AuPS Logo Programme
Contents
Previous Next PDF

Na+ H+ exchanger regulatory factor 2 (NHERF-2) is a scaffold for the plasma membrane Ca2+ ATPase (PMCA)

W.A. Kruger1, G.R. Monteith2, L. Tongpao1 and P. Poronnik1, 1School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia and 2School of Pharmacy, The University of Queensland, St Lucia, QLD 4072, Australia.

Resting cytosolic Ca2+ levels are maintained at nanomolar levels by the sequestration of Ca2+ into intracellular stores or the extrusion of Ca2+ across the plasma membrane by the PMCA. Despite the ubiquitous distribution of PMCA and its pivotal role in Ca2+ signalling, little is known about how PMCA activity is regulated during G protein coupled receptor signalling. There are 4 isoforms of PMCA (1-4) and many splice variants of all isoforms have been identified and all PMCA-b splice variants have a consensus class 1 PSD-95/Dlg/Zo-1 (PDZ) binding motif (Strehler et al., 2001). Protein-protein interactions mediated by PDZ modules are now recognized as playing a key role in spatially constraining many ion channels and transporters into signalling complexes in membrane microdomains (Pawson et al., 1997). Previously, PMCA 2b has been reported to interact with NHERF-2 in a heterologous expression system (DeMarco et al., 2003). This study investigated whether PMCA interacts with NHERF-2 in a native epithelial cell and the physiological significance of this interaction in terms of G-protein mediated Ca2+ signalling via the muscarinic M3 receptor.

This study used the polarised epithelial HT29 cell line which expresses only the M3 isoform of the muscarinic receptor. RT-PCR and Western blotting were used to confirm the presence of both PMCA and NHERF-2 in these cells. Cell surface biotinylations were performed to investigate the changes in levels of PMCA at the plasma membrane following activation of M3 receptor by acetylcholine (ACh). NHERF-2 contains 3 binding domains, PDZ-1, PDZ-2 and the C-terminus. We used GST-fusions of these domains as well as full length NHERF-2 to characterise the interaction between NHERF-2 and PMCA. These interactions were validated in vivo using co-immunoprecipitation with a polyclonal NHERF-2 antibody and subsequent Western blotting with a pan-PMCA antibody. To examine the functional role of NHERF-2, endogenous protein was knocked down by transfecting siRNA plasmids. Changes in intracellular Ca2+ were measured using FURA-2 in a microplate assay.

RT-PCR and Western blots confirmed that HT29 cells expressed both NHERF-2 and PMCA isoform 1 and 4 (n = 3). Importantly, we found that the levels of PMCA at the plasma membrane increased by 62 ± 12% (n = 3) within 1 min of exposure to ACh and returned to control levels within 3 min. GST-pulldown experiments in HT29 cell lysates clearly showed that PMCA interacted with the second PDZ module of NHERF-2 (n=4). Co-immunoprecipitation experiments using HT29 cell lysates confirmed the interaction between NHERF-2 and PMCA occurred under in vivo conditions (n=3). Silencing of NHERF-2 reduced the levels of endogenous NHERF-2 by a 68 ± 10% (n = 3). When we examined the Ca2+ response to ACh in the cells where NHERF-2 had been silenced we observed that the rate of recovery from the peak Ca2+ transient was 50 ± 10% (n = 3; P < 0.05) faster than in control cells.

These data reveal for the first time that the increase in intracellular Ca2+ in response to M3 receptor activation is accompanied by a rapid increase in PMCA at the plasma membrane, presumably due to translocation from subplasmalemmal stores. The functional interaction between NHERF-2 and PMCA may underlie the changes in recovery rate of Ca2+ following exposure to ACh. Further studies will provide new insights into how scaffold proteins may confer specificity in terms of G-protein mediated Ca2+ signalling.

Demarco, S., Chicka, M. & Strehler, E. (2003) Journal of Biological Chemistry 277, 10506-11.

Pawson, T. & Scott, J. (1997) Science 278, 2075-80.

Strehler, E. & Zacharias, D. (2001) Physiological Reviews 81, 21-50.