Elsevier

Peptides

Volume 21, Issue 7, July 2000, Pages 1081-1099
Peptides

Regular paper
Neurohumoral effects of orphanin FQ/nociceptin: relevance to cardiovascular and renal function

https://doi.org/10.1016/S0196-9781(00)00246-1Get rights and content

Abstract

Orphanin FQ/Nociceptin (OFQ/N) is a peptide whose structure resembles that of the endogenous opioid peptides (endorphins). OFQ/N and its receptor are distributed in neural tissue and brain regions involved in the regulation of pituitary hormone release. Functional studies have shown that this peptide evokes a unique pattern of cardiovascular and renal excretory responses. This review will focus on the neural and humoral effects of OFQ/N and how this peptide may participate in the regulation of cardiovascular and renal function.

Introduction

The discovery of the endogenous opioid-like peptide, orphanin FQ/nociceptin (OFQ/N) in brain tissue has stimulated considerable interest in the potential physiological roles that this novel compound may have. Research initially demonstrated that OFQ/N acts selectively to activate the ORL1 (opioid receptor-like 1) receptor to produce either nociceptive and/or analgesic effects, depending on the experimental conditions of the study. Based on these and subsequent investigations, OFQ/N has been shown to have a complex pharmacology with a pattern of physiological responses that are either similar, or in opposition to those elicited by classic opioid peptide systems (i.e. the endorphins).

In addition to exhibiting a unique profile of effects on analgesic/nociceptive processes, it is clear that OFQ/N evokes marked changes in other biologic systems. OFQ/N modulates second messenger pathways, alters neurotransmitter and hormonal release, and evokes end organ responses in central and peripheral tissues. From these observations, OFQ/N has been suggested to mediate a wide variety of physiological processes. The wide distribution of OFQ/N and ORL1 in neuronal tissue suggests that this novel system may exert many of its biologic effects by modulating neurotransmission and the activity of hormonal pathways. The purpose of this article is to review what is known regarding the neural and humoral effects of OFQ/N on cardiovascular and renal function. For those areas where information is available, a brief overview of the neural and humoral effects of OFQ/N on other biologic systems will also be presented.

OFQ/N is a heptadecapeptide that has been isolated from brain tissue and shown to be the endogenous ligand of the orphan ORL1 receptor [85], [100]. The amino acid sequence of OFQ/N most closely resembles that of endogenous opioid peptides with particular similarity to dynorphin A(1–17), a proposed endogenous ligand of the κ-opioid receptor [26]. ORL1 shares a high degree of nucleotide sequence homology with each of the cloned μ, δ, and κ-opioid receptors, but most notably resembles the κ-1 subtype [86].

The structural resemblance of OFQ/N to that of endogenous opioid peptides suggests that these two systems may share common features in regards to function and/or mechanisms of action. Opioid agonists produce profound alterations in cardiovascular and renal function, respiratory rate, and gastrointestinal motility [93]. These and other biologic responses are mediated by the action of opioids to alter a broad range of neural and humoral systems. These include opioid-induced changes in central sympathetic outflow [62], [63] and the secretion/release of hormones from the pituitary [7], [8], [96], heart [41], adrenal glands [58], [96], and kidneys [58].

Neuroanatomical studies have demonstrated that OFQ/N, prepro-OFQ/N (the precursor of OFQ/N), and ORL1 are distributed throughout the CNS in regions known to participate in the regulation of autonomic and cardiovascular function, and fluid and electrolyte balance (refer to other articles in this issue for complete review). This includes expression of OFQ/N, prepro-OFQ/N, and ORL1 transcripts in regions such as the nucleus of the solitary tract, the central amygdala, the hypothalamus, the dorsal and ventral horns of the spinal cord, and in pre-vertebral and para-vertebral ganglia [2], [10], [13], [37], [42], [54], [71], [82], [86], [87], [89], [91]. In addition to the CNS, OFQ/N has been identified in neural tissue innervating the lung [40] and in several peripheral organs, including the kidneys [115].

Section snippets

OFQ/N and cardiovascular function

Arterial blood pressure is controlled by a complex interaction of neural and humoral mechanisms that act collectively to determine the level of heart rate, cardiac contractility, arterial resistance, and water and sodium balance. An alteration in one or more of these controlling systems can result in a change in arterial blood pressure. The autonomic nervous system expresses the activity of neurogenic arcs received from afferent baroreceptor inputs that innervate the nucleus of the solitary

OFQ/N and renal function

Synthetic or endogenous opioid agonists evoke profound changes in renal function (for review see ref. 58). The effects of opioids on the renal handling of water and electrolytes (e.g. sodium) can occur from a peripheral, direct renal, or CNS site of action. From these locations, opioids evoke multiple integrated neural and humoral mechanisms that act collectively to affect renal hemodynamics and/or the renal tubular reabsorption of sodium and water. OFQ/N shares a number of structural,

Conclusions

The studies summarized above demonstrate that OFQ/N can evoke profound cardiovascular responses by pathways that affect the autonomic nervous system. There is good evidence that OFQ/N affects heart rate and arterial blood pressure by interacting with pathways that affect central parasympathetic (heart) and sympathetic outflow (heart, vasculature, and kidneys). OFQ/N exerts a unique profile of physiological responses, including hypotension, bradycardia, diuresis and antinatriuresis. The neural

Acknowledgements

I thank Velga A. Kenigs and Lisa A. Dayan for their valuable technical support in the research performed in my laboratory.

References (121)

  • X Chu et al.

    Inhibition of cardiovascular activity following microinjection of novel opioid-like neuropeptide nociceptin (orphanin FQ) into the rat rosteral ventrolateral medulla

    Brain Res

    (1999)
  • M.A Czapla et al.

    Nociceptin, an endogenous ligand for the ORL1 receptor, has vasodilator activity in the hindquarters vascular bed of the rat

    Peptides

    (1997)
  • M.A Czapla et al.

    Decreases in systemic arterial and hindquarters perfusion pressure in response to nociceptin are not inhibited by naloxone in the rat

    Peptides

    (1997)
  • N Doi et al.

    Inhibition of rat oxytocin and vasopressin supraoptic nucleus neurons by nociceptin in vitro

    Neuroscience

    (1998)
  • N.J Dun et al.

    Nociceptin-like immunoreactivity in autonomic nuclei of the rat spinal cord

    Neurosci Lett

    (1997)
  • K Fukuda et al.

    cDNA cloning and regional distribution of a novel member of the opioid receptor family

    FEBS Lett

    (1994)
  • A.R Gintzler et al.

    Modulation of enkephalin release by nociceptin

    Eur J Pharmacol

    (1997)
  • S Giuliani et al.

    Prejunctional modulation by nociceptin of nerve-mediated inotropic responses in guinea-pig left atrium

    Eur J Pharmacol

    (1997)
  • S Giuliani et al.

    Effect of nociceptin on heart rate and blood pressure in unanesthetized rats

    Eur J Pharmacol

    (1997)
  • B Gumusel et al.

    Nociceptin. An endogenous agonist for central opioid like1 (ORL1) receptors possesses systemic vasorelaxant properties

    Life Sci

    (1997)
  • T Houtani et al.

    Structure and regional distribution of nociceptin/orphanin FQ precursor

    Biochem Biophys Res Commun

    (1996)
  • K Ikeda et al.

    Functional coupling of the nociceptin/orphanin FQ receptor with the G-protein-activated K+ (GIRK) channel

    Brain Res Mol Brain Res

    (1997)
  • M Kato et al.

    Activation of κ-opioid receptors inhibits depolarisation-evoked exocytosis but not the rise in intracellular in secretory nerve terminals of the neurohypophysis

    Brain Res

    (1992)
  • W Kummer et al.

    Nociceptin and its receptor in guinea-pig sympathetic ganglia

    Neurosci Lett

    (1997)
  • C.C Lai et al.

    Nociceptin-like immunoreactivity in the rat dorsal horn and inhibition of substantia gelatinosa neurons

    Neuroscience

    (1997)
  • A Lecci et al.

    Multiple sites of action in the inhibitory effect of nociceptin on micturation reflex

    J Urol

    (2000)
  • B Lin et al.

    Nociceptin receptor activation produces nitric oxide-mediated systemic hypotension

    Life Sci

    (2000)
  • C.A Maggi

    The role of peptides in the regulation of the micturation reflexan update

    Gen Pharmacol

    (1991)
  • M.H Makman et al.

    Presence and characterization of nociceptin (orphanin FQ) receptor binding in adult rat and human fetal hypothalamus

    Brain Res

    (1997)
  • I McFadzean

    The ionic mechanisms underlying opioid actions

    Neuropeptides

    (1988)
  • C Mollereau et al.

    ORL-1, a novel member of the opioid receptor familyCloning, functional expression and localization

    FEBS Lett

    (1994)
  • N.P Murphy et al.

    Intracerebroventricular orphanin FQ/nociceptin suppresses dopamine release in the nucleus accumbens of anaesthetized rats

    Neuroscience

    (1996)
  • S Ozaki et al.

    A potent and highly selective nonpeptidyl nociceptin/orphanin FQ receptor (ORL1) antagonistJ-113397

    Eur J Pharmacol

    (2000)
  • D.I Quigley et al.

    Orphanin FQ is the major OFQ1–17-containing peptide produced in the rodent and monkey hypothalamus

    Peptides

    (1998)
  • F.A Abdulla et al.

    Nociceptin inhibits T-type Ca2+ channel current in rat sensory neurons by G-protein-independent mechanisms

    J Neurosci

    (1997)
  • B Anton et al.

    Immunohistochemical localization of ORL-1 in the central nervous system of the rat

    J Comp Neurol

    (1996)
  • A Ardati et al.

    Interaction of [3H]orphanin FQ and 125I-Tyr14-orphanin FQ with the orphanin FQ receptorKinetics and modulation by cations and guanine nucleotides

    Mol Pharmacol

    (1997)
  • M Baerwolff et al.

    Effects of subpicomolar changes in vasopressin on urinary concentration

    Am J Physiol

    (1988)
  • C.A Barraclough et al.

    Inhibition of the release of pituitary ovulating hormone in the rat by morphine

    Endocrinology

    (1955)
  • R.J Bicknell et al.

    Effects of opioid agonists and antagonists on oxytocin and vasopressin release in vitro

    Neuroendocrinology

    (1985)
  • R Bigoni et al.

    Characterization of nociceptin receptors in the peripheryin vitro and in vivo studies

    Naunyn-Schmiedeberg’s Arch Pharmacol

    (1999)
  • D.P Brooks et al.

    Opiate receptors within the blood-brain barrier mediate kappa agonist-induced water diuresis

    J Pharmacol Exp Ther

    (1993)
  • B Bucher

    ORL1 receptor-mediated inhibition by nociceptin of noradrenaline release from perivascular sympathetic nerve endings of the rat tail artery

    Naunyn-Schmiedeberg’s Arch Pharmacol

    (1998)
  • G Calo’ et al.

    Structure-activity study of the nociceptin(1–13)-NH2 N-terminal tetrapeptide and discovery of a nociceptin receptor antagonist

    J Med Chem

    (1998)
  • G Calo’ et al.

    Pharmacological characterization of nociceptin receptoran in vitro study

    Can J Physiol Pharmacol

    (1997)
  • D.A Carter et al.

    Inhibition of vasopressin secretion by a κ-opioid receptor agonist

    Neuroendocrinol Lett

    (1984)
  • C.-L Chou et al.

    Oxytocin as an antidiuretic hormone II. Role of V2 vasopressin receptor

    Am J Physiol

    (1995)
  • X Chu et al.

    Profound inhibition of cardiomotor neurons in the rat rostral ventrolateral medulla by nociceptin (orphanin FQ)

    NeuroReport

    (1998)
  • O Civelli et al.

    Orphan receptors and their natural ligands

    J Recept Signal Transduct Res

    (1997)
  • M Connor et al.

    Modulation of Ca2+channel currents of acutely dissociated rat periaqueductal grey neurons

    J Physiol (London)

    (1998)
  • Cited by (58)

    • NOP receptors in the prelimbic cortex have an inhibitory influence on cardiovascular responses induced by restraint stress

      2016, Neuropeptides
      Citation Excerpt :

      In agreement to these results, the i.c.v. administration of N/OFQ induced depressor and bradycardiac responses, as well as a reduction in urinary sodium excretion and an increase in urine flow rate(Kapusta et al., 1999). Depressor and bradycardiac responses were also observed after intravenous administration of this peptide (Champion et al., 1997; Giuliani et al., 1997; Kapusta, 2000), due to parasympathetic activation and sympathetic indirect inhibition. In addition, the increase in plasma corticosterone induced by RS was higher after i.c.v. administration of NOP antagonist (Leggett et al., 2007).

    • Endogenous Nociceptin System Involvement in Stress Responses and Anxiety Behavior

      2015, Vitamins and Hormones
      Citation Excerpt :

      Intracerebroventricular (i.c.v.) injection of N/OFQ causes a variety of behavioral effects in rodents (Calò et al., 1998; Gavioli & Calò, 2006; Meunier et al., 1995; Reinscheid et al., 1995) consistent with a broad central nervous system distribution of the peptide and its receptor (Boom et al., 1999; Mollereau et al., 1996; Neal et al., 1999a, 1999b; Nothacker et al., 1996). Diverse central actions of N/OFQ have been reported including regulation of locomotor activity (Rizzi et al., 2001), feeding (Polidori, de Caro, & Massi, 2000), cognition (Higgins et al., 2002; Nabeshima, Noda, & Mamiya, 1999), the response to psychoactive drugs (Ciccocioppo, Angeletti, Panocka, & Massi, 2000; Martin-Fardon, Ciccocioppo, Massi, & Weiss, 2000), and regulation of cardiovascular and renal function (Kapusta, 2000; Kapusta, Dayan, & Kenigs, 2002). As its name suggests, N/OFQ also regulates pain but in a complex way reflecting the intriguing pharmacology of this peptide system.

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

      2015, Vitamins and Hormones
      Citation Excerpt :

      The cardiovascular dysfunction in septic patients is characterized by severe hypotension, which may be mediated at least in part by NO, a potent vasodilator (Bateman, Sharpe, & Ellis, 2003). It was demonstrated that N/OFQ induces hypotension and bradycardia (Kapusta, 2000; Malinowska, Godlewski, & Schlicker, 2002), decreasing heart rate and mean arterial pressure in mice (Burmeister, Ansonoff, Pintar, & Kapusta, 2008). Using the cecal ligation and puncture (CLP) model of sepsis in rats, our group has demonstrated that the mortality rate of septic animals was significantly reduced by the treatment with a single dose of the NOP antagonist UFP-101 (Carvalho et al., 2008).

    • Nociceptin/orphanin FQ receptor antagonists as innovative antidepressant drugs

      2013, Pharmacology and Therapeutics
      Citation Excerpt :

      Few studies are available detailing the effects of NOP receptor antagonists on the cardiovascular and renal systems. Nevertheless, there is a general agreement that N/OFQ produces hypotension and bradycardia (Kapusta, 2000; Malinowska et al., 2002). This occurs following intravenous (Bigoni et al., 1999; Madeddu et al., 1999) and intracerebroventricular (Kapusta et al., 1999) administration, and is absent in NOP(-/-) mice (Burmeister et al., 2008).

    • Nociceptin/orphanin FQ in inflammation and sepsis

      2011, British Journal of Anaesthesia
      Citation Excerpt :

      Endogenous and exogenous opioids have important cardiovascular effects.30 Similarly, N/OFQ causes hypotension and bradycardia when administered i.v. and intracerebroventricularly, as demonstrated by various in vitro and in vivo animal studies and this has been covered extensively.6 31 Different mechanisms of action have been proposed and here we concentrate on the data which suggest an immunovascular link.

    View all citing articles on Scopus

    This work was supported in part by United States Grants DK43337 and DK02605 from the National Institute of Diabetes and Digestive and Kidney Diseases.

    View full text