Involvement of I2-imidazoline binding sites in positive and negative morphine analgesia modulatory effects

https://doi.org/10.1016/j.ejphar.2006.09.031Get rights and content

Abstract

Some studies, suggesting the involvement of I2-imidazoline binding sites (I2-IBS) in morphine analgesia modulation, prompted us to examine on mice antinociceptive assays the effect produced by 1 (phenyzoline), that in view of its high I2-IBS affinity and high I2-IBS selectivity with regard to I1-IBS, α2-adrenoreceptors and μ-opioid receptors might be considered the first interesting I2-IBS ligand. The study was also applied to its ortho phenyl derivative 2 (diphenyzoline), designed and prepared in order to produce a possible modification of the biological profile of 1. Diphenyzoline (2) retains a significant I2-IBS selectivity with regard to I1-IBS, α2-adrenoreceptors and μ-opioid receptors. Moreover, by the functional assays 1 and 2 proved inactive at all α2-adrenoreceptors subtypes up to 10 3 M. As expected, phenyzoline and diphenyzoline, which are structurally related, highlighted an interesting “positive” or “negative”, respectively, morphine analgesia modulatory effect. In fact, 1 (s.c. 10 mg/kg) enhanced morphine analgesia (60% and 40% in mouse tail-flick and mouse hot-plate, respectively), while 2 (s.c. 10 mg/kg) decreased it (− 41% and − 20%, respectively). The ability to decrease morphine analgesia had never been observed before in I2-IBS ligands. These effects were not affected by i.p. treatment of animals with yohimbine (a selective α2-adrenoreceptor antagonist, 0.625 mg/kg) or efaroxan (an I1-IBS/α2-adrenoreceptor antagonist, 1.0 mg/kg). In contrast, they were completely reversed by i.p. treatment of animals with idazoxan (an I2-IBS/α2-adrenoreceptor antagonist, 2 mg/kg). Moreover, compound 2, in mouse tail-flick test, was able to potentiate by 23% the naloxone-induced decrease of morphine analgesia. Therefore, the results of this study indicate the crucial involvement of I2-IBS in the morphine analgesia modulatory effects of 1 and 2.

Introduction

The existence of imidazoline binding sites (IBS) was hypothesized about 20 years ago (Bousquet et al., 1984). Findings from different laboratories have shown that they are widely distributed throughout the tissues of various species, including man, in both central and peripheral nervous systems. The numerous studies with radioligands and those of functional type (Molderings, 1997), apart from confirming the existence of IBS, have also revealed their heterogeneity. At present, they appear to be divided into I1-IBS, recognized preferentially by [3H]-clonidine and related compounds and I2-IBS by [3H]-idazoxan; still uncertain is the existence of I3-IBS subtypes. Even though as a result of immunological studies it has been possible to ascribe the IBS nature to distinct proteins in human and rat brain, the structures of these binding proteins have not been identified yet (Dardonville and Rozas, 2004).

The I1-IBS, located on the plasma membrane of neurons, are present at a fairly high density in the region of the medulla oblungata, that contains the sites of the hypotensive action for imidazoline-like and related drugs (Heemskerk et al., 1998). Pharmacological studies suggested that I1-IBS are involved in the regulation of cardiovascular function (Bousquet, 2001, Molderings and Gothert, 1999), in the modulation of ocular pressure (Ogidigben and Potter, 2001), and in the secretion of renal sodium (Smyth and Penner, 1999).

The I2-IBS, located principally in the outer membrane of mitochondria of peripheral and central tissues (Tesson and Parini, 1991, Tesson et al., 1992) are divided into I2A- and I2B-IBS according to their affinity for amiloride, high or low, respectively (Parini et al., 1996). Biochemical and pharmacological studies suggested a possible structural and functional correlation between I2-IBS and monoamine oxidases (MAOs), two mitochondrial enzymes involved in the oxidate deamination of neurotransmitters (Raddatz et al., 2000, Eglen et al., 1998). I2-IBS are involved in CNS pathologies such as Parkinson's disease (Reynolds et al., 1996), depression (Escriba et al., 1999), tolerance and addiction to opiods (Ruiz-Durantez et al., 2003). It has been described that I2-IBS are present in brain areas involved in perception and response to painful stimuli (Ruggiero et al., 1998).

Since a potentiation of the analgesic effect of morphine by agmatine (proposed as endogenous ligand of IBS) (Kolesnikov et al., 1996) has been observed and a significant decrease of the IBS density in different brain regions after chronic morphine treatment (Su et al., 2001), it is reasonable to hypothesize IBS involvement in the modulation of pain and the pharmacological effects of opioids. However, since agmatine binds to I2-IBS, I1-IBS and α2-adrenoreceptors in many regions of the brain (Li et al., 1994), it could therefore be difficult to ascribe its modulatory activity on morphine antinociception to its binding to one of these receptor types.

Recently a number of studies suggested that some substances displaying high affinity toward the I2-IBS with regard to α2-adrenoceptors such as BFI (Hudson et al., 1995, Alemany et al., 1997) and tracizoline (Pigini et al., 1997), named also valldemossine or LSL 61122 (Ozaita et al., 1997) were able, analogously to agmatine, to regulate opioid-induced analgesia (Sanchez-Blazquez et al., 2000), and to attenuate the development of tolerance and dependence (Boronat et al., 1998). Moreover, the I2-IBS ligands exhibiting no antinociceptive effects by themselves but able to potentiate the analgesic effect of morphine, have been indicated as agonists, while those, such as idazoxan, that by co-treatment completely reverse this potentiation, have been considered antagonists (Sanchez-Blazquez et al., 2000).

Despite the intense efforts in the field, there is still a great deal of uncertainty about the involvement of I2-IBS in the modulation of pain, also because the most commonly used ligands are generally scantly selective with regard to the two main subtypes I1- and I2-IBS, and a possible role of the I1-IBS in analgesia cannot be ruled out (Millan, 2002).

In the present study, our lead is compound 1 (Phenyzoline) (Fig. 1), selected for its very high I2-IBS affinity and high I2-IBS selectivity with regard to I1-IBS and α2-adrenoceptors (pKi I2 = 8.60; pKi I1 = 5.43; pKi α2 = 5.70; I2/I1 = 1479; I2/α2 = 794) (Gentili et al., 2003). Analogously to what was demonstrated by us for ligands interacting with α2-adrenoceptors (Gentili et al., 2002) and I1-IBS (Gentili et al., 2005), in order to produce a possible modulation of biological profile of 1, we designed and prepared its ortho phenyl derivative, compound 2 (Diphenyzoline), already known (Jones and Dimopoulos, 2000) but never considered from this point of view (Fig. 1). The affinity values at I2-IBS, I1-IBS and α2-adrenoceptors of 2 and the values at μ-opioid receptors of 1 and 2 were determined. Compounds 1 and 2 were tested on two algesiometric paradigms: mouse hot-plate and mouse tail-flick tests; the previously studied tracizoline (Pigini et al., 1997) or valldemossine (Sanchez-Blazquez et al., 2000) has been included in this study with the aim to verify the validity of our experimental conditions. To confirm the role played by I2-IBS in the modulation of morphine analgesia, the effects of the pre-treatment with idazoxan (a mixed I2-IBS/α2-adrenoceptor antagonist), efaroxan (a mixed I1-IBS/α2-adrenoceptor antagonist), and yohimbine (a selective α2-adrenoceptor antagonist) were evaluated; a pre-treatment with naloxone (an opioid receptor antagonist) was also performed. In addition, 1 and 2 were subjected to functional assays at α2-adrenoceptor subtypes.

Finally, the molecular structures of 1, 2, tracizoline and idazoxan were superposed.

Section snippets

Animals

Male CD-1 mice (Charles River Breeding Laboratory, Bloomington, MA, USA), weighing 25–35 g were employed. Animals were kept in a room with a reverse 12:12 h light/dark cycle (lights off at 10:00 a.m.), temperature of 20–22 °C and humidity of 45–55%. They were offered free access to tap water and food pellets (4RF18, Mucedola, Settimo Milanese, Italy). The animals were used only in one experimental session. Animal testing was carried out according to the European Community Council Directive of

Radioligand binding assays at I2-IBS, I1-IBS, α2-adrenoceptors and μ-opioid receptors

The affinity values, expressed as pKi, at I2-IBS, I1-IBS, α2-adrenoceptors and μ-opioid receptors of compounds 1 and 2 are reported in Table 1, together with those of tracizoline and naloxone for useful comparison. They show that 2, similarly to 1, displays good affinity for I2-IBS (pKi 6.80), and low affinity for the I1-IBS and α2-adrenoceptors (pKi 5.20 and 5.15, respectively). Both the compounds 1 and 2 display low affinity for μ-opioid receptors (pKi < 5 and 5.7, respectively). High affinity

Discussion

Pain represents a notable medical problem: it could be tackled by the rational design of compounds acting both at the opioid receptors (morphine-like compounds) and non-opioid receptors. Among them, the α2-adrenergic system might also be included, whose agonist ligands, endowed with specific analgesic properties, are also able to synergically interact with opioids. Nevertheless, their side effects such as sedation (Hunter et al., 1997, Lakhlani et al., 1997, Mizobe et al., 1996),

Acknowledgement

The present work was supported by a grant from the University of Camerino and Cofinanziamento MIUR 2005, Italy.

References (49)

  • D.A. Ruggiero et al.

    Immunocytochemical localization of an imidazoline receptor protein in the central nervous system

    Brain Res.

    (1998)
  • A.R.S. Santos et al.

    Mechanisms involved in the antinociception caused by agmatine in mice

    Neuropharmacology

    (2005)
  • F. Tesson et al.

    Identification of an imidazoline-guanidinium receptive site in mitochondria from rabbit cerebral cortex

    Eur. J. Pharmacol.

    (1991)
  • F. Tesson et al.

    Tissue-specific localization of mitochondrial imidazoline-guanidinium receptive site

    Eur. J. Pharmacol.

    (1992)
  • O. Yesilyurt et al.

    Agmatine potentiates the analgesic effect of morphine by an α2-adrenoceptor-mediated mechanism in mice

    Neuropsychopharmacology

    (2001)
  • B.C. Yoburn et al.

    Species differences in μ- and δ-opioid receptors

    Eur. J. Pharmacol.

    (1991)
  • R. Alemany et al.

    Labelling of I2B-imidazoline receptors by [3H]2-(2-benzofuranyl)-2-imidazoline (2-BFI) in rat brain and liver: characterization, regulation and relation to monoamine oxidase enzymes

    Naunyn-Schmiedeberg's Arch. Pharmacol.

    (1997)
  • M.A. Boronat et al.

    Attenuation of tolerance to opioid-induced antinociception and protection against morphine-induced decrease of neurofilament proteins by idazoxan and other I2-imidazoline ligands

    Br. J. Pharmacol.

    (1998)
  • P. Bousquet

    I1 receptors, cardiovascular function, and metabolism

    Am. J. Hypertens.

    (2001)
  • P. Bousquet et al.

    Central cardiovascular effects of alpha adrenergic drugs: differences between catecholamines and imidazolines

    J. Pharmacol. Exp. Ther.

    (1984)
  • Y.C. Cheng et al.

    Relationship between the inhibition constant (Ki) and the concentration of inhibitor which causes 50 percent inhibition (IC50) of an enzymatic reaction

    Biochem. Pharmacol.

    (1973)
  • C. Dardonville et al.

    Imidazoline binding sites and their ligands: an overview of the different chemical structures

    Med. Res. Rev.

    (2004)
  • A. DeLean et al.

    Simultaneous analysis of families of sigmoidal curves: application to bioassay, radioligand assay, and physiological dose–response curves

    Am. J. Physiol.

    (1978)
  • A. Diaz et al.

    Autoradiographic mapping of μ-opioid receptors during opiate tolerance and supersensitivity in the rat central nervous system

    Naunyn-Schmiedeberg's Arch. Pharmacol.

    (2000)
  • Cited by (33)

    • α<inf>2</inf>-agonists and antagonists

      2022, Small Animal Critical Care Medicine
    • Imidazoline I<inf>2</inf> receptors: An update

      2017, Pharmacology and Therapeutics
    • Discriminative stimulus effects of the imidazoline I<inf>2</inf> receptor ligands BU224 and phenyzoline in rats

      2015, European Journal of Pharmacology
      Citation Excerpt :

      Idazoxan is the prototypic compound that was used to define I2 receptors (Eglen et al., 1998). It is often used to block some behavioral effects of other I2 receptor ligands (Ferrari et al., 2011; Gentili et al., 2006; Lanza et al., 2014; Li et al., 2014, 2011; Meregalli et al., 2012; Sanchez-Blazquez et al., 2000; Thorn et al., 2012; Tonello et al., 2012). However, idazoxan fully substitutes for 2-BFI in rats discriminating 2-BFI (Jordan et al., 1996; MacInnes and Handley, 2002).

    • α2 agonists and antagonists

      2014, Small Animal Critical Care Medicine, Second Edition
    View all citing articles on Scopus
    View full text