Skip to main content

EDITORIAL article

Front. Mol. Biosci., 13 May 2022
Sec. Cellular Biochemistry
Volume 9 - 2022 | https://doi.org/10.3389/fmolb.2022.924023

Editorial: Venom Peptides: A Rich Combinatorial Library for Drug Development

  • 1Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
  • 2CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
  • 3Programa de Pós Graduação em Medicina e Biomedicina, Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
  • 4Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil

Venom peptides are exceptional natural molecules in which bioactivities led to venom-derived drugs such as Captopril to treat hypertension (Ferreira et al., 1970; Cushman and Ondetti, 1999), Ziconotide to treat chronic pain (Miljanich, 2004) and Exenatide to treat type-2 diabetes (Furman, 2012). Their proved translation into new therapeutics have attracted researchers seeking molecular tools and drugs for a variety of diseases from neurological disorders to cancer, autoimmunity and infection (Cardoso, 2020). In addition, venom peptides have been studied as pesticides which led to the development of the bioinsecticide Spear comprising a spider peptide (Chong et al., 2007). In this Research Topic in Frontiers of Molecular Biosciences, we captured state-of-the-art research in venom peptides through eight original research papers and five reviews. From spider, snake, scorpion and cone snail venoms applied in venomics and structure-function studies to the pre-clinical efficacy of venom-derived peptides, we navigate through exceptional research findings in the field of venom peptides and their applications into novel pharmacological and biotechnological tools.

Modern venom peptide discovery utilizes venom gland transcriptomics to unravel venom components and their molecular diversity (Ducancel et al., 2014). In Chase et al., this approach was applied to integrate venom peptides libraries from Turridae marine snails to better understand phylogenetic relationships and to identify novel bio-actives. Their findings support the conventional polyphyletic origins of this marine snail family and describes the new genus Purpuraturris. Novel venom peptides were identified and are under consideration for studies of characterization and therapeutic potentials. In another study, Kuhn-Nentwig et al. applied transcriptomics to explore linear peptides in modern spider venoms, a neglected group of membrane active peptides with cytolytic properties contributing to venom activity. These linear peptides were identified only in Ctenidae, Lycosidae, Oxyopidae, Pisauridae, and Zodariidae out of 23 families of spiders studied. Interestingly, in Lycosidae, Oxyopidae, and in the genus Cupiennius, these peptides are highly expressed, indicating functional importance in venom activity.

Linear venom peptides from ant (Touchard et al., 2016; Schifano and Caputo, 2021), spider (Melo-Braga et al., 2020), bee (Memariani and Memariani, 2021) and wasp (Dos Santos Cabrera et al., 2019) venoms have been investigated for antimicrobial properties against superbugs. These often comprise cationic, anionic or amphipathic peptides and display strong activity to penetrate cell membranes (Yacoub et al., 2020). Reis et al. investigated an optimized antimicrobial peptide named LyeTxI-b which is a derivative of LyeTxI isolated from the spider Lycosa erythrognatha. LyeTxI-b had higher affinity for anionic lipids and superior potency against Staphylococcus aureus. From the venom of the scorpion Tityus stigmurus, Melo et al. isolated a multifunctional anionic linear peptide displaying Fe2+ and Cu2+ chelating properties with potential as novel drugs for acute and chronic intoxication, neurodegeneration, haematological and cardiovascular diseases and cancer. Wasps are rich in mastoparans, another group multifunctional linear cationic peptides with therapeutic properties, as revised in Santana et al. These unique peptides have a wide variety of biological effects, including mast cell degranulation, activation of protein G, phospholipase A2, C, and D activation, serotonin and insulin release, antimicrobial, hemolytic, and anticancer activities.

Inhibitory cystine knot (ICK) peptides are predominant in spider and cone snail venoms and display unique therapeutic potential by modulating ion channels and receptors (Cardoso and Lewi, 2018; Cardoso and Lewi, 2019; Cardoso, 2020; Cardoso et al., 2021). The spider ICK peptide PnTx-2-6 was isolated from the Brazilian wandering spider Phoneutria nigriventer as reviewed by Silva et al. In clinical cases and pre-clinical studies, PnTx-2-6 induced priapism occurring via NO/cGMP signalling and neurotoxicity induced by voltage-gated sodium (NaV) channels. An optimized version of PnTx2-6 named PnPP-19 maintained beneficial erectile properties but lost activity against NaVs and hence its side-effects. PnPP-19 was also effective in treating pain via opioid and cannabinoid systems, and glaucoma via modulation of intraocular pressure. Another spider ICK peptide is HwTx-IV isolated from the tarantula Cyriopagopus schmidti that modulates NaVs and has analgesic properties (Xiao et al., 2008; Cardoso and Lewi, 2018; Cardoso and Lewi, 2019). Lopez et al. investigated HwTx-IV properties to inhibit NaV1.7, a potential analgesic target, and NaV1.6, a defined off-target, using synthetic peptide analogues. The ratio of NaV1.7/NaV1.6 inhibition was increased when the E4K mutation was combined to R26A and Y33W, and by substituting the C-terminal amidation by a carboxylated motif. In contrast, this ratio was decreased by E4K alone, or by combined substitutions E1G/E4G or E4K/R26Q. These results demonstrated it is possible to manipulate the selectivity of venom peptides towards specific NaV subtypes to improve the potency and specificity.

Chronic pain, especially visceral pain, was shown treatable with ICK peptides inhibiting NaVs as reported for Tap1a and Tsp1a isolated from the tarantulas Theraphosa apophysis and Trixopelma sp, respectively (Cardoso et al., 2021; Jiang et al., 2021). Hu et al. evaluated the structure-function properties of Tap1a associated to potency and selectivity for NaVs, and to optimize Tap1a with greater in vivo effect. By incorporating residues from optimized NaSpTx1 peptides, optimized Tap1a peptides were designed with greater potency and selectivity for NaV subtypes in chronic pain, as well as greater in vivo effects. Preclinical efficacy in treating several neurological disorders is also demonstrated by the ICK peptide Phα1β isolated from P. nigriventer (Antunes et al., 2021; Silva et al., 2021). Diniz et al. investigated how the analgesic effects induced by Phα1β and morphine modify the central nervous system (CNS) function. Phα1β and morphine analgesic effects have a different profile in the CNS, with Phα1β selectively inhibiting activity in the unilateral motor cortex and cingulate cortex, while morphine treatment led to small and selective inhibition of the bilateral amygdala striatum and accumbens.

Venoms from Araneomorphae spiders differ from Mygalomorphaes by the predominance of large proteins with enzymatic activity that enhances the envenomation process. The therapeutic and biotechnological potential of the venom of the Araneomorphae brown spiders of the genus Loxosceles was reviewed by Gremski et al. These venoms are known to induce dermo necrosis, oedema and haemorrhage that together are defined as loxoscelism. These symptoms are induced by phospholipases D, which have now been engineered to a non-toxic form to produce antivenom or a vaccine to prevent loxoscelism. Other components of these venoms include hyaluronidases, allergen factor, and ICK peptides, amongst others, which can be used as pharmacological tools, insecticides, or drug candidates to tackle cancer and pain.

Snake venoms comprise a range of proteins and peptides that induce hemotoxic, neurotoxic, and cytotoxic effects in snakebite victims (Ferraz et al., 2019). Vasconcelos et al. reviewed the structure-function relationships of snake disintegrins which modulate cell-cell and cell-matrix interactions. In this review, disintegrins were classified into seven groups based on disulphide pattern and sequence signatures, which facilitated identification of new disintegrins. Structural signatures associated to disintegrin-integrin interactions are presented to assist the understanding of their structure-function properties. Nicotine acetylcholine receptors (nAChR) are targets of snake peptides aimed at muscles to induce paralysis (Ferraz et al., 2019). Kasheverov et al. have developed snake peptides labelled with fluorophores to probe and visualize nAChR in cells and tissues. The 3FTX α-cobratoxin and the linear peptide azemiopsin from snake venom were labelled with a small synthetic analogue of green fluorescent protein (GFP) named p-HOBDI-BF2. The introduction of the synthetic GFP preserved the peptide affinity for nAChR while the full-length GFP induced considerable loss of affinity. Although useful in the visualization of nAChR in live cells, the use of synthetic GFP-labelled snake peptides was limited in rodent tissue and in flow cytometry applications.

GPCRs are the targets of almost 30% of drugs currently on the market. This way, studies of venom peptides that can modulate their activity with the purpose of drug development is of particular importance. We end this research topic with a review by Van Baelen et al. discussing venom peptides that target GPCRs and their structure-function properties. These GPCR-targeting venom peptides were divided into two classes: endogenous related agonist-mimicking peptides, and non-endogenous related peptides with agonist, antagonist and allosteric properties. This review highlights the absence of toxicity in some of these venom peptides that display high affinity and selectivity to GPCRs that can be largely exploited for therapeutic development, as well as the importance of using ecologically relevant animal models to decipher the biological role of these venom peptides.

Author Contributions

FC wrote the editorial, DS and MdL revised the editorial.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Antunes, F. T. T., de Souza, A. H., Caminski, E. S., Greggio, S., Venturin, G. T., da Costa, J. C., et al. (2021). Neuroprotective Effects of the CTK 01512-2 Toxin against Neurotoxicity Induced by 3-nitropropionic Acid in Rats. Neurotoxicology 87, 30–42. doi:10.1016/j.neuro.2021.08.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Cardoso, F. C., Castro, J., Grundy, L., Schober, G., Garcia-Caraballo, S., Zhao, T., et al. (2021). A Spider-Venom Peptide with Multitarget Activity on Sodium and Calcium Channels Alleviates Chronic Visceral Pain in a Model of Irritable Bowel Syndrome. Pain 162 (2), 569–581. doi:10.1097/j.pain.0000000000002041

PubMed Abstract | CrossRef Full Text | Google Scholar

Cardoso, F. C., and Lewis, R. J. (2018). Sodium Channels and Pain: from Toxins to Therapies. Br. J. Pharmacol. 175 (12), 2138–2157. doi:10.1111/bph.13962

PubMed Abstract | CrossRef Full Text | Google Scholar

Cardoso, F. C., and Lewis, R. J. (2019). Structure-function and Therapeutic Potential of Spider Venom-Derived Cysteine Knot Peptides Targeting Sodium Channels. Front. Pharmacol. 10, 366. doi:10.3389/fphar.2019.00366

PubMed Abstract | CrossRef Full Text | Google Scholar

Cardoso, F. C. (2020). Multi-targeting Sodium and Calcium Channels Using Venom Peptides for the Treatment of Complex Ion Channels-Related Diseases. Biochem. Pharmacol. 181, 114107. doi:10.1016/j.bcp.2020.114107

PubMed Abstract | CrossRef Full Text | Google Scholar

Chong, Y., Hayes, J. L., Sollod, B., Wen, S., Wilson, D. T., Hains, P. G., et al. (2007). The ω-atracotoxins: Selective Blockers of Insect M-LVA and HVA Calcium Channels. Biochem. Pharmacol. 74 (4), 623–638. doi:10.1016/j.bcp.2007.05.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Cushman, D. W., and Ondetti, M. A. (1999). Design of Angiotensin Converting Enzyme Inhibitors. Nat. Med. 5 (10), 1110–1112. doi:10.1038/13423

PubMed Abstract | CrossRef Full Text | Google Scholar

Dos Santos Cabrera, M. P., Rangel, M., Ruggiero Neto, J., and Konno, K. (2019). Chemical and Biological Characteristics of Antimicrobial α-Helical Peptides Found in Solitary Wasp Venoms and Their Interactions with Model Membranes. Toxins 11 (10), 559. doi:10.3390/toxins11100559

CrossRef Full Text | Google Scholar

Ducancel, F., Durban, J., and Verdenaud, M. (2014). Transcriptomics and Venomics: Implications for Medicinal Chemistry. Future Med. Chem. 6 (15), 1629–1643. doi:10.4155/fmc.14.95

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferraz, C. R., Arrahman, A., Xie, C., Casewell, N. R., Lewis, R. J., Kool, J., et al. (2019). Multifunctional Toxins in Snake Venoms and Therapeutic Implications: From Pain to Hemorrhage and Necrosis. Front. Ecol. Evol. 7, 218. doi:10.3389/fevo.2019.00218

CrossRef Full Text | Google Scholar

Ferreira, S. H., Bartelt, D. C., and Greene, L. J. (1970). Isolation of Bradykinin-Potentiating Peptides from Bothrops jararaca Venom. Biochemistry 9 (13), 2583–2593. doi:10.1021/bi00815a005

PubMed Abstract | CrossRef Full Text | Google Scholar

Furman, B. L. (2012). The Development of Byetta (Exenatide) from the Venom of the Gila Monster as an Anti-diabetic Agent. Toxicon 59 (4), 464–471. doi:10.1016/j.toxicon.2010.12.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, Y., Castro, J., Blomster, L. V., Agwa, A. J., Maddern, J., Schober, G., et al. (2021). Pharmacological Inhibition of the Voltage-Gated Sodium Channel Nav1.7 Alleviates Chronic Visceral Pain in a Rodent Model of Irritable Bowel Syndrome. ACS Pharmacol. Transl. Sci. 4 (4), 1362–1378. doi:10.1021/acsptsci.1c00072

PubMed Abstract | CrossRef Full Text | Google Scholar

Melo-Braga, M. N., De Marco Almeida, F., Dos Santos, D. M., de Avelar Júnior, J. T., Dos Reis, P. V. M., and de Lima, M. E. (2020). Antimicrobial Peptides from Lycosidae (Sundevall, 1833) Spiders. Curr. Protein Pept. Sci. 21 (5), 527–541. doi:10.2174/1389203721666200116091911

PubMed Abstract | CrossRef Full Text | Google Scholar

Memariani, H., and Memariani, M. (2021). Melittin as a Promising Anti-protozoan Peptide: Current Knowledge and Future Prospects. Amb. Expr. 11 (1), 69. doi:10.1186/s13568-021-01229-1

CrossRef Full Text | Google Scholar

Miljanich, G. P. (2004). Ziconotide: Neuronal Calcium Channel Blocker for Treating Severe Chronic Pain. Curr. Med. Chem. 11 (23), 3029–3040. doi:10.2174/0929867043363884

PubMed Abstract | CrossRef Full Text | Google Scholar

Schifano, N. P., and Caputo, G. A. (2021). Investigation of the Role of Hydrophobic Amino Acids on the Structure-Activity Relationship in the Antimicrobial Venom Peptide Ponericin L1. J. Membr. Biol. [Epub ahead of print]. doi:10.1007/s00232-021-00204-y

CrossRef Full Text | Google Scholar

Silva, J. F. d., Binda, N. S., Pereira, E. M. R., Lavor, M. S. L. d., Vieira, L. B., Souza, A. H. d., et al. (2021). Analgesic Effects of Phα1β Toxin: a Review of Mechanisms of Action Involving Pain Pathways. J. Venom. Anim. Toxins Incl. Trop. Dis. 27, e20210001. doi:10.1590/1678-9199-JVATITD-2021-0001

PubMed Abstract | CrossRef Full Text | Google Scholar

Touchard, A., Aili, S., Fox, E., Escoubas, P., Orivel, J., Nicholson, G., et al. (2016). The Biochemical Toxin Arsenal from Ant Venoms. Toxins 8 (1), 30. doi:10.3390/toxins8010030

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, Y., Luo, X., Kuang, F., Deng, M., Wang, M., Zeng, X., et al. (2008). Synthesis and Characterization of Huwentoxin-IV, a Neurotoxin Inhibiting Central Neuronal Sodium Channels. Toxicon 51 (2), 230–239. doi:10.1016/j.toxicon.2007.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Yacoub, T., Rima, M., Karam, M., Sabatier, J.-M., and Fajloun, Z. (2020). Antimicrobials from Venomous Animals: An Overview. Molecules 25 (10), 2402. doi:10.3390/molecules25102402

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: venom peptide, structure-function activity, drug developement, pharmacological tools, novel therapeutic agents

Citation: Cardoso FC, Servent D and de Lima ME (2022) Editorial: Venom Peptides: A Rich Combinatorial Library for Drug Development. Front. Mol. Biosci. 9:924023. doi: 10.3389/fmolb.2022.924023

Received: 20 April 2022; Accepted: 27 April 2022;
Published: 13 May 2022.

Edited and reviewed by:

Cecilia Giulivi, University of California, Davis, United States

Copyright © 2022 Cardoso, Servent and de Lima. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Fernanda C. Cardoso, f.caldascardoso@uq.edu.au

Download