1. Academic Validation
  2. The Tarantula Venom Peptide Eo1a Binds to the Domain II S3-S4 Extracellular Loop of Voltage-Gated Sodium Channel NaV1.8 to Enhance Activation

The Tarantula Venom Peptide Eo1a Binds to the Domain II S3-S4 Extracellular Loop of Voltage-Gated Sodium Channel NaV1.8 to Enhance Activation

  • Front Pharmacol. 2022 Jan 14;12:789570. doi: 10.3389/fphar.2021.789570.
Jennifer R Deuis 1 Lotten Ragnarsson 1 Samuel D Robinson 1 Zoltan Dekan 1 Lerena Chan 1 Ai-Hua Jin 1 Poanna Tran 1 Kirsten L McMahon 1 Shengnan Li 2 John N Wood 2 James J Cox 2 Glenn F King 1 3 Volker Herzig 4 5 Irina Vetter 1 6
Affiliations

Affiliations

  • 1 Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
  • 2 Wolfson Institute for Biomedical Research, University College London, London, United Kingdom.
  • 3 Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, Australia.
  • 4 GeneCology Research Centre, University of the Sunshine Coast, Sippy Downs, QLD, Australia.
  • 5 School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia.
  • 6 School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
Abstract

Venoms from cone snails and arachnids are a rich source of peptide modulators of voltage-gated sodium (NaV) channels, however relatively few venom-derived Peptides with activity at the mammalian NaV1.8 subtype have been isolated. Here, we describe the discovery and functional characterisation of β-theraphotoxin-Eo1a, a peptide from the venom of the Tanzanian black and olive baboon tarantula Encyocratella olivacea that modulates NaV1.8. Eo1a is a 37-residue peptide that increases NaV1.8 peak current (EC50 894 ± 146 nM) and causes a large hyperpolarising shift in both the voltage-dependence of activation (ΔV50-20.5 ± 1.2 mV) and steady-state fast inactivation (ΔV50-15.5 ± 1.8 mV). At a concentration of 10 μM, Eo1a has varying effects on the peak current and channel gating of NaV1.1-NaV1.7, although its activity is most pronounced at NaV1.8. Investigations into the binding site of Eo1a using NaV1.7/NaV1.8 chimeras revealed a critical contribution of the DII S3-S4 extracellular loop of NaV1.8 to toxin activity. Results from this work may form the basis for future studies that lead to the rational design of spider venom-derived Peptides with improved potency and selectivity at NaV1.8.

Keywords

Nav1.8; pain; peptide; spider; voltage-gated sodium channel.

Figures