1
|
Terenteva O, Mostovaya O, Bukharov M, Mukhametzyanov T, Bikmukhametov A, Lyubina A, Voloshina A, Petrov K, Padnya P, Stoikov I. Peptidomimetics based on thiacalixarene with L-tyrosine moieties: Antibacterial activity against methicillin-resistant Staphylococcus aureus and degradation induced by binding to α-chymotrypsin. Bioorg Chem 2025; 160:108434. [PMID: 40187027 DOI: 10.1016/j.bioorg.2025.108434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The design of new antimicrobial agents is an important challenge due to the growing resistance of microorganisms to existing antibiotics. In recent years, the trend towards the development of compounds and materials with (bio)degradable properties has emerged. In this work, we propose and develop a method for the synthesis of new peptidomimetics, i.e., water-soluble macrocyclic quaternary ammonium salts containing L-tyrosine fragments based on p-tert-butylthiacalix[4]arene in various stereoisomeric forms (cone, partial cone, and 1,3-alternate). These compounds have low cytotoxicity (IC50 = 80-267 μM) and high antibacterial activity (MIC = 0.5-15.6 μM) against Gram-positive bacterial strains including methicillin-resistant Staphylococcus aureus (MRSA). The obtained peptidomimetics can bind α-chymotrypsin with the formation of supramolecular systems and their subsequent degradation. Our results demonstrate the first example of multi-action thiacalixarene derivatives with antibacterial activity, protein binding ability and degradation induced by binding to α-chymotrypsin. The obtained results open the possibility of creating multi-action peptidomimetic systems with antimicrobial and biodegradable effect.
Collapse
Affiliation(s)
- Olga Terenteva
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Olga Mostovaya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Mikhail Bukharov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Timur Mukhametzyanov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Azamat Bikmukhametov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Anna Lyubina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Alexandra Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Konstantin Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Pavel Padnya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation.
| | - Ivan Stoikov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation.
| |
Collapse
|
2
|
Barroso RA, Rodrigues T, Campos A, Almeida D, Guardiola FA, Turkina MV, Antunes A. Proteomic Diversity of the Sea Anemone Actinia fragacea: Comparative Analysis of Nematocyst Venom, Mucus, and Tissue-Specific Profiles. Mar Drugs 2025; 23:79. [PMID: 39997203 PMCID: PMC11857728 DOI: 10.3390/md23020079] [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: 01/25/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Sea anemones (Actiniaria, Cnidaria) are promising targets for biomedical research, as they produce unique bioactive compounds, including toxins and antimicrobial peptides (AMPs). However, the diversity and mechanisms underlying their chemical defenses remain poorly understood. In this study, we investigate the proteomic profiles of the unexplored sea anemone Actinia fragacea by analyzing its venom nematocyst extract, tissues, and mucus secretion. A total of 4011 different proteins were identified, clustered into 3383 protein groups. Among the 83 putative toxins detected, actinoporins, neurotoxins, and phospholipase A2 were uncovered, as well as two novel zinc metalloproteinases with two specific domains (ShK) associated with potassium channel inhibition. Common Gene Ontology (GO) terms were related to immune responses, cell adhesion, protease inhibition, and tissue regeneration. Furthermore, 1406 of the 13,276 distinct peptides identified were predicted as potential AMPs, including a putative Aurelin-like AMP localized within the nematocysts. This discovery highlights and strengthens the evidence for a cnidarian-exclusive Aurelin peptide family. Several other bioactive compounds with distinctive defense functions were also detected, including enzymes, pattern recognition proteins (PRPs), and neuropeptides. This study provides the first proteome map of A. fragacea, offering a critical foundation for exploring novel bioactive compounds and valuable insights into its molecular complexity.
Collapse
Affiliation(s)
- Ricardo Alexandre Barroso
- CIIMAR/CIMAR—Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450-208 Porto, Portugal; (R.A.B.); (T.R.); (A.C.)
- Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre 687, 4169-007 Porto, Portugal
| | - Tomás Rodrigues
- CIIMAR/CIMAR—Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450-208 Porto, Portugal; (R.A.B.); (T.R.); (A.C.)
- Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre 687, 4169-007 Porto, Portugal
| | - Alexandre Campos
- CIIMAR/CIMAR—Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450-208 Porto, Portugal; (R.A.B.); (T.R.); (A.C.)
| | - Daniela Almeida
- Department of Zoology and Physical Anthropology, Faculty of Biology, University of Murcia, Campus of International Excellence, Campus Mare Nostrum, 30100 Murcia, Spain;
| | - Francisco A. Guardiola
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Regional Campus of International Excellence “Campus Mare Nostrum”, University of Murcia, 30100 Murcia, Spain;
| | - Maria V. Turkina
- Department of Biomedical and Clinical Sciences, Faculty of Medicine and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden;
| | - Agostinho Antunes
- CIIMAR/CIMAR—Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450-208 Porto, Portugal; (R.A.B.); (T.R.); (A.C.)
- Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre 687, 4169-007 Porto, Portugal
| |
Collapse
|
3
|
Kvetkina AN, Oreshkov SD, Mironov PA, Zaigraev MM, Klimovich AA, Deriavko YV, Menshov AS, Kulbatskii DS, Logashina YA, Andreev YA, Chugunov AO, Kirpichnikov MP, Lyukmanova EN, Leychenko EV, Shenkarev ZO. Sea Anemone Kunitz Peptide HCIQ2c1: Structure, Modulation of TRPA1 Channel, and Suppression of Nociceptive Reaction In Vivo. Mar Drugs 2024; 22:542. [PMID: 39728117 DOI: 10.3390/md22120542] [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: 10/18/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
TRPA1 is a homotetrameric non-selective calcium-permeable channel. It contributes to chemical and temperature sensitivity, acute pain sensation, and development of inflammation. HCIQ2c1 is a peptide from the sea anemone Heteractis magnifica that inhibits serine proteases. Here, we showed that HCIQ2c1 significantly reduces AITC- and capsaicin-induced pain and inflammation in mice. Electrophysiology recordings in Xenopus oocytes expressing rat TRPA1 channel revealed that HCIQ2c1 binds to open TRPA1 and prevents its transition to closed and inhibitor-insensitive 'hyperactivated' states. NMR study of the 15N-labeled recombinant HCIQ2c1 analog described a classical Kunitz-type structure and revealed two dynamic hot-spots (loops responsible for protease binding and regions near the N- and C-termini) that exhibit simultaneous mobility on two timescales (ps-ns and μs-ms). In modelled HCIQ2c1/TRPA1 complex, the peptide interacts simultaneously with one voltage-sensing-like domain and two pore domain fragments from different channel's subunits, and with lipid molecules. The model explains stabilization of the channel in the open conformation and the restriction of 'hyperactivation', which are probably responsible for the observed analgetic activity. HCIQ2c1 is the third peptide ligand of TRPA1 from sea anemones and the first Kunitz-type ligand of this channel. HCIQ2c1 is a prototype of efficient analgesic and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Aleksandra N Kvetkina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
| | - Sergey D Oreshkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Pavel A Mironov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
- Interdisciplinary Scientific and Educational School of Moscow University «Molecular Technologies of the Living Systems and Synthetic Biology», Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Maxim M Zaigraev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Anna A Klimovich
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
| | - Yulia V Deriavko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
| | - Aleksandr S Menshov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
| | - Dmitrii S Kulbatskii
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
| | - Yulia A Logashina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
| | - Yaroslav A Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
| | - Anton O Chugunov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Mikhail P Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
- Interdisciplinary Scientific and Educational School of Moscow University «Molecular Technologies of the Living Systems and Synthetic Biology», Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ekaterina N Lyukmanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
- Interdisciplinary Scientific and Educational School of Moscow University «Molecular Technologies of the Living Systems and Synthetic Biology», Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Shenzhen MSU-BIT University, No. 1, International University Park Road, Dayun New Town, Longgang District, Shenzhen 518172, China
| | - Elena V Leychenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
| | - Zakhar O Shenkarev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| |
Collapse
|
4
|
Hua Z, Liao Y, Fu J, Li X, Xu Q, Lin L, Huang M, Gao B. Revealing the Diversity of Sequences, Structures, and Targets of Peptides from South China Sea Macrodactyla doreensis Based on Transcriptomics. Mar Drugs 2024; 22:470. [PMID: 39452877 PMCID: PMC11509556 DOI: 10.3390/md22100470] [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: 08/23/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
The South China Sea is rich in sea anemone resources, and the protein and peptide components from sea anemone toxins comprise an important treasure trove for researchers to search for leading compounds. This study conducted a comprehensive transcriptomic analysis of the tentacles and column of Macrodactyla doreensis and explored the distribution and diversity of proteins and peptides in depth using bioinformatics, initially constructing a putative protein and peptide database. In this database, typical peptide families are identified through amino acid sequence analysis, and their 3D structures and potential biological activities are revealed through AlphaFold2 modeling and molecular docking. A total of 4239 transcripts were identified, of which the putative protein accounted for 81.53%. The highest content comprised immunoglobulin and a variety of proteases, mainly distributed in the column and related to biological functions. Importantly, the putative peptide accounted for 18.47%, containing ShK domain and Kunitz-type peptides, mainly distributed in the tentacles and related to offensive predatory behavior. Interestingly, 40 putative peptides belonging to eight typical peptide families were identified, and their structures and targets were predicted. This study reveals the diversity and complexity of Macrodactyla doreensis toxins and predicts their structure and targets based on amino acid sequences, providing a feasible approach for research regarding the discovery of peptides with potentially high activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Meiling Huang
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, International Joint Research Center of Human-Machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, School of Pharmacy, Hainan Medical University, Haikou 571199, China; (Z.H.); (Y.L.); (J.F.); (X.L.); (Q.X.); (L.L.)
| | - Bingmiao Gao
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, International Joint Research Center of Human-Machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, School of Pharmacy, Hainan Medical University, Haikou 571199, China; (Z.H.); (Y.L.); (J.F.); (X.L.); (Q.X.); (L.L.)
| |
Collapse
|
5
|
Paul DC, Bhattacharjee M. Revisiting the significance of natural protease inhibitors: A comprehensive review. Int J Biol Macromol 2024; 280:135899. [PMID: 39317291 DOI: 10.1016/j.ijbiomac.2024.135899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/09/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
Protease inhibitors (PIs) function as a natural adversary to proteolytic enzymes. They can diminish or inhibit the catalytic properties of proteases, which are crucial for various tasks in the physiology and metabolism of cellular forms. Protease Inhibitors are low molecular weight (5-25 kDa) stable proteins. Plants are a fair source of PIs, so foods containing PIs remarkably influence human health. PIs are usually present in storage tissues of the plant, although they are present in other aerial parts as well. In plants, protease inhibitors participate in vital functions such as maintaining physiological homeostasis, mobilization of storage proteins, defense systems, apoptosis, and other processes. In recent years, plant-derived PIs have shown promising results in treating various diseases including inflammatory conditions, osteoporosis, cardiovascular issues, and brain disorders. The primary goal of this review is to provide a comprehensive understanding of the characteristics, applications, and challenges associated with natural protease inhibitors in plants, which draws insights from an extensive examination of 80+ research papers with a focus on their potential in agriculture and medicine. By synthesizing findings from an extensive literature review, this work aims to guide future research directions and innovations in leveraging plant-based PIs for sustainable agricultural practices and advanced therapeutic interventions.
Collapse
Affiliation(s)
- Dhiman Chandra Paul
- Programme of Biotechnology, Assam down town University, Panikhaiti, Gandhinagar, Guwahati, Assam 26, India
| | - Minakshi Bhattacharjee
- Programme of Biotechnology, Assam down town University, Panikhaiti, Gandhinagar, Guwahati, Assam 26, India.
| |
Collapse
|
6
|
Ghourchian H, Pecho RDC, Karimi-Dehkordi M, Mazandarani A, Ghajari G, Piri-Gharaghie T. Novel Niosome-Encapsulated 2,5-Diketopiperazine (BHPPD): Synthesis, Formulation, and Anti-breast Cancer Activity. Appl Biochem Biotechnol 2024; 196:3126-3147. [PMID: 37624507 DOI: 10.1007/s12010-023-04687-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
In the course of this investigation, a brand-new noisome-encapsulated 2,5-diketopiperazine (BHPPD) was developed, synthesized, and assessed. Utilizing CCK-8, invasion screens, MTT test, flow cytometry, and cell cycle analysis, we evaluated the anti-breast cancer properties of niosome-encapsulated BHPPD. Apoptosis-related gene expression and cytotoxicity was measured using quantitative real-time PCR and MTT assays. This meta-analysis showed a significant drug-binding affinity for intestinal protease. The spherical mean diameters of the free BHPPD, the F1 niosomal-BHPPD, and the F2 niosomal-BHPPD were all determined to be108.91 ± 4.2, 129.13 ± 7.2 nm, and 149.43 ± 3.2 nm, respectively. Also, it was found that the entrapment efficiency (EE%) of the F1 formulations of BHPPD that was niosome-encapsulated was 81.01 0.09% and that it was 70.22 0.13%, respectively. Early, late, necrotic, and viable MCF-7 cells were present in the cells with F1 formulation in proportions of 38.24%, 34.34%, 4.02%, and 23.40%, respectively. Compared to the control group, the treatment group's expression of the genes P57, Prkca, MDM4, Map2k6, and FADD was considerably greater (P < 0.001). Furthermore, compared to control cells, cells in the treatment group expressed less BCL2 and survival genes (P < 0.001). Moreover, formulations of BHPPD encapsulated in niosomes showed a biocompatible nanoscale delivery method and exhibited little cytotoxicity against the HEK-293 standard cell line. According to the findings, formulations of BHPPD with niosome-encapsulation might be viable for boosting anticancer activity.
Collapse
Affiliation(s)
- Hedieh Ghourchian
- Department of Biology, Faculty of Biological Science, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | | | - Maryam Karimi-Dehkordi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Aynaz Mazandarani
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Ghazal Ghajari
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tohid Piri-Gharaghie
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| |
Collapse
|
7
|
Guo Q, Fu J, Yuan L, Liao Y, Li M, Li X, Yi B, Zhang J, Gao B. Diversity analysis of sea anemone peptide toxins in different tissues of Heteractis crispa based on transcriptomics. Sci Rep 2024; 14:7684. [PMID: 38561372 PMCID: PMC10985097 DOI: 10.1038/s41598-024-58402-2] [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: 11/27/2023] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
Peptide toxins found in sea anemones venom have diverse properties that make them important research subjects in the fields of pharmacology, neuroscience and biotechnology. This study used high-throughput sequencing technology to systematically analyze the venom components of the tentacles, column, and mesenterial filaments of sea anemone Heteractis crispa, revealing the diversity and complexity of sea anemone toxins in different tissues. A total of 1049 transcripts were identified and categorized into 60 families, of which 91.0% were proteins and 9.0% were peptides. Of those 1049 transcripts, 416, 291, and 307 putative proteins and peptide precursors were identified from tentacles, column, and mesenterial filaments respectively, while 428 were identified when the datasets were combined. Of these putative toxin sequences, 42 were detected in all three tissues, including 33 proteins and 9 peptides, with the majority of peptides being ShKT domain, β-defensin, and Kunitz-type. In addition, this study applied bioinformatics approaches to predict the family classification, 3D structures, and functional annotation of these representative peptides, as well as the evolutionary relationships between peptides, laying the foundation for the next step of peptide pharmacological activity research.
Collapse
Affiliation(s)
- Qiqi Guo
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Jinxing Fu
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Lin Yuan
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, School of Pharmacy, Hainan Medical University, Haikou, China
- Department of Pharmacy, 928th Hospital of PLA Joint Logistics Support Force, Haikou, China
| | - Yanling Liao
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Ming Li
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Xinzhong Li
- School of Health and Life Sciences, Teesside University, Middlesbrough, UK
| | - Bo Yi
- Department of Pharmacy, 928th Hospital of PLA Joint Logistics Support Force, Haikou, China
| | - Junqing Zhang
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, School of Pharmacy, Hainan Medical University, Haikou, China.
| | - Bingmiao Gao
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, School of Pharmacy, Hainan Medical University, Haikou, China.
| |
Collapse
|
8
|
Lin Z, Yi X, Ali MM, Zhang L, Wang S, Tian S, Chen F. RNAi-Mediated Suppression of OsBBTI5 Promotes Salt Stress Tolerance in Rice. Int J Mol Sci 2024; 25:1284. [PMID: 38279284 PMCID: PMC10816146 DOI: 10.3390/ijms25021284] [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: 10/19/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
This study explores the impact of RNAi in terms of selectively inhibiting the expression of the OsBBTI5 gene, with the primary objective of uncovering its involvement in the molecular mechanisms associated with salt tolerance in rice. OsBBTI5, belonging to the Bowman-Birk inhibitor (BBI) family gene, is known for its involvement in plant stress responses. The gene was successfully cloned from rice, exhibiting transcriptional self-activation in yeast. A yeast two-hybrid assay confirmed its specific binding to OsAPX2 (an ascorbate peroxidase gene). Transgenic OsBBTI5-RNAi plants displayed insensitivity to varying concentrations of 24-epibrassinolide in the brassinosteroid sensitivity assay. However, they showed reduced root and plant height at high concentrations (10 and 100 µM) of GA3 immersion. Enzyme activity assays revealed increased peroxidase (POD) and superoxide dismutase (SOD) activities and decreased malondialdehyde (MDA) content under 40-60 mM NaCl. Transcriptomic analysis indicated a significant upregulation of photosynthesis-related genes in transgenic plants under salt stress compared to the wild type. Notably, this study provides novel insights, suggesting that the BBI gene is part of the BR signaling pathway, and that OsBBTI5 potentially enhances stress tolerance in transgenic plants through interaction with the salt stress-related gene OsAPX2.
Collapse
Affiliation(s)
- Zhimin Lin
- Fujian Academy of Agricultural Sciences Biotechnology Institute, Fuzhou 350003, China
| | - Xiaoyan Yi
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.Y.); (M.M.A.); (L.Z.); (S.W.); (S.T.)
| | - Muhammad Moaaz Ali
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.Y.); (M.M.A.); (L.Z.); (S.W.); (S.T.)
| | - Lijuan Zhang
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.Y.); (M.M.A.); (L.Z.); (S.W.); (S.T.)
| | - Shaojuan Wang
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.Y.); (M.M.A.); (L.Z.); (S.W.); (S.T.)
| | - Shengnan Tian
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.Y.); (M.M.A.); (L.Z.); (S.W.); (S.T.)
| | - Faxing Chen
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.Y.); (M.M.A.); (L.Z.); (S.W.); (S.T.)
| |
Collapse
|
9
|
Lecaudey LA, Netzer R, Wibberg D, Busche T, Bloecher N. Metatranscriptome analysis reveals the putative venom toxin repertoire of the biofouling hydroid Ectopleura larynx. Toxicon 2024; 237:107556. [PMID: 38072317 DOI: 10.1016/j.toxicon.2023.107556] [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: 07/07/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023]
Abstract
Cnidarians thriving in biofouling communities on aquaculture net pens represent a significant health risk for farmed finfish due to their stinging cells. The toxins coming into contact with the fish, during net cleaning, can adversely affect their behavior, welfare, and survival, with a particularly serious health risk for the gills, causing direct tissue damage such as formation of thrombi and increasing risks of secondary infections. The hydroid Ectopleura larynx is one of the most common fouling organisms in Northern Europe. However, despite its significant economic, environmental, and operational impact on finfish aquaculture, biological information on this species is scarce and its venom composition has never been investigated. In this study, we generated a whole transcriptome of E. larynx, and identified its putative expressed venom toxin proteins (predicted toxin proteins, not functionally characterized) based on in silico transcriptome annotation mining and protein sequence analysis. The results uncovered a broad and diverse repertoire of putative toxin proteins for this hydroid species. Its toxic arsenal appears to include a wide and complex selection of toxin proteins, covering a large panel of potential biological functions that play important roles in envenomation. The putative toxins identified in this species, such as neurotoxins, GTPase toxins, metalloprotease toxins, ion channel impairing toxins, hemorrhagic toxins, serine protease toxins, phospholipase toxins, pore-forming toxins, and multifunction toxins may cause various major deleterious effects in prey, predators, and competitors. These results provide valuable new insights into the venom composition of cnidarians, and venomous marine organisms in general, and offer new opportunities for further research into novel and valuable bioactive molecules for medicine, agronomics and biotechnology.
Collapse
Affiliation(s)
| | - Roman Netzer
- SINTEF Ocean, Aquaculture Department, Brattørkaia 17c, 7010, Trondheim, Norway
| | - Daniel Wibberg
- Center for Biotechnology (CeBiTec), Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Tobias Busche
- Center for Biotechnology (CeBiTec), Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany; Medical School OWL, Bielefeld University, Morgenbreede 1, 33615, Bielefeld, Germany
| | - Nina Bloecher
- SINTEF Ocean, Aquaculture Department, Brattørkaia 17c, 7010, Trondheim, Norway
| |
Collapse
|
10
|
Sonoda GG, Tobaruela EDC, Norenburg J, Fabi JP, Andrade SCS. Venomous Noodles: The Evolution of Toxins in Nemertea through Positive Selection and Gene Duplication. Toxins (Basel) 2023; 15:650. [PMID: 37999513 PMCID: PMC10674772 DOI: 10.3390/toxins15110650] [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: 08/30/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 11/25/2023] Open
Abstract
Some, probably most and perhaps all, members of the phylum Nemertea are poisonous, documented so far from marine and benthic specimens. Although the toxicity of these animals has been long known, systematic studies on the characterization of toxins, mechanisms of toxicity, and toxin evolution for this group are scarce. Here, we present the first investigation of the molecular evolution of toxins in Nemertea. Using a proteo-transcriptomic approach, we described toxins in the body and poisonous mucus of the pilidiophoran Lineus sanguineus and the hoplonemertean Nemertopsis pamelaroeae. Using these new and publicly available transcriptomes, we investigated the molecular evolution of six selected toxin gene families. In addition, we also characterized in silico the toxin genes found in the interstitial hoplonemertean, Ototyphlonemertes erneba, a meiofaunal taxa. We successfully identified over 200 toxin transcripts in each of these species. Evidence of positive selection and gene duplication was observed in all investigated toxin genes. We hypothesized that the increased rates of gene duplications observed for Pilidiophora could be involved with the expansion of toxin genes. Studies concerning the natural history of Nemertea are still needed to understand the evolution of their toxins. Nevertheless, our results show evolutionary mechanisms similar to other venomous groups.
Collapse
Affiliation(s)
- Gabriel Gonzalez Sonoda
- Departamento de Genética e Biologia Evolutiva, IB-Universidade de São Paulo, São Paulo 05508-090, Brazil;
- Instituto Butantan, São Paulo 05503-900, Brazil
| | - Eric de Castro Tobaruela
- Faculdade de Ciências Farmacêuticas, Food Research Center (FoRC), Universidade de São Paulo, São Paulo 05508-080, Brazil; (E.d.C.T.); (J.P.F.)
| | | | - João Paulo Fabi
- Faculdade de Ciências Farmacêuticas, Food Research Center (FoRC), Universidade de São Paulo, São Paulo 05508-080, Brazil; (E.d.C.T.); (J.P.F.)
| | - Sónia C. S. Andrade
- Departamento de Genética e Biologia Evolutiva, IB-Universidade de São Paulo, São Paulo 05508-090, Brazil;
| |
Collapse
|
11
|
Nolasco M, Mariano DOC, Pimenta DC, Biondi I, Branco A. Proteomic analyses of venom from a Spider Hawk, Pepsis decorata. J Venom Anim Toxins Incl Trop Dis 2023; 29:e20220090. [PMID: 37965483 PMCID: PMC10642949 DOI: 10.1590/1678-9199-jvatitd-2022-0090] [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: 01/05/2023] [Accepted: 06/12/2023] [Indexed: 11/16/2023] Open
Abstract
Background The composition of the venom from solitary wasps is poorly known, although these animals are considered sources of bioactive substances. Until the present moment, there is only one proteomic characterization of the venom of wasps of the family Pompilidae and this is the first proteomic characterization for the genus Pepsis. Methods To elucidate the components of Pepsis decorata venom, the present work sought to identify proteins using four different experimental conditions, namely: (A) crude venom; (B) reduced and alkylated venom; (C) trypsin-digested reduced and alkylated venom, and; (D) chymotrypsin-digested reduced and alkylated venom. Furthermore, three different mass spectrometers were used (Ion Trap-Time of Flight, Quadrupole-Time of Flight, and Linear Triple Quadruple). Results Proteomics analysis revealed the existence of different enzymes related to the insect's physiology in the venom composition. Besides toxins, angiotensin-converting enzyme (ACE), hyaluronidase, and Kunitz-type inhibitors were also identified. Conclusion The data showed that the venom of Pepsis decorata is mostly composed of proteins involved in the metabolism of arthropods, as occurs in parasitic wasps, although some classical toxins were recorded, and among them, for the first time, ACE was found in the venom of solitary wasps. This integrative approach expanded the range of compounds identified in protein analyses, proving to be efficient in the proteomic characterization of little-known species. It is our understanding that the current work will provide a solid base for future studies dealing with other Hymenoptera venoms.
Collapse
Affiliation(s)
- Matheus Nolasco
- Graduate Program in Biotechnology, Department of Biological Sciences, State University of Feira de Santana, Feira de Santana, BA, Brazil
| | - Douglas O. C. Mariano
- Laboratory of Biochemistry and Biophysics, Instituto Butantan, São Paulo, SP, Brazil
| | - Daniel C. Pimenta
- Laboratory of Biochemistry and Biophysics, Instituto Butantan, São Paulo, SP, Brazil
| | - Ilka Biondi
- Laboratory of Venomous Animals and Herpetology. Biology Department, State University of Feira de Santana - UEFS, Feira de Santana, BA, Brazil
| | - Alexsandro Branco
- Phytochemistry Laboratory, Health Department, State University of Feira de Santana - UEFS, Feira de Santana, BA, Brazil
| |
Collapse
|
12
|
Wójcik-Mieszawska S, Lewtak K, Skwarek E, Dębowski D, Gitlin-Domagalska A, Nowak J, Wydrych J, Pawelec J, Fiołka MJ. Autophagy of Candida albicans cells after the action of earthworm Venetin-1 nanoparticle with protease inhibitor activity. Sci Rep 2023; 13:14228. [PMID: 37648723 PMCID: PMC10468520 DOI: 10.1038/s41598-023-41281-4] [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: 03/02/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023] Open
Abstract
The present studies show the effect of the Venetin-1 protein-polysaccharide complex obtained from the coelomic fluid of the earthworm Dendrobaena veneta on Candida albicans cells. They are a continuation of research on the mechanisms of action, cellular targets, and modes of cell death. After the action of Venetin-1, a reduced survival rate of the yeast cells was noted. The cells were observed to be enlarged compared to the controls and deformed. In addition, an increase in the number of cells with clearly enlarged vacuoles was noted. The detected autophagy process was confirmed using differential interference contrast, fluorescence microscopy, and transmission electron microscopy. Autophagic vesicles were best visible after incubation of fungus cells with the Venetin-1 complex at a concentration of 50 and 100 µg mL-1. The changes in the vacuoles were accompanied by changes in the size of mitochondria, which is probably related to the previously documented oxidative stress. The aggregation properties of Venetin-1 were characterized. Based on the results of the zeta potential at the Venetin-1/KCl interface, the pHiep = 4 point was determined, i.e. the zeta potential becomes positive above pH = 4 and is negative below this value, which may affect the electrostatic interactions with other particles surrounding Venetin-1.
Collapse
Affiliation(s)
- Sylwia Wójcik-Mieszawska
- Department of Immunobiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland
| | - Kinga Lewtak
- Department of Cell Biology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland.
| | - Ewa Skwarek
- Department of Radiochemistry and Environmental Chemistry, Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Skłodowska University, Lublin, Poland
| | - Dawid Dębowski
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Agata Gitlin-Domagalska
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Jakub Nowak
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jerzy Wydrych
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Jarosław Pawelec
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Marta J Fiołka
- Department of Immunobiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland.
| |
Collapse
|
13
|
Kvetkina A, Pislyagin E, Menchinskaya E, Yurchenko E, Kalina R, Kozlovskiy S, Kaluzhskiy L, Menshov A, Kim N, Peigneur S, Tytgat J, Ivanov A, Ayvazyan N, Leychenko E, Aminin D. Kunitz-Type Peptides from Sea Anemones Protect Neuronal Cells against Parkinson's Disease Inductors via Inhibition of ROS Production and ATP-Induced P2X7 Receptor Activation. Int J Mol Sci 2022; 23:ijms23095115. [PMID: 35563513 PMCID: PMC9103184 DOI: 10.3390/ijms23095115] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is a socially significant disease, during the development of which oxidative stress and inflammation play a significant role. Here, we studied the neuroprotective effects of four Kunitz-type peptides from Heteractis crispa and Heteractis magnifica sea anemones against PD inductors. The peptide HCIQ1c9, which was obtained for the first time, inhibited trypsin less than other peptides due to unfavorable interactions of Arg17 with Lys43 in the enzyme. Its activity was reduced by up to 70% over the temperature range of 60–100 °C, while HCIQ2c1, HCIQ4c7, and HMIQ3c1 retained their conformation and stayed active up to 90–100 °C. All studied peptides inhibited paraquat- and rotenone-induced intracellular ROS formation, in particular NO, and scavenged free radicals outside the cells. The peptides did not modulate the TRPV1 channels but they affected the P2X7R, both of which are considered therapeutic targets in Parkinson’s disease. HMIQ3c1 and HCIQ4c7 almost completely inhibited the ATP-induced uptake of YO-PRO-1 dye in Neuro-2a cells through P2X7 ion channels and significantly reduced the stable calcium response in these cells. The complex formation of the peptides with the P2X7R extracellular domain was determined via SPR analysis. Thus, these peptides may be considered promising compounds to protect neuronal cells against PD inductors, which act as ROS production inhibitors and partially act as ATP-induced P2X7R activation inhibitors.
Collapse
Affiliation(s)
- Aleksandra Kvetkina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Evgeny Pislyagin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Ekaterina Menchinskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Ekaterina Yurchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Rimma Kalina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Sergei Kozlovskiy
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Leonid Kaluzhskiy
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., 119121 Moscow, Russia; (L.K.); (A.I.)
| | - Alexander Menshov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Natalia Kim
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Steve Peigneur
- Toxicology and Pharmacology, Campus Gasthuisberg O&N2, University of Leuven (KU Leuven), Herestraat 49, P.O. Box 922, B-3000 Leuven, Belgium; (S.P.); (J.T.)
| | - Jan Tytgat
- Toxicology and Pharmacology, Campus Gasthuisberg O&N2, University of Leuven (KU Leuven), Herestraat 49, P.O. Box 922, B-3000 Leuven, Belgium; (S.P.); (J.T.)
| | - Alexis Ivanov
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., 119121 Moscow, Russia; (L.K.); (A.I.)
| | - Naira Ayvazyan
- L.A. Orbeli Institute of Physiology, National Academy of Sciences of Armenia, Yerevan 0028, Armenia;
| | - Elena Leychenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
| | - Dmitry Aminin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.K.); (E.P.); (E.M.); (E.Y.); (R.K.); (S.K.); (A.M.); (N.K.); (E.L.)
- Correspondence:
| |
Collapse
|
14
|
AsKC11, a Kunitz Peptide from Anemonia sulcata, Is a Novel Activator of G Protein-Coupled Inward-Rectifier Potassium Channels. Mar Drugs 2022; 20:md20020140. [PMID: 35200669 PMCID: PMC8876855 DOI: 10.3390/md20020140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
(1) Background: G protein-coupled inward-rectifier potassium (GIRK) channels, especially neuronal GIRK1/2 channels, have been the focus of intense research interest for developing drugs against brain diseases. In this context, venom peptides that selectively activate GIRK channels can be seen as a new source for drug development. Here, we report on the identification and electrophysiological characterization of a novel activator of GIRK1/2 channels, AsKC11, found in the venom of the sea anemone Anemonia sulcata. (2) Methods: AsKC11 was purified from the sea anemone venom by reverse-phase chromatography and the sequence was identified by mass spectrometry. Using the two-electrode voltage-clamp technique, the activity of AsKC11 on GIRK1/2 channels was studied and its selectivity for other potassium channels was investigated. (3) Results: AsKC11, a Kunitz peptide found in the venom of A. sulcata, is the first peptide shown to directly activate neuronal GIRK1/2 channels independent from Gi/o protein activity, without affecting the inward-rectifier potassium channel (IRK1) and with only a minor effect on KV1.6 channels. Thus, AsKC11 is a novel activator of GIRK channels resulting in larger K+ currents because of an increased chord conductance. (4) Conclusions: These discoveries provide new insights into a novel class of GIRK activators.
Collapse
|
15
|
Deep-Sea Anemones Are Prospective Source of New Antimicrobial and Cytotoxic Compounds. Mar Drugs 2021; 19:md19120654. [PMID: 34940653 PMCID: PMC8704684 DOI: 10.3390/md19120654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/16/2022] Open
Abstract
The peculiarities of the survival and adaptation of deep-sea organisms raise interest in the study of their metabolites as promising drugs. In this work, the hemolytic, cytotoxic, antimicrobial, and enzyme-inhibitory activities of tentacle extracts from five species of sea anemones (Cnidaria, orders Actiniaria and Corallimorpharia) collected near the Kuril and Commander Islands of the Far East of Russia were evaluated for the first time. The extracts of Liponema brevicorne and Actinostola callosa demonstrated maximal hemolytic activity, while high cytotoxic activity against murine splenocytes and Ehrlich carcinoma cells was found in the extract of Actinostola faeculenta. The extracts of Corallimorphus cf. pilatus demonstrated the greatest activity against Ehrlich carcinoma cells but were not toxic to mouse spleen cells. Sea anemones C. cf. pilatus and Stomphia coccinea are promising sources of antimicrobial and antifungal compounds, being active against Gram-positive bacteria Bacillus subtilis, Staphylococcus aureus, and yeast Candida albicans. Moreover, all sea anemones contain α-galactosidase inhibitors. Peptide mass fingerprinting of L. brevicorne and C. cf. pilatus extracts provided a wide range of peptides, predominantly with molecular masses of 4000–5900 Da, which may belong to a known or new structural class of toxins. The obtained data allow concluding that deep-sea anemones are a promising source of compounds for drug discovery.
Collapse
|
16
|
Kim BY, Lee KS, Lee KY, Yoon HJ, Jin BR. Anti-fibrinolytic activity of a metalloprotease inhibitor from bumblebee (Bombus ignitus) venom. Comp Biochem Physiol C Toxicol Pharmacol 2021; 245:109042. [PMID: 33838314 DOI: 10.1016/j.cbpc.2021.109042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 11/20/2022]
Abstract
Bee venom is a mixture of bioactive components that include proteases and protease inhibitors. A metalloprotease inhibitor has been predicted to be a bumblebee-specific toxin in the venom proteome of Bombus terrestris; however, the identification and functional roles of bee venom metalloprotease inhibitors have not been previously determined. In this study, we identified a bumblebee (B. ignitus) venom metalloprotease inhibitor (BiVMPI) that exhibits anti-fibrinolytic activity. BiVMPI contains a trypsin inhibitor-like cysteine-rich domain that exhibits similarity to inducible metalloprotease inhibitor. Using an anti-BiVMPI antibody raised against a recombinant BiVMPI protein produced in baculovirus-infected insect cells, the presence of BiVMPI in the venom gland and secreted venom of B. ignitus worker bees was confirmed. The recombinant BiVMPI protein demonstrated inhibitory activity against a metalloprotease, trypsin, chymotrypsin, protease K, and plasmin, but not subtilisin A, elastase, or thrombin. Additionally, the recombinant BiVMPI bound to plasmin and inhibited the plasmin-mediated degradation of fibrin, demonstrating an anti-fibrinolytic role for BiVMPI as a bee venom metalloprotease inhibitor. Our results provide the first evidence for the identification and anti-fibrinolytic activity of a metalloprotease inhibitor from bee venom.
Collapse
Affiliation(s)
- Bo Yeon Kim
- College of Natural Resources and Life Science, Dong-A University, Busan 49315, Republic of Korea
| | - Kwang Sik Lee
- College of Natural Resources and Life Science, Dong-A University, Busan 49315, Republic of Korea
| | - Kyeong Yong Lee
- Department of Agricultural Biology, National Academy of Agricultural Science, Wanju 55365, Republic of Korea
| | - Hyung Joo Yoon
- Department of Agricultural Biology, National Academy of Agricultural Science, Wanju 55365, Republic of Korea.
| | - Byung Rae Jin
- College of Natural Resources and Life Science, Dong-A University, Busan 49315, Republic of Korea.
| |
Collapse
|
17
|
Damm M, Hempel BF, Süssmuth RD. Old World Vipers-A Review about Snake Venom Proteomics of Viperinae and Their Variations. Toxins (Basel) 2021; 13:toxins13060427. [PMID: 34204565 PMCID: PMC8235416 DOI: 10.3390/toxins13060427] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Fine-tuned by millions of years of evolution, snake venoms have frightened but also fascinated humanity and nowadays they constitute potential resources for drug development, therapeutics and antivenoms. The continuous progress of mass spectrometry techniques and latest advances in proteomics workflows enabled toxinologists to decipher venoms by modern omics technologies, so-called ‘venomics’. A tremendous upsurge reporting on snake venom proteomes could be observed. Within this review we focus on the highly venomous and widely distributed subfamily of Viperinae (Serpentes: Viperidae). A detailed public literature database search was performed (2003–2020) and we extensively reviewed all compositional venom studies of the so-called Old-World Vipers. In total, 54 studies resulted in 89 venom proteomes. The Viperinae venoms are dominated by four major, four secondary, six minor and several rare toxin families and peptides, respectively. The multitude of different venomics approaches complicates the comparison of venom composition datasets and therefore we differentiated between non-quantitative and three groups of quantitative workflows. The resulting direct comparisons within these groups show remarkable differences on the intra- and interspecies level across genera with a focus on regional differences. In summary, the present compilation is the first comprehensive up-to-date database on Viperinae venom proteomes and differentiating between analytical methods and workflows.
Collapse
Affiliation(s)
- Maik Damm
- Department of Chemistry, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany;
| | - Benjamin-Florian Hempel
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, (BCRT), 10117 Berlin, Germany;
| | - Roderich D. Süssmuth
- Department of Chemistry, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany;
- Correspondence: ; Tel.: +49-(0)30-314-24205
| |
Collapse
|
18
|
Identification of Novel Toxin Genes from the Stinging Nettle Caterpillar Parasa lepida (Cramer, 1799): Insights into the Evolution of Lepidoptera Toxins. INSECTS 2021; 12:insects12050396. [PMID: 33946702 PMCID: PMC8145965 DOI: 10.3390/insects12050396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary Many caterpillar species can produce toxins that cause harmful reactions to humans, varying from mild irritation to death. Currently, there is very limited knowledge about caterpillar toxin diversity, because only a few species have been investigated. We used the transcriptome technique to identify candidate toxin genes from the nettle caterpillar Parasa lepida (Cramer, 1799). It is a common pest of oil palm, coconut, and mango in South and South-East Asia, which can cause severe pain and allergic responses to those in contact with them. We reported 168 candidate toxin genes. Most of them are members of the toxin genes families commonly recruited in animal venoms such as serine protease and serine protease inhibitors. However, we identified 21 novel genes encoding knottin-like peptides expressed at a high level in the transcriptome. Their predicted 3D structures are similar to neurotoxins in scorpion and tarantula. Our study suggests that P. lepida venom contains diverse toxin proteins that potentially cause allergic reactions and pain. This study sheds light on the hidden diversity of toxin proteins in caterpillar lineage, which could be future fruitful new drug sources. Abstract Many animal species can produce venom for defense, predation, and competition. The venom usually contains diverse peptide and protein toxins, including neurotoxins, proteolytic enzymes, protease inhibitors, and allergens. Some drugs for cancer, neurological disorders, and analgesics were developed based on animal toxin structures and functions. Several caterpillar species possess venoms that cause varying effects on humans both locally and systemically. However, toxins from only a few species have been investigated, limiting the full understanding of the Lepidoptera toxin diversity and evolution. We used the RNA-seq technique to identify toxin genes from the stinging nettle caterpillar, Parasa lepida (Cramer, 1799). We constructed a transcriptome from caterpillar urticating hairs and reported 34,968 unique transcripts. Using our toxin gene annotation pipeline, we identified 168 candidate toxin genes, including protease inhibitors, proteolytic enzymes, and allergens. The 21 P. lepida novel Knottin-like peptides, which do not show sequence similarity to any known peptide, have predicted 3D structures similar to tarantula, scorpion, and cone snail neurotoxins. We highlighted the importance of convergent evolution in the Lepidoptera toxin evolution and the possible mechanisms. This study opens a new path to understanding the hidden diversity of Lepidoptera toxins, which could be a fruitful source for developing new drugs.
Collapse
|
19
|
Diversity, molecular mechanisms and structure-activity relationships of marine protease inhibitors-A review. Pharmacol Res 2021; 166:105521. [PMID: 33662574 DOI: 10.1016/j.phrs.2021.105521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 11/23/2022]
Abstract
Marine habitats are well-known for their diverse life forms that are potential sources of novel bioactive compounds. Evidence from existing studies suggests that these compounds contribute significantly to the field of pharmaceuticals, nutraceuticals, and cosmeceuticals. The isolation of natural compounds from a marine environment with protease inhibitory activity has gained importance due to drug discovery potential. Despite the increasing research endeavours focusing on protease inhibitors' design and characterization, many of these compounds have failed to reach final phases of clinical trials. As a result, the search for new sources for the development of protease inhibitors remains pertinent. This review focuses on the diverse marine protease inhibitors and their structure-activity relationships. Furthermore, the potential of marine protease inhibitors in drug discovery and molecular mechanism inhibitor binding are critically discussed.
Collapse
|
20
|
S. mansoni SmKI-1 Kunitz-domain: Leucine point mutation at P1 site generates enhanced neutrophil elastase inhibitory activity. PLoS Negl Trop Dis 2021; 15:e0009007. [PMID: 33465126 PMCID: PMC7846107 DOI: 10.1371/journal.pntd.0009007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/29/2021] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
The Schistosoma mansoni SmKI-1 protein is composed of two domains: a Kunitz-type serine protease inhibitor motif (KD) and a C-terminus domain with no similarity outside the genera. Our previous work has demonstrated that KD plays an essential role in neutrophil elastase (NE) binding blockage, in neutrophil influx and as a potential anti-inflammatory molecule. In order to enhance NE blocking capacity, we analyzed the KD sequence from a structure-function point of view and designed specific point mutations in order to enhance NE affinity. We substituted the P1 site residue at the reactive site for a leucine (termed RL-KD), given its central role for KD's inhibition to NE. We have also substituted a glutamic acid that strongly interacts with the P1 residue for an alanine, to help KD to be buried on NE S1 site (termed EA-KD). KD and the mutant proteins were evaluated in silico by molecular docking to human NE, expressed in Escherichia coli and tested towards its NE inhibitory activity. Both mutated proteins presented enhanced NE inhibitory activity in vitro and RL-KD presented the best performance. We further tested RL-KD in vivo in an experimental model of monosodium urate (MSU)-induced acute arthritis. RL-KD showed reduced numbers of total cells and neutrophils in the mouse knee cavity when compared to KD. Nevertheless, both RL-KD and KD reduced mice hypernociception in a similar fashion. In summary, our results demonstrated that both mutated proteins showed enhanced NE inhibitory activity in vitro. However, RL-KD had a prominent effect in diminishing inflammatory parameters in vivo.
Collapse
|
21
|
Leung TCN, Qu Z, Nong W, Hui JHL, Ngai SM. Proteomic Analysis of the Venom of Jellyfishes Rhopilema esculentum and Sanderia malayensis. Mar Drugs 2020; 18:md18120655. [PMID: 33371176 PMCID: PMC7766711 DOI: 10.3390/md18120655] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/26/2022] Open
Abstract
Venomics, the study of biological venoms, could potentially provide a new source of therapeutic compounds, yet information on the venoms from marine organisms, including cnidarians (sea anemones, corals, and jellyfish), is limited. This study identified the putative toxins of two species of jellyfish—edible jellyfish Rhopilema esculentum Kishinouye, 1891, also known as flame jellyfish, and Amuska jellyfish Sanderia malayensis Goette, 1886. Utilizing nano-flow liquid chromatography tandem mass spectrometry (nLC–MS/MS), 3000 proteins were identified from the nematocysts in each of the above two jellyfish species. Forty and fifty-one putative toxins were identified in R. esculentum and S. malayensis, respectively, which were further classified into eight toxin families according to their predicted functions. Amongst the identified putative toxins, hemostasis-impairing toxins and proteases were found to be the most dominant members (>60%). The present study demonstrates the first proteomes of nematocysts from two jellyfish species with economic and environmental importance, and expands the foundation and understanding of cnidarian toxins.
Collapse
Affiliation(s)
- Thomas C. N. Leung
- State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China;
| | - Zhe Qu
- Simon F.S. Li Marine Science Laboratory, State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China; (Z.Q.); (W.N.)
| | - Wenyan Nong
- Simon F.S. Li Marine Science Laboratory, State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China; (Z.Q.); (W.N.)
| | - Jerome H. L. Hui
- Simon F.S. Li Marine Science Laboratory, State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China; (Z.Q.); (W.N.)
- Correspondence: (J.H.L.H.); (S.M.N.)
| | - Sai Ming Ngai
- State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China;
- Correspondence: (J.H.L.H.); (S.M.N.)
| |
Collapse
|
22
|
Barati M, Javanmardi F, Mousavi Jazayeri SMH, Jabbari M, Rahmani J, Barati F, Nickho H, Davoodi SH, Roshanravan N, Mousavi Khaneghah A. Techniques, perspectives, and challenges of bioactive peptide generation: A comprehensive systematic review. Compr Rev Food Sci Food Saf 2020; 19:1488-1520. [PMID: 33337080 DOI: 10.1111/1541-4337.12578] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 04/03/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Due to the digestible refractory and absorbable structures of bioactive peptides (BPs), they could induce notable biological impacts on the living organism. In this regard, the current study was devoted to providing an overview regarding the available methods for BPs generation by the aid of a systematic review conducted on the published articles up to April 2019. In this context, the PubMed and Scopus databases were screened to retrieve the related publications. According to the results, although the characterization of BPs mainly has been performed using enzymatic and microbial in-vitro methods, they cannot be considered as suitable techniques for further stimulation of digestion in the gastrointestinal tract. Therefore, new approaches for both in-vivo and in-silico methods for BPs identification should be developed to overcome the obstacles that belonged to the current methods. The purpose of this review was to compile the recent analytical methods applied for studying various aspects of food-derived biopeptides, and emphasizing generation at in vitro, in vivo, and in silico.
Collapse
Affiliation(s)
- Meisam Barati
- Student Research Committee, Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fardin Javanmardi
- Department of Food Science and Technology, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Masoumeh Jabbari
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamal Rahmani
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzaneh Barati
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Hamid Nickho
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sayed Hossein Davoodi
- Department of Clinical Nutrition and Dietetic, National Institute and Faculty of Nutrition and Food Technology; Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Mousavi Khaneghah
- Department of Food Science, Faculty of Food Engineering, University of Campinas (UNICAMP), São Paulo, Brazil
| |
Collapse
|
23
|
Kunitz-Type Peptides from the Sea Anemone Heteractis crispa Demonstrate Potassium Channel Blocking and Anti-Inflammatory Activities. Biomedicines 2020; 8:biomedicines8110473. [PMID: 33158163 PMCID: PMC7694175 DOI: 10.3390/biomedicines8110473] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
The Kunitz/BPTI peptide family includes unique representatives demonstrating various biological activities. Electrophysiological screening of peptides HCRG1 and HCRG2 from the sea anemone Heteractis crispa on six Kv1.x channel isoforms and insect Shaker IR channel expressed in Xenopus laevis oocytes revealed their potassium channels blocking activity. HCRG1 and HCRG2 appear to be the first Kunitz-type peptides from sea anemones blocking Kv1.3 with IC50 of 40.7 and 29.7 nM, respectively. In addition, peptides mainly vary in binding affinity to the Kv1.2 channels. It was established that the single substitution, Ser5Leu, in the TRPV1 channel antagonist, HCRG21, induces weak blocking activity of Kv1.1, Kv1.2, and Kv1.3. Apparently, for the affinity and selectivity of Kunitz-fold toxins to Kv1.x isoforms, the number and distribution along their molecules of charged, hydrophobic, and polar uncharged residues, as well as the nature of the channel residue at position 379 (Tyr, Val or His) are important. Testing the compounds in a model of acute local inflammation induced by the introduction of carrageenan administration into mice paws revealed that HCRG1 at doses of 0.1–1 mg/kg reduced the volume of developing edema during 24 h, similar to the effect of the nonsteroidal anti-inflammatory drug, indomethacin, at a dose of 5 mg/kg. ELISA analysis of the animals blood showed that the peptide reduced the synthesis of TNF-α, a pro-inflammatory mediator playing a leading role in the development of edema in this model.
Collapse
|
24
|
Droctové L, Lancien M, Tran VL, Susset M, Jego B, Theodoro F, Kessler P, Mourier G, Robin P, Diarra SS, Palea S, Flahault A, Chorfa A, Corbani M, Llorens-Cortes C, Mouillac B, Mendre C, Pruvost A, Servent D, Truillet C, Gilles N. A snake toxin as a theranostic agent for the type 2 vasopressin receptor. Am J Cancer Res 2020; 10:11580-11594. [PMID: 33052234 PMCID: PMC7545998 DOI: 10.7150/thno.47485] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/04/2020] [Indexed: 01/01/2023] Open
Abstract
Rationale: MQ1, a snake toxin which targets with high nanomolar affinity and absolute selectivity for the type 2 vasopressin receptor (V2R), is a drug candidate for renal diseases and a molecular probe for imaging cells or organs expressing V2R. Methods: MQ1's pharmacological properties were characterized and applied to a rat model of hyponatremia. Its PK/PD parameters were determined as well as its therapeutic index. Fluorescently and radioactively labeled MQ1 were chemically synthesized and associated with moderate loss of affinity. MQ1's dynamic biodistribution was monitored by positron emission tomography. Confocal imaging was used to observe the labeling of three cancer cell lines. Results: The inverse agonist property of MQ1 very efficiently prevented dDAVP-induced hyponatremia in rats with low nanomolar/kg doses and with a very large therapeutic index. PK (plasma MQ1 concentrations) and PD (diuresis) exhibited a parallel biphasic decrease. The dynamic biodistribution showed that MQ1 targets the kidneys and then exhibits a blood and kidney biphasic decrease. Whatever the approach used, we found a T1/2α between 0.9 and 3.8 h and a T1/2β between 25 and 46 h and demonstrated that the kidneys were able to retain MQ1. Finally, the presence of functional V2R expressed at the membrane of cancer cells was, for the first time, demonstrated with a specific fluorescent ligand. Conclusion: As the most selective V2 binder, MQ1 is a new promising drug for aquaresis-related diseases and a molecular probe to visualize in vitro and in vivo V2R expressed physiologically or under pathological conditions.
Collapse
|
25
|
Mourão CBF, Brand GD, Fernandes JPC, Prates MV, Bloch C, Barbosa JARG, Freitas SM, Restano-Cassulini R, Possani LD, Schwartz EF. Head-to-Tail Cyclization after Interaction with Trypsin: A Scorpion Venom Peptide that Resembles Plant Cyclotides. J Med Chem 2020; 63:9500-9511. [PMID: 32787139 DOI: 10.1021/acs.jmedchem.0c00686] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Peptidase inhibitors (PIs) have been broadly studied due to their wide therapeutic potential for human diseases. A potent trypsin inhibitor from Tityus obscurus scorpion venom was characterized and named ToPI1, with 33 amino acid residues and three disulfide bonds. The X-ray structure of the ToPI1:trypsin complex, in association with the mass spectrometry data, indicate a sequential set of events: the complex formation with the inhibitor Lys32 in the trypsin S1 pocket, the inhibitor C-terminal residue Ser33 cleavage, and the cyclization of ToPI1 via a peptide bond between residues Ile1 and Lys32. Kinetic and thermodynamic characterization of the complex was obtained. ToPI1 shares no sequence similarity with other PIs characterized to date and is the first PI with CS-α/β motif described from animal venoms. In its cyclic form, it shares structural similarities with plant cyclotides that also inhibit trypsin. These results bring new insights for studies with venom compounds, PIs, and drug design.
Collapse
Affiliation(s)
- Caroline B F Mourão
- Neuropharma Lab, Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brası́lia, Brasília-DF 70910-900, Brazil.,Instituto Federal de Brası́lia, Campus Ceilándia, Brası́lia-DF 72220-260, Brazil
| | - Guilherme D Brand
- Laboratório de Sı́ntese e Análise de Biomoléculas, LSAB, Instituto de Quı́mica, Universidade de Brası́lia, Brası́lia-DF 70910-900, Brazil
| | - João Paulo C Fernandes
- Laboratório de Biofı́sica Molecular, Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brası́lia, Brası́lia-DF 70910-900, Brazil
| | - Maura V Prates
- Laboratório de Espectrometria de Massa, EMBRAPA Recursos Genéticos e Biotecnologia, Brası́lia-DF 70770-917, Brazil
| | - Carlos Bloch
- Laboratório de Espectrometria de Massa, EMBRAPA Recursos Genéticos e Biotecnologia, Brası́lia-DF 70770-917, Brazil
| | - João Alexandre R G Barbosa
- Laboratório de Biofı́sica Molecular, Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brası́lia, Brası́lia-DF 70910-900, Brazil
| | - Sônia M Freitas
- Laboratório de Biofı́sica Molecular, Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brası́lia, Brası́lia-DF 70910-900, Brazil
| | - Rita Restano-Cassulini
- Instituto de Biotecnologı́a, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Lourival D Possani
- Instituto de Biotecnologı́a, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Elisabeth F Schwartz
- Neuropharma Lab, Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brası́lia, Brasília-DF 70910-900, Brazil
| |
Collapse
|
26
|
Shintre NA, Tamhane VA, Baig UI, Pund AS, Patwardhan RB, Deshpande NM. Diversity of Culturable Actinobacteria Producing Protease Inhibitors Isolated from the Intertidal Zones of Maharashtra, India. Curr Microbiol 2020; 77:3555-3564. [PMID: 32902705 DOI: 10.1007/s00284-020-02174-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/21/2020] [Indexed: 11/26/2022]
Abstract
Phylogenetic diversity of culturable actinobacteria isolated from the intertidal regions of west coast of Maharashtra, India was studied using 16S rRNA gene sequencing. Total of 140 actinobacterial isolates were obtained, which belonged to 14 genera, 10 families and 65 putative species with Streptomyces being the most dominant (63%) genus followed by Nocardiopsis and Micromonospora. Isolates were screened for production of extracellular protease inhibitors (PI) against three pure proteases viz. chymotrypsin, trypsin, subtilisin and a crude extracellular protease from Pseudomonas aeruginosa. Eighty percent of the isolates showed PI activity against at least one of the four proteases, majority of these belonged to genus Streptomyces. Actinobacterial diversity from two sites Ade (17° 52' N, 73° 04' E) and Harnai (17° 48' N, 73° 05' E) with varying anthropological pressure showed that more putative species diversity was obtained from site with lower human intervention i.e. Ade (Shannon's H 3.45) than from Harnai (Shannon's H 2.83), a site with more human intervention. However, in Ade, percentage of isolates not showing PI activity against any of the proteases was close to 21% and that in Harnai was close to 9%. In other words, percentage of PI producers was lower at a site with lesser human intervention.
Collapse
Affiliation(s)
- Neha A Shintre
- Department of Microbiology, M.E.S. Abasaheb Garware College, Karve Road, Pune, Maharashtra, 411004, India
| | - Vaijayanti A Tamhane
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, 411007, India
| | - Ulfat I Baig
- Indian Institute of Science Education and Research, Pune (IISER-P), Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India
| | - Anagha S Pund
- Indian Institute of Science Education and Research, Pune (IISER-P), Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India
| | - Rajashree B Patwardhan
- Department of Microbiology, Haribhai V. Desai College of Commerce, Arts and Science, Pune, Maharashtra, 411002, India
| | - Neelima M Deshpande
- Department of Microbiology, M.E.S. Abasaheb Garware College, Karve Road, Pune, Maharashtra, 411004, India.
| |
Collapse
|
27
|
Zhu W, Gao H, Luo X, Ye X, Ding L, Hao J, Shu Z, Li S, Li J, Chen Z. Cloning and identification of a new multifunctional Ascaris-type peptide from the hemolymph of Buthus martensii Karsch. Toxicon 2020; 184:167-174. [PMID: 32565098 DOI: 10.1016/j.toxicon.2020.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 10/24/2022]
Abstract
Only a few work have been done for peptides from non-venom gland tissues of venomous animals. Here, with the help of the whole body transcriptomic and the hemolymph proteomic data of the Chinese scorpion Buthus martensii Karsch, we identified the first Ascaris-type peptide BmHDP from scorpion hemolymph. The precursor of BmHDP has 80 residues, including a 16 residue signal peptide and a 64 residue mature peptide. The mature peptide has 10 conserved cysteines and adopts a conserved Ascaris-type fold. Using combined inclusion body refolding and biochemical identification strategies, recombinant BmHDP was obtained successfully. Protease inhibitory assays showed that BmHDP inhibited chymotrypsin apparently at a concentration of 8 nM. Patch-clamp experiments showed that BmHDP inhibited the Kv1.3 potassium channel apparently at a concentration of 1000 nM. Coagulation experiment assays showed that BmHDP inhibited intrinsic coagulation pathway apparently at a concentration of 500 nM. To the best of our knowledge, BmHDP is the first Ascaris-type peptide from scorpion hemolymph. Our work highlighted a functional link between scorpion non-venom gland peptides and venom gland toxin peptides, and suggested that scorpion hemolymph might be a new source of bioactive peptides.
Collapse
Affiliation(s)
- Wen Zhu
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Huanhuan Gao
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Xudong Luo
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China; Institute of Biomedicine and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Hubei, China
| | - Xiangdong Ye
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China; Institute of Biomedicine and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Hubei, China
| | - Li Ding
- Department of Clinical Laboratory, Dongfeng Hospital, Hubei University of Medicine, Hubei, China; Institute of Biomedicine and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Hubei, China
| | - Jinbo Hao
- Department of Clinical Laboratory, Shiyan Occupational Disease Hospital, Hubei, China
| | - Zhan Shu
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Shan Li
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Jian Li
- Department of Human Parasitology, College of Basic Medical Sciences, Hubei University of Medicine, Hubei, China
| | - Zongyun Chen
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China; Institute of Biomedicine and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Hubei, China.
| |
Collapse
|
28
|
Finol-Urdaneta RK, Belovanovic A, Micic-Vicovac M, Kinsella GK, McArthur JR, Al-Sabi A. Marine Toxins Targeting Kv1 Channels: Pharmacological Tools and Therapeutic Scaffolds. Mar Drugs 2020; 18:E173. [PMID: 32245015 PMCID: PMC7143316 DOI: 10.3390/md18030173] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/15/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
Toxins from marine animals provide molecular tools for the study of many ion channels, including mammalian voltage-gated potassium channels of the Kv1 family. Selectivity profiling and molecular investigation of these toxins have contributed to the development of novel drug leads with therapeutic potential for the treatment of ion channel-related diseases or channelopathies. Here, we review specific peptide and small-molecule marine toxins modulating Kv1 channels and thus cover recent findings of bioactives found in the venoms of marine Gastropod (cone snails), Cnidarian (sea anemones), and small compounds from cyanobacteria. Furthermore, we discuss pivotal advancements at exploiting the interaction of κM-conotoxin RIIIJ and heteromeric Kv1.1/1.2 channels as prevalent neuronal Kv complex. RIIIJ's exquisite Kv1 subtype selectivity underpins a novel and facile functional classification of large-diameter dorsal root ganglion neurons. The vast potential of marine toxins warrants further collaborative efforts and high-throughput approaches aimed at the discovery and profiling of Kv1-targeted bioactives, which will greatly accelerate the development of a thorough molecular toolbox and much-needed therapeutics.
Collapse
Affiliation(s)
- Rocio K. Finol-Urdaneta
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia;
- Electrophysiology Facility for Cell Phenotyping and Drug Discovery, Wollongong, NSW 2522, Australia
| | - Aleksandra Belovanovic
- College of Engineering and Technology, American University of the Middle East, Kuwait; (A.B.); (M.M.-V.)
| | - Milica Micic-Vicovac
- College of Engineering and Technology, American University of the Middle East, Kuwait; (A.B.); (M.M.-V.)
| | - Gemma K. Kinsella
- School of Food Science and Environmental Health, College of Sciences and Health, Technological University Dublin, D07 ADY7 Dublin, Ireland;
| | - Jeffrey R. McArthur
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia;
| | - Ahmed Al-Sabi
- College of Engineering and Technology, American University of the Middle East, Kuwait; (A.B.); (M.M.-V.)
| |
Collapse
|
29
|
Kvetkina A, Leychenko E, Chausova V, Zelepuga E, Chernysheva N, Guzev K, Pislyagin E, Yurchenko E, Menchinskaya E, Aminin D, Kaluzhskiy L, Ivanov A, Peigneur S, Tytgat J, Kozlovskaya E, Isaeva M. A new multigene HCIQ subfamily from the sea anemone Heteractis crispa encodes Kunitz-peptides exhibiting neuroprotective activity against 6-hydroxydopamine. Sci Rep 2020; 10:4205. [PMID: 32144281 PMCID: PMC7060258 DOI: 10.1038/s41598-020-61034-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/04/2020] [Indexed: 12/14/2022] Open
Abstract
The Kunitz/BPTI-type peptides are ubiquitous in numerous organisms including marine venomous animals. The peptides demonstrate various biological activities and therefore they are the subject of a number of investigations. We have discovered a new HCIQ subfamily belonging to recently described multigene HCGS family of Heteractis crispa Kunitz-peptides. The uniqueness of this subfamily is that the HCIQ precursors contain a propeptide terminating in Lys-Arg (endopeptidase cleavage site) the same as in the neuro- and cytotoxin ones. Moreover, the HCIQ genes contain two introns in contrast to HCGS genes with one intron. As a result of Sanger and amplicon deep sequencings, 24 HCIQ isoforms were revealed. The recombinant peptides for the most prevalent isoform (HCIQ2c1) and for the isoform with the rare substitution Gly17Glu (HCIQ4c7) were obtained. They can inhibit trypsin with Ki 5.2 × 10-8 M and Ki 1.9 × 10-7 M, respectively, and interact with some serine proteinases including inflammatory ones according to the SPR method. For the first time, Kunitz-peptides have shown to significantly increase neuroblastoma cell viability in an in vitro 6-OHDA-induced neurotoxicity model being a consequence of an effective decrease of ROS level in the cells.
Collapse
Affiliation(s)
- Aleksandra Kvetkina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Elena Leychenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia.
| | - Victoria Chausova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Elena Zelepuga
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Nadezhda Chernysheva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Konstantin Guzev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Evgeny Pislyagin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Ekaterina Yurchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Ekaterina Menchinskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Dmitry Aminin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Leonid Kaluzhskiy
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., Moscow, 119121, Russia
| | - Alexis Ivanov
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., Moscow, 119121, Russia
| | - Steve Peigneur
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg, O&N2, Herestraat 49, P.O. Box 922, Leuven, B-3000, Belgium
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg, O&N2, Herestraat 49, P.O. Box 922, Leuven, B-3000, Belgium
| | - Emma Kozlovskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Marina Isaeva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| |
Collapse
|
30
|
Hart CE, Ribeiro JM, Kazimirova M, Thangamani S. Tick-Borne Encephalitis Virus Infection Alters the Sialome of Ixodes ricinus Ticks During the Earliest Stages of Feeding. Front Cell Infect Microbiol 2020; 10:41. [PMID: 32133301 PMCID: PMC7041427 DOI: 10.3389/fcimb.2020.00041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/21/2020] [Indexed: 01/14/2023] Open
Abstract
Ticks are hematophagous arthropods that transmit a number of pathogens while feeding. Among these is tick-borne encephalitis virus (TBEV), a flavivirus transmitted by Ixodes ricinus ticks in the temperate zone of Europe. The infection results in febrile illness progressing to encephalitis and meningitis with a possibility of fatality or long-term neurological sequelae. The composition of tick saliva plays an essential role in the initial virus transmission during tick feeding. Ticks secrete a diverse range of salivary proteins to modulate the host response, such as lipocalins to control the itch and inflammatory response, and both proteases and protease inhibitors to prevent blood coagulation. Here, the effect of viral infection of adult females of Ixodes ricinus was studied with the goal of determining how the virus alters the tick sialome to modulate host tissue response at the site of infection. Uninfected ticks or those infected with TBEV were fed on mice and removed and dissected one- and 3-h post-attachment. RNA from the salivary glands of these ticks, as well as from unfed ticks, was extracted and subjected to next-generation sequencing to determine the expression of key secreted proteins at each timepoint. Genes showing statistically significant up- or down-regulation between infected and control ticks were selected and compared to published literature to ascertain their function. From this, the effect of tick viral infection on the modulation of the tick-host interface was determined. Infected ticks were found to differentially express a number of uncategorized genes, proteases, Kunitz-type serine protease inhibitors, cytotoxins, and lipocalins at different timepoints. These virus-induced changes to the tick sialome may play a significant role in facilitating virus transmission during the early stages of tick feeding.
Collapse
Affiliation(s)
- Charles E. Hart
- SUNY Center for Environmental Health and Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
- Institute for Global Health and Translational Science, SUNY Upstate Medical University, Syracuse, NY, United States
- The Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, United States
| | - Jose M. Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Maria Kazimirova
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Saravanan Thangamani
- SUNY Center for Environmental Health and Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
- Institute for Global Health and Translational Science, SUNY Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
31
|
Kodama RT, Kuniyoshi AK, Silva CCFD, Cajado-Carvalho D, Duzzi B, Mariano DC, Pimenta DC, Borges R, Silva WDD, Portaro FCV. A Kunitz-type peptide from Dendroaspis polylepis venom as a simultaneous inhibitor of serine and cysteine proteases. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20200037. [PMID: 33088285 PMCID: PMC7546081 DOI: 10.1590/1678-9199-jvatitd-2020-0037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/26/2020] [Indexed: 11/22/2022] Open
|
32
|
Cid-Uribe JI, Veytia-Bucheli JI, Romero-Gutierrez T, Ortiz E, Possani LD. Scorpion venomics: a 2019 overview. Expert Rev Proteomics 2019; 17:67-83. [DOI: 10.1080/14789450.2020.1705158] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jimena I. Cid-Uribe
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - José Ignacio Veytia-Bucheli
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Teresa Romero-Gutierrez
- Departamento de Ciencias Computacionales, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Mexico
| | - Ernesto Ortiz
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Lourival D. Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| |
Collapse
|
33
|
Chen X, Leahy D, Van Haeften J, Hartfield P, Prentis PJ, van der Burg CA, Surm JM, Pavasovic A, Madio B, Hamilton BR, King GF, Undheim EAB, Brattsand M, Harris JM. A Versatile and Robust Serine Protease Inhibitor Scaffold from Actinia tenebrosa. Mar Drugs 2019; 17:E701. [PMID: 31842369 PMCID: PMC6950308 DOI: 10.3390/md17120701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022] Open
Abstract
Serine proteases play pivotal roles in normal physiology and a spectrum of patho-physiological processes. Accordingly, there is considerable interest in the discovery and design of potent serine protease inhibitors for therapeutic applications. This led to concerted efforts to discover versatile and robust molecular scaffolds for inhibitor design. This investigation is a bioprospecting study that aims to isolate and identify protease inhibitors from the cnidarian Actinia tenebrosa. The study isolated two Kunitz-type protease inhibitors with very similar sequences but quite divergent inhibitory potencies when assayed against bovine trypsin, chymostrypsin, and a selection of human sequence-related peptidases. Homology modeling and molecular dynamics simulations of these inhibitors in complex with their targets were carried out and, collectively, these methodologies enabled the definition of a versatile scaffold for inhibitor design. Thermal denaturation studies showed that the inhibitors were remarkably robust. To gain a fine-grained map of the residues responsible for this stability, we conducted in silico alanine scanning and quantified individual residue contributions to the inhibitor's stability. Sequences of these inhibitors were then used to search for Kunitz homologs in an A. tenebrosa transcriptome library, resulting in the discovery of a further 14 related sequences. Consensus analysis of these variants identified a rich molecular diversity of Kunitz domains and expanded the palette of potential residue substitutions for rational inhibitor design using this domain.
Collapse
Affiliation(s)
- Xingchen Chen
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Darren Leahy
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Jessica Van Haeften
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Perry Hartfield
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Peter J. Prentis
- School of Earth, Environmental and Biological Sciences, Science and Engineering Faculty, Queensland University of Technology, Brisbane, QLD 4000, Australia;
- Institute for Future Environments, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Chloé A. van der Burg
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Joachim M. Surm
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Ana Pavasovic
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Bruno Madio
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD 4072, Australia; (B.M.); (G.F.K.)
| | - Brett R. Hamilton
- Centre for Advanced Imaging, University of Queensland, St Lucia, QLD 4072, Australia; (B.R.H.); (E.A.B.U.)
| | - Glenn F. King
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD 4072, Australia; (B.M.); (G.F.K.)
| | - Eivind A. B. Undheim
- Centre for Advanced Imaging, University of Queensland, St Lucia, QLD 4072, Australia; (B.R.H.); (E.A.B.U.)
- Centre for Biodiversity Dynamics, Department of Biology, Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, PO Box 1066 Blindern, 0316 Oslo, Norway
| | - Maria Brattsand
- Department of Medical Biosciences, Umeå University, 901 87 Umeå, Sweden;
| | - Jonathan M. Harris
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| |
Collapse
|
34
|
Liang H, Jiang G, Wang T, Zhang J, Liu W, Xu Z, Zhang J, Xiao L. An integrated transcriptomic and proteomic analysis reveals toxin arsenal of a novel Antarctic jellyfish Cyanea sp. J Proteomics 2019; 208:103483. [DOI: 10.1016/j.jprot.2019.103483] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/17/2019] [Accepted: 08/07/2019] [Indexed: 11/27/2022]
|
35
|
Ramírez-Carreto S, Vera-Estrella R, Portillo-Bobadilla T, Licea-Navarro A, Bernaldez-Sarabia J, Rudiño-Piñera E, Verleyen JJ, Rodríguez E, Rodríguez-Almazán C. Transcriptomic and Proteomic Analysis of the Tentacles and Mucus of Anthopleura dowii Verrill, 1869. Mar Drugs 2019; 17:md17080436. [PMID: 31349621 PMCID: PMC6722582 DOI: 10.3390/md17080436] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 02/07/2023] Open
Abstract
Sea anemone venom contains a complex and diverse arsenal of peptides and proteins of pharmacological and biotechnological interest, however, only venom from a few species has been explored from a global perspective to date. In the present study, we identified the polypeptides present in the venom of the sea anemone Anthopleura dowii Verrill, 1869 through a transcriptomic and proteomic analysis of the tentacles and the proteomic profile of the secreted mucus. In our transcriptomic results, we identified 261 polypeptides related to or predicted to be secreted in the venom, including proteases, neurotoxins that could act as either potassium (K+) or sodium (Na+) channels inhibitors, protease inhibitors, phospholipases A2, and other polypeptides. Our proteomic data allowed the identification of 156 polypeptides—48 exclusively identified in the mucus, 20 in the tentacles, and 88 in both protein samples. Only 23 polypeptides identified by tandem mass spectrometry (MS/MS) were related to the venom and 21 exclusively identified in the mucus, most corresponding to neurotoxins and hydrolases. Our data contribute to the knowledge of evolutionary and venomic analyses of cnidarians, particularly of sea anemones.
Collapse
Affiliation(s)
- Santos Ramírez-Carreto
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Cuernavaca, Morelos 62210, México
| | - Rosario Vera-Estrella
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Cuernavaca, Morelos 62210, México
| | - Tobías Portillo-Bobadilla
- Unidad de Bioinformática, Bioestadística y Biología Computacional. Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México-Instituto Nacional De Ciencias Médicas y Nutrición Salvador Zubirán, Calle Vasco de Quiroga 15, Tlalpan, C.P. 14080, Ciudad de México, México
| | - Alexei Licea-Navarro
- Departamento de Innovación Biomédica, CICESE, Carretera Ensenada-Tijuana 3918, Ensenada, BC C.P. 22860, México
| | - Johanna Bernaldez-Sarabia
- Departamento de Innovación Biomédica, CICESE, Carretera Ensenada-Tijuana 3918, Ensenada, BC C.P. 22860, México
| | - Enrique Rudiño-Piñera
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Cuernavaca, Morelos 62210, México
| | - Jerome J Verleyen
- Unidad Universitaria de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Cuernavaca, Morelos 62210, México
| | - Estefanía Rodríguez
- Division of Invertebrate Zoology, American Museum of Natural History, Central Park West at 79th Street, New York, NY 10024, USA
| | - Claudia Rodríguez-Almazán
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Cuernavaca, Morelos 62210, México.
| |
Collapse
|
36
|
Kazemi SM, Sabatier JM. Venoms of Iranian Scorpions (Arachnida, Scorpiones) and Their Potential for Drug Discovery. Molecules 2019; 24:molecules24142670. [PMID: 31340554 PMCID: PMC6680535 DOI: 10.3390/molecules24142670] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/16/2019] [Accepted: 07/20/2019] [Indexed: 12/19/2022] Open
Abstract
Scorpions, a characteristic group of arthropods, are among the earliest diverging arachnids, dating back almost 440 million years. One of the many interesting aspects of scorpions is that they have venom arsenals for capturing prey and defending against predators, which may play a critical role in their evolutionary success. Unfortunately, however, scorpion envenomation represents a serious health problem in several countries, including Iran. Iran is acknowledged as an area with a high richness of scorpion species and families. The diversity of the scorpion fauna in Iran is the subject of this review, in which we report a total of 78 species and subspecies in 19 genera and four families. We also list some of the toxins or genes studied from five species, including Androctonus crassicauda, Hottentotta zagrosensis, Mesobuthus phillipsi, Odontobuthus doriae, and Hemiscorpius lepturus, in the Buthidae and Hemiscorpiidae families. Lastly, we review the diverse functions of typical toxins from the Iranian scorpion species, including their medical applications.
Collapse
Affiliation(s)
- Seyed Mahdi Kazemi
- Zagros Herpetological Institute, No 12, Somayyeh 14 Avenue, 3715688415 Qom, Iran.
| | - Jean-Marc Sabatier
- Institute of NeuroPhysiopathology, UMR 7051, Faculté de Médecine Secteur Nord, 51, Boulevard Pierre Dramard-CS80011, 13344-Marseille Cedex 15, France
| |
Collapse
|
37
|
Madio B, King GF, Undheim EAB. Sea Anemone Toxins: A Structural Overview. Mar Drugs 2019; 17:E325. [PMID: 31159357 PMCID: PMC6627431 DOI: 10.3390/md17060325] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/22/2019] [Accepted: 05/25/2019] [Indexed: 02/06/2023] Open
Abstract
Sea anemones produce venoms of exceptional molecular diversity, with at least 17 different molecular scaffolds reported to date. These venom components have traditionally been classified according to pharmacological activity and amino acid sequence. However, this classification system suffers from vulnerabilities due to functional convergence and functional promiscuity. Furthermore, for most known sea anemone toxins, the exact receptors they target are either unknown, or at best incomplete. In this review, we first provide an overview of the sea anemone venom system and then focus on the venom components. We have organised the venom components by distinguishing firstly between proteins and non-proteinaceous compounds, secondly between enzymes and other proteins without enzymatic activity, then according to the structural scaffold, and finally according to molecular target.
Collapse
Affiliation(s)
- Bruno Madio
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Eivind A B Undheim
- Centre for Advanced Imaging, The University of Queensland, St. Lucia, QLD 4072, Australia.
- Centre for Ecology and Evolutionary Synthesis, Department of Biosciences, University of Oslo, 0316 Oslo, Norway.
| |
Collapse
|
38
|
Ziegman R, Undheim EAB, Baillie G, Jones A, Alewood PF. Investigation of the estuarine stonefish (Synanceia horrida) venom composition. J Proteomics 2019; 201:12-26. [PMID: 30953730 DOI: 10.1016/j.jprot.2019.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 11/29/2022]
Abstract
The Estuarine stonefish (Synanceia horrida) is recognised as one of the most venomous fish species in the world but the overall venom composition has yet to be investigated using in-depth transcriptomic and proteomic methods. To date, known venom components are restricted to a hyaluronidase and a large, pore-forming toxin known as Stonustoxin (SNTX). Transcriptomic sequencing of the venom gland resulted in over 170,000 contigs with only 0.4% that were homologous to putative venom proteins. Integration of the transcriptomic data with proteomic data from the S. horrida venom confirmed the hyaluronidase and SNTX to be present, together with several other protein families including major contributions from C-type lectins. Other protein families observed included peroxiredoxin and several minor protein families such as Golgi-associated plant pathogenesis related proteins, tissue pathway factor inhibitors, and Kazal-type serine protease inhibitors that, although not putative venom proteins, may contribute to the venom's adverse effects. BIOLOGICAL SIGNIFICANCE: Proteomic analysis of milked Synanceia horrida venom, paired with transcriptomic analysis of the venom gland tissue revealed for the first time the composition of one of the world's most dangerous fish venoms. The results demonstrate that the venom is relatively less complex compared to other well-studied venomous animals with a number of unique proteins not previously found in animal venoms.
Collapse
Affiliation(s)
- Rebekah Ziegman
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Eivind A B Undheim
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Gregory Baillie
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Alun Jones
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia.
| |
Collapse
|
39
|
Proteomic Analysis of Novel Components of Nemopilema nomurai Jellyfish Venom: Deciphering the Mode of Action. Toxins (Basel) 2019; 11:toxins11030153. [PMID: 30857234 PMCID: PMC6468547 DOI: 10.3390/toxins11030153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
Nowadays, proliferation of jellyfish has become a severe matter in many coastal areas around the world. Jellyfish Nemopilema nomurai is one of the most perilous organisms and leads to significant deleterious outcomes such as harm to the fishery, damage the coastal equipment, and moreover, its envenomation can be hazardous to the victims. Till now, the components of Nemopilema nomurai venom (NnV) are unknown owing to scant transcriptomics and genomic data. In the current research, we have explored a proteomic approach to identify NnV components and their interrelation with pathological effects caused by the jellyfish sting. Altogether, 150 proteins were identified, comprising toxins and other distinct proteins that are substantial in nematocyst genesis and nematocyte growth by employing two-dimensional gel electrophoresis and matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI/TOF/MS). The identified toxins are phospholipase A2, phospholipase D Li Sic Tox beta IDI, a serine protease, putative Kunitz-type serine protease inhibitor, disintegrin and metalloproteinase, hemolysin, leukotoxin, three finger toxin MALT0044C, allergens, venom prothrombin activator trocarin D, tripeptide Gsp 9.1, and along with other toxin proteins. These toxins are relatively well characterized in the venoms of other poisonous species to induce pathogenesis, hemolysis, inflammation, proteolysis, blood coagulation, cytolysis, hemorrhagic activity, and type 1 hypersensitivity, suggesting that these toxins in NnV can also cause similar deleterious consequences. Our proteomic works indicate that NnV protein profile represents valuable source which leads to better understanding the clinical features of the jellyfish stings. As one of the largest jellyfish in the world, Nemopilema nomurai sting is considered to be harmful to humans due to its potent toxicity. The identification and functional characterization of its venom components have been poorly described and are beyond our knowledge. Here is the first report demonstrating the methodical overview of NnV proteomics research, providing significant information to understand the mechanism of NnV envenomation. Our proteomics findings can provide a platform for novel protein discovery and development of practical ways to deal with jellyfish stings on human beings.
Collapse
|
40
|
Liu W, Mo F, Jiang G, Liang H, Ma C, Li T, Zhang L, Xiong L, Mariottini GL, Zhang J, Xiao L. Stress-Induced Mucus Secretion and Its Composition by a Combination of Proteomics and Metabolomics of the Jellyfish Aurelia coerulea. Mar Drugs 2018; 16:E341. [PMID: 30231483 PMCID: PMC6165293 DOI: 10.3390/md16090341] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/05/2018] [Accepted: 09/09/2018] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Jellyfish respond quickly to external stress that stimulates mucus secretion as a defense. Neither the composition of secreted mucus nor the process of secretion are well understood. METHODS Aurelia coerulea jellyfish were stimulated by removing them from environmental seawater. Secreted mucus and tissue samples were then collected within 60 min, and analyzed by a combination of proteomics and metabolomics using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) and ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS/MS), respectively. RESULTS Two phases of sample collection displayed a quick decrease in volume, followed by a gradual increase. A total of 2421 and 1208 proteins were identified in tissue homogenate and secreted mucus, respectively. Gene Ontology (GO) analysis showed that the mucus-enriched proteins are mainly located in extracellular or membrane-associated regions, while the tissue-enriched proteins are distributed throughout intracellular compartments. Tryptamine, among 16 different metabolites, increased with the largest-fold change value of 7.8 in mucus, which is consistent with its involvement in the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway 'tryptophan metabolism'. We identified 11 metalloproteinases, four serpins, three superoxide dismutases and three complements, and their presence was speculated to be related to self-protective defense. CONCLUSIONS Our results provide a composition profile of proteins and metabolites in stress-induced mucus and tissue homogenate of A. coerulea. This provides insight for the ongoing endeavors to discover novel bioactive compounds. The large increase of tryptamine in mucus may indicate a strong stress response when jellyfish were taken out of seawater and the active self-protective components such as enzymes, serpins and complements potentially play a key role in innate immunity of jellyfish.
Collapse
Affiliation(s)
- Wenwen Liu
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China.
- Department of Marine Biotechnology, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China.
| | - Fengfeng Mo
- Department of Ship Hygiene, Faculty of Navy Medicine, Second Military Medical University, Shanghai 200433, China.
| | - Guixian Jiang
- Clinical Medicine, Grade 2015, Second Military Medical University, Shanghai 200433, China.
| | - Hongyu Liang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China.
- Department of Marine Biotechnology, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China.
| | - Chaoqun Ma
- Department of Marine Biotechnology, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China.
| | - Tong Li
- School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| | - Lulu Zhang
- Department of Marine Biotechnology, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China.
| | - Liyan Xiong
- Department of Traditional Chinese Medicine Identification, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Gian Luigi Mariottini
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Viale Benedetto XV 5, I-16132 Genova, Italy.
| | - Jing Zhang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China.
| | - Liang Xiao
- Department of Marine Biotechnology, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
41
|
Engineering varied serine protease inhibitors by converting P1 site of BF9, a weakly active Kunitz-type animal toxin. Int J Biol Macromol 2018; 120:1190-1197. [PMID: 30172807 DOI: 10.1016/j.ijbiomac.2018.08.178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/14/2018] [Accepted: 08/29/2018] [Indexed: 12/30/2022]
Abstract
Although there were a lot of weakly active animal toxins in the venoms, their values and applications are still mysterious, such as BF9, which is a Kunitz-type toxin isolated from the venom of the elapid snake Bungarus fasciatus. Here, we used BF9 to be a molecular scaffold, and engineered eight BF9-derived peptides by changing P1 site Asn17 of BF9, such as BF9-N17Y and BF9-N17T designed from the polar subfamily, BF9-N17L and BF9-N17G designed from the Non-polar subfamily, BF9-N17D designed from acidic subfamily, and BF9-N17H, BF9-N17K and BF9-N17R designed from basic subfamily. Through enzyme inhibitor experiment assays, we found a potent and selective chymotrypsin inhibitor BF9-N17Y, a potent and selective coagulation factor XIa inhibitor BF9-N17H, and two highly potent coagulation factor XIa inhibitors BF9-N17K and BF9-N17. APTT and PT assays further showed that BF9-N17H, BF9-N17K and BF9-N17R were three novel anticoagulants with selectively intrinsic coagulation pathway inhibitory activity. Considering that natural weakly active animal toxins are also a huge peptide resource, our present work might open a new window about pharmacological applications of weakly active animal toxins, which might be good templates for potent and selective molecular probe and lead drug designs.
Collapse
|
42
|
O'Hara EP, Caldwell GS, Bythell J. Equistatin and equinatoxin gene expression is influenced by environmental temperature in the sea anemone Actinia equina. Toxicon 2018; 153:12-16. [PMID: 30144458 DOI: 10.1016/j.toxicon.2018.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/30/2018] [Accepted: 08/19/2018] [Indexed: 01/21/2023]
Abstract
We examined the gene expression levels of equinatoxin and equistatin in the sea anemone Actinia equina, when reared at varying environmental temperatures for five months. Both genes were significantly downregulated at 10 °C compared to 16 °C but showed no significant change at 22 °C. This provides the first evidence of an effect of temperature on gene expression, but with no effect of increasing temperatures such as those predicted due to climate change.
Collapse
Affiliation(s)
- Emily P O'Hara
- School of Natural and Environmental Sciences, Ridley Building, Newcastle University, NE1 7RU, United Kingdom. e.p.o'
| | - Gary S Caldwell
- School of Natural and Environmental Sciences, Ridley Building, Newcastle University, NE1 7RU, United Kingdom.
| | - John Bythell
- School of Natural and Environmental Sciences, Ridley Building, Newcastle University, NE1 7RU, United Kingdom.
| |
Collapse
|
43
|
Islam SMA, Kearney CM, Baker EJ. Assigning biological function using hidden signatures in cystine-stabilized peptide sequences. Sci Rep 2018; 8:9049. [PMID: 29899538 PMCID: PMC5998126 DOI: 10.1038/s41598-018-27177-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 05/25/2018] [Indexed: 12/19/2022] Open
Abstract
Cystine-stabilized peptides have great utility as they naturally block ion channels, inhibit acetylcholine receptors, or inactivate microbes. However, only a tiny fraction of these peptides has been characterized. Exploration for novel peptides most efficiently starts with the identification of candidates from genome sequence data. Unfortunately, though cystine-stabilized peptides have shared structures, they have low DNA sequence similarity, restricting the utility of BLAST and even more powerful sequence alignment-based annotation algorithms, such as PSI-BLAST and HMMER. In contrast, a supervised machine learning approach may improve discovery and function assignment of these peptides. To this end, we employed our previously described m-NGSG algorithm, which utilizes hidden signatures embedded in peptide primary sequences that define and categorize structural or functional classes of peptides. From the generalized m-NGSG framework, we derived five specific models that categorize cystine-stabilized peptide sequences into specific functional classes. When compared with PSI-BLAST, HMMER and existing function-specific models, our novel approach (named CSPred) consistently demonstrates superior performance in discovery and function-assignment. We also report an interactive version of CSPred, available through download ( https://bitbucket.org/sm_islam/cystine-stabilized-proteins/src ) or web interface (watson.ecs.baylor.edu/cspred), for the discovery of cystine-stabilized peptides of specific function from genomic datasets and for genome annotation. We fully describe, in the Availability section following the Discussion, the quick and simple usage of the CsPred website to automatically deliver function assignments for batch submissions of peptide sequences.
Collapse
Affiliation(s)
- S M Ashiqul Islam
- Institute of Biomedical Studies, Baylor University, Waco, 76798, USA
| | - Christopher Michel Kearney
- Institute of Biomedical Studies, Baylor University, Waco, 76798, USA.,Department of Biology, Baylor University, Waco, 76798, USA
| | - Erich J Baker
- Institute of Biomedical Studies, Baylor University, Waco, 76798, USA. .,Department of Computer Science, Baylor University, Waco, 76798, USA.
| |
Collapse
|
44
|
Bensaoud C, Nishiyama MY, Ben Hamda C, Lichtenstein F, Castro de Oliveira U, Faria F, Loiola Meirelles Junqueira-de-Azevedo I, Ghedira K, Bouattour A, M'Ghirbi Y, Chudzinski-Tavassi AM. De novo assembly and annotation of Hyalomma dromedarii tick (Acari: Ixodidae) sialotranscriptome with regard to gender differences in gene expression. Parasit Vectors 2018; 11:314. [PMID: 29793520 PMCID: PMC5968504 DOI: 10.1186/s13071-018-2874-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/26/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hard ticks are hematophagous ectoparasites characterized by their long-term feeding. The saliva that they secrete during their blood meal is their crucial weapon against host-defense systems including hemostasis, inflammation and immunity. The anti-hemostatic, anti-inflammatory and immune-modulatory activities carried out by tick saliva molecules warrant their pharmacological investigation. The Hyalomma dromedarii Koch, 1844 tick is a common parasite of camels and probably the best adapted to deserts of all hard ticks. Like other hard ticks, the salivary glands of this tick may provide a rich source of many compounds whose biological activities interact directly with host system pathways. Female H. dromedarii ticks feed longer than males, thereby taking in more blood. To investigate the differences in feeding behavior as reflected in salivary compounds, we performed de novo assembly and annotation of H. dromedarii sialotranscriptome paying particular attention to variations in gender gene expression. RESULTS The quality-filtered Illumina sequencing reads deriving from a cDNA library of salivary glands led to the assembly of 15,342 transcripts. We deduced that the secreted proteins included: metalloproteases, glycine-rich proteins, mucins, anticoagulants of the mandanin family and lipocalins, among others. Expression analysis revealed differences in the expression of transcripts between male and female H. dromedarii that might explain the blood-feeding strategies employed by both genders. CONCLUSIONS The annotated sialome of H. dromedarii helps understand the interaction of tick-host molecules during blood-feeding and can lead to the discovery of new pharmacologically active proteins of ticks of the genus Hyalomma.
Collapse
Affiliation(s)
- Chaima Bensaoud
- Université de Tunis El Manar, Institut Pasteur de Tunis, LR11IPT03, Service d'entomologie médicale, 1002, Tunis, Tunisie
| | - Milton Yutaka Nishiyama
- Laboratório Especial de Toxinologia Aplicada, CeTICS, Instituto Butantan, Av. Vital Brazil, 1500, CEP, São Paulo, 05503-900, Brazil
| | - Cherif Ben Hamda
- Université de Tunis El Manar, Institut Pasteur de Tunis, LR11IPT09, Laboratoire de Bioinformatique, Biomathematique et biostatiqtiques, 1002, Tunis, Tunisie
| | - Flavio Lichtenstein
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brazil, 1500, CEP, São Paulo, 05503-900, Brazil
| | - Ursula Castro de Oliveira
- Laboratório Especial de Toxinologia Aplicada, CeTICS, Instituto Butantan, Av. Vital Brazil, 1500, CEP, São Paulo, 05503-900, Brazil
| | - Fernanda Faria
- Laboratório de Biologia Molecular, Instituto Butantan, Av. Vital Brazil, 1500, CEP, São Paulo, 05503-900, Brazil
| | | | - Kais Ghedira
- Université de Tunis El Manar, Institut Pasteur de Tunis, LR11IPT09, Laboratoire de Bioinformatique, Biomathematique et biostatiqtiques, 1002, Tunis, Tunisie
| | - Ali Bouattour
- Université de Tunis El Manar, Institut Pasteur de Tunis, LR11IPT03, Service d'entomologie médicale, 1002, Tunis, Tunisie.
| | - Youmna M'Ghirbi
- Université de Tunis El Manar, Institut Pasteur de Tunis, LR11IPT03, Service d'entomologie médicale, 1002, Tunis, Tunisie
| | | |
Collapse
|
45
|
Santibáñez-López CE, Ontano AZ, Harvey MS, Sharma PP. Transcriptomic Analysis of Pseudoscorpion Venom Reveals a Unique Cocktail Dominated by Enzymes and Protease Inhibitors. Toxins (Basel) 2018; 10:E207. [PMID: 29783636 PMCID: PMC5983263 DOI: 10.3390/toxins10050207] [Citation(s) in RCA: 19] [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: 04/18/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/19/2022] Open
Abstract
Transcriptomic and genomic analyses have illuminated the diversity of venoms in three of the four venomous arachnid orders (scorpions, spiders, and ticks). To date, no venom gland transcriptome analysis has been available for pseudoscorpions, the fourth venomous arachnid lineage. To redress this gap, we sequenced an mRNA library generated from the venom glands of the species Synsphyronus apimelus (Garypidae). High-throughput sequencing by the Illumina protocol, followed by de novo assembly, resulted in a total of 238,331 transcripts. From those, we annotated 131 transcripts, which code for putative peptides/proteins with similar sequences to previously reported venom components available from different arachnid species in protein databases. Transcripts putatively coding for enzymes showed the richest diversity, followed by other venom components such as peptidase inhibitors, cysteine-rich peptides, and thyroglobulin 1-like peptides. Only 11 transcripts were found that code for putatively low molecular mass spider toxins. This study constitutes the first report of the diversity of components within pseudoscorpion venom.
Collapse
Affiliation(s)
- Carlos E Santibáñez-López
- Department of Integrative Biology, University of Wisconsin-Madison, 430 Lincoln Drive, Madison, WI 53706, USA.
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, Ciudad de México C.P. 04510, Mexico.
| | - Andrew Z Ontano
- Department of Integrative Biology, University of Wisconsin-Madison, 430 Lincoln Drive, Madison, WI 53706, USA.
| | - Mark S Harvey
- Department of Terrestrial Zoology, Western Australian Museum, Locked Bag 49, Welshpool DC, Western Australia 6986, Australia.
| | - Prashant P Sharma
- Department of Integrative Biology, University of Wisconsin-Madison, 430 Lincoln Drive, Madison, WI 53706, USA.
| |
Collapse
|
46
|
Sabellastarte magnifica Carboxypeptidase Inhibitor: The first Kunitz inhibitor simultaneously interacting with carboxypeptidases and serine proteases. Biochimie 2018; 150:37-47. [PMID: 29730302 DOI: 10.1016/j.biochi.2018.04.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/29/2018] [Indexed: 01/14/2023]
Abstract
Multi-domain inhibitors capable to block the activity of different classes of proteases are not very common in nature. However, these kinds of molecules are attractive systems for biomedical or biotechnological applications, where two or more different targets need to be neutralized. SmCI, the Sabellastarte magnifica Carboxypeptidase Inhibitor, is a tri-domain BPTI-Kunitz inhibitor capable to inhibit serine proteases and A-like metallocarboxypeptidases. The BPTI-Kunitz family of proteins includes voltage gated channel blockers and inhibitors of serine proteases. SmCI is therefore, the only BPTI-Kunitz protein capable of inhibiting metallocarboxypeptidases. The X-ray structure of the SmCI-carboxypeptidase A complex previously obtained by us, revealed that this enzyme interacts with SmCI N-tail. In the complex, the reactive loops for serine protease inhibition remain fully exposed to the solvent in each domain, suggesting SmCI can simultaneously interact with multiple serine proteases. The twofold goals of this study were: i) to establish serine proteases-SmCI binding stoichiometry, given that the inhibitor is comprised of three potential binding domains; and ii) to determine whether or not SmCI can simultaneously bind both classes of enzymes, to which it binds individually. Our experimental approach included a variety of techniques for the study of protein-protein interactions, using as model enzymes pancreatic trypsin, elastase and carboxypeptidase A. In particular, we combined information obtained from gel filtration chromatography, denaturing electrophoresis, nuclear magnetic resonance spectroscopy and enzyme inhibition assays. Our results show that SmCI is able to bind three trypsin molecules under saturating conditions, but only one elastase interacts with the inhibitor. Additionally, we demonstrated that SmCI can bind serine proteases and carboxypeptidases at the same time (at least in the ratio 1:1:1), becoming the first protease inhibitor that simultaneously blocks these two mechanistic classes of enzymes.
Collapse
|
47
|
Loret EP, Luis J, Nuccio C, Villard C, Mansuelle P, Lebrun R, Villard PH. A Low Molecular Weight Protein from the Sea Anemone Anemonia viridis with an Anti-Angiogenic Activity. Mar Drugs 2018; 16:E134. [PMID: 29671760 PMCID: PMC5923421 DOI: 10.3390/md16040134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/06/2018] [Accepted: 04/12/2018] [Indexed: 01/16/2023] Open
Abstract
Sea anemones are a remarkable source of active principles due to a decentralized venom system. New blood vessel growth or angiogenesis is a very promising target against cancer, but the few available antiangiogenic compounds have limited efficacy. In this study, a protein fraction, purified from tentacles of Anemonia viridis, was able to limit endothelial cells proliferation and angiogenesis at low concentration (14 nM). Protein sequences were determined with Edman degradation and mass spectrometry in source decay and revealed homologies with Blood Depressing Substance (BDS) sea anemones. The presence of a two-turn alpha helix observed with circular dichroism and a trypsin activity inhibition suggested that the active principle could be a Kunitz-type inhibitor, which may interact with an integrin due to an Arginine Glycin Aspartate (RGD) motif. Molecular modeling showed that this RGD motif was well exposed to solvent. This active principle could improve antiangiogenic therapy from existing antiangiogenic compounds binding on the Vascular Endothelial Growth Factor (VEGF).
Collapse
Affiliation(s)
- Erwann P Loret
- Aix-Marseille University (AMU), Université d'Avignon, Centre National de la Recherche Scientifique (CNRS), Institut de la Recherche et du Développement (IRD), Institut Méditerranéen de Biologie et d'Ecologie. CNRS UMR 7263 IRD 237 Faculté de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille, France.
| | - José Luis
- AMU, CNRS, Institut de Neurophysio Pathologie, 13385 Marseille, France.
| | - Christopher Nuccio
- AMU, Institut National de la Santé Et de la Recherche Scientifique, 13385 Marseille, France.
| | - Claude Villard
- AMU, CNRS, Institut de Neurophysio Pathologie, 13385 Marseille, France.
| | - Pascal Mansuelle
- AMU, CNRS Formation de Recherche 3479, Institut de Microbiologie de la Méditerranée, Plateforme Protéomique, 31 Chemin Joseph Aiguier, 13402 Marseille, France.
| | - Régine Lebrun
- AMU, CNRS Formation de Recherche 3479, Institut de Microbiologie de la Méditerranée, Plateforme Protéomique, 31 Chemin Joseph Aiguier, 13402 Marseille, France.
| | - Pierre Henri Villard
- Aix-Marseille University (AMU), Université d'Avignon, Centre National de la Recherche Scientifique (CNRS), Institut de la Recherche et du Développement (IRD), Institut Méditerranéen de Biologie et d'Ecologie. CNRS UMR 7263 IRD 237 Faculté de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille, France.
| |
Collapse
|
48
|
Serine protease inhibitors containing a Kunitz domain: their role in modulation of host inflammatory responses and parasite survival. Microbes Infect 2018; 20:606-609. [PMID: 29355617 DOI: 10.1016/j.micinf.2018.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/09/2018] [Indexed: 11/21/2022]
Abstract
Proteins containing a Kunitz domain have the typical serine protease inhibition function ranging from sea anemone to man. Protease inhibitors play major roles in infection, inflammation disorders and cancer. This review discusses the role of serine proteases containing a Kunitz domain in immunomodulation induced by helminth parasites. Helminth parasites are associated with protection from inflammatory conditions. Therefore, interest has raised whether worm parasites or their products hold potential as drugs for treatment of immunological disorders. Finally, we also propose the use of recombinant SmKI-1 from Schistosoma mansoni as a potential therapeutic molecule to treat inflammatory diseases.
Collapse
|
49
|
Guarnieri MC, de Albuquerque Modesto JC, Pérez CD, Ottaiano TF, Ferreira RDS, Batista FP, de Brito MV, Campos IHMP, Oliva MLV. Zoanthid mucus as new source of useful biologically active proteins. Toxicon 2018; 143:96-107. [PMID: 29360533 DOI: 10.1016/j.toxicon.2018.01.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/02/2018] [Accepted: 01/18/2018] [Indexed: 10/18/2022]
Abstract
Palythoa caribaeorum is a very common colonial zoanthid in the coastal reefs of Brazil. It is known for its massive production of mucus, which is traditionally used in folk medicine by fishermen in northeastern Brazil. This study identified biologically active compounds in P. caribaerum mucus. Crude mucus was collected during low tides by the manual scraping of colonies; samples were maintained in an ice bath, homogenized, and centrifuged at 16,000 g for 1 h at 4 °C; the supernatant (mucus) was kept at -80 °C until use. The enzymatic (proteolytic and phospholipase A2), inhibitory (metallo, cysteine and serine proteases), and hemagglutinating (human erythrocyte) activities were determined. The results showed high levels of cysteine and metallo proteases, intermediate levels of phosholipase A2, low levels of trypsin, and no elastase and chymotrypsin like activities. The mucus showed potent inhibitory activity on snake venom metalloproteases and cysteine proteinase papain. In addition, it showed agglutinating activity towards O+, B+, and A+ erythrocyte types. The hemostatic results showed that the mucus prolongs the aPTT and PT, and strongly inhibited platelet aggregation induced by arachidonic acid, collagen, epinephrine, ADP, and thrombin. The antimicrobial activity was tested on 15 strains of bacteria and fungi through the radial diffusion assay in agar, and no activity was observed. Compounds in P. caribaeorum mucus were analyzed for the first time in this study, and our results show potential pharmacological activities in these compounds, which are relevant for use in physiopathological investigations. However, the demonstration of these activities indicates caution in the use of crude mucus in folk medicine. Furthermore, the present or absent activities identified in this mucus suggest that the studied P. caribaeorum colonies were in thermal stress conditions at the time of sample collection; these conditions may precede the bleaching process in zoanthids. Hence, the use of mucus as an indicator of this process should be evaluated in the future.
Collapse
Affiliation(s)
- Míriam Camargo Guarnieri
- Department of Zoology, Federal University of Pernambuco, Av. Prof Moraes Rego 1235, CEP 50670-901, Cidade Universitária, Recife, PE, Brazil; Biochemistry Department, Federal University of São Paulo, Rua Três de maio 100, CEP 04044-020, Vila Clementino, São Paulo, SP, Brazil.
| | - Jeanne Claíne de Albuquerque Modesto
- Vitória Academic Center, Federal University of Pernambuco, Rua Alto do Reservatório, s/n, CEP 55608-680, Bela Vista, Vitória de Santo Antão, PE, Brazil.
| | - Carlos Daniel Pérez
- Vitória Academic Center, Federal University of Pernambuco, Rua Alto do Reservatório, s/n, CEP 55608-680, Bela Vista, Vitória de Santo Antão, PE, Brazil.
| | - Tatiana Fontes Ottaiano
- Biochemistry Department, Federal University of São Paulo, Rua Três de maio 100, CEP 04044-020, Vila Clementino, São Paulo, SP, Brazil.
| | - Rodrigo da Silva Ferreira
- Biochemistry Department, Federal University of São Paulo, Rua Três de maio 100, CEP 04044-020, Vila Clementino, São Paulo, SP, Brazil.
| | - Fabrício Pereira Batista
- Biochemistry Department, Federal University of São Paulo, Rua Três de maio 100, CEP 04044-020, Vila Clementino, São Paulo, SP, Brazil.
| | - Marlon Vilela de Brito
- Biochemistry Department, Federal University of São Paulo, Rua Três de maio 100, CEP 04044-020, Vila Clementino, São Paulo, SP, Brazil.
| | - Ikaro Henrique Mendes Pinto Campos
- Department of Zoology, Federal University of Pernambuco, Av. Prof Moraes Rego 1235, CEP 50670-901, Cidade Universitária, Recife, PE, Brazil.
| | - Maria Luiza Vilela Oliva
- Biochemistry Department, Federal University of São Paulo, Rua Três de maio 100, CEP 04044-020, Vila Clementino, São Paulo, SP, Brazil.
| |
Collapse
|
50
|
Elucidating the biogeographical variation of the venom of Naja naja (spectacled cobra) from Pakistan through a venom-decomplexing proteomic study. J Proteomics 2018; 175:156-173. [DOI: 10.1016/j.jprot.2017.12.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 11/30/2017] [Accepted: 12/18/2017] [Indexed: 02/08/2023]
|