The sickness behaviour and CNS inflammatory mediator profile induced by systemic challenge of mice with synthetic double-stranded RNA (poly I:C)

https://doi.org/10.1016/j.bbi.2006.12.007Get rights and content

Abstract

Poly inosinic:poly cytidylic acid (poly I:C) is a synthetic double-stranded RNA and is a ligand for the Toll like receptor-3. This receptor is involved in the innate immune response to viral infection and poly I:C has been used to mimic the acute phase of a viral infection. The effects of TLR3 activation on brain function have not been widely studied. In the current study we investigate the spectrum of sickness behavioural changes induced by poly I:C in C57BL/6 mice and the CNS expression of inflammatory mediators that may underlie this. Poly I:C, at doses of 2, 6 and 12 mg/kg, induced a dose–responsive sickness behaviour, decreasing locomotor activity, burrowing and body weight, and caused a mild hyperthermia at 6 h. The 12 mg/kg dose caused significant hypothermia at later times. The Remo400 remote Telemetry system proved a sensitive measure of this biphasic temperature response. The behavioural responses to poly I:C were not significantly blunted upon a second poly I:C challenge either 1 or 3 weeks later. Plasma concentrations of IL-6, TNF-α and IFN-β were markedly elevated and IL-1β was also detectable. Cytokine synthesis within the CNS, as determined by quantitative PCR, was dominated by IL-6, with lesser inductions of IL-1β, TNF-α and IFN-β and there was a clear activation of cyclooxygenase-2 at the brain endothelium. These findings demonstrate clear CNS effects of peripheral TLR3 stimulation and will be useful in studying aspects of the effects of systemic viral infection on brain function in both normal and pathological situations.

Introduction

Poly I:C is a synthetic double-stranded RNA that has been identified as a ligand for the Toll like receptor-3 (TLR3; Alexopoulou et al., 2001). Double-stranded RNA (dsRNA) is a product of viral replication and thus TLR3 is involved in the innate immune response to viral infection (Schulz et al., 2005). Most researchers use the synthetic dsRNA analog poly I:C to stimulate TLR3 since this high molecular weight compound is more potent than the smaller viral dsRNA fragments isolated from viral preparations (Fang et al., 1999). However it is clear that dsRNA detection only represents one part of the innate immune response to viral infection (Schroder and Bowie, 2005), and that there are other pathways through which cells respond to viral infection (Coccia et al., 2004, Edelmann et al., 2004, Li et al., 2005).

A number of researchers have used poly I:C to mimic the acute phase of a viral infection (Guha-Thakurta and Majde, 1997, Traynor et al., 2004) or as a model of chronic fatigue syndrome (Katafuchi et al., 2003, Katafuchi et al., 2005). Stimulation of TLR3 using dsRNA or poly I:C induces type one interferons α and β and many of the acute symptoms of viral infection have been attributed to these cytokines (see Majde, 2000 for review). Whether or not TLR3 activation is an essential part of the anti-viral response, the essential nature of Type I interferons in combating viral infection is undisputed (Muller et al., 1994) and these mediators have been shown to produce a spectrum of behavioural changes (Segall and Crnic, 1990, Crnic and Segall, 1992a, Crnic and Segall, 1992b). However, much like the TLR4 ligand LPS, stimulation of the relevant TLR is likely to produce a wider spectrum of behavioural changes than would any single cytokine induced as a result of its stimulation. To date a limited number of studies of poly I:C induced sickness behaviour have been conducted. It is clear that poly I:C does cause some symptoms of sickness: poly I:C has been shown to induce a fever in rats that is at least partially dependent on IL-1β (Fortier et al., 2004) and mouse studies have reported a biphasic temperature response accompanied by hypoactivity (Traynor et al., 2004). We know quite a lot about how sickness is induced by lipopolysaccharide (LPS) in mice (see Dantzer, 2004 for review) and many of the cytokines implicated in sickness behaviour responses to LPS have also been shown to be induced in vivo after systemic administration of poly I:C (Guha-Thakurta and Majde, 1997, Traynor et al., 2004). Evidence from in vitro studies suggest that a variety of different cell types can respond to poly I:C to synthesize these cytokines (Doyle et al., 2003, Olson and Miller, 2004, Re and Strominger, 2004, Sivori et al., 2004, Scumpia et al., 2005). However, the full spectrum of the sickness behaviour response after systemic poly I:C challenges has not been examined and the CNS expression of cytokines in response to these challenges remains virtually unexplored. A fuller understanding of the CNS consequences of systemic viral infection is desirable both in the study of sickness behaviour mechanisms per se and for the examination of interactions between viral infection and chronic neurodegeneration.

In the current study, we aimed to characterize the sickness behavioural and peripheral and CNS cytokine responses to a range of doses of poly I:C administered intraperitoneally (i.p.) in C57BL/6 mice. The sickness behaviour spectrum examined comprised core-body temperature, hypoactivity and anorexia as well as decreased performance of species-typical behaviours such as burrowing. We also used this experimental system to test the Remo400 remote telemetry system. This system allows remote monitoring of core body temperature using probes considerably smaller than currently available models, and capable of simultaneous monitoring of numerous animals in a single home cage. In addition we have investigated whether C57BL/6 mice show tolerance to the behavioural effects of poly I:C upon repeated challenge with this dsRNA. Aspects of the response to LPS are known to be down-regulated upon repeated LPS challenge but whether a similar phenomenon occurs with TLR3 has not, to our knowledge, been examined in vivo.

We also examined hypothalamic and hippocampal transcription of inflammatory genes in response to intraperitoneal poly I:C. We have assessed CNS transcription of IL-1β, IL-6, TNF-α, and IFN-β, using quantitative PCR and have examined the expression of COX-2 at the cerebral vascular endothelium by immunocytochemistry.

Section snippets

Materials and methods

Poly I:C was obtained from Amersham Biosciences (Little Chalfont, Buckinghamshire, UK). It was prepared for injection by resuspending in sterile saline, heating to 50 °C at a concentration of 2 mg/ml to ensure complete solubilization and then allowing to cool naturally to room temperature to ensure proper annealing of double-stranded RNA. Poly I:C was stored at −20 °C until use. This solution was determined by Limulus assay (LAL) to contain approximately 28 pg LPS/mg poly I:C, or 6.8 pg/mouse. This

Behaviour: open field activity

Open field activity was assessed as described at various times post-challenge with poly I:C at doses of 2, 6 and 12 mg/kg body weight. Repeated measures ANOVA showed a statistically significant effect of time (p < 0.0001, df 4, 144) and an interaction of time and treatment (p = 0.0463, df 12, 144). All three doses showed reduced activity in the open field and this effect was apparent at 4 h and maintained at 8 and 12 h (Fig. 1a). By 12 h animals were in the dark phase of the 12:12 h cycle and were being

Discussion

In this study, we have analysed, in detail, the sickness behaviour pattern induced by intraperitoneal challenge with poly I:C in C57BL/6 mice. We show a profile that is dose-dependent and encompasses alterations in core-body temperature and body weight, changes in locomotor activities and engagement in species-typical behaviours. These changes are accompanied by CNS synthesis of pro-inflammatory cytokines and by activation of the cerebral endothelium.

Conclusion

A clearer knowledge of the acute phase of the innate immune response to viral infection is clearly desirable with respect to its effect on brain function. The production of inflammatory mediators in the brain has a normal homeostatic role as demonstrated from their importance for coordinating sickness behaviour responses (Dantzer, 2004), but they can clearly also have deleterious effects depending on the concentration and duration of their production (Rothwell and Luheshi, 2000). We have been

Acknowledgments

We thank Leah Scott at DSTL Porton Down and Remo Technologies for the use of the Remo400 remote telemetry system. We also wish to gratefully acknowledge the financial support of the Wellcome Trust, the BBSRC and the MS society for their generous financial support of this work.

References (58)

  • C.J. Gordon et al.

    Behavioral thermoregulatory responses of single- and group-housed mice

    Physiol. Behav.

    (1998)
  • Z. Jiang et al.

    Poly(I-C)-induced Toll-like receptor 3 (TLR3)-mediated activation of NFkappa B and MAP kinase is through an interleukin-1 receptor-associated kinase (IRAK)-independent pathway employing the signaling components TLR3-TRAF6-TAK1-TAB2-PKR

    J. Biol. Chem.

    (2003)
  • T. Katafuchi et al.

    Prolonged effects of polyriboinosinic:polyribocytidylic acid on spontaneous running wheel activity and brain interferon-alpha mRNA in rats: a model for immunologically induced fatigue

    Neuroscience

    (2003)
  • K. Li et al.

    Distinct poly(I–C) and virus-activated signaling pathways leading to interferon-beta production in hepatocytes

    J. Biol. Chem.

    (2005)
  • S. Ohdo et al.

    Circadian rhythm of fever induced by interferon-alpha in mice

    Life Sci.

    (1997)
  • N.J. Rothwell et al.

    Interleukin 1 in the brain: biology, pathology and therapeutic target

    Trends Neurosci.

    (2000)
  • M. Schroder et al.

    TLR3 in antiviral immunity: key player or bystander?

    Trends Immunol.

    (2005)
  • T.R. Traynor et al.

    Intratracheal double-stranded RNA plus interferon-gamma: a model for analysis of the acute phase response to respiratory viral infections

    Life Sci.

    (2004)
  • T.R. Traynor et al.

    Sleep and body temperature responses in an acute viral infection model are altered in interferon type I receptor-deficient mice

    Brain Behav. Immun.

    (2006)
  • C. Wietek et al.

    Interferon regulatory factor-3-mediated activation of the interferon-sensitive response element by Toll-like receptor (TLR) 4 but not TLR3 requires the p65 subunit of NF-kappa

    J. Biol. Chem.

    (2003)
  • L. Alexopoulou et al.

    Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3

    Nature

    (2001)
  • A. Billiau et al.

    Tolerability of pure fibroblast interferon in man

    Ann. NY Acad. Sci.

    (1980)
  • L. Capuron et al.

    Association of exaggerated HPA axis response to the initial injection of interferon-alpha with development of depression during interferon-alpha therapy

    Am. J. Psychiatry

    (2003)
  • E.M. Coccia et al.

    Viral infection and Toll-like receptor agonists induce a differential expression of type I and lambda interferons in human plasmacytoid and monocyte-derived dendritic cells

    Eur. J. Immunol.

    (2004)
  • P.G. Conaldi et al.

    Persistent infection of human vascular endothelial cells by group B coxsackieviruses

    J. Infect. Dis.

    (1997)
  • C. Cunningham et al.

    Central and systemic endotoxin challenges exacerbate the local inflammatory response and increase neuronal death during chronic neurodegeneration

    J. Neurosci.

    (2005)
  • C.A. Dinarello et al.

    Mechanisms of fever induced by recombinant human interferon

    J. Clin. Invest.

    (1984)
  • S.E. Doyle et al.

    Toll-like receptor 3 mediates a more potent antiviral response than Toll-like receptor 4

    J. Immunol.

    (2003)
  • J. Fang et al.

    Synthetic influenza viral double-stranded RNA induces an acute-phase response in rabbits

    J. Med. Virol.

    (1999)
  • Cited by (247)

    View all citing articles on Scopus
    View full text