1
|
Basumatary M, Talukdar A, Sharma M, Dutta A, Mukhopadhyay R, Doley R. Exploring the anticancer potential of Cytotoxin 10 from Naja kaouthia venom: Mechanistic insights from breast and lung cancer cell lines. Chem Biol Interact 2024; 403:111254. [PMID: 39321861 DOI: 10.1016/j.cbi.2024.111254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 09/27/2024]
Abstract
Breast and lung cancers are the leading causes of cancer-related deaths in the world. Although considerable progress has been made in the field of cancer therapy, quest to discover potent, safe and cost-effective alternatives especially from natural sources is being pursued. Snake venom, which is a treasure trove of various peptides and proteins including natural toxins that specifically target tissues and receptors in the envenomated victims. Many such proteins are being explored for their therapeutic potential against various diseases including cancers. Here, we report the mechanism of cytotoxic activity of crude venom and a purified protein, Cytotoxin from the monocled cobra (Naja kaouthia), an elapid snake with neurotoxic venom prominently found in the North-East India. The crude venom showed significant cytotoxicity against breast (MCF-7and MDA-MB-231) and lung (A549, NCI-H522) cancer cell lines. Bioassay-guided fractionation using RP-HPLC showed highest cytotoxic activity in peak P9. Liquid chromatography-tandem mass spectrometry (ESI-LC-MS/MS) analysis was employed and the fraction is identified as Cytotoxin 10 which showed comparable cytotoxicity against the experimental cell lines. Cytotoxin 10 also exhibited apoptosis in MCF-7 and A549 cell lines using AO/EtBr and flow cytometry analysis. Expressions of apoptosis related proteins e.g. Bax, Bcl-2, Caspase-7 and PARP were also studied following Cytotoxin 10 treatment in both cell lines. Molecular docking experiments performed to investigate the interactions between Cytotoxin 10 and the apoptotic proteins revealed favourable binding scores compared to their corresponding inhibitors. Interestingly, Cytotoxin 10 inhibited migration and adhesion in a time and dose-dependent manner in both MCF-7 and A549 cells. This is the first report elucidating the mechanism of cytotoxic activity of Cytotoxin 10 purified from Naja kaouthia venom of North-East India origin and could pave the way for development of potential therapeutic strategies against breast and lung cancer.
Collapse
Affiliation(s)
- Mandira Basumatary
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Assam, 784028, India
| | - Amit Talukdar
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Assam, 784028, India
| | - Manoj Sharma
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Assam, 784028, India
| | - Anupam Dutta
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Assam, 784028, India
| | - Rupak Mukhopadhyay
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Assam, 784028, India.
| | - Robin Doley
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Assam, 784028, India.
| |
Collapse
|
2
|
Oliveira VQ, Santos LC, Teixeira SC, Correia TML, Andrade LOSB, Gimenes SNC, Colombini M, Marques LM, Jiménez-Charris E, Freitas-de-Sousa LA, Silva MJB, Magalhães Gusmão ACMD, Ferro EAV, Clissa PB, Melo Rodrigues VD, Lopes DS. Antiangiogenic properties of BthMP, a P-I metalloproteinase from Bothrops moojeni snake venom by VEGF pathway in endothelial cells. Biochem Biophys Res Commun 2024; 706:149748. [PMID: 38460450 DOI: 10.1016/j.bbrc.2024.149748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Angiogenesis is a process that is controlled by a delicate combination of proangiogenic and antiangiogenic molecules and can be disrupted in various illnesses, including cancer. Non-cancerous diseases can also have an abnormal or insufficient vascular growth, inflammation and hypoxia, which exacerbate angiogenesis. These conditions include atherosclerosis, psoriasis, endometriosis, asthma, obesity and AIDS. Based on that, the present work assessed the in vitro and ex vivo antiangiogenic properties stemming from BthMP, a P-I metalloproteinase from Bothrops moojeni snake venom, via the VEGF pathway. BthMP at a concentration of 5 and 40 μg/mL showed no toxicity to endothelial cells (HUVEC) in the MTT assay and was not able to induce necrosis and colony proliferation. Interestingly, BthMP inhibited adhesion, migration and invasion of HUVECs in Matrigel and arrested in vitro angiogenesis by reducing the average number of nodules in toxin-treated cells by 9.6 and 17.32 at 5 and 40 μg/mL, respectively, and the number of tubules by 15.9 at 5 μg/mL and 21.6 at 40 μg/mL in a VEGF-dependent way, an essential proangiogenic property. Furthermore, BthMP inhibited the occurrence of the angiogenic process in an ex vivo aortic ring test by decreasing new vessel formation by 52% at 5 μg/mL and by 66% at 40 μg/mL and by increasing the expression of an antiangiogenic gene, SFLT-1, and decreasing the expression of the proangiogenic genes VEGFA and ANGPT-1. Finally, this toxin reduces the production of nitric oxide, a marker that promotes angiogenesis and VEGF modulation, and decreases the protein expression of VEGFA in the supernatant of the HUVEC culture by about 30 %. These results suggest that BthMP has a promising antiangiogenic property and proves to be a biotechnological mechanism for understanding the antiangiogenic responses induced by snake venom metalloproteinases, which could be applied to a variety of diseases that exhibit an imbalance of angiogenesis mechanisms.
Collapse
Affiliation(s)
- Vinícius Queiroz Oliveira
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | - Luísa Carregosa Santos
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | - Samuel Cota Teixeira
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil.
| | | | | | | | - Mônica Colombini
- Laboratory of Immunopathology, Institute of Butantan, São Paulo, SP, Brazil
| | - Lucas Miranda Marques
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | | | | | - Marcelo José Barbosa Silva
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | | | - Eloisa Amália Vieira Ferro
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | | | - Veridiana de Melo Rodrigues
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia (UFU), Uberlândia-MG, Brazil
| | - Daiana Silva Lopes
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil.
| |
Collapse
|
3
|
Jafari Z, Sadeghi S, Dehaghi MM, Bigham A, Honarmand S, Tavasoli A, Hoseini MHM, Varma RS. Immunomodulatory activities and biomedical applications of melittin and its recent advances. Arch Pharm (Weinheim) 2024; 357:e2300569. [PMID: 38251938 DOI: 10.1002/ardp.202300569] [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: 10/06/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
Melittin (MLT), a peptide containing 26 amino acids, is a key constituent of bee venom. It comprises ∼40%-60% of the venom's dry weight and is the main pricing index for bee venom, being the causative factor of pain. The unique properties of MLT extracted from bee venom have made it a very valuable active ingredient in the pharmaceutical industry as this cationic and amphipathic peptide has propitious effects on human health in diverse biological processes. It has the ability to strongly impact the membranes of cells and display hemolytic activity with anticancer characteristics. However, the clinical application of MLT has been limited by its severe hemolytic activity, which poses a challenge for therapeutic use. By employing more efficient mechanisms, such as modifying the MLT sequence, genetic engineering, and nano-delivery systems, it is anticipated that the limitations posed by MLT can be overcome, thereby enabling its wider application in therapeutic contexts. This review has outlined recent advancements in MLT's nano-delivery systems and genetically engineered cells expressing MLT and provided an overview of where the MLTMLT's platforms are and where they will go in the future with the challenges ahead. The focus is on exploring how these approaches can overcome the limitations associated with MLT's hemolytic activity and improve its selectivity and efficacy in targeting cancer cells. These advancements hold promise for the creation of innovative and enhanced therapeutic approaches based on MLT for the treatment of cancer.
Collapse
Affiliation(s)
- Zohreh Jafari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Mirzarazi Dehaghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy (IPCB-CNR), Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
| | - Shokouh Honarmand
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Afsaneh Tavasoli
- Department of Biotechnology, Faculty of Pharmacy, Alborz University of Medical Sciences, Karaj, Iran
| | - Mostafa Haji Molla Hoseini
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rajender S Varma
- Department of Chemistry, Centre of Excellence for Research in Sustainable Chemistry, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
4
|
Almeida GDO, de Oliveira IS, Arantes EC, Sampaio SV. Snake venom disintegrins update: insights about new findings. J Venom Anim Toxins Incl Trop Dis 2023; 29:e20230039. [PMID: 37818211 PMCID: PMC10561651 DOI: 10.1590/1678-9199-jvatitd-2023-0039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/25/2023] [Indexed: 10/12/2023] Open
Abstract
Snake venom disintegrins are low molecular weight, non-enzymatic proteins rich in cysteine, present in the venom of snakes from the families Viperidae, Crotalidae, Atractaspididae, Elapidae, and Colubridae. This family of proteins originated in venom through the proteolytic processing of metalloproteinases (SVMPs), which, in turn, evolved from a gene encoding an A Disintegrin And Metalloprotease (ADAM) molecule. Disintegrins have a recognition motif for integrins in their structure, allowing interaction with these transmembrane adhesion receptors and preventing their binding to proteins in the extracellular matrix and other cells. This interaction gives disintegrins their wide range of biological functions, including inhibition of platelet aggregation and antitumor activity. As a result, many studies have been conducted in an attempt to use these natural compounds as a basis for developing therapies for the treatment of various diseases. Furthermore, the FDA has approved Tirofiban and Eptifibatide as antiplatelet compounds, and they are synthesized from the structure of echistatin and barbourin, respectively. In this review, we discuss some of the main functional and structural characteristics of this class of proteins and their potential for therapeutic use.
Collapse
Affiliation(s)
- Gabriela de Oliveira Almeida
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Isadora Sousa de Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Eliane Candiani Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
5
|
Gattringer J, Gruber CW, Hellinger R. Peptide modulators of cell migration: Overview, applications and future development. Drug Discov Today 2023; 28:103554. [PMID: 36921670 PMCID: PMC7615922 DOI: 10.1016/j.drudis.2023.103554] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Cell migration is a key physiological process in the development and homeostasis of multicellular organisms; errors in this complex system can trigger the development of cancer or inflammatory disorders. Therefore, modulating cell migration provides opportunities for drug discovery. Peptides are gaining importance on the global therapeutics market, given their unique properties compared with established small-molecule drugs or biologics. In this review, we identified over 470 peptides modulating cell migration and analyzed their characteristics. Over 95% of these peptides are in the discovery or preclinical stage, because the transition of peptide hits into drug leads often results in a bottleneck in the development process. We summarize chemical strategies in (pre-)clinical development to enhance drug-like properties of bioactive peptides.
Collapse
Affiliation(s)
- Jasmin Gattringer
- Medical University of Vienna, Center for Physiology and Pharmacology, Schwarzspanierstrasse 17, A-1090 Vienna, Austria
| | - Christian W Gruber
- Medical University of Vienna, Center for Physiology and Pharmacology, Schwarzspanierstrasse 17, A-1090 Vienna, Austria
| | - Roland Hellinger
- Medical University of Vienna, Center for Physiology and Pharmacology, Schwarzspanierstrasse 17, A-1090 Vienna, Austria.
| |
Collapse
|
6
|
Mahto AK, Kumari S, Akbar S, Paroha S, Sahoo PK, Kumar A, Dewangan RP. Peptide-Based Therapeutics and Drug Delivery Systems. DRUGS AND A METHODOLOGICAL COMPENDIUM 2023:173-211. [DOI: 10.1007/978-981-19-7952-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Abstract
INTRODUCTION Platelets play a key role in arterial thrombosis and antiplatelet therapy is pivotal in the treatment of cardiovascular disease. Current antiplatelet drugs target different pathways of platelet activation and show specific pharmacodynamic and pharmacokinetic characteristics, implicating clinically relevant drug-drug interactions. AREAS COVERED This article reviews the role of platelets in hemostasis and cardiovascular thrombosis, and discusses the key pharmacodynamics, drug-drug interactions and reversal strategies of clinically used antiplatelet drugs. EXPERT OPINION Antiplatelet therapies target distinct pathways of platelet activation: thromboxane A2 synthesis, adenosine diphosphate-mediated signaling, integrin αIIbβ3 (GPIIb/IIIa), thrombin-mediated platelet activation via the PAR1 receptor and phosphodiesterases. Key clinical drug-drug interactions of antiplatelet agents involve acetylsalicylic acid - ibuprofen, clopidogrel - omeprazole, and morphine - oral P2Y12 inhibitors, all of which lead to an attenuated antiplatelet effect. Platelet function and genetic testing and the use of scores (ARC-HBR, PRECISE-DAPT, ESC ischemic risk definition) may contribute to a more tailored antiplatelet therapy. High on-treatment platelet reactivity presents a key problem in the acute management of ST-elevation myocardial infarction (STEMI). A treatment strategy involving early initiation of an intravenous antiplatelet agent may be able to bridge the gap of insufficient platelet inhibition in high ischemic risk patients with STEMI.
Collapse
Affiliation(s)
- Georg Gelbenegger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
van den Kerkhof DL, Nagy M, Wichapong K, Brouns SL, Suylen DP, Hackeng TM, Dijkgraaf I. Unraveling the role of the homoarginine residue in antiplatelet drug eptifibatide in binding to the αIIbβ3 integrin receptor. Thromb Res 2022; 217:96-103. [DOI: 10.1016/j.thromres.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/19/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
|
9
|
Oliveira AL, Viegas MF, da Silva SL, Soares AM, Ramos MJ, Fernandes PA. The chemistry of snake venom and its medicinal potential. Nat Rev Chem 2022; 6:451-469. [PMID: 35702592 PMCID: PMC9185726 DOI: 10.1038/s41570-022-00393-7] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 12/15/2022]
Abstract
The fascination and fear of snakes dates back to time immemorial, with the first scientific treatise on snakebite envenoming, the Brooklyn Medical Papyrus, dating from ancient Egypt. Owing to their lethality, snakes have often been associated with images of perfidy, treachery and death. However, snakes did not always have such negative connotations. The curative capacity of venom has been known since antiquity, also making the snake a symbol of pharmacy and medicine. Today, there is renewed interest in pursuing snake-venom-based therapies. This Review focuses on the chemistry of snake venom and the potential for venom to be exploited for medicinal purposes in the development of drugs. The mixture of toxins that constitute snake venom is examined, focusing on the molecular structure, chemical reactivity and target recognition of the most bioactive toxins, from which bioactive drugs might be developed. The design and working mechanisms of snake-venom-derived drugs are illustrated, and the strategies by which toxins are transformed into therapeutics are analysed. Finally, the challenges in realizing the immense curative potential of snake venom are discussed, and chemical strategies by which a plethora of new drugs could be derived from snake venom are proposed.
Collapse
Affiliation(s)
- Ana L. Oliveira
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Matilde F. Viegas
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Saulo L. da Silva
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Andreimar M. Soares
- Biotechnology Laboratory for Proteins and Bioactive Compounds from the Western Amazon, Oswaldo Cruz Foundation, National Institute of Epidemiology in the Western Amazon (INCT-EpiAmO), Porto Velho, Brazil
- Sao Lucas Universitary Center (UniSL), Porto Velho, Brazil
| | - Maria J. Ramos
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Pedro A. Fernandes
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Contrasting patterns of venom regeneration in a centipede (Scolopendra viridis) and a scorpion (Centruroides hentzi). Toxicon 2022; 210:132-140. [PMID: 35245607 DOI: 10.1016/j.toxicon.2022.02.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 11/23/2022]
Abstract
As biochemical traits with clear fitness consequences, venoms serve a critical ecological role for the animals that produce them. Understanding how venoms are maintained and regenerated after use will, therefore, provide valuable insight into the ecology of venomous animals. Furthermore, most studies on venomous organisms often require removing animals from the wild and waiting extended periods of time between venom extractions. Uncovering the patterns of venom regeneration across different species will likely lead to the development of more efficient venom extraction protocols, reducing both experimental time and the number of animals required. Using reversed-phase high-performance liquid chromatography, we identified asynchronous regeneration of venom protein component abundances in the centipede Scolopendra viridis but found no evidence for asynchronous venom regeneration in the scorpion Centruroides hentzi. We also observed high levels of intraspecific venom variation in C. hentzi, emphasizing the importance of testing for intraspecific venom variation in studies evaluating the synchronicity of venom regeneration. Although the regeneration of relative venom protein component abundances is an asynchronous process in S. viridis, we provide evidence that the presence-absence of major venom components is not an asynchronous process and suggest that studies relying on just the presence/absence of individual proteins (e.g. bioprospecting, drug discovery) could use catch-and-release methods of venom extraction to reduce the number of animals removed from the wild.
Collapse
|
11
|
Vasconcelos AA, Estrada JC, David V, Wermelinger LS, Almeida FCL, Zingali RB. Structure-Function Relationship of the Disintegrin Family: Sequence Signature and Integrin Interaction. Front Mol Biosci 2021; 8:783301. [PMID: 34926583 PMCID: PMC8678471 DOI: 10.3389/fmolb.2021.783301] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/11/2021] [Indexed: 01/09/2023] Open
Abstract
Disintegrins are small cysteine-rich proteins found in a variety of snake venom. These proteins selectively modulate integrin function, heterodimeric receptors involved in cell-cell and cell-matrix interaction that are widely studied as therapeutic targets. Snake venom disintegrins emerged from the snake venom metalloproteinase and are classified according to the sequence size and number of disulfide bonds. Evolutive structure and function diversification of disintegrin family involves a stepwise decrease in the polypeptide chain, loss of cysteine residues, and selectivity. Since the structure elucidation of echistatin, the description of the structural properties of disintegrins has allowed the investigation of the mechanisms involved in integrin-cell-extracellular matrix interaction. This review provides an analysis of the structures of all family groups enabling the description of an expanded classification of the disintegrin family in seven groups. Each group presents a particular disulfide pattern and sequence signatures, facilitating the identification of new disintegrins. The classification was based on the disintegrin-like domain of the human metalloproteinase (ADAM-10). We also present the sequence and structural signatures important for disintegrin-integrin interaction, unveiling the relationship between the structure and function of these proteins.
Collapse
Affiliation(s)
- Ariana A Vasconcelos
- Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Centro Nacional de Ressonância Magnética Nuclear, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Centro Nacional de Ressonância Magnética Nuclear (CNRMN), Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge C Estrada
- Laboratório de Hemostase e Venenos, Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor David
- Laboratório de Hemostase e Venenos, Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana S Wermelinger
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio C L Almeida
- Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Centro Nacional de Ressonância Magnética Nuclear, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Centro Nacional de Ressonância Magnética Nuclear (CNRMN), Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Russolina B Zingali
- Laboratório de Hemostase e Venenos, Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Pathare SJ, Eng W, Lee SJJ, Ramasubramanian AK. Fibrin prestress due to platelet aggregation and contraction increases clot stiffness. BIOPHYSICAL REPORTS 2021; 1:100022. [PMID: 36425457 PMCID: PMC9680775 DOI: 10.1016/j.bpr.2021.100022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/08/2021] [Indexed: 06/16/2023]
Abstract
Efficient hemorrhagic control is attained through the formation of strong and stable blood clots at the site of injury. Although it is known that platelet-driven contraction can dramatically influence clot stiffness, the underlying mechanisms by which platelets assist fibrin in resisting external loads are not understood. In this study, we delineate the contribution of platelet-fibrin interactions to clot tensile mechanics using a combination of new mechanical measurements, image analysis, and structural mechanics simulation. Based on uniaxial tensile test data using custom-made microtensometer and fluorescence microscopy of platelet aggregation and platelet-fibrin interactions, we show that integrin-mediated platelet aggregation and actomyosin-driven platelet contraction synergistically increase the elastic modulus of the clots. We demonstrate that the mechanical and geometric response of an active contraction model of platelet aggregates compacting vicinal fibrin is consistent with the experimental data. The model suggests that platelet contraction induces prestress in fibrin fibers and increases the effective stiffness in both cross-linked and noncross-linked clots. Our results provide evidence for fibrin compaction at discrete nodes as a major determinant of mechanical response to applied loads.
Collapse
Affiliation(s)
| | - Wilson Eng
- Department of Mechanical Engineering, San José State University, San José, California
| | - Sang-Joon J. Lee
- Department of Mechanical Engineering, San José State University, San José, California
| | | |
Collapse
|
13
|
Chérifi F, Laraba-Djebari F. Bioactive Molecules Derived from Snake Venoms with Therapeutic Potential for the Treatment of Thrombo-Cardiovascular Disorders Associated with COVID-19. Protein J 2021; 40:799-841. [PMID: 34499333 PMCID: PMC8427918 DOI: 10.1007/s10930-021-10019-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 01/08/2023]
Abstract
As expected, several new variants of Severe Acute Respiratory Syndrome-CoronaVirus-2 (SARS-CoV-2) emerged and have been detected around the world throughout this Coronavirus Disease of 2019 (COVID-19) pandemic. Currently, there is no specific developed drug against COVID-19 and the challenge of developing effective antiviral strategies based on natural agents with different mechanisms of action becomes an urgent need and requires identification of genetic differences among variants. Such data is used to improve therapeutics to combat SARS-CoV-2 variants. Nature is known to offer many biotherapeutics from animal venoms, algae and plant that have been historically used in traditional medicine. Among these bioresources, snake venom displays many bioactivities of interest such as antiviral, antiplatelet, antithrombotic, anti-inflammatory, antimicrobial and antitumoral. COVID-19 is a viral respiratory sickness due to SARS-CoV-2 which induces thrombotic disorders due to cytokine storm, platelet hyperactivation and endothelial dysfunction. This review aims to: (1) present an overview on the infection, the developed thrombo-inflammatory responses and mechanisms of induced thrombosis of COVID-19 compared to other similar pathogenesis; (2) underline the role of natural compounds such as anticoagulant, antiplatelet and thrombolytic agents; (3) investigate the management of coagulopathy related to COVID-19 and provide insight on therapeutic such as venom compounds. We also summarize the updated advances on antiviral proteins and peptides derived from snake venoms that could weaken coagulopathy characterizing COVID-19.
Collapse
Affiliation(s)
- Fatah Chérifi
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, USTHB, BP 32, El-Alia, Bab Ezzouar, Algiers, Algeria
| | - Fatima Laraba-Djebari
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, USTHB, BP 32, El-Alia, Bab Ezzouar, Algiers, Algeria.
| |
Collapse
|
14
|
Smallwood TB, Clark RJ. Advances in venom peptide drug discovery: where are we at and where are we heading? Expert Opin Drug Discov 2021; 16:1163-1173. [PMID: 33914674 DOI: 10.1080/17460441.2021.1922386] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: Animal venoms are a complex mixture of bioactive molecules that have evolved over millions of years for prey capture and defense from predators. Venom consists of many different types of molecules, with disulfide-rich peptides being a major component in most venoms. The study of these potent and highly selective molecules has led to the development of venom-derived drugs for diseases such as type 2 diabetes mellitus and chronic pain. As technologies have improved, more bioactive peptides have been discovered from venomous animals. Many of these molecules may have applications as tools for understanding normal and disease physiology, therapeutics, cosmetics or in agriculture.Areas covered: This article reviews venom-derived drugs approved by the FDA and venom-derived peptides currently in development. It discusses the challenges faced by venom-derived peptide drugs during drug development and the future for venom-derived peptides.Expert opinion: New techniques such as toxin driven discovery are expanding the pipeline of venom-derived peptides. There are many venom-derived peptides currently in preclinical and clinical trials that would have remained undiscovered using traditional approaches. A renewed focus on venoms, with advances in technology, will broaden the diversity of venom-derived peptide therapeutics and expand our knowledge of their molecular targets.
Collapse
Affiliation(s)
- Taylor B Smallwood
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Richard J Clark
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
15
|
Bordon KDCF, Cologna CT, Fornari-Baldo EC, Pinheiro-Júnior EL, Cerni FA, Amorim FG, Anjolette FAP, Cordeiro FA, Wiezel GA, Cardoso IA, Ferreira IG, de Oliveira IS, Boldrini-França J, Pucca MB, Baldo MA, Arantes EC. From Animal Poisons and Venoms to Medicines: Achievements, Challenges and Perspectives in Drug Discovery. Front Pharmacol 2020; 11:1132. [PMID: 32848750 PMCID: PMC7396678 DOI: 10.3389/fphar.2020.01132] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022] Open
Abstract
Animal poisons and venoms are comprised of different classes of molecules displaying wide-ranging pharmacological activities. This review aims to provide an in-depth view of toxin-based compounds from terrestrial and marine organisms used as diagnostic tools, experimental molecules to validate postulated therapeutic targets, drug libraries, prototypes for the design of drugs, cosmeceuticals, and therapeutic agents. However, making these molecules applicable requires extensive preclinical trials, with some applications also demanding clinical trials, in order to validate their molecular target, mechanism of action, effective dose, potential adverse effects, as well as other fundamental parameters. Here we go through the pitfalls for a toxin-based potential therapeutic drug to become eligible for clinical trials and marketing. The manuscript also presents an overview of the current picture for several molecules from different animal venoms and poisons (such as those from amphibians, cone snails, hymenopterans, scorpions, sea anemones, snakes, spiders, tetraodontiformes, bats, and shrews) that have been used in clinical trials. Advances and perspectives on the therapeutic potential of molecules from other underexploited animals, such as caterpillars and ticks, are also reported. The challenges faced during the lengthy and costly preclinical and clinical studies and how to overcome these hindrances are also discussed for that drug candidates going to the bedside. It covers most of the drugs developed using toxins, the molecules that have failed and those that are currently in clinical trials. The article presents a detailed overview of toxins that have been used as therapeutic agents, including their discovery, formulation, dosage, indications, main adverse effects, and pregnancy and breastfeeding prescription warnings. Toxins in diagnosis, as well as cosmeceuticals and atypical therapies (bee venom and leech therapies) are also reported. The level of cumulative and detailed information provided in this review may help pharmacists, physicians, biotechnologists, pharmacologists, and scientists interested in toxinology, drug discovery, and development of toxin-based products.
Collapse
Affiliation(s)
- Karla de Castro Figueiredo Bordon
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Takeno Cologna
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Ernesto Lopes Pinheiro-Júnior
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe Augusto Cerni
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernanda Gobbi Amorim
- Postgraduate Program in Pharmaceutical Sciences, Vila Velha University, Vila Velha, Brazil
| | | | - Francielle Almeida Cordeiro
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Gisele Adriano Wiezel
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Iara Aimê Cardoso
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isabela Gobbo Ferreira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isadora Sousa de Oliveira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | - Mateus Amaral Baldo
- Health and Science Institute, Paulista University, São José do Rio Pardo, Brazil
| | - Eliane Candiani Arantes
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
16
|
Brzoska T, Vats R, Bennewitz MF, Tutuncuoglu E, Watkins SC, Ragni MV, Neal MD, Gladwin MT, Sundd P. Intravascular hemolysis triggers ADP-mediated generation of platelet-rich thrombi in precapillary pulmonary arterioles. JCI Insight 2020; 5:139437. [PMID: 32544100 DOI: 10.1172/jci.insight.139437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/11/2020] [Indexed: 12/17/2022] Open
Abstract
Patients with hereditary or acquired hemolytic anemias have a high risk of developing in situ thrombosis of the pulmonary vasculature. While pulmonary thrombosis is a major morbidity associated with hemolytic disorders, the etiological mechanism underlying hemolysis-induced pulmonary thrombosis remains largely unknown. Here, we use intravital lung microscopy in mice to assess the pathogenesis of pulmonary thrombosis following deionized water-induced acute intravascular hemolysis. Acute hemolysis triggered the development of αIIbβ3-dependent platelet-rich thrombi in precapillary pulmonary arterioles, which led to the transient impairment of pulmonary blood flow. The hemolysis-induced pulmonary thrombosis was phenocopied with intravascular ADP- but not thrombin-triggered pulmonary thrombosis. Consistent with a mechanism involving ADP release from hemolyzing erythrocytes, the inhibition of platelet P2Y12 purinergic receptor signaling attenuated pulmonary thrombosis and rescued blood flow in the pulmonary arterioles of mice following intravascular hemolysis. These findings are the first in vivo studies to our knowledge to suggest that acute intravascular hemolysis promotes ADP-dependent platelet activation, leading to thrombosis in the precapillary pulmonary arterioles, and that thrombin generation most likely does not play a significant role in the pathogenesis of acute hemolysis-triggered pulmonary thrombosis.
Collapse
Affiliation(s)
- Tomasz Brzoska
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ravi Vats
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Margaret F Bennewitz
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, West Virginia, USA
| | - Egemen Tutuncuoglu
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Simon C Watkins
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Margaret V Ragni
- Department of Medicine, University of Pittsburgh, Hemophilia Center of Western Pennsylvania, Pittsburgh, Pennsylvania, USA
| | | | - Mark T Gladwin
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pulmonary Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Prithu Sundd
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Division of Pulmonary Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
17
|
Memariani H, Memariani M, Moravvej H, Shahidi-Dadras M. Melittin: a venom-derived peptide with promising anti-viral properties. Eur J Clin Microbiol Infect Dis 2020; 39:5-17. [PMID: 31422545 PMCID: PMC7224078 DOI: 10.1007/s10096-019-03674-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/08/2019] [Indexed: 12/23/2022]
Abstract
Despite tremendous advances in the development of anti-viral therapeutics, viral infections remain a chief culprit accounting for ongoing morbidity and mortality worldwide. Natural products, in particular animal venoms, embody a veritable cornucopia of exotic constituents, suggesting an immensurable source of anti-infective drugs. In this context, melittin, the principal constituent in the venom of the European honeybee Apis mellifera, has been demonstrated to exert anti-cancer, anti-inflammatory, anti-diabetic, anti-infective, and adjuvant properties. To our knowledge, there is no review appertaining to effects of melittin against viruses, prompting us to synopsize experimental investigations on its anti-viral activity throughout the past decades. Accumulating evidence indicates that melittin curbs infectivity of a diverse array of viruses including coxsackievirus, enterovirus, influenza A viruses, human immunodeficiency virus (HIV), herpes simplex virus (HSV), Junín virus (JV), respiratory syncytial virus (RSV), vesicular stomatitis virus (VSV), and tobacco mosaic virus (TMV). However, medication safety, different routes of administrations, and molecular mechanisms behind the anti-viral activity of melittin should be scrutinized in future studies.
Collapse
Affiliation(s)
- Hamed Memariani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Memariani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamideh Moravvej
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
18
|
Tran HNT, Tran P, Deuis JR, Agwa AJ, Zhang AH, Vetter I, Schroeder CI. Enzymatic Ligation of a Pore Blocker Toxin and a Gating Modifier Toxin: Creating Double-Knotted Peptides with Improved Sodium Channel NaV1.7 Inhibition. Bioconjug Chem 2019; 31:64-73. [DOI: 10.1021/acs.bioconjchem.9b00744] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hue N. T. Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Poanna Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer R. Deuis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Akello J. Agwa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alan H. Zhang
- Center for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Christina I. Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
19
|
Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins (Basel) 2019; 11:toxins11100564. [PMID: 31557973 PMCID: PMC6832721 DOI: 10.3390/toxins11100564] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/20/2019] [Accepted: 09/22/2019] [Indexed: 12/16/2022] Open
Abstract
Animal venoms are used as defense mechanisms or to immobilize and digest prey. In fact, venoms are complex mixtures of enzymatic and non-enzymatic components with specific pathophysiological functions. Peptide toxins isolated from animal venoms target mainly ion channels, membrane receptors and components of the hemostatic system with high selectivity and affinity. The present review shows an up-to-date survey on the pharmacology of snake-venom bioactive components and evaluates their therapeutic perspectives against a wide range of pathophysiological conditions. Snake venoms have also been used as medical tools for thousands of years especially in tradition Chinese medicine. Consequently, snake venoms can be considered as mini-drug libraries in which each drug is pharmacologically active. However, less than 0.01% of these toxins have been identified and characterized. For instance, Captopril® (Enalapril), Integrilin® (Eptifibatide) and Aggrastat® (Tirofiban) are drugs based on snake venoms, which have been approved by the FDA. In addition to these approved drugs, many other snake venom components are now involved in preclinical or clinical trials for a variety of therapeutic applications. These examples show that snake venoms can be a valuable source of new principle components in drug discovery.
Collapse
|
20
|
Machado ART, Aissa AF, Ribeiro DL, Ferreira RS, Sampaio SV, Antunes LMG. BjussuLAAO-II induces cytotoxicity and alters DNA methylation of cell-cycle genes in monocultured/co-cultured HepG2 cells. J Venom Anim Toxins Incl Trop Dis 2019; 25:e147618. [PMID: 31131003 PMCID: PMC6527400 DOI: 10.1590/1678-9199-jvatitd-1476-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/22/2018] [Indexed: 12/18/2022] Open
Abstract
Background: The use of animal venoms and their toxins as material sources for
biotechnological applications has received much attention from the
pharmaceutical industry. L-amino acid oxidases from snake venoms (SV-LAAOs)
have demonstrated innumerous biological effects and pharmacological
potential against different cancer types. Hepatocellular carcinoma has
increased worldwide, and the aberrant DNA methylation of liver cells is a
common mechanism to promote hepatic tumorigenesis. Moreover, tumor
microenvironment plays a major role in neoplastic transformation. To
elucidate the molecular mechanisms responsible for the cytotoxic effects of
SV-LAAO in human cancer cells, this study aimed to evaluate the cytotoxicity
and the alterations in DNA methylation profiler in the promoter regions of
cell-cycle genes induced by BjussuLAAO-II, an LAAO from Bothrops
jaracussu venom, in human hepatocellular carcinoma (HepG2)
cells in monoculture and co-culture with endothelial (HUVEC) cells. Methods: BjussuLAAO-II concentrations were 0.25, 0.50, 1.00 and 5.00 μg/mL. Cell
viability was assessed by MTT assay and DNA methylation of the promoter
regions of 22 cell-cycle genes by EpiTect Methyl II PCR array. Results: BjussuLAAO-II decreased the cell viability of HepG2 cells in monoculture at
all concentrations tested. In co-culture, 1.00 and 5.00 μg/mL induced
cytotoxicity (p < 0.05). BjussuLAAO-II increased the
methylation of CCND1 and decreased the methylation of
CDKN1A in monoculture and GADD45A in
both cell-culture models (p < 0.05). Conclusion: Data showed BjussuLAAO-II induced cytotoxicity and altered DNA methylation of
the promoter regions of cell-cycle genes in HepG2 cells in monoculture and
co-culture models. We suggested the analysis of DNA methylation profile of
GADD45A as a potential biomarker of the cell cycle
effects of BjussuLAAO-II in cancer cells. The tumor microenvironment should
be considered to comprise part of biotechnological strategies during the
development of snake-toxin-based novel drugs.
Collapse
Affiliation(s)
- Ana Rita Thomazela Machado
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Alexandre Ferro Aissa
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Diego Luis Ribeiro
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Rui Seabra Ferreira
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University - UNESP, Botucatu, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Lusânia Maria Greggi Antunes
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| |
Collapse
|
21
|
Mariottini GL, Grice ID. Natural Compounds and Drug Discovery: Can Cnidarian Venom Play a Role? Cent Nerv Syst Agents Med Chem 2019; 19:114-118. [PMID: 30827266 DOI: 10.2174/1871524919666190227234834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/22/2019] [Accepted: 01/26/2019] [Indexed: 11/22/2022]
Abstract
Natural compounds extracted from organisms and microorganisms are an important resource for the development of drugs and bioactive molecules. Many such compounds have made valuable contributions in diverse fields such as human health, pharmaceutics and industrial applications. Presently, however, research on investigating natural compounds from marine organisms is scarce. This is somewhat surprising considering that the marine environment makes a major contribution to Earth's ecosystems and consequently possesses a vast storehouse of diverse marine species. Interestingly, of the marine bioactive natural compounds identified to date, many are venoms, coming from Cnidarians (jellyfish, sea anemones, corals). Cnidarians are therefore particularly interesting marine species, producing important biological compounds that warrant further investigation for their development as possible therapeutic agents. From an experimental aspect, this review aims to emphasize and update the current scientific knowledge reported on selected biological activity (antiinflammatory, antimicrobial, antitumoral, anticoagulant, along with several less studied effects) of Cnidarian venoms/extracts, highlighting potential aspects for ongoing research towards their utilization in human therapeutic approaches.
Collapse
Affiliation(s)
- Gian Luigi Mariottini
- Department of Earth, Environment and Life Sciences, University of Genova, Genova, Italy
| | - Irwin Darren Grice
- Institute for Glycomics and School of Medical Science, Griffith University, Southport, Queensland, Australia
| |
Collapse
|
22
|
Otvos RA, Still KBM, Somsen GW, Smit AB, Kool J. Drug Discovery on Natural Products: From Ion Channels to nAChRs, from Nature to Libraries, from Analytics to Assays. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2019; 24:362-385. [PMID: 30682257 PMCID: PMC6484542 DOI: 10.1177/2472555218822098] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/16/2018] [Accepted: 12/07/2018] [Indexed: 12/21/2022]
Abstract
Natural extracts are complex mixtures that may be rich in useful bioactive compounds and therefore are attractive sources for new leads in drug discovery. This review describes drug discovery from natural products and in explaining this process puts the focus on ion-channel drug discovery. In particular, the identification of bioactives from natural products targeting nicotinic acetylcholine receptors (nAChRs) and serotonin type 3 receptors (5-HT3Rs) is discussed. The review is divided into three parts: "Targets," "Sources," and "Approaches." The "Targets" part will discuss the importance of ion-channel drug targets in general, and the α7-nAChR and 5-HT3Rs in particular. The "Sources" part will discuss the relevance for drug discovery of finding bioactive compounds from various natural sources such as venoms and plant extracts. The "Approaches" part will give an overview of classical and new analytical approaches that are used for the identification of new bioactive compounds with the focus on targeting ion channels. In addition, a selected overview is given of traditional venom-based drug discovery approaches and of diverse hyphenated analytical systems used for screening complex bioactive mixtures including venoms.
Collapse
Affiliation(s)
- Reka A. Otvos
- The Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Kristina B. M. Still
- The Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Govert W. Somsen
- The Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jeroen Kool
- The Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Oliveira ISD, Manzini RV, Ferreira IG, Cardoso IA, Bordon KDCF, Machado ART, Antunes LMG, Rosa JC, Arantes EC. Cell migration inhibition activity of a non-RGD disintegrin from Crotalus durissus collilineatus venom. J Venom Anim Toxins Incl Trop Dis 2018; 24:28. [PMID: 30377432 PMCID: PMC6195974 DOI: 10.1186/s40409-018-0167-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/05/2018] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND In recent decades, snake venom disintegrins have received special attention due to their potential use in anticancer therapy. Disintegrins are small and cysteine-rich proteins present in snake venoms and can interact with specific integrins to inhibit their activities in cell-cell and cell-ECM interactions. These molecules, known to inhibit platelet aggregation, are also capable of interacting with certain cancer-related integrins, and may interfere in important processes involved in carcinogenesis. Therefore, disintegrin from Crotalus durissus collilineatus venom was isolated, structurally characterized and evaluated for its toxicity and ability to interfere with cell proliferation and migration in MDA-MB-231, a human breast cancer cell line. METHODS Based on previous studies, disintegrin was isolated by FPLC, through two chromatographic steps, both on reversed phase C-18 columns. The isolated disintegrin was structurally characterized by Tris-Tricine-SDS-PAGE, mass spectrometry and N-terminal sequencing. For the functional assays, MTT and wound-healing assays were performed in order to investigate cytotoxicity and effect on cell migration in vitro, respectively. RESULTS Disintegrin presented a molecular mass of 7287.4 Da and its amino acid sequence shared similarity with the disintegrin domain of P-II metalloproteases. Using functional assays, the disintegrin showed low cytotoxicity (15% and 17%, at 3 and 6 μg/mL, respectively) after 24 h of incubation and in the wound-healing assay, the disintegrin (3 μg/mL) was able to significantly inhibit cell migration (24%, p < 0.05), compared to negative control. CONCLUSION Thus, our results demonstrate that non-RGD disintegrin from C. d. collilineatus induces low cytotoxicity and inhibits migration of human breast cancer cells. Therefore, it may be a very useful molecular tool for understanding ECM-cell interaction cancer-related mechanisms involved in an important integrin family that highlights molecular aspects of tumorigenesis. Also, non-RGD disintegrin has potential to serve as an agent in anticancer therapy or adjuvant component combined with other anticancer drugs.
Collapse
Affiliation(s)
- Isadora Sousa de Oliveira
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Av. do Café s/n°, Monte Alegre, Ribeirão Preto, SP 14040-903 Brazil
| | - Rafaella Varzoni Manzini
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Av. do Café s/n°, Monte Alegre, Ribeirão Preto, SP 14040-903 Brazil
| | - Isabela Gobbo Ferreira
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Av. do Café s/n°, Monte Alegre, Ribeirão Preto, SP 14040-903 Brazil
| | - Iara Aimê Cardoso
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Av. do Café s/n°, Monte Alegre, Ribeirão Preto, SP 14040-903 Brazil
| | - Karla de Castro Figueiredo Bordon
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Av. do Café s/n°, Monte Alegre, Ribeirão Preto, SP 14040-903 Brazil
| | - Ana Rita Thomazela Machado
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP Brazil
| | - Lusânia Maria Greggi Antunes
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP Brazil
| | - José Cesar Rosa
- Protein Chemistry Center and Department of Molecular and Cell Biology and Pathogenic Bioagents, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP Brazil
| | - Eliane Candiani Arantes
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Av. do Café s/n°, Monte Alegre, Ribeirão Preto, SP 14040-903 Brazil
| |
Collapse
|
24
|
A microengineered vascularized bleeding model that integrates the principal components of hemostasis. Nat Commun 2018; 9:509. [PMID: 29410404 PMCID: PMC5802762 DOI: 10.1038/s41467-018-02990-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/11/2018] [Indexed: 01/12/2023] Open
Abstract
Hemostasis encompasses an ensemble of interactions among platelets, coagulation factors, blood cells, endothelium, and hemodynamic forces, but current assays assess only isolated aspects of this complex process. Accordingly, here we develop a comprehensive in vitro mechanical injury bleeding model comprising an "endothelialized" microfluidic system coupled with a microengineered pneumatic valve that induces a vascular "injury". With perfusion of whole blood, hemostatic plug formation is visualized and "in vitro bleeding time" is measured. We investigate the interaction of different components of hemostasis, gaining insight into several unresolved hematologic issues. Specifically, we visualize and quantitatively demonstrate: the effect of anti-platelet agent on clot contraction and hemostatic plug formation, that von Willebrand factor is essential for hemostasis at high shear, that hemophilia A blood confers unstable hemostatic plug formation and altered fibrin architecture, and the importance of endothelial phosphatidylserine in hemostasis. These results establish the versatility and clinical utility of our microfluidic bleeding model.
Collapse
|
25
|
Chopra G, Samudrala R. Exploring Polypharmacology in Drug Discovery and Repurposing Using the CANDO Platform. Curr Pharm Des 2017; 22:3109-23. [PMID: 27013226 DOI: 10.2174/1381612822666160325121943] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/01/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND Traditional drug discovery approaches focus on a limited set of target molecules for treatment against specific indications/diseases. However, drug absorption, dispersion, metabolism, and excretion (ADME) involve interactions with multiple protein systems. Drugs approved for particular indication(s) may be repurposed as novel therapeutics for others. The severely declining rate of discovery and increasing costs of new drugs illustrate the limitations of the traditional reductionist paradigm in drug discovery. METHODS We developed the Computational Analysis of Novel Drug Opportunities (CANDO) platform based on a hypothesis that drugs function by interacting with multiple protein targets to create a molecular interaction signature that can be exploited for therapeutic repurposing and discovery. We compiled a library of compounds that are human ingestible with minimal side effects, followed by an 'all-compounds' vs 'all-proteins' fragment-based multitarget docking with dynamics screen to construct compound-proteome interaction matrices that were then analyzed to determine similarity of drug behavior. The proteomic signature similarity of drugs is then ranked to make putative drug predictions for all indications in a shotgun manner. RESULTS We have previously applied this platform with success in both retrospective benchmarking and prospective validation, and to understand the effect of druggable protein classes on repurposing accuracy. Here we use the CANDO platform to analyze and determine the contribution of multitargeting (polypharmacology) to drug repurposing benchmarking accuracy. Taken together with the previous work, our results indicate that a large number of protein structures with diverse fold space and a specific polypharmacological interactome is necessary for accurate drug predictions using our proteomic and evolutionary drug discovery and repurposing platform. CONCLUSION These results have implications for future drug development and repurposing in the context of polypharmacology.
Collapse
Affiliation(s)
- Gaurav Chopra
- Department of Chemistry, Purdue University, West Lafayette, IN, USA.
| | - Ram Samudrala
- Department of Biomedical Informatics, SUNY, Buffalo, NY, USA.
| |
Collapse
|
26
|
Gorai B, Sivaraman T. Delineating residues for haemolytic activities of snake venom cardiotoxin 1 from Naja naja as probed by molecular dynamics simulations and in vitro validations. Int J Biol Macromol 2017; 95:1022-1036. [DOI: 10.1016/j.ijbiomac.2016.10.091] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/26/2016] [Indexed: 02/05/2023]
|
27
|
Shariati H, Sanei H, Pourmoghadas A, Salehizadeh L, Amirpour A. Immediate outcomes of eptifibatide therapy during intracoronary stent implantation. Adv Biomed Res 2016; 5:204. [PMID: 28217642 PMCID: PMC5220753 DOI: 10.4103/2277-9175.196831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 10/15/2014] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The objective of the present study was to assess the major immediate outcomes of eptifibatide therapy during intracoronary stent implantation. MATERIALS AND METHODS In an interventional study, patients undergoing percutaneous coronary intervention (PCI) were randomized into either the eptifibatide (n = 100) or the control (n = 107) group. In each group, demographic and clinical characteristics such as cardiac death, stent thrombosis (ST), myocardial infarction (MI), rates of target lesion and vessel revascularization, cerebral vascular accident (CVA), and emergency coronary artery bypass grafting (CABG) were recorded. RESULTS The overall rates of major adverse events such as mortality, Stent thrombosis (ST), Myocardial Infarction (MI), target lesion revascularization (TLR), target vessel revascularization (TVR), CVA, and emergency CABG within 24 h after stent implantation were low and comparable between the two groups; P > 0.05 considered significant for all comparisons. CONCLUSION There were no statistical differences between the clinical outcomes of groups administered with single-dose intracoronary eptifibatide and control groups among patients undergoing PCI during stent implantation.
Collapse
Affiliation(s)
- Hooman Shariati
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Sanei
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Pourmoghadas
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Salehizadeh
- Interventional Cardiologist, Chamran Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Afshin Amirpour
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
28
|
Harnessing the knowledge of animal toxins to generate drugs. Pharmacol Res 2016; 112:30-36. [PMID: 26826284 DOI: 10.1016/j.phrs.2016.01.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/07/2016] [Accepted: 01/12/2016] [Indexed: 11/20/2022]
Abstract
Animal toxins present high selectivity and specificity for their molecular targets, and have long been considered as prototypes for developing novel drugs, with some successful cases. In this regard, the variety of molecules found in animal venoms, which can be capable of affecting vital physiological systems, have providing the development of studies focusing on turning those molecules (toxins) into therapeutics to treat several diseases, such as chronic pain, hypertension, thrombosis, cancer, and so on. However, some important issues have been responsible for disrupting the toxin-based drug discovery projects. In this review, we have briefly highlighted the development of drugs based on animal toxins, discussing some successful cases as well as the main causes of failure, pointing out the recent strategies applied to overcome the difficulties related to the translational process in this kind of development scenario.
Collapse
|
29
|
Caporali A, Meloni M, Nailor A, Mitić T, Shantikumar S, Riu F, Sala-Newby GB, Rose L, Besnier M, Katare R, Voellenkle C, Verkade P, Martelli F, Madeddu P, Emanueli C. p75(NTR)-dependent activation of NF-κB regulates microRNA-503 transcription and pericyte-endothelial crosstalk in diabetes after limb ischaemia. Nat Commun 2015; 6:8024. [PMID: 26268439 PMCID: PMC4538859 DOI: 10.1038/ncomms9024] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 07/09/2015] [Indexed: 12/28/2022] Open
Abstract
The communication between vascular endothelial cells (ECs) and pericytes in the microvasculature is fundamental for vascular growth and homeostasis; however, these processes are disrupted by diabetes. Here we show that modulation of p75(NTR) expression in ECs exposed to high glucose activates transcription of miR-503, which negatively affects pericyte function. p75(NTR) activates NF-κB to bind the miR-503 promoter and upregulate miR-503 expression in ECs. NF-κB further induces activation of Rho kinase and shedding of endothelial microparticles carrying miR-503, which transfer miR-503 from ECs to vascular pericytes. The integrin-mediated uptake of miR-503 in the recipient pericytes reduces expression of EFNB2 and VEGFA, resulting in impaired migration and proliferation. We confirm operation of the above mechanisms in mouse models of diabetes, in which EC-derived miR-503 reduces pericyte coverage of capillaries, increased permeability and impaired post-ischaemic angiogenesis in limb muscles. Collectively, our data demonstrate that miR-503 regulates pericyte-endothelial crosstalk in microvascular diabetic complications.
Collapse
Affiliation(s)
- Andrea Caporali
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Marco Meloni
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
| | - Audrey Nailor
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
| | - Tijana Mitić
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
| | - Saran Shantikumar
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
| | - Federica Riu
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
| | | | - Lorraine Rose
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Marie Besnier
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
| | - Rajesh Katare
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, Milan 20097, Italy
| | - Paul Verkade
- Wolfson Bioimaging Facility, University of Bristol, Bristol BS2 8HW, UK
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, Milan 20097, Italy
| | - Paolo Madeddu
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
| | - Costanza Emanueli
- School of Clinical Sciences, Bristol Heart Institute, Bristol BS2 8HW, UK
- National Institute of Heart and Lung, Imperial College of London, London SW7 2AZ, UK
| |
Collapse
|
30
|
Secondary structure, a missing component of sequence-based minimotif definitions. PLoS One 2012; 7:e49957. [PMID: 23236358 PMCID: PMC3517595 DOI: 10.1371/journal.pone.0049957] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 10/15/2012] [Indexed: 12/27/2022] Open
Abstract
Minimotifs are short contiguous segments of proteins that have a known biological function. The hundreds of thousands of minimotifs discovered thus far are an important part of the theoretical understanding of the specificity of protein-protein interactions, posttranslational modifications, and signal transduction that occur in cells. However, a longstanding problem is that the different abstractions of the sequence definitions do not accurately capture the specificity, despite decades of effort by many labs. We present evidence that structure is an essential component of minimotif specificity, yet is not used in minimotif definitions. Our analysis of several known minimotifs as case studies, analysis of occurrences of minimotifs in structured and disordered regions of proteins, and review of the literature support a new model for minimotif definitions that includes sequence, structure, and function.
Collapse
|
31
|
Anticoagulant activity of Moon jellyfish (Aurelia aurita) tentacle extract. Toxicon 2012; 60:719-23. [DOI: 10.1016/j.toxicon.2012.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 05/09/2012] [Accepted: 05/17/2012] [Indexed: 11/18/2022]
|
32
|
King GF. Venoms as a platform for human drugs: translating toxins into therapeutics. Expert Opin Biol Ther 2011; 11:1469-84. [PMID: 21939428 DOI: 10.1517/14712598.2011.621940] [Citation(s) in RCA: 392] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION An extraordinarily diverse range of animals have evolved venoms for predation, defence, or competitor deterrence. The major components of most venoms are peptides and proteins that are often protease-resistant due to their disulfide-rich architectures. Some of these toxins have become valuable as pharmacological tools and/or therapeutics due to their extremely high specificity and potency for particular molecular targets. There are currently six FDA-approved drugs derived from venom peptides or proteins. AREAS COVERED This article surveys the current pipeline of venom-derived therapeutics and critically examines the potential of peptide and protein drugs derived from venoms. Emerging trends are identified, including an increasing industry focus on disulfide-rich venom peptides and the use of a broader array of molecular targets in order to develop venom-based therapeutics for treating a wider range of clinical conditions. EXPERT OPINION Key technical advances in combination with a renewed industry-wide focus on biologics have converged to provide a larger than ever pipeline of venom-derived therapeutics. Disulfide-rich venom peptides obviate some of the traditional disadvantages of therapeutic peptides and some may be suitable for oral administration. Moreover, some venom peptides can breach the blood brain barrier and translocate across cell membranes, which opens up the possibility of exploiting molecular targets not previously accessible to peptide drugs.
Collapse
Affiliation(s)
- Glenn F King
- The University of Queensland, Institute for Molecular Bioscience, 306 Carmody Road, St Lucia, Queensland 4072, Australia.
| |
Collapse
|
33
|
Kaluski E. The Role of Glycoprotein IIb/IIIa Inhibitors- A Promise Not Kept? Curr Cardiol Rev 2011; 4:84-91. [PMID: 19936282 PMCID: PMC2779356 DOI: 10.2174/157340308784245793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Revised: 05/17/2007] [Accepted: 05/30/2007] [Indexed: 11/22/2022] Open
Abstract
For over one decade Glycoproteins IIb/IIIa inhibitors (GPI) have been administered to prevent coronary artery thrombosis. Initially these agents were used for acute coronary syndromes and subsequently as adjunctive pharmacotherapy for percutaneous coronary interventions (PCIs). Most benefit of GPI emerged from reduction of ischemic events: mostly non-q-wave myocardial infarctions (NQWMIs) during PCI. However, individual randomized clinical trials could not demonstrate that any of these agents could significantly reduce mortality in any clinical subset of patients. Studies of employing prolonged oral GPI administration resulted in excessive death. The non-homogenous statistically-significant reduction of ischemic endpoints was accompanied by an excess of bleeding, vascular complications, and thrombocytopenia. The clinical and ecomomic burden of major bleeding and thrombocytopenia is substantial. The ACUITY trial has initiate a new debate regarding the efficacy and safety of GPI. Selective “patient-tailored” use of GPI limited to moderate-high risk PCI patients with low bleeding propensity is suggested. Research of new algorithms emphasizing abbreviated GPI administration, careful access site and bleeding surveillance, in conjunction with lower doses of unfractionated heparin or new and safer anti-thrombins may further enhance patient safety.
Collapse
Affiliation(s)
- Edo Kaluski
- Department of Cardiology, University Medical Center, University of Medicine and Dentistry, Newark, NJ, USA
| |
Collapse
|
34
|
Kaluski E, Haider B, Milo-Cotter O, Klapholz M. Glycoprotein IIb/IIIa inhibitors: questioning indications and treatment algorithms. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2007; 8:281-8. [PMID: 18053951 DOI: 10.1016/j.carrev.2007.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Accepted: 03/30/2007] [Indexed: 11/27/2022]
Abstract
Glycoprotein inhibitors (GPI) are viewed as beneficial adjunctive pharmacotherapy agents for percutaneous coronary interventions (PCIs). The major benefit of GPI is derived from the reduction of ischemic events (mostly non-Q-wave myocardial infarctions) during PCI. There is no single randomized clinical trial demonstrating that any of these agents significantly reduces mortality in any clinical subset of patients. Studies of sustained oral GPI resulted in excessive death and myocardial infarctions. Reduction of ischemic end points was counteracted by excessive bleeding, vascular complications, and thrombocytopenia. These complications bear considerable medical and economic impact. The Acute Catheterization and Early Intervention Triage Strategy trial demonstrated that GPI, when added to heparin, enoxaparine, or bivalirudin, do not reduce mortality or ischemic events but significantly increase bleeding complications. Major bleeding resulted in threefold mortality at 1 year. In view of available data, the use of GPI should be limited to moderate-risk to high-risk PCI patients with low bleeding propensity. Protocols of abbreviated GPI administration and careful bleeding surveillance, in conjunction with lower doses of unfractionated heparin or new and possibly safer antithrombins, can potentially improve patient safety.
Collapse
Affiliation(s)
- Edo Kaluski
- Department of Cardiology, University Medical Center, University of Medicine and Dentistry, Newark, NJ 07101, USA.
| | | | | | | |
Collapse
|
35
|
Rajagopalan N, Pung YF, Zhu YZ, Wong PTH, Kumar PP, Kini RM. Beta-cardiotoxin: a new three-finger toxin from Ophiophagus hannah (king cobra) venom with beta-blocker activity. FASEB J 2007; 21:3685-95. [PMID: 17616557 DOI: 10.1096/fj.07-8658com] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Snake venoms have provided a number of novel ligands with therapeutic potential. We have constructed a partial cDNA library from the mRNA of Ophiophagus hannah (king cobra) venom gland tissue and identified five new genes encoding proteins belonging to the three-finger toxin family of snake venom proteins. We have isolated and characterized one of these beta-sheet containing proteins with a mass of 7012.43 +/- 0.91 Da from the venom. The protein was nonlethal up to a dose of 10 mg/kg when injected intraperitoneally into Swiss albino mice. However, it induces labored breathing and death at a dose of 100 mg/kg. It does not show any hemolytic or anticoagulant activity. It caused a dose-dependent decrease of heart rate in vivo (anesthetized Sprague-Dawley rats) and also ex vivo (Langendorff isolated rat heart). This is in contrast to classical cardiotoxins from snake venom that increase the heart rate in animals. Radioligand displacement studies showed that this protein targets beta-adrenergic receptors with a binding affinity (Ki) of 5.3 and 2.3 microM toward beta1 and beta2 subtypes, respectively, to bring about its effect, and hence, it was named as beta-cardiotoxin. This is the first report of a natural exogenous beta-blocker.
Collapse
Affiliation(s)
- Nandhakishore Rajagopalan
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Science Dr. 4, Singapore 117543
| | | | | | | | | | | |
Collapse
|
36
|
Birrell GW, Earl STH, Wallis TP, Masci PP, de Jersey J, Gorman JJ, Lavin MF. The Diversity of Bioactive Proteins in Australian Snake Venoms. Mol Cell Proteomics 2007; 6:973-86. [PMID: 17317661 DOI: 10.1074/mcp.m600419-mcp200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Australian elapid snakes are among the most venomous in the world. Their venoms contain multiple components that target blood hemostasis, neuromuscular signaling, and the cardiovascular system. We describe here a comprehensive approach to separation and identification of the venom proteins from 18 of these snake species, representing nine genera. The venom protein components were separated by two-dimensional PAGE and identified using mass spectrometry and de novo peptide sequencing. The venoms are complex mixtures showing up to 200 protein spots varying in size from <7 to over 150 kDa and in pI from 3 to >10. These include many proteins identified previously in Australian snake venoms, homologs identified in other snake species, and some novel proteins. In many cases multiple trains of spots were typically observed in the higher molecular mass range (>20 kDa) (indicative of post-translational modification). Venom proteins and their post-translational modifications were characterized using specific antibodies, phosphoprotein- and glycoprotein-specific stains, enzymatic digestion, lectin binding, and antivenom reactivity. In the lower molecular weight range, several proteins were identified, but the predominant species were phospholipase A2 and alpha-neurotoxins, both represented by different sequence variants. The higher molecular weight range contained proteases, nucleotidases, oxidases, and homologs of mammalian coagulation factors. This information together with the identification of several novel proteins (metalloproteinases, vespryns, phospholipase A2 inhibitors, protein-disulfide isomerase, 5'-nucleotidases, cysteine-rich secreted proteins, C-type lectins, and acetylcholinesterases) aids in understanding the lethal mechanisms of elapid snake venoms and represents a valuable resource for future development of novel human therapeutics.
Collapse
Affiliation(s)
- Geoff W Birrell
- The Queensland Institute of Medical Research, P. O. Royal Brisbane Hospital, Brisbane 4029, Australia
| | | | | | | | | | | | | |
Collapse
|
37
|
Banerjee Y, Mizuguchi J, Iwanaga S, Kini RM. Hemextin AB Complex, a Unique Anticoagulant Protein Complex from Hemachatus haemachatus (African Ringhals Cobra) Venom That Inhibits Clot Initiation and Factor VIIa Activity. J Biol Chem 2005; 280:42601-11. [PMID: 16204244 DOI: 10.1074/jbc.m508987200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
During injury or trauma, blood coagulation is initiated by the interaction of factor VIIa (FVIIa) in the blood with freshly exposed tissue factor (TF) to form the TF.FVIIa complex. However, unwanted clot formation can lead to death and debilitation due to vascular occlusion, and hence, anticoagulants are important for the treatment of thromboembolic disorders. Here, we report the isolation and characterization of two synergistically acting anticoagulant proteins, hemextins A and B, from the venom of Hemachatus haemachatus (African Ringhals cobra). N-terminal sequences and CD spectra of the native proteins indicate that these proteins belong to the three-finger toxin family. Hemextin A (but not hemextin B) exhibits mild anticoagulant activity. However, hemextin B forms a complex (hemextin AB complex) with hemextin A and synergistically enhances its anticoagulant potency. Prothrombin time assay showed that these two proteins form a 1:1 complex. Complex formation was supported by size-exclusion chromatography. Using a "dissection approach," we determined that hemextin A and the hemextin AB complex prolong clotting by inhibiting TF.FVIIa activity. The site of anticoagulant effects was supported by their inhibitory effect on the reconstituted TF.FVIIa complex. Furthermore, we demonstrated their specificity of inhibition by studying their effects on 12 serine proteases; the hemextin AB complex potently inhibited the amidolytic activity of FVIIa in the presence and absence of soluble TF. Kinetic studies showed that the hemextin AB complex is a noncompetitive inhibitor of soluble TF.FVIIa amidolytic activity, with a Ki of 50 nm. Isothermal titration calorimetric studies showed that the hemextin AB complex binds directly to FVIIa with a binding constant of 1.62 x 10(5) m(-1). The hemextin AB complex is the first reported natural inhibitor of FVIIa that does not require a scaffold to mediate its inhibitory activity. Molecular interactions of the hemextin AB complex with FVIIa/TF.FVIIa will provide a new paradigm in the search for anticoagulants that inhibit the initiation of blood coagulation.
Collapse
Affiliation(s)
- Yajnavalka Banerjee
- Protein Science Laboratory, Department of Biological Sciences, Faculty of Science, National University of Singapore Singapore 117543
| | | | | | | |
Collapse
|
38
|
Brouwers FM, Oyen WJG, Boerman OC, Barrett JA, Verheugt FWA, Corstens FHM, Van der Meer JWM. Evaluation of Tc-99m-labeled glycoprotein IIb/IIIa receptor antagonist DMP444 SPECT in patients with infective endocarditis. Clin Nucl Med 2003; 28:480-4. [PMID: 12911097 DOI: 10.1097/01.rlu.0000067508.82824.75] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE Infective endocarditis (IE) is characterized by aggregation of activated platelets, fibrin, and bacteria. DMP444, a high-affinity glycoprotein IIb/IIIa receptor antagonist, binds to the fibrinogen-binding domain of activated platelets, depicting a key feature of IE. Tc-99m DMP444 scintigraphy was studied in a group of patients with possible IE. METHODS Tc-99m DMP444 (600 MBq; 16 mCi) planar and SPECT images of the heart were recorded in patients with possible IE for as long as 6 hours after injection. Results were compared to echocardiography and the Duke classification. RESULTS Sixteen patients (age range, 37 to 78 years) participated. DMP444 imaging was positive on SPECT in five patients, and all had definite endocarditis (affecting both prosthetic and native valves). Eleven patients were DMP444 negative, seven with no proof of IE. The remaining four patients were classified as having IE, but three had been receiving adequate intravenous antibiotic regimens for > or = 2 weeks at the time of scintigraphy and one had Q-fever endocarditis. CONCLUSIONS DMP444 SPECT allows in vivo visualization of IE if it is performed within 1 to 2 weeks after the start of antibiotic treatment. Given the high affinity of DMP444 for activated platelets, the results indicate the involvement of activated platelets in early IE.
Collapse
|