1
|
Takjoo R, Wilson DT, Le Quilliec J, Schmidt CA, Zhao G, Liddell MJ, Shaikh NY, Sunagar K, Loukas A, Smout MJ, Daly NL. Structural analysis of an Asterias rubens peptide indicates the presence of a disulfide-directed β-hairpin fold. FEBS Open Bio 2025; 15:415-426. [PMID: 39561265 PMCID: PMC11891777 DOI: 10.1002/2211-5463.13931] [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: 05/24/2024] [Revised: 10/19/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
Sea stars are an abundant group of marine invertebrates that display remarkably robust regenerative capabilities throughout all life stages. Numerous proteins and peptides have been identified in a proteome study on the coelomic fluid (biofluid) of the common sea star Asterias rubens, which appear to be involved with the wound-healing response in the organism. However, the three-dimensional structure and function of several of these injury-responsive peptides, including the peptide KASH2, are yet to be investigated. Here, we show that the KASH2 peptide adopts a disulfide-directed β-hairpin fold (DDH). The DDH motif appears to be evolutionarily related to the inhibitor cystine knot motif, which is one of the most widespread disulfide-rich peptide folds. The DDH motif was originally thought to be restricted to arachnids, but our study suggests that as a result of convergent evolution it could also have originated in sea stars. Although the widely conserved DDH fold has potential cross-phyla wound-healing capacity, we have shown that KASH2 does not enhance the proliferation of human fibroblasts, a simple method for wound-healing re-epithelialisation screening. Therefore, additional research is necessary to determine the role of KASH2 in the sea stars.
Collapse
Affiliation(s)
- Rozita Takjoo
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - David T. Wilson
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - Justine Le Quilliec
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
- Univ Brest, École Supérieure d'Ingénieurs en Agroalimentaire de Bretagne atlantiquePlouzanéFrance
| | - Casey A. Schmidt
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - Guangzu Zhao
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | | | - Naeem Y. Shaikh
- Evolutionary Venomics Lab, Centre for Ecological SciencesIndian Institute of ScienceBangaloreIndia
| | - Kartik Sunagar
- Evolutionary Venomics Lab, Centre for Ecological SciencesIndian Institute of ScienceBangaloreIndia
| | - Alex Loukas
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - Michael J. Smout
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - Norelle L. Daly
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| |
Collapse
|
2
|
Freuville L, Matthys C, Quinton L, Gillet JP. Venom-derived peptides for breaking through the glass ceiling of drug development. Front Chem 2024; 12:1465459. [PMID: 39398192 PMCID: PMC11468230 DOI: 10.3389/fchem.2024.1465459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
Venoms are complex mixtures produced by animals and consist of hundreds of components including small molecules, peptides, and enzymes selected for effectiveness and efficacy over millions of years of evolution. With the development of venomics, which combines genomics, transcriptomics, and proteomics to study animal venoms and their effects deeply, researchers have identified molecules that selectively and effectively act against membrane targets, such as ion channels and G protein-coupled receptors. Due to their remarkable physico-chemical properties, these molecules represent a credible source of new lead compounds. Today, not less than 11 approved venom-derived drugs are on the market. In this review, we aimed to highlight the advances in the use of venom peptides in the treatment of diseases such as neurological disorders, cardiovascular diseases, or cancer. We report on the origin and activity of the peptides already approved and provide a comprehensive overview of those still in development.
Collapse
Affiliation(s)
- Lou Freuville
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Chloé Matthys
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| | - Loïc Quinton
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Jean-Pierre Gillet
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| |
Collapse
|
3
|
Tytgat J. A personal view on the history of toxins: From ancient times to artificial intelligence. Toxicon 2024; 248:108034. [PMID: 39038662 DOI: 10.1016/j.toxicon.2024.108034] [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: 05/02/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
Bioactive substances found in plants, microorganisms and animals have fascinated mankind since time immemorial. This review will focus on the progress that has been made over the centuries and our growing insights. The developments relate to both the discovery and characterization of novel bioactive substances, as well as the ceaseless implementation of refined techniques, the use of high-end instruments and breakthroughs in artificial intelligence with deep learning-based computational methods. As these approaches possess great translational potential, with many applications in different fields, such as therapeutic, diagnostic and agrochemical use, there is a good rationale to continue investing in toxinology-related research.
Collapse
Affiliation(s)
- Jan Tytgat
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg, ON2, PO Box 922, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
4
|
Ciesiołkiewicz A, Lizandra Perez J, Skalniak L, Noceń P, Berlicki Ł. Miniprotein engineering for inhibition of PD-1/PD-L1 interaction. Protein Sci 2024; 33:e5106. [PMID: 39012010 PMCID: PMC11250529 DOI: 10.1002/pro.5106] [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: 01/15/2024] [Revised: 06/07/2024] [Accepted: 06/23/2024] [Indexed: 07/17/2024]
Abstract
Miniproteins constitute an excellent basis for the development of structurally demanding functional molecules. The engrailed homeodomain, a three-helix-containing miniprotein, was applied as a scaffold for constructing programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) interaction inhibitors. PD-L1 binders were initially designed using the computer-aided approach and subsequently optimized iteratively. The conformational stability was assessed for each obtained miniprotein using circular dichroism spectroscopy, indicating that numerous mutations could be introduced. The formation of a sizable hydrophobic surface at the inhibitor that fits the molecular target imposed the necessity for the incorporation of additional charged amino acid residues to retain its appropriate solubility. Finally, the miniprotein effectively binding to PD-L1 (KD = 51.4 nM) that inhibits PD-1/PD-L1 interaction in cell-based studies with EC50 = 3.9 μM, was discovered.
Collapse
Affiliation(s)
| | - Juan Lizandra Perez
- Department of Bioorganic ChemistryWrocław University of Science and TechnologyWrocławPoland
| | | | - Paweł Noceń
- Department of Bioorganic ChemistryWrocław University of Science and TechnologyWrocławPoland
| | - Łukasz Berlicki
- Department of Bioorganic ChemistryWrocław University of Science and TechnologyWrocławPoland
| |
Collapse
|
5
|
Raffaelli T, Wilson DT, Dutertre S, Giribaldi J, Vetter I, Robinson SD, Thapa A, Widi A, Loukas A, Daly NL. Structural analysis of a U-superfamily conotoxin containing a mini-granulin fold: Insights into key features that distinguish between the ICK and granulin folds. J Biol Chem 2024; 300:107203. [PMID: 38508311 PMCID: PMC11035057 DOI: 10.1016/j.jbc.2024.107203] [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: 11/16/2023] [Revised: 02/28/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024] Open
Abstract
We are entering an exciting time in structural biology where artificial intelligence can be used to predict protein structures with greater accuracy than ever before. Extending this level of accuracy to the predictions of disulfide-rich peptide structures is likely to be more challenging, at least in the short term, given the tight packing of cysteine residues and the numerous ways that the disulfide bonds can potentially be linked. It has been previously shown in many cases that several disulfide bond connectivities can be accommodated by a single set of NMR-derived structural data without significant violations. Disulfide-rich peptides are prevalent throughout nature, and arguably the most well-known are those present in venoms from organisms such as cone snails. Here, we have determined the first three-dimensional structure and disulfide connectivity of a U-superfamily cone snail venom peptide, TxVIIB. TxVIIB has a VI/VII cysteine framework that is generally associated with an inhibitor cystine knot (ICK) fold; however, AlphaFold predicted that the peptide adopts a mini-granulin fold with a granulin disulfide connectivity. Our experimental studies using NMR spectroscopy and orthogonal protection of cysteine residues indicate that TxVIIB indeed adopts a mini-granulin fold but with the ICK disulfide connectivity. Our findings provide structural insight into the underlying features that govern formation of the mini-granulin fold rather than the ICK fold and will provide fundamental information for prediction algorithms, as the subtle complexity of disulfide isomers may be not adequately addressed by the current prediction algorithms.
Collapse
Affiliation(s)
- Tiziano Raffaelli
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - David T Wilson
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | | | | | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia; School of Pharmacy, The University of Queensland, Queensland, Australia
| | - Samuel D Robinson
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Ashvriya Thapa
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia; School of Pharmacy, The University of Queensland, Queensland, Australia
| | - Antin Widi
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Norelle L Daly
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia.
| |
Collapse
|
6
|
Hua X, Yao J, Liu X, Liu Q, Deng Y, Li S, Valdivia CR, Wang F, Pozzolini M, Shou Z, Valdivia HH, Xiao L. Comparison of the structure-function of five newly members of the calcin family. Int J Biol Macromol 2024; 260:129424. [PMID: 38219929 DOI: 10.1016/j.ijbiomac.2024.129424] [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: 08/30/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Calcins are a group of scorpion toxin peptides specifically binding to ryanodine receptors (RyRs) with high affinity, and have the ability to activate and stabilize RyR in a long-lasting subconductance state. Five newly calcins synthesized compounds exhibit typical structural characteristics of a specific family through chemical synthesis and virtual analysis. As the calcins from the same species, Petersiicalcin1 and Petersiicalcin2, Jendekicalcin2 and Jendekicalcin3, have only one residue difference. Both Petersiicalcin1 and Petersiicalcin2 exhibited different affinities in stimulating [3H]ryanodine binding, but the residue mutation resulted in a 2.7 folds difference. Other calcins also exhibited a stimulatory effect on [3H]ryanodine binding to RyR1, however, their affinities were significantly lower than that of Petersiiicalcin1 and Petersiiicalcin2. The channel domain of RyR1 was found to be capable of binding with the basic residues of these calcins, which also exhibited interactions with the S6 helices on RyR1. Dynamic simulations were conducted for Petersiicalcin1 and Petersiicalcin2, which demonstrated their ability to form a highly stable conformation and resulting in an asymmetric tetramer structure of RyR1. The discovery of five newly calcins further enriches the diversity of the natural calcin family, which provides more native peptides for the structure-function analysis between calcin and RyRs.
Collapse
Affiliation(s)
- Xiaoyu Hua
- Department of Occupational and Environmental Health, Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Jinchi Yao
- Department of Occupational and Environmental Health, Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; School of Life Sciences, Liaoning Normal University, Dalian 116081, China
| | - Xinyan Liu
- Department of Traditional Chinese Medicine Surgery, the First Affiliated Hospital of the Navy Medical University, Shanghai, 200433, China
| | - Qing Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, Taigu 030801, China
| | - Yuchen Deng
- Department of Dermatology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Songhua Li
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Carmen R Valdivia
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| | - Fei Wang
- Department of Occupational and Environmental Health, Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Marina Pozzolini
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Via Pastore 3, 16132 Genova, Italy.
| | - Zhaoyong Shou
- Faculty of Health Service, Nacal Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Héctor H Valdivia
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| | - Liang Xiao
- Department of Occupational and Environmental Health, Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| |
Collapse
|
7
|
Aslam L, Kaur R, Hussain S, Kapoor N, Mahajan R. LC-MS/MS identification and structural characterization of isolated cyclotides from precursor sequences of Viola odorata L. petiole tissue using computational approach. J Biosci 2022. [DOI: 10.1007/s12038-022-00283-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
8
|
Miniproteins in medicinal chemistry. Bioorg Med Chem Lett 2022; 71:128806. [PMID: 35660515 DOI: 10.1016/j.bmcl.2022.128806] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 11/20/2022]
Abstract
Miniproteins exhibit great potential as scaffolds for drug candidates because of their well-defined structure and good synthetic availability. Because of recently described methodologies for their de novo design, the field of miniproteins is emerging and can provide molecules that effectively bind to problematic targets, i.e., those that have been previously considered to be undruggable. This review describes methodologies for the development of miniprotein scaffolds and for the construction of biologically active miniproteins.
Collapse
|
9
|
Yoon KA, Kim WJ, Lee S, Yang HS, Lee BH, Lee SH. Comparative analyses of the venom components in the salivary gland transcriptomes and saliva proteomes of some heteropteran insects. INSECT SCIENCE 2022; 29:411-429. [PMID: 34296820 DOI: 10.1111/1744-7917.12955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 06/13/2023]
Abstract
Salivary gland-specific transcriptomes of nine heteropteran insects with distinct feeding strategies (predaceous, hematophagous, and phytophagous) were analyzed and annotated to compare and identify the venom components as well as their expression profiles. The transcriptional abundance of venom genes was verified via quantitative real-time PCR. Hierarchical clustering of 30 representative differentially expressed venom genes from the nine heteropteran species revealed unique groups of salivary gland-specific genes depending on their feeding strategy. The commonly transcribed genes included a paralytic neurotoxin (arginine kinase), digestive enzymes (cathepsin and serine protease), an anti-inflammatory protein (cystatin), hexamerin, and an odorant binding protein. Both predaceous and hematophagous (bed bug) heteropteran species showed relatively higher transcription levels of genes encoding proteins involved in proteolysis and cytolysis, whereas phytophagous heteropterans exhibited little or no expression of these genes, but had a high expression of vitellogenin, a multifunctional allergen. Saliva proteomes from four representative species were also analyzed. All venom proteins identified via saliva proteome analysis were annotated using salivary gland transcriptome data. The proteomic expression profiles of venom proteins were in good agreement with the salivary gland-specific transcriptomic profiles. Our results indicate that profiling of the salivary gland transcriptome provides important information on the composition and evolutionary features of venoms depending on their feeding strategy.
Collapse
Affiliation(s)
- Kyungjae Andrew Yoon
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | | | - Seungki Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon, Korea
| | - Hee-Sun Yang
- National Institute of Biological Resources, Environmental Research Complex, Incheon, Korea
| | - Byoung-Hee Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon, Korea
| | - Si Hyeock Lee
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
- Department of Agricultural Biology, Seoul National University, Seoul, Korea
| |
Collapse
|
10
|
Luo R, Liu H, Cheng Z. Protein scaffolds: Antibody alternative for cancer diagnosis and therapy. RSC Chem Biol 2022; 3:830-847. [PMID: 35866165 PMCID: PMC9257619 DOI: 10.1039/d2cb00094f] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/23/2022] [Indexed: 12/01/2022] Open
Abstract
Although antibodies are well developed and widely used in cancer therapy and diagnostic fields, some defects remain, such as poor tissue penetration, long in vivo metabolic retention, potential cytotoxicity, patent limitation, and high production cost. These issues have led scientists to explore and develop novel antibody alternatives. Protein scaffolds are small monomeric proteins with stable tertiary structures and mutable residues, which emerged in the 1990s. By combining robust gene engineering and phage display techniques, libraries with sufficient diversity could be established for target binding scaffold selection. Given the properties of small size, high affinity, and excellent specificity and stability, protein scaffolds have been applied in basic research, and preclinical and clinical fields over the past two decades. To date, more than 20 types of protein scaffolds have been developed, with the most frequently used being affibody, adnectin, ANTICALIN®, DARPins, and knottin. In this review, we focus on the protein scaffold applications in cancer therapy and diagnosis in the last 5 years, and discuss the pros and cons, and strategies of optimization and design. Although antibodies are well developed and widely used in cancer therapy and diagnostic fields, some defects remain, such as poor tissue penetration, long in vivo metabolic retention, potential cytotoxicity, patent limitation, and high production cost.![]()
Collapse
Affiliation(s)
- Renli Luo
- Department of Molecular Medicine, College of Life and Health Sciences, Northeastern University Shenyang China
| | - Hongguang Liu
- Department of Molecular Medicine, College of Life and Health Sciences, Northeastern University Shenyang China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- Drug Discovery Shandong Laboratory, Bohai Rim Advanced Research Institute for Drug Discovery Yantai Shandong 264117 China
| |
Collapse
|
11
|
Hellinger R, Muratspahić E, Devi S, Koehbach J, Vasileva M, Harvey PJ, Craik DJ, Gründemann C, Gruber CW. Importance of the Cyclic Cystine Knot Structural Motif for Immunosuppressive Effects of Cyclotides. ACS Chem Biol 2021; 16:2373-2386. [PMID: 34592097 PMCID: PMC9286316 DOI: 10.1021/acschembio.1c00524] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cyclotide T20K inhibits the proliferation of human immune cells and is currently in clinical trials for multiple sclerosis. Here, we provide novel functional data and mechanistic insights into structure-activity relationships of T20K. Analogs with partial or complete reduction of the cystine knot had loss of function in proliferation experiments. Similarly, an acyclic analog of T20K was inactive in lymphocyte bioassays. The lack of activity of non-native peptide analogs appears to be associated with the ability of cyclotides to interact with and penetrate cell membranes, since cellular uptake studies demonstrated fast fractional transfer only of the native peptide into the cytosol of human immune cells. Therefore, structural differences between cyclic and linear native folded peptides were investigated by NMR to elucidate structure-activity relationships. Acyclic T20K had a less rigid backbone and considerable structural changes in loops 1 and 6 compared to the native cyclic T20K, supporting the idea that the cyclic cystine knot motif is a unique bioactive scaffold. This study provides evidence that this structural motif in cyclotides governs bioactivity, interactions with and transport across biological membranes, and the structural integrity of these peptides. These observations could be useful to understand the structure-activity of other cystine knot proteins due to the structural conservation of the cystine knot motif across evolution and to provide guidance for the design of novel cyclic cysteine-stabilized molecules.
Collapse
Affiliation(s)
- Roland Hellinger
- Center for
Physiology and Pharmacology, Medical University
of Vienna, Schwarzspanierstr. 17, Vienna 1090, Austria
| | - Edin Muratspahić
- Center for
Physiology and Pharmacology, Medical University
of Vienna, Schwarzspanierstr. 17, Vienna 1090, Austria
| | - Seema Devi
- Institute
for Infection Prevention and Hospital Epidemiology, Center for Complementary
Medicine, Faculty of Medicine, University
of Freiburg, Breisacher Str. 115B, Freiburg 79106, Germany
| | - Johannes Koehbach
- Institute
for Molecular Bioscience, Australian Research Council Centre of Excellence
for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Mina Vasileva
- Center for
Physiology and Pharmacology, Medical University
of Vienna, Schwarzspanierstr. 17, Vienna 1090, Austria
| | - Peta J. Harvey
- Institute
for Molecular Bioscience, Australian Research Council Centre of Excellence
for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David J. Craik
- Institute
for Molecular Bioscience, Australian Research Council Centre of Excellence
for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Carsten Gründemann
- Translational
Complementary Medicine, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstr. 80, Basel 4056, Switzerland
| | - Christian W. Gruber
- Center for
Physiology and Pharmacology, Medical University
of Vienna, Schwarzspanierstr. 17, Vienna 1090, Austria
| |
Collapse
|
12
|
Pan NC, Zhang T, Hu S, Liu C, Wang Y. Fast desensitization of acetylcholine receptors induced by a spider toxin. Channels (Austin) 2021; 15:507-515. [PMID: 34374321 PMCID: PMC8366537 DOI: 10.1080/19336950.2021.1961459] [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] [Indexed: 11/09/2022] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) are members of the “cys-loop” ligand-gated ion channel superfamily that play important roles in both the peripheral and central system. At the neuromuscular junction, the endplate current is induced by ACh binding and nAChR activation, and then, the current declines to a small steady state, even though ACh is still bound to the receptors. The kinetics of nAChRs with high affinity for ACh but no measurable ion conductance is called desensitization. This adopted desensitization of nAChR channel currents might be an important mechanism for protecting cells against uncontrolled excitation. This study aimed to show that Grammostola spatulata toxin (GsMTx4), which was first purified and characterized from the venom of the tarantula Grammostola spatulata (now genus Phixotricus), can facilitate the desensitization of nAChRs in murine C2C12 myotubes. To examine the details, muscle-type nAChRs, which are expressed heterologously in HEK293T cells, were studied. A single channel current was recorded under the cell-attached configuration, and the channel activity (NPo) decayed much faster after the addition of GsMTx-4 to the pipette solution. The channel kinetics were further analyzed, and GsMTx-4 affected the channel activity of nAChRs by prolonging the closing time without affecting channel conductance or opening activity. The interaction between nAChRs embedded in the lipid membrane and toxin inserted into the membrane may contribute to the conformational change in the receptor and thus change the channel activity. This new property of GsMTx-4 may lead to a better understanding of the desensitization of ligand-gated channels and disease therapy.
Collapse
Affiliation(s)
- Na Clara Pan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neuromodulation, Beijing, China
| | - Tingting Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neuromodulation, Beijing, China
| | - Shimin Hu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neuromodulation, Beijing, China
| | - Chunyan Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neuromodulation, Beijing, China
| | - Yuping Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neuromodulation, Beijing, China.,Centre of Epilepsy, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Chen T, Tang S, Hecht ES, Yen CW, Andersen N, Chin S, Cadang L, Roper B, Estevez A, Rohou A, Chang D, Dai L, Liu P, Al-Sayah M, Nagapudi K, Lin F, Famili A, Hu C, Kuhn R, Stella C, Crittenden CM, Gruenhagen JA, Venkatramani C, Hannoush RN, Leung D, Vandlen R, Yehl P. Discovery of a dual pathway aggregation mechanism for a therapeutic constrained peptide. J Pharm Sci 2021; 110:2362-2371. [PMID: 33652014 DOI: 10.1016/j.xphs.2020.12.041] [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: 11/11/2020] [Accepted: 12/07/2020] [Indexed: 10/22/2022]
Abstract
Constrained peptides (CPs) have emerged as attractive candidates for drug discovery and development. To fully unlock the therapeutic potential of CPs, it is crucial to understand their physical stability and minimize the formation of aggregates that could induce immune responses. Although amyloid like aggregates have been researched extensively, few studies have focused on aggregates from other peptide scaffolds (e.g., CPs). In this work, a streamlined approach to effectively profile the nature and formation pathway of CP aggregates was demonstrated. Aggregates of various sizes were detected and shown to be amorphous. Though no major changes were found in peptide structure upon aggregation, these aggregates appeared to have mixed natures, consisting of primarily non-covalent aggregates with a low level of covalent species. This co-existence phenomenon was also supported by two kinetic pathways observed in time- and temperature-dependent aggregation studies. Furthermore, a stability study with 8 additional peptide variants exhibited good correlation between aggregation propensity and peptide hydrophobicity. Therefore, a dual aggregation pathway was proposed, with the non-covalent aggregates driven by hydrophobic interactions, whereas the covalent ones formed through disulfide scrambling. Overall, the workflow presented here provides a powerful strategy for comprehensive characterization of peptide aggregates and understanding their mechanisms of formation.
Collapse
Affiliation(s)
- Tao Chen
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Shijia Tang
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Elizabeth S Hecht
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Chun-Wan Yen
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Nisana Andersen
- Protein Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Steven Chin
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Lance Cadang
- Protein Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Brian Roper
- Protein Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Alberto Estevez
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Alexis Rohou
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Debby Chang
- Department of Drug Delivery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Lu Dai
- Protein Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Peter Liu
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Mohammad Al-Sayah
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Karthik Nagapudi
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Fiona Lin
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Amin Famili
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Chloe Hu
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Robert Kuhn
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Cinzia Stella
- Protein Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Christopher M Crittenden
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Jason A Gruenhagen
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Cadapakam Venkatramani
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Dennis Leung
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Richard Vandlen
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Peter Yehl
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| |
Collapse
|
14
|
Shahin-Kaleybar B, Niazi A, Afsharifar A, Nematzadeh G, Yousefi R, Retzl B, Hellinger R, Muratspahić E, Gruber CW. Isolation of Cysteine-Rich Peptides from Citrullus colocynthis. Biomolecules 2020; 10:E1326. [PMID: 32948080 PMCID: PMC7565491 DOI: 10.3390/biom10091326] [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: 08/15/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
The plant Citrullus colocynthis, a member of the squash (Cucurbitaceae) family, has a long history in traditional medicine. Based on the ancient knowledge about the healing properties of herbal preparations, plant-derived small molecules, e.g., salicylic acid, or quinine, have been integral to modern drug discovery. Additionally, many plant families, such as Cucurbitaceae, are known as a rich source for cysteine-rich peptides, which are gaining importance as valuable pharmaceuticals. In this study, we characterized the C. colocynthis peptidome using chemical modification of cysteine residues, and mass shift analysis via matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry. We identified the presence of at least 23 cysteine-rich peptides in this plant, and eight novel peptides, named citcol-1 to -8, with a molecular weight between ~3650 and 4160 Da, were purified using reversed-phase high performance liquid chromatography (HPLC), and their amino acid sequences were determined by de novo assignment of b- and y-ion series of proteolytic peptide fragments. In silico analysis of citcol peptides revealed a high sequence similarity to trypsin inhibitor peptides from Cucumis sativus, Momordica cochinchinensis, Momordica macrophylla and Momordica sphaeroidea. Using genome/transcriptome mining it was possible to identify precursor sequences of this peptide family in related Cucurbitaceae species that cluster into trypsin inhibitor and antimicrobial peptides. Based on our analysis, the presence or absence of a crucial Arg/Lys residue at the putative P1 position may be used to classify these common cysteine-rich peptides by functional properties. Despite sequence homology and the common classification into the inhibitor cysteine knot family, these peptides appear to have diverse and additional bioactivities yet to be revealed.
Collapse
Affiliation(s)
- Behzad Shahin-Kaleybar
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (B.S.-K.); (B.R.); (R.H.); (E.M.)
- Department of Plant Biotechnology, Shiraz University, Shiraz 7144165186, Iran;
| | - Ali Niazi
- Department of Plant Biotechnology, Shiraz University, Shiraz 7144165186, Iran;
| | - Alireza Afsharifar
- Department of Plant Protection, Shiraz University, Shiraz 7144165186, Iran;
| | | | - Reza Yousefi
- Department of Biology, Shiraz University, Shiraz 7194684795, Iran;
| | - Bernhard Retzl
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (B.S.-K.); (B.R.); (R.H.); (E.M.)
| | - Roland Hellinger
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (B.S.-K.); (B.R.); (R.H.); (E.M.)
| | - Edin Muratspahić
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (B.S.-K.); (B.R.); (R.H.); (E.M.)
| | - Christian W. Gruber
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (B.S.-K.); (B.R.); (R.H.); (E.M.)
| |
Collapse
|
15
|
Pro-Inflammatory Signaling Upregulates a Neurotoxic Conotoxin-Like Protein Encrypted Within Human Endogenous Retrovirus-K. Cells 2020; 9:cells9071584. [PMID: 32629888 PMCID: PMC7407490 DOI: 10.3390/cells9071584] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/20/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
Motor neuron degeneration and spinal cord demyelination are hallmark pathological events in Amyotrophic Lateral Sclerosis (ALS). Endogenous retrovirus-K (ERVK) expression has an established association with ALS neuropathology, with murine modeling pointing to a role for the ERVK envelope (env) gene in disease processes. Here, we describe a novel viral protein cryptically encoded within the ERVK env transcript, which resembles two distinct cysteine-rich neurotoxic proteins: conotoxin proteins found in marine snails and the Human Immunodeficiency Virus (HIV) Tat protein. Consistent with Nuclear factor-kappa B (NF-κB)-induced retrotransposon expression, the ERVK conotoxin-like protein (CTXLP) is induced by inflammatory signaling. CTXLP is found in the nucleus, impacting innate immune gene expression and NF-κB p65 activity. Using human autopsy specimens from patients with ALS, we further showcase CTXLP expression in degenerating motor cortex and spinal cord tissues, concomitant with inflammation linked pathways, including enhancement of necroptosis marker mixed lineage kinase domain-like (MLKL) protein and oligodendrocyte maturation/myelination inhibitor Nogo-A. These findings identify CTXLP as a novel ERVK protein product, which may act as an effector in ALS neuropathology.
Collapse
|
16
|
Anti-tumoral effect of scorpion peptides: Emerging new cellular targets and signaling pathways. Cell Calcium 2019; 80:160-174. [DOI: 10.1016/j.ceca.2019.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/10/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022]
|
17
|
Arthropod venoms: Biochemistry, ecology and evolution. Toxicon 2018; 158:84-103. [PMID: 30529476 DOI: 10.1016/j.toxicon.2018.11.433] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 11/20/2018] [Accepted: 11/26/2018] [Indexed: 12/17/2022]
Abstract
Comprising of over a million described species of highly diverse invertebrates, Arthropoda is amongst the most successful animal lineages to have colonized aerial, terrestrial, and aquatic domains. Venom, one of the many fascinating traits to have evolved in various members of this phylum, has underpinned their adaptation to diverse habitats. Over millions of years of evolution, arthropods have evolved ingenious ways of delivering venom in their targets for self-defence and predation. The morphological diversity of venom delivery apparatus in arthropods is astounding, and includes extensively modified pedipalps, tail (telson), mouth parts (hypostome), fangs, appendages (maxillulae), proboscis, ovipositor (stinger), and hair (urticating bristles). Recent investigations have also unravelled an astonishing venom biocomplexity with molecular scaffolds being recruited from a multitude of protein families. Venoms are a remarkable bioresource for discovering lead compounds in targeted therapeutics. Several components with prospective applications in the development of advanced lifesaving drugs and environment friendly bio-insecticides have been discovered from arthropod venoms. Despite these fascinating features, the composition, bioactivity, and molecular evolution of venom in several arthropod lineages remains largely understudied. This review highlights the prevalence of venom, its mode of toxic action, and the evolutionary dynamics of venom in Arthropoda, the most speciose phylum in the animal kingdom.
Collapse
|
18
|
CPP-Ts: a new intracellular calcium channel modulator and a promising tool for drug delivery in cancer cells. Sci Rep 2018; 8:14739. [PMID: 30282983 PMCID: PMC6170434 DOI: 10.1038/s41598-018-33133-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/21/2018] [Indexed: 12/16/2022] Open
Abstract
Scorpion sting envenoming impacts millions of people worldwide, with cardiac effects being one of the main causes of death on victims. Here we describe the first Ca2+ channel toxin present in Tityus serrulatus (Ts) venom, a cell penetrating peptide (CPP) named CPP-Ts. We show that CPP-Ts increases intracellular Ca2+ release through the activation of nuclear InsP3R of cardiomyocytes, thereby causing an increase in the contraction frequency of these cells. Besides proposing a novel subfamily of Ca2+ active toxins, we investigated its potential use as a drug delivery system targeting cancer cell nucleus using CPP-Ts’s nuclear-targeting property. To this end, we prepared a synthetic CPP-Ts sub peptide14–39 lacking pharmacological activity which was directed to the nucleus of specific cancer cell lines. This research identifies a novel subfamily of Ca2+ active toxins and provides new insights into biotechnological applications of animal venoms.
Collapse
|
19
|
Romero-Gutiérrez MT, Santibáñez-López CE, Jiménez-Vargas JM, Batista CVF, Ortiz E, Possani LD. Transcriptomic and Proteomic Analyses Reveal the Diversity of Venom Components from the Vaejovid Scorpion Serradigitus gertschi. Toxins (Basel) 2018; 10:E359. [PMID: 30189638 PMCID: PMC6162517 DOI: 10.3390/toxins10090359] [Citation(s) in RCA: 23] [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: 08/10/2018] [Revised: 08/29/2018] [Accepted: 09/01/2018] [Indexed: 12/22/2022] Open
Abstract
To understand the diversity of scorpion venom, RNA from venomous glands from a sawfinger scorpion, Serradigitus gertschi, of the family Vaejovidae, was extracted and used for transcriptomic analysis. A total of 84,835 transcripts were assembled after Illumina sequencing. From those, 119 transcripts were annotated and found to putatively code for peptides or proteins that share sequence similarities with the previously reported venom components of other species. In accordance with sequence similarity, the transcripts were classified as potentially coding for 37 ion channel toxins; 17 host defense peptides; 28 enzymes, including phospholipases, hyaluronidases, metalloproteases, and serine proteases; nine protease inhibitor-like peptides; 10 peptides of the cysteine-rich secretory proteins, antigen 5, and pathogenesis-related 1 protein superfamily; seven La1-like peptides; and 11 sequences classified as "other venom components". A mass fingerprint performed by mass spectrometry identified 204 components with molecular masses varying from 444.26 Da to 12,432.80 Da, plus several higher molecular weight proteins whose precise masses were not determined. The LC-MS/MS analysis of a tryptic digestion of the soluble venom resulted in the de novo determination of 16,840 peptide sequences, 24 of which matched sequences predicted from the translated transcriptome. The database presented here increases our general knowledge of the biodiversity of venom components from neglected non-buthid scorpions.
Collapse
Affiliation(s)
- Maria Teresa Romero-Gutiérrez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Carlos Eduardo Santibáñez-López
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
- Department of Integrative Biology, University of Wisconsin⁻Madison, Madison, WI 53706, USA.
| | - Juana María Jiménez-Vargas
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Cesar Vicente Ferreira Batista
- Laboratorio Universitario de Proteómica, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Ernesto Ortiz
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Lourival Domingos Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| |
Collapse
|
20
|
The diversity of venom components of the scorpion species Paravaejovis schwenkmeyeri (Scorpiones: Vaejovidae) revealed by transcriptome and proteome analyses. Toxicon 2018; 151:47-62. [DOI: 10.1016/j.toxicon.2018.06.085] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
|
21
|
Structural diversity of arthropod venom toxins. Toxicon 2018; 152:46-56. [DOI: 10.1016/j.toxicon.2018.07.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/29/2018] [Accepted: 07/19/2018] [Indexed: 11/19/2022]
|
22
|
Möller C, Dovell S, Melaun C, Marí F. Definition of the R-superfamily of conotoxins: Structural convergence of helix-loop-helix peptidic scaffolds. Peptides 2018; 107:75-82. [PMID: 30040981 DOI: 10.1016/j.peptides.2018.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/13/2018] [Accepted: 06/18/2018] [Indexed: 10/28/2022]
Abstract
The F14 conotoxins define a four-cysteine, three-loop conotoxin scaffold that produce tightly folded structures held together by two disulfide bonds with a CCCC arrangement (conotoxin framework 14). Here we describe the precursors of the F14 conotoxins from the venom of Conus anabathrum and Conus villepinii. Using transcriptomic and cDNA cloning analysis, the full-length of the precursors of flf14a and flf14b from the transcriptome of C. anabathrum revealed a unique signal sequence that defines the new conotoxin R-superfamily. Using the signal sequence as a primer, we cloned seven additional previously undescribed toxins of the R-superfamily from C. villepinii. The propeptide regions of the R-conotoxins are unusually long and with prevalent proline residues in repeating pentads which qualifies them as Pro-rich motifs (PRMs), which can be critical for protein-protein interactions or they can be cleaved to release short linear peptides that may be part of the envenomation mélange. Additionally, we determined the three-dimensional structure of vil14a by solution 1H-NMR and found that the structure of this conotoxin displays a cysteine-stabilized α-helix-loop-helix (Cs α/α) fold. The structure is well-defined over the helical regions (backbone RMSD for residues 2-13 and 17-26 is 0.63 ± 0.14 Å), with conformational flexibility in the triple Gly region of the second loop as well as the N- and C- termini. Structurally, the F14 conotoxins overlap with the Cs α/α scorpion toxins and other peptidic natural products, and in spite of their different exogenomic origins, there is convergence into this scaffold from several classes of living organisms that express these peptides.
Collapse
Affiliation(s)
- Carolina Möller
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431-0991, USA
| | - Sanaz Dovell
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431-0991, USA
| | - Christian Melaun
- Justus Liebig Universität Giessen, Institut für Allg. Zoologie und Entwicklungsbiologie, Giessen, Germany
| | - Frank Marí
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431-0991, USA; Marine Biochemical Sciences, Chemical Sciences Division, National Institute of Standards and Technology, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC 29412, USA.
| |
Collapse
|
23
|
Diniz MRV, Paiva ALB, Guerra-Duarte C, Nishiyama MY, Mudadu MA, de Oliveira U, Borges MH, Yates JR, Junqueira-de-Azevedo IDL. An overview of Phoneutria nigriventer spider venom using combined transcriptomic and proteomic approaches. PLoS One 2018; 13:e0200628. [PMID: 30067761 PMCID: PMC6070231 DOI: 10.1371/journal.pone.0200628] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 06/29/2018] [Indexed: 01/23/2023] Open
Abstract
Phoneutria nigriventer is one of the largest existing true spiders and one of the few considered medically relevant. Its venom contains several neurotoxic peptides that act on different ion channels and chemical receptors of vertebrates and invertebrates. Some of these venom toxins have been shown as promising models for pharmaceutical or biotechnological use. However, the large diversity and the predominance of low molecular weight toxins in this venom have hampered the identification and deep investigation of the less abundant toxins and the proteins with high molecular weight. Here, we combined conventional and next-generation cDNA sequencing with Multidimensional Protein Identification Technology (MudPIT), to obtain an in-depth panorama of the composition of P. nigriventer spider venom. The results from these three approaches showed that cysteine-rich peptide toxins are the most abundant components in this venom and most of them contain the Inhibitor Cysteine Knot (ICK) structural motif. Ninety-eight sequences corresponding to cysteine-rich peptide toxins were identified by the three methodologies and many of them were considered as putative novel toxins, due to the low similarity to previously described toxins. Furthermore, using next-generation sequencing we identified families of several other classes of toxins, including CAPs (Cysteine Rich Secretory Protein-CRiSP, antigen 5 and Pathogenesis-Related 1-PR-1), serine proteinases, TCTPs (translationally controlled tumor proteins), proteinase inhibitors, metalloproteinases and hyaluronidases, which have been poorly described for this venom. This study provides an overview of the molecular diversity of P. nigriventer venom, revealing several novel components and providing a better basis to understand its toxicity and pharmacological activities.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Biomarkers, Tumor/chemistry
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- DNA, Complementary/metabolism
- High-Throughput Nucleotide Sequencing
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Peptides/metabolism
- Proteomics
- Sequence Alignment
- Sequence Analysis, DNA
- Spider Venoms/metabolism
- Spiders/genetics
- Spiders/metabolism
- Toxins, Biological/genetics
- Toxins, Biological/metabolism
- Transcriptome
- Tumor Protein, Translationally-Controlled 1
Collapse
Affiliation(s)
- Marcelo R. V. Diniz
- Laboratório de Toxinologia Molecular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Ana L. B. Paiva
- Laboratório de Toxinologia Molecular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Clara Guerra-Duarte
- Laboratório de Toxinologia Molecular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Milton Y. Nishiyama
- Laboratório Especial de Toxinologia Aplicada, CeTICS, Instituto Butantan, São Paulo, SP, Brazil
| | | | - Ursula de Oliveira
- Laboratório Especial de Toxinologia Aplicada, CeTICS, Instituto Butantan, São Paulo, SP, Brazil
| | - Márcia H. Borges
- Laboratório de Toxinologia Molecular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - John R. Yates
- Department of Chemical Physiology and Molecular and Cellular Neurobiology, The Scripps Research Institute, La Jolla, California, United States of America
| | | |
Collapse
|
24
|
Zhao J, Yuan S, Gao B, Zhu S. Molecular diversity of fungal inhibitor cystine knot peptides evolved by domain repeat and fusion. FEMS Microbiol Lett 2018; 365:5046422. [PMID: 29961831 DOI: 10.1093/femsle/fny158] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/26/2018] [Indexed: 12/23/2022] Open
Affiliation(s)
- Jingru Zhao
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Shouli Yuan
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Bin Gao
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Shunyi Zhu
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| |
Collapse
|
25
|
Patel S, Barkell AM, Gupta D, Strong SL, Bruton S, Muskett FW, Addis PW, Renshaw PS, Slocombe PM, Doyle C, Clargo A, Taylor RJ, Prosser CE, Henry AJ, Robinson MK, Waters LC, Holdsworth G, Carr MD. Structural and functional analysis of Dickkopf 4 (Dkk4): New insights into Dkk evolution and regulation of Wnt signaling by Dkk and Kremen proteins. J Biol Chem 2018; 293:12149-12166. [PMID: 29925589 PMCID: PMC6078440 DOI: 10.1074/jbc.ra118.002918] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/11/2018] [Indexed: 11/06/2022] Open
Abstract
Dickkopf (Dkk) family proteins are important regulators of Wnt signaling pathways, which play key roles in many essential biological processes. Here, we report the first detailed structural and dynamics study of a full-length mature Dkk protein (Dkk4, residues 19–224), including determination of the first atomic-resolution structure for the N-terminal cysteine-rich domain (CRD1) conserved among Dkk proteins. We discovered that CRD1 has significant structural homology to the Dkk C-terminal cysteine-rich domain (CRD2), pointing to multiple gene duplication events during Dkk family evolution. We also show that Dkk4 consists of two independent folded domains (CRD1 and CRD2) joined by a highly flexible, nonstructured linker. Similarly, the N-terminal region preceding CRD1 and containing a highly conserved NXI(R/K) sequence motif was shown to be dynamic and highly flexible. We demonstrate that Dkk4 CRD2 mediates high-affinity binding to both the E1E2 region of low-density lipoprotein receptor–related protein 6 (LRP6 E1E2) and the Kremen1 (Krm1) extracellular domain. In contrast, the N-terminal region alone bound with only moderate affinity to LRP6 E1E2, consistent with binding via the conserved NXI(R/K) motif, but did not interact with Krm proteins. We also confirmed that Dkk and Krm family proteins function synergistically to inhibit Wnt signaling. Insights provided by our integrated structural, dynamics, interaction, and functional studies have allowed us to refine the model of synergistic regulation of Wnt signaling by Dkk proteins. Our results indicate the potential for the formation of a diverse range of ternary complexes comprising Dkk, Krm, and LRP5/6 proteins, allowing fine-tuning of Wnt-dependent signaling.
Collapse
Affiliation(s)
- Saleha Patel
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Alice M Barkell
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Deepti Gupta
- UCB, 208 Bath Road, Slough SL1 3WE, United Kingdom
| | - Sarah L Strong
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Shaun Bruton
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Frederick W Muskett
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Philip W Addis
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Philip S Renshaw
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | | | - Carl Doyle
- UCB, 208 Bath Road, Slough SL1 3WE, United Kingdom
| | | | | | - Christine E Prosser
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; UCB, 208 Bath Road, Slough SL1 3WE, United Kingdom
| | | | | | - Lorna C Waters
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom.
| | | | - Mark D Carr
- Leicester Institute of Structural and Chemical Biology, Lancaster Road, Leicester LE1 7HB, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom.
| |
Collapse
|
26
|
Classes, Databases, and Prediction Methods of Pharmaceutically and Commercially Important Cystine-Stabilized Peptides. Toxins (Basel) 2018; 10:toxins10060251. [PMID: 29921767 PMCID: PMC6024828 DOI: 10.3390/toxins10060251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Abstract
Cystine-stabilized peptides represent a large family of peptides characterized by high structural stability and bactericidal, fungicidal, or insecticidal properties. Found throughout a wide range of taxa, this broad and functionally important family can be subclassified into distinct groups dependent upon their number and type of cystine bonding patters, tertiary structures, and/or their species of origin. Furthermore, the annotation of proteins related to the cystine-stabilized family are under-represented in the literature due to their difficulty of isolation and identification. As a result, there are several recent attempts to collate them into data resources and build analytic tools for their dynamic prediction. Ultimately, the identification and delivery of new members of this family will lead to their growing inclusion into the repertoire of commercial viable alternatives to antibiotics and environmentally safe insecticides. This review of the literature and current state of cystine-stabilized peptide biology is aimed to better describe peptide subfamilies, identify databases and analytics resources associated with specific cystine-stabilized peptides, and highlight their current commercial success.
Collapse
|
27
|
dos Santos-Pinto JRA, Perez-Riverol A, Lasa AM, Palma MS. Diversity of peptidic and proteinaceous toxins from social Hymenoptera venoms. Toxicon 2018; 148:172-196. [DOI: 10.1016/j.toxicon.2018.04.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 12/20/2022]
|
28
|
Abstract
Antibodies have proved to be a valuable mode of therapy for numerous diseases, mainly owing to their high target binding affinity and specificity. Unfortunately, antibodies are also limited in several respects, chief amongst those being the extremely high cost of manufacture. Therefore, non-antibody binding proteins have long been sought after as alternative therapies. New binding protein scaffolds are constantly being designed or discovered with some already approved for human use by the FDA. This review focuses on protein scaffolds that are either already being used in humans or are currently being evaluated in clinical trials. Although not all are expected to be approved, the significant benefits ensure that these molecules will continue to be investigated and developed as therapeutic alternatives to antibodies. Based on the location of the amino acids that mediate ligand binding, we place all the protein scaffolds under clinical development into two general categories: scaffolds with ligand-binding residues located in exposed flexible loops, and those with the binding residues located in protein secondary structures, such as α-helices. Scaffolds that fall under the first category include adnectins, anticalins, avimers, Fynomers, Kunitz domains, and knottins, while those belonging to the second category include affibodies, β-hairpin mimetics, and designed ankyrin repeat proteins (DARPins). Most of these scaffolds are thermostable and can be easily produced in microorganisms or completely synthesized chemically. In addition, many of these scaffolds derive from human proteins and thus possess very low immunogenic potential. Additional advantages and limitations of these protein scaffolds as therapeutics compared to antibodies will be discussed.
Collapse
Affiliation(s)
- Rudo Simeon
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, TX, 77845, USA
| | - Zhilei Chen
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, TX, 77845, USA.
| |
Collapse
|
29
|
Geron M, Hazan A, Priel A. Animal Toxins Providing Insights into TRPV1 Activation Mechanism. Toxins (Basel) 2017; 9:toxins9100326. [PMID: 29035314 PMCID: PMC5666373 DOI: 10.3390/toxins9100326] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 02/06/2023] Open
Abstract
Beyond providing evolutionary advantages, venoms offer unique research tools, as they were developed to target functionally important proteins and pathways. As a key pain receptor in the nociceptive pathway, transient receptor potential vanilloid 1 (TRPV1) of the TRP superfamily has been shown to be a target for several toxins, as a way of producing pain to deter predators. Importantly, TRPV1 is involved in thermoregulation, inflammation, and acute nociception. As such, toxins provide tools to understand TRPV1 activation and modulation, a critical step in advancing pain research and the development of novel analgesics. Indeed, the phytotoxin capsaicin, which is the spicy chemical in chili peppers, was invaluable in the original cloning and characterization of TRPV1. The unique properties of each subsequently characterized toxin have continued to advance our understanding of functional, structural, and biophysical characteristics of TRPV1. By building on previous reviews, this work aims to provide a comprehensive summary of the advancements made in TRPV1 research in recent years by employing animal toxins, in particular DkTx, RhTx, BmP01, Echis coloratus toxins, APHCs and HCRG21. We examine each toxin’s functional aspects, behavioral effects, and structural features, all of which have contributed to our current knowledge of TRPV1. We additionally discuss the key features of TRPV1’s outer pore domain, which proves to be the target of the currently discussed toxins.
Collapse
Affiliation(s)
- Matan Geron
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Adina Hazan
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Avi Priel
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| |
Collapse
|
30
|
Xiao L, Gurrola GB, Zhang J, Valdivia CR, SanMartin M, Zamudio FZ, Zhang L, Possani LD, Valdivia HH. Structure-function relationships of peptides forming the calcin family of ryanodine receptor ligands. J Gen Physiol 2017; 147:375-94. [PMID: 27114612 PMCID: PMC4845687 DOI: 10.1085/jgp.201511499] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 03/21/2016] [Indexed: 12/22/2022] Open
Abstract
Calcins are a novel family of scorpion peptides that bind with high affinity to ryanodine receptors (RyRs) and increase their activity by inducing subconductance states. Here, we provide a comprehensive analysis of the structure-function relationships of the eight calcins known to date, based on their primary sequence, three-dimensional modeling, and functional effects on skeletal RyRs (RyR1). Primary sequence alignment and evolutionary analysis show high similarity among all calcins (≥78.8% identity). Other common characteristics include an inhibitor cysteine knot (ICK) motif stabilized by three pairs of disulfide bridges and a dipole moment (DM) formed by positively charged residues clustering on one side of the molecule and neutral and negatively charged residues segregating on the opposite side. [(3)H]Ryanodine binding assays, used as an index of the open probability of RyRs, reveal that all eight calcins activate RyR1 dose-dependently with Kd values spanning approximately three orders of magnitude and in the following rank order: opicalcin1 > opicalcin2 > vejocalcin > hemicalcin > imperacalcin > hadrucalcin > maurocalcin >> urocalcin. All calcins significantly augment the bell-shaped [Ca(2+)]-[(3)H]ryanodine binding curve with variable effects on the affinity constants for Ca(2+) activation and inactivation. In single channel recordings, calcins induce the appearance of a subconductance state in RyR1 that has a unique fractional value (∼20% to ∼60% of the full conductance state) but bears no relationship to binding affinity, DM, or capacity to stimulate Ca(2+) release. Except for urocalcin, all calcins at 100 nM concentration stimulate Ca(2+) release and deplete Ca(2+) load from skeletal sarcoplasmic reticulum. The natural variation within the calcin family of peptides offers a diversified set of high-affinity ligands with the capacity to modulate RyRs with high dynamic range and potency.
Collapse
Affiliation(s)
- Liang Xiao
- Department of Marine Biotechnology, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China Center for Arrhythmia Research, Cardiovascular Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Georgina B Gurrola
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62271, México Center for Arrhythmia Research, Cardiovascular Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Jing Zhang
- Center for Arrhythmia Research, Cardiovascular Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Carmen R Valdivia
- Center for Arrhythmia Research, Cardiovascular Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Mario SanMartin
- Center for Arrhythmia Research, Cardiovascular Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Fernando Z Zamudio
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62271, México Center for Arrhythmia Research, Cardiovascular Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Liming Zhang
- Department of Marine Biotechnology, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| | - Lourival D Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62271, México
| | - Héctor H Valdivia
- Center for Arrhythmia Research, Cardiovascular Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
31
|
Ravipati AS, Poth AG, Troeira Henriques S, Bhandari M, Huang YH, Nino J, Colgrave ML, Craik DJ. Understanding the Diversity and Distribution of Cyclotides from Plants of Varied Genetic Origin. JOURNAL OF NATURAL PRODUCTS 2017; 80:1522-1530. [PMID: 28471681 DOI: 10.1021/acs.jnatprod.7b00061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cyclotides are a large family of naturally occurring plant-derived macrocyclic cystine-knot peptides, with more than 400 having been identified in species from the Violaceae, Rubiaceae, Cucurbitaceae, Fabaceae, and Solanaceae families. Nevertheless, their specialized distribution within the plant kingdom remains poorly understood. In this study, the diversity of cyclotides was explored through the screening of 197 plants belonging to 43 different families. In total, 28 cyclotides were sequenced from 15 plant species, one of which belonged to the Rubiaceae and 14 to the Violaceae. Every Violaceae species screened contained cyclotides, but they were only sparsely represented in Rubiaceae and nonexistent in other families. The study thus supports the hypothesis that cyclotides are ubiquitous in the Violaceae, and it adds to the list of plants found to express kalata S and cycloviolacin O12. Finally, previous studies suggested the existence of cyclotide isoforms with either an Asn or an Asp at the C-terminal processing site of the cyclotide domain within the precursor proteins. Here we found that despite the discovery of a few cyclotides genuinely containing an Asp in loop 6 as evidenced by gene sequencing, deamidation of Asn during enzymatic digestion resulted in the artifactual presence of Asp isoforms. This result is consistent with studies suggesting that peptides can undergo deamidation after being subjected to external factors, including pH, temperature, and enzymatic digestion.
Collapse
Affiliation(s)
- Anjaneya S Ravipati
- Institute for Molecular Bioscience, The University of Queensland , Brisbane 4072, Queensland Australia
| | - Aaron G Poth
- Institute for Molecular Bioscience, The University of Queensland , Brisbane 4072, Queensland Australia
| | - Sónia Troeira Henriques
- Institute for Molecular Bioscience, The University of Queensland , Brisbane 4072, Queensland Australia
| | - Murari Bhandari
- Institute for Molecular Bioscience, The University of Queensland , Brisbane 4072, Queensland Australia
| | - Yen-Hua Huang
- Institute for Molecular Bioscience, The University of Queensland , Brisbane 4072, Queensland Australia
| | - Jaime Nino
- Universidad Tecnológica de Pereira , Cra 27 No 10-02-Los Álamos, Pereira, Risaralda, Colombia
| | - Michelle L Colgrave
- Commonwealth Scientific and Industrial Research Organization, Agriculture and Food, St Lucia 4067, Queensland, Australia
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland , Brisbane 4072, Queensland Australia
| |
Collapse
|
32
|
Oldrati V, Koua D, Allard PM, Hulo N, Arrell M, Nentwig W, Lisacek F, Wolfender JL, Kuhn-Nentwig L, Stöcklin R. Peptidomic and transcriptomic profiling of four distinct spider venoms. PLoS One 2017; 12:e0172966. [PMID: 28306751 PMCID: PMC5357004 DOI: 10.1371/journal.pone.0172966] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/13/2017] [Indexed: 11/18/2022] Open
Abstract
Venom based research is exploited to find novel candidates for the development of innovative pharmacological tools, drug candidates and new ingredients for cosmetic and agrochemical industries. Moreover, venomics, as a well-established approach in systems biology, helps to elucidate the genetic mechanisms of the production of such a great molecular biodiversity. Today the advances made in the proteomics, transcriptomics and bioinformatics fields, favor venomics, allowing the in depth study of complex matrices and the elucidation even of minor compounds present in minute biological samples. The present study illustrates a rapid and efficient method developed for the elucidation of venom composition based on NextGen mRNA sequencing of venom glands and LC-MS/MS venom proteome profiling. The analysis of the comprehensive data obtained was focused on cysteine rich peptide toxins from four spider species originating from phylogenetically distant families for comparison purposes. The studied species were Heteropoda davidbowie (Sparassidae), Poecilotheria formosa (Theraphosidae), Viridasius fasciatus (Viridasiidae) and Latrodectus mactans (Theridiidae). This led to a high resolution profiling of 284 characterized cysteine rich peptides, 111 of which belong to the Inhibitor Cysteine Knot (ICK) structural motif. The analysis of H. davidbowie venom revealed a high richness in term of venom diversity: 95 peptide sequences were identified; out of these, 32 peptides presented the ICK structural motif and could be classified in six distinct families. The profiling of P. formosa venom highlighted the presence of 126 peptide sequences, with 52 ICK toxins belonging to three structural distinct families. V. fasciatus venom was shown to contain 49 peptide sequences, out of which 22 presented the ICK structural motif and were attributed to five families. The venom of L. mactans, until now studied for its large neurotoxins (Latrotoxins), revealed the presence of 14 cysteine rich peptides, out of which five were ICK toxins belonging to the CSTX superfamily. This in depth profiling of distinct ICK peptide families identified across the four spider species highlighted the high conservation of these neurotoxins among spider families.
Collapse
Affiliation(s)
- Vera Oldrati
- Atheris SA, Chemin d’Alcire 1, Plan-les-Ouates, Geneva, Switzerland
- School of Pharmaceutical Sciences, EPGL, University of Geneva, University of Lausanne, 1, Rue Michel-Servet, Geneva 4, Switzerland
- * E-mail:
| | - Dominique Koua
- Atheris SA, Chemin d’Alcire 1, Plan-les-Ouates, Geneva, Switzerland
| | - Pierre-Marie Allard
- School of Pharmaceutical Sciences, EPGL, University of Geneva, University of Lausanne, 1, Rue Michel-Servet, Geneva 4, Switzerland
| | - Nicolas Hulo
- University of Geneva, CMU, 1, Rue Michel Servet, Geneva 4, Switzerland
- Atheris Laboratories, Chemin d’Alcire 1, Plan-les-Ouates, Geneva, Switzerland
| | - Miriam Arrell
- Atheris SA, Chemin d’Alcire 1, Plan-les-Ouates, Geneva, Switzerland
| | - Wolfgang Nentwig
- University of Bern, Institute of Ecology and Evolution, 6, Baltzerstrasse, Bern, Switzerland
| | - Frédérique Lisacek
- University of Geneva, CMU, 1, Rue Michel Servet, Geneva 4, Switzerland
- SIB Swiss Institute of Bioinformatics, CUI, 7, Route de Drize, Geneva, Switzerland
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, EPGL, University of Geneva, University of Lausanne, 1, Rue Michel-Servet, Geneva 4, Switzerland
| | - Lucia Kuhn-Nentwig
- University of Bern, Institute of Ecology and Evolution, 6, Baltzerstrasse, Bern, Switzerland
| | - Reto Stöcklin
- Atheris SA, Chemin d’Alcire 1, Plan-les-Ouates, Geneva, Switzerland
| |
Collapse
|
33
|
Vargas-Jaimes L, Xiao L, Zhang J, Possani LD, Valdivia HH, Quintero-Hernández V. Recombinant expression of Intrepicalcin from the scorpion Vaejovis intrepidus and its effect on skeletal ryanodine receptors. Biochim Biophys Acta Gen Subj 2017; 1861:936-946. [PMID: 28159581 DOI: 10.1016/j.bbagen.2017.01.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 01/22/2017] [Accepted: 01/30/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Scorpion venoms contain toxins that modulate ionic channels, among which are the calcins, a small group of short, basic peptides with an Inhibitor Cystine Knot (ICK) motif that target calcium release channels/ryanodine receptors (RyRs) with high affinity and selectivity. Here we describe the heterologous expression of Intrepicalcin, identified by transcriptomic analysis of venomous glands from Vaejovis intrepidus. METHODS Recombinant Intrepicalcin was obtained in Escherichia coli BL21-DE3 (periplasm) by fusing the Intrepicalcin gene to sequences coding for signal-peptide, thioredoxin, His-tag and enterokinase cleavage site. RESULTS [3H]Ryanodine binding, used as a functional index of RyR activity, revealed that recombinant Intrepicalcin activates skeletal RyR (RyR1) dose-dependently with Kd=17.4±4.0nM. Intrepicalcin significantly augments the bell-shaped [Ca2+]-[3H]ryanodine binding curve at all [Ca2+] ranges, as is characteristic of the calcins. In single channel recordings, Intrepicalcin induces the appearance of a subconductance state in RyR1 with a fractional value ∼55% of the full conductance state, very close to that of Vejocalcin. Furthermore, Intrepicalcin stimulates Ca2+ release at an initial dose=45.3±2.5nM, and depletes ~50% of Ca2+ load from skeletal sarcoplasmic reticulum vesicles. CONCLUSIONS We conclude that active recombinant Intrepicalcin was successfully obtained without the need of manual oxidation, enabling it to target RyR1s with high affinity. GENERAL SIGNIFICANCE This is the first calcin heterologously expressed in the periplasma of Escherichia coli BL21-DE3, shown to be pharmacologically effective, thus paving the way for the generation of Intrepicalcin variants that are required for structure-function relationship studies of calcins and RyRs.
Collapse
Affiliation(s)
- Leonel Vargas-Jaimes
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62271, México
| | - Liang Xiao
- Department of Marine Biotechnology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China.,Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jing Zhang
- Department of Marine Biotechnology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Lourival D Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62271, México
| | - Héctor H Valdivia
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Verónica Quintero-Hernández
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62271, México.,CONACYT- Laboratorio de Ecología Molecular Microbiana, Centro de Investigaciones en Ciencias Microbiológicas-Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, C.P. 72570, Puebla, México
| |
Collapse
|
34
|
Park S, Yoo KO, Marcussen T, Backlund A, Jacobsson E, Rosengren KJ, Doo I, Göransson U. Cyclotide Evolution: Insights from the Analyses of Their Precursor Sequences, Structures and Distribution in Violets ( Viola). FRONTIERS IN PLANT SCIENCE 2017; 8:2058. [PMID: 29326730 PMCID: PMC5741643 DOI: 10.3389/fpls.2017.02058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/17/2017] [Indexed: 05/20/2023]
Abstract
Cyclotides are a family of plant proteins that are characterized by a cyclic backbone and a knotted disulfide topology. Their cyclic cystine knot (CCK) motif makes them exceptionally resistant to thermal, chemical, and enzymatic degradation. By disrupting cell membranes, the cyclotides function as host defense peptides by exhibiting insecticidal, anthelmintic, antifouling, and molluscicidal activities. In this work, we provide the first insight into the evolution of this family of plant proteins by studying the Violaceae, in particular species of the genus Viola. We discovered 157 novel precursor sequences by the transcriptomic analysis of six Viola species: V. albida var. takahashii, V. mandshurica, V. orientalis, V. verecunda, V. acuminata, and V. canadensis. By combining these precursor sequences with the phylogenetic classification of Viola, we infer the distribution of cyclotides across 63% of the species in the genus (i.e., ~380 species). Using full precursor sequences from transcriptomes, we show an evolutionary link to the structural diversity of the cyclotides, and further classify the cyclotides by sequence signatures from the non-cyclotide domain. Also, transcriptomes were compared to cyclotide expression on a peptide level determined using liquid chromatography-mass spectrometry. Furthermore, the novel cyclotides discovered were associated with the emergence of new biological functions.
Collapse
Affiliation(s)
- Sungkyu Park
- Division of Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ki-Oug Yoo
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea
| | - Thomas Marcussen
- Department of Biosciences, Centre for Ecological and Evolutionary Synthesis, University of Oslo, Oslo, Norway
| | - Anders Backlund
- Division of Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Erik Jacobsson
- Division of Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - K. Johan Rosengren
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Inseok Doo
- Biotech Research Team, Biotech Research Center of Dong-A Pharm Co Ltd., Seoul, South Korea
| | - Ulf Göransson
- Division of Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- *Correspondence: Ulf Göransson
| |
Collapse
|
35
|
Wang X, Gao B, Zhu S. Exon Shuffling and Origin of Scorpion Venom Biodiversity. Toxins (Basel) 2016; 9:toxins9010010. [PMID: 28035955 PMCID: PMC5308243 DOI: 10.3390/toxins9010010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/13/2016] [Accepted: 12/21/2016] [Indexed: 12/01/2022] Open
Abstract
Scorpion venom is a complex combinatorial library of peptides and proteins with multiple biological functions. A combination of transcriptomic and proteomic techniques has revealed its enormous molecular diversity, as identified by the presence of a large number of ion channel-targeted neurotoxins with different folds, membrane-active antimicrobial peptides, proteases, and protease inhibitors. Although the biodiversity of scorpion venom has long been known, how it arises remains unsolved. In this work, we analyzed the exon-intron structures of an array of scorpion venom protein-encoding genes and unexpectedly found that nearly all of these genes possess a phase-1 intron (one intron located between the first and second nucleotides of a codon) near the cleavage site of a signal sequence despite their mature peptides remarkably differ. This observation matches a theory of exon shuffling in the origin of new genes and suggests that recruitment of different folds into scorpion venom might be achieved via shuffling between body protein-coding genes and ancestral venom gland-specific genes that presumably contributed tissue-specific regulatory elements and secretory signal sequences.
Collapse
Affiliation(s)
- Xueli Wang
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects & Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China.
| | - Bin Gao
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects & Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China.
| | - Shunyi Zhu
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects & Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China.
| |
Collapse
|
36
|
Venom Gland Transcriptomic and Proteomic Analyses of the Enigmatic Scorpion Superstitionia donensis (Scorpiones: Superstitioniidae), with Insights on the Evolution of Its Venom Components. Toxins (Basel) 2016; 8:toxins8120367. [PMID: 27941686 PMCID: PMC5198561 DOI: 10.3390/toxins8120367] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/28/2016] [Accepted: 12/01/2016] [Indexed: 11/28/2022] Open
Abstract
Venom gland transcriptomic and proteomic analyses have improved our knowledge on the diversity of the heterogeneous components present in scorpion venoms. However, most of these studies have focused on species from the family Buthidae. To gain insights into the molecular diversity of the venom components of scorpions belonging to the family Superstitioniidae, one of the neglected scorpion families, we performed a transcriptomic and proteomic analyses for the species Superstitionia donensis. The total mRNA extracted from the venom glands of two specimens was subjected to massive sequencing by the Illumina protocol, and a total of 219,073 transcripts were generated. We annotated 135 transcripts putatively coding for peptides with identity to known venom components available from different protein databases. Fresh venom collected by electrostimulation was analyzed by LC-MS/MS allowing the identification of 26 distinct components with sequences matching counterparts from the transcriptomic analysis. In addition, the phylogenetic affinities of the found putative calcins, scorpines, La1-like peptides and potassium channel κ toxins were analyzed. The first three components are often reported as ubiquitous in the venom of different families of scorpions. Our results suggest that, at least calcins and scorpines, could be used as molecular markers in phylogenetic studies of scorpion venoms.
Collapse
|
37
|
Cremonez CM, Maiti M, Peigneur S, Cassoli JS, Dutra AAA, Waelkens E, Lescrinier E, Herdewijn P, de Lima ME, Pimenta AMC, Arantes EC, Tytgat J. Structural and Functional Elucidation of Peptide Ts11 Shows Evidence of a Novel Subfamily of Scorpion Venom Toxins. Toxins (Basel) 2016; 8:toxins8100288. [PMID: 27706049 PMCID: PMC5086648 DOI: 10.3390/toxins8100288] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/25/2016] [Accepted: 09/26/2016] [Indexed: 12/31/2022] Open
Abstract
To date, several families of peptide toxins specifically interacting with ion channels in scorpion venom have been described. One of these families comprise peptide toxins (called KTxs), known to modulate potassium channels. Thus far, 202 KTxs have been reported, belonging to several subfamilies of KTxs (called α, β, γ, κ, δ, and λ-KTxs). Here we report on a previously described orphan toxin from Tityus serrulatus venom, named Ts11. We carried out an in-depth structure-function analysis combining 3D structure elucidation of Ts11 and electrophysiological characterization of the toxin. The Ts11 structure is highlighted by an Inhibitor Cystine Knot (ICK) type scaffold, completely devoid of the classical secondary structure elements (α-helix and/or β-strand). This has, to the best of our knowledge, never been described before for scorpion toxins and therefore represents a novel, 6th type of structural fold for these scorpion peptides. On the basis of their preferred interaction with voltage-gated K channels, as compared to all the other targets tested, it can be postulated that Ts11 is the first member of a new subfamily, designated as ε-KTx.
Collapse
Affiliation(s)
- Caroline M Cremonez
- Laboratório de Toxinas Animais, Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto 14040-903, São Paulo, Brasil.
| | - Mohitosh Maiti
- Laboratory for Medicinal Chemistry, Rega Institute for Medical Research, University of Leuven (KU Leuven), P.O. Box 922, Leuven 3000, Belgium.
| | - Steve Peigneur
- Toxicology & Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg O&N2, P.O. Box 922, Leuven 3000, Belgium.
| | - Juliana Silva Cassoli
- Laboratório de Venenos e Toxinas Animais, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brasil.
| | - Alexandre A A Dutra
- Laboratório de Venenos e Toxinas Animais, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brasil.
| | - Etienne Waelkens
- Laboratory of Protein Phosphorylation and Proteomics, University of Leuven (KU Leuven), P.O. Box 922, Leuven 3000, Belgium.
| | - Eveline Lescrinier
- Laboratory for Medicinal Chemistry, Rega Institute for Medical Research, University of Leuven (KU Leuven), P.O. Box 922, Leuven 3000, Belgium.
| | - Piet Herdewijn
- Laboratory for Medicinal Chemistry, Rega Institute for Medical Research, University of Leuven (KU Leuven), P.O. Box 922, Leuven 3000, Belgium.
| | - Maria Elena de Lima
- Laboratório de Venenos e Toxinas Animais, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brasil.
| | - Adriano M C Pimenta
- Laboratório de Venenos e Toxinas Animais, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brasil.
| | - Eliane C Arantes
- Laboratório de Toxinas Animais, Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto 14040-903, São Paulo, Brasil.
| | - Jan Tytgat
- Toxicology & Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg O&N2, P.O. Box 922, Leuven 3000, Belgium.
| |
Collapse
|
38
|
Kintzing JR, Cochran JR. Engineered knottin peptides as diagnostics, therapeutics, and drug delivery vehicles. Curr Opin Chem Biol 2016; 34:143-150. [PMID: 27642714 DOI: 10.1016/j.cbpa.2016.08.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 12/18/2022]
Abstract
Inhibitor cystine-knots, also known as knottins, are a structural family of ultra-stable peptides with diverse functions. Knottins and related backbone-cyclized peptides called cyclotides contain three disulfide bonds connected in a particular arrangement that endows these peptides with high thermal, proteolytic, and chemical stability. Knottins have gained interest as candidates for non-invasive molecular imaging and for drug development as they can possess the pharmacological properties of small molecules and the target affinity and selectively of protein biologics. Naturally occurring knottins are clinically approved for treating chronic pain and GI disorders. Combinatorial methods are being used to engineer knottins that can bind to other clinically relevant targets in cancer, and inflammatory and cardiac disease. This review details recent examples of engineered knottin peptides; their use as molecular imaging agents, therapeutics, and drug delivery vehicles; modifications that can be introduced to improve peptide folding and bioactivity; and future perspectives and challenges in the field.
Collapse
Affiliation(s)
- James R Kintzing
- Department of Bioengineering, Stanford University, United States
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, United States; Department of Chemical Engineering, Stanford University, United States.
| |
Collapse
|
39
|
Kwon S, Bosmans F, Kaas Q, Cheneval O, Conibear AC, Rosengren KJ, Wang CK, Schroeder CI, Craik DJ. Efficient enzymatic cyclization of an inhibitory cystine knot-containing peptide. Biotechnol Bioeng 2016; 113:2202-12. [PMID: 27093300 DOI: 10.1002/bit.25993] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/04/2016] [Accepted: 04/11/2016] [Indexed: 01/01/2023]
Abstract
Disulfide-rich peptides isolated from cone snails are of great interest as drug leads due to their high specificity and potency toward therapeutically relevant ion channels and receptors. They commonly contain the inhibitor cystine knot (ICK) motif comprising three disulfide bonds forming a knotted core. Here we report the successful enzymatic backbone cyclization of an ICK-containing peptide κ-PVIIA, a 27-amino acid conopeptide from Conus purpurascens, using a mutated version of the bacterial transpeptidase, sortase A. Although a slight loss of activity was observed compared to native κ-PVIIA, cyclic κ-PVIIA is a functional peptide that inhibits the Shaker voltage-gated potassium (Kv) channel. Molecular modeling suggests that the decrease in potency may be related to the loss of crucial, but previously unidentified electrostatic interactions between the N-terminus of the peptide and the Shaker channel. This hypothesis was confirmed by testing an N-terminally acetylated κ-PVIIA, which shows a similar decrease in activity. We also investigated the conformational dynamics and hydrogen bond network of cyc-PVIIA, both of which are important factors to be considered for successful cyclization of peptides. We found that cyc-PVIIA has the same conformational dynamics, but different hydrogen bond network compared to those of κ-PVIIA. The ability to efficiently cyclize ICK peptides using sortase A will enable future protein engineering for this class of peptides and may help in the development of novel therapeutic molecules. Biotechnol. Bioeng. 2016;113: 2202-2212. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Soohyun Kwon
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld, 4072, Australia
| | - Frank Bosmans
- Department of Physiology and Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Quentin Kaas
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld, 4072, Australia
| | - Olivier Cheneval
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld, 4072, Australia
| | - Anne C Conibear
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld, 4072, Australia
| | - K Johan Rosengren
- The University of Queensland, School of Biomedical Sciences, Brisbane, Qld, Australia
| | - Conan K Wang
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld, 4072, Australia
| | - Christina I Schroeder
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld, 4072, Australia.
| | - David J Craik
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld, 4072, Australia.
| |
Collapse
|
40
|
Zhong J, Zeng XC, Zeng X, Nie Y, Zhang L, Wu S, Bao A. Transcriptomic analysis of the venom glands from the scorpion Hadogenes troglodytes revealed unique and extremely high diversity of the venom peptides. J Proteomics 2016; 150:40-62. [PMID: 27519694 DOI: 10.1016/j.jprot.2016.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/25/2016] [Accepted: 08/06/2016] [Indexed: 12/14/2022]
Abstract
Hadogenes is a genus of large African scorpions with 18 described species. However, little is known about the venom peptide composition of any species from Hadogenes so far. Here, we fully explored the composition of venom gland peptides from Hadogenes troglodytes using transcriptomic approach. We discovered 121 novel peptides from the scorpion, including 20 new-type peptides cross-linked with one, two, three, four or seven disulfide bridges, respectively, 11 novel K+-channel toxin-like peptides, 2 novel ryanodine receptors-specific toxin-like peptides, a unique peptide containing the cysteine knots of spider toxins, 15 novel La1-like toxins, 3 novel TIL domain-containing peptides, 5 novel peptides with atypical cysteine patterns, 19 novel antimicrobial peptides, 6 novel cysteine-free peptides and 39 new-type cysteine-free peptides. Among them, the new-type peptides are largely dominant; this highlights the unique diversity of the venom gland peptides from H. troglodytes. Some of the new peptides would serve as new molecular probes for the investigations of cellular ion channels and other receptors, or offer new templates for the development of therapeutic drugs for the treatment of ion channel-associated diseases, and infections caused by antibiotics-resistant pathogens. BIOLOGICAL SIGNIFICANCE In this study, we fully explored the composition of venom gland peptides from the scorpion Hadogenes troglodytes using transcriptomic approach. We discovered a total of 121 novel peptides from the venom glands of the scorpion, of which new-type peptides are largely dominant. These data highlight the unique diversity of the venom gland peptides from the scorpion H. troglodytes, gain insights into new mechanisms for the scorpion to subdue its prey and predators, and enlarge the protein database of scorpion venom glands. The discovery of a lot of novel peptides provides new templates for the development of therapeutic drugs, and offers new molecular materials for the basic researches of various cellular receptors, and for the evolutionary investigations of scorpion toxins.
Collapse
Affiliation(s)
- Jie Zhong
- Department of Biological Science and Technology, School of Environmental Studies & State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences, Wuhan 430074, PR China
| | - Xian-Chun Zeng
- Department of Biological Science and Technology, School of Environmental Studies & State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences, Wuhan 430074, PR China.
| | - Xin Zeng
- Department of Biological Science and Technology, School of Environmental Studies & State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences, Wuhan 430074, PR China
| | - Yao Nie
- Department of Biological Science and Technology, School of Environmental Studies & State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences, Wuhan 430074, PR China
| | - Lei Zhang
- Department of Biological Science and Technology, School of Environmental Studies & State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences, Wuhan 430074, PR China
| | - Shifen Wu
- Department of Biological Science and Technology, School of Environmental Studies & State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences, Wuhan 430074, PR China
| | - Aorigele Bao
- Department of Biological Science and Technology, School of Environmental Studies & State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences, Wuhan 430074, PR China
| |
Collapse
|
41
|
Rong M, Liu J, Zhang M, Wang G, Zhao G, Wang G, Zhang Y, Hu K, Lai R. A sodium channel inhibitor ISTX-I with a novel structure provides a new hint at the evolutionary link between two toxin folds. Sci Rep 2016; 6:29691. [PMID: 27407029 PMCID: PMC4942781 DOI: 10.1038/srep29691] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/23/2016] [Indexed: 12/15/2022] Open
Abstract
Members of arachnida, such as spiders and scorpions, commonly produce venom with specialized venom glands, paralyzing their prey with neurotoxins that specifically target ion channels. Two well-studied motifs, the disulfide-directed hairpin (DDH) and the inhibitor cystine knot motif (ICK), are both found in scorpion and spider toxins. As arachnids, ticks inject a neurotoxin-containing cocktail from their salivary glands into the host to acquire a blood meal, but peptide toxins acting on ion channels have not been observed in ticks. Here, a new neurotoxin (ISTX-I) that acts on sodium channels was identified from the hard tick Ixodes scapularis and characterized. ISTX-I exhibits a potent inhibitory function with an IC50 of 1.6 μM for sodium channel Nav1.7 but not other sodium channel subtypes. ISTX-I adopts a novel structural fold and is distinct from the canonical ICK motif. Analysis of the ISTX-I, DDH and ICK motifs reveals that the new ISTX-I motif might be an intermediate scaffold between DDH and ICK, and ISTX-I is a clue to the evolutionary link between the DDH and ICK motifs. These results provide a glimpse into the convergent evolution of neurotoxins from predatory and blood-sucking arthropods.
Collapse
Affiliation(s)
- Mingqiang Rong
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Kunming Yunnan 650223, China
| | - Jiangxin Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Meilin Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Kunming Yunnan 650223, China
| | - Gan Wang
- Life Sciences College of Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Gang Zhao
- Yunnan Academy of Grassland and Animal Science, Xiaoshao, Kunming 650212, China
| | - Guodong Wang
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Yaping Zhang
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Kaifeng Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Kunming Yunnan 650223, China.,Life Sciences College of Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| |
Collapse
|
42
|
In cellulo phosphorylation induces pharmacological reprogramming of maurocalcin, a cell-penetrating venom peptide. Proc Natl Acad Sci U S A 2016; 113:E2460-8. [PMID: 27071086 DOI: 10.1073/pnas.1517342113] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The venom peptide maurocalcin (MCa) is atypical among toxins because of its ability to rapidly translocate into cells and potently activate the intracellular calcium channel type 1 ryanodine receptor (RyR1). Therefore, MCa is potentially subjected to posttranslational modifications within recipient cells. Here, we report that MCa Thr(26) belongs to a consensus PKA phosphorylation site and can be phosphorylated by PKA both in vitro and after cell penetration in cellulo. Unexpectedly, phosphorylation converts MCa from positive to negative RyR1 allosteric modulator. Thr(26) phosphorylation leads to charge neutralization of Arg(24), a residue crucial for MCa agonist activity. The functional effect of Thr(26) phosphorylation is partially mimicked by aspartyl mutation. This represents the first case, to our knowledge, of both ex situ posttranslational modification and pharmacological reprogramming of a small natural cystine-rich peptide by target cells. So far, phosphorylated MCa is the first specific negative allosteric modulator of RyR1, to our knowledge, and represents a lead compound for further development of phosphatase-resistant analogs.
Collapse
|
43
|
Walker AA, Weirauch C, Fry BG, King GF. Venoms of Heteropteran Insects: A Treasure Trove of Diverse Pharmacological Toolkits. Toxins (Basel) 2016; 8:43. [PMID: 26907342 PMCID: PMC4773796 DOI: 10.3390/toxins8020043] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 01/25/2016] [Accepted: 01/26/2016] [Indexed: 11/16/2022] Open
Abstract
The piercing-sucking mouthparts of the true bugs (Insecta: Hemiptera: Heteroptera) have allowed diversification from a plant-feeding ancestor into a wide range of trophic strategies that include predation and blood-feeding. Crucial to the success of each of these strategies is the injection of venom. Here we review the current state of knowledge with regard to heteropteran venoms. Predaceous species produce venoms that induce rapid paralysis and liquefaction. These venoms are powerfully insecticidal, and may cause paralysis or death when injected into vertebrates. Disulfide-rich peptides, bioactive phospholipids, small molecules such as N,N-dimethylaniline and 1,2,5-trithiepane, and toxic enzymes such as phospholipase A2, have been reported in predatory venoms. However, the detailed composition and molecular targets of predatory venoms are largely unknown. In contrast, recent research into blood-feeding heteropterans has revealed the structure and function of many protein and non-protein components that facilitate acquisition of blood meals. Blood-feeding venoms lack paralytic or liquefying activity but instead are cocktails of pharmacological modulators that disable the host haemostatic systems simultaneously at multiple points. The multiple ways venom is used by heteropterans suggests that further study will reveal heteropteran venom components with a wide range of bioactivities that may be recruited for use as bioinsecticides, human therapeutics, and pharmacological tools.
Collapse
Affiliation(s)
- Andrew A Walker
- Institute for Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Christiane Weirauch
- Department of Entomology, University of California, Riverside, CA 92521, USA.
| | - Bryan G Fry
- School of Biological Sciences, The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Glenn F King
- Institute for Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
44
|
Synthetic Cystine-Knot Miniproteins - Valuable Scaffolds for Polypeptide Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 917:121-44. [PMID: 27236555 DOI: 10.1007/978-3-319-32805-8_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Peptides with the cystine-knot architecture, often termed knottins, are promising scaffolds for biomolecular engineering. These unique molecules combine diverse bioactivities with excellent structural, thermal, and proteolytical stability. Being different in the composition and structure of their amino acid backbone, knottins share the same core element, namely cystine knot, which is built by six cysteine residues forming three disulfides upon oxidative folding. This motif ensures a notably rigid framework that highly tolerates both rational and combinatorial changes in the primary structure. Being accessible through recombinant production and total chemical synthesis, cystine-knot miniproteins can be endowed with novel bioactivities by variation of surface-exposed loops and incorporation of non-natural elements within their non-conserved regions towards the generation of tailor-made peptidic compounds. In this chapter the topology of cystine-knot peptides, their synthesis and applications for diagnostics and therapy is discussed.
Collapse
|
45
|
Trancriptomic approach reveals the molecular diversity of Hottentotta conspersus (Buthidae) venom. Toxicon 2015; 99:73-9. [DOI: 10.1016/j.toxicon.2015.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/20/2015] [Accepted: 03/25/2015] [Indexed: 11/21/2022]
|
46
|
Nguyen PQT, Luu TT, Bai Y, Nguyen GKT, Pervushin K, Tam JP. Allotides: Proline-Rich Cystine Knot α-Amylase Inhibitors from Allamanda cathartica. JOURNAL OF NATURAL PRODUCTS 2015; 78:695-704. [PMID: 25832441 DOI: 10.1021/np500866c] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Cystine knot α-amylase inhibitors belong to a knottin family of peptidyl inhibitors of 30-32 residues and contain two to four prolines. Thus far, only four members of the group of cystine knot α-amylase inhibitors have been characterized. Herein, the discovery and characterization of five cystine knot α-amylase inhibitors, allotides C1-C5 (Ac1-Ac5) (1-5), from the medicinal plant Allamanda cathartica are reported using both proteomic and genomic methods. Proteomic analysis showed that 1-5 are 30 amino acids in length with three or four proline residues. NMR determination of 4 revealed that it has two cis- and one trans-proline residues and adopts two equally populated conformations in solution. Determination of disulfide connectivity of 2 by differential S-reduction and S-alkylation provided clues of its unfolding process. Genomic analysis showed that allotide precursors contain a three-domain arrangement commonly found in plant cystine knot peptides with conserved residues flanking the processing sites of the mature allotide domain. This work expands the number of known cystine knot α-amylase inhibitors and furthers the understanding of both the structural and biological diversity of this type of knottin family.
Collapse
Affiliation(s)
- Phuong Q T Nguyen
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Thuy T Luu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Yang Bai
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Giang K T Nguyen
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Konstantin Pervushin
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - James P Tam
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| |
Collapse
|
47
|
Chen J, Xu Y, San M, Cao Z, Li W, Wu Y, Chen Z. Cloning and Genomic Characterization of a Natural Insecticidal Peptide LaIT1 with Unique DDH Structural Fold. J Biochem Mol Toxicol 2015; 29:207-12. [DOI: 10.1002/jbt.21686] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 11/14/2014] [Accepted: 12/10/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Jing Chen
- Department of Biochemistry and Molecular Biology; Institute of Basic Medical Sciences; Hubei University of Medicine; Hubei People's Republic of China
- State Key Laboratory of Virology; College of Life Sciences; Wuhan University; Hubei People's Republic of China
| | - Yue Xu
- Department of Biochemistry and Molecular Biology; Institute of Basic Medical Sciences; Hubei University of Medicine; Hubei People's Republic of China
| | - Mingkui San
- Department of Biochemistry and Molecular Biology; Institute of Basic Medical Sciences; Hubei University of Medicine; Hubei People's Republic of China
| | - Zhijian Cao
- State Key Laboratory of Virology; College of Life Sciences; Wuhan University; Hubei People's Republic of China
| | - Wenxin Li
- State Key Laboratory of Virology; College of Life Sciences; Wuhan University; Hubei People's Republic of China
| | - Yingliang Wu
- State Key Laboratory of Virology; College of Life Sciences; Wuhan University; Hubei People's Republic of China
| | - Zongyun Chen
- Department of Biochemistry and Molecular Biology; Institute of Basic Medical Sciences; Hubei University of Medicine; Hubei People's Republic of China
- State Key Laboratory of Virology; College of Life Sciences; Wuhan University; Hubei People's Republic of China
| |
Collapse
|
48
|
Touchard A, Koh JMS, Aili SR, Dejean A, Nicholson GM, Orivel J, Escoubas P. The complexity and structural diversity of ant venom peptidomes is revealed by mass spectrometry profiling. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2015; 29:385-396. [PMID: 26349460 DOI: 10.1002/rcm.7116] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/03/2014] [Accepted: 12/03/2014] [Indexed: 06/05/2023]
Abstract
RATIONALE Compared with other animal venoms, ant venoms remain little explored. Ants have evolved complex venoms to rapidly immobilize arthropod prey and to protect their colonies from predators and pathogens. Many ants have retained peptide-rich venoms that are similar to those of other arthropod groups. METHODS With the goal of conducting a broad and comprehensive survey of ant venom peptide diversity, we investigated the peptide composition of venoms from 82 stinging ant species from nine subfamilies using matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry (MALDI-TOFMS). We also conducted an in-depth investigation of eight venoms using reversed-phase high-performance liquid chromatography (RP-HPLC) separation coupled with offline MALDI-TOFMS. RESULTS Our results reveal that the peptide compositions of ant venom peptidomes from both poneroid and formicoid ant clades comprise hundreds of small peptides (<4 kDa), while large peptides (>4 kDa) are also present in the venom of formicoids. Chemical reduction revealed the presence of disulfide-linked peptides in most ant subfamilies, including peptides structured by one, two or three disulfide bonds as well as dimeric peptides reticulated by three disulfide bonds. CONCLUSIONS The biochemical complexity of ant venoms, associated with an enormous ecological and taxonomic diversity, suggests that stinging ant venoms constitute a promising source of bioactive molecules that could be exploited in the search for novel drug and biopesticide leads.
Collapse
Affiliation(s)
- Axel Touchard
- CNRS, UMR Ecologie des Forêts de Guyane (EcoFoG), Campus Agronomique, BP 316, 97379, Kourou Cedex, France
| | - Jennifer M S Koh
- Neurotoxin Research Group, School of Medical & Molecular Biosciences, University of Technology, Sydney, NSW, Australia
| | - Samira R Aili
- Neurotoxin Research Group, School of Medical & Molecular Biosciences, University of Technology, Sydney, NSW, Australia
| | - Alain Dejean
- CNRS, UMR Ecologie des Forêts de Guyane (EcoFoG), Campus Agronomique, BP 316, 97379, Kourou Cedex, France
- Laboratoire Écologie Fonctionnelle et Environnement, Université de Toulouse, Toulouse, France
| | - Graham M Nicholson
- Neurotoxin Research Group, School of Medical & Molecular Biosciences, University of Technology, Sydney, NSW, Australia
| | - Jérôme Orivel
- CNRS, UMR Ecologie des Forêts de Guyane (EcoFoG), Campus Agronomique, BP 316, 97379, Kourou Cedex, France
| | | |
Collapse
|
49
|
Park HG, Kyung SS, Lee KS, Kim BY, Choi YS, Yoon HJ, Kwon HW, Je YH, Jin BR. Dual function of a bee (Apis cerana) inhibitor cysteine knot peptide that acts as an antifungal peptide and insecticidal venom toxin. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 47:247-53. [PMID: 25106915 DOI: 10.1016/j.dci.2014.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 07/31/2014] [Accepted: 08/01/2014] [Indexed: 05/27/2023]
Abstract
Inhibitor cysteine knot (ICK) peptides exhibit ion channel blocking, insecticidal, and antimicrobial activities, but currently, no functional roles for bee-derived ICK peptides have been identified. In this study, a bee (Apis cerana) ICK peptide (AcICK) that acts as an antifungal peptide and as an insecticidal venom toxin was identified. AcICK contains an ICK fold that is expressed in the epidermis, fat body, or venom gland and is present as a 6.6-kDa peptide in bee venom. Recombinant AcICK peptide (expressed in baculovirus-infected insect cells) bound directly to Beauveria bassiana and Fusarium graminearum, but not to Escherichia coli or Bacillus thuringiensis. Consistent with these findings, AcICK showed antifungal activity, indicating that AcICK acts as an antifungal peptide. Furthermore, AcICK expression is induced in the fat body and epidermis after injection with B. bassiana. These results provide insight into the role of AcICK during the innate immune response following fungal infection. Additionally, we show that AcICK has insecticidal activity. Our results demonstrate a functional role for AcICK in bees: AcICK acts as an antifungal peptide in innate immune reactions in the body and as an insecticidal toxin in venom. The finding that the AcICK peptide functions with different mechanisms of action in the body and in venom highlights the two-pronged strategy that is possible with the bee ICK peptide.
Collapse
Affiliation(s)
- Hee Geun Park
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, Republic of Korea
| | - Seung Su Kyung
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, Republic of Korea
| | - Kwang Sik Lee
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, Republic of Korea
| | - Bo Yeon Kim
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, Republic of Korea
| | - Yong Soo Choi
- Department of Agricultural Biology, National Academy of Agricultural Science, Suwon, Republic of Korea
| | - Hyung Joo Yoon
- Department of Agricultural Biology, National Academy of Agricultural Science, Suwon, Republic of Korea
| | - Hyung Wook Kwon
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Yeon Ho Je
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Byung Rae Jin
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, Republic of Korea.
| |
Collapse
|
50
|
Holocyclotoxin-1, a cystine knot toxin from Ixodes holocyclus. Toxicon 2014; 90:308-17. [DOI: 10.1016/j.toxicon.2014.08.068] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 08/20/2014] [Accepted: 08/20/2014] [Indexed: 01/31/2023]
|