Chapter Four - Restoring the Spinal Pain Gate: GABAA Receptors as Targets for Novel Analgesics

https://doi.org/10.1016/bs.apha.2014.11.007Get rights and content

Abstract

GABAA receptors (GABAARs) and glycine receptors are key elements of the spinal control of nociception and pain. Compromised functioning of these two transmitter systems contributes to chronic pain states. Restoring their proper function through positive allosteric modulators should constitute a rational approach to the treatment of chronic pain syndromes involving diminished inhibitory spinal pain control. Although classical benzodiazepines (i.e., full agonists at the benzodiazepine binding site of GABAARs) potentiate synaptic inhibition in spinal pain controlling circuits, they lack clinically relevant analgesic activity in humans. Recent data obtained from experiments in GABAAR point-mutated mice suggests dose-limiting sedative effects of classical nonspecific benzodiazepines as the underlying cause. Experiments in genetically engineered mice resistant to the sedative effects of classical benzodiazepines and studies with novel less sedating benzodiazepines have indeed shown that profound antihyperalgesia can be obtained at least in preclinical pain models. Present evidence suggests that compounds with high intrinsic activity at α2-GABAAR and minimal agonistic activity at α1-GABAAR should possess relevant antihyperalgesic activity without causing unwanted sedation. On-going preclinical studies in genetically engineered mice and clinical trials with more selective benzodiazepine site agonists should soon provide additional insights into this emerging topic.

Introduction

Chronic pain is a severe medical condition affecting millions of patients worldwide. It is almost generally accepted that neuronal and synaptic plasticity occurring at different levels of the neuraxis are major contributors to chronic pain (Luo et al., 2014, Sandkühler, 2009, Zeilhofer, Witschi and Johansson, 2009) (for a schematic illustration of the pain pathway, see Fig. 1). Some of these neuroplastic changes occur already in the peripheral terminals of nociceptors, which sense noxious stimuli arriving at the skin or in other peripheral tissues and convey them to the central nervous system. The central terminals of these nociceptors innervate the substantia gelatinosa (lamina II) of the spinal dorsal horn, or the trigeminal nucleus of the brainstem in case of those nociceptors coming from the facial skin or the meninges. From there, signals are propagated through various relay stations in the brainstem, midbrain, and thalamus to several cortical areas which give rise to the conscious sensation of pain. One site that has attracted particular attention in pain-related neuroplasticity is the spinal dorsal horn, which constitutes as the first site of synaptic integration in the pain pathway. Neurons located in the spinal dorsal horn integrate primary afferent sensory signals of painful and non-painful modalities with input from descending fiber tracts, which can either inhibit or facilitate pain. Inhibitory interneurons have been attributed a critical role in this process already in the gate-control-theory of pain (Fig. 2; Melzack & Wall, 1965). Although some of the synaptic connections proposed in the original scheme do apparently not exist, plenty of evidence indicates that compromising the function of inhibitory dorsal horn neurons induces symptoms reminiscent of chronic pain syndromes in humans. Animals develop an exaggerated sensitivity to painful stimuli (hyperalgesia), they respond with withdrawal responses upon exposure to stimuli, which are normally not felt as painful (allodynia), and they also show signs of spontaneous discomfort. Many lines of evidence indicate that typical causes of chronic pain such as inflammation or neuropathies compromise the function of inhibitory interneurons in the spinal dorsal horn through different mechanisms (for a review, see Zeilhofer, Benke, & Yévenes, 2012). According to this concept, a facilitation of inhibitory neurotransmission should be a rational strategy for the treatment of many chronic pain states. Yet, none of the established analgesics act- through a facilitation of inhibitory neurotransmission. In the following text, we will review mechanisms of pain-related spinal disinhibition and evidence supporting the concept that novel subtype-selective benzodiazepine agonists would be suitable for the treatment of chronic pain syndromes. In the context of this review, we use the term “benzodiazepine” for all agonists at the benzodiazepine binding site of γ-aminobutyric acid type A receptors (GABAARs) independent of their chemical structure. It should also be mentioned here that GABAARs exist, which are resistant to modulation by classical benzodiazepines. These receptors contain α4 or α6 subunits instead of α1, α2, α3, or α5, or a γ1 or δ subunit instead of the γ2 subunit. These benzodiazepine-insensitive receptors are quite abundant in several brain regions (e.g., thalamus and cerebellum), but their expression in the spinal cord is sparse.

Section snippets

Synaptic Disinhibition in Pathological Pain

Fast synaptic inhibition in the spinal dorsal horn is mediated by GABA and glycine acting respectively at GABAAR and strychnine-sensitive GlyRs. Plenty of evidence indicates that blockade of spinal GABAARs or GlyRs produces signs of allodynia and spontaneous pain (Beyer et al., 1985, Miraucourt et al., 2007, Roberts et al., 1986). More recent studies provided insights into the mechanism of this sensitization on the level of dorsal horn neuronal circuits. The most consistent observation in these

Spinal GABAAR Subtypes Mediating Antihyperalgesia: Evidence from Genetically Engineered Mice

Analgesic or antihyperalgesic actions of benzodiazepines occur after local spinal injection suggesting that these effects are mediated by GABAARs expressed in the spinal cord. To identify the GABAAR subtypes responsible for these antihyperalgesic effects, “knock-in” mice were investigated, in which the α1, α2, α3, or α5-GABAAR subunits had been rendered diazepam-insensitive through the introduction of a histamine to arginine (H/R) point mutation (Knabl et al., 2008; for information on the

Mechanisms of Spinal Benzodiazepine-Mediated Antihyperalgesia

Immunohistochemistry studies have identified specific spinal distribution patterns of GABAAR subunits (Bohlhalter et al., 1996, Paul et al., 2012; Fig. 4A–D). These receptors are expressed on intrinsic dorsal horn neurons and on the central terminals of primary sensory nociceptors. Spinal antihyperalgesia may therefore originate either from classical postsynaptic inhibition mediated by GABAARs on intrinsic dorsal horn neurons or from GABAARs on nociceptor terminals which mediate presynaptic

Antihyperalgesic Action of Benzodiazepines with Improved Subtype Specificity: Preclinical Studies

A number of benzodiazepines with reduced activity at α1-GABAARs have been developed in the last two decades mainly in the quest for nonsedating anxiolytics (for a comprehensive list, see Rudolph & Knoflach, 2011). Because benzodiazepine-mediated anxiolysis and antihyperalgesia share a similar dependence on GABAAR subtypes, some of these compounds were also tested in pain studies (Table 1). It should be mentioned here that α1-sparing compounds are sometimes referred to as “α2/3 selective” (e.g.,

Clinical Studies on Antihyperalgesia by Benzodiazepines

The preclinical studies discussed above performed in mice resistant to the sedative effects of benzodiazepines demonstrate that classical benzodiazepines do in principle exert profound antihyperalgesic actions but only at doses, which normally induce strong sedation. Less-sedating benzodiazepines exhibited antihyperalgesic efficacy at nonsedating doses also in wild-type mice.

In human patients, classical nonselective benzodiazepines do not exert relevant analgesic (or antihyperalgesic) actions

Which GABAAR subtypes should be targeted for optimal analgesia with minimal side-effects?

Work in the GABAAR H/R point-mutated mice suggests that α2-, α3-, and α5-GABAARs contribute to spinal benzodiazepine antihyperalgesia. It is however not clear whether only one subtype (i.e., α2-GABAARs) should be targeted for optimal antihyperalgesia or whether simultaneous activity at more than one subunit would be advantageous. In the absence of fully selective subtype-specific drugs, investigations on mice carrying more than one point-mutated GABAAR subtype should be informative. When taking

Conclusion

There is compelling evidence from preclinical studies in rodents to support that non-sedative benzodiazepines with improved subtype specificity exert antihyperalgesic effects. Available clinical data are consistent with this view. Current knowledge suggests that robust antihyperalgesic activity with low sedative properties requires a high intrinsic activity at α2-GABAARs (or possibly also at α3-/α5-GABAARs) and very low activity at α1-GABAARs. The optimal profile of such a drug in terms of GABAA

Conflict of Interest Statement

The authors declare that they have no conflict of interest.

Acknowledgment

The research of H. U. Z. has been supported by grants from the Swiss National Science Foundation and by an Advanced Investigator Grant from the European Research Council (DHISP; 250128).

References (95)

  • J. Knabl et al.

    Genuine antihyperalgesia by systemic diazepam revealed by experiments in GABAA receptor point-mutated mice

    Pain

    (2009)
  • D.M. Kullmann et al.

    Presynaptic, extrasynaptic and axonal GABAA receptors in the CNS: Where and why?

    Progress in Biophysics & Molecular Biology

    (2005)
  • C. Luo et al.

    Synaptic plasticity in pathological pain

    Trends in Neurosciences

    (2014)
  • G. Munro et al.

    A question of balance–positive versus negative allosteric modulation of GABAA receptor subtypes as a driver of analgesic efficacy in rat models of inflammatory and neuropathic pain

    Neuropharmacology

    (2011)
  • S. Pauli et al.

    Occupancy of the central benzodiazepine receptors during benzodiazepine treatment determined by PET

    European Neuropsychopharmacology

    (1991)
  • T.J. Price et al.

    Protein expression and mRNA cellular distribution of the NKCC1 cotransporter in the dorsal root and trigeminal ganglia of the rat

    Brain Research

    (2006)
  • S. Reichl et al.

    Peripheral and spinal GABAergic regulation of incisional pain in rats

    Pain

    (2012)
  • A. Ribeiro-Da-Silva et al.

    Distribution of glomeruli with fluoride-resistant acid phosphatase (FRAP)-containing terminals in the substantia gelatinosa of the rat

    Brain Research

    (1986)
  • L.A. Roberts et al.

    Nociceptive responses to altered GABAergic activity at the spinal cord

    Life Sciences

    (1986)
  • E. Sigel et al.

    Structure, function, and modulation of GABAA receptors

    The Journal of Biological Chemistry

    (2012)
  • P. Skolnick

    Anxioselective anxiolytics: On a quest for the holy grail

    Trends in Pharmacological Sciences

    (2012)
  • M.A. Tatsuo et al.

    Midazolam-induced hyperalgesia in rats: Modulation via GABAA receptors at supraspinal level

    European Journal of Pharmacology

    (1999)
  • C. Vidal et al.

    Stress hyperalgesia in rats: An experimental animal model of anxiogenic hyperalgesia in human

    Life Sciences

    (1982)
  • H.U. Zeilhofer et al.

    GABAergic analgesia—New insights from mutant mice and subtype-selective agonists

    Trends in Pharmacological Sciences

    (2009)
  • B. Abrahamsen et al.

    The cell and molecular basis of mechanical, cold, and inflammatory pain

    Science

    (2008)
  • N. Agarwal et al.

    Conditional gene deletion in primary nociceptive neurons of trigeminal ganglia and dorsal root ganglia

    Genesis

    (2004)
  • S. Ahmadi et al.

    PGE2 selectively blocks inhibitory glycinergic neurotransmission onto rat superficial dorsal horn neurons

    Nature Neuroscience

    (2002)
  • F.J. Alvarez et al.

    Ultrastructural morphology, synaptic relationships, and CGRP immunoreactivity of physiologically identified C-fiber terminals in the monkey spinal cord

    The Journal of Comparative Neurology

    (1993)
  • J. Andre et al.

    Involvement of cholecystokininergic systems in anxiety-induced hyperalgesia in male rats: Behavioral and biochemical studies

    The Journal of Neuroscience

    (2005)
  • J.R. Atack et al.

    Preclinical and clinical pharmacology of TPA023B, a GABAA receptor α2/α3 subtype-selective partial agonist

    Journal of Psychopharmacology

    (2011)
  • J.R. Atack et al.

    MRK-409 (MK-0343), a GABAA receptor subtype-selective partial agonist, is a non-sedating anxiolytic in preclinical species but causes sedation in humans

    Journal of Psychopharmacology

    (2011)
  • J.R. Atack et al.

    TPA023 [7-(1,1-Dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluor ophenyl)-1,2,4-triazolo[4,3-b]pyridazine], an agonist selective for α2- and α3-containing GABAA receptors, is a nonsedating anxiolytic in rodents and primates

    The Journal of Pharmacology and Experimental Therapeutics

    (2006)
  • N.A. Ator et al.

    Reducing abuse liability of GABAA/benzodiazepine ligands via selective partial agonist efficacy at α1 and α2/3 subtypes

    The Journal of Pharmacology and Experimental Therapeutics

    (2010)
  • H. Baba et al.

    Peripheral inflammation facilitates Aβ fiber-mediated synaptic input to the substantia gelatinosa of the adult rat spinal cord

    The Journal of Neuroscience

    (1999)
  • M.L. Baccei et al.

    Development of GABAergic and glycinergic transmission in the neonatal rat dorsal horn

    The Journal of Neuroscience

    (2004)
  • E. Balic et al.

    The α5(H105R) mutation impairs α5 selective binding properties by altered positioning of the α5 subunit in GABAA receptors containing two distinct types of α subunits

    Journal of Neurochemistry

    (2009)
  • S. Bohlhalter et al.

    Laminar compartmentalization of GABAA-receptor subtypes in the spinal cord: An immunohistochemical study

    The Journal of Neuroscience

    (1996)
  • Y. Carrasquillo et al.

    Activation of the extracellular signal-regulated kinase in the amygdala modulates pain perception

    The Journal of Neuroscience

    (2007)
  • I. Colin et al.

    Localization of components of glycinergic synapses during rat spinal cord development

    The Journal of Comparative Neurology

    (1998)
  • J.A. Coull et al.

    BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain

    Nature

    (2005)
  • J.A. Coull et al.

    Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain

    Nature

    (2003)
  • F. Crestani et al.

    Molecular targets for the myorelaxant action of diazepam

    Molecular Pharmacology

    (2001)
  • G.R. Dawson et al.

    An inverse agonist selective for α5 subunit-containing GABAA receptors enhances cognition

    The Journal of Pharmacology and Experimental Therapeutics

    (2006)
  • S.L. de Haas et al.

    Pharmacodynamic and pharmacokinetic effects of MK-0343, a GABAA α2,3 subtype selective agonist, compared to lorazepam and placebo in healthy male volunteers

    Journal of Psychopharmacology

    (2008)
  • D.A. Fishbain et al.

    Clonazepam open clinical treatment trial for myofascial syndrome associated chronic pain

    Pain Medicine

    (2000)
  • S. Harkins et al.

    Administration of clonazepam in the treatment of TMD and associated myofascial pain: A double-blind pilot study

    Journal of Craniomandibular Disorders

    (1991)
  • J.A. Harris et al.

    Effects of benzodiazepine microinjection into the amygdala or periaqueductal gray on the expression of conditioned fear and hypoalgesia in rats

    Behavioral Neuroscience

    (1995)
  • Cited by (39)

    • The imidazodiazepine, KRM-II-81: An example of a newly emerging generation of GABAkines for neurological and psychiatric disorders

      2022, Pharmacology Biochemistry and Behavior
      Citation Excerpt :

      With either chemoexcitant, highly significant reductions in firing rate were produced by KRM-II-81. Despite the pervasive role of GABA in pain regulation (Hammond and Drower, 1984; Dirig and Yaksh, 1995; Enna and McCarson, 2006; Zeilhofer et al., 2015; Etlin et al., 2016), GABAkines have not been effectively made into medicines for pain moderation or long-term therapy. There is, however, a huge gap in the need for new pain medications that typically rely on opioid receptor mechanisms (O'Brien and Roman, 2021).

    • ‘Proximity frequencies’ a new parameter to evaluate the profile of GABA<inf>A</inf>R modulators

      2021, Bioorganic and Medicinal Chemistry Letters
      Citation Excerpt :

      In particular, regarding the GABAA subtype receptors (α1-3,5-β2 γ2 GABAARs), modulated by benzodiazepine-like and non-benzodiazepine ligands, the type of isoform alpha addresses not only the affinity but also the pharmacological activity. Thus, the α1-subunit containing GABAARs mediate the sedating and hypnotic effect, while the α2-subunit receptors mediate the anxiolytic and antihyperalgesic effect; the α3-subunit receptors seem involved in the myorelaxant activity and the α 5-subunit receptors show modulation of cognitive processes related memory.11–14 In this manuscript, we reported the studies of further exploration of the pyrazolo[1,5-a]quinazoline scaffold.

    View all citing articles on Scopus
    View full text