Huwentoxin

Last updated

Huwentoxins (HWTX) are a group of neurotoxic peptides found in the venom of the Chinese bird spider Haplopelma schmidti . The species was formerly known as Haplopelma huwenum, Ornithoctonus huwena and Selenocosmia huwena. [1] While structural similarity can be found among several of these toxins, HWTX as a group possess high functional diversity.

Contents

Sources

Huwentoxins are neurotoxic peptides produced by the Chinese bird spider, Haplopelma schmidti. [2]

Overview

The venom of H. schmidti contains a large variety of neurotoxins, which function to paralyze the spider's prey. So far, 14 of the isolated primarily neurotoxic peptide components have been characterized and investigated. In the following, two subfamilies of the HWTX are described: those targeting voltage-gated calcium channels, and those targeting voltage-gated sodium channels.

Toxins targeting voltage-gated calcium channels (VGCC)

Huwentoxin-I

HWTX-I is the most abundant toxic component in the venom of H. schmidti. It inhibits presynaptic N-type Ca2+ channels.

Chemistry

The molecular weight of HWTX-I is 3750 Da. The toxin comprises 33 residues, including six cysteines that form three disulfide linkages. [3] [4] These were assigned as Cys2-Cys17, Cys9-Cys22, and Cys16-Cys29 and are buried within the molecule. [5] The molecule adopts a compact structure consisting of a small triple-stranded antiparallel beta-sheet and five beta-turns. It was found that the structure contains an inhibitor cystine knot (ICK) motif. To form this motif, three disulfide bridges are needed. Two of them create a loop through which the third disulfide bridge passes. [4] [6] [7] The structure of HWTX-I is very stable, secondary structure elements do not significantly alter under different pH conditions or after heating. [8]

Mode of action

HWTX-I selectively inhibits N-type HVA channels. [9] A recent study found that HWTX-I also inhibits Na+ channels. [10]

Effects

In mice the intraperitoneal LD50 of HWTX-I is 0.70 mg/kg, the intracisternal LD50 has been determined as 9.40 µg/kg. Neurotoxic symptoms after intraperitoneal injection were gasping, excitation, spastic paralysis of the hindlimb and asynergia. [3] [11]

HWTX-I is a potential novel analgesic pharmaceutic. [12] Epidural administration of HWTX-I in rats with chronic neuropathic pain blocked heat hyperalgesia and mechanical allodynia in the injured hindpaw of rats, indicating that epidurally administered HWTX-I could alleviate neuropathic pain. [13] Cytosolic Ca2+ overload is one of the primary factors for inflammatory cells activation, therefore Ca2+ channel blockers can have a potential role as an anti-inflammatory drug. HWTX-I can relieve pain in the inflammatory joints and eliminate arthrocele to some degree. In a rat model of rheumatoid arthritis HWTX-I is able to decrease the concentration of tumor necrosis factor α (TNF-α) in serum and decrease the mRNA expression level interleukin 1β (IL-1β) and interleukin 6 (IL-6). [14]

Primary sequences of the peptide isolated from the venom of spider H. schmidti. The disulfide bridge linkage pattern is shown with the connector lines. Cysteines are shown in red. An asterisk indicates that the C-terminal carboxyl group is amidated. HWTX amino acid sequences.png
Primary sequences of the peptide isolated from the venom of spider H. schmidti. The disulfide bridge linkage pattern is shown with the connector lines. Cysteines are shown in red. An asterisk indicates that the C-terminal carboxyl group is amidated.

Huwentoxin-X

HWTX-X is the smallest peptide among the huwentoxins so far isolated.

Chemistry

HWTX-X has a molecular mass of 2931 Da. It comprises 28 amino acid residues, including six cysteine residues forming three disulfide bridges. Like most huwentoxins, it adopts the ICK motif. [15] HWTX-X shows little homology with other huwentoxins, however, it can cause reversible blockage of N-type Ca2+ channels in rat dorsal root ganglion cells under whole-cell voltage clamp conditions. It does show more than 50% homology with the toxin Ptu1 from the assassin bug Peirates turpis and ω-conotoxin SVIA from the Conus striatus , two N-type Ca2+ blockers. [16]

Mode of action

HWTX-X has selectivity for isoforms of N-type Ca2+ channels, compared with ω-conotoxins GVIA and MVIIA.

Effects

HWTX-X specifically blocks GVIA-sensitive, N-type Ca2+ channels in rat dorsal root ganglion cells. It does not block L-type Ca2+ channels. While structurally similar to ω-conotoxins that block the twitch response to electrical nerve stimulation, HWTX-X has no effect on the twitch response of rat vas deferens. [15]

Toxins targeting voltage-gated sodium channels (VGSC)

Huwentoxin-II

HWTX-II is an insecticidal peptide and is structurally unusual compared to the other HWTX in that it lacks the typical ICK motif.

Chemistry

HWTX-II consists of 37 amino acid residues including six cysteines involved in three disulfide bridges. [17] The disulfide linkage of HWTX-II was assigned as Cys4-Cys18, Cys8-Cys29 and Cys23-Cys34, forming a 1-3,2-5 and 4-6 disulfide connectivity. [18] The three-dimensional structure of HWTX II contains two beta-turns (Cys4-Ser7 and Lys24-Trp27) and a double stranded antiparallel beta-sheet (Tryp27-Cys29 and Cys34-Lys36).

Mode of action

HWTX-II was able to reversibly paralyze cockroaches for several hours, with a median knockdown dose ED50 of 127 ± 54 µg/g. [19] HWTX-II blocks neuromuscular transmission in the isolated mouse nerve diaphragm preparation and acts cooperatively to potentiate the activity of HWTX I. [18]

Effects

The toxin can paralyze cockroaches.

Huwentoxin-III

HWTX-III is a selective inhibitor of insect voltage-gated Na+ channels. It has a natural mutant named HWTX-IIIa, the sequence of which is only truncated a tryptophan (Trp33) residue from C-terminal of HWTX-III [20] This mutant does not have the same effects as HWTX-III, suggesting that Trp33 is an important residue related to the biological function of HWTX-III.

Chemistry

HWTX III contains 33 residues, including six cysteine residues, which form three disulfide bridges. It has a molecular weight of 3853 Da. [20]

Mode of action

HWTX-III inhibits voltage-gated Na+ channels on dorsal unpaired median (DUM) neurons (concentration of toxin at half-maximal inhibition (IC50) ≈1.106 µmol/L) in a similar way as tetrodotoxin (TTX). HWTX-III has no effect on the kinetics of activation and inactivation.

Effects

HWTX-III showed no effect on the activation and inactivation kinetics of the insect neuron VGSCs, and also no change in the ion selectivity of the channels. However, it can reversibly paralyze cockroaches, depressing the amplitude of the Na+ currents on cockroach DUM neurons. [21] HWTX-III is able to enhance smooth muscle reactions elicited by nerve stimulation of the isolated rat vas deferens. [20]

Huwentoxin-IV

HWTX-IV is an inhibitor of tetrodotoxin (TTX) -sensitive voltage-gated Na+ channels.

Chemistry

HWTX-IV comprises 35 amino acid residues, with three disulfide bridges, belonging to the ICK motif structural family. Its molecular weight is 4108 Da. The C-terminal carboxyl group of this toxin is amidated. The disulfide linkage of HWTX-IV is Cys-2–Cys-17, Cys-9–Cys-24, and Cys-16–Cys-31, adopting a 1–4, 2–5, 3–6 disulfide pattern.

Mode of action

HWTX-IV specifically blocks neuronal TTX-sensitive voltage-gated Na+ channels in adult rat dorsal root ganglion neurons while having no effect on TTX-resistant voltage-gated Na+ channels. HWTX-IV inhibits these channels by binding to receptor site 4 and trapping the domain II voltage sensor in the closed configuration. HWTX-IV is a gating modifier that is likely to functionally behave as a simple channel inhibitor. Obvious gating modifier type behavior was only observed in unlikely conditions: extreme depolarizations or very prolonged strong depolarizations. [22]

Effects

Experiments have revealed that HWTX-IV introduces a significant upgrade to the pain threshold in rats. With HWTX-IV selectively blocking TTX-sensitive voltage-gated Na+ channels (which are involved in the pain pathways), it is hoped that it can be applied in pain therapy. HWTX-IV at high doses elicited no effects in cockroaches. Nor does it target VGSCs in cardiac or skeletal muscle of both amphibians and mammals. [23]

Huwentoxin-VII, -VIII

HWTX-VII and HWTX-VIII are insecticidal peptides with amino acid sequences and bioactivities similar to that of HWTX-II.

Chemistry

HWTX-VII and HWTX-VIII are composed of 35 and 36 amino acid residues respectively, both including six cysteines. They might adopt similar structural scaffolding and the same disulfide bridge pattern as HWTX-II. [24]

Mode of action

Both HWTX-VII and HWTX-VIII block neuromuscular transmission in the isolated mouse phrenic nerve-diaphraghm preparation and act cooperatively with HWTX-I.

Effects

Both toxins paralyze locusts and kill mice through intracerebroventricular injection.

See also

Related Research Articles

Conotoxin

A conotoxin is one of a group of neurotoxic peptides isolated from the venom of the marine cone snail, genus Conus.

Cyriopagopus hainanus is a species of spider in the family Theraphosidae (tarantulas), found in China. It is one of a number of species from China and Vietnam known as "Chinese bird spider". It produces a venom containing numerous compounds capable of blocking neurotransmitters, including neurotoxic peptides called hainantoxins.

Delta atracotoxin

Delta atracotoxin is a low-molecular-weight neurotoxic polypeptide found in the venom of the Sydney funnel-web spider.

Maurotoxin

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.

Spider toxin Family of toxins produced by spiders

Spider toxins are a family of proteins produced by spiders which function as neurotoxins. The mechanism of many spider toxins is through blockage of calcium channels.

Birtoxin is a neurotoxin from the venom of the South African Spitting scorpion. By changing sodium channel activation, the toxin promotes spontaneous and repetitive firing much like pyrethroid insecticides do

BeKm-1 is a toxin from the Central Asian scorpion Buthus eupeus. BeKm-1 acts by selectively inhibiting the human Ether-à-go-go Related Gene (hERG) channels, which are voltage gated potassium ion channels.

Jingzhaotoxin proteins are part of a venom secreted by Chilobrachys jingzhao, the Chinese tarantula. and act as neurotoxins. There are several subtypes of jingzhaotoxin, which differ in terms of channel selectivity and modification characteristics. All subspecies act as gating modifiers of sodium channels and/or, to a lesser extent, potassium channels.

Heteroscodratoxin-1

Heteroscodratoxin-1 is a neurotoxin produced by the venom glands of Heteroscodra maculata that shifts the activation threshold of voltage-gated potassium channels and the inactivation of Nav1.1 sodium channels to more positive potentials.

Raventoxins are neurotoxins from the venom of the spider Macrothele raveni.

Hainantoxins (HNTX) are neurotoxins from the venom of the Chinese bird spider Haplopelma hainanum. Hainantoxins specifically inhibit tetrodotoxin-sensitive Voltage-gated sodium channels, thereby causing blockage of neuromuscular transmission and paralysis. Currently, 13 different hainantoxins are known, but only HNTX-I, -II, -III, -IV and -V have been investigated in detail.

Covalitoxin-II is a peptide toxin that is produced by the spider Coremiocnemis validus. It can induce excitatory, non-lethal behavioral symptoms like quivering and jerking in crickets.

CSTX is a name given to a group of closely related neurotoxic peptides present in the venom of the wandering spider Cupiennius salei. There are twenty types so far described for this protein group. However, some are reclassified into cupiennins group of toxin, including CSTX-3, -4, -5, and -6, because of their chemical affinity. The first thirteen were isolated and identified in 1994 by Lucia Kuhn-Nentwig, Johann Schaller, and Wolfgang Nentwig of the Zoological Institute at the University of Bern, Switzerland. The different types are most likely the products of splicing variant of the same gene. They are all L-type calcium channel blockers, and also exhibit cytolytic activity by forming an alpha-helix across the cell membrane in mammalian neurons. They also inhibit voltage-gated calcium channels in insect neurons.

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.

Agelenin, also called U1-agatoxin-Aop1a, is an antagonist of the presynaptic P-type calcium channel in insects. This neurotoxic peptide consists of 35 amino acids and can be isolated from the venom of the spider Allagelena opulenta.

Spinoxin is a 34-residue peptide neurotoxin isolated from the venom of the Malaysian black scorpion Heterometrus spinifer. It is part of the α-KTx6 subfamily and exerts its effects by inhibiting voltage-gated potassium channels, specifically Kv1.2 and Kv1.3.

LmαTX5 is an α-scorpion toxin which inhibits the fast inactivation of voltage-gated sodium channels. It has been identified through transcriptome analysis of the venom gland of Lychas mucronatus, also known as the Chinese swimming scorpion – a scorpion species which is widely distributed in Southeast Asia.

μ-THTX-Cl6a, also known as Cl6a, is a 33-residue peptide toxin extracted from the venom of the spider Cyriopagopus longipes. The toxin acts as an inhibitor of the tetrodotoxin-sensitive (TTX-S) voltage-gated sodium channel (NaV1.7), thereby causing sustained reduction of NaV1.7 currents.

DKK-Sp1

DKK-SP1 is one of the 100,000 neurotoxins present in the Buthusmartensii Karsch (BmK) scorpion. DKK-SP1 targets the voltage-gated sodium channel Nav1.8 by inhibiting its current. It has been used in traditional Chinese medicine for centuries by treating many diseases like epilepsy and cancer.

Prototoxin-1, or Beta/omega-theraphotoxin-Tp1a, is a 35-amino-acid peptide neurotoxin extracted from the venom of the tarantula Thrixopelma pruriens. Prototoxin-1 belongs to the inhibitory cystine knot (ICK) family of peptide toxins, which have been known to potently inhibit voltage-gated ion channels. Prototoxin-1 selectively blocks low voltage threshold “T-type” calcium channels., voltage gated sodium channels and the nociceptor cation channel TRPA1. Due to its unique ability to bind to TRPA1, Prototoxin-1 has been implicated as a valuable pharmacological reagent with potential applications in clinical contexts with regards to pain and inflammation

References

  1. "Taxon details Haplopelma schmidti von Wirth, 1991". World Spider Catalog. Natural History Museum Bern. Retrieved 2016-05-17.
  2. Wang JF, Peng XJ, Xie LP. A new species of the genus Selenocosmia from south China. Acta Sci. Nat. Univ. Norm. Hunan. 1993;16(1):51-54
  3. 1 2 Liang SP, Zhang DY, Pan X, Chen Q, Zhou PA. Properties and amino acid sequence of huwentoxin-I, a neurotoxin purified from the venom of the Chinese bird spider Selenocosmia huwena. Toxicon. 1993 Aug;31(8):969-78.
  4. 1 2 Qu Y, Liang S, Ding J, Liu X, Zhang R, Gu X. Proton nuclear magnetic resonance studies on huwentoxin-I from the venom of the spider Selenocosmia huwena: 2. Three-dimensional structure in solution. J Protein Chem. 1997 Aug;16(6):565-74.
  5. Zhang D, Liang S. Assignment of the three disulfide bridges of huwentoxin-I, a neurotoxin from the spider selenocosmia huwena. J Protein Chem. 1993 Dec;12(6):735-40.
  6. Pallaghy PK, Nielsen KJ, Craik DJ, Norton RS. A common structural motif incorporating a cystine knot and a triple-stranded beta-sheet in toxic and inhibitory polypeptides. Protein Sci. 1994 Oct;3(10):1833-9.
  7. Norton RS, Pallaghy PK. The cystine knot structure of ion channel toxins and related polypeptides. Toxicon. 1998 Nov;36(11):1573-83.
  8. Liang SP, Zong X, Luo JC, Jing H, Gu XC. Secondary structure study of huwentoxin-I, a neurotoxin from the venom of spider Selenocosmia huwena. Acta Scientiarum Naturalium Universitatis Pekinenisis. 1993;29:668-74.
  9. Peng K, Chen XD, Liang SP. The effect of Huwentoxin-I on Ca(2+) channels in differentiated NG108-15 cells, a patch-clamp study. Toxicon. 2001 Apr;39(4):491-8.
  10. Wang M, Rong M, Xiao Y, Liang S. The effects of huwentoxin-I on the voltage-gated sodium channels of rat hippocampal and cockroach dorsal unpaired median neurons. Peptides. 2012 Mar;34(1):19-25.
  11. Zhou PA, Xie XJ, Li M, Yang DM, Xie ZP, Zong X, et al. Blockade of neuromuscular transmission by huwentoxin-I, purified from the venom of the Chinese bird spider Selenocosmia huwena. Toxicon. 1997 Jan;35(1):39-45.
  12. Che N, Wang L, Gao Y, An C. Soluble expression and one-step purification of a neurotoxin Huwentoxin-I in Escherichia coli. Protein Expr Purif. 2009 Jun;65(2):154-9.
  13. Luo ZM, Li LL, Zhang J, Xie HY, Shen JQ, Liang SP. Effects of epidural administration of HWTX-I in rats with chronic neuropathic pain. Chin J Pain Med. 2002;8:219-24.
  14. Wen Tao Z, Gu Yang T, Ying R, Mao Cai W, Lin L, Chi Miao L, et al. The antinociceptive efficacy of HWTX-I epidurally administered in rheumatoid arthritis rats. Int J Sports Med. 2011 Nov;32(11):869-74.
  15. 1 2 Liu Z, Dai J, Dai L, Deng M, Hu Z, Hu W, et al. Function and solution structure of Huwentoxin-X, a specific blocker of N-type calcium channels, from the Chinese bird spider Ornithoctonus huwena. J Biol Chem. 2006 Mar 31;281(13):8628-35.
  16. Bernard C, Corzo G, Mosbah A, Nakajima T, Darbon H. Solution structure of Ptu1, a toxin from the assassin bug Peirates turpis that blocks the voltage-sensitive calcium channel N-type. Biochemistry. 2001 Oct 30;40(43):12795-800.
  17. Anette SN. Tarantula (Eurypelma californicum) venom, a multicomponent system. Biol Chem Hoppe-Seyler. 1989;370:485-98.
  18. 1 2 Shu Q, Liang SP. Purification and characterization of huwentoxin-II, a neurotoxic peptide from the venom of the Chinese bird spider Selenocosmia huwena. J Pept Res. 1999 May;53(5):486-91.
  19. Shu Q, Lu SY, Gu XC, Liang SP. Sequence-specific Assignment of (1)H-NMR Resonance and Determination of the Secondary Structure of HWTX-II. Sheng Wu Hua Xue Yu Sheng Wu Wu Li Xue Bao (Shanghai). 2001;33(1):65-70.
  20. 1 2 3 Huang RH, Liu ZH, Liang SP. Purification and characterization of a neurotoxic peptide huwentoxin-III and a natural inactive mutant from the venom of the spider Selenocosmia huwena Wang (Ornithoctonus huwena Wang). Sheng Wu Hua Xue Yu Sheng Wu Wu Li Xue Bao (Shanghai). 2003 Nov;35(11):976-80.
  21. Wang RL, Yi S, Liang SP. Mechanism of action of two insect toxins huwentoxin-III and hainantoxin-VI on voltage-gated sodium channels. J Zhejiang Univ Sci B. 2010 Jun;11(6):451-7.
  22. Xiao Y, Bingham JP, Zhu W, Moczydlowski E, Liang S, Cummins TR. Tarantula huwentoxin-IV inhibits neuronal sodium channels by binding to receptor site 4 and trapping the domain ii voltage sensor in the closed configuration. J Biol Chem. 2008 Oct 3;283(40):27300-13.
  23. Peng K, Shu Q, Liu Z, Liang S. Function and solution structure of huwentoxin-IV, a potent neuronal tetrodotoxin (TTX)-sensitive sodium channel antagonist from Chinese bird spider Selenocosmia huwena. J Biol Chem. 2002 Dec 6;277(49):47564-71.
  24. Dai J, Liang SP. Purification and characterization of HWTX-VII and HWTX-VIII: two novel insecticidal neurotoxins from the Chinese bird spider Selenocosmia huwena. Chin J Biochem Mol Biol. 2003;19:71-5.