Elsevier

Immunology Letters

Volume 92, Issues 1–2, 29 March 2004, Pages 55-66
Immunology Letters

Review
Ion channels and lymphocyte activation

https://doi.org/10.1016/j.imlet.2003.11.020Get rights and content

Abstract

The ion channels expressed by T lymphocytes play key roles in the control of the membrane potential and calcium signaling, thereby affecting signal transduction pathways that lead to the activation of these cells following antigenic stimulation. Disruption of these pathways can attenuate or prevent the response of T-cells to antigenic challenge resulting in immune suppression. Studies using ion channel blockers of high affinity and specificity have shown that this interference can be achieved at the level of ion channels. Suppression of immune functions by channel blockers has been demonstrated in vitro and in vivo. New information about the molecular structure of ion channels facilitates the design of more potent and more specific inhibitors. Thus, T-cell ion channels are likely to serve as targets for immunomodulatory drugs in the near future. Here, the biophysical properties, tissue distribution, regulation of expression, molecular pharmacology and role in T-cell activation of the voltage-gated Kv1.3 and the Ca2+-activated IKCa1 potassium channels and those of the Ca+ release-activated Ca2+ (CRAC) channel are reviewed.

Introduction

Clonal expansion of lymphocytes, in vivo, is required to generate an efficient generalized immune response to a specific antigen. Binding of peptide-loaded MHC to the T-cell receptor–CD3 complex (TCR/CD3) results in the recruitment and activation of protein tyrosine kinases, such as members of Src, Zap-70/Syk, Tec and Csk families of non-receptor tyrosine kinases, and the concomitant activation of phospholipase C-γ (PLCγ). PLCγ cleaves phosphatidylinositol 4,5-bisphosphate into diacylglycerol and 1,4,5-inositol trisphosphate (IP3), and in turn, initiates two signaling pathways in lymphocyte activation. Diacylglycerol activates the protein kinase C (PKC) pathway, particularly through protein kinase Cθ, which leads to the phosphorylation of several intracellular substrates and the triggering of transcription via the assembly of the Fos/Jun transcription factor complex on AP1 elements in several genes. The second pathway, initiated by the generation of IP3, governs the sustained elevation of the cytosolic free Ca2+ concentration ([Ca2+]i) required for efficient signal transduction. The calcium signal activates the Ca2+-calmodulin-dependent phospahatase calcineurin. Calcineurin then dephosphorylates the transcription factor NF-AT thereby enabling it to accumulate in the nucleus and bind to the promoter element of the interleukin-2 (IL-2) gene. Activation of IL-2 gene and IL-2 expression is a critical commitment point beyond which further T-cell activation becomes antigen independent. A sustained Ca2+ signal is required to keep NF-AT in the nucleus in the transcriptionally active state.

The sustained Ca2+ signal relies on the operation of the ion channels in T-cells, especially the voltage-gated Kv1.3 and the Ca2+-activated IKCa1 potassium channels and the Ca2+ release-activated Ca2+ (CRAC) channel. In this paper we review the role of these ion channels in T-cell activation in addition to their biophysical properties, tissue distribution, regulation, pharmacology and their connection to certain diseases.

Section snippets

Intracellular and plasma membrane Ca2+ channels participating in the generation of the Ca2+ signal

Two sequentially coupled mechanisms contribute to the generation of the Ca2+ signal in T-cells. The first event is the binding of IP3 to the IP3-receptor (IP3R) located in the membrane of the endoplasmic reticulum. IP3R engagement with its ligand results in the release of Ca2+ stored in the lumen of the ER causing a rise in the [Ca2+]i from a resting level of ∼100 nM to a peak concentration of ∼500 nM (reviewed in [1]).

In addition to the extensively studied IP3–IP3R system, a cyclic ADP-ribose

Membrane potential dependence of the Ca2+ signal, the role of potassium channels

An interesting, physiologically important feature of the CRAC channel is its membrane potential-independent gating and its inward rectification [5]. This means that once the CRAC channels are activated the electrochemical driving force for Ca2+ will determine the magnitude of the inward current. The driving force increases with membrane hyperpolarization, which, along with the inward rectification of the current, results in larger Ca2+ current at negative membrane potentials. This unusual

Tissue distribution of Kv1.3 and IKCa1 channels, regulation of their expression

The tissue distribution of Kv1.3 channels is relatively restricted to the immune system and the central nervous system. In the immune system very interesting changes in K+ channel expression accompany the proliferation, maturation and differentiation of T-cells. Subset specific expression of ion channels in murine thymocytes was described relatively early [59]. Immature CD4+CD8+ cells express ∼300 Kv1.3 channels; the expression level of this channel drops dramatically (∼20 channel per cell) upon

Production of high affinity and high specificity blockers of Kv1.3 and IKCa1 channels: potential immunosuppressors

Soon after the characterization of Kv1.3 channels and Ca2+-activated K+ channels in lymphocytes it was discovered that channel-blocking agents are able to inhibit T-cell activation, including secretion of lymphokines, cell proliferation, and killing of target cells in vitro as well as in vivo (see above). These initial observations led to the search for high specificity and high affinity blockers of Kv1.3 and IKCa1 channels (reviewed recently in [52], [74], [75]). Research in this field was

New perspective: regulation of Kv1.3 channels by supramolecular clusters important in T-cell signaling

Interaction of T lymphocytes with professional antigen presenting cells is required for in vivo activation of T-cells. This interaction takes place at a specialized intercellular contact, called immunological synapse (IS), where the encounter causes proteins to segregate into micrometer-scale domains [86]. Formation of the IS between T-cells and antigen presenting cells recruits LFA-1 in the periphery and TCR/CD3, CD28 and CD2 membrane proteins in the center of the supramolecular activation

Concluding remarks

Critical information accumulated in the past two decades about the biophysical and pharmacological properties of Kv1.3 and IKCa1 channels. The function of these channels in regulating the membrane potential and Ca2+ signaling is well supported. Discovery of potent and selective blockers of Kv1.3 and IKCa1 along with the characterization of subset specific expression of these channels led to successful and selective suppression of lymphocyte activation in vitro and most importantly, in vivo as

Acknowledgements

This work was supported by grants from the following agencies: OTKA TS040773, T043087, F035251, ETT 222/2003, 010/2001. The authors would like to thank János Matkó (Eötvös Loránd University, Budapest) for helpful discussions and critical reading of the manuscript.

References (100)

  • N.J Logsdon et al.

    A novel gene, hKCa4, encodes the calcium-activated potassium channel in human T lymphocytes

    J. Biol. Chem.

    (1997)
  • C.M Fanger et al.

    Calmodulin mediates calcium-dependent activation of the intermediate conductance KCa channel, IKCa1

    J. Biol. Chem.

    (1999)
  • B Sarkadi et al.

    Membrane depolarization selectively inhibits receptor-operated calcium channels in human (Jurkat) T lymphocytes

    Biochim. Biophys. Acta

    (1990)
  • K Kalman et al.

    ShK-Dap22, a potent Kv1.3-specific immunosuppressive polypeptide

    J. Biol. Chem.

    (1998)
  • M.D Cahalan et al.

    Ion channels in the immune system as targets for immunosuppression

    Curr. Opin. Biotechnol.

    (1997)
  • C.M Fanger et al.

    Calcium-activated potassium channels sustain calcium signaling in T lymphocytes. Selective blockers and manipulated channel expression levels

    J. Biol. Chem.

    (2001)
  • S Ghanshani et al.

    Up-regulation of the IKCa1 potassium channel during T-cell activation. Molecular mechanism and functional consequences

    J. Biol. Chem.

    (2000)
  • C Beeton et al.

    A novel fluorescent toxin to detect and investigate Kv1.3 channel up-regulation in chronically activated T lymphocytes

    J. Biol. Chem.

    (2003)
  • A.A Alizadeh et al.

    Genomic-scale gene expression profiling of normal and malignant immune cells

    Curr. Opin. Immunol.

    (2000)
  • R.O Koch et al.

    Complex subunit assembly of neuronal voltage-gated K+ channels. Basis for high-affinity toxin interactions and pharmacology

    J. Biol. Chem.

    (1997)
  • T Ohno-Shosaku et al.

    Presence of the voltage-gated potassium channels sensitive to charybdotoxin in inhibitory presynaptic terminals of cultured rat hippocampal neurons

    Neurosci. Lett.

    (1996)
  • J Aiyar et al.

    Topology of the pore-region of a K+ channel revealed by the NMR-derived structures of scorpion toxins

    Neuron

    (1995)
  • M.L Garcia et al.

    Potassium channels: from scorpion venoms to high-resolution structure

    Toxicon

    (2001)
  • B Attali et al.

    Cloning, functional expression, and regulation of two K+ channels in human T lymphocytes

    J. Biol. Chem.

    (1992)
  • M Dauplais et al.

    On the convergent evolution of animal toxins. Conservation of a diad of functional residues in potassium channel-blocking toxins with unrelated structures

    J. Biol. Chem.

    (1997)
  • C.V Batista et al.

    Two novel toxins from the Amazonian scorpion Tityus cambridgei that block Kv1.3 and Shaker B K(+)-channels with distinctly different affinities

    Biochim. Biophys. Acta

    (2002)
  • M Peter et al.

    Blockage of human T lymphocyte Kv1.3 channels by Pi1, a novel class of scorpion toxin

    Biochem. Biophys. Res. Commun.

    (2000)
  • D.M Davis

    Assembly of the immunological synapse for T cells and NK cells

    Trends Immunol.

    (2002)
  • P.A van der Merwe

    Formation and function of the immunological synapse

    Curr. Opin. Immunol.

    (2002)
  • J.R Martens et al.

    Differential targeting of Shaker-like potassium channels to lipid rafts

    J. Biol. Chem.

    (2000)
  • J.R Martens et al.

    Isoform-specific localization of voltage-gated K+ channels to distinct lipid raft populations. Targeting of Kv1.5 to caveolae

    J. Biol. Chem.

    (2001)
  • J Bock et al.

    Ceramide inhibits the potassium channel Kv1.3 by the formation of membrane platforms

    Biochem. Biophys. Res. Commun.

    (2003)
  • I Szabo et al.

    Tyrosine phosphorylation-dependent suppression of a voltage-gated K+ channel in T lymphocytes upon Fas stimulation

    J. Biol. Chem.

    (1996)
  • T Hanada et al.

    Human homologue of the Drosophila discs large tumor suppressor binds to p56lck tyrosine kinase and Shaker type Kv1.3 potassium channel in T lymphocytes

    J. Biol. Chem.

    (1997)
  • T McCormack et al.

    The effects of Shaker beta-subunits on the human lymphocyte K+ channel Kv1.3

    J. Biol. Chem.

    (1999)
  • J Matko

    K+ channels and T-cell synapses: the molecular background for efficient immunomodulation is shaping up

    Trends Pharmacol. Sci.

    (2003)
  • R.S Lewis

    Calcium signaling mechanisms in T lymphocytes

    Annu. Rev. Immunol.

    (2001)
  • A.H Guse et al.

    Characterization of cyclic adenosine diphosphate-ribose-induced Ca2+ release in T lymphocyte cell lines

    J. Immunol.

    (1995)
  • A.H Guse et al.

    Regulation of calcium signalling in T lymphocytes by the second messenger cyclic ADP-ribose

    Nature

    (1999)
  • A Zweifach et al.

    Mitogen-regulated Ca2+ current of T lymphocytes is activated by depletion of intracellular Ca2+ stores

    Proc. Natl. Acad. Sci. U.S.A.

    (1993)
  • H.H Kerschbaum et al.

    Single-channel recording of a store-operated Ca2+ channel in Jurkat T lymphocytes

    Science

    (1999)
  • A.F Fomina et al.

    Single channel properties and regulated expression of Ca(2+) release-activated Ca(2+) (CRAC) channels in human T cells

    J. Cell Biol.

    (2000)
  • J.A Kozak et al.

    Distinct properties of CRAC and MIC channels in RBL cells

    J. Gen. Phys.

    (2002)
  • M Prakriya et al.

    Separation and characterization of currents through store-operated CRAC channels and Mg2+-inhibited cation (MIC) channels

    J. Gen. Phys.

    (2002)
  • B Minke et al.

    TRP channel proteins and signal transduction

    Phys. Rev.

    (2002)
  • C Randriamampita et al.

    Emptying of intracellular Ca2+ stores releases a novel small messenger that stimulates Ca2+ influx

    Nature

    (1993)
  • Z Su et al.

    A store-operated nonselective cation channel in lymphocytes is activated directly by Ca(2+) influx factor and diacylglycerol

    Am. J. Phys. Cell Phys.

    (2001)
  • J.W Putney et al.

    Mechanisms of capacitative calcium entry

    J. Cell Sci.

    (2001)
  • A.H Guse

    Ca2+ signaling in T-lymphocytes

    Crit. Rev. Immunol.

    (1998)
  • K Kiselyov et al.

    Regulation of Ca2+-release-activated Ca2+ current (Icrac) by ryanodine receptors in inositol 1,4,5-trisphosphate-receptor-deficient DT40 cells

    Biochem. J.

    (2001)
  • Cited by (103)

    • The voltage-gated potassium channel K<inf>V</inf>1.3 as a therapeutic target for venom-derived peptides

      2020, Biochemical Pharmacology
      Citation Excerpt :

      No effects on odour detection were reported in the Phase 1 trial of the potent peptide inhibitor of KV1.3 dalazatide, described further in the next section [165]. In closing this section, we note that the expression of KV1.3 was initially thought to be restricted to the immune system and the central nervous system [166,167]. Several publications suggest, however, that KV1.3 expression is much more widespread [3].

    View all citing articles on Scopus
    View full text