Mapping Functional Domains of Chloride Intracellular Channel (CLIC) Proteins in Vivo

https://doi.org/10.1016/j.jmb.2006.04.046Get rights and content

Abstract

Chloride intracellular channel (CLIC) proteins are small proteins distantly related to the omega family of glutathione S-transferases (GSTs). CLIC proteins are expressed in a wide variety of tissues in multicellular organisms and are targeted to specific cellular membranes. Members of this family are capable in vitro of changing conformation from a globular, soluble state to a membrane-inserted state in which they provide chloride conductance. The structural basis for in vivo CLIC protein function, however, is not well understood. We have mapped the functional domains of CLIC family members using an in vivo assay for membrane localization and function of CLIC proteins in the nematode Caenorhabditis elegans. A < 70 amino acid N-terminal domain is a key determinant of membrane localization and function of invertebrate CLIC proteins. This domain, which we term the ‘‘PTM‘‘ domain, named after an amphipathic putative transmembrane helix contained within it, directs distinct C. elegans CLIC homologs to distinct subcellular membranes. We find that within the PTM region, the cysteine residues required for GST-type activity are unnecessary for invertebrate CLIC function, but that specific residues within the proposed transmembrane helix are necessary for correct targeting and protein function. We find that among all tested invertebrate CLIC proteins, function appears to be completely conserved despite striking differences in the charged residues contained within the amphipathic helix. This indicates that these residues do not contribute to anion selectivity as previously suggested. We find that outside the PTM region, the remaining three-quarters of CLIC protein sequence is functionally equivalent not only among vertebrate and invertebrate CLIC proteins, but also among the more distantly related GST-omega and GST-sigma proteins. The PTM region thus provides both targeting information and CLIC functional specificity, possibly adapting GST-type proteins to function as ion channels.

Introduction

Chloride intracellular channel (CLIC) proteins are small proteins of approximately 240 residues that are distantly related to the omega family of glutathione S-transferases (GSTs).1 CLIC proteins are expressed in a wide variety of tissues in multicellular organisms. Distinct CLIC proteins localize to distinct cellular membranes, including the plasma membrane, Golgi membrane, mitochondria, the nuclear membrane, dense core vesicles, lysosomal membranes, cell–cell junctions, and the luminal membrane of the excretory canal cell of the nematode Caenorhabditis elegans.2., 3., 4., 5., 6., 7. CLIC proteins have been shown to play roles in diverse processes including bone resorption,8 regulation of cell motility,9 apoptosis,4., 10., 11., 12. β-amyloid-induced neurotoxicity,13 and tubulogenesis6 (Figure 1).

Based on several lines of evidence, it has been proposed that CLIC proteins may function as chloride channels. The first CLIC protein to be identified, p64, was isolated by its ability to bind the known chloride channel inhibitor indanyloxyacetic acid (IAA).14., 15., 16. Since then, many CLIC proteins have been shown to confer chloride channel activity in transfected cells.2., 17., 18., 19. While CLIC proteins lack an identifiable signal sequence, epitope tagging experiments have demonstrated that transfected HsCLIC1 and HsCLIC4 can adopt an integral membrane conformation, spanning the membrane an odd number of times with the N terminus on the extracellular side.5., 20. Moreover, it has been shown that purified recombinant HsCLIC1 can insert into artificial lipid bilayers and confer chloride channel activity.21., 22., 23.Finally, proteins of the CLIC family have been associated with physiological processes requiring anion flux, such as water secretion and bone resorption.6., 8., 24.

In spite of these lines of evidence, the molecular function of CLIC proteins is still poorly understood, both on a biochemical and physiological level. Recent work indicates that, in vitro, rat CLIC1 may form non-selective pores rather than anion-selective channels.25 Furthermore, it remains possible that CLIC proteins function in vivo as accessory proteins to anion channels, or play an altogether different role, such as interacting with the actin cytoskeleton.7., 26., 27., 28. Thus, several open questions about CLIC proteins remain. First, is the physiological role of CLIC proteins indeed that of an ion channel and if so, an anion channel or a non-selective pore? One strategy to address this issue at least in part is to define domains and residues required for CLIC translocation and conductance in vitro, and to determine if these are also required for the in vivo activity of CLIC proteins. Second, if CLIC proteins do function as anion channels or non-selective pores, what is the biophysical mechanism of conduction? Given that CLIC proteins are much smaller than and show no sequence homology with the well characterized ClC family of chloride channels,29 they may utilize a novel mechanism of ion conductance. Third, given that there are six vertebrate CLIC homologs which are expressed in different cell types and localized to different cell membranes, do distinct CLIC proteins have distinct functions, or do they differ only in their expression patterns and membrane targeting properties? Similarly, does the function of vertebrate and invertebrate CLIC proteins differ? Lastly, how are CLIC proteins, which do not contain recognizable signal or sorting sequences, targeted to intracellular membranes?

We have previously described the identification of the genes exc-4 and exl-1, which encode the two CLIC protein orthologs in C. elegans.6 Genetic removal of exc-4 causes a cystic defect in the unicellular excretory canal cell of C. elegans6 (Figure 1). This defect has been proposed to model polycystic kidney disease.30., 31. We have shown that EXC-4 is localized to and required at the luminal membrane of the excretory canal cell to establish tubular architecture following cell hollowing and to maintain tubular architecture following development6 (Figure 1). These results have enabled us to establish an in vivo assay for CLIC protein localization and function, by expressing wild-type or mutated CLIC protein homologs in the C. elegans excretory canal cell and asking whether these correctly localize to the intracellular luminal membrane and provide rescuing activity in exc-4 null mutant animals.

We report here the results of using this in vivo assay to define residues and functional domains specifically required for CLIC protein targeting and function within a physiologically relevant, cellular context in C. elegans. We show that a minimal 66 amino acid N-terminal domain, termed the PTM domain, named after a putative transmembrane helix contained within it, is a key determinant of both membrane localization and CLIC-specific protein function. Within this region, an amphipathic α-helix containing positively charged residues has been proposed to function as a pore-lining helix.19,25,32 We describe a mutational analysis of this helix using a functional in vivo assay. Moreover, we demonstrate that outside the PTM region, the C-terminal three-quarters of CLIC proteins are functionally equivalent among vertebrate and invertebrate CLIC proteins and, surprisingly, also among the more distantly related GST-omega and GST-sigma proteins. Our in vivo assay for CLIC function in C. elegans thus reveals that the PTM region provides CLIC-type functional specificity in addition to directing specific membrane targeting. Finally, we show that differences in membrane targeting by functionally equivalent CLIC proteins in C. elegans are determined by both cellular context and differences in protein sequence.

Section snippets

Phylogenetic and sequence analysis of CLIC proteins

Chloride intracellular channel (CLIC) proteins share both sequence and structural homology with the omega family of GST proteins1., 32. but appear to have distinct functions in cellular biology.33., 34. Vertebrate CLIC proteins were originally identified as chloride channels;2., 14., 15., 16., 17., 19. recently, additional invertebrate CLIC homologs have been discovered.6 Using several complete invertebrate and vertebrate genome datasets, we undertook a comprehensive phylogenetic analysis,

Discussion

We reported here the results of using an in vivo assay to define residues and functional domains required for CLIC protein function in C. elegans. Previous in vitro studies aimed at defining residues important for CLIC activity have focused on the role of Cys24 at the start of h1 in HsCLIC1. This residue has been proposed to be required either for channel formation in the case of pre-oxidised proteins37 or for redox-regulation of channel gating through disulfide bond formation between

C. elegans strains

Bristol N2 wild-type. NJ469 exc-4(rh133)I. RB960, exl-1(ok857) II. RT258 unc-119(ed3)III; pwIs50 [unc-119(+), lmp-1::gfp]. GS1912 arIs37[pmyo-3::ssGFP] I; dpy-20(e1282) IV.

Phylogenetic analysis

The following protein sequences were used for sequence and phylogenetic analysis (NCBI ENTREZ accession numbers are given in parentheses): CG6776 (NP_648234), CbGST-44 (CAE71235), CeGST-44 (CAB07572), HsGSTo1 (P78417), HsGSTo2 (NP_899062), HsPGDS (NP_055300), CG8938 (NP_725653), CeGST-36 (NP_509652), CbGST-36 (CAE58723), p64 (

Acknowledgements

We are indebted to Shana Posy for providing essential help with the phylogenetic analysis. We thank the C. elegans knockout consortium in Oklahoma for the ok857 allele, Yuji Kohara for the cDNA clone yk604b8; B. Grant for the strain RT258; C. de la Cova for Drosophila 3rd instar cDNA; S. Breit for providing the plasmid pPD95.75-CLIC1; S. Breit, P. Curmi, and D. Littler for helpful and stimulating discussions and for sharing of unpublished data and R. Ashley, S. Breit, P. Curmi, and D. Littler

References (56)

  • B.M. Tulk et al.

    CLIC-1 functions as a chloride channel when expressed and purified from bacteria

    J. Biol. Chem.

    (2000)
  • K. Warton et al.

    Recombinant CLIC1 (NCC27) assembles in lipid bilayers via a pH-dependent two-state process to form chloride ion channels with identical characteristics to those observed in Chinese hamster ovary cells expressing CLIC1

    J. Biol. Chem.

    (2002)
  • T. Nishizawa et al.

    Molecular cloning and characterization of a novel chloride intracellular channel-related protein, parchorin, expressed in water-secreting cells

    J. Biol. Chem.

    (2000)
  • M. Berryman et al.

    CLIC-5A functions as a chloride channel in vitro and associates with the cortical actin cytoskeleton in vitro and in vivo

    J. Biol. Chem.

    (2004)
  • M. Buechner et al.

    Cystic canal mutants in Caenorhabditis elegans are defective in the apical membrane domain of the renal (excretory) cell

    Dev. Biol.

    (1999)
  • M. Buechner

    Tubes and the single C. elegans excretory cell

    Trends Cell Biol.

    (2002)
  • S.J. Harrop et al.

    Crystal structure of a soluble form of the intracellular chloride ion channel CLIC1 (NCC27) at 1.4-Å resolution

    J. Biol. Chem.

    (2001)
  • P.G. Board et al.

    Identification, characterization, and crystal structure of the Omega class glutathione transferases

    J. Biol. Chem.

    (2000)
  • P.G. Board et al.

    CLIC-2 modulates cardiac ryanodine receptor Ca2+ release channels

    Int. J. Biochem. Cell Biol.

    (2004)
  • I. Nilsson et al.

    Proline-induced disruption of a transmembrane alpha-helix in its natural environment

    J. Mol. Biol.

    (1998)
  • D.R. Littler et al.

    The intracellular chloride ion channel protein CLIC1 undergoes a redox-controlled structural transition

    J. Biol. Chem.

    (2004)
  • A. Schinzel et al.

    Bcl-2 family members: Integrators of survival and death signals in physiology and pathology [corrected]

    Biochim. Biophys. Acta

    (2004)
  • M.C. Wilce et al.

    Structure and function of glutathione S-transferases

    Biochim. Biophys. Acta

    (1994)
  • B. Agianian et al.

    Structure of a Drosophila sigma class glutathione S-transferase reveals a novel active site topography suited for lipid peroxidation products

    J. Mol. Biol.

    (2003)
  • M.W. Parker et al.

    Pore-forming protein toxins: From structure to function

    Prog. Biophys. Mol. Biol.

    (2005)
  • J.I. Kourie et al.

    Biophysical and molecular properties of annexin-formed channels

    Prog. Biophys. Mol. Biol.

    (2000)
  • C. Notredame et al.

    T-Coffee: A novel method for fast and accurate multiple sequence alignment

    J. Mol. Biol.

    (2000)
  • J.Z. Chuang et al.

    A 29 kDa intracellular chloride channel p64H1 is associated with large dense-core vesicles in rat hippocampal neurons

    J. Neurosci.

    (1999)
  • Cited by (42)

    • Epithelial morphogenesis, tubulogenesis and forces in organogenesis

      2021, Current Topics in Developmental Biology
    • A conserved GXXXG motif in the transmembrane domain of CLIC proteins is essential for their cholesterol-dependant membrane interaction

      2019, Biochimica et Biophysica Acta - General Subjects
      Citation Excerpt :

      Interestingly, analysis of the Catalogue of Somatic Mutations in Cancer (COSMIC) has shown approximately 93 cancer-associated mutations in the longest CLIC6 splice variant (704 amino acids long) [57,58]. Upon further observation it was noted that none of the cancer associated positions directly mediated the formation of the inter-domain interface; however, several were found to be localised to the PTMD (Arg384, Leu385 and Leu389) and to the β1-α1 loop (Gly373 and Gly377), and helix α6 (His540) of the TRX and α helical domains, respectively [31,57]. This is significant given that the murine CLIC6 residues Gly373 and Gly377, are equivalent to Gly18 and Gly22 in the human CLIC1 or Gly481 and Gly485 in the human CLIC6 GXXXG motif [57].

    • CLIC proteins, ezrin, radixin, moesin and the coupling of membranes to the actin cytoskeleton: A smoking gun?

      2014, Biochimica et Biophysica Acta - Biomembranes
      Citation Excerpt :

      Some of the proteins now known to cause this mutation include: Sma-1–β spectrin [140]; Exc-5–FGD1, a GEF for Cdc42 [141–143]; Exc-7–ELAV, a splicing factor that binds mRNA for Sma-1 and Exc-3 [144,145]; and Exc-9–Zn-binding LIM domain protein [146]. The discovery that Exc-4 was a member of the CLIC protein family was a major advance in understanding CLIC function [17,18]. Unlike mammalian CLICs, Exc-4 does not have a detectible cytosolic component but lines the apical tubular membrane in the excretory cell and is completely membrane associated.

    View all citing articles on Scopus
    View full text