Link to the University of Pittsburgh Homepage
Link to the University Library System Homepage Link to the Contact Us Form

ENAC REGULATION IN THE KIDNEY: THE ROLE OF ANKYRIN G

Klemens, Christine A. (2017) ENAC REGULATION IN THE KIDNEY: THE ROLE OF ANKYRIN G. Doctoral Dissertation, University of Pittsburgh. (Unpublished)

[img]
Preview
PDF
Download (2MB) | Preview

Abstract

The epithelial sodium channel (ENaC) is the limiting entry point for Na+ reabsorption in the distal kidney nephron and is regulated by numerous hormones, including the mineralocorticoid hormone aldosterone. Previously we identified ankyrin G (AnkG), a cytoskeletal protein involved in vesicular transport, as a novel aldosterone-induced protein that can alter Na+ transport in mouse cortical collecting duct cells. AnkG is highly expressed in the kidney, particularly in the distal nephron. Increasing AnkG expression increases ENaC activity while depleting AnkG reduces ENaC-mediated Na+ transport. The underlying mechanism presiding over this effect; however, was unknown. Here we report that AnkG expression directly regulates Na+ transport by altering ENaC activity in the apical membrane. These changes are due to a change in ENaC directly rather than through alterations to the Na+ driving force created by Na+K+ATPase. Using a constitutively open mutant of ENaC and surface biotinylation, we demonstrate that the augmentation of Na+ transport is caused predominantly by increasing the number of ENaCs at the surface rather than alterations to open probability. To determine the mechanism of AnkG action on ENaC surface number, changes in rates of internalization, recycling, and membrane delivery were investigated. AnkG did not alter ENaC delivery to the membrane from biosynthetic pathways or removal by endocytosis; however, AnkG did alter ENaC insertion from constitutive recycling pathways. We also investigated the potential role of a putative AnkG binding domain in the C-terminus of βENaC, and whether single-site mutations of a charged residue and two regulatory phosphorylation sites could disrupt AnkG augmentation of ENaC current. We did not find any significant evidence that this region is essential for AnkG-ENaC interaction. These findings provide a mechanism to account for the role of AnkG in the regulation of Na+ transport in the distal kidney nephron.


Share

Citation/Export:
Social Networking:
Share |

Details

Item Type: University of Pittsburgh ETD
Status: Unpublished
Creators/Authors:
CreatorsEmailPitt UsernameORCID
Klemens, Christine A.
ETD Committee:
TitleMemberEmail AddressPitt UsernameORCID
Committee ChairKleyman, Thomas R.
Thesis AdvisorButterworth, Michael B.
Committee MemberDeFranco, Donald B.
Committee MemberDevor, Daniel C.
Committee MemberKwiatkowski, Adam V.
Date: 18 May 2017
Date Type: Publication
Defense Date: 11 April 2017
Approval Date: 18 May 2017
Submission Date: 17 May 2017
Access Restriction: No restriction; Release the ETD for access worldwide immediately.
Number of Pages: 158
Institution: University of Pittsburgh
Schools and Programs: School of Medicine > Cell Biology and Molecular Physiology
Degree: PhD - Doctor of Philosophy
Thesis Type: Doctoral Dissertation
Refereed: Yes
Uncontrolled Keywords: Kidney ENaC Epithelial Sodium Channel Ankyrin AnkG ANK3 Ion Transport Trafficking
Date Deposited: 18 May 2017 15:07
Last Modified: 18 May 2017 15:07
URI: http://d-scholarship.pitt.edu/id/eprint/32040

Metrics

Monthly Views for the past 3 years

Plum Analytics


Actions (login required)

View Item View Item