Elsevier

Peptides

Volume 28, Issue 11, November 2007, Pages 2243-2252
Peptides

Regulation of nociceptin/orphaninFQ secretion by immune cells and functional modulation of interleukin-2

https://doi.org/10.1016/j.peptides.2007.09.004Get rights and content

Abstract

Previous research has demonstrated that numerous populations of immune cell, including lymphocytes, synthesize nociceptin (N/OFQ) precursor mRNA although little is known regarding the immunological role of N/OFQ. In the present study we have demonstrated significant effects of mitogens, pro-inflammatory cytokines, cyclic AMP analogues, glucocorticoids and CRF on N/OFQ secretion by rat splenocytes in vitro. N/OFQ (10−14 to 10−10 M) was also shown to inhibit proliferation of Con A-activated splenocytes and production of IL-2 in vitro. In summary we have shown how a variety of stimuli relevant to inflammation can regulate endogenous N/OFQ secretion by splenocytes in vitro. We also suggest that N/OFQ may promote anti-inflammatory actions via suppression of IL-2 in vivo.

Introduction

Accumulating evidence supports the existence of a bi-directional communication network between the nervous and immune systems involving reciprocal feedback coordinated by a common set of signaling molecules including glucocorticoid hormones, peptides and neurotransmitters. The neuropeptide, nociceptin/orphaninFQ (N/OFQ) has recently been shown to modulate several parameters of the immune system. N/OFQ is a heptadecapeptide that is evolutionarily related to the opioid peptide family [29], [38] and exhibits a high degree of homology with the dynorphin A opioid system. Despite the close structural similarity, there is no functional cross talk between opioid and N/OFQ systems at the receptor level, in fact N/OFQ activates its own G-protein-coupled receptor, NOP. N/OFQ has been shown to modulate a wide range of CNS functions in rodents including monoamine neurotransmitter release [28], [41], the hypothalamo–pituitary–adrenal (HPA) axis [6], [23], [24] and other endocrine functions [16], [33], in addition to locomotor [9], [21] and anxiety-related behaviors [7], [14].

Outside of the CNS, a major location of N/OFQ-NOP synthesis is the peripheral immune system. Various neuropeptides have been localized to lymphoid cells and therefore may contribute to pro- and anti-inflammatory responses. Calcitonin-gene related peptide, substance P, somatostatin and neuromedin-U can directly stimulate T cells to produce cytokines [15], [25]. RT-PCR analyses indicate the capacity of various immunocyte populations to synthesize mRNA for the precursor of N/OFQ, preproN/OFQ, and many immune cell types express the full-length NOP receptor mRNA including normal circulating lymphocytes, polymorphonuclear cells and monocytes in addition to T, B and monocytic cell lines [36], [42], however relatively little is known regarding the functional significance of immune-derived N/OFQ and NOP. Emerging evidence shows that the immune NOP receptor modulates proliferation of human lymphocytes in vitro [37], [48], regulates antibody production [12] and neutrophil chemotaxis [42]. In addition, N/OFQ precursor knockout mice show attenuated lymphoid organ expression of cytokine transcripts (TNFα and IL-1β) following challenge with staphylococcal enterotoxin A (SEA) and N/OFQ peptide (100 μg) given by peripheral injection in normal mice also stimulates levels of circulating TNFα in response to SEA [11]. These recent findings suggest that N/OFQ precursor gene products, including N/OFQ itself, are able to modulate immune functions in vivo. Interestingly, recent in vitro studies show that exogenously applied N/OFQ peptide in culture reduces TNFα secretion by staphylococcal enterotoxin B (SEB)-activated T cells [48] denoting divergence between in vitro and in vivo experimental findings. Interaction between N/OFQ and glucocorticoids in vivo may therefore be an important consideration given that N/OFQ precursor knockout mice display elevated basal and post-stress plasma glucocorticoid levels compared to wild-type controls [20]. N/OFQ has the potential to modulate immune function via regulation of glucocorticoid secretion [23], [24] and via direct actions on immune cells. One possibility is that immune-derived N/OFQ may exert paracrine or autocrine regulation over local inflammatory responses or N/OFQ may interact with peripheral nerves to modulate pain transmission.

Like opioids, N/OFQ may serve to regulate both innate and adaptive immunity. Cytokines are the products of innate responses, and these can help to determine the particular type of adaptive response. One mechanism by which N/OFQ could modulate the immune system is by regulation of cytokines, an aspect that warrants further study. The adaptive immune response involves the activation of T cells, of which there are two subsystems, the Th1 and Th2. The classification of these two polarized forms of CD4+ T cells is based on cytokine production patterns. Th1 cells mainly produce TNFβ, IL-2, IL-12, IL-18 and thereby promote cell-mediated immunity, while Th2 cells principally secrete IL-4, IL-5, IL-6 and IL-10 and promote humoral responses. TNFα is derived from both Th1 and Th2 cells. Th1 and Th2 cells are counterregulatory, hence the system is balanced between cell-mediated and humoral immunity. Maintenance of the appropriate Th1/Th2 balance is critical for protective immunity and disruption of this equilibrium has been implicated in a variety of disorders including chronic stress and autoimmunity. To address the role of N/OFQ in cytokine regulation, the effects of N/OFQ on production of IL-2 by splenic lymphocytes was investigated. In this study we targeted IL-2 as it is known to have broad effects on diverse types of cells of the immune system. IL-2 has both autocrine and paracrine functions. It amplifies T cell responses, supports growth, enhances activation-induced apoptosis, stimulates antibody production and regulates T cell tolerance [26].

Cytokines and corticotrophin-releasing factor (CRF) release opioids from immune cells [40]. Potent peripheral analgesia due to direct injection of CRF can be blocked by both antibodies directed against opioid peptides, including endomorphin-2 and β-endorphin [22] and opioid receptor antagonists, including naloxone [31]. The release of opioid peptides from lymphocytes is calcium-dependent and opioid receptor specific [3]. Despite observations of elevations in plasma N/OFQ in chronic pain conditions [19], little is known regarding the origin of plasma N/OFQ and indeed whether N/OFQ in immune cells is released and if so, under what conditions. A further aim of this study was therefore to investigate the factors and mechanisms governing secretion of N/OFQ in lymphocyte cultures. This study addressed the regulatory potential of glucocorticoids, cytokines and other inflammatory mediators on N/OFQ secretion by rat lymphocyte cultures.

Section snippets

Animals

Male Wistar rats (200–250 g) were obtained from Harlan (Harlan, Blackthorn, Oxfordshire) and after arrival in the housing facility were allowed to acclimatize to the colony conditions for a week prior to the onset of the experiment. Rats were group-reared (four rats per cage) in standard sawdust-lined cages in a holding room subject to a fixed photoperiod (lights on 07:00–19:00 h). Standard rat chow and water were available ad libitum. Ambient temperature of the housing room was 22–23 °C.

Splenocyte culture

Rats were

Effect of proliferative stimuli on N/OFQ secretion

To determine whether N/OFQ production could be modulated by cell activation, cell suspensions were incubated in the absence or presence of the T cell mitogen Con A (5 μg/ml) or the B cell mitogen LPS (25 μg/ml) compared to control over time (0, 6, 18, 24 and 48 h). Cell culture supernatants were collected and analyzed for N/OFQ-like immunoreactivity by ELISA. N/OFQ secretion did not change significantly over time in unstimulated culture supernatants (see Fig. 1). Treatment with mitogens resulted

Discussion

Recently the neuropeptide N/OFQ has been demonstrated to modulate several immune parameters including proliferation of human PBMCs [37], [48], monocyte [47] and neutrophil chemotaxis [42] and mast cell histamine release [18]. These studies were based on the observed effects of exogenously applied N/OFQ whereas the functional significance of endogenous N/OFQ in monocytic and lymphocytic cells is less well understood. In the present study we addressed the regulation of endogenous N/OFQ production

Acknowledgement

This study was supported by funds from the University of Bristol.

References (52)

  • J. Mika et al.

    Relationship of pronociceptin/orphanin FQ and the nociceptin receptor ORL1 with substance P and calcitonin gene-related peptide expression in dorsal root ganglion of the rat

    Neurosci. Lett.

    (2003)
  • J. Nemeth et al.

    Inhibition of nociceptin on sensory neuropeptide release and mast cell-mediated plasma extravasation in rats

    Eur. J. Pharmacol.

    (1998)
  • C. Netti et al.

    Endocrine effects of centrally injected nociceptin in the rat

    Brain Res.

    (2002)
  • E. Okuda-Ashitaka et al.

    Nocistatin: a novel neuropeptide encoded by the gene for the nociceptin/orphaninFQ precursor

    Peptides

    (2000)
  • M.S. Pampusch et al.

    Expression of nociceptin/OFQ receptor and prepro-nociceptin/OFQ in lymphoid tissues

    Peptides

    (2000)
  • J. Peluso et al.

    Distribution of nociceptin/orphaninFQ receptor transcript in human central nervous system and immune cells

    J. Neuroimmunol.

    (1998)
  • J. Peluso et al.

    OrphaninFQ/nociceptin binds to functionally coupled ORL1 receptors on human immune cell lines and alters peripheral blood mononuclear cell proliferation

    Brain Res. Bull.

    (2001)
  • Y. Saito et al.

    Molecular cloning and characterization of a novel form of neuropeptide gene as a developmentally regulated molecule

    J. Biol. Chem.

    (1996)
  • E. Schlicker et al.

    Nociceptin/orphanin FQ and neurotransmitter release in the central nervous system

    Peptides

    (2000)
  • L. Sun et al.

    VIP inhibits IL-2 and IL-4 production through different mechanisms in T cells activated by the T cell receptor/CD3 complex

    J. Neuroimmunol.

    (1993)
  • N. Takayama et al.

    Morphine-induced overexpression of prepro-nociceptin/orphanin FQ in cultured astrocytes

    Peptides

    (2005)
  • S. Trombella et al.

    Nociceptin/orphanin FQ stimulates human monocyte chemotaxis via NOP receptor activation

    Peptides

    (2005)
  • P.S. Waits et al.

    Nociceptin/orphaninFQ modulates human T cell function in vitro

    J. Neuroimmunol.

    (2004)
  • M.J. Wick et al.

    Expression of alternate forms of brain opioid ‘orphan’ receptor mRNA in activated human peripheral blood lymphocytes and lymphocytic lines

    Mol. Brain Res.

    (1995)
  • G. Xie et al.

    The promoter region of human prepro-nociceptin gene and its regulation by cyclic AMP and steroid hormones

    Gene

    (1999)
  • B. Buzas et al.

    Activity and cyclic AMP-dependent regulation of nociceptin/orphanin FQ gene expression in primary neuronal and astrocyte cultures

    J. Neurochem.

    (1998)
  • Cited by (29)

    • Phorbol-12-myristate-13-acetate induces nociceptin in human Mono Mac 6 cells via multiple transduction signalling pathways

      2016, British Journal of Anaesthesia
      Citation Excerpt :

      Nociceptin secreted by blood leucocytes may trigger the expression of its receptor and influence the release of inflammatory mediators which contribute to the process of inflammation and/or pain. 31 36–38 In addition, a potential relationship between nociceptin and MAPKs has been reported. 31 39 40 Controlling inflammatory diseases via the peripheral nociceptin-NOP system has been suggested as a possible approach in the development of new agents in the treatment of inflammation and pain. 2 4 5 23 31

    • Endogenous Nociceptin System Involvement in Stress Responses and Anxiety Behavior

      2015, Vitamins and Hormones
      Citation Excerpt :

      N/OFQ may also stimulate proinflammatory responses by triggering mast cell histamine release and increased vascular permeability in vivo (Kimura et al., 2000), although reports of inhibitory effects of N/OFQ on mast cell-mediated plasma extravasation exist (Németh et al., 1998) and we have shown that N/OFQ modulates rat splenocyte proliferation and proinflammatory cytokine production in vitro (Miller & Fulford, 2007). Powerful inflammatory stimuli, such as bacterial endotoxin, lipolysaccharide, appear to stimulate splenocyte release of N/OFQ (Miller & Fulford, 2007), thus immune-derived N/OFQ may act as a paracrine/autocrine regulator of local inflammation or could interact with NOP receptors on peripheral nerves to modulate nociceptive neuronal transmission in vivo. Endogenous N/OFQ function is also implicated in the response to systemic inflammation following lipopolysaccharide administration.

    • Nociceptin/Orphanin FQ-NOP Receptor System in Inflammatory and Immune-Mediated Diseases

      2015, Vitamins and Hormones
      Citation Excerpt :

      For example, Buzas et al. (2002) reported that inflammatory mediators, such as lipopolysaccharide (LPS), IL-1β, and TNF-α, increase ppN/OFQ mRNA and N/OFQ production in astrocytes. Miller and Fulford (2007) showed that those same cytokines enhanced the N/OFQ production in rat splenocytes. These data clearly demonstrated the influence of the immune system in the N/OFQ-NOP receptor pathway, particularly in conditions of proinflammatory response and cell activation as highlighted in the Table 1.

    • Exploring LPS-induced sepsis in rats and mice as a model to study potential protective effects of the nociceptin/orphanin FQ system

      2014, Peptides
      Citation Excerpt :

      One reason for this could be that the changes that occur in the nociceptin system are occurring at a time after the animals in our experiments have reached the permitted limit of illness severity. In vitro LPS-treated splenocytes show a significant increase in N/OFQ secretion compared to controls at 24 h after stimulation [12] and unpublished in vitro work from our lab found upregulation of ppN/OFQ mRNA in a precursor eosinophilic cell line did not occur until 48 h post LPS-treatment. A recent study using a CLP model of sepsis in rats found increased pp N/OFQ mRNA at 72 h, and the authors suggested that a model of 5–7 days might be required [11].

    View all citing articles on Scopus
    View full text