Elsevier

Brain Research

Volume 726, Issues 1–2, 8 July 1996, Pages 160-166
Brain Research

Blockade of lipopolysaccharide-induced fever by subdiaphragmatic vagotomy in guinea pigs

https://doi.org/10.1016/0006-8993(96)00326-5Get rights and content

Abstract

It is generally believed that fever is mediated by certain cytokines produced by immune cells activated by exogenous pyrogens, e.g., lipopolysaccharides (LPS), released into the circulation and transported to the brain. There, the cytokines are thought to stimulate prostaglandin (PG) E2 production within the organum vasculosum laminae terminalis region. PGE, then may act as a febrigenic mediator locally or in the surrounding preoptic area (POA). However, whereas the increases in preoptic PGE2 and body (core) temperature (Tc) following the intravenous (i.0 administration of LPS correlate temporally, cytokine levels in blood lag both these increases. From recent data in the literature, we have conjectured that a possible, alternative communication pathway between the i.v. LPS-activated immune system and brain PGE2 may be provided by the vagi. To test this possibility, we measured the levels of PGE2 in the extracellular fluid of the POA (collected by microdialysis) of conscious, subdiaphragmatically vagotomized or sham-operated guinea pigs following LPS administration (2 μg/kg; i.v.); controls received pyrogen-free saline (PFS). The effluents from the microdialysis probes were collected over 30-min periods throughout the experiments and the samples analyzed by radioimmunoassay; (Tc) was monitored continuously using thermocouples inserted 5 cm into the colon. LPS induced a biphasic fall in Tc and failed to increase preoptic PGE2 levels in the vagotomized guinea pigs (n = 10), whereas in their sham-operated controls (n = 10) it induced increases in both preoptic PGE2 and (Tc) within 15 min after its injection; PFS (n = 13) had no effect on either variable. We postulate that peripheral immune cell-derived signals may be transmitted via the vagi to the medulla. From other data, we suggest further that they may be conveyed from here via the ventral noradrenergic bundle to the POA region, where the released norepinephrine induces the local synthesis of PGE2 and, hence, fever onset.

References (49)

  • van DeventerS.J.H. et al.

    Intestinal endotoxemia: clinical significance

    Gastroenterology

    (1988)
  • WanW. et al.

    Neural and biochemical mediators of endotoxin and stress-inducedc-fos expression in the rat brain

    Brain Res. Bull.

    (1994)
  • WatkinsL.R. et al.

    Characterization of cytokine-induced hyperalgesia

    Brain Res.

    (1994)
  • AderemA.A. et al.

    Bacterial lipopolysaccharides prime macrophages for enhanced release of arachidonic acid metabolites

    J. Exp. Med.

    (1986)
  • CoceaniF.

    Prostaglandins and fever: facts and controversies

  • ColemanR.A. et al.

    Prostanoids and their receptors

  • CunninghamE.T. et al.

    Interleukin-1 receptors in the brain and endocrine tissues

    Immunol. Today

    (1993)
  • DeckerK.

    Biologically active products of stimulated liver macrophages (Kupffer cells)

    Eur. J. Biochem.

    (1990)
  • DeRijkR. et al.

    Selective depletion of macrophages prevents increased plasma interleukin-1 concentrations and pituitaryadrenal activation in response to endotoxin administration in rats

    Endocrinology

    (1991)
  • DunnA.J.

    Endotoxin-induced activation of cerebral catecholamine and serotonin metabolism: comparsion with interleukin-1

    J. Pharmacol. Exp. Ther.

    (1992)
  • ElmquistJ.K. et al.

    Anatomic characterization of the ventral medial peroptic area: involvement in the febrile reaction

    Soc. Neurosci. Abst.

    (1995)
  • EriccsonA. et al.

    A functional anatomical analysis of central pathways subserving the effects of interleukin-1 on stress-related neuroendocrine neurons

    J. Neurosci.

    (1994)
  • FosterK.D. et al.

    Fever, tumor necrosis factor and interleukin-6 in young, mature and aged, Fischer 344 rats

    Am. J. Physiol.

    (1992)
  • GaykemaR.P.A. et al.

    Subdiaphragmatic vagotomy suppresses endotoxin-induced activation of hypothalamic corticotropin-releasing hormone neurons and ACTH secretion

    Endocrinology

    (1995)
  • Cited by (176)

    • Body thermal responses and the vagus nerve

      2019, Neuroscience Letters
      Citation Excerpt :

      In rats, subdiaphragmatic vagotomy attenuated or eliminated IL-1β or LPS-triggered hyperthermia [78,109,110]. Similar results were observed in guinea pigs [36,92]. Later, numerous contradictory evidences that support or against the role of the vagus nerve in pyrogen-induced fever were reported.

    • The OVLT initiates the fall in arterial pressure evoked by high dose lipopolysaccharide: Evidence that dichotomous, dose-related mechanisms mediate endotoxic hypotension

      2015, Journal of Neuroimmunology
      Citation Excerpt :

      Together, these data suggest that the ‘cut off’ for vagal mediation of LPS hypotension is between 5 and 15 mg/kg LPS. These experiments were predicated on extensive evidence that the mechanism responsible for the thermoregulatory effects of LPS and cytokines is dose-dependent: very low LPS doses (1 μg/kg i.v.) cause fever by activating vagus nerve afferents whereas higher doses (30 μg/kg i.v.) activate receptors in the OVLT, area postrema and, perhaps, other circumventricular organs (Sehic and Blatteis, 1996; Romanovsky et al., 1997, 2005; Hansen et al., 2001; Blatteis et al., 2005). The vagus nerve is also thought to mediate some of the behavioral and endocrine effects of LPS.

    View all citing articles on Scopus
    View full text