Dendrotoxin

Last updated

Dendrotoxins are a class of presynaptic neurotoxins produced by mamba snakes ( Dendroaspis ) that block particular subtypes of voltage-gated potassium channels in neurons, thereby enhancing the release of acetylcholine at neuromuscular junctions. Because of their high potency and selectivity for potassium channels, dendrotoxins have proven to be extremely useful as pharmacological tools for studying the structure and function of these ion channel proteins.

Contents

Sequence alignment of dendrotoxins and BPTI. Amino acid residues with similar properties are colored accordingly. Sequence alignment dendrotoxins.jpg
Sequence alignment of dendrotoxins and BPTI. Amino acid residues with similar properties are colored accordingly.

Dendrotoxins have been shown to block particular subtypes of voltage-gated potassium (K+) channels in neuronal tissue.[ citation needed ] In the nervous system, voltage-gated K+ channels control the excitability of nerves and muscles by controlling the resting membrane potential and by repolarizing the membrane during action potentials. Dendrotoxin has been shown to bind the nodes of Ranvier of motor neurons [1] and to block the activity of these potassium channels. In this way, dendrotoxins prolong the duration of action potentials and increase acetylcholine release at the neuromuscular junction, which may result in muscle hyperexcitability and convulsive symptoms.

Dendrotoxin structure

3D model of alpha-dendrotoxin. colored in red are positively charged amino acid residues in the N-terminus and the b-turn region that are believed to be important for potassium channel binding. Dendrotoxin alpha.jpg
3D model of alpha-dendrotoxin. colored in red are positively charged amino acid residues in the N-terminus and the β-turn region that are believed to be important for potassium channel binding.

Dendrotoxins are ~7kDa proteins consisting of a single peptide chain of approximately 57-60 amino acids. Several homologues of alpha-dendrotoxin have been isolated, all possessing a slightly different sequence. However, the molecular architecture and folding conformation of these proteins are all very similar. Dendrotoxins possess a very short 310-helix near the N-terminus of the peptide, while a two turn alpha-helix occurs near the C-terminus. A two-stranded antiparallel β-sheet occupies the central part of the molecular structure. These two β-strands are connected by a distorted β-turn region [2] that is thought to be important for the binding activity of the protein. All dendrotoxins are cross-linked by three disulfide bridges, which add stability to the protein and greatly contribute to its structural conformation. The cysteine residues forming these disulfide bonds have been conserved among all members of the dendrotoxin family, and are located at C7-C57, C16-C40, and C32-C53 (numbering according to alpha-dendrotoxin).

The dendrotoxins are structurally homologous to the Kunitz-type serine protease inhibitors, including bovine pancreatic trypsin inhibitor (BPTI). Alpha-dendrotoxin and BPTI have been shown to have 35% sequence identity as well as identical disulfide bonds. Despite the structural homology between these two proteins, dendrotoxins do not appear to exhibit any measurable inhibitory protease activity like BPTI. This loss of activity appears to result from the absence of key amino acid residues that produce structural differences that hinder the key interactions necessary for the protease activity seen in BPTI.

Dendrotoxins are basic proteins that possess a net positive charge when present in neutral pH. Most of the positively charged amino acid residues of dendrotoxins are located in the lower part of the structure, creating a cationic domain on one side of the protein. Positive charge results from lysine (Lys) and arginine (Arg) residues that are concentrated in three primary regions of the protein: near the N-terminus (Arg3, Arg4, Lys5), near the C-terminus (Arg54, Arg55) and at the narrow β-turn region (Lys28, Lys29, Lys30). [3] It is believed that these positively charged residues can play a critical role in dendrotoxin binding activity, as they can make potential interactions with the anionic sites (negatively charged amino acids) in the pore of potassium channels.

Biological activity

Pharmacology

A single dendrotoxin molecule associates reversibly with a potassium channel in order to exert its inhibitory effect. It is proposed that this interaction is mediated by electrostatic interactions between the positively charged amino acid residues in the cationic domain of dendrotoxin and the negatively charged residues in the ion channel pore. Potassium channels, similar to other cation-selective channels, are believed to have a cloud of negative charges that precede the opening to the channel pore that help conduct potassium ions through the permeation pathway. It is generally believed (though not proven) that a dendrotoxin molecules bind to anionic sites near the extracellular surface of the channel and physically occlude the pore, thereby preventing ion conductance. However, Imredy and MacKinnon [4] have proposed that delta-dendrotoxin may have an off-center binding site on their target proteins, and may inhibit the channel by altering the structure of the channel, rather than physically blocking the pore.

Biologically important residues

Many studies have attempted to identify which amino acid residues are important for binding activity of dendrotoxins to their potassium channel targets. Harvey et al. [5] used residue-specific modifications to identify positively charged residues that were crucial to the blocking activity of dendrotoxin-I. They reported that acetylation of Lys5 near the N-terminal region and Lys29 in the beta-turn region led to substantial decreases in DTX-I binding affinity. Similar results have been shown with dendrotoxin-K using site-directed mutagenesis to substitute positively charged lysine and arginine residues to neutral alanines. These results, along with many others, have implicated that the positively charged lysines in the N-terminal half, particularly Lys5 in the 310-helix, play a very important role in the dendrotoxin binding to their potassium channel targets. The lysine residues in the β-turn region has provided more confounding results, appearing to be biologically critical in some dendrotoxin homologues and not necessary for others. Furthermore, mutation of the entire lysine triplet (K28-K29-K30) to Ala-Ala-Gly in alpha-DTX resulted in very little change in biological activity.

There is a general agreement that the conserved lysine residue near the N-terminus (Lys5 in alpha-DTX) is crucial for the biological activity of all dendrotoxins, while additional residues, such as those in the beta-turn region, might play a role in dendrotoxin specificity by mediating the interactions of individual toxins to their individual target sites. This not only helps explain the stringent specificity of some dendrotoxins for different subtypes of voltage-gated K+ channels, but also accounts for differences in the potency of dendrotoxins for common K+ channels. For example, Wang et al. [6] showed that the interaction of dendrotoxin-K with KV1.1 is mediated by its lysine residues in both the N-terminus and the β-turn region, while alpha-dendrotoxin appears to interact with its target solely through the N-terminus. This less expansive interactive domain may help explain why alpha-dendrotoxin is less discriminative while dendrotoxin-K is strictly selective for KV1.1.

Uses in research

Potassium channels of vertebrate neurons display a high degree of diversity that allows neurons to precisely tune their electrical signaling properties by expression of different combinations of potassium channel subunits. Furthermore, because they regulate ionic flux across biological membranes, they are important in many aspects of cellular regulation and signal transduction of different cell types. Therefore, voltage-gated potassium channels are targets for a wide range of potent biological toxins from such organisms as snakes, scorpions, sea anemones, and cone snails. Thus, venom purification has led to the isolation of peptide toxins such as the dendrotoxins, which have become useful pharmacological tools for the study of potassium channels. Because of their potency and selectivity for different subtypes of potassium channels, dendrotoxins have become useful as molecular probes for the structural and functional study of these proteins. This may help improve our understanding of the roles played by individual channel types, as well as assist in the pharmacological classification of these diverse channel types. [7] Furthermore, the availability of radiolabelled dendrotoxins provides a tool for the screening of other sources in a search for new potassium channel toxins, such as the kalicludine class of potassium channel toxins in sea anemones. Lastly, the structural information provided by dendrotoxins may provide clues to the synthesis of therapeutic compounds that may target particular classes of potassium channels. Dendrotoxin I has also been used to help purify and characterize the K+ channel protein to which it binds via different binding assay and chromatography techniques. [8]

Related Research Articles

<span class="mw-page-title-main">Maurotoxin</span> Toxin in scorpions

Maurotoxin is a peptide toxin from the venom of the Tunisian chactoid scorpion Scorpio maurus palmatus, from which it was first isolated and from which the chemical gets its name. It acts by blocking several types of voltage-gated potassium channel.

<span class="mw-page-title-main">KCNA4</span> Protein-coding gene in the species Homo sapiens

Potassium voltage-gated channel subfamily A member 4 also known as Kv1.4 is a protein that in humans is encoded by the KCNA4 gene. It contributes to the cardiac transient outward potassium current (Ito1), the main contributing current to the repolarizing phase 1 of the cardiac action potential.

<span class="mw-page-title-main">KCNB1</span> Protein-coding gene in the species Homo sapiens

Potassium voltage-gated channel, Shab-related subfamily, member 1, also known as KCNB1 or Kv2.1, is a protein that, in humans, is encoded by the KCNB1 gene.

<span class="mw-page-title-main">Cobatoxin</span> Chemical compound

Cobatoxin is a toxin present in the venom of the scorpion Centruroides noxius. It blocks two potassium channel subtypes; voltage-gated and calcium-activated channels.

Kaliotoxin (KTX) inhibits potassium flux through the Kv1.3 voltage-gated potassium channel and calcium-activated potassium channels by physically blocking the channel-entrance and inducing a conformational change in the K+-selectivity filter of the channel.

<span class="mw-page-title-main">Pandinotoxin</span> Chemical compound

Pandinotoxins are toxins from the venom of the emperor scorpion Pandinus imperator. They are selective blockers of voltage-gated potassium channels

Parabutoxin (PBTx) is a Shaker-related voltage-gated K+ channel (Kvα1) inhibitor purified from different Parabuthus scorpion species found in southern Africa. It occurs in different forms: parabutoxin 1 (PBTx1), parabutoxin 2 (PBTx2), parabutoxin 3 (PBTx3) and parabutoxin (PBTx10). The different variants have different affinities towards Kvα1 channels.

Butantoxin (BuTX) is a compound of the venom of three Brazilian and an Argentinean scorpion species of the genus Tityus. Butantoxin reversibly blocks the voltage-gated K+ channels Shaker B and Kv1.2, and the Ca2+-activated K+ channelsKCa 1.1 and KCa 3.1.

<span class="mw-page-title-main">Pandinus imperator (Pi3) toxin</span>

Pi3 toxin is a purified peptide derivative of the Pandinus imperator scorpion venom. It is a potent blocker of voltage-gated potassium channel, Kv1.3 and is closely related to another peptide found in the venom, Pi2.

Tamulotoxin is a venomous neurotoxin from the Indian Red Scorpion.

<span class="mw-page-title-main">Ball and chain inactivation</span> Model in neuroscience

In neuroscience, ball and chain inactivation is a model to explain the fast inactivation mechanism of voltage-gated ion channels. The process is also called hinged-lid inactivation or N-type inactivation. A voltage-gated ion channel can be in three states: open, closed, or inactivated. The inactivated state is mainly achieved through fast inactivation, by which a channel transitions rapidly from an open to an inactivated state. The model proposes that the inactivated state, which is stable and non-conducting, is caused by the physical blockage of the pore. The blockage is caused by a "ball" of amino acids connected to the main protein by a string of residues on the cytoplasmic side of the membrane. The ball enters the open channel and binds to the hydrophobic inner vestibule within the channel. This blockage causes inactivation of the channel by stopping the flow of ions. This phenomenon has mainly been studied in potassium channels and sodium channels.

BgK is a neurotoxin found within secretions of the sea anemone Bunodosomagranulifera which blocks voltage-gated potassium channels, thus inhibiting neuronal repolarization.

Kaliseptine (AsKS) is a neurotoxin which can be found in the snakelocks anemone Anemonia viridis. It belongs to a class of sea anemone neurotoxins that inhibits voltage-gated potassium channels.

HsTx1 is a toxin from the venom of the scorpion Heterometrus spinifer. HsTx1 is a very potent inhibitor of the rat Kv1.3 voltage-gated potassium channel.

Pi4 is a short toxin from the scorpion Pandinus imperator that blocks specific potassium channels.

Kalicludine (AsKC) is a blocker of the voltage-dependent potassium channel Kv1.2 found in the snakeslocks anemone Anemonia viridis, which it uses to paralyse prey.

<span class="mw-page-title-main">ImKTx88</span>

ImKTx88 is a selective inhibitor of the Kv1 ion channel family that can be isolated from the venom of the Isometrus maculatus. This peptide belongs to the α-KTx subfamily and is classified as a pore-blocking toxin.

BmP02, also known as α-KTx 9.1 or Bmkk(6), is a toxin from the Buthus Martensi Karsch (BmK) scorpion. The toxin acts on potassium channels, blocking Kv1.3 and slowing the deactivation of Kv4.2. BmP02 is not toxic to humans or mice.

<span class="mw-page-title-main">OSK3</span>

OSK3, from the venom of the scorpion Orthochirus scrobiculosus, is a potassium channel blocker that belongs to the α-KTx8 subfamily and targets the voltage-gated potassium channels KCNA2 (Kv1.2), and KCNA3 (Kv1.3).

Kunitz-type serine protease inhibitor APEKTx1 is a peptide toxin derived from the sea anemone Anthopleura elegantissima. This toxin has a dual function, acting as an serine protease inhibitor and as a pore blocker of Kv1.1, a shaker related voltage-gated potassium channel.

References

  1. Gasparini S, Danse J-M, Licoq A, Pinkasfeld S, Zinn-Justin S, Young LC, C.L. de Medeiros C, Rowan EG, Harvey AL, and Me’nez A (1998). Delineation of the Functional Site of alpha-dendrotoxin: The functional topographies of dendrotoxins are different but share a conserved core with those of other KV1 potassium channel-blocking toxins. Journal of Biological Chemistry 273:25393-25403
  2. Katoh E, Nishio H, Inui T, Nishiuchi Y, Kimura T, Sakakibara S, Yamazaki T (2000). Structural Basis for the Biological Activity of Dendrotoxin-I, a Potent Potassium Channel Blocker. Biopolymers 54:44-57
  3. Swaminathan P, Hariharan M, Murali R, Singh CU (1996). Molecular Structure, Conformational Analysis, and Structure-Activity Studies of Dendrotoxin and Its Homologues Using Molecular Mechanics and Molecular Dynamics Techniques. Journal of Medicinal Chemistry. 39:2141-2155
  4. Imredy JP, and MacKinnon R (2000). Energetic and Structural Interactions between delta-Dendrotoxin and a Voltage-gated Potassium Channel. Journal of Molecular Biology 296:1283-1294
  5. Harvey AL, Rowan EG, Vatanpour H, Engstrom A, Westerlund B, Karlsson E (1997). Changes to biological activity following acetylation of dendrotoxin I from Dendroaspis polylepis (black mamba). ' 35:1263-1273
  6. Wang FC, Bell N, Reid P, Smith LA, McIntosh P, Robertson B, and Dolly JO (1999). Identification of residues in dendrotoxin K responsible for its discrimination between neuronal K+ channels containing KV1.1 and 1.2 alpha subunits. European Journal of Biochemistry 263:222-229
  7. Yoshida S and Matsumoto S (2005). Effects of alpha-dendrotoxin on K+ currents and action potentials in tetrodotoxin-resistant adult rat trigeminal ganglion neurons. Journal of Pharmacology and Experimental Therapeutics 314:437-445
  8. Rehm, H.; Lazdunski, M. (1988-07-01). "Purification and subunit structure of a putative K+-channel protein identified by its binding properties for dendrotoxin I." Proceedings of the National Academy of Sciences. 85 (13): 4919–4923. Bibcode:1988PNAS...85.4919R. doi: 10.1073/pnas.85.13.4919 . ISSN   0027-8424. PMC   80549 . PMID   2455300.