1
|
French S, Da Silva R, Storm J, Wastika CE, Cullen I, Have MT, Hughes GL, Modahl CM. Exploiting venom toxins in paratransgenesis to prevent mosquito-borne disease. Parasit Vectors 2025; 18:32. [PMID: 39881388 PMCID: PMC11776213 DOI: 10.1186/s13071-025-06663-9] [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: 09/25/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Mosquitoes are responsible for the transmission of numerous pathogens, including Plasmodium parasites, arboviruses and filarial worms. They pose a significant risk to public health with over 200 million cases of malaria per annum and approximately 4 billion people at risk of arthropod-borne viruses (arboviruses). Mosquito populations are geographically expanding into temperate regions and their distribution is predicted to continue increasing. Mosquito symbionts, including fungi, bacteria and viruses, have desirable traits for mosquito disease control including spreading horizontally and vertically through mosquito populations and potentially colonising multiple important vector species. Paratransgenesis, genetic modification of mosquito symbionts with effectors to target the pathogen rather than the vector, is a promising strategy to prevent the spread of mosquito-borne diseases. A variety of effectors can be expressed but venom toxins are excellent effector candidates because they are target specific, potent and stable. However, the only toxins to be explored in mosquito paratransgenesis to date are scorpine and mutated phospholipase A2. To enhance the scope, effectiveness and durability of paratransgenesis, an expanded arsenal of effectors is required. This review discusses other potential toxin effectors for future paratransgenesis studies based on prior in vitro and in vivo antiparasitic and antiviral studies and highlights the need for further research and investment in this area. In terms of mosquito-borne diseases, paratransgenesis strategies have been developed to target Plasmodium. We postulate the potential to apply this principle to target arboviruses using antiviral toxin effectors.
Collapse
Affiliation(s)
- Stephanie French
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Rachael Da Silva
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Janet Storm
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Christida E Wastika
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Tropical Disease, Liverpool School of Tropical Medicine, Liverpool, UK
| | - India Cullen
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Martijn Ten Have
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Grant L Hughes
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Tropical Disease, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Cassandra M Modahl
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, UK
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Tropical Disease, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
2
|
Owliaee I, Khaledian M, Shojaeian A, Madanchi H, Yarani R, Boroujeni AK, Shoushtari M. Antimicrobial Peptides Against Arboviruses: Mechanisms, Challenges, and Future Directions. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10430-0. [PMID: 39776036 DOI: 10.1007/s12602-024-10430-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
This review delves into the potential of antimicrobial peptides (AMPs) as promising candidates for combating arboviruses, focusing on their mechanisms of antiviral activity, challenges, and future directions. AMPs have shown promise in preventing arbovirus attachment to host cells, inducing interferon production, and targeting multiple viral stages, illustrating their multifaceted impact on arbovirus infections. Structural elucidation of AMP-viral complexes is explored to deepen the understanding of molecular determinants governing viral neutralization, paving the way for structure-guided design. Furthermore, this review highlights the potential of AMP-based combination therapies to create synergistic effects that enhance overall treatment outcomes while minimizing the likelihood of resistance development. Challenges such as susceptibility to proteases, toxicity, and scalable production are discussed alongside strategies to address these limitations. Additionally, the expanding applications of AMPs as vaccine adjuvants and antiviral delivery systems are emphasized, underscoring their versatility beyond direct antiviral functions.
Collapse
Affiliation(s)
- Iman Owliaee
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, 65178-38736, Iran
- Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, 65178-38736, Iran
| | - Mehran Khaledian
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, 65178-38736, Iran
- Department of Medical Entomology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, 65178-38736, Iran
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Hamid Madanchi
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, 35147-99442, Iran
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 13169-43551, Iran
| | - Reza Yarani
- Interventional Radiology Innovation at Stanford (IRIS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Armin Khaghani Boroujeni
- Skin Disease and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Shoushtari
- Department of Virology, Pasteur Institute of Iran, Tehran, 13169-43551, Iran.
| |
Collapse
|
3
|
Hboub H, Ben Mrid R, Bouchmaa N, Oukkache N, El Fatimy R. An in-depth exploration of snake venom-derived molecules for drug discovery in advancing antiviral therapeutics. Heliyon 2024; 10:e37321. [PMID: 39323826 PMCID: PMC11422003 DOI: 10.1016/j.heliyon.2024.e37321] [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: 02/26/2024] [Revised: 07/20/2024] [Accepted: 09/01/2024] [Indexed: 09/27/2024] Open
Abstract
Snake venom is a cocktail and rich source of various bioactive compounds that have been extensively studied for their potential as pharmaceutical agents due to their diverse chemical structures and wide range of biological activities. In light of the emergency and the re-emergence of viral infectious diseases that threaten human health and economic systems, exploring new fertile and rich fields such as snake venom is an attractive path for anti-viral drug discovery, especially in the lack of effective vaccines. Although 85 % of reported antiviral molecules belong to the phospholipase A2 (PLA2) family, other protein families including L-amino acid oxidases (LAAO), disintegrins, metalloproteases (SVMPs), and cathelicidins have also shown antiviral activity. Thus, in this review, we have highlighted the antiviral properties of compounds derived from snake venom and their mechanisms of action against virus classes like HIV, Coronaviridae, Flaviviridae, and Paramyxoviridae. Although the initial research emphasis has been on Retroviridae (HIV) and Flaviviridae viruses, it is crucial to extend the exploration of the potential of these compounds to other viruses. The utilization of snake venom-derived compounds as antivirals shows significant promise for the development of novel therapeutics to address viral infections. However, a more in-depth investigation is necessary to fully assess the potential of these compounds against other viruses and unveil the mechanisms underlying their action.
Collapse
Affiliation(s)
- Hicham Hboub
- Institute of Biological Sciences (ISSB), Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | - Reda Ben Mrid
- Institute of Biological Sciences (ISSB), Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | - Najat Bouchmaa
- Institute of Biological Sciences (ISSB), Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | - Naoual Oukkache
- Laboratory of Venoms and Toxins, Pasteur Institute of Morocco, Casablanca, 20360, Morocco
| | - Rachid El Fatimy
- Institute of Biological Sciences (ISSB), Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| |
Collapse
|
4
|
Zhou H, Zou L, Ren H, Shen Z, Lin Y, Cai H, Zhang J. Cathelicidin-BF regulates the AMPK/SIRT1/NF-κB pathway to ameliorate murine osteoarthritis: In vitro and in vivo studie. Int Immunopharmacol 2024; 134:112201. [PMID: 38718660 DOI: 10.1016/j.intimp.2024.112201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 06/03/2024]
Abstract
Osteoarthritis (OA) is a chronic degenerative disease with a significant prevalence that causes cartilage damage and can lead to disability. The main factors contributing to the onset and progression of OA include inflammation and degeneration of the extracellular matrix. Cathelicidin-BF (BF-30), a natural peptide derived from Bungarus fasciatus venom, has shown multiple important pharmacological effects. However, the action mechanism of BF-30 in OA treatment remains to be elucidated. In this research, X-ray and Safranin O staining were employed to evaluate the imageology and histomorphology differences in the knee joints of mice in vivo. Techniques such as Western blot analysis, RT-qPCR, ELISA, and immunofluorescence staining were applied to examine gene and protein level changes in in vitro experiments. It was found that BF-30 significantly decreased inflammation and enhanced extracellular matrix metabolism. For the first time, it was demonstrated that the positive effects of BF-30 are mediated through the activation of the AMPK/SIRT1/NF-κB pathway. Moreover, when BF-30 was co-administered with Compound C, an AMPK inhibitor, the therapeutic benefits of BF-30 were reversed in both in vivo and in vitro settings. In conclusion, the findings suggest that BF-30 could be a novel therapeutic agent for OA improvement.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China; Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200031, China.
| | - Linfang Zou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China
| | - Hui Ren
- Central Laboratory, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zhenyu Shen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China
| | - Yuanqu Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China
| | - Haikang Cai
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200031, China.
| | - Jingdong Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China.
| |
Collapse
|
5
|
Díaz-Gómez JL, Martín-Estal I, Rivera-Aboytes E, Gaxiola-Muñíz RA, Puente-Garza CA, García-Lara S, Castorena-Torres F. Biomedical applications of synthetic peptides derived from venom of animal origin: A systematic review. Biomed Pharmacother 2024; 170:116015. [PMID: 38113629 DOI: 10.1016/j.biopha.2023.116015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Development of therapeutic agents that have fewer adverse effects and have higher efficacy for diseases, such as cancer, metabolic disorders, neurological diseases, infections, cardiovascular diseases, and respiratory diseases, are required. Recent studies have focused on identifying novel sources for pharmaceutical molecules to develop therapies against these diseases. Among the sources for potentially new therapies, animal venom-derived molecules have generated much interest. Various animal venom-derived proteins and peptides have been isolated, identified, synthesized, and tested to develop drugs. Venom-derived peptides have several biomedical properties, such as proapoptotic, cell migration, and autophagy regulation activities in cancer cell models; induction of vasodilation by nitric oxide and regulation of angiotensin II; modification of insulin response by controlling calcium and potassium channels; regulation of pain receptor activity; modulation of immune cell activity; alteration of motor neuron activity; degradation or inhibition of β-amyloid plaque formation; antibacterial, antifungal, antiviral, and antiprotozoal activities; increase in sperm motility and potentiation of erectile function; reduction of intraocular pressure; anticoagulation, fibrinolytic, and antithrombotic activities; etc. This systematic review compiles these biomedical properties and potential biomedical applications of synthesized animal venom-derived peptides reported in the latest research. In addition, the limitations and areas of opportunity in this research field are discussed so that new studies can be developed based on the data presented.
Collapse
Affiliation(s)
- Jorge L Díaz-Gómez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Irene Martín-Estal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Elizabeth Rivera-Aboytes
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| | - Ramón Alonso Gaxiola-Muñíz
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - César A Puente-Garza
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| | - Silverio García-Lara
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| | - Fabiola Castorena-Torres
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico.
| |
Collapse
|
6
|
Mwangi J, Kamau PM, Thuku RC, Lai R. Design methods for antimicrobial peptides with improved performance. Zool Res 2023; 44:1095-1114. [PMID: 37914524 PMCID: PMC10802102 DOI: 10.24272/j.issn.2095-8137.2023.246] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/20/2023] [Indexed: 11/03/2023] Open
Abstract
The recalcitrance of pathogens to traditional antibiotics has made treating and eradicating bacterial infections more difficult. In this regard, developing new antimicrobial agents to combat antibiotic-resistant strains has become a top priority. Antimicrobial peptides (AMPs), a ubiquitous class of naturally occurring compounds with broad-spectrum antipathogenic activity, hold significant promise as an effective solution to the current antimicrobial resistance (AMR) crisis. Several AMPs have been identified and evaluated for their therapeutic application, with many already in the drug development pipeline. Their distinct properties, such as high target specificity, potency, and ability to bypass microbial resistance mechanisms, make AMPs a promising alternative to traditional antibiotics. Nonetheless, several challenges, such as high toxicity, lability to proteolytic degradation, low stability, poor pharmacokinetics, and high production costs, continue to hamper their clinical applicability. Therefore, recent research has focused on optimizing the properties of AMPs to improve their performance. By understanding the physicochemical properties of AMPs that correspond to their activity, such as amphipathicity, hydrophobicity, structural conformation, amino acid distribution, and composition, researchers can design AMPs with desired and improved performance. In this review, we highlight some of the key strategies used to optimize the performance of AMPs, including rational design and de novo synthesis. We also discuss the growing role of predictive computational tools, utilizing artificial intelligence and machine learning, in the design and synthesis of highly efficacious lead drug candidates.
Collapse
Affiliation(s)
- James Mwangi
- Key Laboratory of Bioactive Peptides of Yunnan Province, Engineering Laboratory of Peptides of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Centre for Non-Human Primates, Kunming Primate Research Centre, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Centre, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Peter Muiruri Kamau
- Key Laboratory of Bioactive Peptides of Yunnan Province, Engineering Laboratory of Peptides of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Centre for Non-Human Primates, Kunming Primate Research Centre, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Centre, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Rebecca Caroline Thuku
- Key Laboratory of Bioactive Peptides of Yunnan Province, Engineering Laboratory of Peptides of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Centre for Non-Human Primates, Kunming Primate Research Centre, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Centre, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ren Lai
- Key Laboratory of Bioactive Peptides of Yunnan Province, Engineering Laboratory of Peptides of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Centre for Non-Human Primates, Kunming Primate Research Centre, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Centre, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- Centre for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, Guangdong 511458, China. E-mail:
| |
Collapse
|
7
|
Fernandez GJ, Ramírez-Mejía JM, Castillo JA, Urcuqui-Inchima S. Vitamin D modulates expression of antimicrobial peptides and proinflammatory cytokines to restrict Zika virus infection in macrophages. Int Immunopharmacol 2023; 119:110232. [PMID: 37150017 DOI: 10.1016/j.intimp.2023.110232] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 05/09/2023]
Abstract
Although the impact of Zika virus (ZIKV) infection on human health has been well documented, we still have no vaccine or effective treatment. This fact highlights the importance of searching for alternative therapy for treating ZIKV. To search for ZIKV antivirals, we examined the effect of vitamin D in monocyte-derived macrophages (MDMs) differentiated in the presence of vitamin D (D3-MDM) and explored the molecular mechanisms by analyzing transcriptional profiles. Our data show the restriction of ZIKV infection in D3-MDMs as compared to MDMs. Transcriptional profiles show that vitamin D alters about 19% of Zika response genes (8.2% diminished and 10.8% potentiated). Among the genes with diminished expression levels, we found proinflammatory cytokines and chemokines such as IL6, TNF, IL1A, IL1B, and IL12B, CCL1, CCL4, CCL7, CXCL3, CXCL6, and CXCL8. On the other hand, genes with potentiated expression were related to degranulation such as Lysozyme, cathelicidin (CAMP), and Serglycin. Since the CAMP gene encodes the antimicrobial peptide LL-37, we treated MDMs with LL-37 and infected them with ZIKV. The results showed a decrease in the proportion of infected cells. Our data provide new insights into the role of vitamin D in restricting ZIKV infection in macrophages that are mediated by induction of cathelicidin/LL-37 expression and downregulation of proinflammatory genes. Results highlight the biological relevance of vitamin D-inducible peptides as an antiviral treatment for Zika fever.
Collapse
Affiliation(s)
- Geysson Javier Fernandez
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia; Grupo Biología y Control de Enfermedades Infecciosas, Universidad de Antioquia UdeA, Medellín, Colombia.
| | - Julieta M Ramírez-Mejía
- CIBIOP Group, Department of Applied Sciences and Engineering, Universidad EAFIT, Medellín, Antioquia, Colombia.
| | - Jorge Andrés Castillo
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia; Grupo de enfermedades infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia.
| |
Collapse
|
8
|
Jangpromma N, Konkchaiyaphum M, Punpad A, Sosiangdi S, Daduang S, Klaynongsruang S, Tankrathok A. Rational Design of RN15m4 Cathelin Domain-Based Peptides from Siamese Crocodile Cathelicidin Improves Antimicrobial Activity. Appl Biochem Biotechnol 2023; 195:1096-1108. [PMID: 36327032 DOI: 10.1007/s12010-022-04210-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Antimicrobial peptides are becoming a new generation of antibiotics due to their therapeutic potential and ability to decrease drug-resistant bacteria development. Cathelicidins are known as effective peptides of vertebrate immunity that play crucial roles in the defensive strategy against pathogens. To improve its potency, the RN15 antibacterial peptide derived from the cathelin domain of Crocodylus siamensis cathelicidin has been modified and its antimicrobial properties investigated. Peptides were derived by template-based and physicochemical designation. The RN15 derivative peptides were predicted through their structure modeling, antimicrobial potency, and peptide-membrane calculation. The antimicrobial and cytotoxic activities of candidate peptides were investigated. Simultaneous consideration of physicochemical characteristics, secondary structure modeling, and the result of antimicrobial peptide tools prediction indicated that RN15m4 peptide was a candidate derivative antimicrobial peptide. The RN15m4 peptide expresses antimicrobial activity against most Gram-positive and Gram-negative bacteria and fungi with a lower minimum inhibition concentration (MIC) than the parent peptide. Besides, the time-killing assay shows that the designed peptide performed its ability to quickly kill bacteria better than the original peptide. Scanning electron microscopy (SEM) displayed the destruction of the bacterial cell membrane caused by the RN15m4 peptide. In addition, the RN15m4 peptide exhibits low hemolytic activity and low cytotoxic activity as good as the template peptide. The RN15m4 peptide performs a range of antimicrobial activities with low cell toxicity. Our study has illustrated the combination approach to peptide design for potent antibiotic peptide discovery.
Collapse
Affiliation(s)
- Nisachon Jangpromma
- Faculty of Science, Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Khon Kaen University, 40000, Khon Kaen, Thailand.,Faculty of Science, Department of Integrated Science, Khon Kaen University, 40000, Khon Kaen, Thailand
| | - Monruedee Konkchaiyaphum
- Faculty of Science, Department of Biochemistry, Khon Kaen University, 40000, Khon Kaen, Thailand
| | - Arpaporn Punpad
- Faculty of Agricultural Technology, Department of Biotechnology, Kalasin University, 46000, Kalasin, Thailand
| | - Sirinthip Sosiangdi
- Faculty of Science, Department of Biochemistry, Khon Kaen University, 40000, Khon Kaen, Thailand
| | - Sakda Daduang
- Faculty of Science, Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Khon Kaen University, 40000, Khon Kaen, Thailand.,Faculty of Pharmaceutical Sciences, Division of Pharmacognosy and Toxicology, Khon Kaen University, 40000, Khon Kaen, Thailand
| | - Sompong Klaynongsruang
- Faculty of Science, Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Khon Kaen University, 40000, Khon Kaen, Thailand.,Faculty of Science, Department of Biochemistry, Khon Kaen University, 40000, Khon Kaen, Thailand
| | - Anupong Tankrathok
- Faculty of Science, Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Khon Kaen University, 40000, Khon Kaen, Thailand. .,Faculty of Agricultural Technology, Department of Biotechnology, Kalasin University, 46000, Kalasin, Thailand.
| |
Collapse
|
9
|
Zoladek J, Burlaud-Gaillard J, Chazal M, Desgraupes S, Jeannin P, Gessain A, Pardigon N, Hubert M, Roingeard P, Jouvenet N, Afonso PV. Human Claudin-Derived Peptides Block the Membrane Fusion Process of Zika Virus and Are Broad Flavivirus Inhibitors. Microbiol Spectr 2022; 10:e0298922. [PMID: 36040168 PMCID: PMC9603178 DOI: 10.1128/spectrum.02989-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/15/2022] [Indexed: 01/04/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that emerged in the Pacific islands in 2007 and spread to the Americas in 2015. The infection remains asymptomatic in most cases but can be associated with severe neurological disorders. Despite massive efforts, no specific drug or vaccine against ZIKV infection is available to date. Claudins are tight-junction proteins that favor the entry of several flaviviruses, including ZIKV. In this study, we identified two peptides derived from the N-terminal sequences of claudin-7 and claudin-1, named CL7.1 and CL1.1, respectively, that inhibited ZIKV infection in a panel of human cell lines. Using cell-to-cell fusion assays, we demonstrated that these peptides blocked the ZIKV E-mediated membrane fusion. A comparison of the antiviral efficacy of CL1.1 and CL7.1 pointed to the importance of the peptide amphipathicity. Electron microscopic analysis revealed that CL1.1 altered the ultrastructure of the viral particles likely by binding the virus lipid envelope. However, amphipathicity could not fully explain the antiviral activity of CL1.1. In silico docking simulations suggested that CL1.1 may also interact with the E protein, near its stem region. Overall, our data suggested that claudin-derived peptides inhibition may be linked to simultaneous interaction with the E protein and the viral lipid envelope. Finally, we found that CL1.1 also blocked infection by yellow fever and Japanese encephalitis viruses but not by HIV-1 or SARS-CoV-2. Our results provide a basis for the future development of therapeutics against a wide range of endemic and emerging flaviviruses. IMPORTANCE Zika virus (ZIKV) is a flavivirus transmitted by mosquito bites that have spread to the Pacific Islands and the Americas over the past decade. The infection remains asymptomatic in most cases but can cause severe neurological disorders. ZIKV is a major public health threat in areas of endemicity, and there is currently no specific antiviral drug or vaccine available. We identified two antiviral peptides deriving from the N-terminal sequences of claudin-7 and claudin-1 with the latter being the most effective. These peptides block the envelope-mediated membrane fusion. Our data suggested that the inhibition was likely achieved by simultaneously interacting with the viral lipid envelope and the E protein. The peptides also inhibited other flaviviruses. These results could provide the basis for the development of therapies that might target a wide array of flaviviruses from current epidemics and possibly future emergences.
Collapse
Affiliation(s)
- Jim Zoladek
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Julien Burlaud-Gaillard
- Inserm U1259 MAVIVH, Université de Tours and CHRU de Tours, Tours, France
- Plate-Forme IBiSA de Microscopie Electronique, Université de Tours and CHRU de Tours, Tours, France
| | - Maxime Chazal
- Unité Signalisation Antivirale, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Sophie Desgraupes
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Patricia Jeannin
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Antoine Gessain
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Nathalie Pardigon
- Groupe Arbovirus, Unité Environnement et Risques Infectieux, Institut Pasteur, Université Paris Cité, Paris, France
| | - Mathieu Hubert
- Unité Virus et Immunité, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Philippe Roingeard
- Inserm U1259 MAVIVH, Université de Tours and CHRU de Tours, Tours, France
- Plate-Forme IBiSA de Microscopie Electronique, Université de Tours and CHRU de Tours, Tours, France
| | - Nolwenn Jouvenet
- Unité Signalisation Antivirale, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Philippe V. Afonso
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| |
Collapse
|
10
|
Wang J, Jiang B, Wang K, Dai J, Dong C, Wang Y, Zhang P, Li M, Xu W, Wei L. A cathelicidin antimicrobial peptide from Hydrophis cyanocinctus inhibits Zika virus infection by downregulating expression of a viral entry factor. J Biol Chem 2022; 298:102471. [PMID: 36089062 PMCID: PMC9530963 DOI: 10.1016/j.jbc.2022.102471] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 11/05/2022] Open
Abstract
Zika virus (ZIKV) is a re-emerging flavivirus that causes conditions such as microcephaly and testis damage. The spread of ZIKV has become a major public health concern. Recent studies indicated that antimicrobial peptides are an ideal source for screening antiviral candidates with broad-spectrum antiviral activities, including against ZIKV. We herein found that Hc-CATH, a cathelicidin antimicrobial peptide identified from the sea snake Hydrophis cyanocinctus in our previous work, conferred protection against ZIKV infection in host cells and showed preventative efficacy and therapeutic efficacy in C57BL/6J mice, Ifnar1−/− mice, and pregnant mice. Intriguingly, we revealed that Hc-CATH decreased the susceptibility of host cells to ZIKV by downregulating expression of AXL, a TAM (TYRO3, AXL and MERTK) family kinase receptor that mediates ZIKV infection, and subsequently reversed the negative regulation of AXL on host’s type I interferon response. Furthermore, we showed that the cyclo-oxygenase-2/prostaglandin E2/adenylyl cyclase/protein kinase A pathway was involved in Hc-CATH-mediated AXL downregulation, and Hc-CATH in addition directly inactivated ZIKV particles by disrupting viral membrane. Finally, while we found Hc-CATH did not act on the late stage of ZIKV infection, structure–function relationship studies revealed that α-helix and phenylalanine residues are key structural requirements for its protective efficacy against initial ZIKV infection. In summary, we demonstrate that Hc-CATH provides prophylactic and therapeutic efficacy against ZIKV infection via downregulation of AXL, as well as inactivating the virion. Our findings reveal a novel mechanism of cathelicidin against viral infection and highlight the potential of Hc-CATH to prevent and treat ZIKV infection.
Collapse
Affiliation(s)
- Jing Wang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Bingyan Jiang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Kezhen Wang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jianfeng Dai
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Chunsheng Dong
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Yipeng Wang
- Department of Biopharmaceuticals, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Peng Zhang
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Li
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China.
| | - Lin Wei
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
11
|
Zupin L, dos Santos-Silva CA, Al Mughrbi ARH, Vilela LMB, Benko-Iseppon AM, Crovella S. Bioactive Antimicrobial Peptides: A New Weapon to Counteract Zoonosis. Microorganisms 2022; 10:1591. [PMID: 36014009 PMCID: PMC9414035 DOI: 10.3390/microorganisms10081591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Zoonoses have recently become the center of attention of the general population and scientific community. Notably, more than 30 new human pathogens have been identified in the last 30 years, 75% of which can be classified as zoonosis. The complete eradication of such types of infections is far out of reach, considering the limited understanding of animal determinants in zoonoses and their causes of emergence. Therefore, efforts must be doubled in examining the spread, persistence, and pathogenicity of zoonosis and studying possible clinical interventions and antimicrobial drug development. The search for antimicrobial bioactive compounds has assumed great emphasis, considering the emergence of multi-drug-resistant microorganisms. Among the biomolecules of emerging scientific interest are antimicrobial peptides (AMPs), potent biomolecules that can potentially act as important weapons against infectious diseases. Moreover, synthetic AMPs are easily tailored (bioinformatically) to target specific features of the pathogens to hijack, inducing no or very low resistance. Although very promising, previous studies on SAMPs' efficacy are still at their early stages. Indeed, further studies and better characterization on their mechanism of action with in vitro and in vivo assays are needed so as to proceed to their clinical application on human beings.
Collapse
Affiliation(s)
- Luisa Zupin
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | | | | | - Livia Maria Batista Vilela
- Centro de Biociências, Departamento de Genética, Universidade Federal de Pernambuco, Recife 50670-420, Brazil
| | - Ana Maria Benko-Iseppon
- Centro de Biociências, Departamento de Genética, Universidade Federal de Pernambuco, Recife 50670-420, Brazil
| | - Sergio Crovella
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha 2713, Qatar
| |
Collapse
|
12
|
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
|
13
|
Yu Y, Si L, Meng Y. Flavivirus Entry Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:171-197. [PMID: 35412141 DOI: 10.1007/978-981-16-8702-0_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Flaviviruses, including Dengue virus, Zika virus, Yellow fever virus, Japanese encephalitis virus, West Nile virus, cause thousands of deaths and millions of illnesses each year. The large outbreak of ZIKV in 2016 reminds us that flaviviruses can pose a serious threat to human safety and public health as emerging and re-emerging viruses. However, there are no specific drugs approved for the treatment of flavivirus infections. Due to no need to enter the cells, viral entry inhibitors have the unique advantage in suppressing viral infections. Flaviviruses bind to receptors and attach to the cell surface, then enter the endosome in a clathrin-dependent manner and finalizes the viral entry process after fusion with the cell membrane in a low pH environment. Small molecules, antibodies or peptides can inhibit flavivirus entry by targeting the above processes. Here, we focus on flavivirus entry inhibitors with well-defined target and antiviral activity. We hope that our review will provide a theoretical basis for flavivirus treatment and drug research and help to accelerate the clinical application of flavivirus entry inhibitors.
Collapse
Affiliation(s)
- Yufeng Yu
- Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Lulu Si
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Meng
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Dali University, Dali, Yunnan, China
| |
Collapse
|
14
|
Antiviral Effects of Animal Toxins: Is There a Way to Drugs? Int J Mol Sci 2022; 23:ijms23073634. [PMID: 35408989 PMCID: PMC8998278 DOI: 10.3390/ijms23073634] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/18/2022] Open
Abstract
Viruses infect all types of organisms, causing viral diseases, which are very common in humans. Since viruses use the metabolic pathways of their host cells to replicate, they are difficult to eradicate without affecting the cells. The most effective measures against viral infections are vaccinations and antiviral drugs, which selectively inhibit the viral replication cycle. Both methods have disadvantages, which requires the development of new approaches to the treatment of viral diseases. In the study of animal venoms, it was found that, in addition to toxicity, venoms exhibit other types of biological activity, including an antiviral one, the first mention of which dates back to middle of the last century, but detailed studies of their antiviral activity have been conducted over the past 15 years. The COVID-19 pandemic has reinforced these studies and several compounds with antiviral activity have been identified in venoms. Some of them are very active and can be considered as the basis for antiviral drugs. This review discusses recent antiviral studies, the found compounds with high antiviral activity, and the possible mechanisms of their action. The prospects for using the animal venom components to create antiviral drugs, and the expected problems and possible solutions are also considered.
Collapse
|
15
|
van Harten RM, Tjeerdsma-van Bokhoven JL, de Greeff A, Balhuizen MD, van Dijk A, Veldhuizen EJ, Haagsman HP, Scheenstra MR. d-enantiomers of CATH-2 enhance the response of macrophages against Streptococcus suis serotype 2. J Adv Res 2022; 36:101-112. [PMID: 35127168 PMCID: PMC8799869 DOI: 10.1016/j.jare.2021.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/20/2021] [Accepted: 05/23/2021] [Indexed: 11/30/2022] Open
Abstract
D-CATH-2 has strong antimicrobial activities towards multiple S.suis strains. D-CATH-2 ameliorates macrophage function. DCATH-2 binds LTA. DCATH-2 has prophylactic effect against S. suis infection in vivo.
Introduction Due to the increase of antibiotic resistant bacterial strains, there is an urgent need for development of alternatives to antibiotics. Cathelicidins can be such an alternative to antibiotics having both a direct antimicrobial capacity as well as an immunomodulatory function. Previously, the full d-enantiomer of chicken cathelicidin-2 (d-CATH-2) has shown to prophylactically protect chickens against infection 7 days post hatch when administered in ovo three days before hatch. Objectives To further evaluate d-CATH-2 in mammals as a candidate for an alternative to antibiotics. In this study, the prophylactic capacity of d-CATH-2 and two truncated derivatives, d-C(1–21) and d-C(4–21), was determined in mammalian cells. Methods Antibacterial assays; immune cell differentiation and modulation; cytotoxicity, isothermal titration calorimetry; in vivo prophylactic capacity of peptides in an S. suis infection model. Results d-CATH-2 and its derivatives were shown to have a strong direct antibacterial capacity against four different S. suis serotype 2 strains (P1/7, S735, D282, and OV625) in bacterial medium and even stronger in cell culture medium. In addition, d-CATH-2 and its derivatives ameliorated the efficiency of mouse bone marrow-derived macrophages (BMDM) and skewed mouse bone marrow-derived dendritic cells (BMDC) towards cells with a more macrophage-like phenotype. The peptides directly bind lipoteichoic acid (LTA) and inhibit LTA-induced activation of macrophages. In addition, S. suis killed by the peptide was unable to further activate mouse macrophages, which indicates that S. suis was eliminated by the previously reported silent killing mechanism. Administration of d-C(1–21) at 24 h or 7 days before infection resulted in a small prophylactic protection with reduced disease severity and reduced mortality of the treated mice. Conclusion d-enantiomers of CATH-2 show promise as anti-infectives against pathogenic S. suis for application in mammals.
Collapse
Affiliation(s)
- Roel M. van Harten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, the Netherlands
| | | | - Astrid de Greeff
- Wageningen Bioveterinary Research, Wageningen University & Research, Houtribweg 39, 8221 RA Lelystad, the Netherlands
| | - Melanie D. Balhuizen
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, the Netherlands
| | - Albert van Dijk
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, the Netherlands
| | - Edwin J.A. Veldhuizen
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, the Netherlands
- Corresponding author.
| | - Henk P. Haagsman
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, the Netherlands
| | - Maaike R. Scheenstra
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
16
|
Endogenous cathelicidin is required for protection against ZIKV-caused testis damage via inactivating virons. Antiviral Res 2022; 198:105248. [PMID: 35038500 DOI: 10.1016/j.antiviral.2022.105248] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 12/18/2022]
Abstract
Cathelicidins have been shown to effectively inhibit flavivirus replication in vitro. However, the effects of mouse and human endogenous cathelicidins on flavivirus infection in vivo are rarely known. We herein found that mouse endogenous cathelicidin CRAMP was significantly up-regulated upon Zika virus (ZIKV) infection. CRAMP deficiency markedly exacerbated ZIKV replication in testis, and aggravated ZIKV-induced testicular damage and ZIKV-induced spermatic damage in mice, indicating that endogenous cathelicidin is required for protection against ZIKV-caused male infertility in mice. In vitro antiviral assay showed that both mouse cathelidin CRAMP and human cathelicidin LL-37 obviously reduced ZIKV-caused cytopathic effect and inhibited ZIKV replication in Vero cells. Antiviral mechanism revealed that they both directly inactivated ZIKV virons by binding to ZIKV virons and inducing the leakage of ZIKV genomic RNA, consequently inactivated ZIKV virons. In vivo antiviral assay indicated that both of them effectively inhibited ZIKV replication in C57BL/6J and IFNα/β receptor-deficient (Ifnar1-/-) mice when CRAMP or LL-37 was intravenously injected in parallel with or at 1 h after intravenous injection of ZIKV, implying that mouse cathelidin CRAMP and human cathelicidin LL-37 effectively inactivated ZIKV particles and exhibited therapeutic potential against ZIKV infection in vivo. Our findings reveal that endogenous cahtelicidin CRAMP and LL-37 act as inactivators of ZIKV, and effectively protect against ZIKV replication and ZIKV-induced male infertility, highlighting their potential for therapy of ZIKV infection.
Collapse
|
17
|
Xiong W, Li J, Feng Y, Chai J, Wu J, Hu Y, Tian M, Lu W, Xu X, Zou M. Brevinin-2GHk, a Peptide Derived from the Skin of Fejervarya limnocharis, Inhibits Zika Virus Infection by Disrupting Viral Integrity. Viruses 2021; 13:v13122382. [PMID: 34960651 PMCID: PMC8708736 DOI: 10.3390/v13122382] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/25/2021] [Indexed: 12/11/2022] Open
Abstract
Several years have passed since the Zika virus (ZIKV) pandemic reoccurred in 2015–2016. However, there is still a lack of proved protective vaccines or effective drugs against ZIKV. The peptide brevinin-2GHk (BR2GK), pertaining to the brevinin-2 family of antimicrobial peptides, has been reported to exhibit only weak antibacterial activity, and its antiviral effects have not been investigated. Thus, we analyzed the effect of BR2GK on ZIKV infection. BR2GK showed significant inhibitory activity in the early and middle stages of ZIKV infection, with negligible cytotoxicity. Furthermore, BR2GK was suggested to bind with ZIKV E protein and disrupt the integrity of the envelope, thus directly inactivating ZIKV. In addition, BR2GK can also penetrate the cell membrane, which may contribute to inhibition of the middle stage of ZIKV infection. BR2GK blocked ZIKV E protein expression with an IC50 of 3.408 ± 0.738 μΜ. In summary, BR2GK was found to be a multi-functional candidate and a potential lead compound for further development of anti-ZIKV drugs.
Collapse
Affiliation(s)
- Weichen Xiong
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jingyan Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yifei Feng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yunrui Hu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Maolin Tian
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wancheng Lu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Correspondence: (X.X.); (M.Z.)
| | - Min Zou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Correspondence: (X.X.); (M.Z.)
| |
Collapse
|
18
|
Damour A, Garcia M, Cho HS, Larivière A, Lévêque N, Park C, Bodet C. Characterisation of Antiviral Activity of Cathelicidins from Naked Mole Rat and Python bivittatus on Human Herpes Simplex Virus 1. Pharmaceuticals (Basel) 2021; 14:ph14080715. [PMID: 34451812 PMCID: PMC8398704 DOI: 10.3390/ph14080715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Hg-CATH and Pb-CATH4 are cathelicidins from Heterocephalus glaber and Python bivittatus that have been previously identified as potent antibacterial peptides. However, their antiviral properties were not previously investigated. In this study, their activity against the herpes simplex virus (HSV)-1 was evaluated during primary human keratinocyte infection. Both of them significantly reduced HSV-1 DNA replication and production of infectious viral particles in keratinocytes at noncytotoxic concentrations, with the stronger activity of Pb-CATH4. These peptides did not show direct virucidal activity and did not exhibit significant immunomodulatory properties, except for Pb-CATH4, which exerted a moderate proinflammatory action. All in all, our results suggest that Hg-CATH and Pb-CATH4 could be potent candidates for the development of new therapies against HSV-1.
Collapse
Affiliation(s)
- Alexia Damour
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
| | - Magali Garcia
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, 86021 Poitiers, France
| | - Hye-Sun Cho
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea; (H.-S.C.); (C.P.)
| | - Andy Larivière
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, 86021 Poitiers, France
| | - Nicolas Lévêque
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, 86021 Poitiers, France
| | - Chankyu Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea; (H.-S.C.); (C.P.)
| | - Charles Bodet
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
- Correspondence:
| |
Collapse
|
19
|
Rádis-Baptista G. Cell-Penetrating Peptides Derived from Animal Venoms and Toxins. Toxins (Basel) 2021; 13:147. [PMID: 33671927 PMCID: PMC7919042 DOI: 10.3390/toxins13020147] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/31/2021] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Abstract
Cell-penetrating peptides (CPPs) comprise a class of short polypeptides that possess the ability to selectively interact with the cytoplasmic membrane of certain cell types, translocate across plasma membranes and accumulate in the cell cytoplasm, organelles (e.g., the nucleus and mitochondria) and other subcellular compartments. CPPs are either of natural origin or de novo designed and synthesized from segments and patches of larger proteins or designed by algorithms. With such intrinsic properties, along with membrane permeation, translocation and cellular uptake properties, CPPs can intracellularly convey diverse substances and nanomaterials, such as hydrophilic organic compounds and drugs, macromolecules (nucleic acids and proteins), nanoparticles (nanocrystals and polyplexes), metals and radionuclides, which can be covalently attached via CPP N- and C-terminals or through preparation of CPP complexes. A cumulative number of studies on animal toxins, primarily isolated from the venom of arthropods and snakes, have revealed the cell-penetrating activities of venom peptides and toxins, which can be harnessed for application in biomedicine and pharmaceutical biotechnology. In this review, I aimed to collate examples of peptides from animal venoms and toxic secretions that possess the ability to penetrate diverse types of cells. These venom CPPs have been chemically or structurally modified to enhance cell selectivity, bioavailability and a range of target applications. Herein, examples are listed and discussed, including cysteine-stabilized and linear, α-helical peptides, with cationic and amphipathic character, from the venom of insects (e.g., melittin, anoplin, mastoparans), arachnids (latarcin, lycosin, chlorotoxin, maurocalcine/imperatoxin homologs and wasabi receptor toxin), fish (pardaxins), amphibian (bombesin) and snakes (crotamine and cathelicidins).
Collapse
Affiliation(s)
- Gandhi Rádis-Baptista
- Laboratory of Biochemistry and Biotechnology, Institute for Marine Sciences, Federal University of Ceara, Fortaleza 60165-081, Brazil
| |
Collapse
|