1
|
Lins J, Miloslavina YA, Avrutina O, Theiss F, Hofmann S, Kolmar H, Buntkowsky G. Enhancing Sensitivity of Nuclear Magnetic Resonance in Biomolecules: Parahydrogen-Induced Hyperpolarization in Synthetic Disulfide-Rich Miniproteins. J Am Chem Soc 2024; 146:35175-35184. [PMID: 39662885 DOI: 10.1021/jacs.4c11589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Hyperpolarization of small peptides by parahydrogen-induced polarization (PHIP) to increase the sensitivity of nuclear magnetic resonance (NMR) techniques is well established, while its application to larger biopolymers is still a mainly unexplored area. A particular challenge is the presence of folding-essential disulfide bridges. They tend to form metal complexes, thus hampering catalytic hydrogenation, a prerequisite for PHIP. We applied the PHIP technique to enhance NMR signal intensity in cystine-knot miniproteins─highly ordered peptide architectures covalently stabilized by three disulfides. To achieve PHIP, we introduced an l-propargyl tyrosine label at different positions in three synthetic open-chain variants of a natural trypsin inhibitor MCoTI-II. For the folded cystine knot, we observed NMR signal enhancements of up to 499 in methanol, 307 in a D2O-methanol mixture, and 964 for the cysteine-bearing reduced precursor. Trypsin inhibition assays elucidated that introducing a PHIP label into the terminal regions is preferable to alterations within the functional loop to preserve bioactivity. Substitution of the native tyrosine resulted in the highest bioactivity. A drastic reduction in PHIP enhancement was observed in the presence of trypsin due to slower hydrogenation, conditioned by the accessibility of the label within an enzyme-inhibitor complex.
Collapse
Affiliation(s)
- Jonas Lins
- Eduard-Zintl-Institut für Anorganische und Physikalische Chemie, Technische Universität Darmstadt, Peter-Grünberg-Straße 8, 64287 Darmstadt, Germany
| | - Yuliya A Miloslavina
- Eduard-Zintl-Institut für Anorganische und Physikalische Chemie, Technische Universität Darmstadt, Peter-Grünberg-Straße 8, 64287 Darmstadt, Germany
| | - Olga Avrutina
- Clemens-Schöpf-Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Peter-Grünberg-Straße 4, 64287 Darmstadt, Germany
| | - Franziska Theiss
- Eduard-Zintl-Institut für Anorganische und Physikalische Chemie, Technische Universität Darmstadt, Peter-Grünberg-Straße 8, 64287 Darmstadt, Germany
| | - Sarah Hofmann
- Clemens-Schöpf-Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Peter-Grünberg-Straße 4, 64287 Darmstadt, Germany
| | - Harald Kolmar
- Clemens-Schöpf-Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Peter-Grünberg-Straße 4, 64287 Darmstadt, Germany
| | - Gerd Buntkowsky
- Eduard-Zintl-Institut für Anorganische und Physikalische Chemie, Technische Universität Darmstadt, Peter-Grünberg-Straße 8, 64287 Darmstadt, Germany
| |
Collapse
|
2
|
Dresler J, Herzig V, Vilcinskas A, Lüddecke T. Enlightening the toxinological dark matter of spider venom enzymes. NPJ BIODIVERSITY 2024; 3:25. [PMID: 39271930 PMCID: PMC11399385 DOI: 10.1038/s44185-024-00058-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024]
Abstract
Spiders produce highly adapted venoms featuring a complex mixture of biomolecules used mainly for hunting and defense. The most prominent components are peptidic neurotoxins, a major focus of research and drug development, whereas venom enzymes have been largely neglected. Nevertheless, investigation of venom enzymes not only reveals insights into their biological functions, but also provides templates for future industrial applications. Here we compared spider venom enzymes validated at protein level contained in the VenomZone database and from all publicly available proteo-transcriptomic spider venom datasets. We assigned reported enzymes to cellular processes and known venom functions, including toxicity, prey pre-digestion, venom preservation, venom component activation, and spreading factors. Our study unveiled extensive discrepancy between public databases and publications with regard to enzyme coverage, which impedes the development of novel spider venom enzyme-based applications. Uncovering the previously unrecognized abundance and diversity of venom enzymes will open new avenues for spider venom biodiscovery.
Collapse
Affiliation(s)
- Josephine Dresler
- Animal Venomics Lab, Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Gießen, Germany.
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt a. M., Germany.
| | - Volker Herzig
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia
| | - Andreas Vilcinskas
- Animal Venomics Lab, Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Gießen, Germany
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt a. M., Germany
- Institute for Insect Biotechnology, Justus-Liebig-University of Giessen, Gießen, Germany
| | - Tim Lüddecke
- Animal Venomics Lab, Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Gießen, Germany.
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt a. M., Germany.
| |
Collapse
|
3
|
Walker AA, Chin YKY, Guo S, Jin J, Wilbrink E, Goudarzi MH, Wirth H, Gordon E, Weirauch C, King GF. Structure and bioactivity of an insecticidal trans-defensin from assassin bug venom. Structure 2024; 32:1348-1357.e4. [PMID: 38889720 DOI: 10.1016/j.str.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/30/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024]
Abstract
Disulfide-rich peptides such as defensins play diverse roles in immunity and ion channel modulation, as well as constituting the bioactive components of many animal venoms. We investigated the structure and bioactivity of U-RDTX-Pp19, a peptide previously discovered in venom of the assassin bug Pristhesancus plagipennis. Recombinant Pp19 (rPp19) was found to possess insecticidal activity when injected into Drosophila melanogaster. A bioinformatic search revealed that domains homologous to Pp19 are produced by assassin bugs and diverse other arthropods. rPp19 co-eluted with native Pp19 isolated from P. plagipennis, which we found is more abundant in hemolymph than venom. We solved the three-dimensional structure of rPp19 using 2D 1H NMR spectroscopy, finding that it adopts a disulfide-stabilized structure highly similar to known trans-defensins, with the same cystine connectivity as human α-defensin (I-VI, II-IV, and III-V). The structure of Pp19 is unique among reported structures of arthropod peptides.
Collapse
Affiliation(s)
- Andrew A Walker
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; Centre of Excellence for Innovations in Protein and Peptide Science, St Lucia, QLD 4072, Australia.
| | - Yanni K-Y Chin
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Shaodong Guo
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Jiayi Jin
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Evienne Wilbrink
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; Fontys University of Applied Sciences, Eindhoven 5612 AR, the Netherlands
| | - Mohaddeseh Hedayati Goudarzi
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; Centre of Excellence for Innovations in Protein and Peptide Science, St Lucia, QLD 4072, Australia
| | - Hayden Wirth
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; Centre of Excellence for Innovations in Protein and Peptide Science, St Lucia, QLD 4072, Australia
| | - Eric Gordon
- Department of Entomology, University of California Riverside, Riverside, CA 92521, USA
| | - Christiane Weirauch
- Department of Entomology, University of California Riverside, Riverside, CA 92521, USA
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; Centre of Excellence for Innovations in Protein and Peptide Science, St Lucia, QLD 4072, Australia.
| |
Collapse
|
4
|
Marone Fassolo E, Guo S, Wang Y, Rosa S, Herzig V. Genetically encoded libraries and spider venoms as emerging sources for crop protective peptides. J Pept Sci 2024; 30:e3600. [PMID: 38623834 DOI: 10.1002/psc.3600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/17/2024]
Abstract
Agricultural crops are targeted by various pathogens (fungi, bacteria, and viruses) and pests (herbivorous arthropods). Antimicrobial and insecticidal peptides are increasingly recognized as eco-friendly tools for crop protection due to their low propensity for resistance development and the fact that they are fully biodegradable. However, historical challenges have hindered their development, including poor stability, limited availability, reproducibility issues, high production costs, and unwanted toxicity. Toxicity is a primary concern because crop-protective peptides interact with various organisms of environmental and economic significance. This review focuses on the potential of genetically encoded peptide libraries like the use of two-hybrid-based methods for antimicrobial peptides identification and insecticidal spider venom peptides as two main approaches for targeting plant pathogens and pests. We discuss some key findings and challenges regarding the practical application of each strategy. We conclude that genetically encoded peptide library- and spider venom-derived crop protective peptides offer a sustainable and environmentally responsible approach for addressing modern crop protection needs in the agricultural sector.
Collapse
Affiliation(s)
| | - Shaodong Guo
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Yachen Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Stefano Rosa
- Department of Biosciences, University of Milan, Milan, Italy
- Department of Chemistry & Chemical Biology, Northeastern University, Boston, Massachusetts, USA
| | - Volker Herzig
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| |
Collapse
|
5
|
Jiménez J, Kemmerer M, King GF, Polston JE, Bonning BC. Coat protein of a whitefly-vectored plant virus as a delivery system to target whitefly. Microb Biotechnol 2024; 17:e14468. [PMID: 38635158 PMCID: PMC11025618 DOI: 10.1111/1751-7915.14468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
The sweet potato whitefly Bemisia tabaci (Hemiptera: Aleyrodidae) is responsible for significant crop losses and presents one of the greatest challenges for global agricultural pest management. Management of whitefly populations and associated plant viral diseases is hindered by widespread whitefly resistance to chemical insecticides. An alternative control approach involves the use of insect-specific neurotoxins, but these require delivery from the whitefly gut into the haemocoel. Here we demonstrate that the coat protein (CP) of a begomovirus, Tomato yellow leaf curl virus, is sufficient for delivery of fused proteins into the whitefly haemocoel without virion assembly. Following feeding on the recombinant CP-P-mCherry fusion (where -P- is a proline-rich linker), mCherry fluorescence was detected in the dorsal aorta and pericardial cells of the whitefly, but not in those of whitefly fed on negative control treatments, indicating effective CP-mediated delivery of mCherry into the whitefly haemocoel. Significant mortality was observed in whiteflies fed on a fusion of CP-P to the insect-specific neurotoxin Hv1a, but not in whiteflies fed on CP-P fused to a disarmed Hv1a mutant. Begomovirus coat protein - insect neurotoxin fusions hold considerable potential for transgenic resistance to whitefly providing valuable tools for whitefly management.
Collapse
Affiliation(s)
- Jaime Jiménez
- Department of Entomology and NematologyUniversity of FloridaGainesvilleFloridaUSA
- Present address:
Instituto de Ciencias Agrarias – Consejo Superior de Investigaciones Científicas (ICA‐CSIC)MadridSpain
| | - Mariah Kemmerer
- Department of Entomology and NematologyUniversity of FloridaGainesvilleFloridaUSA
- Present address:
Department of Biological SciencesUniversity of DelawareNewarkDelawareUSA
| | - Glenn F. King
- Centre for Future Medicines, Institute for Molecular BioscienceThe University of QueenslandBrisbaneQueenslandAustralia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQueenslandAustralia
| | - Jane E. Polston
- Department of Plant PathologyUniversity of FloridaGainesvilleFloridaUSA
| | - Bryony C. Bonning
- Department of Entomology and NematologyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
6
|
Gao X, Kaluarachchi H, Zhang Y, Hwang S, Hannoush RN. A phage-displayed disulfide constrained peptide discovery platform yields novel human plasma protein binders. PLoS One 2024; 19:e0299804. [PMID: 38547072 PMCID: PMC10977726 DOI: 10.1371/journal.pone.0299804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/15/2024] [Indexed: 04/02/2024] Open
Abstract
Disulfide constrained peptides (DCPs) show great potential as templates for drug discovery. They are characterized by conserved cysteine residues that form intramolecular disulfide bonds. Taking advantage of phage display technology, we designed and generated twenty-six DCP phage libraries with enriched molecular diversity to enable the discovery of ligands against disease-causing proteins of interest. The libraries were designed based on five DCP scaffolds, namely Momordica charantia 1 (Mch1), gurmarin, Asteropsin-A, antimicrobial peptide-1 (AMP-1), and potato carboxypeptidase inhibitor (CPI). We also report optimized workflows for screening and producing synthetic and recombinant DCPs. Examples of novel DCP binders identified against various protein targets are presented, including human IgG Fc, serum albumin, vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor (PDGF). We identified DCPs against human IgG Fc and serum albumin with sub-micromolar affinity from primary panning campaigns, providing alternative tools for potential half-life extension of peptides and small protein therapeutics. Overall, the molecular diversity of the DCP scaffolds included in the designed libraries, coupled with their distinct biochemical and biophysical properties, enables efficient and robust identification of de novo binders to drug targets of therapeutic relevance.
Collapse
Affiliation(s)
- Xinxin Gao
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Peptide Therapeutics, Genentech, South San Francisco, California, United States of America
| | - Harini Kaluarachchi
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
| | - Yingnan Zhang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Biological Chemistry, Genentech, South San Francisco, California, United States of America
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Peptide Therapeutics, Genentech, South San Francisco, California, United States of America
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
| |
Collapse
|
7
|
Wang Y, Guo S, Ventura T, Jain R, Robinson KE, Mitter N, Herzig V. Development of a soybean leaf disc assay for determining oral insecticidal activity in the lepidopteran agricultural pest Helicoverpa armigera. Toxicon 2024; 238:107588. [PMID: 38147939 DOI: 10.1016/j.toxicon.2023.107588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 12/28/2023]
Abstract
Pest insects pose a heavy burden on global agricultural industries with small molecule insecticides being predominantly used for their control. Unwanted side effects and resistance development plagues most small molecule insecticides such as the neonicotinoids, which have been reported to be harmful to honeybees. Bioinsecticides like Bacillus thuringiensis (Bt) toxins can be used as environmentally-friendly alternatives. Arachnid venoms comprise another promising source of bioinsecticides, containing a multitude of selective and potent insecticidal toxins. Unfortunately, no standardised insect models are currently available to assess the suitability of insecticidal agents under laboratory conditions. Thus, we aimed to develop a laboratory model that closely mimics field conditions by employing a leaf disk assay (LDA) for oral application of insecticidal agents in a bioassay tray format. Neonate larvae of the cotton bollworm (Helicoverpa armigera) were fed with soybean (Glycine max) leaves that were treated with different insecticidal agents. We observed dose-dependent insecticidal effects for Bt toxin and the neonicotinoid insecticide imidacloprid, with imidacloprid exhibiting a faster response. Furthermore, we identified several insecticidal arachnid venoms that were active when co-applied with sub-lethal doses of Bt toxin. We propose the H. armigera LDA as a suitable tool for assessing the insecticidal effects of insecticidal agents against lepidopterans.
Collapse
Affiliation(s)
- Yachen Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia
| | - Shaodong Guo
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia
| | - Tomer Ventura
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia
| | - Ritesh Jain
- Centre for Horticultural Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Karl E Robinson
- Centre for Horticultural Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Neena Mitter
- Centre for Horticultural Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Volker Herzig
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia.
| |
Collapse
|
8
|
Hernandez Duran L, Wilson DT, Rymer TL. Exploring behavioral traits over different contexts in four species of Australian funnel-web spiders. Curr Zool 2023; 69:766-774. [PMID: 37876639 PMCID: PMC10591153 DOI: 10.1093/cz/zoac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 10/04/2022] [Indexed: 10/26/2023] Open
Abstract
Australian funnel-web spiders are arguably the most venomous spiders in the world, with much research focusing on this aspect of their biology. However, other aspects related to their life history, ecology and behaviour have been overlooked. For the first time, we assessed repeatability, namely risk-taking behaviour, aggressiveness and activity in the contexts of predation, conspecific tolerance and exploration of a new territory in four species of Australian funnel-web spiders: two are closely related, Hadronyche valida and H. infensa, and two have overlapping distributions but occupy different habitats, H. cerberea and Atrax robustus. We also compared behaviors between species. At the species level, we found that H. valida showed consistency in risk-taking behavior when exposed to a predator stimulus, aggressiveness against conspecifics, and exploration of a new territory. In contrast, in the other species, only A. robustus showed repeatability in the context of exploration of a new territory. These results suggest that some behavioral traits are likely more flexible than others, and that the repeatability of behaviors may be species-specific in funnel-webs. When we compared species, we found differences in risk-taking behavior and defensiveness. This study provides novel insights to understanding variation in behavioral traits within and between species of funnel-web spiders, suggesting that some behavioral traits are likely context and/or species dependent, as a result of their evolutionary history. These findings provide key insights for understanding the ecological role of behavior and venom deployment in venomous animals, and a greater understanding of behavior in these medically significant and iconic spiders that are of conservation concern.
Collapse
Affiliation(s)
- Linda Hernandez Duran
- College of Science and Engineering, James Cook University, P.O. Box 6811, Cairns, QLD 4870, Australia
- Centre for Tropical Environmental and Sustainability Sciences, James Cook University, Cairns, QLD 4870, Australia
| | - David Thomas Wilson
- Centre for Molecular Therapeutics, Australian Institute for Tropical Health and Medicine, James Cook University, Cairns, QLD 4878, Australia
| | - Tasmin Lee Rymer
- College of Science and Engineering, James Cook University, P.O. Box 6811, Cairns, QLD 4870, Australia
- Centre for Tropical Environmental and Sustainability Sciences, James Cook University, Cairns, QLD 4870, Australia
| |
Collapse
|
9
|
Oparin PB, Nikodimov SS, Vassilevski AA. Venoms with oral toxicity towards insects. Toxicon 2023; 235:107308. [PMID: 37797725 DOI: 10.1016/j.toxicon.2023.107308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/07/2023]
Abstract
Animal venoms are a promising source of potential bioinsecticides. To find hits with pronounced oral insect toxicity, we screened 82 venoms using Achroia grisella (Lepidoptera) and Tenebrio molitor (Coleoptera) larvae, and adult Drosophila melanogaster (Diptera). We also injected the most potent venoms in adult D. melanogaster to compare their efficiency in different routes of administration. 18 venoms from spiders and snakes show high oral toxicity and can be further exploited to isolate new insecticides.
Collapse
Affiliation(s)
- Peter B Oparin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya, Moscow, 117997, Russia
| | - Sergei S Nikodimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya, Moscow, 117997, Russia
| | - Alexander A Vassilevski
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya, Moscow, 117997, Russia.
| |
Collapse
|
10
|
Lyons K, Dugon MM, Boyd A, Healy K. Venom extraction method influences venom composition and potency in the giant house spider Eratigena atrica (C. L. Koch, 1843). Toxicon 2023; 234:107303. [PMID: 37775046 DOI: 10.1016/j.toxicon.2023.107303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Extraction is the first step when investigating venom composition and function. In small invertebrates, widely used extraction methods include electrostimulation and venom gland extraction, however, the influence of these methods on composition and toxicology is poorly understood. Using the Giant House Spider Eratigena atrica as a model, we show that electrostimulation and venom gland removal extraction methods produce different protein profiles as assessed by Coomassie-stained SDS-PAGE and significantly different potencies in the cricket Acheta domesticus.
Collapse
Affiliation(s)
- Keith Lyons
- Macroecology Lab, School of Natural Sciences, Ryan Institute, University of Galway, H91 TK33, Galway, Ireland.
| | - Michel M Dugon
- Venom Systems & Proteomics Lab, School of Natural Sciences, Ryan Institute, University of Galway, H91 TK33, Galway, Ireland
| | - Aoife Boyd
- Pathogenic Mechanisms Research Group, School of Natural Sciences, Ryan Institute, University of Galway, H91 TK33, Galway, Ireland
| | - Kevin Healy
- Macroecology Lab, School of Natural Sciences, Ryan Institute, University of Galway, H91 TK33, Galway, Ireland
| |
Collapse
|
11
|
de Jesus-López E, Cuéllar-Balleza L, Díaz-Peña LF, Luna-Vázquez FJ, Ibarra-Alvarado C, García-Arredondo JA. Vasodilator activity of Poecilotheria ornata venom involves activation of the NO/cGMP pathway and inhibition of calcium influx to vascular smooth muscle cells. Toxicon X 2023; 19:100159. [PMID: 37251689 PMCID: PMC10220391 DOI: 10.1016/j.toxcx.2023.100159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/01/2023] [Accepted: 05/15/2023] [Indexed: 05/31/2023] Open
Abstract
Tarantula venoms may be a natural source of new vasodilator components useful in pharmacological research. Moreover, biological function data of the venoms are important to enhance the knowledge about the biodiversity and evolution of these species. The present study aims to describe the vasodilatory activity induced by the venom of Poecilotheria ornata on isolated rat aortic rings. This venom induced a vasodilator activity that was significantly reduced after incubation with L-NAME or ODQ. Measurements of nitrite concentrations on rat aorta homogenates showed that the venom significantly increased the basal levels. Moreover, the venom attenuates the contraction induced by calcium. These results suggest that P. ornata venom contains a mixture of vasodilator components that act through the activation of the nitric oxide/cGMP pathway, as well as, through an endothelium-independent mechanism that involves the calcium influx into vascular smooth muscle cells.
Collapse
Affiliation(s)
- Enrique de Jesus-López
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario S/N, 76010, Querétaro, Mexico
| | - Luis Cuéllar-Balleza
- Aracnario, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. de las Ciencias S/N, 76230, Juriquilla, Querétaro, Mexico
| | - Luis Fernando Díaz-Peña
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario S/N, 76010, Querétaro, Mexico
| | - Francisco Javier Luna-Vázquez
- Departamento de Investigación Química y Farmacológica de Productos Naturales, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario S/N, 76010, Querétaro, Mexico
| | - César Ibarra-Alvarado
- Departamento de Investigación Química y Farmacológica de Productos Naturales, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario S/N, 76010, Querétaro, Mexico
| | - José Alejandro García-Arredondo
- Departamento de Investigación Química y Farmacológica de Productos Naturales, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario S/N, 76010, Querétaro, Mexico
| |
Collapse
|
12
|
Tran P, Crawford T, Ragnarsson L, Deuis JR, Mobli M, Sharpe SJ, Schroeder CI, Vetter I. Structural Conformation and Activity of Spider-Derived Inhibitory Cystine Knot Peptide Pn3a Are Modulated by pH. ACS OMEGA 2023; 8:26276-26286. [PMID: 37521635 PMCID: PMC10373202 DOI: 10.1021/acsomega.3c02664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023]
Abstract
Numerous spider venom-derived gating modifier toxins exhibit conformational heterogeneity during purification by reversed-phase high-performance liquid chromatography (RP-HPLC). This conformational exchange is especially peculiar for peptides containing an inhibitor cystine knot motif, which confers excellent structural stability under conditions that are not conducive to disulfide shuffling. This phenomenon is often attributed to proline cis/trans isomerization but has also been observed in peptides that do not contain a proline residue. Pn3a is one such peptide forming two chromatographically distinguishable peaks that readily interconvert following the purification of either conformer. The nature of this exchange was previously uncharacterized due to the fast rate of conversion in solution, making isolation of the conformers impossible. In the present study, an N-terminal modification of Pn3a enabled the isolation of the individual conformers, allowing activity assays to be conducted on the individual conformers using electrophysiology. The conformers were analyzed separately by nuclear magnetic resonance spectroscopy (NMR) to study their structural differences. RP-HPLC and NMR were used to study the mechanism of exchange. The later-eluting conformer was the active conformer with a rigid structure that corresponds to the published structure of Pn3a, while NMR analysis revealed the earlier-eluting conformer to be inactive and disordered. The exchange was found to be pH-dependent, arising in acidic solutions, possibly due to reversible disruption and formation of intramolecular salt bridges. This study reveals the nature of non-proline conformational exchange observed in Pn3a and possibly other disulfide-rich peptides, highlighting that the structure and activity of some disulfide-stabilized peptides can be dramatically susceptible to disruption.
Collapse
Affiliation(s)
- Poanna Tran
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
| | - Theo Crawford
- Centre
for Advanced Imaging, The University of
Queensland, Brisbane, Queensland 4072, Australia
| | - Lotten Ragnarsson
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer R. Deuis
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
| | - Mehdi Mobli
- Centre
for Advanced Imaging, The University of
Queensland, Brisbane, Queensland 4072, Australia
| | - Simon J. Sharpe
- Molecular
Medicine Program, Research Institute, The
Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department
of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Christina I. Schroeder
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
- Center
for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702-1201, United States
- Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Irina Vetter
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4102, Australia
| |
Collapse
|
13
|
Ahmed J, Walker AA, Perdomo HD, Guo S, Nixon SA, Vetter I, Okoh HI, Shehu DM, Shuaibu MN, Ndams IS, King GF, Herzig V. Two Novel Mosquitocidal Peptides Isolated from the Venom of the Bahia Scarlet Tarantula ( Lasiodora klugi). Toxins (Basel) 2023; 15:418. [PMID: 37505687 PMCID: PMC10467143 DOI: 10.3390/toxins15070418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/16/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
Effective control of diseases transmitted by Aedes aegypti is primarily achieved through vector control by chemical insecticides. However, the emergence of insecticide resistance in A. aegypti undermines current control efforts. Arachnid venoms are rich in toxins with activity against dipteran insects and we therefore employed a panel of 41 spider and 9 scorpion venoms to screen for mosquitocidal toxins. Using an assay-guided fractionation approach, we isolated two peptides from the venom of the tarantula Lasiodora klugi with activity against adult A. aegypti. The isolated peptides were named U-TRTX-Lk1a and U-TRTX-Lk2a and comprised 41 and 49 residues with monoisotopic masses of 4687.02 Da and 5718.88 Da, respectively. U-TRTX-Lk1a exhibited an LD50 of 38.3 pmol/g when injected into A. aegypti and its modeled structure conformed to the inhibitor cystine knot motif. U-TRTX-Lk2a has an LD50 of 45.4 pmol/g against adult A. aegypti and its predicted structure conforms to the disulfide-directed β-hairpin motif. These spider-venom peptides represent potential leads for the development of novel control agents for A. aegypti.
Collapse
Affiliation(s)
- Jamila Ahmed
- Department of Zoology, Ahmadu Bello University Zaria, Kaduna 810107, Nigeria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Andrew A. Walker
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, University of Queensland, Brisbane, QLD 4072, Australia
| | - Hugo D. Perdomo
- School of Biological Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Shaodong Guo
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, University of Queensland, Brisbane, QLD 4072, Australia
| | - Samantha A. Nixon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, University of Queensland, Brisbane, QLD 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Hilary I. Okoh
- Department of Animal and Environmental Biology, Federal University Oye-Ekiti, Oye 371104, Nigeria
| | - Dalhatu M. Shehu
- Department of Zoology, Ahmadu Bello University Zaria, Kaduna 810107, Nigeria
| | - Mohammed N. Shuaibu
- Department of Biochemistry, Ahmadu Bello University Zaria, Kaduna 810107, Nigeria
- Centre for Biotechnology Research and Training, Ahmadu Bello University Zaria, Kaduna 810107, Nigeria
| | - Iliya S. Ndams
- Department of Zoology, Ahmadu Bello University Zaria, Kaduna 810107, Nigeria
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, University of Queensland, Brisbane, QLD 4072, Australia
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
- School of Science, Technology, and Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
| |
Collapse
|
14
|
A subfraction obtained from the venom of the tarantula Poecilotheria regalis contains inhibitor cystine knot peptides and induces relaxation of rat aorta by inhibiting L-type voltage-gated calcium channels. Toxicon X 2023; 18:100151. [PMID: 36873112 PMCID: PMC9978846 DOI: 10.1016/j.toxcx.2023.100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
Venoms from tarantulas contain low molecular weight vasodilatory compounds whose biological action is conceived as part of the envenomation strategy due to its propagative effects. However, some properties of venom-induced vasodilation do not match those described by such compounds, suggesting that other toxins may cooperate with these ones to produce the observed biological effect. Owing to the distribution and function of voltage-gated ion channels in blood vessels, disulfide-rich peptides isolated from venoms of tarantulas could be conceived into potential vasodilatory compounds. However, only two peptides isolated from spider venoms have been investigated so far. This study describes for the first time a subfraction containing inhibitor cystine knot peptides, PrFr-I, obtained from the venom of the tarantula Poecilotheria regalis. This subfraction induced sustained vasodilation in rat aortic rings independent of vascular endothelium and endothelial ion channels. Furthermore, PrFr-I decreased calcium-induced contraction of rat aortic segments and reduced extracellular calcium influx to chromaffin cells by the blockade of L-type voltage-gated calcium channels. This mechanism was unrelated to the activation of potassium channels from vascular smooth muscle, since vasodilation was not affected in the presence of TEA, and PrFr-I did not modify the conductance of the voltage-gated potassium channel Kv10.1. This work proposes a new envenomating function of peptides from venoms of tarantulas, and establishes a new mechanism for venom-induced vasodilation.
Collapse
Key Words
- ACh, acetylcholine
- ADP, adenosine diphosphate
- Cav, voltage-gated calcium channel
- DMEM, Dulbecco's modified eagle's medium
- DRP, disulfide-rich peptide
- EC50, half maximal effective concentration
- Emax, maximum effect
- FBS, fetal bovine serum
- HPLC, high-performance liquid chromatography
- ICK peptide
- ICK, inhibitor cystine knot
- IKCa, intermediate conductance calcium-activated potassium channel
- Kv, voltage-gated potassium channel
- L-type calcium voltage-gated calcium
- LC-MS/MS, liquid chromatography-tandem mass spectrometer
- MALDI, matrix-assisted desorption ionization
- Nav, voltage-gated sodium channel
- Phe, phenylephrine
- Poecilotheria regalis, venom
- RP, reverse phase
- Rat aorta
- SEM, standard error of the mean
- SKCa, small conductance calcium-activated potassium channel
- SNP, sodium nitroprusside
- TEA, tetraethylammonium
- TOF, time of flight
- TRP, transient receptor potential
- Vasodilation
- Vh, holding-voltage
- endothelium-intact (E+), endothelium-denuded (E−)
Collapse
|
15
|
Qin C, Yang X, Zhang Y, Deng G, Huang X, Zuo Z, Sun F, Cao Z, Chen Z, Wu Y. Functional Characterization of a New Degradation Peptide BmTX4-P1 from Traditional Chinese Scorpion Medicinal Material. Toxins (Basel) 2023; 15:toxins15050340. [PMID: 37235373 DOI: 10.3390/toxins15050340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Thermally processed Buthus martensii Karsch scorpion is an important traditional Chinese medical material that has been widely used to treat various diseases in China for over one thousand years. Our recent work showed that thermally processed Buthus martensii Karsch scorpions contain many degraded peptides; however, the pharmacological activities of these peptides remain to be studied. Here, a new degraded peptide, BmTX4-P1, was identified from processed Buthus martensii Karsch scorpions. Compared with the venom-derived wild-type toxin peptide BmTX4, BmTX4-P1 missed some amino acids at the N-terminal and C-terminal regions, while containing six conserved cysteine residues, which could be used to form disulfide bond-stabilized α-helical and β-sheet motifs. Two methods (chemical synthesis and recombinant expression) were used to obtain the BmTX4-P1 peptide, named sBmTX4-P1 and rBmTX4-P1. Electrophysiological experimental results showed that sBmTX4-P1 and rBmTX4-P1 exhibited similar activities to inhibit the currents of hKv1.2 and hKv1.3 channels. In addition, the experimental electrophysiological results of recombinant mutant peptides of BmTX4-P1 indicated that the two residues of BmTX4-P1 (Lys22 and Tyr31) were the key residues for its potassium channel inhibitory activity. In addition to identifying a new degraded peptide, BmTX4-P1, from traditional Chinese scorpion medicinal material with high inhibitory activities against the hKv1.2 and hKv1.3 channels, this study also provided a useful method to obtain the detailed degraded peptides from processed Buthus martensii Karsch scorpions. Thus, the study laid a solid foundation for further research on the medicinal function of these degraded peptides.
Collapse
Affiliation(s)
- Chenhu Qin
- College of Life Sciences, Wuhan University, Wuhan 430072, China
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hubei University of Medicine, Shiyan 442000, China
| | - Xuhua Yang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yuanyuan Zhang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Gang Deng
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xin Huang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zheng Zuo
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Fang Sun
- College of Life Sciences, Wuhan University, Wuhan 430072, China
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hubei University of Medicine, Shiyan 442000, China
| | - Zhijian Cao
- College of Life Sciences, Wuhan University, Wuhan 430072, China
- Center for BioDrug Research, Wuhan University, Wuhan 430072, China
| | - Zongyun Chen
- College of Life Sciences, Wuhan University, Wuhan 430072, China
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hubei University of Medicine, Shiyan 442000, China
| | - Yingliang Wu
- College of Life Sciences, Wuhan University, Wuhan 430072, China
- Center for BioDrug Research, Wuhan University, Wuhan 430072, China
| |
Collapse
|
16
|
Sjakste N, Gajski G. A Review on Genotoxic and Genoprotective Effects of Biologically Active Compounds of Animal Origin. Toxins (Basel) 2023; 15:165. [PMID: 36828477 PMCID: PMC9961038 DOI: 10.3390/toxins15020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Envenomation by animal venoms remains a serious medical and social problem, especially in tropical countries. On the other hand, animal venoms are widely used as a source of biologically active compounds for the development of novel drugs. Numerous derivatives of animal venoms are already used in clinical practice. When analysing the mechanisms of action of animal venoms, attention is usually focused on the main target of the venom's enzymes and peptides such as neurotoxic, cytotoxic or haemorrhagic effects. In the present review, we would like to draw attention to the "hidden" effects of animal venoms and their derivatives in regard to DNA damage and/or protection against DNA damage. Alkaloids and terpenoids isolated from sponges such as avarol, ingenamine G or variolin B manifest the capability to bind DNA in vitro and produce DNA breaks. Trabectidin, isolated from a sea squirt, also binds and damages DNA. A similar action is possible for peptides isolated from bee and wasp venoms such as mastoparan, melectin and melittin. However, DNA lesions produced by the crude venoms of jellyfish, scorpions, spiders and snakes arise as a consequence of cell membrane damage and the subsequent oxidative stress, whereas certain animal venoms or their components produce a genoprotective effect. Current research data point to the possibility of using animal venoms and their components in the development of various potential therapeutic agents; however, before their possible clinical use the route of injection, molecular target, mechanism of action, exact dosage, possible side effects and other fundamental parameters should be further investigated.
Collapse
Affiliation(s)
- Nikolajs Sjakste
- Department of Medical Biochemistry, Faculty of Medicine, University of Latvia, 1004 Riga, Latvia
- Genetics and Bioinformatics, Institute of Biology, University of Latvia, 1004 Riga, Latvia
| | - Goran Gajski
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| |
Collapse
|
17
|
Sukiran NA, Pyati P, Willis CE, Brown AP, Readshaw JJ, Fitches EC. Enhancing the oral and topical insecticidal efficacy of a commercialized spider venom peptide biopesticide via fusion to the carrier snowdrop lectin (Galanthus nivalis agglutinin). PEST MANAGEMENT SCIENCE 2023; 79:284-294. [PMID: 36161468 PMCID: PMC10091797 DOI: 10.1002/ps.7198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/23/2022] [Accepted: 09/26/2022] [Indexed: 05/30/2023]
Abstract
BACKGROUND Spear®-T sold as a contact foliar spray for the control of glasshouse pests such as aphids, thrips, spider mites and whiteflies, contains the recombinant spider venom peptide GS-ω/κ-HxTx-Hv1h (named as GS-ω/κ-HxTx-Hv1a by Vestaron) as the active ingredient. Here we investigate whether fusion of the peptide to snowdrop lectin, (Galanthus nivalis agglutinin; GNA) enhances the efficacy of this venom peptide towards aphid pests. RESULTS Recombinant GS-ω/κ-HxTx-Hv1h (HxTx-Hv1h) and an HxTx-Hv1h/GNA fusion protein were produced using the yeast Pichia pastoris. Purified proteins showed comparable toxicity when injected into lepidopteran (Mamestra brassicae) larvae, but significant differences in oral and contact activity towards aphids. HxTx-Hv1h had comparable acute oral toxicity to pea (Acyrthosiphon pisum) and peach potato (Myzus persicae) aphids with respective Day (2) median lethal concentration (LC50 ) values of 111 and 108 μm derived from diet assays. The fusion protein also showed comparable oral toxicity to both species but D2 LC50 values were >3-fold lower (35 and 33 μm for pea and peach potato aphids, respectively) as compared to HxTx-Hv1h. Topically applied toxin and fusion protein, but not GNA, caused significant reductions in pea aphid survival. Contact effects on mortality were significantly greater for aphids exposed to fusion protein as compared to toxin alone. Whole aphid fluorescence microscopy and immunoblotting suggest that improved efficacy is due to enhanced persistence of HxTx-Hv1h when fused to GNA following internalisation of ingested or topically applied proteins. CONCLUSIONS This is the first study to report on the insecticidal activity of HxTx-Hv1h towards aphids and results suggest that a fusion protein-based approach offers opportunities to significantly enhance oral and contact efficacy of naturally derived toxins, such as HxTx-Hv1h, towards crop pests. © 2022 The Authors. Pest Management Science published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
| | - Prashant Pyati
- School of BiosciencesUniversity of DurhamDurhamUK
- Ajeet Seeds Pvt. Ltd.Plant Biotechnology Research CentreAurangabadIndia
| | - Caitlin E Willis
- School of BiosciencesUniversity of DurhamDurhamUK
- Biointeractions & Crop Protection DepartmentRothamstead ResearchHarpendenUK
| | | | | | | |
Collapse
|
18
|
Michálek O, Walker AA, Šedo O, Zdráhal Z, King GF, Pekár S. Composition and toxicity of venom produced by araneophagous white-tailed spiders (Lamponidae: Lampona sp.). Sci Rep 2022; 12:21597. [PMID: 36517485 PMCID: PMC9751281 DOI: 10.1038/s41598-022-24694-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
Prey-specialised spiders are adapted to capture specific prey items, including dangerous prey. The venoms of specialists are often prey-specific and less complex than those of generalists, but their venom composition has not been studied in detail. Here, we investigated the venom of the prey-specialised white-tailed spiders (Lamponidae: Lampona), which utilise specialised morphological and behavioural adaptations to capture spider prey. We analysed the venom composition using proteo-transcriptomics and taxon-specific toxicity using venom bioassays. Our analysis identified 208 putative toxin sequences, comprising 103 peptides < 10 kDa and 105 proteins > 10 kDa. Most peptides belonged to one of two families characterised by scaffolds containing eight or ten cysteine residues. Toxin-like proteins showed similarity to galectins, leucine-rich repeat proteins, trypsins and neprilysins. The venom of Lampona was shown to be more potent against the preferred spider prey than against alternative cricket prey. In contrast, the venom of a related generalist was similarly potent against both prey types. These data provide insights into the molecular adaptations of venoms produced by prey-specialised spiders.
Collapse
Affiliation(s)
- Ondřej Michálek
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic.
| | - Andrew A Walker
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Ondrej Šedo
- Research Group Proteomics, Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Zbyněk Zdráhal
- Research Group Proteomics, Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Stano Pekár
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic
| |
Collapse
|
19
|
Conformations of disulfides are conserved in inhibitory cystine knot (ICK) motif polypeptides. Toxicon 2022; 219:106926. [PMID: 36167143 DOI: 10.1016/j.toxicon.2022.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/24/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022]
Abstract
The inhibitory cystine knot (ICK) motif is an evolutionarily optimized disulfide-rich peptide motif widely present in diverse phyla with distinct biological functions. Cysteine disulfides are highly conserved in the ICK motif with C1-C4 (Disulfide-I), C2-C5(Disulfide-II), and C3-C6(Disulfide-III) connectivities in a sequence. Disulfide-I and disulfide-II form a loop and the disulfide-III tethers through the loop forming a knotted fold. The current report has analysed the conformation of disulfides in the ICK motif using the side-chain torsional angles of cysteine disulfide. In crystal structures: 88% of Disulfide-I have (+,-)SynRHHook, 92% of Disulfide-II have (+,-)RHSpiral, and 100% of Disulfide-III have (-,-)LHSpiral conformations. In NMR structures, conformational diversity has been observed for each of the cysteine disulfides of the ICK motif. The highest percentage occurrence in NMR structures: 27% of Disulfide-I have (+,-)SynRHHook, 36% of Disulfide-II have (+,-)RHSpiral, and 50% of Disulfide-III have (-,-)LHSpiral conformations. In the view of the method of identification of disulfides between cysteine residues using NMR spectroscopy, the NMR structure represents an ensemble of conformations of disulfides instead of specific disulfide conformation. The retention of the conformation in both X-ray and NMR structures supports the conservation of conformation of disulfides in the ICK motif. The tendency to exhibit specific conformation of disulfide even with variations in 3D structures supports the evolutionarily optimized nature of the ICK motif.
Collapse
|
20
|
Mellado G, Espinoza N, Garate JA, Neely A. Spider Toxin SNX-482 Gating Modifier Spontaneously Partitions in the Membrane Guided by Electrostatic Interactions. MEMBRANES 2022; 12:membranes12060595. [PMID: 35736302 PMCID: PMC9231009 DOI: 10.3390/membranes12060595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
Abstract
Spider toxin SNX-482 is a cysteine-rich peptide that interferes with calcium channel activity by binding to voltage-sensing domains of the CaV2.3 subtype. Two mechanisms dominate the binding process of cysteine-rich peptides.: direct binding from the aqueous phase or through lateral diffusion from the membrane, the so-called reduction in dimensionality mechanism. In this work, via coarse-grained and atomistic molecular dynamics simulations, we have systematically studied the spontaneous partitioning of SNX-482 with membranes of different anionic compositions and explored via diffusional analysis both binding mechanisms. Our simulations revealed a conserved protein patch that inserts in the membrane, a preference for binding towards partially negatively charged membranes, and that electrostatics guides membrane binding by incrementing and aligning the molecular dipole. Finally, diffusivity calculations showed that the toxin diffusion along the membrane plane is an order of magnitude slower than the aqueous phase suggesting that the critical factor in determining the SNX-482-CaV2.3 binding mechanism is the affinity between the membrane and SNX-482.
Collapse
Affiliation(s)
- Guido Mellado
- Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso 2362735, Chile; (G.M.); (N.E.)
- Biofisica y Biología Computacional, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso 2362735, Chile
| | - Nicolas Espinoza
- Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso 2362735, Chile; (G.M.); (N.E.)
- Biofisica y Biología Computacional, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso 2362735, Chile
- Millennium Nucleus in NanoBioPhysics, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso 2360102, Chile
| | - Jose Antonio Garate
- Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso 2362735, Chile; (G.M.); (N.E.)
- Millennium Nucleus in NanoBioPhysics, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso 2360102, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Santiago 7750000, Chile
- Correspondence: (J.A.G.); (A.N.)
| | - Alan Neely
- Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaiso, Valparaiso 2362735, Chile; (G.M.); (N.E.)
- Correspondence: (J.A.G.); (A.N.)
| |
Collapse
|
21
|
Herzig V, Chen YC, Chin YKY, Dekan Z, Chang YW, Yu HM, Alewood PF, Chen CC, King GF. The Tarantula Toxin ω-Avsp1a Specifically Inhibits Human CaV3.1 and CaV3.3 via the Extracellular S3-S4 Loop of the Domain 1 Voltage-Sensor. Biomedicines 2022; 10:biomedicines10051066. [PMID: 35625803 PMCID: PMC9138389 DOI: 10.3390/biomedicines10051066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 12/01/2022] Open
Abstract
Inhibition of T-type calcium channels (CaV3) prevents development of diseases related to cardiovascular and nerve systems. Further, knockout animal studies have revealed that some diseases are mediated by specific subtypes of CaV3. However, subtype-specific CaV3 inhibitors for therapeutic purposes or for studying the physiological roles of CaV3 subtypes are missing. To bridge this gap, we employed our spider venom library and uncovered that Avicularia spec. (“Amazonas Purple”, Peru) tarantula venom inhibited specific T-type CaV channel subtypes. By using chromatographic and mass-spectrometric techniques, we isolated and sequenced the active toxin ω-Avsp1a, a C-terminally amidated 36 residue peptide with a molecular weight of 4224.91 Da, which comprised the major peak in the venom. Both native (4.1 μM) and synthetic ω-Avsp1a (10 μM) inhibited 90% of CaV3.1 and CaV3.3, but only 25% of CaV3.2 currents. In order to investigate the toxin binding site, we generated a range of chimeric channels from the less sensitive CaV3.2 and more sensitive CaV3.3. Our results suggest that domain-1 of CaV3.3 is important for the inhibitory effect of ω-Avsp1a on T-type calcium channels. Further studies revealed that a leucine of T-type calcium channels is crucial for the inhibitory effect of ω-Avsp1a.
Collapse
Affiliation(s)
- Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (Y.K.-Y.C.); (Z.D.); (P.F.A.)
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
- Correspondence: (V.H.); (C.-C.C.); (G.F.K.); Tel.: +61-7-5456-5382 (V.H.); +886-2-2652-3522 (C.-C.C.); +61-7-3346-2025 (G.F.K.)
| | - Yong-Cyuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (Y.-C.C.); (Y.-W.C.)
| | - Yanni K.-Y. Chin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (Y.K.-Y.C.); (Z.D.); (P.F.A.)
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zoltan Dekan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (Y.K.-Y.C.); (Z.D.); (P.F.A.)
| | - Yu-Wang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (Y.-C.C.); (Y.-W.C.)
| | - Hui-Ming Yu
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan;
| | - Paul F. Alewood
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (Y.K.-Y.C.); (Z.D.); (P.F.A.)
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (Y.-C.C.); (Y.-W.C.)
- Correspondence: (V.H.); (C.-C.C.); (G.F.K.); Tel.: +61-7-5456-5382 (V.H.); +886-2-2652-3522 (C.-C.C.); +61-7-3346-2025 (G.F.K.)
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (Y.K.-Y.C.); (Z.D.); (P.F.A.)
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD 4072, Australia
- Correspondence: (V.H.); (C.-C.C.); (G.F.K.); Tel.: +61-7-5456-5382 (V.H.); +886-2-2652-3522 (C.-C.C.); +61-7-3346-2025 (G.F.K.)
| |
Collapse
|
22
|
A Deep Learning Approach with Data Augmentation to Predict Novel Spider Neurotoxic Peptides. Int J Mol Sci 2021; 22:ijms222212291. [PMID: 34830173 PMCID: PMC8619404 DOI: 10.3390/ijms222212291] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
As major components of spider venoms, neurotoxic peptides exhibit structural diversity, target specificity, and have great pharmaceutical potential. Deep learning may be an alternative to the laborious and time-consuming methods for identifying these peptides. However, the major hurdle in developing a deep learning model is the limited data on neurotoxic peptides. Here, we present a peptide data augmentation method that improves the recognition of neurotoxic peptides via a convolutional neural network model. The neurotoxic peptides were augmented with the known neurotoxic peptides from UniProt database, and the models were trained using a training set with or without the generated sequences to verify the augmented data. The model trained with the augmented dataset outperformed the one with the unaugmented dataset, achieving accuracy of 0.9953, precision of 0.9922, recall of 0.9984, and F1 score of 0.9953 in simulation dataset. From the set of all RNA transcripts of Callobius koreanus spider, we discovered neurotoxic peptides via the model, resulting in 275 putative peptides of which 252 novel sequences and only 23 sequences showing homology with the known peptides by Basic Local Alignment Search Tool. Among these 275 peptides, four were selected and shown to have neuromodulatory effects on the human neuroblastoma cell line SH-SY5Y. The augmentation method presented here may be applied to the identification of other functional peptides from biological resources with insufficient data.
Collapse
|
23
|
Dashevsky D, Rodriguez J. A Short Review of the Venoms and Toxins of Spider Wasps (Hymenoptera: Pompilidae). Toxins (Basel) 2021; 13:toxins13110744. [PMID: 34822528 PMCID: PMC8622703 DOI: 10.3390/toxins13110744] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Parasitoid wasps represent the plurality of venomous animals, but have received extremely little research in proportion to this taxonomic diversity. The lion’s share of investigation into insect venoms has focused on eusocial hymenopterans, but even this small sampling shows great promise for the development of new active substances. The family Pompilidae is known as the spider wasps because of their reproductive habits which include hunting for spiders, delivering a paralyzing sting, and entombing them in burrows with one of the wasp’s eggs to serve as food for the developing larva. The largest members of this family, especially the tarantula hawks of the genus Pepsis, have attained notoriety for their large size, dramatic coloration, long-term paralysis of their prey, and incredibly painful defensive stings. In this paper we review the existing research regarding the composition and function of pompilid venoms, discuss parallels from other venom literatures, identify possible avenues for the adaptation of pompilid toxins towards human purposes, and future directions of inquiry for the field.
Collapse
|
24
|
Stability and Safety of Inhibitor Cystine Knot Peptide, GTx1-15, from the Tarantula Spider Grammostola rosea. Toxins (Basel) 2021; 13:toxins13090621. [PMID: 34564625 PMCID: PMC8473062 DOI: 10.3390/toxins13090621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 12/19/2022] Open
Abstract
Inhibitor cystine knot (ICK) peptides are knotted peptides with three intramolecular disulfide bonds that affect several types of ion channels. Some are proteolytically stable and are promising scaffolds for drug development. GTx1-15 is an ICK peptide that inhibits the voltage-dependent calcium channel Cav3.1 and the voltage-dependent sodium channels Nav1.3 and Nav1.7. As a model molecule to develop an ICK peptide drug, we investigated several important pharmaceutical characteristics of GTx1-15. The stability of GTx1-15 in rat and human blood plasma was examined, and no GTx1-15 degradation was observed in either rat or human blood plasma for 24 h in vitro. GTx1-15 in blood circulation was detected for several hours after intravenous and intramuscular administration, indicating high stability in plasma. The thermal stability of GTx1-15 as examined by high thermal incubation and protein thermal shift assays indicated that GTx1-15 possesses high heat stability. The cytotoxicity and immunogenicity of GTx1-15 were examined using the human monocytic leukemia cell line THP-1. GTx1-15 showed no cytotoxicity or immunogenicity even at high concentrations. These results indicate that GTx1-15 itself is suitable for peptide drug development and as a peptide library scaffold.
Collapse
|
25
|
Lüddecke T, Herzig V, von Reumont BM, Vilcinskas A. The biology and evolution of spider venoms. Biol Rev Camb Philos Soc 2021; 97:163-178. [PMID: 34453398 DOI: 10.1111/brv.12793] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022]
Abstract
Spiders are diverse, predatory arthropods that have inhabited Earth for around 400 million years. They are well known for their complex venom systems that are used to overpower their prey. Spider venoms contain many proteins and peptides with highly specific and potent activities suitable for biomedical or agrochemical applications, but the key role of venoms as an evolutionary innovation is often overlooked, even though this has enabled spiders to emerge as one of the most successful animal lineages. In this review, we discuss these neglected biological aspects of spider venoms. We focus on the morphology of spider venom systems, their major components, biochemical and chemical plasticity, as well as ecological and evolutionary trends. We argue that the effectiveness of spider venoms is due to their unprecedented complexity, with diverse components working synergistically to increase the overall potency. The analysis of spider venoms is difficult to standardize because they are dynamic systems, fine-tuned and modified by factors such as sex, life-history stage and biological role. Finally, we summarize the mechanisms that drive spider venom evolution and highlight the need for genome-based studies to reconstruct the evolutionary history and physiological networks of spider venom compounds with more certainty.
Collapse
Affiliation(s)
- Tim Lüddecke
- Department for Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Ohlebergsweg 12, Gießen, 35392, Germany.,LOEWE Centre for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, Frankfurt am Main, 60325, Germany
| | - Volker Herzig
- GeneCology Research Centre, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia.,School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia
| | - Björn M von Reumont
- LOEWE Centre for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, Frankfurt am Main, 60325, Germany.,Institute for Insect Biotechnology, Justus-Liebig University Giessen, Heinrich-Buff-Ring 26-32, Gießen, 35392, Germany
| | - Andreas Vilcinskas
- Department for Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Ohlebergsweg 12, Gießen, 35392, Germany.,LOEWE Centre for Translational Biodiversity Genomics (TBG), Senckenberganlage 25, Frankfurt am Main, 60325, Germany.,Institute for Insect Biotechnology, Justus-Liebig University Giessen, Heinrich-Buff-Ring 26-32, Gießen, 35392, Germany
| |
Collapse
|
26
|
Jiang Y, Castro J, Blomster LV, Agwa AJ, Maddern J, Schober G, Herzig V, Chow CY, Cardoso FC, Demétrio De Souza França P, Gonzales J, Schroeder CI, Esche S, Reiner T, Brierley SM, King GF. Pharmacological Inhibition of the Voltage-Gated Sodium Channel Na V1.7 Alleviates Chronic Visceral Pain in a Rodent Model of Irritable Bowel Syndrome. ACS Pharmacol Transl Sci 2021; 4:1362-1378. [PMID: 34423271 DOI: 10.1021/acsptsci.1c00072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Indexed: 12/12/2022]
Abstract
The human nociceptor-specific voltage-gated sodium channel 1.7 (hNaV1.7) is critical for sensing various types of somatic pain, but it appears not to play a primary role in acute visceral pain. However, its role in chronic visceral pain remains to be determined. We used assay-guided fractionation to isolate a novel hNaV1.7 inhibitor, Tsp1a, from tarantula venom. Tsp1a is 28-residue peptide that potently inhibits hNaV1.7 (IC50 = 10 nM), with greater than 100-fold selectivity over hNaV1.3-hNaV1.6, 45-fold selectivity over hNaV1.1, and 24-fold selectivity over hNaV1.2. Tsp1a is a gating modifier that inhibits NaV1.7 by inducing a hyperpolarizing shift in the voltage-dependence of channel inactivation and slowing recovery from fast inactivation. NMR studies revealed that Tsp1a adopts a classical knottin fold, and like many knottin peptides, it is exceptionally stable in human serum. Remarkably, intracolonic administration of Tsp1a completely reversed chronic visceral hypersensitivity in a mouse model of irritable bowel syndrome. The ability of Tsp1a to reduce visceral hypersensitivity in a model of irritable bowel syndrome suggests that pharmacological inhibition of hNaV1.7 at peripheral sensory nerve endings might be a viable approach for eliciting analgesia in patients suffering from chronic visceral pain.
Collapse
Affiliation(s)
- Yan Jiang
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Linda V Blomster
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Akello J Agwa
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Jessica Maddern
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Gudrun Schober
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Paula Demétrio De Souza França
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Otorhinolaryngology & Head and Neck Surgery, Federal University of São Paulo, São Paulo 04021-001, Brazil
| | - Junior Gonzales
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | | | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Radiology, Weill Cornell Medical College, New York, New York 10021, United States
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
27
|
Cook Sangar ML, Girard EJ, Hopping G, Yin C, Pakiam F, Brusniak MY, Nguyen E, Ruff R, Gewe MM, Byrnes-Blake K, Nairn NW, Miller DM, Mehlin C, Strand AD, Mhyre AJ, Correnti CE, Strong RK, Simon JA, Olson JM. A potent peptide-steroid conjugate accumulates in cartilage and reverses arthritis without evidence of systemic corticosteroid exposure. Sci Transl Med 2021; 12:12/533/eaay1041. [PMID: 32132215 DOI: 10.1126/scitranslmed.aay1041] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 01/23/2020] [Indexed: 12/12/2022]
Abstract
On-target, off-tissue toxicity limits the systemic use of drugs that would otherwise reduce symptoms or reverse the damage of arthritic diseases, leaving millions of patients in pain and with limited physical mobility. We identified cystine-dense peptides (CDPs) that rapidly accumulate in cartilage of the knees, ankles, hips, shoulders, and intervertebral discs after systemic administration. These CDPs could be used to concentrate arthritis drugs in joints. A cartilage-accumulating peptide, CDP-11R, reached peak concentration in cartilage within 30 min after administration and remained detectable for more than 4 days. Structural analysis of the peptides by crystallography revealed that the distribution of positive charge may be a distinguishing feature of joint-accumulating CDPs. In addition, quantitative whole-body autoradiography showed that the disulfide-bonded tertiary structure is critical for cartilage accumulation and retention. CDP-11R distributed to joints while carrying a fluorophore imaging agent or one of two different steroid payloads, dexamethasone (dex) and triamcinolone acetonide (TAA). Of the two payloads, the dex conjugate did not advance because the free drug released into circulation was sufficient to cause on-target toxicity. In contrast, the CDP-11R-TAA conjugate alleviated joint inflammation in the rat collagen-induced model of rheumatoid arthritis while avoiding toxicities that occurred with nontargeted steroid treatment at the same molar dose. This conjugate shows promise for clinical development and establishes proof of concept for multijoint targeting of disease-modifying therapeutic payloads.
Collapse
Affiliation(s)
- Michelle L Cook Sangar
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Emily J Girard
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Gene Hopping
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Chunfeng Yin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Fiona Pakiam
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mi-Youn Brusniak
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Elizabeth Nguyen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Raymond Ruff
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mesfin M Gewe
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | - Christopher Mehlin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Andrew D Strand
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Andrew J Mhyre
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Colin E Correnti
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Roland K Strong
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Julian A Simon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - James M Olson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
28
|
Trim CM, Byrne LJ, Trim SA. Utilisation of compounds from venoms in drug discovery. PROGRESS IN MEDICINAL CHEMISTRY 2021; 60:1-66. [PMID: 34147202 DOI: 10.1016/bs.pmch.2021.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Difficult drug targets are becoming the normal course of business in drug discovery, sometimes due to large interacting surfaces or only small differences in selectivity regions. For these, a different approach is merited: compounds lying somewhere between the small molecule and the large antibody in terms of many properties including stability, biodistribution and pharmacokinetics. Venoms have evolved over millions of years to be complex mixtures of stable molecules derived from other somatic molecules, the stability comes from the pressure to be ready for delivery at a moment's notice. Snakes, spiders, scorpions, jellyfish, wasps, fish and even mammals have evolved independent venom systems with complex mixtures in their chemical arsenal. These venom-derived molecules have been proven to be useful tools, such as for the development of antihypotensive angiotensin converting enzyme (ACE) inhibitors and have also made successful drugs such as Byetta® (Exenatide), Integrilin® (Eptifibatide) and Echistatin. Only a small percentage of the available chemical space from venoms has been investigated so far and this is growing. In a new era of biological therapeutics, venom peptides present opportunities for larger target engagement surface with greater stability than antibodies or human peptides. There are challenges for oral absorption and target engagement, but there are venom structures that overcome these and thus provide substrate for engineering novel molecules that combine all desired properties. Venom researchers are characterising new venoms, species, and functions all the time, these provide great substrate for solving the challenges presented by today's difficult targets.
Collapse
Affiliation(s)
- Carol M Trim
- Faculty of Science, Engineering and Social Sciences, Natural and Applied Sciences, School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, Kent, United Kingdom
| | - Lee J Byrne
- Faculty of Science, Engineering and Social Sciences, Natural and Applied Sciences, School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, Kent, United Kingdom
| | | |
Collapse
|
29
|
Gremski LH, Matsubara FH, Polli NLC, Antunes BC, Schluga PHDC, da Justa HC, Minozzo JC, Wille ACM, Senff-Ribeiro A, Veiga SS. Prospective Use of Brown Spider Venom Toxins as Therapeutic and Biotechnological Inputs. Front Mol Biosci 2021; 8:706704. [PMID: 34222343 PMCID: PMC8247472 DOI: 10.3389/fmolb.2021.706704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/04/2021] [Indexed: 11/20/2022] Open
Abstract
Brown spider (genus Loxosceles) venoms are mainly composed of protein toxins used for predation and defense. Bites of these spiders most commonly produce a local dermonecrotic lesion with gravitational spread, edema and hemorrhage, which together are defined as cutaneous loxoscelism. Systemic loxoscelism, such as hematological abnormalities and renal injury, are less frequent but more lethal. Some Loxosceles venom toxins have already been isolated and extensively studied, such as phospholipases D (PLDs), which have been recombinantly expressed and were proven to reproduce toxic activities associated to the whole venom. PLDs have a notable potential to be engineered and converted in non-toxic antigens to produce a new generation of antivenoms or vaccines. PLDs also can serve as tools to discover inhibitors to be used as therapeutic agents. Other Loxosceles toxins have been identified and functionally characterized, such as hyaluronidases, allergen factor, serpin, TCTP and knottins (ICK peptides). All these toxins were produced as recombinant molecules and are biologically active molecules that can be used as tools for the potential development of chemical candidates to tackle many medical and biological threats, acting, for instance, as antitumoral, insecticides, analgesic, antigens for allergy tests and biochemical reagents for cell studies. In addition, these recombinant toxins may be useful to develop a rational therapy for loxoscelism. This review summarizes the main candidates for the development of drugs and biotechnological inputs that have been described in Brown spider venoms.
Collapse
Affiliation(s)
| | | | | | - Bruno Cesar Antunes
- Department of Cell Biology, Federal University of Paraná, Curitiba, Brazil.,Production and Research Center of Immunobiological Products, State Department of Health, Piraquara, Brazil
| | | | | | - João Carlos Minozzo
- Production and Research Center of Immunobiological Products, State Department of Health, Piraquara, Brazil
| | - Ana Carolina Martins Wille
- Department of Structural, Molecular Biology and Genetics, State University of Ponta Grossa, Ponta Grossa, Brazil
| | | | | |
Collapse
|
30
|
Giribaldi J, Smith JJ, Schroeder CI. Recent developments in animal venom peptide nanotherapeutics with improved selectivity for cancer cells. Biotechnol Adv 2021; 50:107769. [PMID: 33989705 DOI: 10.1016/j.biotechadv.2021.107769] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023]
Abstract
Animal venoms are a rich source of bioactive peptides that efficiently modulate key receptors and ion channels involved in cellular excitability to rapidly neutralize their prey or predators. As such, they have been a wellspring of highly useful pharmacological tools for decades. Besides targeting ion channels, some venom peptides exhibit strong cytotoxic activity and preferentially affect cancer over healthy cells. This is unlikely to be driven by an evolutionary impetus, and differences in tumor cells and the tumor microenvironment are probably behind the serendipitous selectivity shown by some venom peptides. However, strategies such as bioconjugation and nanotechnologies are showing potential to improve their selectivity and potency, thereby paving the way to efficiently harness new anticancer mechanisms offered by venom peptides. This review aims to highlight advances in nano- and chemotherapeutic tools and prospective anti-cancer drug leads derived from animal venom peptides.
Collapse
Affiliation(s)
- Julien Giribaldi
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Jennifer J Smith
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Christina I Schroeder
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| |
Collapse
|
31
|
Ryan RYM, Seymour J, Loukas A, Lopez JA, Ikonomopoulou MP, Miles JJ. Immunological Responses to Envenomation. Front Immunol 2021; 12:661082. [PMID: 34040609 PMCID: PMC8141633 DOI: 10.3389/fimmu.2021.661082] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/01/2021] [Indexed: 01/05/2023] Open
Abstract
Venoms are complex mixtures of toxic compounds delivered by bite or sting. In humans, the consequences of envenomation range from self-limiting to lethal. Critical host defence against envenomation comprises innate and adaptive immune strategies targeted towards venom detection, neutralisation, detoxification, and symptom resolution. In some instances, venoms mediate immune dysregulation that contributes to symptom severity. This review details the involvement of immune cell subtypes and mediators, particularly of the dermis, in host resistance and venom-induced immunopathology. We further discuss established venom-associated immunopathology, including allergy and systemic inflammation, and investigate Irukandji syndrome as a potential systemic inflammatory response. Finally, this review characterises venom-derived compounds as a source of immune modulating drugs for treatment of disease.
Collapse
Affiliation(s)
- Rachael Y. M. Ryan
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
- School of Environment and Sciences, Griffith University, Nathan, QLD, Australia
| | - Jamie Seymour
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - Alex Loukas
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - J. Alejandro Lopez
- School of Environment and Sciences, Griffith University, Nathan, QLD, Australia
- QIMR Berghofer Medical Research Institute, The University of Queensland, Herston, QLD, Australia
| | - Maria P. Ikonomopoulou
- Translational Venomics Group, Madrid Institute for Advanced Studies (IMDEA) in Food, CEI UAM+CSIC, Madrid, Spain
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - John J. Miles
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
32
|
Bell J, Sukiran NA, Walsh S, Fitches EC. The insecticidal activity of recombinant nemertide toxin α-1 from Lineus longissimus towards pests and beneficial species. Toxicon 2021; 197:79-86. [PMID: 33852905 DOI: 10.1016/j.toxicon.2021.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 01/05/2023]
Abstract
The nemertide toxins from the phylum Nemertea are a little researched family of neurotoxins with potential for development as biopesticides. Here we report the recombinant production of nemertide α-1 (α-1), a 65-residue inhibitor cystine knot (ICK) peptide from Lineus longissimus, known to target insect voltage-gated sodium channels. The insecticidal activity of α-1 was assessed and compared with the well characterised ICK venom peptide, ω-atracotoxin/hexatoxin-Hv1a (Hv1a). α-1 elicited potent spastic paralysis when injected into cabbage moth (Mamestra brassicae) larvae; conferring an ED50 3.90 μg/larva (10.30 nmol/g larva), followed by mortality (60% within 48 h after 10 μg injection). By comparison, injection of M. brassicae larvae with recombinant Hv1a produced short-lived flaccid paralysis with an ED50 over 6 times greater than that of α-1 at 26.20 μg/larva (64.70 nmol/g larva). Oral toxicity of α-1 was demonstrated against two aphid species (Myzus persicae and Acyrthosiphon pisum), with respective LC50 values of 0.35 and 0.14 mg/mL, some 6-fold lower than those derived for recombinant Hv1a. When delivered orally to M. brassicae larvae, α-1 caused both paralysis (ED50 11.93 μg/larva, 31.5 nmol/g larva) and mortality. This contrasts with the lack of oral activity of Hv1a, which when fed to M. brassicae larvae had no effect on feeding or survival. Hv1a has previously been shown to be non-toxic by injection to the beneficial honeybee (Apis mellifera). By contrast, rapid paralysis and 100% mortality was observed following injection of α-1 (31.6 nmol/g insect). These results demonstrate the great potential of naturally occurring non-venomous peptides, such as α-1, for development as novel effective biopesticides, but equally highlights the importance of understanding the phyletic specificity of a given toxin at an early stage in the quest to discover and develop safe and sustainable pesticides.
Collapse
Affiliation(s)
- Jack Bell
- Durham University, Department of Biosciences, Stockton Rd, Durham, DH1 3LE, UK.
| | - Nur Afiqah Sukiran
- Durham University, Department of Biosciences, Stockton Rd, Durham, DH1 3LE, UK
| | - Stephen Walsh
- Durham University, Department of Biosciences, Stockton Rd, Durham, DH1 3LE, UK
| | - Elaine C Fitches
- Durham University, Department of Biosciences, Stockton Rd, Durham, DH1 3LE, UK
| |
Collapse
|
33
|
Animal Venoms-Curse or Cure? Biomedicines 2021; 9:biomedicines9040413. [PMID: 33921205 PMCID: PMC8068803 DOI: 10.3390/biomedicines9040413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 12/16/2022] Open
Abstract
An estimated 15% of animals are venomous, with representatives spread across the majority of animal lineages. Animals use venoms for various purposes, such as prey capture and predator deterrence. Humans have always been fascinated by venomous animals in a Janus-faced way. On the one hand, humans have a deeply rooted fear of venomous animals. This is boosted by their largely negative image in public media and the fact that snakes alone cause an annual global death toll in the hundreds of thousands, with even more people being left disabled or disfigured. Consequently, snake envenomation has recently been reclassified by the World Health Organization as a neglected tropical disease. On the other hand, there has been a growth in recent decades in the global scene of enthusiasts keeping venomous snakes, spiders, scorpions, and centipedes in captivity as pets. Recent scientific research has focussed on utilising animal venoms and toxins for the benefit of humanity in the form of molecular research tools, novel diagnostics and therapeutics, biopesticides, or anti-parasitic treatments. Continued research into developing efficient and safe antivenoms and promising discoveries of beneficial effects of animal toxins is further tipping the scales in favour of the “cure” rather than the “curse” prospect of venoms.
Collapse
|
34
|
Tran P, Schroeder CI. Enzymatic Ligation of Disulfide-Rich Animal Venom Peptides: Using Sortase A to Form Double-Knotted Peptides. Methods Mol Biol 2021; 2355:83-92. [PMID: 34386952 DOI: 10.1007/978-1-0716-1617-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Sortase A is a thiol transpeptidase expressed by Gram-positive bacteria. This enzyme is capable of site-specifically ligating peptides containing the C-terminal recognition motif LPXTG to peptides containing an N-terminal polyglycine sequence, forming a native peptide bond. Here, we describe the preparation and application of sortase A to the ligation of two individually folded disulfide-rich animal venom peptides in order to form a heterodimeric double-knotted peptide with a native peptide linker. This method is mild enough to preserve the structures and disulfide connectivities of the peptides during ligation. We employed a highly efficient sortase A pentamutant (SrtA5°), which brings the reaction to completion within 15 min with a ~50-80% yield of ligated peptide.
Collapse
Affiliation(s)
- Poanna Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
- National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| |
Collapse
|
35
|
Chen M, Peng S, Wang L, Yang L, Si Y, Zhou X, Zhang Y, Liu Z. Recombinant PaurTx-3, a spider toxin, inhibits sodium channels and decreases membrane excitability in DRG neurons. Biochem Biophys Res Commun 2020; 533:958-964. [PMID: 33004176 DOI: 10.1016/j.bbrc.2020.09.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/23/2020] [Indexed: 10/23/2022]
Abstract
Voltage-gated sodium channels are critical for the generation and propagation of action potentials. Gating modifier toxins from spider venom can modulate the gating mechanism of sodium channels and thus have potential as drug leads. Here, we established expression of the gating modifier toxin PaurTx-3, a sodium channel inhibitor found in the venom of the spider Phrixotrichus auratus. Whole-cell voltage-clamp recordings indicated that recombinant PaurTx-3 (rPaurTx-3) inhibited Nav1.4, Nav1.5, and Nav1.7 currents with IC50 values of 61 nM, 72 nM, and 25 nM, respectively. Furthermore, rPaurTx-3 irreversibly inhibited Nav1.7 currents, but had 60-70% recovery in Nav1.4 and Nav1.5 after washing with a bath solution. rPaurTx-3 also hyperpolarized the voltage-dependent steady-state inactivation curve and significantly slowed recovery from fast inactivation of Nav1.7. Current-clamp recordings showed that rPaurTx-3 suppressed small DRG neuron activity. The biological activity assay findings for rPaurTx-3 support its potent pharmacological effect in Nav1.7 and small DRG neurons.
Collapse
Affiliation(s)
- Minzhi Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Shuijiao Peng
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Li Wang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Li Yang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yuxin Si
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Xi Zhou
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| | - Yunxiao Zhang
- Key Laboratory of Hunan Province for Advanced Carbon-based Functional Materials, School of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang, 414006, Hunan, China.
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
36
|
Herzig V, Cristofori-Armstrong B, Israel MR, Nixon SA, Vetter I, King GF. Animal toxins - Nature's evolutionary-refined toolkit for basic research and drug discovery. Biochem Pharmacol 2020; 181:114096. [PMID: 32535105 PMCID: PMC7290223 DOI: 10.1016/j.bcp.2020.114096] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/27/2022]
Abstract
Venomous animals have evolved toxins that interfere with specific components of their victim's core physiological systems, thereby causing biological dysfunction that aids in prey capture, defense against predators, or other roles such as intraspecific competition. Many animal lineages evolved venom systems independently, highlighting the success of this strategy. Over the course of evolution, toxins with exceptional specificity and high potency for their intended molecular targets have prevailed, making venoms an invaluable and almost inexhaustible source of bioactive molecules, some of which have found use as pharmacological tools, human therapeutics, and bioinsecticides. Current biomedically-focused research on venoms is directed towards their use in delineating the physiological role of toxin molecular targets such as ion channels and receptors, studying or treating human diseases, targeting vectors of human diseases, and treating microbial and parasitic infections. We provide examples of each of these areas of venom research, highlighting the potential that venom molecules hold for basic research and drug development.
Collapse
Affiliation(s)
- Volker Herzig
- School of Science & Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia; Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia.
| | | | - Mathilde R Israel
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Samantha A Nixon
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
37
|
Toxin-like neuropeptides in the sea anemone Nematostella unravel recruitment from the nervous system to venom. Proc Natl Acad Sci U S A 2020; 117:27481-27492. [PMID: 33060291 DOI: 10.1073/pnas.2011120117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The sea anemone Nematostella vectensis (Anthozoa, Cnidaria) is a powerful model for characterizing the evolution of genes functioning in venom and nervous systems. Although venom has evolved independently numerous times in animals, the evolutionary origin of many toxins remains unknown. In this work, we pinpoint an ancestral gene giving rise to a new toxin and functionally characterize both genes in the same species. Thus, we report a case of protein recruitment from the cnidarian nervous to venom system. The ShK-like1 peptide has a ShKT cysteine motif, is lethal for fish larvae and packaged into nematocysts, the cnidarian venom-producing stinging capsules. Thus, ShK-like1 is a toxic venom component. Its paralog, ShK-like2, is a neuropeptide localized to neurons and is involved in development. Both peptides exhibit similarities in their functional activities: They provoke contraction in Nematostella polyps and are toxic to fish. Because ShK-like2 but not ShK-like1 is conserved throughout sea anemone phylogeny, we conclude that the two paralogs originated due to a Nematostella-specific duplication of a ShK-like2 ancestor, a neuropeptide-encoding gene, followed by diversification and partial functional specialization. ShK-like2 is represented by two gene isoforms controlled by alternative promoters conferring regulatory flexibility throughout development. Additionally, we characterized the expression patterns of four other peptides with structural similarities to studied venom components and revealed their unexpected neuronal localization. Thus, we employed genomics, transcriptomics, and functional approaches to reveal one venom component, five neuropeptides with two different cysteine motifs, and an evolutionary pathway from nervous to venom system in Cnidaria.
Collapse
|
38
|
Touchard A, Mendel HC, Boulogne I, Herzig V, Braga Emidio N, King GF, Triquigneaux M, Jaquillard L, Beroud R, De Waard M, Delalande O, Dejean A, Muttenthaler M, Duplais C. Heterodimeric Insecticidal Peptide Provides New Insights into the Molecular and Functional Diversity of Ant Venoms. ACS Pharmacol Transl Sci 2020; 3:1211-1224. [PMID: 33344898 DOI: 10.1021/acsptsci.0c00119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Indexed: 12/14/2022]
Abstract
Ants use venom for predation, defense, and communication; however, the molecular diversity, function, and potential applications of ant venom remains understudied compared to other venomous lineages such as arachnids, snakes and cone snails. In this work, we used a multidisciplinary approach that encompassed field work, proteomics, sequencing, chemical synthesis, structural analysis, molecular modeling, stability studies, and in vitro and in vivo bioassays to investigate the molecular diversity of the venom of the Amazonian Pseudomyrmex penetrator ants. We isolated a potent insecticidal heterodimeric peptide Δ-pseudomyrmecitoxin-Pp1a (Δ-PSDTX-Pp1a) composed of a 27-residue long A-chain and a 33-residue long B-chain cross-linked by two disulfide bonds in an antiparallel orientation. We chemically synthesized Δ-PSDTX-Pp1a, its corresponding parallel AA and BB homodimers, and its monomeric chains and demonstrated that Δ-PSDTX-Pp1a had the most potent insecticidal effects in blowfly assays (LD50 = 3 nmol/g). Molecular modeling and circular dichroism studies revealed strong α-helical features, indicating its cytotoxic effects could derive from cell membrane pore formation or disruption. The native heterodimer was substantially more stable against proteolytic degradation (t 1/2 = 13 h) than its homodimers or monomers (t 1/2 < 20 min), indicating an evolutionary advantage of the more complex structure. The proteomic analysis of Pseudomyrmex penetrator venom and in-depth characterization of Δ-PSDTX-Pp1a provide novel insights in the structural complexity of ant venom and further exemplifies how nature exploits disulfide-bond formation and dimerization to gain an evolutionary advantage via improved stability, a concept that is highly relevant for the design and development of peptide therapeutics, molecular probes, and bioinsecticides.
Collapse
Affiliation(s)
- Axel Touchard
- CNRS, UMR Ecofog, AgroParisTech, Cirad, INRAE, Université des Antilles, Université de Guyane, Kourou 97310, France
| | - Helen C Mendel
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Isabelle Boulogne
- Université de ROUEN, UFR des Sciences et Techniques, Laboratoire Glycobiologie et Matrice Extracellulaire Végétale, UPRES-EA 4358, Fédération de Recherche Normandie Végétal FED 4277, Mont-Saint-Aignan 76821, France
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.,GeneCology Research Centre, School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland 4556, Australia
| | - Nayara Braga Emidio
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | | | - Lucie Jaquillard
- Smartox Biotechnology, 6 rue des Platanes, Saint Egrève 38120, France
| | - Rémy Beroud
- Smartox Biotechnology, 6 rue des Platanes, Saint Egrève 38120, France
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, Saint Egrève 38120, France.,Université de Nantes, CNRS, INSERM, L'institut du thorax, Nantes 44000, France.,LabEx, Ion Channels, Science & Therapeutics, Valbonne 06560, France
| | - Olivier Delalande
- Institute of Genetics and Development of Rennes (IGDR), CNRS UMR 6290, Université de Rennes Faculté de Pharmacie, 2 avenue du Professeur Léon Bernard, Rennes 35043, France
| | - Alain Dejean
- CNRS, UMR Ecofog, AgroParisTech, Cirad, INRAE, Université des Antilles, Université de Guyane, Kourou 97310, France.,Ecolab, Université de Toulouse, CNRS, INPT, UPS, Toulouse 31058, France
| | - Markus Muttenthaler
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.,Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Christophe Duplais
- CNRS, UMR Ecofog, AgroParisTech, Cirad, INRAE, Université des Antilles, Université de Guyane, Kourou 97310, France
| |
Collapse
|
39
|
Zhang Q, Si Y, Yang L, Wang L, Peng S, Chen Y, Chen M, Zhou X, Liu Z. Two Novel Peptide Toxins from the Spider Cyriopagopus longipes Inhibit Tetrodotoxin-Sensitive Sodium Channels. Toxins (Basel) 2020; 12:toxins12090529. [PMID: 32824960 PMCID: PMC7551932 DOI: 10.3390/toxins12090529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/16/2020] [Indexed: 12/24/2022] Open
Abstract
Sodium channels play a critical role in the generation and propagation of action potentials in excitable tissues, such as nerves, cardiac muscle, and skeletal muscle, and are the primary targets of toxins found in animal venoms. Here, two novel peptide toxins (Cl6a and Cl6b) were isolated from the venom of the spider Cyriopagopus longipes and characterized. Cl6a and Cl6b were shown to be inhibitors of tetrodotoxin-sensitive (TTX-S), but not TTX-resistant, sodium channels. Among the TTX-S channels investigated, Cl6a and Cl6b showed the highest degree of inhibition against NaV1.7 (half-maximal inhibitory concentration (IC50) of 11.0 ± 2.5 nM and 18.8 ± 2.4 nM, respectively) in an irreversible manner that does not alter channel activation, inactivation, or repriming kinetics. Moreover, analysis of NaV1.7/NaV1.8 chimeric channels revealed that Cl6b is a site 4 neurotoxin. Site-directed mutagenesis analysis indicated that D816, V817, and E818 observably affected the efficacy of the Cl6b-NaV1.7 interaction, suggesting that these residues might directly affect the interaction of NaV1.7 with Cl6b. Taken together, these two novel peptide toxins act as potent and sustained NaV1.7 blockers and may have potential in the pharmacological study of sodium channels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xi Zhou
- Correspondence: (X.Z); (Z.L.)
| | | |
Collapse
|
40
|
Crook ZR, Girard E, Sevilla GP, Merrill M, Friend D, Rupert PB, Pakiam F, Nguyen E, Yin C, Ruff RO, Hopping G, Strand AD, Finton KAK, Coxon M, Mhyre AJ, Strong RK, Olson JM. A TfR-Binding Cystine-Dense Peptide Promotes Blood-Brain Barrier Penetration of Bioactive Molecules. J Mol Biol 2020; 432:3989-4009. [PMID: 32304700 PMCID: PMC9569163 DOI: 10.1016/j.jmb.2020.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 04/01/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023]
Abstract
The impenetrability of the blood-brain barrier (BBB) to most conventional drugs impedes the treatment of central nervous system (CNS) disorders. Interventions for diseases like brain cancer, neurodegeneration, or age-associated inflammatory processes require varied approaches to CNS drug delivery. Cystine-dense peptides (CDPs) have drawn recent interest as drugs or drug-delivery vehicles. Found throughout the phylogenetic tree, often in drug-like roles, their size, stability, and protein interaction capabilities make CDPs an attractive mid-size biologic scaffold to complement conventional antibody-based drugs. Here, we describe the identification, maturation, characterization, and utilization of a CDP that binds to the transferrin receptor (TfR), a native receptor and BBB transporter for the iron chaperone transferrin. We developed variants with varying binding affinities (KD as low as 216 pM), co-crystallized it with the receptor, and confirmed murine cross-reactivity. It accumulates in the mouse CNS at ~25% of blood levels (CNS blood content is only ~1%-6%) and delivers neurotensin, an otherwise non-BBB-penetrant neuropeptide, at levels capable of modulating CREB signaling in the mouse brain. Our work highlights the utility of CDPs as a diverse, easy-to-screen scaffold family worthy of inclusion in modern drug discovery strategies, demonstrated by the discovery of a candidate CNS drug delivery vehicle ready for further optimization and preclinical development.
Collapse
Affiliation(s)
- Zachary R Crook
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Emily Girard
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Gregory P Sevilla
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Morgan Merrill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Della Friend
- Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Peter B Rupert
- Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Fiona Pakiam
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Elizabeth Nguyen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Chunfeng Yin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Raymond O Ruff
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Gene Hopping
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Andrew D Strand
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Kathryn A K Finton
- Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Margo Coxon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Andrew J Mhyre
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Roland K Strong
- Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - James M Olson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, USA.
| |
Collapse
|
41
|
Rupasinghe DB, Herzig V, Vetter I, Dekan Z, Gilchrist J, Bosmans F, Alewood PF, Lewis RJ, King GF. Mutational analysis of ProTx-I and the novel venom peptide Pe1b provide insight into residues responsible for selective inhibition of the analgesic drug target Na V1.7. Biochem Pharmacol 2020; 181:114080. [PMID: 32511987 DOI: 10.1016/j.bcp.2020.114080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/30/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022]
Abstract
Management of chronic pain presents a major challenge, since many currently available treatments lack efficacy and have problems such as addiction and tolerance. Loss of function mutations in the SCN9A gene lead to a congenital inability to feel pain, with no other sensory deficits aside from anosmia. SCN9A encodes the voltage-gated sodium (NaV) channel 1.7 (NaV1.7), which has been identified as a primary pain target. However, in developing NaV1.7-targeted analgesics, extreme care must to be taken to avoid off-target activity on other NaV subtypes that are critical for survival. Since spider venoms are an excellent source of NaV channel modulators, we screened a panel of spider venoms to identify selective NaV1.7 inhibitors. This led to identification of two novel NaV modulating venom peptides (β/μ-theraphotoxin-Pe1a and β/μ-theraphotoxin-Pe1b (Pe1b) from the arboreal tarantula Phormingochilus everetti. A third peptide isolated from the tarantula Bumba pulcherrimaklaasi was identical to the well-known ProTx-I (β/ω-theraphotoxin-Tp1a) from the tarantula Thrixopelma pruriens. A tethered toxin (t-toxin)-based alanine scanning strategy was used to determine the NaV1.7 pharmacophore of ProTx-I. We designed several ProTx-I and Pe1b analogues, and tested them for activity and NaV channel subtype selectivity. Several analogues had improved potency against NaV1.7, and altered specificity against other NaV channels. These analogues provide a foundation for development of Pe1b as a lead molecule for therapeutic inhibition of NaV1.7.
Collapse
Affiliation(s)
- Darshani B Rupasinghe
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4105, Australia
| | - Zoltan Dekan
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - John Gilchrist
- Department of Physiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Frank Bosmans
- Department of Physiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
42
|
Chow CY, Absalom N, Biggs K, King GF, Ma L. Venom-derived modulators of epilepsy-related ion channels. Biochem Pharmacol 2020; 181:114043. [PMID: 32445870 DOI: 10.1016/j.bcp.2020.114043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022]
Abstract
Epilepsy is characterised by spontaneous recurrent seizures that are caused by an imbalance between neuronal excitability and inhibition. Since ion channels play fundamental roles in the generation and propagation of action potentials as well as neurotransmitter release at a subset of excitatory and inhibitory synapses, their dysfunction has been linked to a wide variety of epilepsies. Indeed, these unique proteins are the major biological targets for antiepileptic drugs. Selective targeting of a specific ion channel subtype remains challenging for small molecules, due to the high level of homology among members of the same channel family. As a consequence, there is a growing trend to target ion channels with biologics. Venoms are the best known natural source of ion channel modulators, and venom peptides are increasingly recognised as potential therapeutics due to their high selectivity and potency gained through millions of years of evolutionary selection pressure. Here we describe the major ion channel families involved in the pathogenesis of various types of epilepsy, including voltage-gated Na+, K+, Ca2+ channels, Cys-loop receptors, ionotropic glutamate receptors and P2X receptors, and currently available venom-derived peptides that target these channel proteins. Although only a small number of venom peptides have successfully progressed to the clinic, there is reason to be optimistic about their development as antiepileptic drugs, notwithstanding the challenges associated with development of any class of peptide drug.
Collapse
Affiliation(s)
- Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nathan Absalom
- Brain and Mind Centre, School of Pharmacy, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW 2050, Australia
| | - Kimberley Biggs
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Linlin Ma
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia.
| |
Collapse
|
43
|
Structural venomics reveals evolution of a complex venom by duplication and diversification of an ancient peptide-encoding gene. Proc Natl Acad Sci U S A 2020; 117:11399-11408. [PMID: 32398368 DOI: 10.1073/pnas.1914536117] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Spiders are one of the most successful venomous animals, with more than 48,000 described species. Most spider venoms are dominated by cysteine-rich peptides with a diverse range of pharmacological activities. Some spider venoms contain thousands of unique peptides, but little is known about the mechanisms used to generate such complex chemical arsenals. We used an integrated transcriptomic, proteomic, and structural biology approach to demonstrate that the lethal Australian funnel-web spider produces 33 superfamilies of venom peptides and proteins. Twenty-six of the 33 superfamilies are disulfide-rich peptides, and we show that 15 of these are knottins that contribute >90% of the venom proteome. NMR analyses revealed that most of these disulfide-rich peptides are structurally related and range in complexity from simple to highly elaborated knottin domains, as well as double-knot toxins, that likely evolved from a single ancestral toxin gene.
Collapse
|
44
|
Crook ZR, Sevilla GP, Mhyre AJ, Olson JM. Mammalian Surface Display Screening of Diverse Cystine-Dense Peptide Libraries for Difficult-to-Drug Targets. Methods Mol Biol 2020; 2070:363-396. [PMID: 31625107 DOI: 10.1007/978-1-4939-9853-1_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Many diseases are mediated by targets that are not amenable to conventional small-molecule drug approaches. While antibody-based drugs have undeniable utility, peptides of the 1-9 kDa size range (10-80 amino acids) have drawn interest as alternate drug scaffolds This is born of a desire to identify compounds with the advantages of antibody-based therapeutics (affinity, potency, specificity, and ability to disrupt protein:protein interactions) without all of their liabilities (large size, expensive manufacturing, and necessity of humanization). Of these alternate scaffolds, cystine-dense peptides (CDPs) have several specific benefits. Due to their stable intra-chain disulfide bridges, CDPs often demonstrate resistance to heat and proteolysis, along with low immunogenicity. These properties do not require chemical modifications, permitting CDP screening by conventional genetic means. The cystine topology of a typical CDP requires an oxidative environment, and we have found that the mammalian secretory pathway is most effective at allowing diverse CDPs to achieve a stable fold. As such, high-diversity screens to identify CDPs that interact with targets of interest can be efficiently conducted using mammalian surface display. In this protocol, we present the theory and tools to conduct a mammalian surface display screen for CDPs that bind with targets of interest, including the steps to validate binding and mature the affinity of preliminary candidates. With these methods, CDPs of all kinds can be brought to bear against targets that would benefit from a peptide-based intervention.
Collapse
Affiliation(s)
- Zachary R Crook
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Gregory P Sevilla
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Andrew J Mhyre
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - James M Olson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|
45
|
Tran HNT, Tran P, Deuis JR, Agwa AJ, Zhang AH, Vetter I, Schroeder CI. Enzymatic Ligation of a Pore Blocker Toxin and a Gating Modifier Toxin: Creating Double-Knotted Peptides with Improved Sodium Channel NaV1.7 Inhibition. Bioconjug Chem 2019; 31:64-73. [DOI: 10.1021/acs.bioconjchem.9b00744] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hue N. T. Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Poanna Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer R. Deuis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Akello J. Agwa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alan H. Zhang
- Center for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Christina I. Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
46
|
Morlighem JÉRL, Radis-Baptista G. The Place for Enzymes and Biologically Active Peptides from Marine Organisms for Application in Industrial and Pharmaceutical Biotechnology. Curr Protein Pept Sci 2019; 20:334-355. [PMID: 30255754 DOI: 10.2174/1389203719666180926121722] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/10/2018] [Accepted: 09/15/2018] [Indexed: 01/07/2023]
Abstract
Since the beginning of written history, diverse texts have reported the use of enzymatic preparations in food processing and have described the medicinal properties of crude and fractionated venoms to treat various diseases and injuries. With the biochemical characterization of enzymes from distinct sources and bioactive polypeptides from animal venoms, the last sixty years have testified the advent of industrial enzymology and protein therapeutics, which are currently applicable in a wide variety of industrial processes, household products, and pharmaceuticals. Bioprospecting of novel biocatalysts and bioactive peptides is propelled by their unsurpassed properties that are applicable for current and future green industrial processes, biotechnology, and biomedicine. The demand for both novel enzymes with desired characteristics and novel peptides that lead to drug development, has experienced a steady increase in response to the expanding global market for industrial enzymes and peptidebased drugs. Moreover, although largely unexplored, oceans and marine realms, with their unique ecosystems inhabited by a large variety of species, including a considerable number of venomous animals, are recognized as untapped reservoirs of molecules and macromolecules (enzymes and bioactive venom-derived peptides) that can potentially be converted into highly valuable biopharmaceutical products. In this review, we have focused on enzymes and animal venom (poly)peptides that are presently in biotechnological use, and considering the state of prospection of marine resources, on the discovery of useful industrial biocatalysts and drug leads with novel structures exhibiting selectivity and improved performance.
Collapse
Affiliation(s)
- Jean-Étienne R L Morlighem
- Institute for Marine Sciences, Federal University of Ceara, Av da Abolicao 3207. Fortaleza/CE. 60165081, Brazil
| | - Gandhi Radis-Baptista
- Institute for Marine Sciences, Federal University of Ceara, Av da Abolicao 3207. Fortaleza/CE. 60165081, Brazil
| |
Collapse
|
47
|
King GF. Tying pest insects in knots: the deployment of spider-venom-derived knottins as bioinsecticides. PEST MANAGEMENT SCIENCE 2019; 75:2437-2445. [PMID: 31025461 DOI: 10.1002/ps.5452] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/20/2019] [Accepted: 04/25/2019] [Indexed: 06/09/2023]
Abstract
Spider venoms are complex chemical arsenals that contain a rich variety of insecticidal toxins. However, the major toxin class in many spider venoms is disulfide-rich peptides known as knottins. The knotted three-dimensional fold of these mini-proteins provides them with exceptional chemical and thermal stability as well as resistance to proteases. In contrast with other bioinsecticides, which are often slow-acting, spider knottins are fast-acting neurotoxins. In addition to being potently insecticidal, some knottins have exceptional taxonomic selectivity, being lethal to key agricultural pests but innocuous to vertebrates and beneficial insects such as bees. The intrinsic oral activity of these peptides, combined with the ability of aerosolized knottins to penetrate insect spiracles, has enabled them to be developed commercially as eco-friendly bioinsecticides. Moreover, it has been demonstrated that spider-knottin transgenes can be used to engineer faster-acting entomopathogens and insect-resistant crops. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
48
|
Drukewitz SH, Bokelmann L, Undheim EAB, von Reumont BM. Toxins from scratch? Diverse, multimodal gene origins in the predatory robber fly Dasypogon diadema indicate a dynamic venom evolution in dipteran insects. Gigascience 2019; 8:giz081. [PMID: 31289835 PMCID: PMC6615979 DOI: 10.1093/gigascience/giz081] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/07/2019] [Accepted: 06/14/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Venoms and the toxins they contain represent molecular adaptations that have evolved on numerous occasions throughout the animal kingdom. However, the processes that shape venom protein evolution are poorly understood because of the scarcity of whole-genome data available for comparative analyses of venomous species. RESULTS We performed a broad comparative toxicogenomic analysis to gain insight into the genomic mechanisms of venom evolution in robber flies (Asilidae). We first sequenced a high-quality draft genome of the hymenopteran hunting robber fly Dasypogon diadema, analysed its venom by a combined proteotranscriptomic approach, and compared our results with recently described robber fly venoms to assess the general composition and major components of asilid venom. We then applied a comparative genomics approach, based on 1 additional asilid genome, 10 high-quality dipteran genomes, and 2 lepidopteran outgroup genomes, to reveal the evolutionary mechanisms and origins of identified venom proteins in robber flies. CONCLUSIONS While homologues were identified for 15 of 30 predominant venom protein in the non-asilid genomes, the remaining 15 highly expressed venom proteins appear to be unique to robber flies. Our results reveal that the venom of D. diadema likely evolves in a multimodal fashion comprising (i) neofunctionalization after gene duplication, (ii) expression-dependent co-option of proteins, and (iii) asilid lineage-specific orphan genes with enigmatic origin. The role of such orphan genes is currently being disputed in evolutionary genomics but has not been discussed in the context of toxin evolution. Our results display an unexpected dynamic venom evolution in asilid insects, which contrasts the findings of the only other insect toxicogenomic evolutionary analysis, in parasitoid wasps (Hymenoptera), where toxin evolution is dominated by single gene co-option. These findings underpin the significance of further genomic studies to cover more neglected lineages of venomous taxa and to understand the importance of orphan genes as possible drivers for venom evolution.
Collapse
Affiliation(s)
- Stephan Holger Drukewitz
- Institute for Biology, University of Leipzig, Talstrasse 33, 04103 Leipzig, Germany
- Project group Bioresources, Animal Venomics, Fraunhofer Institute for Molecular Biology and Applied Ecology, Winchesterstrasse 2, 35392 Gießen, Germany
| | - Lukas Bokelmann
- Evolutionary Genetics Department, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, D-04103 Leipzig, Germany
| | - Eivind A B Undheim
- Centre for Advanced Imaging, The University of Queensland, St. Lucia, QLD 4072, Australia
- Centre for Ecology and Evolutionary Synthesis, Department of Biosciences, University of Oslo, PO Box 1066 Blindern, 0316 Oslo, Norway
| | - Björn M von Reumont
- Project group Bioresources, Animal Venomics, Fraunhofer Institute for Molecular Biology and Applied Ecology, Winchesterstrasse 2, 35392 Gießen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Institute for Insect Biotechnology, Justus Liebig University, Heinrich Buff Ring 58, 35394 Gießen, Germany
| |
Collapse
|
49
|
Pore-Forming Proteins from Cnidarians and Arachnids as Potential Biotechnological Tools. Toxins (Basel) 2019; 11:toxins11060370. [PMID: 31242582 PMCID: PMC6628452 DOI: 10.3390/toxins11060370] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/31/2022] Open
Abstract
Animal venoms are complex mixtures of highly specialized toxic molecules. Cnidarians and arachnids produce pore-forming proteins (PFPs) directed against the plasma membrane of their target cells. Among PFPs from cnidarians, actinoporins stand out for their small size and molecular simplicity. While native actinoporins require only sphingomyelin for membrane binding, engineered chimeras containing a recognition antibody-derived domain fused to an actinoporin isoform can nonetheless serve as highly specific immunotoxins. Examples of such constructs targeted against malignant cells have been already reported. However, PFPs from arachnid venoms are less well-studied from a structural and functional point of view. Spiders from the Latrodectus genus are professional insect hunters that, as part of their toxic arsenal, produce large PFPs known as latrotoxins. Interestingly, some latrotoxins have been identified as potent and highly-specific insecticides. Given the proteinaceous nature of these toxins, their promising future use as efficient bioinsecticides is discussed throughout this Perspective. Protein engineering and large-scale recombinant production are critical steps for the use of these PFPs as tools to control agriculturally important insect pests. In summary, both families of PFPs, from Cnidaria and Arachnida, appear to be molecules with promising biotechnological applications.
Collapse
|
50
|
Denisov SS, Ippel JH, Heinzmann ACA, Koenen RR, Ortega-Gomez A, Soehnlein O, Hackeng TM, Dijkgraaf I. Tick saliva protein Evasin-3 modulates chemotaxis by disrupting CXCL8 interactions with glycosaminoglycans and CXCR2. J Biol Chem 2019; 294:12370-12379. [PMID: 31235521 PMCID: PMC6699855 DOI: 10.1074/jbc.ra119.008902] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/11/2019] [Indexed: 01/22/2023] Open
Abstract
Chemokines are a group of chemotaxis proteins that regulate cell trafficking and play important roles in immune responses and inflammation. Ticks are blood-sucking parasites that secrete numerous immune-modulatory agents in their saliva to evade host immune responses. Evasin-3 is a small salivary protein that belongs to a class of chemokine-binding proteins isolated from the brown dog tick, Rhipicephalus sanguineus. Evasin-3 has been shown to have a high affinity for chemokines CXCL1 and CXCL8 and to diminish inflammation in mice. In the present study, solution NMR spectroscopy was used to investigate the structure of Evasin-3 and its CXCL8–Evasin-3 complex. Evasin-3 is found to disrupt the glycosaminoglycan-binding site of CXCL8 and inhibit the interaction of CXCL8 with CXCR2. Structural data were used to design two novel CXCL8-binding peptides. The linear tEv3 17–56 and cyclic tcEv3 16–56 dPG Evasin-3 variants were chemically synthesized by solid-phase peptide synthesis. The affinity of these newly synthesized variants to CXCL8 was measured by surface plasmon resonance biosensor analysis. The Kd values of tEv3 17–56 and tcEv3 16–56 dPG were 27 and 13 nm, respectively. Both compounds effectively inhibited CXCL8-induced migration of polymorphonuclear neutrophils. The present results suggest utility of synthetic Evasin-3 variants as scaffolds for designing and fine-tuning new chemokine-binding agents that suppress immune responses and inflammation.
Collapse
Affiliation(s)
- Stepan S Denisov
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht, 6229 ER, Maastricht, The Netherlands
| | - Johannes H Ippel
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht, 6229 ER, Maastricht, The Netherlands
| | - Alexandra C A Heinzmann
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht, 6229 ER, Maastricht, The Netherlands
| | - Rory R Koenen
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht, 6229 ER, Maastricht, The Netherlands
| | - Almudena Ortega-Gomez
- Institute for Cardiovascular Prevention, Ludwig Maximilian University, 80336, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, Ludwig Maximilian University, 80336, Munich, Germany; German Center for Cardiovascular Research, 13316, Berlin, Germany; Partner Site Munich Heart Alliance, 80802 Munich, Germany; Department of Physiology and Pharmacology and Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Tilman M Hackeng
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht, 6229 ER, Maastricht, The Netherlands
| | - Ingrid Dijkgraaf
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|