Elsevier

Neuropharmacology

Volume 53, Issue 2, August 2007, Pages 197-206
Neuropharmacology

Review
Targeting the cholinergic system as a therapeutic strategy for the treatment of pain

https://doi.org/10.1016/j.neuropharm.2007.04.002Get rights and content

Abstract

Acetylcholine mediates its effects through both the nicotinic acetylcholine receptors (ligand-gated ion channels) and the G protein-coupled muscarinic receptors. It plays pivotal roles in a diverse array of physiological processes and its activity is controlled through enzymatic degradation by acetylcholinesterase. The effects of receptor agonists and enzyme inhibitors, collectively termed cholinomimetics, in antinociception/analgesia are well established. These compounds successfully inhibit pain signaling in both humans and animals and are efficacious in a number of different preclinical and clinical pain models, suggesting a broad therapeutic potential. In this review we examine and discuss the evidence for the therapeutic exploitation of the cholinergic system as an approach to treat pain.

Introduction

Acetylcholine mediates its effects through both the nicotinic acetylcholine receptors (ligand-gated ion channels) and the G protein-coupled muscarinic receptors. It plays pivotal roles in a diverse array of physiological processes and its activity is controlled through enzymatic degradation by acetylcholinesterase. The effects of receptor agonists and enzyme inhibitors, collectively termed cholinomimetics, in antinociception/analgesia are well established. These compounds successfully inhibit pain signaling in both humans and animals and are efficacious in a number of different preclinical and clinical pain models (see Table 1), suggesting a broad therapeutic potential. In this review we examine and discuss the evidence for the therapeutic exploitation of the cholinergic system as an approach to treat pain.

Section snippets

Mechanisms of cholinomimetic analgesia

A major site of action for cholinomimetics in analgesia is the spinal cord. Intrathecal cholinergic agents cause antinociception by mimicking the release of acetylcholine (ACh) from the spinal cholinergic nerves. Painful stimuli are known to increase ACh in the spinal cord, as seen in the CSF of anesthetized sheep following noxious stimulation (Eisenach et al., 1996). This ACh is released from the cholinergic interneurons in the dorsal horn, which are activated by the inhibitory descending

Acetylcholinesterase

The hydrolysis of ACh by acetylcholinesterase (AChE) has a key role in limiting the activation of both nicotinic and muscarinic receptors. Overactivity of these receptors causes a multitude of adverse physiological effects and can result in respiratory failure and death. However, cholinesterase inhibitors have shown activity in clinical trials for pain.

Muscarinic acetylcholine receptors

The slow actions of acetylcholine are mediated through the muscarinic acetylcholine receptors (mAChRs), a family of five related G protein-coupled receptors, which are expressed throughout the CNS and in peripheral tissues (Caulfield and Birdsall, 1998). M1, 3 and 5 are linked to phospholipase C via Gq and their stimulation leads to formation of inositol phosphates and a consequent increase in intracellular calcium. M2 and M4 couple through the inhibitory G protein, Gi/o and inhibit formation

Nicotinic acetylcholine receptors

The fast actions of acetylcholine are mediated by its interaction with the nicotinic acetylcholine receptors (nAChRs), a family of pentameric ligand-gated ion channels composed of one or more of sixteen different subunits (α, β, etc.) and distributed widely throughout the CNS and the periphery. In the CNS the predominant nAChRs are the homomeric α7 receptor and the heteromeric α4/β2 receptor representing α-bungarotoxin sensitive and insensitive receptors, respectively, providing a convenient

Conclusions

The cholinergic system offers a number of tractable targets for the development of pain therapeutics. We have discussed the wealth of literature suggesting that cholinomimetics have broad therapeutic potential for efficacy against a number of clinically relevant pain states including inflammatory, neuropathic, visceral pain and pain due to arthritis.

To date, efforts to produce subtype-selective agents have been largely unsuccessful representing the primary obstacle to the successful entry of an

Acknowledgement

We would like to thank Dr. Garth Whiteside, Wyeth Neurosciences, for his valuable comments on our manuscript.

References (98)

  • A.P.C. Guimaraes et al.

    Modulation of carbachol-induced antinociception from the rat periaqueductal gray

    Brain Research Bulletin

    (2000)
  • S.E. Harte et al.

    Involvement of the intralaminar parafascicular nucleus in muscarinic-induced antinociception in rats

    Brain Research

    (2004)
  • K. Honda et al.

    Involvement of M3 muscarinic receptors of the spinal cord in formalin-induced nociception in mice

    Brain Research

    (2000)
  • M. Iyadomi et al.

    Presynaptic inhibition by baclofen of miniature EPSCs and IPSCs in substantia gelatinosa neurons of the adult rat spinal dorsal horn

    Pain

    (2000)
  • K. Koga et al.

    Intrathecal clonidine inhibits mechanical allodynia via activation of the spinal muscarinic M1 receptor in streptozotocin-induced diabetic mice

    European Journal of Pharmacology

    (2004)
  • R.J. Langley et al.

    Central but not the peripheral action of cholinergic compounds suppresses the immune system

    Journal of Neuroimmunology

    (2004)
  • N.B. Lawand et al.

    Nicotinic cholinergic receptors: potential targets for inflammatory pain relief

    Pain

    (1999)
  • B.G. Livett et al.

    Therapeutic applications of conotoxins that target the neuronal nicotininc acetylcholine receptor

    Toxicon

    (2006)
  • J.J. Lynch et al.

    ABT-594 (a nicotinic acetylcholine agonist): anti-allodynia in a rat chemotherapy-induced pain model

    European Journal of Pharmacology

    (2005)
  • E. Mulugeta et al.

    Loss of muscarinic M4 receptors in spinal cord of arthritic rats: implications for a role of M4 receptors in pain response

    Brain Research

    (2003)
  • T. Nishiyama et al.

    Spinally mediated analgesic and receptor binding affinity of epibatidine analogs

    European Journal of Pharmacology

    (2003)
  • E. Nizri et al.

    Anti-inflammatory properties of cholinergic up-regulation: a new role for acetylcholinesterase inhibitors

    Neuropharmacology

    (2006)
  • J. Nomura et al.

    The presence and functions of muscarinic receptors in human T cells: the involvement in IL-2 and IL-2 receptor system

    Life Sciences

    (2003)
  • W.A. Prado et al.

    Antinociceptive effects of bethanechol or dimethylphenylpiperazinium in models of phasic or incisional pain in rats

    Brain Research

    (2004)
  • T.S. Rao et al.

    Evaluation of anti-nociceptive effects of neuronal nicotinic acetylcholine receptor (NAChR) ligands in the rat tail-flick assay

    Neuropharmacology

    (1996)
  • A.L. Sabb et al.

    Discovery of a highly potent, functionally-selective muscarinic M1 agonist, WAY-132983 using rational drug design and receptor modelling

    Bioorganic Medincinal Chemistry Letters

    (1999)
  • P. Sauerberg et al.

    Muscarinic agonists as analgesics. Antinociceptive activity versus M1 activity: SAR of alkylthio-TZTP's and related 1,2,5-thiadiazole analogs

    Life Sciences

    (1995)
  • H.E. Shannon et al.

    Antihyperalgesic effects of the muscarinic receptor ligand vedaclidine in models involving central sensitization in rats

    Pain

    (2001)
  • J.P. Sullivan et al.

    A-85380: in vitro pharmacological properties of a novel, high-affinity α4/β2 nicotinic acetylcholine receptor ligand

    Neuropharmacology

    (1996)
  • A.M. Tata et al.

    Expression of muscarinic M2 receptor mRNA in dorsal root ganglia of neonatal rat

    Brain Research

    (1999)
  • A.M. Tata et al.

    Muscarinic receptor subtypes expression in rat and chick dorsal root ganglia

    Molecular Brain Research

    (2000)
  • S.K. Tayebati et al.

    Radioligand binding assay of M1–M5 muscarinic cholinergic receptor subtypes in human peripheral blood lymphocytes

    Journal of Neuroimmunology

    (1999)
  • S.K. Tayebati et al.

    Immunochemical and immunocytochemical characterization of cholinergic markers in human peripheral blood lymphocytes

    Journal of Neuroimmunology

    (2002)
  • H. Wang et al.

    Chronic neuropathic pain is accompanied by global changes in gene expression and shares pathobiology with neurodegenerative diseases

    Neuroscience

    (2002)
  • Y. Wang et al.

    Antinociceptive effects of choline against acute and inflammatory pain

    Neuroscience

    (2005)
  • J. Wess et al.

    Muscarinic receptor subtypes mediating central and peripheral antinociception studied with muscarinic receptor knockout mice: a review

    Life Sciences

    (2003)
  • I. Wessler et al.

    Non-neuronal acetylcholine, a locally acting molecule, widely distributed in biological systems: expression and function in humans

    Pharmacology and Therapeutics

    (1998)
  • M. Zhuo et al.

    Spinal cholinergic and monaminergic receptors mediate descending inhibition from the nuclei reticularis gigantocellularis and giganocellularis pars alpha in the rat

    Brain Research

    (1990)
  • R.A. Almeida et al.

    Low-dose intrathecal neostigmine enhanced the analgesic effect of intrathecal morphine following gynecologic surgery

    Anesthesiology

    (2003)
  • A.W. Bannon et al.

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

    Science

    (1998)
  • A.W. Bannon et al.

    ABT-594 [(R)-5-2(azetidinylmethoxy)-2-chloropyridine]: a novel, orally effective antinociceptive agent acting via neuronal nicotinic acetylcholine receptors: II. in vivo characterization. Journal of Pharmacology and Experimental

    Therapeutics

    (1998)
  • K.J. Berkley

    Sex difference in pain

    Behavioural Brain Sciences

    (1997)
  • N. Bernardini et al.

    Rat dorsal root ganglia express M1–M4 muscarinic receptor proteins

    Journal of the Peripheral Nervous System

    (1999)
  • N. Bernardini et al.

    Excitatory nicotinic and desensitizing muscarinic (M2) effects on C-nociceptors in isolated rat skin

    Journal of Neuroscience

    (2001)
  • N. Bernardini et al.

    Muscarinic M2 receptors on peripheral nerve endings: a molecular target of antinociception

    Journal of Neuroscience

    (2002)
  • L.V. Borovikova et al.

    Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin

    Nature

    (2000)
  • H. Bouaziz et al.

    Postoperative analgesia from intrathecal neostigmine in sheep

    Anesthesia and Analgesia

    (1995)
  • T.J. Brennan et al.

    Characterization of a rat model of incisional pain

    Pain

    (1996)
  • F.I. Carroll et al.

    Synthesis, nicotinic acetylcholine receptor binding, and antinociceptive properties of 2-exo-2-(2′,3′-disubstituted 5′-pyridinyl)-7-azabicyclo[2.2.1]heptanes: epibatidine analogues

    Journal of Medicinal Chemistry

    (2002)
  • Cited by (90)

    • Antinociceptive effects of potent, selective and brain penetrant muscarinic M<inf>4</inf> positive allosteric modulators in rodent pain models

      2020, Brain Research
      Citation Excerpt :

      One such approach, with a broad array of tractable targets, could be to modulate the cholinergic system (Widman et al., 1985). A review of the literature suggests that agonists and enzyme inhibitors targeting either nicotinic ligand-gated ion channels or G-protein coupled muscarinic receptors, have efficacy in a variety of preclinical and clinical pain models, and could offer therapeutic approaches for clinically relevant pain targets including acute, inflammatory, and neuropathic pain (Jones and Dunlop, 2007). Based upon the literature, muscarinic acetylcholine (ACh) receptors (mAChRs) contribute to modulation of pain both directly and indirectly.

    • Differential effect of sex on pain severity and smoking behavior and processes

      2019, Addictive Behaviors
      Citation Excerpt :

      Pain has been shown to motivate smoking (Ditre & Brandon, 2008; Parkerson & Asmundson, 2016) and pain patients endorse the use of cigarettes to cope with pain (Hooten et al., 2011; Patterson et al., 2012). Indeed, smokers may use cigarettes to manage pain in part due to the analgesic effects of nicotine (Ditre, Heckman, Zale, Kosiba, & Maisto, 2016; Jones & Dunlop, 2007). Conversely, cigarette smoking has been implicated in the onset and exacerbation of painful conditions, including chronic low back pain and rheumatoid arthritis (Shiri, Karppinen, Leino-Arjas, Solovieva, & Viikari-Juntura, 2010; Sugiyama et al., 2009; USDHHS, 2014).

    View all citing articles on Scopus
    View full text