Background Image
Table of Contents Table of Contents
Previous Page  52 / 76 Next Page
Information
Show Menu
Previous Page 52 / 76 Next Page
Page Background

CARDIOVASCULAR JOURNAL OF AFRICA • Volume 27, No 6, November/December 2016

382

AFRICA

Surprisingly, FTY720 exerted a considerably different effect

on infarct size (Fig. 9), in contrast to the effects seen on

functional recovery after regional ischaemia. Administration of

1

µ

M FTY720 as a pre-treatment showed a very strong tendency

to increase IFS (control: 39.89

±

3.93% vs PreFTY: 51.73

±

2.36%,

n

=

6;

p

=

0.066), while administration during reperfusion

limited the development of infarction (control: 39.89

±

3.93% vs

PostFTY: 23.96

±

3.99%,

n

=

6–7;

p

<

0.05).

In contrast to this dependence on the time point of

administration relative to sustained ischaemia, 2.5

µ

M FTY720

consistently reduced IFS (control: 39.89

±

3.93% vs PreFTY:

29.97

±

1.03% and PostFTY720: 30.45

±

2.16%,

n

=

6;

p

<

0.05).

The area at risk, relative to the total area, did not differ among

any of the groups.

We are therefore left with these two disparate observations:

on one hand, 1

µ

M FTY720 had no effect on post-ischaemic

functional recovery and 2.5

µ

M reduced functional recovery,

while on the other hand FTY720 caused a significant reduction

in IFS, with the exception of the increase observed when 1

µ

M

FTY720 was administered prior to ischaemia.

Discussion

Recently there has been great interest in the sphingosine

analogue, FTY720, mostly because of its immunomodulatory

functions, but also because of its potential to stimulate similar

pathways to sphingosine-1-phosphate. In this context, several

researchers have investigated its potential to confer protection

against myocardial ischaemia/reperfusion injury, however, with

divergent results.

In this study we investigated the effects of two different

concentrations of FTY720, administered prior to ischaemia

or during initial reperfusion in two models of ischaemia with

function and infarct size as endpoints. We found that 1

µ

M

FTY720 exerted no effect on functional recovery regardless of

the time of administration, although pre-treatment augmented

infarction, while reperfusion treatment reduced IFS. Increasing

the dose to 2.5

µ

M proved severely detrimental to functional

recovery, although it was associated with an unexpected

reduction in IFS.

FTY720 is a sphingosine analogue, therefore it can elicit

similar effects to sphingosine and it can also be phosphorylated by

intracellular SK2 to generate a phosphorylated form (P-FTY720)

similar to S1P, which can exit the cell of origin and then bind on

any one of the S1P receptors (except receptor 2).

53

This leads

to a conundrum when administering and experimenting with

FTY720, which has also been reported by others,

30,37

namely, are

the effects observed due to FTY720 simulating sphingosine or

P-FTY720 mimicking S1P?

Although we were unable to distinguish between these

two forms of the drug in our experimental setup, there is an

observation and an argument, which both point to a possible

significant involvement of P-FTY720. First, a significant increase

in coronary flow was observed during the administration of

FTY720 at both dosages tested. This vasodilatory effect has

been linked to the activation of S1P receptor 1 and/or 3 in

the endothelium, which then recruits a PI3-kinase pathway,

as well as a Ca

2+

-mediated mechanism to activate endothelial

nitric oxide synthase (eNOS) to produce nitric oxide (NO),

which ultimately facilitates vascular relaxation.

54-56

Although

the IC50 values for P-FTY720 binding to both S1P1 and S1P3

are extremely low (0.21

±

0.17 nM for S1P1 and 5.0

±

2.7 nM

for S1P3

53

), the observed vasodilation in our experimental

hearts nonetheless implies the presence of P-FTY720 in the

system. This possibility is further confirmed by the fact that

the enzyme responsible for phosphorylating FTY270, SK2, is

the predominant sphingosine kinase in the heart

57,58

and is also

activated by FTY720.

57

It is therefore probable that the relatively

high dose of FTY720 employed in this study would further

stimulate FTY720 phosphorylation.

In this regard, Tölle and colleagues

55

found that SK2 in

human umbilical vein endothelial cells (HUVECs) and whole

aortae converted FTY720 to P-FTY720 at a notable rate: 70%

of 1

µ

M FTY720 was phosphorylated after two minutes, and

90% after 10 minutes. Taken together, it is highly likely that a

large portion of the FTY720 administered in our study had been

phosphorylated.

The S1P receptors are all G-coupled protein receptors, which,

when activated, can induce the activation of several pathways

associated with cardioprotection, such as protein kinase C

(PKC), phosphatidylinositol-3-kinase (PI3-kinase) and protein

kinase B (PKB/Akt).

20-24,59

It is therefore no surprise that several

researchers have investigated the ability of S1P to limit infarct

size.

21-24

Even unphosphorylated sphingosine has been associated

with cardioprotection if administered concentrations are low

enough (~0.4

µ

M

22

).

These observations have led to the question whether FTY720

can also be used to confer protection. In 2009, Hofmann and

colleagues addressed this in an

ex vivo

rat heart preparation and

found that although FTY720 increased post-ischaemic function,

it failed to limit IFS,

32

a result repeated in an

in vivo

model

of ischaemia.

29

In 2011, however, Egom

et al

.

31

illustrated the

ability of FTY720 to increase cell viability in isolated neonatal

rat cardiomyocytes exposed to either simulated ischaemia or

hypoxia. These observations were subsequently confirmed by

Vessey and co-workers,

30

who reported that 600 nM FTY720,

administered as a post-conditioning intervention, exerted an

infarct-sparing effect in an isolated mouse heart model exposed

to global ischaemia.

Wang and colleagues

60

administered FTY720 for a period

of one or three weeks in a mouse model and found that this

relatively chronic treatment regime also reduced IFS. Therefore

our results agree with the latter set of findings that FTY720

is able to reduce infarct size. These results can probably be

explained by the expected activation of S1P receptor-mediated

pro-survival pathways and/or suppressed inflammation in the

heart, as has been shown by others (although we did not assess

the mechanism of protection).

The surprising exception was pre-treatment with 1

µ

M, which,

at best, did not influence IFS, while not having an effect on post-

ischaemic functional recovery. To our knowledge, this is the first

study to have experimented with the acute administration of

FTY720 prior to ischaemia, although both Hofmann

et al.

29

and

Wang

et al

.

60

administered the drug to their respective animal

models for a substantial period of time (from one day once off

to three weeks’ chronic treatment) prior to ischaemia. We did

not investigate the mechanism by which FTY720 pre-treatment

influences injury. We however propose that it can be explained, at

least in part, by the concurrent activation of protein phosphatase

2A (PP2A). The concentration of FTY720 used in the present