02.01.2015 Views

Book of abstracts - British Neuroscience Association

Book of abstracts - British Neuroscience Association

Book of abstracts - British Neuroscience Association

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

10.02<br />

The effect <strong>of</strong> PAT (thymulin related peptide) on pain related<br />

behavior and cytokine production in the brain in a rat model <strong>of</strong><br />

neuroinflammation.<br />

Safieh-Garabedian B, Poole S, Mazih B, Saade N E<br />

<strong>Neuroscience</strong> Program, American University <strong>of</strong> Beirut, Bliss Street,<br />

Beirut, Lebanon. Parenterals Section, NIBSC,, Blanche lane, South<br />

Mimms, Potters Bar, Herts EN6 3QG, UK, ,<br />

Despite the relative protection <strong>of</strong> the brain, intracerebral inflammation<br />

can be detrimental for neurons. Our aim in this study, was to<br />

investigate the possible protective role <strong>of</strong> PAT against inflammation.<br />

Brain inflammation was induced in Sprague-Dawley rats by<br />

intracerebroventricular (ICV) injection <strong>of</strong> endotoxin (1µg/5µl). Separate<br />

groups <strong>of</strong> rats (n= 5-6 each) were pretreated with different doses <strong>of</strong><br />

PAT (ICV; 0.2µg, 1µg and 5µg) 30 minutes before ET. One group <strong>of</strong><br />

rats was treated with PAT only and sham operated animals served as<br />

controls. The effects <strong>of</strong> pretreatment with PAT were assessed on pain<br />

related behavior and on the increased intracerebral levels <strong>of</strong> cytokines<br />

induced by ICV injection <strong>of</strong> ET. Under deep anesthesia, tissue samples<br />

were isolated from different brain areas (hippocampus, diencephalon<br />

and brainstem) and were processed for the determination <strong>of</strong><br />

interleukins (IL)-6, IL-8, IL-10 and NGF concentrations by ELISA. ET<br />

injection produced sustained thermal and cold hyperalgesia and<br />

significant upregulation <strong>of</strong> the levels <strong>of</strong> cytokines and NGF.<br />

Pretreatment with PAT prevented in a dose-dependent manner the ETinduced<br />

hyperalgesia and altered the cytokine levels in different areas<br />

<strong>of</strong> the brain. The results suggest that PAT might have an important<br />

protective role <strong>of</strong> the brain against inflammation.<br />

(Supported by grants from URB and Lebanese National Scientific<br />

Research Council)<br />

10.03<br />

Systemic inflammation alters expression <strong>of</strong> the kynurenine pathway<br />

enzymes indolamine 2,3-dioxygenase and kynurenine 3-<br />

monooxygenase in rat brain<br />

Connor T J, Starr N, Harkin A<br />

Trinity College Institute <strong>of</strong> <strong>Neuroscience</strong>, University <strong>of</strong> Dublin, Trinity<br />

College, Dublin 2, Ireland<br />

Indoleamine 2,3-dioxygenase (IDO) is the rate limiting enzyme in the<br />

kynurenine pathway; the main pathway for tryptophan metabolism in the<br />

brain. Here we demonstrate that systemic administration <strong>of</strong> the<br />

inflammagen bacterial lipopolysaccharide (LPS) to rats induces IDO mRNA<br />

expression in cortex and hippocampus 4hr post administration. Induction <strong>of</strong><br />

IDO was associated with a large increase in expression <strong>of</strong> the proinflammatory<br />

cytokines TNF-alpha and IL-6, with a modest increase in IFNgamma<br />

mRNA expression. Following its synthesis by IDO, kynurenine can<br />

be further metabolised down one <strong>of</strong> two pathways. Specifically, the enzyme<br />

kynurenine-3-monooxygenase (KMO) metabolises kynurenine into the free<br />

radical generator 3-hydroxykynurenine; a compound the can be further<br />

metabolized to form the excitotoxic N-methyl-D-aspartate receptor agonist<br />

quinolinic acid. Alternatively, the enzyme kynurenine aminotransferase<br />

(KAT II) metabolises kynurenine into kynurenic acid; a neuroprotective<br />

compound with NMDA receptor antagonist properties. Consequently,<br />

maintaining an appropriate balance between these two arms <strong>of</strong> the<br />

kynurenine pathway is critical for a healthy brain. Here we demonstrate that<br />

LPS suppressed KMO mRNA expression 4hr post administration, with a<br />

rebound increase observed at 24hr post LPS. In contrast, LPS failed to<br />

alter KAT II expression. This is the first demonstration that an inflammatory<br />

stimulus impacts upon expression <strong>of</strong> KMO; an enzyme that drives<br />

kynurenine metabolism in a neurotoxic direction. We speculate that the<br />

reduction in KMO expression observed 4hr post LPS may represent a<br />

physiological mechanism to limit production <strong>of</strong> neurotoxic products<br />

following IDO induction in response to an inflammatory insult.<br />

10.04<br />

Noradrenaline reuptake inhibitors inhibit neuroinflammation<br />

induced by a systemic inflammatory challenge<br />

O`Sullivan J B, Harkin A, Connor T J<br />

Trinity College Institute <strong>of</strong> <strong>Neuroscience</strong>, University <strong>of</strong> Dublin, Trinity<br />

College, Dublin 2, Ireland.<br />

Evidence suggests that the monoamine neurotransmitter<br />

noradrenaline elicits anti-inflammatory actions in the central nervous<br />

system (CNS), and consequently may play an endogenous<br />

neuroprotective role in CNS disorders where inflammatory events<br />

contribute to pathology. In line with this hypothesis, we demonstrate<br />

that noradrenaline suppresses expression <strong>of</strong> the pro-inflammatory<br />

cytokines IL-1beta and TNF-alpha and induction <strong>of</strong> iNOS/nitric oxide<br />

production from mixed glial cultures prepared from rat cortex, in<br />

response to the inflammagen bacterial lipopolysaccharide (LPS). As<br />

previous studies indicate that the noradrenaline reuptake inhibitor<br />

(NRI) desipramine has anti-inflammatory properties, we examined the<br />

ability <strong>of</strong> desipramine and more selective NRI’s to alter glial proinflammatory<br />

cytokine production. However, treatment <strong>of</strong> mixed glial<br />

cells with NRI’s largely failed to alter inflammatory events induced by<br />

LPS. In contrast to the in vitro situation, acute in vivo treatment <strong>of</strong> rats<br />

with NRI’s elicited an anti-inflammatory effect in the CNS<br />

characterised by reduced mRNA expression <strong>of</strong> the pro-inflammatory<br />

cytokines IL-1beta and TNF-alpha and iNOS in cortex in response to<br />

systemic LPS administration. The data also suggest that in vivo<br />

treatment with NRI’s inhibited microglial activation in the cortex<br />

indicated by reduced expression <strong>of</strong> the microglial activation makers<br />

CD40 and CD11b. These data indicate that NRI’s do not have a direct<br />

modulatory effect on the inflammatory response in glial cells, however<br />

when administered in vivo can limit inflammatory events in the brain.<br />

Overall, this study has yielded significant insights into the ability <strong>of</strong><br />

noradrenaline augmentation strategies to limit neuroinflammation.<br />

10.05<br />

The PPARã agonist, rosiglitazone, attenuates LPS-induced changes in<br />

vitro in an IL-4-dependent manner.<br />

Griffin R J, Loane D J, Deighan B F, Lynch M A<br />

Trinity College Institute <strong>of</strong> <strong>Neuroscience</strong>, Physiology Department, Trinity<br />

College, Dublin 2, Ireland.<br />

Inflammatory changes in the brain contribute to neuronal deficits associated<br />

with age and neurodegenerative conditions. These changes include<br />

activation <strong>of</strong> microglia and the associated release <strong>of</strong> proinflammatory<br />

cytokines. Evidence from this and other laboratories suggest that<br />

rosiglitazone, a selective peroxisome proliferator-activated receptor gamma<br />

(PPARγ) agonist, exerts an anti-inflammatory action in brain.<br />

We investigated the modulatory effect <strong>of</strong> rosiglitazone on LPS-induced<br />

changes in cultured rat cortical glia and report that it significantly reduced<br />

the LPS-triggered increases in interleukin-1β (IL-1β) and tumour necrosis<br />

factor-α (TNFα) (p < 0.001). The mechanisms underlying the antiinflammatory<br />

effects <strong>of</strong> rosiglitazone are not thoroughly understood but our<br />

data show that treatment with rosiglitazone increases hippocampal<br />

concentration <strong>of</strong> the anti-inflammatory cytokine IL-4. To address this further,<br />

we investigated the effect <strong>of</strong> rosiglitazone on LPS-induced changes in<br />

primary glial cells prepared from 1-day old wild-type and IL-4-/- mice. The<br />

data show that rosiglitazone attenuates the LPS-induced increase in IL-1β<br />

concentration (p < 0.05) and the expression <strong>of</strong> major histocomaptabilty<br />

complex class (MHC) II mRNA (p < 0.05) in cells prepared from in wild-type<br />

but not IL-4-/- mice. In contrast, rosiglitazone attenuated the LPS-induced<br />

increase to a similar extent in both preparations and had no effect on the<br />

LPS-induced increase on IL-6. Our findings confirm an anti-inflammatory<br />

role for rosiglitazone and suggest that the action may be mediated by IL-4.<br />

Acknowledgements: Science Foundation Ireland, The Higher Education<br />

Authority Ireland (PRTLI), G03007 8AA.<br />

Page 17/101 - 10/05/2013 - 11:11:03

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!