Dynein regulates Kv7.4 channel trafficking from the cell membrane

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Dynein regulates Kv7.4 channel trafficking from the cell membrane. / van der Horst, Jennifer; Rognant, Salomé; Abbott, Geoffrey W; Ozhathil, Lijo Cherian; Hägglund, Per; Barrese, Vincenzo; Chuang, Christine Y; Jespersen, Thomas; Davies, Michael J; Greenwood, Iain A; Gourdon, Pontus; Aalkjær, Christian; Jepps, Thomas A.

In: Journal of General Physiology, Vol. 153, No. 3, e202012760, 2021.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

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', Journal of General Physiology, vol. 153, no. 3, e202012760. https://doi.org/10.1085/JGP.202012760

APA

van der Horst, J., Rognant, S., Abbott, G. W., Ozhathil, L. C., Hägglund, P., Barrese, V., Chuang, C. Y., Jespersen, T., Davies, M. J., Greenwood, I. A., Gourdon, P., Aalkjær, C., & Jepps, T. A. (2021). Dynein regulates Kv7.4 channel trafficking from the cell membrane. Journal of General Physiology, 153(3), [e202012760]. https://doi.org/10.1085/JGP.202012760

Vancouver

van der Horst J, Rognant S, Abbott GW, Ozhathil LC, Hägglund P, Barrese V et al. Dynein regulates Kv7.4 channel trafficking from the cell membrane. Journal of General Physiology. 2021;153(3). e202012760. https://doi.org/10.1085/JGP.202012760

Author

van der Horst, Jennifer ; Rognant, Salomé ; Abbott, Geoffrey W ; Ozhathil, Lijo Cherian ; Hägglund, Per ; Barrese, Vincenzo ; Chuang, Christine Y ; Jespersen, Thomas ; Davies, Michael J ; Greenwood, Iain A ; Gourdon, Pontus ; Aalkjær, Christian ; Jepps, Thomas A. / Dynein regulates Kv7.4 channel trafficking from the cell membrane. In: Journal of General Physiology. 2021 ; Vol. 153, No. 3.

Bibtex

@article{3900c060bbd64cb7a98c4eb0ffe49f43,
title = "Dynein regulates Kv7.4 channel trafficking from the cell membrane",
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.",
author = "{van der Horst}, Jennifer and Salom{\'e} Rognant and Abbott, {Geoffrey W} and Ozhathil, {Lijo Cherian} and Per H{\"a}gglund and Vincenzo Barrese and Chuang, {Christine Y} and Thomas Jespersen and Davies, {Michael J} and Greenwood, {Iain A} and Pontus Gourdon and Christian Aalkj{\ae}r and Jepps, {Thomas A}",
note = "(Ekstern)",
year = "2021",
doi = "10.1085/JGP.202012760",
language = "English",
volume = "153",
journal = "Journal of General Physiology",
issn = "0022-1295",
publisher = "Rockefeller University Press",
number = "3",

}

RIS

TY - JOUR

T1 - Dynein regulates Kv7.4 channel trafficking from the cell membrane

AU - van der Horst, Jennifer

AU - Rognant, Salomé

AU - Abbott, Geoffrey W

AU - Ozhathil, Lijo Cherian

AU - Hägglund, Per

AU - Barrese, Vincenzo

AU - Chuang, Christine Y

AU - Jespersen, Thomas

AU - Davies, Michael J

AU - Greenwood, Iain A

AU - Gourdon, Pontus

AU - Aalkjær, Christian

AU - Jepps, Thomas A

N1 - (Ekstern)

PY - 2021

Y1 - 2021

N2 - 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.

AB - 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.

UR - http://www.scopus.com/inward/record.url?scp=85101299544&partnerID=8YFLogxK

U2 - 10.1085/JGP.202012760

DO - 10.1085/JGP.202012760

M3 - Journal article

C2 - 33533890

AN - SCOPUS:85101299544

VL - 153

JO - Journal of General Physiology

JF - Journal of General Physiology

SN - 0022-1295

IS - 3

M1 - e202012760

ER -

ID: 257748257