SORA

Advancing, promoting and sharing knowledge of health through excellence in teaching, clinical practice and research into the prevention and treatment of illness

Dynein regulates Kv7.4 channel trafficking from the cell membrane.

van der Horst, J; Rognant, S; Abbott, GW; Ozhathil, LC; Hägglund, P; Barrese, V; Chuang, CY; Jespersen, T; Davies, MJ; Greenwood, IA; et al. van der Horst, J; Rognant, S; Abbott, GW; Ozhathil, LC; Hägglund, P; Barrese, V; Chuang, CY; Jespersen, T; Davies, MJ; Greenwood, IA; Gourdon, P; Aalkjær, C; Jepps, TA (2021) Dynein regulates Kv7.4 channel trafficking from the cell membrane. J Gen Physiol, 153 (3). e202012760. ISSN 1540-7748 https://doi.org/10.1085/jgp.202012760
SGUL Authors: Greenwood, Iain Andrew

[img]
Preview
PDF Published Version
Available under License Creative Commons Attribution Non-commercial Share Alike.

Download (3MB) | Preview

Abstract

The dynein motor protein transports proteins away from the cell membrane along the microtubule network. Recently, we found the microtubule network was important for regulating the membrane abundance of voltage-gated Kv7.4 potassium channels in vascular smooth muscle. Here, we aimed to investigate the influence of dynein on the microtubule-dependent internalization of the Kv7.4 channel. Patch-clamp recordings from HEK293B cells showed Kv7.4 currents were increased after inhibiting dynein function with ciliobrevin D or by coexpressing p50/dynamitin, which specifically interferes with dynein motor function. Mutation of a dynein-binding site in the Kv7.4 C terminus increased the Kv7.4 current and prevented p50 interference. Structured illumination microscopy, proximity ligation assays, and coimmunoprecipitation showed colocalization of Kv7.4 and dynein in mesenteric artery myocytes. Ciliobrevin D enhanced mesenteric artery relaxation to activators of Kv7.2-Kv7.5 channels and increased membrane abundance of Kv7.4 protein in isolated smooth muscle cells and HEK293B cells. Ciliobrevin D failed to enhance the negligible S-1-mediated relaxations after morpholino-mediated knockdown of Kv7.4. Mass spectrometry revealed an interaction of dynein with caveolin-1, confirmed using proximity ligation and coimmunoprecipitation assays, which also provided evidence for interaction of caveolin-1 with Kv7.4, confirming that Kv7.4 channels are localized to caveolae in mesenteric artery myocytes. Lastly, cholesterol depletion reduced the interaction of Kv7.4 with caveolin-1 and dynein while increasing the overall membrane expression of Kv7.4, although it attenuated the Kv7.4 current in oocytes and interfered with the action of ciliobrevin D and channel activators in arterial segments. Overall, this study shows that dynein can traffic Kv7.4 channels in vascular smooth muscle in a mechanism dependent on cholesterol-rich caveolae.

Item Type: Article
Additional Information: © 2021 van der Horst et al. This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the first six months after the publication date (see http://www.rupress.org/terms/). After six months it is available under a Creative Commons License (Attribution–Noncommercial–Share Alike 4.0 International license, as described at https://creativecommons.org/licenses/by-nc-sa/4.0/).
Keywords: 0606 Physiology, 1116 Medical Physiology, Physiology
SGUL Research Institute / Research Centre: Academic Structure > Molecular and Clinical Sciences Research Institute (MCS)
Journal or Publication Title: J Gen Physiol
ISSN: 1540-7748
Language: eng
Dates:
DateEvent
1 March 2021Published
3 February 2021Published Online
8 January 2021Accepted
Publisher License: Creative Commons: Attribution-Noncommercial-Share Alike 4.0
Projects:
Project IDFunderFunder ID
CF16-0136Carlsberg FoundationUNSPECIFIED
R323-2018-367Lundbeck FoundationUNSPECIFIED
9039-00409BDanmarks Frie ForskningsfondUNSPECIFIED
801199Horizon 2020UNSPECIFIED
NNF13OC0004294Novo Nordiskhttp://dx.doi.org/10.13039/501100004191
NNF18OC0031634Novo Nordiskhttp://dx.doi.org/10.13039/501100004191
GM130377National Institute of General Medical Scienceshttp://dx.doi.org/10.13039/100000057
NS107671National Institute of Neurological Disorders and Strokehttp://dx.doi.org/10.13039/100000065
PubMed ID: 33533890
Go to PubMed abstract
URI: https://openaccess.sgul.ac.uk/id/eprint/112943
Publisher's version: https://doi.org/10.1085/jgp.202012760

Actions (login required)

Edit Item Edit Item