Abstract

In response to diverse stimuli, two-pore-domain potassium channel TREK-2 regulates cellular excitability, and hence plays a key role in mediating neuropathic pain, mood disorders and ischemia through. Although more and more input modalities are found to achieve their modulations via acting on the channel, the potential role of subunit interaction in these modulations remains to be explored. In the current study, the deletion (lack of proximal C-terminus, ΔpCt) or point mutation (G312A) was introduced into TREK-2 subunits to limit K+ conductance and used to report subunit stoichiometry. The constructs were then combined with wild type (WT) subunit to produce concatenated dimers with defined composition, and the gating kinetics of these channels to 2-Aminoethoxydiphenyl borate (2-APB) and extracellular pH (pHo) were characterized. Our results show that combination of WT and ΔpCt/G312A subunits reserves similar gating properties to that of WT dimmers, suggesting that the WT subunit exerts dominant and positive effects on the mutated one, and thus the two subunits controls channel gating via a concerted cooperative manner. Further introduction of ΔpCt into the latter subunit of heterodimeric channel G312A-WT or G312A-G312A attenuated their sensitivity to 2-APB and pHo alkalization, implicating that these signals were transduced by a cis-type mechanism. Together, our findings elucidate the mechanisms for how the two subunits control the pore gating of TREK-2, in which both intersubunit concerted cooperative and cis-type manners modulate the allosteric regulations induced by 2-APB and pHo alkalization.

Highlights

  • Two-pore domain K+ (K2P) channels, the last discovered K+ channel family, are major contributors to background K+ conductance by producing ‘‘leak’’ currents, and play a predominant role in stabilizing resting membrane potential as well as regulating cellular excitability

  • We investigated the effects of these mutations or deletions in the context of tandem-linked dimers on both 2-Aminoethoxydiphenyl borate (2-APB) pathway and pHo pathways, and found that concerted cooperative intersubunit interaction and cis-type transduction along single subunit underlie the allosteric regulations of TREK-2 channels

  • By determining the effects of 2-APB and extracellular alkalization on concatenated dimers comprised of wild type (WT) TREK-2 and loss of function mutants (∆proximal C-terminus (pCt) and G312A), we demonstrate that the two subunits gate channel in a cooperative concerted way, and show that the allosteric regulations evoked by 2-APB and pHo changes occur in a cis-type mechanism

Read more

Summary

Introduction

Two-pore domain K+ (K2P) channels, the last discovered K+ channel family, are major contributors to background K+ conductance by producing ‘‘leak’’ currents, and play a predominant role in stabilizing resting membrane potential as well as regulating cellular excitability. In addition to central nervous system (Lesage et al, 2000), the channel is highly expressed in dorsal root ganglia (DRG) and trigeminal ganglia (Kang and Kim, 2006; Yamamoto et al, 2009; Acosta et al, 2014). In DRG C-fiber nociceptors, the functions of TREK-2 are involved in hyperpolarizing membrane potential and limiting spontaneous pain (Kang and Kim, 2006; Acosta et al, 2014). TREK-2 expression is up-regulated in the astrocytic membrane (Rivera-Pagán et al, 2015), cortical and hippocampal neurons (Li et al, 2005). TREK-2 has been implicated to be a new potential therapeutic target for treating neuropathic pain, mood disorders and ischemia

Objectives
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