Elsevier

Toxicon

Volume 49, Issue 2, February 2007, Pages 213-230
Toxicon

Tarantula toxins interacting with voltage sensors in potassium channels

https://doi.org/10.1016/j.toxicon.2006.09.024Get rights and content

Abstract

Voltage-activated ion channels open and close in response to changes in membrane voltage, a process that is crucial for electrical signaling in the nervous system. The venom from many poisonous creatures contains a diverse array of small protein toxins that bind to voltage-activated channels and modify the gating mechanism. Hanatoxin and a growing number of related tarantula toxins have been shown to inhibit activation of voltage-activated potassium (Kv) channels by interacting with their voltage-sensing domains. This review summarizes our current understanding of the mechanism by which these toxins alter gating, the location of the toxin receptor within Kv channels and the disposition of this receptor with respect to the lipid membrane. The conservation of tarantula toxin receptors among voltage-activated ion channels will also be discussed.

Introduction

Voltage-activated ion channels open and close in response to changes in membrane voltage, a process that underlies their fundamental roles in the generation and propagation of electrical signals within the nervous system. Not surprisingly, these crucial ion channels are popular targets for the wide array of protein toxins found in the venom of poisonous creatures. For example, venom from arachnids (spiders and scorpions), anthozoans (sea anemone), mollusks (cone snails) and reptiles (snakes) contain toxins that bind to voltage-activated potassium (Kv), sodium (Nav) and calcium (Cav) channels (Possani et al., 2000; Rash and Hodgson, 2002; Srinivasan et al., 2002; Terlau and Olivera, 2004). The most widely appreciated biological function of toxins targeting voltage-activated channels is to paralyze prey, either by inhibiting (Nav and Cav inhibitors) or excessively stimulating (Kv inhibitors and Nav activators) transmission at synapses within the nervous system.

The toxins that interact with voltage-activated ion channels can be thought of as working through two distinct targeting mechanisms. In the first mechanism the toxin binds to the outer vestibule of the ion conduction pore and inhibits the flow of ions. The best studied examples of this mechanism are the scorpion toxins charybodotoxin and agitoxin (Miller, 1995), two particularly interesting Kv channel inhibitors that were used to find the pore forming region of potassium channels (MacKinnon and Miller, 1989). In the second type of mechanism the toxin binds to a region of the channel that changes conformation during gating and influences the gating mechanism by altering the relative stability of closed, open or inactivated states. The α- and β-scorpion Nav channel toxins from Centruroides sculpturatus scorpion venom were the earliest studied protein toxins that modify gating of voltage-activated ion channels (Koppenhofer and Schmidt, 1968a, Koppenhofer and Schmidt, 1968b; Cahalan, 1975; Wang and Strichartz, 1983). This review primarily focuses on the more recently discovered family of toxins that modify gating of Kv channels, with occasional reference to related studies on toxins that interact with Cav and Nav channels.

The X-ray structure of the mammalian Kv1.2 channel (Long et al., 2005) nicely illustrates the two primary regions of the channel that are targeted by toxins (Fig. 1). As the structure shows, Kv channels are tetramers, a fact that was first established from experiments with the pore blocking scorpion toxin, agitoxin (MacKinnon, 1991). Each Kv channel subunit contains 6 transmembrane segments, termed S1–S6, arranged into two types of domains; a single pore domain formed by the S5–S6 regions from the four subunits (yellow helices), and four surrounding voltage-sensing domains, each one formed by the S1–S4 helices from a single subunit. The pore domain houses the K+ selective ion conduction pathway (demarcated by the red potassium ions in Fig. 1B,C), and the receptor for pore blocking toxins that bind to the extracellular vestibule near the selectivity filter. In the case of charybdotoxin, a Lys residue on the active surface of the toxin projects into the pore of the channel and interacts with potassium ions bound within the selectivity filter (Anderson et al., 1988; MacKinnon and Miller, 1988; Park and Miller, 1992). Many other types of K+ channels only contain the equivalent of the pore domain of Kv channels (Jan and Jan, 1997), and pore blocking scorpion and bee toxins similarly bind to the extracellular entrance of the pore in these simpler channels (Lu and MacKinnon, 1997; Jin and Lu, 1998), highlighting the conserved nature of potassium channel pores. The pore domain also contains the S6 activation gate, an intracellular region of the ion conduction pore that prevents the flow of ions in the closed state (Fig. 1B) (Yellen, 2002). The S6 gate region of the protein does not appear to be directly targeted by protein toxins, presumably because poisonous creatures deposit their venom into the extracellular solution and the S6 gate is located on the opposite side of the membrane.

The voltage-sensing domain is a second region of the channel that is widely targeted by toxins, in this case by the class of toxins that bind to and inhibit the channel by stabilizing a particular state in the gating pathway. The process of voltage-dependent gating in Kv channels is thought to involve several relatively independent motions of the four voltage-sensing domains between resting (R) and activated (A) states, followed by a concerted opening transition where the S6 gate moves from a closed (C) to an open (O) state (Scheme 1; (Ledwell and Aldrich, 1999)). There is growing evidence to suggest that the S4 segment within the voltage-sensing domains also moves during the concerted opening transition (Smith-Maxwell et al., 1998a, Smith-Maxwell et al., 1998b; Ledwell and Aldrich, 1999; del Camino et al., 2005; Pathak et al., 2005), and therefore a toxin that binds to the voltage-sensing domains could in principle inhibit channel activation by stabilizing any of the states prior to the open state.

Hanatoxin is the founding member of a family of toxins that bind to the voltage sensing domains in Kv channels and inhibit opening of these channels. This 35 residue protein toxin was originally isolated from the venom of the Chilean rose tarantula (Grammostola spatulata) during a search for new inhibitors of the Kv2.1 channel (Swartz and MacKinnon, 1995). The selectivity of hanatoxin is far from absolute, as it also inhibits Kv4.2 channels with similar affinity compared to Kv2.1 (see also Section 5.1). Although hanatoxin is one of the most extensively studied toxins targeting the voltage sensing domains in Kv channels (see below), the list of newer toxins in this class is rapidly growing (Fig. 2). The closely related SGTx1 from Scodra griseipes (Marvin et al., 1999; Lee et al., 2004) also inhibits Kv2.1 and systematic mutagenesis on this toxin has identified the active face of the molecule (Wang et al., 2004). The recently discovered guangxitoxin (GxTx1E) from Plesiophrictus guangxiensis has particularly high affinity for Kv2.1 (Herrington et al., 2006), which should open up promising opportunities for better understanding the complex between this family of toxins and voltage-sensing domains (see Section 2.3). Stromatoxin (ScTx1) from Stromatopelma calceata and heteroscodratoxins (HmTx1,2) from Heteroscodra maculate target both Kv2 and Kv4 channels (Escoubas et al., 2002), whereas the heteropodatoxins (HpTx1-3) from Heteropoda venatoria (Sanguinetti et al., 1997; Zarayskiy et al., 2005), phrixotoxins (PaTx1,2) from Phrixotrichus auratus (Diochot et al., 1999) and TLTx1-3 from Theraphosa leblondi venom (Ebbinghaus et al., 2004) seem to primarily target Kv4 channels. One of the more recent additions to this family of toxins is VsTx1, a toxin that was isolated from Grammostola spatulata venom when screening for activity against the KvAP channel (Ruta et al., 2003; Ruta and MacKinnon, 2004), an archebacterial Kv channel for which several X-ray crystal structures are available (Jiang et al., 2003a; Lee et al., 2005). The amino acid sequences of the Kv channel toxins discussed thus far show varying similarity, ranging from 82% to 30%, and all have six cysteine residues that form 3 disulfide bonds at the core of the molecules (Fig. 2; see also Section 3). It is rather interesting that with the exception of HpTx1-3, all of these Kv channel gating modifier toxins were isolated from tarantula venom. Although it is a mystery why these particular creatures are so fond of producing gating modifier toxins (and why they appear uninterested in making pore blocking protein toxins), it is clear that their venom is a particularly rich and relatively untapped repository of these toxins.

It has recently become evident that sea anemone also produce gating modifier toxins that interact with Kv channels. BDS I and II from Anemonia sulcata inhibit Kv3 channels (Diochot et al., 1998) and the related APETx1 from Anthopleura elegantissima venom inhibit HERG Kv channels (Diochot et al., 2003; Restano-Cassulini et al., 2006). The sequences of these anemone toxins are quite different than other Kv channel toxins discussed here, but a growing body of work suggests that they probably inhibit through a related mechanism (Diochot et al., 2003; Yeung et al., 2005).

Section snippets

Mechanism of channel inhibition

The first indication that protein toxins inhibit Kv channels through a mechanism other than pore occlusion came from experiments on hanatoxin where transfer of the outer vestibule (S5–S6 linker) of Kv2.1 into a relatively toxin insensitive channel failed to render the recipient channel sensitive to the toxin (Swartz and MacKinnon, 1995). Binding of a Kv2.1 toxin to regions outside the pore could have been predicted from earlier studies showing that the Kv2.1 channel contains several

Structures of Kv channel voltage sensor toxins

NMR solution structures have been solved for five of the Kv channel voltage sensor toxins listed in Fig. 2, including hanatoxin1, SGTx1, HpTx2, PaTx1 and VSTx1 (Bernard et al., 2000; Takahashi et al., 2000; Chagot et al., 2004; Lee et al., 2004; Jung et al., 2005). Structures are also available for the related toxins GrTx-SIA (Takeuchi et al., 2002) and GsMTx4 (Oswald et al., 2002), inhibitors of Cav channels and stretch activated channels, respectively.

Membrane partitioning of voltage sensor toxins

Based on the X-ray structures of the KvAP channel and biotin accessibility studies (Jiang et al., 2003a, Jiang et al., 2003b; Ruta et al., 2005), MacKinnon and colleagues proposed that the voltage sensor paddle moves relatively large distances through the lipid bilayer in response to changes in membrane voltage. The voltage sensor paddle, a helix-turn-helix motif composed of both S3b and S4 helices, is positioned largely within the inner leaflet of the membrane at negative membrane voltages in

Conservation and adaptation of voltage-sensing domains

One of the more interesting aspects of gating modifier toxins is that many structurally diverse toxins interact with the equivalent region of different voltage-activated cation channels. For example, the mutagenesis results identifying the hanatoxin receptors on Kv channels are remarkably consistent with similar efforts to identify the receptors for gating modifier toxins on Nav and Cav channels. Mutations at E1613 in the fourth repeat of the Nav 1.2 channel alters the binding affinity of α

Acknowledgments

I thank AbdulRasheed Alabi, Frank Bosmans, Dmitriy Krepkiy, Mirela Milescu and L. Revell Phillips for helpful discussions. I also thank Mirela Milescu for making Fig. 7, Fig. 8. This work was supported by the Intramural Research Program of the NINDS, NIH.

References (96)

  • R.S. Norton et al.

    The cystine knot structure of ion channel toxins and related polypeptides

    Toxicon

    (1998)
  • R.E. Oswald et al.

    Solution structure of peptide toxins that block mechanosensitive ion channels

    J. Biol. Chem.

    (2002)
  • C.S. Park et al.

    Interaction of charybdotoxin with permeant ions inside the pore of a K+ channel

    Neuron

    (1992)
  • L.D. Possani et al.

    Peptides and genes coding for scorpion toxins that affect ion-channels

    Biochimie

    (2000)
  • L.D. Rash et al.

    Pharmacology and biochemistry of spider venoms

    Toxicon

    (2002)
  • J.C. Rogers et al.

    Molecular determinants of high affinity binding of alpha-scorpion toxin and sea anemone toxin in the S3–S4 extracellular loop in domain IV of the Na+ channel alpha subunit

    J. Biol. Chem.

    (1996)
  • V. Ruta et al.

    Calibrated measurement of gating-charge arginine displacement in the KvAP voltage-dependent K+ channel

    Cell

    (2005)
  • G. Schreiber et al.

    Energetics of protein–protein interactions: analysis of the barnase–barstar interface by single mutations and double mutant cycles

    J. Mol. Biol.

    (1995)
  • K.J. Shon et al.

    kappa-Conotoxin PVIIA is a peptide inhibiting the shaker K+ channel

    J. Biol. Chem.

    (1998)
  • J.J. Smith et al.

    Differential phospholipid binding by site 3 and site 4 toxins: Implications for structural variability between voltage-sensitive sodium channel domains

    J. Biol. Chem.

    (2005)
  • K.N. Srinivasan et al.

    SCORPION, a molecular database of scorpion toxins

    Toxicon

    (2002)
  • K.J. Swartz et al.

    An inhibitor of the Kv2.1 potassium channel isolated from the venom of a Chilean tarantula

    Neuron

    (1995)
  • K.J. Swartz et al.

    Hanatoxin modifies the gating of a voltage-dependent K+ channel through multiple binding sites

    Neuron

    (1997)
  • K.J. Swartz et al.

    Mapping the receptor site for hanatoxin, a gating modifier of voltage- dependent K+ channels

    Neuron

    (1997)
  • H. Takahashi et al.

    Solution structure of hanatoxin1, a gating modifier of voltage-dependent K+ channels: common surface features of gating modifier toxins

    J. Mol. Biol.

    (2000)
  • K. Takeuchi et al.

    Solution structure of w-grammotoxin SIA, a gating modifier of P/Q and N type calcium channels

    J. Mol. Biol.

    (2002)
  • V.V. Zarayskiy et al.

    Heteropoda toxin 2 is a gating modifier toxin specific for voltage-gated K+ channels of the Kv4 family

    Toxicon

    (2005)
  • A.A. Alabi et al.

    Compatibility of the paddle region revealed by chimeras between archebacterial and eukaryotic Kv channels

    Biophys. J.

    (2006)
  • C.S. Anderson et al.

    Charybdotoxin block of single Ca2+-activated K+ channels. Effects of channel gating, voltage, and ionic strength

    J. Gen. Physiol.

    (1988)
  • C. Bernard et al.

    Solution structure of hpTX2, a toxin from Heteropoda venatoria spider that blocks Kv4.2 potassium channel

    Protein Sci.

    (2000)
  • A. Boccaccio et al.

    Binding of kappa-conotoxin PVIIA to Shaker K+ channels reveals different K+ and Rb+ occupancies within the ion channel pore

    J. Gen. Physiol.

    (2004)
  • L.M. Boland et al.

    omega-Conotoxin block of N-type calcium channels in frog and rat sympathetic neurons

    J. Neurosci.

    (1994)
  • Y. Bourne et al.

    Crystal structure of a Cbtx-AChBP complex reveals essential interactions between snake alpha-neurotoxins and nicotinic receptors

    Embo. J.

    (2005)
  • M.D. Cahalan

    Modification of sodium channel gating in frog myelinated nerve fibres by Centruroides sculpturatus scorpion venom

    J Physiol. (Lond).

    (1975)
  • P.H. Celie et al.

    Crystal structure of nicotinic acetylcholine receptor homolog AChBP in complex with an alpha-conotoxin PnIA variant

    Nat. Struct. Mol. Biol.

    (2005)
  • B. Chagot et al.

    Solution structure of Phrixotoxin 1, a specific peptide inhibitor of Kv4 potassium channels from the venom of the theraphosid spider Phrixotrichus auratus

    Protein Sci.

    (2004)
  • M.Z. Chou et al.

    Resiniferatoxin binds to the capsaicin receptor (TRPV1) near the extracellular side of the S4 transmembrane domain

    Biochemistry

    (2004)
  • R.S. Chuang et al.

    Inhibition of T-type voltage-gated calcium channels by a new scorpion toxin

    Nat. Neurosci.

    (1998)
  • K.B. Craven et al.

    CNG and HCN channels: two peas, one pod

    Annu. Rev. Physiol,

    (2006)
  • D. del Camino et al.

    Status of the intracellular gate in the activated-not-open state of shaker K+ channels

    J. Gen. Physiol.

    (2005)
  • S. Diochot et al.

    Effects of phrixotoxins on the Kv4 family of potassium channels and implications for the role of Ito1 in cardiac electrogenesis

    Br. J. Pharmacol.

    (1999)
  • S. Diochot et al.

    APETx1, a new toxin from the sea anemone Anthopleura elegantissima, blocks voltage-gated human ether-a-go-go-related gene potassium channels

    Mol. Pharmacol.

    (2003)
  • P.T. Ellinor et al.

    Structural determinants of the blockade of N-type calcium channels by a peptide neurotoxin

    Nature

    (1994)
  • P. Escoubas et al.

    Novel tarantula toxins for subtypes of voltage-dependent potassium channels in the Kv2 and Kv4 subfamilies

    Mol. Pharmacol.

    (2002)
  • E. Garcia et al.

    A marine snail neurotoxin shares with scorpion toxins a convergent mechanism of blockade on the pore of voltage-gated K channels

    J. Gen. Physiol.

    (1999)
  • J. Herrington et al.

    Blockers of the delayed-rectifier potassium current in pancreatic beta-cells enhance glucose-dependent insulin secretion

    Diabetes

    (2006)
  • P. Hidalgo et al.

    Revealing the architecture of a K+ channel pore through mutant cycles with a peptide inhibitor

    Science

    (1995)
  • L.D. Islas et al.

    Voltage sensitivity and gating charge in Shaker and Shab family potassium channels

    J. Gen. Physiol.

    (1999)
  • Cited by (142)

    View all citing articles on Scopus
    View full text