Background Image
Table of Contents Table of Contents
Previous Page  53 / 76 Next Page
Information
Show Menu
Previous Page 53 / 76 Next Page
Page Background

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

AFRICA

383

study was notably higher than those used by others. Egom

et

al

.

28

used as little as 25 nM, Hofmann

et al

.

32

used 500 nM, while

Vessey

et al

.

30

used 600 nM. Our dose was based on work done

on FTY720 as an activator of PP2A in the setting of cancer and

cancerous cell lines.

In 2003, Matsuoka and colleagues

38

reported that 8

µ

M

FTY720 could suppress the phosphorylation of PKB/Akt, Bad

and p70S6 kinase in T-cell leukaemia cells through the activation

of PP2A. Others have also reported on the ability of FTY720

to activate PP2A in the range of 2.5 to 10

µ

M.

36,37,39

Recently it

was reported that FTY720 established this activation by binding

to SET (Suvar3-9, enhancer of zeste, trithorax), an endogenous

inhibitor of PP2A, thereby mediating the dissociation of

SET from PP2A.

37,39

This mechanism is however dependent

on FTY720 not being phosphorylated, since P-FTY720 has

the potential to phosphorylate SET through a Jak2-mediated

pathway, thereby enhancing its PP2A inhibitory function.

37

Others have however also shown that FTY720 can activate

PP2A through the activation of a Pak1-mediated signalling

pathway.

61-63

It is therefore possible that the increase in infarct

size that we observed in the 1-

µ

M pre-treatment group was due

to the activation of PP2A, which theoretically could suppress

the phosphorylation-mediated pro-survival pathways, also

co-activated by FTY720, which we have observed (unpublished

data) and others have reported.

21,2230-32,63

In contrast to this, in the 2.5-

µ

M pre-treatment group,

pro-survival activation probably dominated over PP2A

activation, thereby inducing protection. As will be discussed

in the following paragraphs, 2.5

µ

M FTY720 also induced a

profound reduction in post-ischaemic functional ability, thereby

reducing energy demand and also potentially contributing to a

reduction in IFS.

Contrary to the infarct-sparing effects associated with

FTY720 treatment in our study, we found that 1

µ

M exerted

no effect on functional recovery, while 2.5

µ

M significantly

suppressed post-ischaemic function in both the GI, as well as

RI models. This is in contrast to Hofmann

et al

.

32

and Vessey

et al.

,

30

who both showed that FTY720 maintained functional

ability after ischaemia at doses of 500 nM, 600 nM and 1

µ

M.

It seems as if FTY720 has a dose-dependent effect, with an

increase in FTY720 concentration being detrimental to post-

ischaemic function. This was a surprising finding since, in the

context of cancer research, it has been reported that relatively

high doses of FTY720, administered chronically to mouse

models, did not exert any toxic effects.

36,37

Neither of these studies

however investigated isolated heart performance or resistance to

ischaemic stress.

The concentrations of FTY720 used in our study were also

not as high as those of sphingosine, which have been shown

by others to be detrimental: Suzuki and colleagues

64

reported

that 10 or 20

µ

M sphingosine induced apoptosis in several

cell lines, while Karliner

18

specified 5

µ

M of sphingosine as

cardiotoxic. With regard to the phosphorylated form, Theilmeier

and co-workers

59

reported that a dose as high as 10

µ

M of S1P

protected neonatal rat cardiomyocytes from apoptosis in a model

of glucose and growth factor withdrawal. It is therefore unlikely

that the FTY720 concentrations that we used were toxic. A

possible explanation for our seemingly controversial results may

be found in the perfusion model used, as well as the effects of

S1P receptor stimulation on heart function

per se.

Our experimental model differed from those used by others.

Hofmann

32

showed cardioprotection in retrogradely perfused

rat hearts and human myocardial muscle strip preparations,

while Vessey

et al.

30

used an isolated retrogradely perfused

mouse heart model, with FTY720 administered as a post-

conditioning intervention (four cycles of five seconds’ ischaemia

and reperfusion at the onset of reperfusion). We however

utilised an isolated working rat heart preparation, which has an

additional energy demand

65

and free radical exposure

66

associated

with it. This more challenging setting than normal retrograde

perfusion might explain the inability of FTY720 to protect

functional capacity in our model, and even contribute to its loss.

Although not investigated by us, it has been reported by

others that S1P receptor activation has the potential to suppress

both heart rate, as well as contractility, largely through effects on

intracellular calcium ion (Ca

2+

) dynamics. We propose that these

mechanisms, at least in part, explain the observed detrimental

effects of FTY720 on post-ischaemic function as follows. Heart

tissue expresses S1P receptors 1, 2 and 3.

67

Of these, it is only 1

and 3 that can be activated by P-FTY720.

53

It has been reported

that activation of these receptors in the heart, especially receptor

3, induces a reduction in heart rate

68-70

through the activation of

the inwardly rectifying atrial potassium ion channel (IKACh),

thereby allowing an increased inward flux of potassium ions into

the cell and hyperpolarising the sarcolemma.

70,71

Activation of the S1P receptor 1 also exerts a negative

inotropic effect, through a reduction in the availability of

intracellular Ca

2+

from the sarcoplasmic reticulum (SR) for the

initiation of contraction. Two mechanisms have been shown to

be involved in this inotropic effect: (1) similarly to the reduction

in heart rate, activation of IKACh leads to the hyperpolarisation

of the sarcolemma, leading to a subsequent reduction in action

potential duration, which in turn implies a reduced influx of

Ca

2+

into the cardiomyocytes, thereby reducing the stimulus for

the Ca

2+

-induced release of Ca

2+

from the SR; (2) linking with

the previous mechanism, S1P has been shown to reduce Ca

2+

flux through the L-type Ca

2+

channel, thereby also diminishing

the potency of Ca

2+

-induced Ca

2+

release. This reduction in flow

through the L-type Ca

2+

channel could be due to a Gi-mediated

reduction in cyclic AMP levels, associated with the stimulation

of the S1P receptors.

67,72,73

Ironically, these same mechanisms that

reduce intracellular Ca

2+

levels might also limit Ca

2+

overload

during reperfusion, thereby contributing to the infarct-limiting

effects of FTY720.

In view of the effect of S1P activation on heart rate (as

discussed above), the divergent results that have been generated

regarding the effect of FTY720 on rhythmicity,

28,29

and the

relevance of arrhythmia in the pathology of myocardial I/R

injury, it is a limitation of this study that we did not include the

incidence of arrhythmia in early reperfusion as an additional

endpoint.

We speculate that the FTY720-mediated reduction in

intracellular Ca

2+

levels associated with a general reduction in

functional ability, in combination with the dual stressor of I/R

and work-mode perfusion could explain the severely detrimental

effects of FTY720 on post-ischaemic functional recovery. It

would therefore be interesting to assess the effects of acute

FTY720 administration at these doses in hearts either not

exposed to ischaemia and reperfusion, and/or not exposed to

work-mode perfusion.