Abstract

See Basbaum (doi:10.1093/brain/awx227) for a scientific commentary on this article. Peripheral neuropathic pain arises as a consequence of injury to sensory neurons; the development of ectopic activity in these neurons is thought to be critical for the induction and maintenance of such pain. Local anaesthetics and anti-epileptic drugs can suppress hyperexcitability; however, these drugs are complicated by unwanted effects on motor, central nervous system and cardiac function, and alternative more selective treatments to suppress hyperexcitability are therefore required. Here we show that a glutamate-gated chloride channel modified to be activated by low doses of ivermectin (but not glutamate) is highly effective in silencing sensory neurons and reversing neuropathic pain-related hypersensitivity. Activation of the glutamate-gated chloride channel expressed in either rodent or human induced pluripotent stem cell-derived sensory neurons in vitro potently inhibited their response to both electrical and algogenic stimuli. We have shown that silencing is achieved both at nerve terminals and the soma and is independent of membrane hyperpolarization and instead likely mediated by lowering of the membrane resistance. Using intrathecal adeno-associated virus serotype 9-based delivery, the glutamate-gated chloride channel was successfully targeted to mouse sensory neurons in vivo, resulting in high level and long-lasting expression of the channel selectively in sensory neurons. This enabled reproducible and reversible modulation of thermal and mechanical pain thresholds in vivo; analgesia was observed for 3 days after a single systemic dose of ivermectin. We did not observe any motor or proprioceptive deficits and noted no reduction in cutaneous afferent innervation or upregulation of the injury marker ATF3 following prolonged glutamate-gated chloride channel expression. Established mechanical and cold pain-related hypersensitivity generated by the spared nerve injury model of neuropathic pain was reversed by ivermectin treatment. The efficacy of ivermectin in ameliorating behavioural hypersensitivity was mirrored at the cellular level by a cessation of ectopic activity in sensory neurons. These findings demonstrate the importance of aberrant afferent input in the maintenance of neuropathic pain and the potential for targeted chemogenetic silencing as a new treatment modality in neuropathic pain.

Highlights

  • Neuropathic pain arises as a consequence of a lesion or disease of the somatosensory nervous system and affects 7–10% of the general population (Colloca et al, 2017)

  • Such abnormalities have been demonstrated in multiple animal models of peripheral nerve injury (Kajander and Bennett, 1992; Boucher et al, 2000; Wu et al, 2001) as well as in patients suffering from peripheral neuropathic pain through the use of microneurography (Serra et al, 2012)

  • To assess the efficacy of GluCl for silencing sensory neurons, we first performed whole-cell patch clamp recordings of dissociated mouse dorsal root ganglia (DRG) neurons electroporated with GluCl plasmid DNA

Read more

Summary

Introduction

Neuropathic pain arises as a consequence of a lesion or disease of the somatosensory nervous system and affects 7–10% of the general population (Colloca et al, 2017). A key pathophysiological driver for the induction and maintenance of neuropathic pain in such conditions is the development of hyperexcitability of sensory neurons, which develop ectopic activity (Wall and Gutnick, 1974; Wall and Devor, 1983; Kajander and Bennett, 1992; Boucher et al, 2000; Wu et al, 2001) and an abnormal response to natural stimuli (Shim et al, 2005; Smith et al, 2013) Such abnormalities have been demonstrated in multiple animal models of peripheral nerve injury (Kajander and Bennett, 1992; Boucher et al, 2000; Wu et al, 2001) as well as in patients suffering from peripheral neuropathic pain through the use of microneurography (Serra et al, 2012). This hyperexcitability occurs due to profound changes in the expression as well as altered trafficking and post-translational modifications of ion channels within sensory neurons (Waxman and Zamponi, 2014)

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call