Inhibition of Inactive States of Tetrodotoxin-Sensitive Sodium Channels Reduces Spontaneous Firing of C-Fiber Nociceptors and Produces Analgesia in Formalin and Complete Freund's Adjuvant Models of Pain

PLoS One. 2015 Sep 17;10(9):e0138140. doi: 10.1371/journal.pone.0138140. eCollection 2015.

Abstract

While genetic evidence shows that the Nav1.7 voltage-gated sodium ion channel is a key regulator of pain, it is unclear exactly how Nav1.7 governs neuronal firing and what biophysical, physiological, and distribution properties of a pharmacological Nav1.7 inhibitor are required to produce analgesia. Here we characterize a series of aminotriazine inhibitors of Nav1.7 in vitro and in rodent models of pain and test the effects of the previously reported "compound 52" aminotriazine inhibitor on the spiking properties of nociceptors in vivo. Multiple aminotriazines, including some with low terminal brain to plasma concentration ratios, showed analgesic efficacy in the formalin model of pain. Effective concentrations were consistent with the in vitro potency as measured on partially-inactivated Nav1.7 but were far below concentrations required to inhibit non-inactivated Nav1.7. Compound 52 also reversed thermal hyperalgesia in the complete Freund's adjuvant (CFA) model of pain. To study neuronal mechanisms, electrophysiological recordings were made in vivo from single nociceptive fibers from the rat tibial nerve one day after CFA injection. Compound 52 reduced the spontaneous firing of C-fiber nociceptors from approximately 0.7 Hz to 0.2 Hz and decreased the number of action potentials evoked by suprathreshold tactile and heat stimuli. It did not, however, appreciably alter the C-fiber thresholds for response to tactile or thermal stimuli. Surprisingly, compound 52 did not affect spontaneous activity or evoked responses of Aδ-fiber nociceptors. Results suggest that inhibition of inactivated states of TTX-S channels, mostly likely Nav1.7, in the peripheral nervous system produces analgesia by regulating the spontaneous discharge of C-fiber nociceptors.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Action Potentials / physiology
  • Analgesia / methods
  • Analgesics / therapeutic use*
  • Animals
  • Formaldehyde / pharmacology
  • Freund's Adjuvant / pharmacology
  • Male
  • NAV1.7 Voltage-Gated Sodium Channel / metabolism*
  • Nerve Fibers, Unmyelinated / metabolism*
  • Nociceptors / metabolism*
  • Pain / chemically induced
  • Pain / drug therapy*
  • Pain Management
  • Pain Measurement / methods
  • Patch-Clamp Techniques
  • Rats
  • Rats, Sprague-Dawley
  • Sodium Channel Blockers / pharmacology
  • Tetrodotoxin / pharmacology*

Substances

  • Analgesics
  • NAV1.7 Voltage-Gated Sodium Channel
  • Scn9a protein, rat
  • Sodium Channel Blockers
  • Formaldehyde
  • Tetrodotoxin
  • Freund's Adjuvant

Grants and funding

Authors DJM, HB, MC, EFD, LH, DJ, XL, JM, CM, BW, ABM, and SIM were Amgen employees at the time the work was performed. This work was fully supported by Amgen Inc. Amgen Inc. provided support in the form of salaries for these authors, but did not have any additional role in the study design, data collection and analysis, decision to publish, or preparation of this manuscript.