Repository logo
 

Supramolecular clustering of the cardiac sodium channel Nav1.5 in HEK293F cells, with and without the auxiliary β3-subunit.

Accepted version
Peer-reviewed

Type

Article

Change log

Authors

Salvage, Samantha C 
Rees, Johanna S 
Hirsch, Michael 
Wang, Lin 

Abstract

Voltage-gated sodium channels comprise an ion-selective α-subunit and one or more associated β-subunits. The β3-subunit (encoded by the SCN3B gene) is an important physiological regulator of the heart-specific sodium channel, Nav1.5. We have previously shown that when expressed alone in HEK293F cells, the full-length β3-subunit forms trimers in the plasma membrane. We extend this result with biochemical assays and use the proximity ligation assay (PLA) to identify oligomeric β3-subunits, not just at the plasma membrane, but throughout the secretory pathway. We then investigate the corresponding clustering properties of the α-subunit and the effects upon these of the β3-subunits. The oligomeric status of the Nav1.5 α-subunit in vivo, with or without the β3-subunit, has not been previously investigated. Using super-resolution fluorescence imaging, we show that under conditions typically used in electrophysiological studies, the Nav1.5 α-subunit assembles on the plasma membrane of HEK293F cells into spatially localized clusters rather than individual and randomly dispersed molecules. Quantitative analysis indicates that the β3-subunit is not required for this clustering but β3 does significantly change the distribution of cluster sizes and nearest-neighbor distances between Nav1.5 α-subunits. However, when assayed by PLA, the β3-subunit increases the number of PLA-positive signals generated by anti-(Nav1.5 α-subunit) antibodies, mainly at the plasma membrane. Since PLA can be sensitive to the orientation of proteins within a cluster, we suggest that the β3-subunit introduces a significant change in the relative alignment of individual Nav1.5 α-subunits, but the clustering itself depends on other factors. We also show that these structural and higher-order changes induced by the β3-subunit do not alter the degree of electrophysiological gating cooperativity between Nav1.5 α-subunits. Our data provide new insights into the role of the β3-subunit and the supramolecular organization of sodium channels, in an important model cell system that is widely used to study Nav channel behavior.

Description

Keywords

Hodgkin Huxley kinetics, super-resolution imaging, voltage-gated sodium channel Nav1.5, β3-subunit, Cell Membrane, Electrophysiology, HEK293 Cells, Humans, Immunoprecipitation, Kinetics, NAV1.5 Voltage-Gated Sodium Channel, Patch-Clamp Techniques, Protein Subunits

Journal Title

FASEB J

Conference Name

Journal ISSN

0892-6638
1530-6860

Volume Title

34

Publisher

Wiley

Rights

All rights reserved
Sponsorship
British Heart Foundation (None)
Wellcome Trust (105727/Z/14/Z)
We would like to thank the Gurdon Institute Imaging Facility for use of their microscope and general assistance. This work was supported by a British Heart Foundation grant (PG/14/79/31102) to APJ and CLHH, The Wellcome Trust, award number: 105727/Z/14/Z to CLHH and a Medical Research Council grant (MR/K015591/1) to CLF, RAL, and STFC.