Elsevier

Neuropharmacology

Volume 95, August 2015, Pages 37-49
Neuropharmacology

Role of α5-containing nicotinic receptors in neuropathic pain and response to nicotine

https://doi.org/10.1016/j.neuropharm.2015.02.012Get rights and content

Highlights

  • α5 nicotinic receptors are not involved in neuropathic pain nociception in mice.

  • α5 nicotinic receptors are not upregulated in neuropathic pain.

  • α5 nicotinic receptors are involved in nicotine thermal analgesic tolerance.

Abstract

Nicotinic receptors in the central nervous system (nAChRs) are known to play important roles in pain processing and modulate behavioral responses to analgesic drugs, including nicotine. The presence of the α5-neuronal nicotinic accessory subunit in the nicotinic receptor complex is increasingly understood to modulate reward and aversive states, addiction, and possibly pathological pain. In the current study, using α5-knockout (KO) mice and subunit-specific antibodies, we assess the role of α5-containing neuronal nicotinic receptors in neuropathic pain and in the analgesic response to nicotine. After chronic constriction injury (CCI) or partial sciatic nerve ligation (PSNL), no differences in mechanical, heat, or cold hyperalgesia were found in wild-type (WT) versus α5-KO littermate mice. The number of α5-containing nAChRs was decreased (rather than increased) after CCI in the spinal cord and in the thalamus. Nevertheless, thermal analgesic response to nicotine was marginally reduced in CCI α5-KO mice at 4 days after CCI, but not at later timepoints or after PSNL. Interestingly, upon daily intermittent nicotine injections in unoperated mice, WT animals developed tolerance to nicotine-induced analgesia to a larger extent than α5-KO mice. Our results suggest that α5-containing nAChRs mediate analgesic tolerance to nicotine but do not play a major role in neuropathic pain.

Introduction

Neuronal nicotinic acetylcholine receptors (nAChRs) are pentameric ligand-gated channels formed from multiple α (α2–α10) and β subunits (β2–β4) in various combinations that are widely but not uniformly distributed in the peripheral and central nervous system. Heteropentameric nAChRs with an α3β4 backbone prevail in the PNS, whereas α4β2 receptors are more numerous in most parts of the CNS. Both the pharmacological and biochemical properties of nAChRs are critically determined by their subunit composition. Multiple neurobehavioral changes and effects have been attributed to nicotinic receptors in the CNS (Jacob et al., 2013, Dani and Bertrand, 2007, Hurst et al., 2013), including analgesia, allodynia, and pathological pain (Lawand et al., 1999, Bartolini et al., 2011, Umana et al., 2013, Hurst et al., 2013).

Various nicotinic agonists, e.g. epibatidine and related compounds, are potent analgesics acting at the spinal and supraspinal level (Khan et al., 1998, Khan et al., 2001, Bannon et al., 1998, Damaj et al., 1998). Substances such as epibatidine and ABT-594 have been known for quite some time to be equally or more potent analgesics than morphine, depending on the assay (Bannon and Jarboe, 1978). Nicotinic agonist antinociceptive effects have also been shown in animal models of postoperative (Rowley et al., 2008) and of neuropathic pain (Di Cesare et al., 2013, Abdin et al., 2006, Pacini et al., 2010). To date, several types of nAChRs have been implicated in mediating these effects, namely receptors containing the subunits α4 and β2 (Marubio et al., 1999, Khan et al., 2001), α3 (Young et al., 2008, Albers et al., 2014), α5 (Jackson et al., 2010) and α7 (Feuerbach et al., 2009). In vivo evidence for receptors containing the above subunits has been provided by the use of receptor-selective agonists and antagonists, as well as with mice carrying deletions of distinct nAChR subunit genes. Based on molecular modeling, desensitization of α4β2α5 receptors has recently been proposed as the mechanism which mediates the analgesic effect of nicotinic agonists (Zhang et al., 2012). Paradoxically, positive allosteric modulation using novel compounds acting on various nAChRs have also been shown to have potent effects in animal behavioral studies (Uteshev, 2014, Pandya and Yakel, 2013, Rode et al., 2012). For example, the positive α4β2 allosteric modulator NS-9283 can potentiate the analgesic efficacy of the epibatidine analogue ABT-594 (Zhu et al., 2011). Although analgesic effects have to date most often been reported to be due to action at α4β2 containing receptors, recent studies suggest that this subunit combination can be deemed as necessary but not necessarily sufficient to produce analgesia (Gao et al., 2010).

A number of studies have furthermore suggested that nAChRs are directly involved in the pain processing of noxious stimuli and in neuropathic pain. Hence, deletion of the β2 subunit lowers the mechanical and thermal nociceptive thresholds in β2-KO mice (Yalcin et al., 2011), knockdown of α5-containing receptors by intrathecal antisense oligonucleotides moderately reduces allodynia (Vincler and Eisenach, 2005), and hyperalgesia in a nicotine withdrawal model is lost in α7-KO mice (Jackson et al., 2008). After spinal nerve ligation in rats, spinal α5 receptor upregulation has also been reported (Vincler and Eisenach, 2004, Young et al., 2008).

Our work focuses on further studying the role of α5-containing receptors in neuropathic pain and in mediating the analgesic effects of nicotine. α5 is considered an accessory subunit as it can only form functional receptors when co-expressed with a principal subunit (such as α2, α3, or α4) and one complementary subunit (β2 or β4, e.g. as α4β2α5 or α3β4α5 receptors) (Wang et al., 1996, Gerzanich et al., 1998, Ramirez-Latorre et al., 1996). Recent studies using specific antibodies have localized the α5 subunit in various CNS regions, including the substantia nigra pars compacta, medial habenula, interpeduncular nucleus (IPN), striatum, thalamus, prefrontal cortex, hippocampus, and the spinal cord in both rats and mice (Mao et al., 2008, David et al., 2010, Grady et al., 2009, Scholze et al., 2012, Beiranvand et al., 2014). α5 assembles into α3β4 receptors in the superior cervical ganglion (SCG) (Mao et al., 2006, David et al., 2010), whereas in CNS regions such as the hippocampus, the striatum, the cerebral cortex, or the thalamus, α5 is found in combination with the subunits α4 and β2 (Mao et al., 2008). In the habenula, α5 co-assembles with both β2 and β4 to form the α3α5β4β2 complex (Grady et al., 2009, Scholze et al., 2012), while in the IPN α5 subunits co-assemble with β2, but not β4 (Grady et al., 2009, Beiranvand et al., 2014). The presence of α5 can profoundly impact the overall pharmacological and physiological properties of the receptor complex. Effects include altered calcium permeability, increased sensitivity to allosteric modulators, altered receptor desensitization, altered single-channel properties, or altered agonist-mediated responses such as effects on the potency and efficacy of agonists (Ciuraszkiewicz et al., 2013, Tapia et al., 2007, Kuryatov et al., 2008). Two tests for thermal sensitivity testing involving spinal and supraspinal mechanisms show that effects of nicotine are largely reduced in α5-KO mice (Jackson et al., 2010).

In the current study, we test whether α5-KO mice differ from their WT littermates in two well-established models of neuropathic pain and in their responses to analgesic doses of nicotine. We furthermore measure the overall number of hetero-pentameric nAChRs and the expression of distinct receptors containing the subunits β2-, β4-, and α5 by means of immunoprecipitation in the lumbar spinal cord, thalamus, hippocampus, habenula, striatum, and the IPN after peripheral nerve injury. We found no differences in the development of neuropathic pain between WT and α5-KO mice, and only minor changes in the expression of nicotinic receptors after peripheral nerve injury. The thermal analgesic effects of acute nicotine administration were also only marginally different. However, when tested in unoperated mice, WT animals developed tolerance to nicotine-induced analgesia to a larger extent than α5-KO mice.

Section snippets

Animals

For behavioral experiments (see exception below) and all biochemical assays, adult male littermate WT mice and mice with a deletion of the α5 nAChR subunit gene (α5-KO) (Wang et al., 2002) were used. Mice used in this study were backcrossed into C57Bl/6J background for at least 7 generations after germ line transmission. For most of the experiments, KO and WT mice were littermates from heterozygous breeding pairs and genotyped at weaning (18 days after birth). When probing for nicotine

Development of neuropathic pain behaviors is not altered in mice with deletions of the α5 nicotinic receptor subunit

Throughout all experiments, WT and α5-KO animals could not be distinguished by the blinded experimenter and did not show any differences in weight gain. Furthermore, prior to any procedure, control WT and α5-KO mice did not significantly differ in their reaction to heat, cold, or mechanical stimuli (Fig. 1). The development and maintenance of neuropathic pain was compared in α5-KO animals and their WT littermates in two different, widely used animal models: chronic constriction injury (CCI) and

Discussion

The use of nicotinic agonists as an alternative to conventional analgesics for the treatment of acute and pathologic pain is of major clinical interest. It is known that chronic pain patients have high smoking rates (Fishbain et al., 2013) and that smokers are more likely to show chronic pain disorders and have more intense pain (Palmer et al., 2003, Patterson et al., 2012). Studies conducted particularly in postoperative pain have shown mixed significant clinical analgesic effects of nicotine (

Acknowledgments

We would like to thank Gabriele Koth and Karin Schwarz for technical assistance with immunoprecipitation experiments and Jürgen Sandkühler for initial project discussions. Generation of the subunit-specific nAChR antibodies was supported by a grant from the Austrian Science Fund (P19325-B09 to P.S.).

References (97)

  • C. Gotti et al.

    Brain nicotinic acetylcholine receptors: native subtypes and their relevance

    Trends Pharmacol. Sci.

    (2006)
  • K. Hargreaves et al.

    A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia

    Pain

    (1988)
  • P. Honore et al.

    Analgesic profile of intrathecal P2X3 antisense oligonucleotide treatment in chronic inflammatory and neuropathc pain states in rats

    Pain

    (2002)
  • R. Hurst et al.

    Nicotinic acetylcholine receptors: from basic science to therapeutics

    Pharmacol. Ther.

    (2013)
  • K. Kawashima et al.

    Critical roles of acetylcholine and the muscarinic and nicotinic acetylcholine receptors in the regulation of immune function

    Life Sci.

    (2012)
  • I.M. Khan et al.

    Nociceptive and antinociceptive responses to intrathecally administered nicotinic agonists

    Neuropharmacology

    (1998)
  • N.B. Lawand et al.

    Nicotinic cholinergic receptors: potential targets for inflammatory pain relief

    Pain

    (1999)
  • A.B. Malmberg et al.

    Partial sciatic nerve injury in the mouse as a model of neuropathic pain: behavioral and neuroanatomical correlates

    Pain

    (1998)
  • M.J. Marks

    Genetic matters: thirty years of progress using mouse models in nicotinic research

    Biochem. Pharmacol.

    (2013)
  • J.M. McIntosh et al.

    Alpha9 nicotinic acetylcholine receptors and the treatment of pain

    Biochem. Pharmacol.

    (2009)
  • F.J.P. Miao et al.

    Central terminals of nociceptors are targets for nicotine suppression of inflammation

    Neuroscience

    (2004)
  • A. Pacini et al.

    Protective effect of alpha7 nAChR: behavioural and morphological features on neuropathy

    Pain

    (2010)
  • A.A. Pandya et al.

    Effects of neuronal nicotinic acetylcholine receptor allosteric modulators in animal behavior studies

    Biochem. Pharmacol.

    (2013)
  • A.L. Patterson et al.

    Smoking cigarettes as a coping strategy for chronic pain is associated with greater pain intensity and poorer pain-related function

    J. Pain

    (2012)
  • M.R. Picciotto et al.

    Nicotinic receptors in the brain: links between molecular biology and behavior

    Neuropsychopharmacol

    (2000)
  • F. Rode et al.

    Positive allosteric modulation of α4β2 nAChR agonist induced behaviour

    Brain Res.

    (2012)
  • M.C. Rowbotham et al.

    A randomized, double-blind, placebo-controlled trial evaluating the efficacy and safety of ABT-594 in patients with diabetic peripheral neuropathic pain

    Pain

    (2009)
  • L.E. Rueter et al.

    Peripheral and central sites of action for A-85380 in the spinal nerve ligation model of neuropathic pain

    Pain

    (2003)
  • Z. Seltzer et al.

    A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury

    Pain

    (1990)
  • I.C. Umana et al.

    Neuronal nicotinic receptors as analgesic targets: it's a winding road

    Biochem. Pharmacol.

    (2013)
  • V.V. Uteshev

    The therapeutic promise of positive allosteric modulation of nicotinic receptors

    Eur. J. Pharmacol.

    (2014)
  • M. Vincler et al.

    Plasticity of spinal nicotinic acetylcholine receptors following spinal nerve ligation

    Neurosci. Res.

    (2004)
  • M.A. Vincler et al.

    Knock down of the α 5 nicotinic acetylcholine receptor in spinal nerve-ligated rats alleviates mechanical allodynia

    Pharmacol. Biochem. Behav.

    (2005)
  • F. Wang et al.

    Assembly of human neuronal nicotinic receptor α5 subunits with α3, β2, and β4 subunits

    J. Biol. Chem.

    (1996)
  • G. Wu et al.

    A-317491, a selective P2X3/P2X2/3 receptor antagonist reverses inflammatory mechanical hyperalgesia through action at peripheral receptors in rats

    Eur. J. Pharm.

    (2004)
  • I. Yalcin et al.

    Nociceptive thresholds are controlled through spinal β2-subunit-containing nicotinic acetylcholine receptors

    Pain

    (2011)
  • T. Young et al.

    Peripheral nerve injury alters spinal nicotinic acetylcholine receptor pharmacology

    Eur. J. Pharmacol.

    (2008)
  • J. Zhang et al.

    Analgesic effects mediated by neuronal nicotinic acetylcholine receptor agonists: correlation with desensitization of α4β2* receptors

    Eur. J. Pharm. Sci.

    (2012)
  • C.Z. Zhu

    Potentiation of analgesic efficacy but not side effects: co-administration of an α4β2 neuronal nicotinic acetylcholine receptor agonist and its positive allosteric modulator in experimental models of pain in rats

    Biochem. Pharmacol.

    (2011)
  • K.M. Albers et al.

    Artemin growth factor increases nicotinic cholinergic receptor subunit expression and activity in nociceptive sensory neurons

    Mol. Pain

    (2014)
  • S.D. AlSharari et al.

    The antinociceptive effects of nicotinic partial agonists varenicline and sazetidine-A in murine acute and tonic pain models

    J. Pharmacol. Exp. Ther.

    (2012)
  • A.W. Bannon et al.

    Broad-spectrum, non-opioid analgesic activity by selective modulation of neuronal nicotinic acetylcholine receptors

    Science

    (1998)
  • M.J. Bannon et al.

    Reconsideration of a test for dopaminergic stimulation: inability of apomorphine to induce mouse jumping

    Experientia

    (1978)
  • A. Bartolini et al.

    Analgesic and antineuropathic drugs acting through central cholinergic mechanisms

    Recent Pat. CNS Drug Discov.

    (2011)
  • F. Beiranvand et al.

    Nicotinic acetylcholine receptors control acetylcholine and noradrenaline release in the rodent habenulo-interpeduncular complex

    Br. J. Pharmacol.

    (2014)
  • N.L. Benowitz

    Nicotine and postoperative management of pain

    Anesth. Analg.

    (2008)
  • R.T. Boyd et al.

    Nicotinic acetylcholine receptor mRNA in dorsal root ganglion neurons

    J. Neurobiol.

    (1991)
  • A.R. Caggiula et al.

    Different methods of assessing nicotine-induced antinociception may engage different neural mechanisms

    Psychopharmacology (Berl.)

    (1995)
  • Cited by (8)

    • Activation and Inactivation of Nicotinic Receptnors in the Dorsal Hippocampal Region Restored Negative Effects of Total (TSD) and REM Sleep Deprivation (RSD) on Memory Acquisition, Locomotor Activity and Pain Perception

      2020, Neuroscience
      Citation Excerpt :

      Note that, the effects of sleep disorders, TSD (total SD) and RSD (REM SD) on pain perception are inconsistent (Lautenbacher et al., 2006). Interestingly, nAchRs are involved in pain information processing (Xanthos et al., 2015). Previous study has shown that nAchRs play a significant role in mediating stress-induced analgesia in mice (Ghasemzadeh and Rezayof, 2015).

    • Altered nocifensive behavior in animal models of autism spectrum disorder: The role of the nicotinic cholinergic system

      2016, Neuropharmacology
      Citation Excerpt :

      That nAChRs subunits have a role on nocifensive behavior and on the antinociceptive effects of nicotine has also been previously shown (Bagdas et al., 2015; Damaj et al., 1999; Freitas et al., 2015; Saika et al., 2015; Wieskopf et al., 2015; Xanthos et al., 2015; Yalcin et al., 2011). In studies using nAChR agonists and antagonists as well as mice with nAChR subunits null mutations, researchers have shown that nAChRs containing α3, α4, α5, α6, α7, β2, and β4 subunits are involved in modulating nocifensive behaviors in response to acute noxious thermal stimuli (Damaj et al., 1999, 2007; Marubio et al., 1999) and in response to inflammatory and neuropathic processes (AlSharari et al., 2012; Bagdas et al., 2015; Damaj et al., 1999; Freitas et al., 2015; Saika et al., 2015; Wieskopf et al., 2015; Xanthos et al., 2015; Yalcin et al., 2011). Others have also shown that mice with null mutations of α4, α5, or β2 have decreased response to the analgesic effects of nicotine, but intact response to the antinociceptive effects of morphine (Damaj et al., 2007; Marubio et al., 1999).

    View all citing articles on Scopus
    View full text