1
|
Memariani H, Memariani M. New Frontiers in Fighting Mycobacterial Infections: Venom-Derived Peptides. Probiotics Antimicrob Proteins 2025; 17:1217-1235. [PMID: 39828882 DOI: 10.1007/s12602-025-10455-z] [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] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Notwithstanding the indefatigable endeavors to develop effective anti-mycobacterial therapies, mycobacterial infections still present a tough problem for medicine today. The problem is further complicated by the disquieting surge of drug-resistant mycobacterial pathogens, which considerably narrows the existing therapeutic options. Thus, there is a genuine need to discover novel anti-mycobacterial drugs. Animal venoms are considered a treasure trove of structurally variable and biologically active peptides, which may hold promise for therapeutic applications. Over the past two decades, abundant evidence has been amassed regarding anti-mycobacterial effects of various peptides derived from the venoms of honeybees, wasps, scorpions, pseudoscorpions, cone snails, and snakes. This review intends to consolidate the state-of-the-art knowledge on the anti-mycobacterial peptides of animal venoms and to sketch potentially fruitful directions for future investigations. The available data indicate that micromolar concentrations of particular venom-derived peptides can effectively inhibit the in vitro growth of Mycobacterium tuberculosis and non-tuberculous mycobacteria. The proposed mechanisms of action of venom-derived peptides include reduced activity of plasma membrane ATPase, depolarization of the cell membrane, disruption of the cell wall, and increased generation of reactive oxygen species. Interestingly, administering certain peptides (≤ 2 mg/kg body weight) through daily intraperitoneal injections to mice for 8 consecutive days resulted in lower levels of mycobacterial infections and inflammation, hitting two targets with one arrow. Indubitably, such peptides can usher in new possibilities for the prevention and treatment of recalcitrant mycobacterial infections.
Collapse
Affiliation(s)
- Hamed Memariani
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mojtaba Memariani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
Germoush MO, Fouda M, Mantargi MJS, Sarhan M, AlRashdi BM, Massoud D, Altyar AE, Abdel-Daim MM. Exploring the SARS-CoV-2 spike protein destabilizer toxin from the scorpion, spider, and wasp group of toxins as a promising candidate for the identification of pharmacophores against viral infections. Open Vet J 2025; 15:69-84. [PMID: 40092205 PMCID: PMC11910296 DOI: 10.5455/ovj.2024.v15.i1.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/27/2024] [Indexed: 03/19/2025] Open
Abstract
Background The SARS-CoV-2 virus is the infectious agent that causes coronavirus illness (COVID-19). The majority of virus-infected individuals will recover without the need for special care after experiencing mild-to-moderate respiratory symptoms. Some people, nevertheless, will get quite sick and need medical help. The choice of COVID-19 treatment should be made individually. The severity of the illness and the chance that it will worsen will determine the decision. Therefore, developing more potent medications is always a primary goal. Finding more effective drugs is a top priority. In this regard, natural animal toxins, such as toxin derived from scorpions, spiders, and wasps, have been found to include compounds that have significant therapeutic properties. Specifically, targeting the spike protein which acts as a gateway for the vires to enter the human or animal cells. Aim This study focuses on the ability of toxins to destabilize the spike protein of the SARS-CoV-2 virus, which is responsible for the SARS-CoV-2 pandemic. Methods The active protein structure of the SARS-CoV-2, the toxins chosen obtained from the RCSB-protein data bank (PDB), and the molecular structures of toxins that were not proteins were either obtained from PubChem or downloaded as computer structure models from RCSB-PDB. Using molecular docking software such as "PyRx," analyzers such as "BIOVIA-Discovery studios" and "Pymol," and various techniques, the evaluation of the interactions between the spike protein and toxin was performed, to find possible pharmacophores that might serve as a foundation for upcoming medication development. The protein-ligand complex was put to test through the molecular dynamic (MD) simulation via visual molecular dynamics /nanoscale molecular dynamics application to determine the complex stability. Results The current research findings reveal intriguing binding affinities and interaction patterns between the toxin and the SARS-CoV-2 spike protein, where the complex was identified to be stable throughout the study resembling the cellular conditions via MD simulations. We discuss the implications of these interactions and how they might interfere viral infection and entry. Conclusion The current study sheds light on a promising avenue for the development of antiviral therapies, leveraging natural venoms as a source of inspiration for pharmacophore-based drug discovery opposing viral infections.
Collapse
Affiliation(s)
- Mousa O. Germoush
- Biology Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Maged Fouda
- Biology Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Mohammad J. S. Mantargi
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Moustafa Sarhan
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al Hofuf, Saudi Arabia
- Department of Zoology, Faculty of Science, Al-Azhar University, Assuit, Egypt
| | - Barakat M. AlRashdi
- Biology Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Diaa Massoud
- Biology Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Ahmed E. Altyar
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
3
|
Germoush MO, Fouda M, Mantargi MJS, Sarhan M, AlRashdi BM, Massoud D, Altyar AE, Abdel-Daim MM. Exploring the SARS-CoV-2 spike protein destabilizer toxin from the scorpion, spider, and wasp group of toxins as a promising candidate for the identification of pharmacophores against viral infections. Open Vet J 2025; 15:69-84. [PMID: 40092205 PMCID: PMC11910296 DOI: 10.5455/ovj.2025.v15.i1.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/27/2024] [Indexed: 04/11/2025] Open
Abstract
Background The SARS-CoV-2 virus is the infectious agent that causes coronavirus illness (COVID-19). The majority of virus-infected individuals will recover without the need for special care after experiencing mild-to-moderate respiratory symptoms. Some people, nevertheless, will get quite sick and need medical help. The choice of COVID-19 treatment should be made individually. The severity of the illness and the chance that it will worsen will determine the decision. Therefore, developing more potent medications is always a primary goal. Finding more effective drugs is a top priority. In this regard, natural animal toxins, such as toxin derived from scorpions, spiders, and wasps, have been found to include compounds that have significant therapeutic properties. Specifically, targeting the spike protein which acts as a gateway for the vires to enter the human or animal cells. Aim This study focuses on the ability of toxins to destabilize the spike protein of the SARS-CoV-2 virus, which is responsible for the SARS-CoV-2 pandemic. Methods The active protein structure of the SARS-CoV-2, the toxins chosen obtained from the RCSB-protein data bank (PDB), and the molecular structures of toxins that were not proteins were either obtained from PubChem or downloaded as computer structure models from RCSB-PDB. Using molecular docking software such as "PyRx," analyzers such as "BIOVIA-Discovery studios" and "Pymol," and various techniques, the evaluation of the interactions between the spike protein and toxin was performed, to find possible pharmacophores that might serve as a foundation for upcoming medication development. The protein-ligand complex was put to test through the molecular dynamic (MD) simulation via visual molecular dynamics /nanoscale molecular dynamics application to determine the complex stability. Results The current research findings reveal intriguing binding affinities and interaction patterns between the toxin and the SARS-CoV-2 spike protein, where the complex was identified to be stable throughout the study resembling the cellular conditions via MD simulations. We discuss the implications of these interactions and how they might interfere viral infection and entry. Conclusion The current study sheds light on a promising avenue for the development of antiviral therapies, leveraging natural venoms as a source of inspiration for pharmacophore-based drug discovery opposing viral infections.
Collapse
Affiliation(s)
- Mousa O. Germoush
- Biology Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Maged Fouda
- Biology Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Mohammad J. S. Mantargi
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Moustafa Sarhan
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al Hofuf, Saudi Arabia
- Department of Zoology, Faculty of Science, Al-Azhar University, Assuit, Egypt
| | - Barakat M. AlRashdi
- Biology Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Diaa Massoud
- Biology Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Ahmed E. Altyar
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
4
|
Abd El Maksoud EA, Rady MH, Mahmoud AGT, Hamza D, Seadawy MG, Essa EE. Potential therapeutic biomolecules of hymenopteran venom against SARS-CoV-2 from Egyptian patients. Sci Rep 2024; 14:15363. [PMID: 38965389 PMCID: PMC11224265 DOI: 10.1038/s41598-024-65038-9] [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: 03/09/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024] Open
Abstract
The therapeutic potential of insect-derived bioactive molecules as anti-SARS-CoV-2 agents has shown promising results. Hymenopteran venoms, notably from Apis mellifera (honeybee) and Vespa orientalis (oriental wasp), were examined for the first time in an in vitro setting for their potential anti-COVID-19 activity. This assessment utilized an immunodiagnostic system to detect the SARS-CoV-2 nucleocapsid antigen titer reduction. Further analyses, including cytotoxicity assays, plaque reduction assays, and in silico docking-based screening, were performed to evaluate the efficacy of the most potent venom. Results indicated that bee and wasp venoms contain bioactive molecules with potential therapeutic effects against SARS-CoV-2.Nevertheless, the wasp venom exhibited superior efficacy compared to bee venom, achieving a 90% maximal (EC90) concentration effect of antigen depletion at 0.184 mg/mL, in contrast to 2.23 mg/mL for bee venom. The cytotoxicity of the wasp venom was assessed on Vero E6 cells 48 h post-treatment using the MTT assay. The CC 50 of the cell growth was 0.16617 mg/mL for Vero E6 cells. The plaque reduction assay of wasp venom revealed 50% inhibition (IC50) at a 0.208 mg/mL concentration. The viral count at 50% inhibition was 2.5 × 104 PFU/mL compared to the initial viral count of 5 × 104 PFU/mL. In silico data for the wasp venom revealed a strong attraction to binding sites on the ACE2 protein, indicating ideal interactions. This substantiates the potential of wasp venom as a promising viral inhibitor against SARS-CoV-2, suggesting its consideration as a prospective natural preventive and curative antiviral drug. In conclusion, hymenopteran venoms, particularly wasp venom, hold promise as a source of potential therapeutic biomolecules against SARS-CoV-2. More research and clinical trials are needed to evaluate these results and investigate their potential for translation into innovative antiviral therapies.
Collapse
Affiliation(s)
- Eman A Abd El Maksoud
- Armed Forces Laboratories of Medical Research, El-Khalifa El-Maamoun, Mansheya El-Bakry, Heliopolis, Cairo Governorate, Egypt
| | - Magda H Rady
- Entomology Department Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Ahmed Gad Taha Mahmoud
- Microbiology Department, Armed Forces Laboratories of Medical Research, El-Khalifa El-Maamoun, Mansheya El-Bakry, Heliopolis, Cairo Governorate, Egypt
| | - Dalia Hamza
- Zoonoses Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Mohamed G Seadawy
- Biological Prevention Department, Chemical Warfare, Egypt Army, Cairo, Egypt
- Entomology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Eman E Essa
- Entomology Department Faculty of Science, Ain Shams University, Cairo, 11566, Egypt.
- Entomology Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
5
|
Vilas Boas LCP, Buccini DF, Berlanda RLA, Santos BDPO, Maximiano MR, Lião LM, Gonçalves S, Santos NC, Franco OL. Antiviral Activities of Mastoparan-L-Derived Peptides against Human Alphaherpesvirus 1. Viruses 2024; 16:948. [PMID: 38932240 PMCID: PMC11209138 DOI: 10.3390/v16060948] [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: 03/09/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Human alphaherpesvirus 1 (HSV-1) is a significantly widespread viral pathogen causing recurrent infections that are currently incurable despite available treatment protocols. Studies have highlighted the potential of antimicrobial peptides sourced from Vespula lewisii venom, particularly those belonging to the mastoparan family, as effective against HSV-1. This study aimed to demonstrate the antiviral properties of mastoparans, including mastoparan-L [I5, R8], mastoparan-MO, and [I5, R8] mastoparan, against HSV-1. Initially, Vero cell viability was assessed in the presence of these peptides, followed by the determination of antiviral activity, mechanism of action, and dose-response curves through plaque assays. Structural analyses via circular dichroism and nuclear magnetic resonance were conducted, along with evaluating membrane fluidity changes induced by [I5, R8] mastoparan using fluorescence-labeled lipid vesicles. Cytotoxic assays revealed high cell viability (>80%) at concentrations of 200 µg/mL for mastoparan-L and mastoparan-MO and 50 µg/mL for [I5, R8] mastoparan. Mastoparan-MO and [I5, R8] mastoparan exhibited over 80% HSV-1 inhibition, with up to 99% viral replication inhibition, particularly in the early infection stages. Structural analysis indicated an α-helical structure for [I5, R8] mastoparan, suggesting effective viral particle disruption before cell attachment. Mastoparans present promising prospects for HSV-1 infection control, although further investigation into their mechanisms is warranted.
Collapse
Affiliation(s)
- Liana Costa Pereira Vilas Boas
- Pós-Graduação em Patologia Molecular, Campus Darcy Ribeiro, Universidade de Brasília, Brasília 70910-900, DF, Brazil
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
| | - Danieli Fernanda Buccini
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
- Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande 79117-900, MS, Brazil
| | - Rhayfa Lorrayne Araújo Berlanda
- Pós-Graduação em Patologia Molecular, Campus Darcy Ribeiro, Universidade de Brasília, Brasília 70910-900, DF, Brazil
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
| | - Bruno de Paula Oliveira Santos
- Laboratório de Ressonância Magnética Nuclear, Instituto de Química, Universidade Federal de Goiás, Goiânia 74690-900, GO, Brazil
| | - Mariana Rocha Maximiano
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
- Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande 79117-900, MS, Brazil
| | - Luciano Morais Lião
- Laboratório de Ressonância Magnética Nuclear, Instituto de Química, Universidade Federal de Goiás, Goiânia 74690-900, GO, Brazil
| | - Sónia Gonçalves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal (N.C.S.)
| | - Nuno C. Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal (N.C.S.)
| | - Octávio Luiz Franco
- Pós-Graduação em Patologia Molecular, Campus Darcy Ribeiro, Universidade de Brasília, Brasília 70910-900, DF, Brazil
- Centro de Análises Bioquímicas e Proteômicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-760, DF, Brazil
- Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande 79117-900, MS, Brazil
| |
Collapse
|
6
|
da Silva Sanches PR, Sanchez-Velazquez R, Batista MN, Carneiro BM, Bittar C, De Lorenzo G, Rahal P, Patel AH, Cilli EM. Antiviral Evaluation of New Synthetic Bioconjugates Based on GA-Hecate: A New Class of Antivirals Targeting Different Steps of Zika Virus Replication. Molecules 2023; 28:4884. [PMID: 37446546 PMCID: PMC10343505 DOI: 10.3390/molecules28134884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
Re-emerging arboviruses represent a serious health problem due to their rapid vector-mediated spread, mainly in urban tropical areas. The 2013-2015 Zika virus (ZIKV) outbreak in South and Central America has been associated with cases of microcephaly in newborns and Guillain-Barret syndrome. We previously showed that the conjugate gallic acid-Hecate (GA-FALALKALKKALKKLKKALKKAL-CONH2)-is an efficient inhibitor of the hepatitis C virus. Here, we show that the Hecate peptide is degraded in human blood serum into three major metabolites. These metabolites conjugated with gallic acid were synthesized and their effect on ZIKV replication in cultured cells was evaluated. The GA-metabolite 5 (GA-FALALKALKKALKKL-COOH) was the most efficient in inhibiting two ZIKV strains of African and Asian lineage at the stage of both virus entry (virucidal and protective) and replication (post-entry). We also demonstrate that GA-metabolite 5 does not affect cell growth after 7 days of continuous treatment. Thus, this study identifies a new synthetic antiviral compound targeting different steps of ZIKV replication in vitro and with the potential for broad reactivity against other flaviviruses. Our work highlights a promising strategy for the development of new antivirals based on peptide metabolism and bioconjugation.
Collapse
Affiliation(s)
- Paulo Ricardo da Silva Sanches
- School of Pharmaceutical Science, São Paulo State University, Araraquara 14800-903, SP, Brazil
- MRC—University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G12 8QQ, UK; (R.S.-V.); (G.D.L.); (A.H.P.)
- Institute of Chemistry, São Paulo State University, Araraquara 14800-900, SP, Brazil
| | - Ricardo Sanchez-Velazquez
- MRC—University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G12 8QQ, UK; (R.S.-V.); (G.D.L.); (A.H.P.)
| | - Mariana Nogueira Batista
- Laboratory of Virology and Infectious Diseases, The Rockefeller University, New York, NY 10065, USA; (M.N.B.)
| | - Bruno Moreira Carneiro
- School of Health Science, Federal University of Rondonópolis, Rondonópolis 78736-900, MT, Brazil;
| | - Cintia Bittar
- School of Health Science, Federal University of Rondonópolis, Rondonópolis 78736-900, MT, Brazil;
| | - Giuditta De Lorenzo
- MRC—University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G12 8QQ, UK; (R.S.-V.); (G.D.L.); (A.H.P.)
| | - Paula Rahal
- Institute of Bioscience, Humanities and Exact Science, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil;
| | - Arvind H. Patel
- MRC—University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G12 8QQ, UK; (R.S.-V.); (G.D.L.); (A.H.P.)
| | - Eduardo Maffud Cilli
- Institute of Chemistry, São Paulo State University, Araraquara 14800-900, SP, Brazil
| |
Collapse
|
7
|
Oliveira I, Ferreira I, Jacob B, Cardenas K, Cerni F, Baia-da-Silva D, Arantes E, Monteiro W, Pucca M. Harnessing the Power of Venomous Animal-Derived Toxins against COVID-19. Toxins (Basel) 2023; 15:159. [PMID: 36828473 PMCID: PMC9967918 DOI: 10.3390/toxins15020159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/11/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Animal-derived venoms are complex mixtures of toxins triggering important biological effects during envenomings. Although venom-derived toxins are known for their potential of causing harm to victims, toxins can also act as pharmacological agents. During the COVID-19 pandemic, there was observed an increase in in-depth studies on antiviral agents, and since, to date, there has been no completely effective drug against the global disease. This review explores the crosstalk of animal toxins and COVID-19, aiming to map potential therapeutic agents derived from venoms (e.g., bees, snakes, scorpions, etc.) targeting COVID-19.
Collapse
Affiliation(s)
- Isadora Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Isabela Ferreira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Beatriz Jacob
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Kiara Cardenas
- Medical School, Federal University of Roraima, Boa Vista 69310-000, RR, Brazil
| | - Felipe Cerni
- Health Sciences Postgraduate Program, Federal University of Roraima, Boa Vista 69310-000, RR, Brazil
| | - Djane Baia-da-Silva
- Institute of Clinical Research Carlos Borborema, Dr. Heitor Vieira Dourado Tropical Medicine Foundation, Manaus 69850-000, AM, Brazil
- Postgraduate Program in Tropical Medicine, School of Health Sciences, Amazonas State University, Manaus 69850-000, AM, Brazil
- Department of Collective Health, Faculty of Medicine, Federal University of Amazonas, Manaus 69077-000, AM, Brazil
- Leônidas and Maria Deane Institute, Fiocruz Amazônia, Manaus 69057-070, AM, Brazil
- Faculty of Pharmacy, Nilton Lins University, Manaus 69058-040, AM, Brazil
| | - Eliane Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Wuelton Monteiro
- Institute of Clinical Research Carlos Borborema, Dr. Heitor Vieira Dourado Tropical Medicine Foundation, Manaus 69850-000, AM, Brazil
- Postgraduate Program in Tropical Medicine, School of Health Sciences, Amazonas State University, Manaus 69850-000, AM, Brazil
| | - Manuela Pucca
- Medical School, Federal University of Roraima, Boa Vista 69310-000, RR, Brazil
- Health Sciences Postgraduate Program, Federal University of Roraima, Boa Vista 69310-000, RR, Brazil
- Postgraduate Program in Tropical Medicine, School of Health Sciences, Amazonas State University, Manaus 69850-000, AM, Brazil
| |
Collapse
|
8
|
Saini J, Kaur P, Malik N, Lakhawat SS, Sharma PK. Antimicrobial peptides: A promising tool to combat multidrug resistance in SARS CoV2 era. Microbiol Res 2022; 265:127206. [PMID: 36162150 PMCID: PMC9491010 DOI: 10.1016/j.micres.2022.127206] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/26/2022] [Accepted: 09/16/2022] [Indexed: 10/25/2022]
Abstract
COVID-19 (Coronavirus Disease 2019), a life-threatening viral infection, is caused by a highly pathogenic virus named SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2). Currently, no treatment is available for COVID-19; hence there is an urgent need to find effective therapeutic drugs to combat COVID-19 pandemic. Considering the fact that the world is facing a major issue of antimicrobial drug resistance, naturally occurring compounds have the potential to achieve this goal. Antimicrobial peptides (AMPs) are naturally occurring antimicrobial agents which are effective against a wide variety of microbial infections. Therefore, the use of AMPs is an attractive therapeutic strategy for the treatment of SARS-CoV-2 infection. This review sheds light on the potential of antimicrobial peptides as antiviral agents followed by a comprehensive description of effective antiviral peptides derived from various natural sources found to be effective against SARS-CoV and other respiratory viruses. It also highlights the mechanisms of action of antiviral peptides with special emphasis on their effectiveness against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jasleen Saini
- Department of Biotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, India
| | - Pritpal Kaur
- Department of Biotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, India
| | - Naveen Malik
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | | | | |
Collapse
|
9
|
de Santana CJC, Pires Júnior OR, Fontes W, Palma MS, Castro MS. Mastoparans: A Group of Multifunctional α-Helical Peptides With Promising Therapeutic Properties. Front Mol Biosci 2022; 9:824989. [PMID: 35813822 PMCID: PMC9263278 DOI: 10.3389/fmolb.2022.824989] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Biologically active peptides have been attracting increasing attention, whether to improve the understanding of their mechanisms of action or in the search for new therapeutic drugs. Wasp venoms have been explored as a remarkable source for these molecules. In this review, the main findings on the group of wasp linear cationic α-helical peptides called mastoparans were discussed. These compounds have a wide variety of biological effects, including mast cell degranulation, activation of protein G, phospholipase A2, C, and D activation, serotonin and insulin release, and antimicrobial, hemolytic, and anticancer activities, which could lead to the development of new therapeutic agents.
Collapse
Affiliation(s)
- Carlos José Correia de Santana
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Osmindo Rodrigues Pires Júnior
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Mário Sérgio Palma
- Department of Basic and Applied Biology, Institute of Biosciences of Rio Claro, São Paulo State University, UNESP, Rio Claro, Brazil
| | - Mariana S. Castro
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
- *Correspondence: Mariana S. Castro,
| |
Collapse
|
10
|
Animal venoms as a source of antiviral peptides active against arboviruses: a systematic review. Arch Virol 2022; 167:1763-1772. [PMID: 35723756 DOI: 10.1007/s00705-022-05494-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 04/04/2022] [Indexed: 11/02/2022]
Abstract
Arthropod-borne viruses (arboviruses), such as Zika virus (ZIKV), chikungunya virus (CHIKV), dengue virus (DENV), yellow fever virus (YFV), and West Nile virus (WNV), are pathogens of global importance. Therefore, there has been an increasing need for new drugs for the treatment of these viral infections. In this context, antimicrobial peptides (AMPs) obtained from animal venoms stand out as promising compounds because they exhibit strong antiviral activity against emerging arboviral pathogens. Thus, we systematically searched and critically analyzed in vitro and in vivo studies that evaluated the anti-arbovirus effect of peptide derivatives from toxins produced by vertebrates and invertebrates. Thirteen studies that evaluated the antiviral action of 10 peptides against arboviruses were included in this review. The peptides were derived from the venom of scorpions, spiders, wasps, snakes, sea snails, and frogs and were tested against DENV, ZIKV, YFV, WNV, and CHIKV. Despite the high structural variety of the peptides included in this study, their antiviral activity appears to be associated with the presence of positive charges, an excess of basic amino acids (mainly lysine), and a high isoelectric point (above 8). These peptides use different antiviral mechanisms, the most common of which is the inhibition of viral replication, release, entry, or fusion. Moreover, peptides with virucidal and cytoprotective (pre-treatment) effects were also identified. In conclusion, animal-venom-derived peptides stand out as a promising alternative in the search and development of prototype antivirals against arboviruses.
Collapse
|
11
|
Broad-Spectrum Antiviral Activity of the Amphibian Antimicrobial Peptide Temporin L and Its Analogs. Int J Mol Sci 2022; 23:ijms23042060. [PMID: 35216177 PMCID: PMC8878748 DOI: 10.3390/ijms23042060] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 12/04/2022] Open
Abstract
The COVID-19 pandemic has evidenced the urgent need for the discovery of broad-spectrum antiviral therapies that could be deployed in the case of future emergence of novel viral threats, as well as to back up current therapeutic options in the case of drug resistance development. Most current antivirals are directed to inhibit specific viruses since these therapeutic molecules are designed to act on a specific viral target with the objective of interfering with a precise step in the replication cycle. Therefore, antimicrobial peptides (AMPs) have been identified as promising antiviral agents that could help to overcome this limitation and provide compounds able to act on more than a single viral family. We evaluated the antiviral activity of an amphibian peptide known for its strong antimicrobial activity against both Gram-positive and Gram-negative bacteria, namely Temporin L (TL). Previous studies have revealed that TL is endowed with widespread antimicrobial activity and possesses marked haemolytic activity. Therefore, we analyzed TL and a previously identified TL derivative (Pro3, DLeu9 TL, where glutamine at position 3 is replaced with proline, and the D-Leucine enantiomer is present at position 9) as well as its analogs, for their activity against a wide panel of viruses comprising enveloped, naked, DNA and RNA viruses. We report significant inhibition activity against herpesviruses, paramyxoviruses, influenza virus and coronaviruses, including SARS-CoV-2. Moreover, we further modified our best candidate by lipidation and demonstrated a highly reduced cytotoxicity with improved antiviral effect. Our results show a potent and selective antiviral activity of TL peptides, indicating that the novel lipidated temporin-based antiviral agents could prove to be useful additions to current drugs in combatting rising drug resistance and epidemic/pandemic emergencies.
Collapse
|
12
|
Zhao H, Yuen KY. Broad-spectrum Respiratory Virus Entry Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:137-153. [DOI: 10.1007/978-981-16-8702-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Sarhan M, El-Bitar AMH, Mohammadein A, Elshehaby M, Hotta H. Virucidal activity of oriental hornet Vespa orientalis venom against hepatitis C virus. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20210039. [PMID: 34868283 PMCID: PMC8609606 DOI: 10.1590/1678-9199-jvatitd-2021-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022] Open
Abstract
Background Hepatitis C virus (HCV) infection is a major worldwide health problem that can cause liver fibrosis and hepatocellular carcinoma (HCC). The clinical treatment of HCV infection mainly relies on the use of direct-acting antivirals (DAAs) that are usually expensive and have side effects. Therefore, achieving the discovery of more successful agents is always urgent. In this context, antiviral compounds that inhibit viral infections and disease progression with important therapeutic activities have been identified in animal venoms including arthropod toxins. This indicates that arthropod venoms represent a good natural source of promising candidates for new antivirals. Methods The antiviral activity of the wasp venom (WV), isolated from the Oriental hornet (Vespa orientalis), was assessed using cell culture technique with human hepatocellular carcinoma-derived cell line (Huh7it-1) and the recombinant strain of HCV genotype 2a (JFH1). Results The results revealed that WV inhibited HCV infectivity with 50% inhibitory concentration (IC50) of 10 ng/mL, while the 50% cytotoxic concentration (CC50) was 11,000 ng/mL. Time of addition experiment showed that the WV blocked HCV attachment/entry to the cells probably through virucidal effect. On the other hand, the venom showed no inhibitory effect on HCV replication. Conclusion WV can inhibit the entry stage of HCV infection at non-cytotoxic concentrations. Therefore, it could be considered a potential candidate for characterization of natural anti-HCV agents targeting the entry step.
Collapse
Affiliation(s)
- Moustafa Sarhan
- Molecular biology Laboratory, Zoology Department, Faculty of Science, Al-Azhar University, Assiut, Egypt.,Department of Microbiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Alaa M H El-Bitar
- Department of Microbiology, Kobe University Graduate School of Medicine, Kobe, Japan.,Zoology Department, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | - Amaal Mohammadein
- Zoology Department, Faculty of Science, Zagazig University, Zagazig, Egypt.,Department of Biology, College of Science, Taeif University, Saudi Arabia
| | - Mohammed Elshehaby
- Zoology Department, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | - Hak Hotta
- Department of Microbiology, Kobe University Graduate School of Medicine, Kobe, Japan.,Faculty of Clinical Nutrition and Dietetics, Konan Women's University, Kobe, Japan
| |
Collapse
|
14
|
Gentili V, Turrin G, Marchetti P, Rizzo S, Schiuma G, Beltrami S, Cristofori V, Illuminati D, Compagnin G, Trapella C, Rizzo R, Bortolotti D, Fantinati A. Synthesis and biological evaluation of novel rhodanine-based structures with antiviral activity towards HHV-6 virus. Bioorg Chem 2021; 119:105518. [PMID: 34861628 DOI: 10.1016/j.bioorg.2021.105518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/04/2021] [Accepted: 11/23/2021] [Indexed: 11/02/2022]
Abstract
An increased awareness of diseases associated with Human herpesvirus 6 (HHV-6) infection or reactivation has resulted in a growing interest in the evaluation of the best treatment options available for the clinical management of HHV-6 disease. However, no compound has yet been approved exclusively for HHV-6 infection treatment. For this reason, the identification of anti-HHV6 compounds provides a valuable opportunity for developing efficient antiviral therapies. A possible target for antiviral drugs is the virus-cell fusion step. In this study, we synthetized potential fusion intermediates inhibitors based on the rhodanine structure. The obtained derivatives were tested for cytotoxicity and for antiviral activity in human cells infected with HHV6. Level of infection was monitored by viral DNA quantification at different time points up to 7 days post infection. Among the synthetized derivatives, 9e showed a significative inhibitory effect on viral replication that lasted over 7 days, probably attributable to the particular combination of hydrophilic and hydrophobic substituents to the rhodanine moiety. Our results support the use of these amphipathic fusion inhibitors for the treatment of HHV-6 infections.
Collapse
Affiliation(s)
- Valentina Gentili
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Giulia Turrin
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Paolo Marchetti
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Sabrina Rizzo
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Giovanna Schiuma
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Silvia Beltrami
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Virginia Cristofori
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Davide Illuminati
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Greta Compagnin
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Claudio Trapella
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy.
| | - Roberta Rizzo
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy.
| | - Daria Bortolotti
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Anna Fantinati
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| |
Collapse
|
15
|
Tonk M, Růžek D, Vilcinskas A. Compelling Evidence for the Activity of Antiviral Peptides against SARS-CoV-2. Viruses 2021; 13:v13050912. [PMID: 34069206 PMCID: PMC8156787 DOI: 10.3390/v13050912] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022] Open
Abstract
Multiple outbreaks of epidemic and pandemic viral diseases have occurred in the last 20 years, including those caused by Ebola virus, Zika virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The emergence or re-emergence of such diseases has revealed the deficiency in our pipeline for the discovery and development of antiviral drugs. One promising solution is the extensive library of antimicrobial peptides (AMPs) produced by all eukaryotic organisms. AMPs are widely known for their activity against bacteria, but many possess additional antifungal, antiparasitic, insecticidal, anticancer, or antiviral activities. AMPs could therefore be suitable as leads for the development of new peptide-based antiviral drugs. Sixty therapeutic peptides had been approved by the end of 2018, with at least another 150 in preclinical or clinical development. Peptides undergoing clinical trials include analogs, mimetics, and natural AMPs. The advantages of AMPs include novel mechanisms of action that hinder the evolution of resistance, low molecular weight, low toxicity toward human cells but high specificity and efficacy, the latter enhanced by the optimization of AMP sequences. In this opinion article, we summarize the evidence supporting the efficacy of antiviral AMPs and discuss their potential to treat emerging viral diseases including COVID-19.
Collapse
Affiliation(s)
- Miray Tonk
- Institute for Insect Biotechnology, Justus Liebig University of Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany;
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
| | - Daniel Růžek
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic;
- Biology Centre of the Czech Academy of Sciences, Institute of Parasitology, Branisovska 31, 37005 Ceske Budejovice, Czech Republic
| | - Andreas Vilcinskas
- Institute for Insect Biotechnology, Justus Liebig University of Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany;
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Ohlebergsweg 12, 35392 Giessen, Germany
- Correspondence:
| |
Collapse
|
16
|
Ashaolu TJ, Nawaz A, Walayat N, Khalifa I. Potential "biopeptidal" therapeutics for severe respiratory syndrome coronaviruses: a review of antiviral peptides, viral mechanisms, and prospective needs. Appl Microbiol Biotechnol 2021; 105:3457-3470. [PMID: 33876282 PMCID: PMC8054851 DOI: 10.1007/s00253-021-11267-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/25/2021] [Accepted: 04/04/2021] [Indexed: 01/10/2023]
Abstract
Although great advances have been made on large-scale manufacturing of vaccines and antiviral-based drugs, viruses persist as the major cause of human diseases nowadays. The recent pandemic of coronavirus disease-2019 (COVID-19) mounts a lot of stress on the healthcare sector and the scientific society to search continuously for novel components with antiviral possibility. Herein, we narrated the different tactics of using biopeptides as antiviral molecules that could be used as an interesting alternative to treat COVID-19 patients. The number of peptides with antiviral effects is still low, but such peptides already displayed huge potentials to become pharmaceutically obtainable as antiviral medications. Studies showed that animal venoms, mammals, plant, and artificial sources are the main sources of antiviral peptides, when bioinformatics tools are used. This review spotlights bioactive peptides with antiviral activities against human viruses, especially the coronaviruses such as severe acute respiratory syndrome (SARS) virus, Middle East respiratory syndrome (MERS) virus, and severe acute respiratory syndrome coronavirus 2 (SARS-COV-2 or SARS-nCOV19). We also showed the data about well-recognized peptides that are still under investigations, while presenting the most potent ones that may become medications for clinical use.
Collapse
Affiliation(s)
- Tolulope Joshua Ashaolu
- Institute of Research and Development, Duy Tan University, Da Nang, 550000 Vietnam
- Faculty of Environmental and Chemical Engineering, Duy Tan University, Da Nang, 550000 Vietnam
| | - Asad Nawaz
- Jiangsu Key Laboratory of Crop Genetics and Physiology, Key Laboratory of Plant Functional Genomics of the Ministry of Education, College of Agriculture, Yangzhou University, Yangzhou, People’s Republic of China
| | - Noman Walayat
- Department of Food Science and Engineering, College of Ocean, Zhejiang University of Technology, Hangzhou, People’s Republic of China
| | - Ibrahim Khalifa
- Food Technology Department, Faculty of Agriculture, Banha University, 13736, Moshtohor, Cairo, Egypt
| |
Collapse
|
17
|
Mast Cell Functions Linking Innate Sensing to Adaptive Immunity. Cells 2020; 9:cells9122538. [PMID: 33255519 PMCID: PMC7761480 DOI: 10.3390/cells9122538] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Although mast cells (MCs) are known as key drivers of type I allergic reactions, there is increasing evidence for their critical role in host defense. MCs not only play an important role in initiating innate immune responses, but also influence the onset, kinetics, and amplitude of the adaptive arm of immunity or fine-tune the mode of the adaptive reaction. Intriguingly, MCs have been shown to affect T-cell activation by direct interaction or indirectly, by modifying the properties of antigen-presenting cells, and can even modulate lymph node-borne adaptive responses remotely from the periphery. In this review, we provide a summary of recent findings that explain how MCs act as a link between the innate and adaptive immunity, all the way from sensing inflammatory insult to orchestrating the final outcome of the immune response.
Collapse
|
18
|
Colella F, Scillitani G, Pierri CL. Sweet as honey, bitter as bile: Mitochondriotoxic peptides and other therapeutic proteins isolated from animal tissues, for dealing with mitochondrial apoptosis. Toxicology 2020; 447:152612. [PMID: 33171268 DOI: 10.1016/j.tox.2020.152612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
Mitochondria are subcellular organelles involved in cell metabolism and cell life-cycle. Their role in apoptosis regulation makes them an interesting target of new drugs for dealing with cancer or rare diseases. Several peptides and proteins isolated from animal and plant sources are known for their therapeutic properties and have been tested on cancer cell-lines and xenograft murine models, highlighting their ability in inducing cell-death by triggering mitochondrial apoptosis. Some of those molecules have been even approved as drugs. Conversely, many other bioactive compounds are still under investigation for their proapoptotic properties. In this review we report about a group of peptides, isolated from animal venoms, with potential therapeutic properties related to their ability in triggering mitochondrial apoptosis. This class of compounds is known with different names, such as mitochondriotoxins or mitocans.
Collapse
Affiliation(s)
- Francesco Colella
- Laboratory of Biochemistry, Molecular and Structural Biology, Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125, Bari, Italy
| | | | - Ciro Leonardo Pierri
- Laboratory of Biochemistry, Molecular and Structural Biology, Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125, Bari, Italy; BROWSer S.r.l. (https://browser-bioinf.com/) c/o Department of Biosciences, Biotechnologies, Biopharmaceutics, University "Aldo Moro" of Bari, Via E. Orabona, 4, 70126, Bari, Italy.
| |
Collapse
|
19
|
Herzig V, Cristofori-Armstrong B, Israel MR, Nixon SA, Vetter I, King GF. Animal toxins - Nature's evolutionary-refined toolkit for basic research and drug discovery. Biochem Pharmacol 2020; 181:114096. [PMID: 32535105 PMCID: PMC7290223 DOI: 10.1016/j.bcp.2020.114096] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/27/2022]
Abstract
Venomous animals have evolved toxins that interfere with specific components of their victim's core physiological systems, thereby causing biological dysfunction that aids in prey capture, defense against predators, or other roles such as intraspecific competition. Many animal lineages evolved venom systems independently, highlighting the success of this strategy. Over the course of evolution, toxins with exceptional specificity and high potency for their intended molecular targets have prevailed, making venoms an invaluable and almost inexhaustible source of bioactive molecules, some of which have found use as pharmacological tools, human therapeutics, and bioinsecticides. Current biomedically-focused research on venoms is directed towards their use in delineating the physiological role of toxin molecular targets such as ion channels and receptors, studying or treating human diseases, targeting vectors of human diseases, and treating microbial and parasitic infections. We provide examples of each of these areas of venom research, highlighting the potential that venom molecules hold for basic research and drug development.
Collapse
Affiliation(s)
- Volker Herzig
- School of Science & Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia; Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia.
| | | | - Mathilde R Israel
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Samantha A Nixon
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
20
|
Zhao H, To KKW, Sze KH, Yung TTM, Bian M, Lam H, Yeung ML, Li C, Chu H, Yuen KY. A broad-spectrum virus- and host-targeting peptide against respiratory viruses including influenza virus and SARS-CoV-2. Nat Commun 2020; 11:4252. [PMID: 32843628 DOI: 10.21203/rs.3.rs-18687/v1] [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: 03/09/2020] [Accepted: 07/27/2020] [Indexed: 05/22/2023] Open
Abstract
The 2019 novel respiratory virus (SARS-CoV-2) causes COVID-19 with rapid global socioeconomic disruptions and disease burden to healthcare. The COVID-19 and previous emerging virus outbreaks highlight the urgent need for broad-spectrum antivirals. Here, we show that a defensin-like peptide P9R exhibited potent antiviral activity against pH-dependent viruses that require endosomal acidification for virus infection, including the enveloped pandemic A(H1N1)pdm09 virus, avian influenza A(H7N9) virus, coronaviruses (SARS-CoV-2, MERS-CoV and SARS-CoV), and the non-enveloped rhinovirus. P9R can significantly protect mice from lethal challenge by A(H1N1)pdm09 virus and shows low possibility to cause drug-resistant virus. Mechanistic studies indicate that the antiviral activity of P9R depends on the direct binding to viruses and the inhibition of virus-host endosomal acidification, which provides a proof of concept that virus-binding alkaline peptides can broadly inhibit pH-dependent viruses. These results suggest that the dual-functional virus- and host-targeting P9R can be a promising candidate for combating pH-dependent respiratory viruses.
Collapse
Affiliation(s)
- Hanjun Zhao
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Kelvin K W To
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
- Li Ka Shing Faculty of Medicine, Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Kong-Hung Sze
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Timothy Tin-Mong Yung
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Mingjie Bian
- School of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Hoiyan Lam
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Man Lung Yeung
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Cun Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Hin Chu
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Kwok-Yung Yuen
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China.
- Li Ka Shing Faculty of Medicine, Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China.
| |
Collapse
|
21
|
Zhao H, To KKW, Sze KH, Yung TTM, Bian M, Lam H, Yeung ML, Li C, Chu H, Yuen KY. A broad-spectrum virus- and host-targeting peptide against respiratory viruses including influenza virus and SARS-CoV-2. Nat Commun 2020; 11:4252. [PMID: 32843628 PMCID: PMC7447754 DOI: 10.1038/s41467-020-17986-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/27/2020] [Indexed: 02/04/2023] Open
Abstract
The 2019 novel respiratory virus (SARS-CoV-2) causes COVID-19 with rapid global socioeconomic disruptions and disease burden to healthcare. The COVID-19 and previous emerging virus outbreaks highlight the urgent need for broad-spectrum antivirals. Here, we show that a defensin-like peptide P9R exhibited potent antiviral activity against pH-dependent viruses that require endosomal acidification for virus infection, including the enveloped pandemic A(H1N1)pdm09 virus, avian influenza A(H7N9) virus, coronaviruses (SARS-CoV-2, MERS-CoV and SARS-CoV), and the non-enveloped rhinovirus. P9R can significantly protect mice from lethal challenge by A(H1N1)pdm09 virus and shows low possibility to cause drug-resistant virus. Mechanistic studies indicate that the antiviral activity of P9R depends on the direct binding to viruses and the inhibition of virus-host endosomal acidification, which provides a proof of concept that virus-binding alkaline peptides can broadly inhibit pH-dependent viruses. These results suggest that the dual-functional virus- and host-targeting P9R can be a promising candidate for combating pH-dependent respiratory viruses.
Collapse
Affiliation(s)
- Hanjun Zhao
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Kelvin K W To
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
- Li Ka Shing Faculty of Medicine, Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Kong-Hung Sze
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Timothy Tin-Mong Yung
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Mingjie Bian
- School of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Hoiyan Lam
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Man Lung Yeung
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Cun Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Hin Chu
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China
| | - Kwok-Yung Yuen
- Li Ka Shing Faculty of Medicine, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong, China.
- Li Ka Shing Faculty of Medicine, Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China.
| |
Collapse
|
22
|
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
|
23
|
Yacoub T, Rima M, Karam M, Sabatier JM, Fajloun Z. Antimicrobials from Venomous Animals: An Overview. Molecules 2020; 25:molecules25102402. [PMID: 32455792 PMCID: PMC7287856 DOI: 10.3390/molecules25102402] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 01/17/2023] Open
Abstract
The inappropriate or excessive use of antimicrobial agents caused an emerging public health problem due to the resulting resistance developed by microbes. Therefore, there is an urgent need to develop effective antimicrobial strategies relying on natural agents with different mechanisms of action. Nature has been known to offer many bioactive compounds, in the form of animal venoms, algae, and plant extracts that were used for decades in traditional medicine. Animal venoms and secretions have been deeply studied for their wealth in pharmaceutically promising molecules. As such, they were reported to exhibit many biological activities of interest, such as antibacterial, antiviral, anticancer, and anti-inflammatory activities. In this review, we summarize recent findings on the antimicrobial activities of crude animal venoms/secretions, and describe the peptides that are responsible of these activities.
Collapse
Affiliation(s)
- Tania Yacoub
- Department of Biology, University of Balamand, Kalhat, Al-Kurah, P.O. box 100 Tripoli, Lebanon; (T.Y.); (M.K.)
| | - Mohamad Rima
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS U7104, Université de Strasbourg, 67400 Illkirch, France;
| | - Marc Karam
- Department of Biology, University of Balamand, Kalhat, Al-Kurah, P.O. box 100 Tripoli, Lebanon; (T.Y.); (M.K.)
| | - Jean-Marc Sabatier
- Université Aix-Marseille, Institut de NeuroPhysiopathologie, UMR 7051, Faculté de Médecine Secteur Nord, 51, Boulevard Pierre Dramard-CS80011, 13344-Marseille CEDEX 15, France
- Correspondence: (J.-M.S.); (Z.F.)
| | - Ziad Fajloun
- Faculty of Sciences 3, Lebanese University, Michel Slayman Tripoli Campus, Ras Maska 1352, Lebanon
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and its Applications, Doctoral School for Sciences and Technology, Lebanese University, El Mittein Street, 1300 Tripoli, Lebanon
- Correspondence: (J.-M.S.); (Z.F.)
| |
Collapse
|
24
|
Agarwal G, Gabrani R. Antiviral Peptides: Identification and Validation. Int J Pept Res Ther 2020; 27:149-168. [PMID: 32427225 PMCID: PMC7233194 DOI: 10.1007/s10989-020-10072-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/30/2020] [Accepted: 05/08/2020] [Indexed: 12/26/2022]
Abstract
Despite rapid advances in the human healthcare, the infection caused by certain viruses results in high morbidity and mortality accentuate the importance for development of new antivirals. The existing antiviral drugs are limited, due to their inadequate response, increased rate of resistance and several adverse side effects. Therefore, one of the newly emerging field “peptide-based therapeutics” against viruses is being explored and seems promising. Over the last few years, a lot of scientific effort has been made for the identification of novel and potential peptide-based therapeutics using various advanced technologies. Consequently, there are more than 60 approved peptide drugs available for sale in the market of United States, Europe, Japan, and some Asian countries. Moreover, the number of peptide drugs undergoing the clinical trials is rising gradually year by year. The peptide-based antiviral therapeutics have been approved for the Human immunodeficiency virus (HIV), Influenza virus and Hepatitis virus (B and C). This review enlightens the various peptide sources and the different approaches that have contributed to the search of potential antiviral peptides. These include computational approaches, natural and biological sources (library based high throughput screening) for the identification of lead peptide molecules against their target. Further the applications of few advanced techniques based on combinatorial chemistry and molecular biology have been illustrated to measure the binding parameters such as affinity and kinetics of the screened interacting partners. The employment of these advanced techniques can contribute to investigate antiviral peptide therapeutics for emerging infections.
Collapse
Affiliation(s)
- Garima Agarwal
- Department of Biotechnology, Center for Emerging Diseases, Jaypee Institute of Information Technology, Noida, UP 201309 India
| | - Reema Gabrani
- Department of Biotechnology, Center for Emerging Diseases, Jaypee Institute of Information Technology, Noida, UP 201309 India
| |
Collapse
|
25
|
Vilas Boas LCP, Campos ML, Berlanda RLA, de Carvalho Neves N, Franco OL. Antiviral peptides as promising therapeutic drugs. Cell Mol Life Sci 2019; 76:3525-3542. [PMID: 31101936 PMCID: PMC7079787 DOI: 10.1007/s00018-019-03138-w] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/04/2019] [Accepted: 05/07/2019] [Indexed: 01/28/2023]
Abstract
While scientific advances have led to large-scale production and widespread distribution of vaccines and antiviral drugs, viruses still remain a major cause of human diseases today. The ever-increasing reports of viral resistance and the emergence and re-emergence of viral epidemics pressure the health and scientific community to constantly find novel molecules with antiviral potential. This search involves numerous different approaches, and the use of antimicrobial peptides has presented itself as an interesting alternative. Even though the number of antimicrobial peptides with antiviral activity is still low, they already show immense potential to become pharmaceutically available antiviral drugs. Such peptides can originate from natural sources, such as those isolated from mammals and from animal venoms, or from artificial sources, when bioinformatics tools are used. This review aims to shed some light on antimicrobial peptides with antiviral activities against human viruses and update the data about the already well-known peptides that are still undergoing studies, emphasizing the most promising ones that may become medicines for clinical use.
Collapse
Affiliation(s)
| | - Marcelo Lattarulo Campos
- Centro de Análises Bioquímicas e Proteômicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, 70790-160, Brazil
- Departamento de Botânica e Ecologia, Instituto de Biociências, Universidade Federal de Mato Grosso, Cuiabá, MT, 78060-900, Brazil
| | - Rhayfa Lorrayne Araujo Berlanda
- Centro de Análises Bioquímicas e Proteômicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, 70790-160, Brazil
| | - Natan de Carvalho Neves
- Centro de Análises Bioquímicas e Proteômicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, 70790-160, Brazil
| | - Octávio Luiz Franco
- Universidade de Brasília, Pós-Graduação em Patologia Molecular, Campus Darcy Ribeiro, Brasília, DF, 70910-900, Brazil.
- Centro de Análises Bioquímicas e Proteômicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, 70790-160, Brazil.
- S-Inova Biotech, Pós-graduação em Biotecnologia Universidade Católica Dom Bosco, Campo Grande, MS, 79117-900, Brazil.
| |
Collapse
|
26
|
Optimized Mucosal Modified Vaccinia Virus Ankara Prime/Soluble gp120 Boost HIV Vaccination Regimen Induces Antibody Responses Similar to Those of an Intramuscular Regimen. J Virol 2019; 93:JVI.00475-19. [PMID: 31068425 DOI: 10.1128/jvi.00475-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/01/2019] [Indexed: 12/29/2022] Open
Abstract
The benefits of mucosal vaccines over injected vaccines are difficult to ascertain, since mucosally administered vaccines often induce serum antibody responses of lower magnitude than those induced by injected vaccines. This study aimed to determine if mucosal vaccination using a modified vaccinia virus Ankara expressing human immunodeficiency virus type 1 (HIV-1) gp120 (MVAgp120) prime and a HIV-1 gp120 protein boost could be optimized to induce serum antibody responses similar to those induced by an intramuscularly (i.m.) administered MVAgp120 prime/gp120 boost to allow comparison of an i.m. immunization regimen to a mucosal vaccination regimen for the ability to protect against a low-dose rectal simian-human immunodeficiency virus (SHIV) challenge. A 3-fold higher antigen dose was required for intranasal (i.n.) immunization with gp120 to induce serum anti-gp120 IgG responses not significantly different than those induced by i.m. immunization. gp120 fused to the adenovirus type 2 fiber binding domain (gp120-Ad2F), a mucosal targeting ligand, exhibited enhanced i.n. immunogenicity compared to gp120. MVAgp120 was more immunogenic after i.n. delivery than after gastric or rectal delivery. Using these optimized vaccines, an i.n. MVAgp120 prime/combined i.m. (gp120) and i.n. (gp120-Ad2F) boost regimen (i.n./i.m.-plus-i.n.) induced serum anti-gp120 antibody titers similar to those induced by the intramuscular prime/boost regimen (i.m./i.m.) in rabbits and nonhuman primates. Despite the induction of similar systemic anti-HIV-1 antibody responses, neither the i.m./i.m. nor the i.n./i.m.-plus-i.n. regimen protected against a repeated low-dose rectal SHIV challenge. These results demonstrate that immunization regimens utilizing the i.n. route are able to induce serum antigen-specific antibody responses similar to those induced by systemic immunization.IMPORTANCE Mucosal vaccination is proposed as a method of immunization able to induce protection against mucosal pathogens that is superior to protection provided by parenteral immunization. However, mucosal vaccination often induces serum antigen-specific immune responses of lower magnitude than those induced by parenteral immunization, making the comparison of mucosal and parenteral immunization difficult. We identified vaccine parameters that allowed an immunization regimen consisting of an i.n. prime followed by boosters administered by both i.n. and i.m. routes to induce serum antibody responses similar to those induced by i.m. prime/boost vaccination. Additional studies are needed to determine the potential benefit of mucosal immunization for HIV-1 and other mucosally transmitted pathogens.
Collapse
|
27
|
Xu J, Chen S, Jin J, Ma L, Guo M, Zhou C, Dou J. Inhibition of peptide BF-30 on influenza A virus infection in vitro/vivo by causing virion membrane fusion. Peptides 2019; 112:14-22. [PMID: 30447229 DOI: 10.1016/j.peptides.2018.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022]
Abstract
Influenza A virus is a leading cause of mortality in humans and poses a global health emergency due to its newly adapted and resistant strains. Thus, there is an urgency to develop novel anti-influenza drugs. Peptides are a type of biological molecule having a wide range of inhibitory effects against bacteria, fungi, viruses and cancer cells. The prospects of several peptides and their mechanisms of action have received significant attention. BF-30, a 30 amino acid residue peptide isolated from the venom of the snake, Bungarus fasciatus, is reported to have antibacterial and antitumor activities. Here, we demonstrated that the 50% cytotoxic concentration (CC50) of the peptide to MDCK cells is 67.7 μM. While BF-30 could inhibit the influenza virus strains H1N1, H3N2 and the oseltamivir-resistant strain H1N1, in vitro, with 50% effective concentration (EC50) of 5.2, 7.4 and 18.9 μM, respectively. In animal experiments, mice treated with BF-30 showed 50% survival at a dosage of 4 μM, with an approximately 2 log viral titer decrease in the lung. However, further studies showed that BF-30 worked on only the virus invasion stage, and inhibited the influenza virus infection by causing virion membrane fusion rather than interacting with hemagglutinin or neuraminidase. These results demonstrated that the peptide BF-30 exhibited an effective inhibitory activity against the influenza A virus and could be a promising candidate for influenza virus therapy.
Collapse
Affiliation(s)
- Jun Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| | - Shuo Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| | - Jing Jin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| | - Min Guo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| | - Changlin Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| | - Jie Dou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| |
Collapse
|
28
|
Cagno V, Tintori C, Civra A, Cavalli R, Tiberi M, Botta L, Brai A, Poli G, Tapparel C, Lembo D, Botta M. Novel broad spectrum virucidal molecules against enveloped viruses. PLoS One 2018; 13:e0208333. [PMID: 30532192 PMCID: PMC6285983 DOI: 10.1371/journal.pone.0208333] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 11/06/2018] [Indexed: 01/01/2023] Open
Abstract
Viral infections are an important cause of death worldwide. Unfortunately, there is still a lack of antiviral drugs or vaccines for a large number of viruses, and this represents a remarkable challenge particularly for emerging and re-emerging viruses. For this reason, the identification of broad spectrum antiviral compounds provides a valuable opportunity for developing efficient antiviral therapies. Here we report on a class of rhodanine and thiobarbituric derivatives displaying a broad spectrum antiviral activity against seven different enveloped viruses including an HSV-2 acyclovir resistant strain with favorable selectivity indexes. Due to their selective action on enveloped viruses and to their lipid oxidation ability, we hypothesize a mechanism on the viral envelope that affects the fluidity of the lipid bilayer, thus compromising the efficiency of virus-cell fusion and preventing viral entry.
Collapse
Affiliation(s)
- Valeria Cagno
- Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
- Department of Molecular Microbiology, University of Geneva, Geneva, Switzerland
| | - Cristina Tintori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Andrea Civra
- Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Marika Tiberi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Lorenzo Botta
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Annalaura Brai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Lead Discovery Siena S.r.l., Castelnuovo Berardenga, Siena, Italy
| | - Giulio Poli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Caroline Tapparel
- Department of Molecular Microbiology, University of Geneva, Geneva, Switzerland
| | - David Lembo
- Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
| | - Maurizio Botta
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Lead Discovery Siena S.r.l., Castelnuovo Berardenga, Siena, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
29
|
Yuan L, Zhang S, Wang Y, Li Y, Wang X, Yang Q. Surfactin Inhibits Membrane Fusion during Invasion of Epithelial Cells by Enveloped Viruses. J Virol 2018; 92:e00809-18. [PMID: 30068648 PMCID: PMC6189506 DOI: 10.1128/jvi.00809-18] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/25/2018] [Indexed: 01/15/2023] Open
Abstract
Because membrane fusion is a crucial step in the process by which enveloped viruses invade host cells, membrane fusion inhibitors can be effective drugs against enveloped viruses. We found that surfactin from Bacillus subtilis can suppress the proliferation of porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) in epithelial cells at a relatively low concentration range (15 to 50 μg/ml), without cytotoxicity or viral membrane disruption. Membrane fusion inhibition experiments demonstrate that surfactin treatment significantly reduces the rate at which the virus fuses to the cell membrane. Thermodynamic experiments show that the incorporation of small amounts of surfactin hinders the formation of negative curvature by lamellar-phase lipids, suggesting that surfactin acts a membrane fusion inhibitor. A fluorescent lipopeptide similar to surfactin was synthesized, and its ability to insert into the viral membrane was confirmed by spectroscopy. In vivo experiments have shown that oral administration of surfactin to piglets protects against PEDV infection. In conclusion, our study indicates that surfactin is a membrane fusion inhibitor with activity against enveloped viruses. As the first reported naturally occurring wedge lipid membrane fusion inhibitor, surfactin is likely to be a prototype for the development of a broad range of novel antiviral drugs.IMPORTANCE Membrane fusion inhibitors are a rapidly emerging class of antiviral drugs that inhibit the infection process of enveloped viruses. They can be classified, on the basis of the viral components targeted, as fusion protein targeting or membrane lipid targeting. Lipid-targeting membrane fusion inhibitors have a broader antiviral spectrum and are less likely to select for drug-resistant mutations. Here we show that surfactin is a membrane fusion inhibitor and has a strong antiviral effect. The insertion of surfactin into the viral envelope lipids reduces the probability of viral fusion. We also demonstrate that oral administration of surfactin protects piglets from PEDV infection. Surfactin is the first naturally occurring wedge lipid membrane fusion inhibitor that has been identified and may be effective against many viruses beyond the scope of this study. Understanding its mechanism of action provides a foundation for the development of novel antiviral agents.
Collapse
Affiliation(s)
- Lvfeng Yuan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Shuai Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Yongheng Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Yuchen Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Xiaoqing Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
30
|
Co-protoporphyrin IX and Sn-protoporphyrin IX inactivate Zika, Chikungunya and other arboviruses by targeting the viral envelope. Sci Rep 2018; 8:9805. [PMID: 29955082 PMCID: PMC6023862 DOI: 10.1038/s41598-018-27855-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/31/2018] [Indexed: 12/17/2022] Open
Abstract
The global situation of diseases transmitted by arthropod-borne viruses such as Dengue (DENV), Yellow Fever (YFV), Chikungunya (CHIKV) and Zika (ZIKV) viruses is alarming and treatment of human infection by these arboviruses faces several challenges. The discovery of broad-spectrum antiviral molecules, able to inactivate different groups of viruses, is an interesting approach. The viral envelope is a common structure among arboviruses, being a potential target for antivirals. Porphyrins are amphipathic molecules able to interact with membranes and absorb light, being widely used in photodynamic therapy. Previously, we showed that heme, Co-protoporphyrin IX (CoPPIX) and Sn-protoporphyrin IX (SnPPIX) directly inactivate DENV and YFV infectious particles. Here we demonstrate that the antiviral activity of these porphyrins can be broadened to CHIKV, ZIKV, Mayaro virus, Sindbis virus and Vesicular Stomatitis virus. Porphyrin treatment causes viral envelope protein loss, affecting viral morphology, adsorption and entry into target cells. Also, light-stimulation enhanced the SnPPIX activity against all tested arboviruses. In summary, CoPPIX and SnPPIX were shown to be efficient broad-spectrum compounds to inactivate medically and veterinary important viruses.
Collapse
|
31
|
Gomes B, Augusto MT, Felício MR, Hollmann A, Franco OL, Gonçalves S, Santos NC. Designing improved active peptides for therapeutic approaches against infectious diseases. Biotechnol Adv 2018; 36:415-429. [PMID: 29330093 DOI: 10.1016/j.biotechadv.2018.01.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/13/2017] [Accepted: 01/06/2018] [Indexed: 12/25/2022]
Abstract
Infectious diseases are one of the main causes of human morbidity and mortality. In the last few decades, pathogenic microorganisms' resistance to conventional drugs has been increasing, and it is now pinpointed as a major worldwide health concern. The need to search for new therapeutic options, as well as improved treatment outcomes, has therefore increased significantly, with biologically active peptides representing a new alternative. A substantial research effort is being dedicated towards their development, especially due to improved biocompatibility and target selectivity. However, the inherent limitations of peptide drugs are restricting their application. In this review, we summarize the current status of peptide drug development, focusing on antiviral and antimicrobial peptide activities, highlighting the design improvements needed, and those already being used, to overcome the drawbacks of the therapeutic application of biologically active peptides.
Collapse
Affiliation(s)
- Bárbara Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Marcelo T Augusto
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Mário R Felício
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Axel Hollmann
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal; Laboratory of Molecular Microbiology, Institute of Basic and Applied Microbiology, National University of Quilmes, Bernal, Buenos Aires, Argentina; Laboratory of Biointerfaces and Biomimetic Systems, CITSE, National University of Santiago del Estero-CONICET, Santiago del Estero, Argentina
| | - Octávio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília, Brasília, DF, Brazil; S-Inova Biotech, Pós-graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | - Sónia Gonçalves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
32
|
Vilas Boas LCP, de Lima LMP, Migliolo L, Mendes GDS, de Jesus MG, Franco OL, Silva PA. Linear antimicrobial peptides with activity against herpes simplex virus 1 and Aichi virus. Biopolymers 2017; 108. [PMID: 27161201 DOI: 10.1002/bip.22871] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 04/30/2016] [Accepted: 05/04/2016] [Indexed: 01/01/2023]
Abstract
Viruses are the major cause of disease and mortality worldwide. Nowadays there are treatments based on antivirals or prophylaxis with vaccines. However, the rising number of reports of viral resistance to current antivirals and the emergence of new types of virus has concerned the scientific community. In this scenario, the search for alternative treatments has led scientists to the discovery of antimicrobial peptides (AMPs) derived from many different sources. Since some of them have shown antiviral activities, here we challenged 10 synthetic peptides from different animal and plant sources against, herpes simplex virus 1 (HSV-1), and Aichi virus. Among them, the highlight was Pa-MAP from the polar fish Pleuronectes americanus, which caused around 90% of inhibition of the HSV with a selectivity index of 5 and a virucidal mechanism of action. Moreover, LL-37 from human neutrophils showed 96% of inhibition against the Aichi virus, showing a selectivity index of 3.4. The other evaluated peptides did not show significant antiviral activity. In conclusion, the present study demonstrated that Pa-MAP seems to be a reliable candidate for a possible alternative drug to treat HSV-1 infections. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 108: 1-6, 2017.
Collapse
Affiliation(s)
- Liana Costa Pereira Vilas Boas
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| | - Lídia Maria Pinto de Lima
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| | - Ludovico Migliolo
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil.,S-Inova Biotech, Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | - Gabriele Dos Santos Mendes
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| | - Maianne Gonçalves de Jesus
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| | - Octávio Luiz Franco
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil.,S-Inova Biotech, Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | - Paula Andréia Silva
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| |
Collapse
|
33
|
Silva JC, Neto LM, Neves RC, Gonçalves JC, Trentini MM, Mucury-Filho R, Smidt KS, Fensterseifer IC, Silva ON, Lima LD, Clissa PB, Vilela N, Guilhelmelli F, Silva LP, Rangel M, Kipnis A, Silva-Pereira I, Franco OL, Junqueira-Kipnis AP, Bocca AL, Mortari MR. Evaluation of the antimicrobial activity of the mastoparan Polybia-MPII isolated from venom of the social wasp Pseudopolybia vespiceps testacea (Vespidae, Hymenoptera). Int J Antimicrob Agents 2017; 49:167-175. [PMID: 28108242 DOI: 10.1016/j.ijantimicag.2016.11.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 10/05/2016] [Accepted: 11/10/2016] [Indexed: 02/07/2023]
Abstract
Mastoparans, a class of peptides found in wasp venom, have significant effects following a sting as well as useful applications in clinical practice. Among these is their potential use in the control of micro-organisms that cause infectious diseases with a significant impact on society. Thus, the present study describes the isolation and identification of a mastoparan peptide from the venom of the social wasp Pseudopolybia vespiceps and evaluated its antimicrobial profile against bacteria (Staphylococcus aureus and Mycobacterium abscessus subsp. massiliense), fungi (Candida albicans and Cryptococcus neoformans) and in vivo S. aureus infection. The membrane pore-forming ability was also assessed. The mastoparan reduced in vitro and ex vivo mycobacterial growth by 80% at 12.5 µM in infected peritoneal macrophages but did not affect the shape of bacterial cells at the dose tested (6.25 µM). The peptide also showed potent action against S. aureus in vitro (EC50 and EC90 values of 1.83 µM and 2.90 µM, respectively) and reduced the in vivo bacterial load after 6 days of topical treatment (5 mg/kg). Antifungal activity was significant, with EC50 and EC90 values of 12.9 µM and 15.3 µM, respectively, for C. albicans, and 11 µM and 22.70 µM, respectively, for C. neoformans. Peptides are currently attracting interest for their potential in the design of antimicrobial drugs, particularly due to the difficulty of micro-organisms in developing resistance to them. In this respect, Polybia-MPII proved to be highly effective, with a lower haemolysis rate compared with peptides of the same family.
Collapse
Affiliation(s)
- Juliana C Silva
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Lázaro M Neto
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Rogério C Neves
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Jaqueline C Gonçalves
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Monalisa M Trentini
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Ricardo Mucury-Filho
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Karina S Smidt
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Isabel C Fensterseifer
- Center for Proteomics and Biochemical Analyses, Genomic Science and Biotechnology Graduate Program, Catholic University of Brasília, Brasília, Brazil; Molecular Pathology Graduate Program, University of Brasilia, Brasília, Brazil
| | - Osmar N Silva
- Center for Proteomics and Biochemical Analyses, Genomic Science and Biotechnology Graduate Program, Catholic University of Brasília, Brasília, Brazil; Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Lilian D Lima
- Laboratory of Immunopathology, Butantan Institute, Sao Paulo, SP, Brazil
| | - Patricia B Clissa
- Laboratory of Immunopathology, Butantan Institute, Sao Paulo, SP, Brazil
| | - Nathália Vilela
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Fernanda Guilhelmelli
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Luciano P Silva
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil; Laboratory of Mass Spectrometry, Embrapa Genetic Resources and Biotechnology, Brasília, Brazil
| | - Marisa Rangel
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil; Laboratory of Immunopathology, Butantan Institute, Sao Paulo, SP, Brazil
| | - André Kipnis
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Ildinete Silva-Pereira
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Octavio L Franco
- Center for Proteomics and Biochemical Analyses, Genomic Science and Biotechnology Graduate Program, Catholic University of Brasília, Brasília, Brazil; Molecular Pathology Graduate Program, University of Brasilia, Brasília, Brazil; Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil; S-Inova Biotech, Graduate Biotechnology Program, Dom Bosco Catholic University, Campo Grande, Brazil
| | - Ana P Junqueira-Kipnis
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Anamelia L Bocca
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Márcia R Mortari
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.
| |
Collapse
|
34
|
da Mata ÉCG, Mourão CBF, Rangel M, Schwartz EF. Antiviral activity of animal venom peptides and related compounds. J Venom Anim Toxins Incl Trop Dis 2017; 23:3. [PMID: 28074089 PMCID: PMC5217322 DOI: 10.1186/s40409-016-0089-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/30/2016] [Indexed: 12/11/2022] Open
Abstract
Viruses exhibit rapid mutational capacity to trick and infect host cells, sometimes assisted through virus-coded peptides that counteract host cellular immune defense. Although a large number of compounds have been identified as inhibiting various viral infections and disease progression, it is urgent to achieve the discovery of more effective agents. Furthermore, proportionally to the great variety of diseases caused by viruses, very few viral vaccines are available, and not all are efficient. Thus, new antiviral substances obtained from natural products have been prospected, including those derived from venomous animals. Venoms are complex mixtures of hundreds of molecules, mostly peptides, that present a large array of biological activities and evolved to putatively target the biochemical machinery of different pathogens or host cellular structures. In addition, non-venomous compounds, such as some body fluids of invertebrate organisms, exhibit antiviral activity. This review provides a panorama of peptides described from animal venoms that present antiviral activity, thereby reinforcing them as important tools for the development of new therapeutic drugs.
Collapse
Affiliation(s)
- Élida Cleyse Gomes da Mata
- Laboratory of Toxinology, Department of Physiological Sciences, University of Brasília, Brasília, DF 70910-900 Brazil
| | | | - Marisa Rangel
- Laboratory of Toxinology, Department of Physiological Sciences, University of Brasília, Brasília, DF 70910-900 Brazil ; Laboratory of Immunopathology, Butantan Institute, São Paulo, SP 05508-900 Brazil
| | - Elisabeth Ferroni Schwartz
- Laboratory of Toxinology, Department of Physiological Sciences, University of Brasília, Brasília, DF 70910-900 Brazil
| |
Collapse
|
35
|
Irazazabal LN, Porto WF, Ribeiro SM, Casale S, Humblot V, Ladram A, Franco OL. Selective amino acid substitution reduces cytotoxicity of the antimicrobial peptide mastoparan. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2699-2708. [DOI: 10.1016/j.bbamem.2016.07.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 06/28/2016] [Accepted: 07/12/2016] [Indexed: 12/19/2022]
|
36
|
MP-V1 from the Venom of Social Wasp Vespula vulgaris Is a de Novo Type of Mastoparan that Displays Superior Antimicrobial Activities. Molecules 2016; 21:512. [PMID: 27104500 PMCID: PMC6273832 DOI: 10.3390/molecules21040512] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 03/19/2016] [Accepted: 04/12/2016] [Indexed: 01/10/2023] Open
Abstract
Mastoparans from the venom of social wasps have attracted considerable attention as effective antibiotic candidates. In this study, mastoparan V1 (MP-V1) from Vespula vulgaris was first disclosed to have a peptide amino acid sequence distinct from typical mastoparans and its biochemical properties and antimicrobial effects were compared with those of typical mastoparans MP-L, -X(V) and -B. Circular dichroism (CD) spectroscopy revealed that MP-V1 and -X(V) form more stable α-helical conformations in lipid membrane-like environments than MP-L and -B. In parallel, these two also showed more effective antimicrobial activities against the pathogens than did MP-L and -B. Although MP-V1 had a less stable α-helical conformation than MP-X(V), it showed stronger antimicrobial effects against Streptococcus mutans and Salmonella enterica than MP-X(V). In the meantime, analysis of hemolytic activity revealed a range of doses (~50 μM) that exhibited little potent cytotoxicity on human erythrocytes. Finally, the atypical MP-V1 peptide amino acid sequence provided important clues to understanding its antimicrobial mechanism from a structural perspective. Therefore, it has been concluded that MP-V1 is a de novo type of mastoparan with superior antimicrobial activities against both pathogenic bacteria and fungi, which may be useful in developing multipurpose antimicrobial drugs against infectious diseases.
Collapse
|
37
|
Zare-Zardini H, Taheri-Kafrani A, Ordooei M, Ebrahimi L, Tolueinia B, Soleimanizadeh M. Identification and biochemical characterization of a new antibacterial and antifungal peptide derived from the insect Sphodromantis viridis. BIOCHEMISTRY (MOSCOW) 2016; 80:433-40. [PMID: 25869360 DOI: 10.1134/s0006297915040069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Antimicrobial peptides are members of the immune system that protect the host from infection. In this study, a potent and structurally novel antimicrobial peptide was isolated and characterized from praying mantis Sphodromantis viridis. This 14-amino acid peptide was purified by RP-HPLC. Tandem mass spectrometry was used for sequencing this peptide, and the results showed that the peptide belongs to the Mastoparan family. The peptide was named Mastoparan-S. Mastoparan-S demonstrated that it has antimicrobial activities against a broad spectrum of microorganisms (Gram-positive and Gram-negative bacteria and fungi), and it was found to be more potent than common antibiotics such as kanamycin. Mastoparan-S showed higher antimicrobial activity against Gram-negative bacteria compared to Gram-positive ones and fungi. The minimum inhibitory concentration (MIC) values of Mastoparan-S are 15.1-28.3 µg/ml for bacterial and 19.3-24.6 µg/ml for fungal pathogens. In addition, this newly described peptide showed low hemolytic activity against human red blood cells. The in vitro cytotoxicity of Mastoparan-S was also evaluated on monolayer of normal human cells (HeLa) by MTT assay, and the results illustrated that Mastoparan-S had significant cytotoxicity at concentrations higher than 40 µg/ml and had no any cytotoxicity at the MIC (≤30 µg/ml). The findings of the present study reveal that this newly described peptide can be introduced as an appropriate candidate for treatment of topical infection.
Collapse
Affiliation(s)
- Hadi Zare-Zardini
- Young Researchers and Elite Club, Yazd Branch, Islamic Azad University, Yazd, Iran
| | | | | | | | | | | |
Collapse
|
38
|
The Gαo Activator Mastoparan-7 Promotes Dendritic Spine Formation in Hippocampal Neurons. Neural Plast 2015; 2016:4258171. [PMID: 26881110 PMCID: PMC4736189 DOI: 10.1155/2016/4258171] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 07/26/2015] [Accepted: 08/27/2015] [Indexed: 11/17/2022] Open
Abstract
Mastoparan-7 (Mas-7), an analogue of the peptide mastoparan, which is derived from wasp venom, is a direct activator of Pertussis toxin- (PTX-) sensitive G proteins. Mas-7 produces several biological effects in different cell types; however, little is known about how Mas-7 influences mature hippocampal neurons. We examined the specific role of Mas-7 in the development of dendritic spines, the sites of excitatory synaptic contact that are crucial for synaptic plasticity. We report here that exposure of hippocampal neurons to a low dose of Mas-7 increases dendritic spine density and spine head width in a time-dependent manner. Additionally, Mas-7 enhances postsynaptic density protein-95 (PSD-95) clustering in neurites and activates Gαo signaling, increasing the intracellular Ca2+ concentration. To define the role of signaling intermediates, we measured the levels of phosphorylated protein kinase C (PKC), c-Jun N-terminal kinase (JNK), and calcium-calmodulin dependent protein kinase IIα (CaMKIIα) after Mas-7 treatment and determined that CaMKII activation is necessary for the Mas-7-dependent increase in dendritic spine density. Our results demonstrate a critical role for Gαo subunit signaling in the regulation of synapse formation.
Collapse
|
39
|
Abstract
Effective antivirals have been developed against specific viruses, such as HIV, Hepatitis C virus and influenza virus. This 'one bug-one drug' approach to antiviral drug development can be successful, but it may be inadequate for responding to an increasing diversity of viruses that cause significant diseases in humans. The majority of viral pathogens that cause emerging and re-emerging infectious diseases are membrane-enveloped viruses, which require the fusion of viral and cell membranes for virus entry. Therefore, antivirals that target the membrane fusion process represent new paradigms for broad-spectrum antiviral discovery. In this Review, we discuss the mechanisms responsible for the fusion between virus and cell membranes and explore how broad-spectrum antivirals target this process to prevent virus entry.
Collapse
Affiliation(s)
- Frederic Vigant
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, #1124, New York, New York 10029, USA
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, #1124, New York, New York 10029, USA
| |
Collapse
|
40
|
Vila-Farrés X, López-Rojas R, Pachón-Ibáñez ME, Teixidó M, Pachón J, Vila J, Giralt E. Sequence-activity relationship, and mechanism of action of mastoparan analogues against extended-drug resistant Acinetobacter baumannii. Eur J Med Chem 2015; 101:34-40. [PMID: 26114809 DOI: 10.1016/j.ejmech.2015.06.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 06/04/2015] [Accepted: 06/07/2015] [Indexed: 10/23/2022]
Abstract
The treatment of some infectious diseases can currently be very challenging since the spread of multi-, extended- or pan-resistant bacteria has considerably increased over time. On the other hand, the number of new antibiotics approved by the FDA has decreased drastically over the last 30 years. The main objective of this study was to investigate the activity of wasp peptides, specifically mastoparan and some of its derivatives against extended-resistant Acinetobacter baumannii. We optimized the stability of mastoparan in human serum since the specie obtained after the action of the enzymes present in human serum is not active. Thus, 10 derivatives of mastoparan were synthetized. Mastoparan analogues (guanidilated at the N-terminal, enantiomeric version and mastoparan with an extra positive charge at the C-terminal) showed the same activity against Acinetobacter baumannii as the original peptide (2.7 μM) and maintained their stability to more than 24 h in the presence of human serum compared to the original compound. The mechanism of action of all the peptides was carried out using a leakage assay. It was shown that mastoparan and the abovementioned analogues were those that released more carboxyfluorescein. In addition, the effect of mastoparan and its enantiomer against A. baumannii was studied using transmission electron microscopy (TEM). These results suggested that several analogues of mastoparan could be good candidates in the battle against highly resistant A. baumannii infections since they showed good activity and high stability.
Collapse
Affiliation(s)
- Xavier Vila-Farrés
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Rafael López-Rojas
- Biomedical Institute of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Maria Eugenia Pachón-Ibáñez
- Biomedical Institute of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Jerónimo Pachón
- Biomedical Institute of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain
| | - Jordi Vila
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain; Department of Clinical Microbiology, CDB, Hospital Clinic, School of Medicine, University of Barcelona, Barcelona, Spain.
| | - Ernest Giralt
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Department of Organic Chemistry, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
41
|
Zhu JD, Meng W, Wang XJ, Wang HCR. Broad-spectrum antiviral agents. Front Microbiol 2015; 6:517. [PMID: 26052325 PMCID: PMC4440912 DOI: 10.3389/fmicb.2015.00517] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/09/2015] [Indexed: 12/24/2022] Open
Abstract
Development of highly effective, broad-spectrum antiviral agents is the major objective shared by the fields of virology and pharmaceutics. Antiviral drug development has focused on targeting viral entry and replication, as well as modulating cellular defense system. High throughput screening of molecules, genetic engineering of peptides, and functional screening of agents have identified promising candidates for development of optimal broad-spectrum antiviral agents to intervene in viral infection and control viral epidemics. This review discusses current knowledge, prospective applications, opportunities, and challenges in the development of broad-spectrum antiviral agents.
Collapse
Affiliation(s)
- Jun-Da Zhu
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University Beijing, China
| | - Wen Meng
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University Beijing, China
| | - Xiao-Jia Wang
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University Beijing, China
| | - Hwa-Chain R Wang
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville TN, USA
| |
Collapse
|
42
|
Moreno M, Giralt E. Three valuable peptides from bee and wasp venoms for therapeutic and biotechnological use: melittin, apamin and mastoparan. Toxins (Basel) 2015; 7:1126-50. [PMID: 25835385 PMCID: PMC4417959 DOI: 10.3390/toxins7041126] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/18/2015] [Accepted: 03/25/2015] [Indexed: 12/14/2022] Open
Abstract
While knowledge of the composition and mode of action of bee and wasp venoms dates back 50 years, the therapeutic value of these toxins remains relatively unexploded. The properties of these venoms are now being studied with the aim to design and develop new therapeutic drugs. Far from evaluating the extensive number of monographs, journals and books related to bee and wasp venoms and the therapeutic effect of these toxins in numerous diseases, the following review focuses on the three most characterized peptides, namely melittin, apamin, and mastoparan. Here, we update information related to these compounds from the perspective of applied science and discuss their potential therapeutic and biotechnological applications in biomedicine.
Collapse
Affiliation(s)
- Miguel Moreno
- Chemistry and Molecular Pharmacology, Institute for Research in Biomedicine (IRB Barcelona), Baldiri i Reixac, 10, Barcelona 08028, Spain.
| | - Ernest Giralt
- Chemistry and Molecular Pharmacology, Institute for Research in Biomedicine (IRB Barcelona), Baldiri i Reixac, 10, Barcelona 08028, Spain.
| |
Collapse
|
43
|
Martinez JP, Sasse F, Brönstrup M, Diez J, Meyerhans A. Antiviral drug discovery: broad-spectrum drugs from nature. Nat Prod Rep 2015; 32:29-48. [PMID: 25315648 DOI: 10.1039/c4np00085d] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Covering: up to April 2014. The development of drugs with broad-spectrum antiviral activities is a long pursued goal in drug discovery. It has been shown that blocking co-opted host-factors abrogates the replication of many viruses, yet the development of such host-targeting drugs has been met with scepticism mainly due to toxicity issues and poor translation to in vivo models. With the advent of new and more powerful screening assays and prediction tools, the idea of a drug that can efficiently treat a wide range of viral infections by blocking specific host functions has re-bloomed. Here we critically review the state-of-the-art in broad-spectrum antiviral drug discovery. We discuss putative targets and treatment strategies, with particular focus on natural products as promising starting points for antiviral lead development.
Collapse
Affiliation(s)
- J P Martinez
- Infection Biology Group, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
44
|
Piras AM, Sandreschi S, Maisetta G, Esin S, Batoni G, Chiellini F. Chitosan nanoparticles for the linear release of model cationic Peptide. Pharm Res 2015; 32:2259-65. [PMID: 25559891 DOI: 10.1007/s11095-014-1615-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE The present study is focused on the development of a model drug delivery system (DDS) based on Chitosan (CS) nanoparticles using Renin substrate I (RSI) as model agent. RSI shares the main chemical-physical features of several biologically active antimicrobial peptides (AMPs). AMPs have a great therapeutic potential that is hampered by their lability in the biological fluids and as such they are perfect candidates for DDS. The development studies of quality DDS loaded with AMPs would require highly sensitive and specific quantification assays. The use of RSI allowed for the fine-tuning and optimization of the formulation parameters to promote the hydrophobic interactions between CS and the cationic peptide, favour the loading of the active ingredient and enhance the release properties of the carrier. METHODS RSI was encapsulated in chitosan NPs by mean of ionic gelation and a chromogenic enzymatic essay was carried out for the release kinetics evaluation. RESULTS The developed formulations displayed almost 100% of encapsulation efficacy, low burst percentages, and a linear release of the model peptide. A release model was created showing a direct dependence on both the amount of RSI and NPs radius. CONCLUSIONS Although CS has always been formulated with negatively charged active agents (e.g. oligonucleotides or anionic proteins), the use of ionotropic gelation in presence of a small cationic active agent promoted the formation of "core-shell" NPs. The described model, with tuneable linear release rates, appears eligible for further exploitation such as the loading of therapeutically active AMPs.
Collapse
Affiliation(s)
- Anna Maria Piras
- Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via Giuseppe Moruzzi 3, 56124, Pisa, Italy
| | | | | | | | | | | |
Collapse
|