes with its natural analog sphingosine was sufficient to block TranswellTM chemotaxis to S1P directed subsequent investigations to the underlying mechanism. Sphingosine is known to inhibit protein kinase C (PKC), and PKC inhibition with nanomolar concentrations of staurosporine, calphostin C, and GF109203X down-regulated surface expression of S1P1 but not S1P4 in transfected rat hepatoma HTC4 cells. The PKC activator phorbol 12-myristate 13-acetate partially rescued FTY720-induced down-regulation of the S1P1 receptor, linking PKC activation with S1P1 receptor surface expression. FTY720, but not FTY720 phosphate, efficiently inhibited PKC. Cell-based efficacy was obvious with 10 nm FTY720, and in vivo treatment of mice with 0.3–3 mg/kg/day FTY720 showed increasing concentration-dependent effectiveness. PKC inhibition therefore may contribute to lymphopenia by down-regulating S1P1 receptor cell surface expression independently from its activation.
Article
Full-text available
PrimerBank (http://pga.mgh.harvard.edu/primerbank/) is a public resource for the retrieval of human and mouse primer pairs for gene expression analysis by PCR and Quantitative PCR (QPCR). A total of 306 800 primers covering most known human and mouse genes can be accessed from the PrimerBank database, together with information on these primers such as Tm, location on the transcript and amplicon size. For each gene, at least one primer pair has been designed and in many cases alternative primer pairs exist. Primers have been designed to work under the same PCR conditions, thus facilitating high-throughput QPCR. There are several ways to search for primers for the gene(s) of interest, such as by: GenBank accession number, NCBI protein accession number, NCBI gene ID, PrimerBank ID, NCBI gene symbol or gene description (keyword). In all, 26 855 primer pairs covering most known mouse genes have been experimentally validated by QPCR, agarose gel analysis, sequencing and BLAST, and all validation data can be freely accessed from the PrimerBank web site.
Article
Efficient and versatile protocols for the synthesis of chiral analogues of the novel immunomodulator FTY720 and its phosphate are described. These synthetic procedures allow for broad structural variation and deliver essential tools to further elucidate FTY720's novel mechanism of action.
Article
Sphingolipid long-chain base phosphates (LCBPs) regulate cell proliferation, movement and differentiation in higher eukaryotes. To study the function of LCBPs in Saccharomyces cerevisiae, we inactivated LCBP breakdown pathways. Elimination of both the Dpl1 lyase and the Lcb3 phosphatase pathways by gene deletion was lethal, indicating that these enzymes regulate LCBP levels to prevent accumulation. Lethality was prevented by eliminating the major LCB kinase, Lcb4p, which synthesizes LCBPs, but not by eliminating the minor LCB kinase, Lcb5p. These data imply that death results from an accumulation of LCBPs made by the Lcb4p kinase. By regulating Lcb4 kinase activity, we found that cell death correlates with LCBP accumulation and that C18 dihydrosphingosine-1-P (DHS-P) and C20 DHS-P are most likely the killing molecules. LCB levels were found to be most elevated in a strain lacking Lcb4 kinase, Dpl1 lyase and Lcb3 phosphatase activity. Analysis of mutant strains suggests that the C18 and C20 species of LCBPs are preferentially degraded by the Lcb3 phosphate phosphatase, while the Dpl1 lyase prefers C16 DHS-P as a substrate. These and other data indicate the existence of an unknown mechanism(s) for regulating LCB levels. Our results demonstrate that LCBPs may be used in some circumstances to regulate yeast cell growth.
Article
The sphingolipid metabolite sphingosine 1-phosphate (S1P) plays an essential function in the egress of T cells from the thymus and secondary lymphoid organs. The novel immunomodulating agent FTY720 is phosphorylated in vivo to the functional form FTY720 phosphate (FTY720-P), which is structurally similar to S1P. FTY720-P inhibits the S1P-mediated T cell egress as an agonist of S1P receptors. FTY720-P is not stable in plasma and is dephosphorylated to FTY720. In the present study, we investigated activities toward FTY720-P of LPP family members (LPP1, LPP1a, LPP2, and LPP3), which exhibit broad substrate specificity. Of the four, LPP1a, the splicing isoform of LPP1, had the highest activity toward FTY720-P, and the highest affinity. Among blood-facing cells tested, only endothelial cells displayed high phosphatase activity for FTY720-P. Significant levels of LPP1a expression were found in endothelial cells, suggesting that LPP1a is important for the dephosphorylation of FTY720-P in plasma.