Elsevier

Cellular Signalling

Volume 20, Issue 10, October 2008, Pages 1769-1779
Cellular Signalling

Identification of a novel di-leucine motif mediating K+/Cl cotransporter KCC2 constitutive endocytosis

https://doi.org/10.1016/j.cellsig.2008.06.011Get rights and content

Abstract

The neuron-specific potassium-chloride cotransporter 2 (KCC2) plays a crucial role, by controlling chloride extrusion, in the development and maintenance of inhibitory neurotransmission. Although it is now well established that activity-dependent mechanisms can down regulate KCC2 gene expression, the role of post-translational mechanisms in controlling KCC2 expression, specifically at the cell-surface, are poorly understood. We therefore set out to identify the mechanisms and motifs regulating KCC2 endocytosis, one important pathway that may control KCC2 membrane expression. Using a fluorescence-based assay, we show KCC2 when expressed in HEK293 cells is constitutively internalized via a dynamin- and clathrin-dependent pathway. Consistent with this, we demonstrate KCC2 from adult mouse brain associates in vivo with the clathrin-binding adaptor protein-2 (AP-2) complex. Using an endocytosis reporter system, we identify the presence of an autonomous endocytosis motif in the carboxyl cytoplasmic terminus of KCC2. By site-directed mutagenesis we define this novel KCC2 endocytic motif as a non-canonical di-leucine motif, 657LLXXEE662. Finally by mutating this motif in the context of full-length KCC2 we demonstrate that this novel KCC2 endocytic motif is essential for the constitutive internalization of KCC2 and for binding to the AP-2 complex. Subsequent sequence analysis reveals this motif is highly conserved between the closely related K+/Cl family members that mediate chloride efflux, but absent from the more distant related cotransporters controlling chloride influx. In conclusion, our results indicate constitutive internalization of KCC2 is clathrin-mediated and dependent on the binding of AP-2 to this novel endocytic motif. Furthermore, that this process appears to be an evolutionarily conserved mechanism amongst functionally homologous cotransporters.

Introduction

Fast inhibitory neurotransmission is mediated by two classes of ligand gated Cl channels, the γ-amino-butyric acid type-A receptor (GABAAR) and the glycine receptor (GlyR) [1], [2]. In each case Cl influx is triggered upon channel opening resulting in hyperpolarization of the postsynaptic membrane. This in turn, leads to a reduction in the likelihood of further neurotransmitter release [1], [2]. In the majority of neurons, Cl influx and fast hyperpolarizing inhibition are critically dependent on low intracellular chloride concentrations ([Cl]i). The neuron-specific K+–Cl cotransporter, KCC2, has now been identified as an essential protein in establishing and maintaining this low [Cl]i, by controlling Cl extrusion [3].

During central nervous system (CNS) development, KCC2 gene expression is upregulated and underlies the transition of GABA and glycine responses from the immature depolarizing to the hyperpolarizing responses found in adults [3]. This transition occurs as the overall developmental increase in KCC2 activity leads to a reduction in [Cl]i, which at resting membrane potentials promotes Cl influx (hyperpolarization) rather than efflux upon ion channel opening [3]. Further, in mature neurons a reduction in KCC2 gene expression, via antisense oligonucleotide suppression, leads to an increase in [Cl]i and a shift in GABAAR responses to immature depolarizing [3]. These observations have demonstrated that dynamic regulation of KCC2 gene expression can alter the direction of GABAAR and GlyR signaling. Consistent with this and its essential role in inhibition, KCC2 knockout mice die shortly after birth [4]. In addition, a loss of KCC2 expression is recognized as a contributing factor in the pathological conditions of chronic pain [5], nerve injury [6] and epilepsy [7], [8].

Besides the above-mentioned regulation of KCC2 gene expression, short-term modulation of the KCC2 protein has also been demonstrated. Several kinase activities can modulate KCC2 Cl transport activity in both immature [9], [10], [11], [12] and mature [13] hippocampal neurons. The precise molecular mechanisms involved however, have yet to be eluded. In addition a rapid loss of KCC2 cell surface expression has been demonstrated under conditions of increased interictal activity [14] and oxidative stress [15]. Indicating that regulation of KCC2 membrane trafficking may be a crucial mechanism by which KCC2 function can be controlled.

KCC2 is a 12 transmembrane protein with both amino and carboxyl intracellular termini and belongs to the cation-chloride cotransporter (CCC) superfamily, which consists of one Na+–Cl cotransporter (NCC), two Na+–K+–Cl cotransporters (NKCCs), and four K+–Cl cotransporters (KCCs). Different CCCs exert opposite Cl transport activities, with NCC and NKCCs taking up Cl, while KCCs extrude Cl [16]. The functional unit of CCCs is most likely to be a dimer, as homo- and hetero-dimerization have now been demonstrated for NKCC [17], NCC [18] and KCC family proteins [19]. The molecular mechanisms regulating the membrane trafficking of any CCC family member however are presently unknown, albeit they play essential roles in controlling chloride homeostasis in multiple tissues.

The cellular mechanisms controlling the cell-surface expression of many transporters and their membrane internalization, in particular, have been shown to have a profound and dynamic effect on overall transporter activity [20], [21]. As one requirement for protein internalization is an interaction with the cellular endocytic machinery, the identification of the molecular motifs governing these interactions has in several cases revealed pivotal regulatory domains within these proteins [22], [23], [24]. How KCC2 membrane expression is controlled, specifically the cellular mechanism and the molecular motifs contributing to its membrane internalization are presently unknown. Therefore in the present study we set out to investigate the mechanisms controlling the membrane internalization of KCC2. Here we report our findings, using an array of endocytosis reporter systems and site-directed mutagenesis, of the cellular mechanisms and the molecular motif within KCC2 directing its constitutive internalization from the plasma membrane.

Section snippets

Antibodies

The following antibodies were used, mouse monoclonal anti-HA (HA.11, Covance, Berkeley, CA, USA), rabbit polyclonal anti-KCC2 (Upstate, Lake Placid, NY, USA), mouse monoclonal anti-IL2 receptor alpha (Tac; kindly provided by Julie Donaldson, NIH, Bethesda, MD, USA), mouse monoclonal anti-alpha adaptin-clone 8 for immunoblotting (BD Biosciences, Mississauga, ON Canada) and anti-alpha adaptin-clone AP.6 for immunoprecipitation (Affinity BioReagents, Golden CO USA). Donkey anti-mouse and

Endogenous neuronal KCC2 interacts with the clathrin-mediated endocytic machinery

Clathrin-mediated endocytosis (CME) is a prominent mechanism by which plasma membrane proteins are internalized. It involves the recruitment of membrane proteins (cargo) by the adaptor protein-2 (AP-2), to clathrin-coated pits, which are subsequently pinched from the plasma membrane to form internalized endocytic vesicles [28]. To examine whether neuronal KCC2 interacts with the CME machinery, we tested whether the AP-2 complex is bound endogenously to neuronal KCC2 by co-immunoprecipitation.

Discussion

KCC2 is essential in the vast majority of CNS neurons for the development and maintenance of inhibitory neurotransmission [3], [4], [5], [6], [7]. In addition loss of KCC2 expression and function is associated with several neuropathological conditions including chronic pain [5], nerve injury [6] and epilepsy [7]. Given this seemingly crucial role for KCC2 in the mature nervous system, it was surprising how little was known of the cellular mechanisms controlling KCC2 stability and function, in

Acknowledgements

We thank Julie Donaldson, Yves Rouille, Robert lodge, Mark McNiven, Matthew Mulvey, Robert Harvey, Peter McPherson and Stephane Laporte for constructs and antibodies. This work was supported, in part by the Canadian Institute of Health Research (CIHR) operating grant awarded to, F. B. (MOP-62822) and J.P. (MOP-49590). F.B. and D.B. are CIHR-Canada Research Chairs-Tier II recipients.

References (43)

  • W. Sieghart

    Adv. Pharmacol.

    (2006)
  • C.A. Hubner et al.

    Neuron

    (2001)
  • H. Fiumelli et al.

    Neuron

    (2005)
  • J.A. Payne et al.

    Trends Neurosci.

    (2003)
  • J.C. de Jong et al.

    J. Biol. Chem.

    (2003)
  • S. D'Souza et al.

    J. Biol. Chem.

    (1998)
  • T. Grampp et al.

    J. Biol. Chem.

    (2007)
  • A. Kyrozis et al.

    J. Neurosci. Methods

    (1995)
  • Q. Yao et al.

    J. Mol. Biol.

    (2005)
  • J.L. Martys et al.

    J. Biol. Chem.

    (1995)
  • P.K. Tan et al.

    J. Biol. Chem.

    (1998)
  • M.J. Bergeron et al.

    J. Biol. Chem.

    (2006)
  • A. Mercado et al.

    J Biol Chem

    (2006)
  • C.F. Simard et al.

    J. Biol. Chem.

    (2007)
  • H. Betz

    Q. Rev. Biophys.

    (1992)
  • C. Rivera et al.

    Nature

    (1999)
  • J.A. Coull et al.

    Nature

    (2003)
  • J. Nabekura et al.

    J. Neurosci.

    (2002)
  • N.S. Woo et al.

    Hippocampus

    (2002)
  • G. Huberfeld et al.

    J. Neurosci.

    (2007)
  • K. Inoue et al.

    J. Neurochem.

    (2006)
  • Cited by (38)

    • Structural changes in the extracellular loop 2 of the murine KCC2 potassium chloride cotransporter modulate ion transport

      2021, Journal of Biological Chemistry
      Citation Excerpt :

      Having confirmed the conservation of the K+-binding site in KCC2, we next made use of a previously reported KCC2b clone with an extracellular 3xHA tag (KCC2HA) between TM3 and TM4. The tag with a length of 37 amino acid residues is inserted at the very end of EL2 in front of TM4 (31) (Fig. 1A). Importantly, this variant supports transport activity indistinguishable from KCC2bWT (Fig. 2A).

    • KCC2 membrane diffusion tunes neuronal chloride homeostasis

      2020, Neuropharmacology
      Citation Excerpt :

      In addition, the C-terminus of KCC2 contains a motif responsible for the isotonic activity of the transporter (Acton et al., 2012; Bergeron et al., 2006; Mercado et al., 2006). KCC2 and NKCC1 N- and C- termini also participate in the regulation of membrane trafficking (Lee et al., 2010, 2007; Zhao et al., 2008), basolateral and apical sorting in polarized cells (Carmosino et al., 2008) and oligomerization (Casula et al., 2009, 2001; Parvin et al., 2007; Simard et al., 2004; Warmuth et al., 2009). Furthermore, two KCC2 and NKCC1 (a and b) isoforms have been identified, which differ by the presence of additional amino acid sequences in KCC2a and NKCC1a on their N- and C- termini respectively (Fig. 1).

    • Current structural view on potassium chloride co-transporters

      2020, Neuronal Chloride Transporters in Health and Disease
    • Protein interaction partners of neuronal chloride transporters

      2020, Neuronal Chloride Transporters in Health and Disease
    • Transport-dependent and independent functions of KCC2 at excitatory synapses

      2020, Neuronal Chloride Transporters in Health and Disease
    View all citing articles on Scopus
    View full text