251
|
Drayton M, Alford MA, Pletzer D, Haney EF, Machado Y, Luo HD, Overall CM, Kizhakkedathu JN, Hancock REW, Straus SK. Enzymatically releasable polyethylene glycol - host defense peptide conjugates with improved activity and biocompatibility. J Control Release 2021; 339:220-231. [PMID: 34597746 DOI: 10.1016/j.jconrel.2021.09.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/13/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
Host defense peptides (HDPs) have been the subject of great interest for the treatment of multidrug-resistant bacterial infections due to their multimodal activity and low induction of resistance. However, aggregation, toxicity, and short biological half-life have limited their applicability for clinical treatment. Many methods have been explored to alleviate these issues, such as polymer (e.g., polyethylene glycol (PEG)) conjugation, but these are often accompanied by reductions in the activity of the HDP. Here, we detail the design of a novel PEG-HDP conjugate incorporating an enzymatic cleavage sequence targeting matrix metalloproteinases (MMPs) that accumulate at sites of inflammation and infection. Addition of the cleavage sequence onto either the N- or the C-terminal region of the parent peptide (peptide 73, a derivative of the HDP aurein 2.2) was explored to determine the location for optimal antimicrobial activity following MMP cleavage; furthermore, the susceptibility of the peptide to MMP cleavage after conjugation to 2 kDa or 5 kDa PEG was examined. The top candidate, L73, utilized an N-terminal cleavage site that was subsequently conjugated to a 2 kDa PEG polymer. Both L73 and the conjugate exhibited no antimicrobial activity in vitro until cleaved by purified MMP, which liberated a peptide fragment with 16- or 63-fold improved activity, respectively, corresponding to a minimum inhibitory concentration (MIC) of 8 μg/mL, comparable to that of peptide 73 (4 μg/mL). Furthermore, PEG conjugation improved the blood compatibility and reduced the aggregation tendency of the HDP in vitro, indicating enhanced biocompatibility. When administered as a single subcutaneous dose (~3.6 mg, or a peptide concentration of 142 mg/kg) in a mouse abscess model of high-density methicillin-resistant Staphylococcus aureus (MRSA) infection, the conjugate displayed strong activity, reducing abscess size and bacterial load by 73.3% and 58-fold, respectively. This activity was completely lost when the cleavage site was rendered resistant to MMPs by the substitution of two d-amino acids, supporting the hypothesis that antimicrobial activity was dependent on cleavage by MMPs, which were shown here to increasingly accumulate at the abscess site up to 18 h post infection. Finally, the conjugate displayed biocompatibility in vivo, with no identifiable toxicity or aggregation.
Collapse
Affiliation(s)
- Matthew Drayton
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada; Centre for Blood Research, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Morgan A Alford
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver, BC V6T 1Z4, Canada
| | - Daniel Pletzer
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver, BC V6T 1Z4, Canada; Department of Microbiology and Immunology, University of Otago, 720 Cumberland St, Dunedin, New Zealand
| | - Evan F Haney
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver, BC V6T 1Z4, Canada
| | - Yoan Machado
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Haiming D Luo
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada; Centre for Blood Research, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Christopher M Overall
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, 2350 Health Sciences Mall, Vancouver V6T 1Z3, Canada; The School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver, BC V6T 1Z4, Canada
| | - Suzana K Straus
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada.
| |
Collapse
|
252
|
Singh V, Shrivastava S, Kumar Singh S, Kumar A, Saxena S. StaBle-ABPpred: a stacked ensemble predictor based on biLSTM and attention mechanism for accelerated discovery of antibacterial peptides. Brief Bioinform 2021; 23:6423526. [PMID: 34750606 DOI: 10.1093/bib/bbab439] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/06/2021] [Accepted: 09/24/2021] [Indexed: 01/29/2023] Open
Abstract
Due to the rapid emergence of multi-drug resistant (MDR) bacteria, existing antibiotics are becoming ineffective. So, researchers are looking for alternatives in the form of antibacterial peptides (ABPs) based medicines. The discovery of novel ABPs using wet-lab experiments is time-consuming and expensive. Many machine learning models have been proposed to search for new ABPs, but there is still scope to develop a robust model that has high accuracy and precision. In this work, we present StaBle-ABPpred, a stacked ensemble technique-based deep learning classifier that uses bidirectional long-short term memory (biLSTM) and attention mechanism at base-level and an ensemble of random forest, gradient boosting and logistic regression at meta-level to classify peptides as antibacterial or otherwise. The performance of our model has been compared with several state-of-the-art classifiers, and results were subjected to analysis of variance (ANOVA) test and its post hoc analysis, which proves that our model performs better than existing classifiers. Furthermore, a web app has been developed and deployed at https://stable-abppred.anvil.app to identify novel ABPs in protein sequences. Using this app, we identified novel ABPs in all the proteins of the Streptococcus phage T12 genome. These ABPs have shown amino acid similarities with experimentally tested antimicrobial peptides (AMPs) of other organisms. Hence, they could be chemically synthesized and experimentally validated for their activity against different bacteria. The model and app developed in this work can be further utilized to explore the protein diversity for identifying novel ABPs with broad-spectrum activity, especially against MDR bacterial pathogens.
Collapse
Affiliation(s)
- Vishakha Singh
- Department of Computer Science and Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India
| | - Sameer Shrivastava
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122, Uttar Pradesh, India
| | - Sanjay Kumar Singh
- Department of Computer Science and Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India
| | - Abhinav Kumar
- Department of Computer Science and Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India
| | - Sonal Saxena
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122, Uttar Pradesh, India
| |
Collapse
|
253
|
Luna-Reyes I, Pérez-Hernández EG, Delgado-Coello B, Mas-Oliva J. Peptides as Therapeutic Molecules to Neutralize Gram-negative Bacterial Lipopolysaccharides in Sepsis and Septic Shock. Arch Med Res 2021; 52:798-807. [DOI: 10.1016/j.arcmed.2021.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 08/03/2021] [Indexed: 11/16/2022]
|
254
|
Etayash H, Hancock REW. Host Defense Peptide-Mimicking Polymers and Polymeric-Brush-Tethered Host Defense Peptides: Recent Developments, Limitations, and Potential Success. Pharmaceutics 2021; 13:1820. [PMID: 34834239 PMCID: PMC8621177 DOI: 10.3390/pharmaceutics13111820] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/17/2022] Open
Abstract
Amphiphilic antimicrobial polymers have attracted considerable interest as structural mimics of host defense peptides (HDPs) that provide a broad spectrum of activity and do not induce bacterial-drug resistance. Likewise, surface engineered polymeric-brush-tethered HDP is considered a promising coating strategy that prevents infections and endows implantable materials and medical devices with antifouling and antibacterial properties. While each strategy takes a different approach, both aim to circumvent limitations of HDPs, enhance physicochemical properties, therapeutic performance, and enable solutions to unmet therapeutic needs. In this review, we discuss the recent advances in each approach, spotlight the fundamental principles, describe current developments with examples, discuss benefits and limitations, and highlight potential success. The review intends to summarize our knowledge in this research area and stimulate further work on antimicrobial polymers and functionalized polymeric biomaterials as strategies to fight infectious diseases.
Collapse
Affiliation(s)
| | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver, BC V6T 1Z4, Canada;
| |
Collapse
|
255
|
Bin Hafeez A, Jiang X, Bergen PJ, Zhu Y. Antimicrobial Peptides: An Update on Classifications and Databases. Int J Mol Sci 2021; 22:11691. [PMID: 34769122 PMCID: PMC8583803 DOI: 10.3390/ijms222111691] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Antimicrobial peptides (AMPs) are distributed across all kingdoms of life and are an indispensable component of host defenses. They consist of predominantly short cationic peptides with a wide variety of structures and targets. Given the ever-emerging resistance of various pathogens to existing antimicrobial therapies, AMPs have recently attracted extensive interest as potential therapeutic agents. As the discovery of new AMPs has increased, many databases specializing in AMPs have been developed to collect both fundamental and pharmacological information. In this review, we summarize the sources, structures, modes of action, and classifications of AMPs. Additionally, we examine current AMP databases, compare valuable computational tools used to predict antimicrobial activity and mechanisms of action, and highlight new machine learning approaches that can be employed to improve AMP activity to combat global antimicrobial resistance.
Collapse
Affiliation(s)
- Ahmer Bin Hafeez
- Centre of Biotechnology and Microbiology, University of Peshawar, Peshawar 25120, Pakistan;
| | - Xukai Jiang
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (X.J.); (P.J.B.)
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, China
| | - Phillip J. Bergen
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (X.J.); (P.J.B.)
| | - Yan Zhu
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (X.J.); (P.J.B.)
| |
Collapse
|
256
|
Nunes LGP, Reichert T, Machini MT. His-Rich Peptides, Gly- and His-Rich Peptides: Functionally Versatile Compounds with Potential Multi-Purpose Applications. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10302-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
257
|
Róg T, Girych M, Bunker A. Mechanistic Understanding from Molecular Dynamics in Pharmaceutical Research 2: Lipid Membrane in Drug Design. Pharmaceuticals (Basel) 2021; 14:1062. [PMID: 34681286 PMCID: PMC8537670 DOI: 10.3390/ph14101062] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/17/2022] Open
Abstract
We review the use of molecular dynamics (MD) simulation as a drug design tool in the context of the role that the lipid membrane can play in drug action, i.e., the interaction between candidate drug molecules and lipid membranes. In the standard "lock and key" paradigm, only the interaction between the drug and a specific active site of a specific protein is considered; the environment in which the drug acts is, from a biophysical perspective, far more complex than this. The possible mechanisms though which a drug can be designed to tinker with physiological processes are significantly broader than merely fitting to a single active site of a single protein. In this paper, we focus on the role of the lipid membrane, arguably the most important element outside the proteins themselves, as a case study. We discuss work that has been carried out, using MD simulation, concerning the transfection of drugs through membranes that act as biological barriers in the path of the drugs, the behavior of drug molecules within membranes, how their collective behavior can affect the structure and properties of the membrane and, finally, the role lipid membranes, to which the vast majority of drug target proteins are associated, can play in mediating the interaction between drug and target protein. This review paper is the second in a two-part series covering MD simulation as a tool in pharmaceutical research; both are designed as pedagogical review papers aimed at both pharmaceutical scientists interested in exploring how the tool of MD simulation can be applied to their research and computational scientists interested in exploring the possibility of a pharmaceutical context for their research.
Collapse
Affiliation(s)
- Tomasz Róg
- Department of Physics, University of Helsinki, 00014 Helsinki, Finland;
| | - Mykhailo Girych
- Department of Physics, University of Helsinki, 00014 Helsinki, Finland;
| | - Alex Bunker
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland;
| |
Collapse
|
258
|
Rodríguez AA, Otero-González A, Ghattas M, Ständker L. Discovery, Optimization, and Clinical Application of Natural Antimicrobial Peptides. Biomedicines 2021; 9:1381. [PMID: 34680498 PMCID: PMC8533436 DOI: 10.3390/biomedicines9101381] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Antimicrobial peptides (AMPs) are widespread in multicellular organisms. These structurally diverse molecules are produced as the first line of defense against pathogens such as bacteria, viruses, fungi, and parasites. Also known as host defense peptides in higher eukaryotic organisms, AMPs display immunomodulatory and anticancer activities. During the last 30 years, technological advances have boosted the research on antimicrobial peptides, which have also attracted great interest as an alternative to tackling the antimicrobial resistance scenario mainly provoked by some bacterial and fungal pathogens. However, the introduction of natural AMPs in clinical trials faces challenges such as proteolytic digestion, short half-lives, and cytotoxicity upon systemic and oral application. Therefore, some strategies have been implemented to improve the properties of AMPs aiming to be used as effective therapeutic agents. In the present review, we summarize the discovery path of AMPs, focusing on preclinical development, recent advances in chemical optimization and peptide delivery systems, and their introduction into the market.
Collapse
Affiliation(s)
- Armando A. Rodríguez
- Core Facility for Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Maretchia Ghattas
- Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11511, Egypt;
| | - Ludger Ständker
- Core Facility for Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
259
|
Cao F, Ma G, Mei L, Zhu G, Song M, Qin Q, Jiao M. Development of disulfide bond crosslinked antimicrobial peptide hydrogel. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
260
|
Erdem Büyükkiraz M, Kesmen Z. Recombinant expression and coexpression of oyster defensin and proline-rich peptide in Komagataella phaffii. Biotechnol Appl Biochem 2021; 69:1998-2007. [PMID: 34586650 DOI: 10.1002/bab.2262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 09/19/2021] [Indexed: 11/06/2022]
Abstract
Proline-rich peptide (CgPrp) and defensin (CgDef), oyster (Crassostrea gigas)-originated antimicrobial peptides (AMPs), were produced by the recombinant technique in Komagataella phaffii GS115 cells. For this purpose, the nucleotide sequences encoding the CgPrp and CgDef peptides were synthesized by the recursive PCR technique, and ligated in pPICZaA expression vector. Additionally, the expression cassettes of pPICZαA-CgDef and pPICZαA-CgPrp were combined using in vitro multimer ligation strategy to construct the coexpression vector pPICZaA-CgPrp-CgDef. The expression and coexpression vectors transformed into K. phaffii GS115 cells by electroporation. At the end of the 0.5% methanol-induced expression stage for 96 h, the recombinant peptides were purified from the culture medium. The concentrations of purified peptides were changed between 1.05 and 1.21 mg/L. The recombinant peptides successfully inhibited the growth of tested Gram-positive bacterial strains belonging to Staphylococcus aureus, methicillin-resistant S. aureus (MRSA), Listeria monocytogenes, and Bacillus cereus. The minimum inhibitory concentrations (MIC) of recombinant CgPrp, CgDef, and CgPrp-CgDef peptides against tested bacteria were in the range of 12.50-25.00, 18.75-75.00, and 5.80-11.60 pg/μl, respectively. The results of the study proved that the recombinant CgPrp, CgDef, and CgPrp-CgDef peptides expressed in K. phaffii might have good potential for the inhibition of common Gram-positive pathogenic bacteria, including drug-resistant MRSA.
Collapse
Affiliation(s)
| | - Zülal Kesmen
- Faculty of Engineering, Food Engineering Department, Erciyes University, Kayseri, Turkey
| |
Collapse
|
261
|
Zhang L. Interaction of Human β Defensin Type 3 (hBD-3) with Different PIP2-Containing Membranes, a Molecular Dynamics Simulation Study. J Chem Inf Model 2021; 61:4670-4686. [PMID: 34473496 DOI: 10.1021/acs.jcim.1c00805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Human β defensin type 3 (hBD-3) is a cysteine-rich small antibacterial peptide. It belongs to the human innate immune system. hBD-3 has six cysteine residues, which form three pairs of disulfide bonds, and those bonds break in the reducing condition. It is known that hBD-3 can interact with bacterial membrane, and even eukaryotic cell membrane, which has a low concentration of phosphatidylinositol 4,5-bisphosphate (PIP2) lipids. PIP2 is a vital component in cell membranes and has been found to play important roles during antimicrobial peptide (AMP) interaction with membranes. To understand the functional mechanism of hBD-3 interacting with PIP2-containing membranes, the binding structures of hBD-3 on 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) bilayers mixed with 10% of PIP2 were predicted using two kinds of methods. The first one is by placing the hBD-3 monomer in different orientations above the POPC + 10%PIP2 membrane to set up five different initial simulation systems and performing long-term simulations on each to predict the most stable binding structure. It was found that hBD-3 analogue binds on the mixed lipid membrane on the two loop regions. The second method is by running long-term simulations on one or nine hBD-3 dimers binding on POPC mixed with 10%PIP2 lipid bilayer starting from the solid-state NMR (ssNMR)-suggested orientation. The dimer dissociated, and the most stable binding of hBD-3 in wild-type on the mixed membrane is also through the two loop regions, which agrees with the prediction from both the first method and the lipid self-assembly result. The PIP2 lipids can form long-lasting hydrogen bonds with positively charged residues such as Arg and Lys on hBD-3, thus forming clusters with hBD-3. As a comparison, hBD-3 dimers binding with a combined bilayer having 1,2-palmitoyl-oleoyl-sn-glycero-3-phosphoserine (POPS) on the upper and POPC on the lower leaflets and the combined POPS + POPC bilayer mixing with 10%PIP2 were also studied. The long-term simulation result shows that hBD-3 can bind with the heads of negatively charged POPS and PIP2 lipids and form hydrogen bonds. The stable binding sites of hBD-3 on PIP2 or POPS mixed bilayers are still on the two loop regions. On the combined POPS + POPC mixed with 10%PIP2 bilayer, the binding of hBD-3 with PIP2 lipids became less stable and fewer because of the competition of binding with the POPS lipids. Besides that, binding with hBD-3 can decrease the membrane thickness of the POPC + PIP2, POPS + POPC, and POPS + POPC + PIP2 bilayers and make POPS and PIP2 lipids more flexible based on the order parameter calculations. Our results supply molecular insight on AMP binding with different membranes and can help understand the functional mechanism of hBD-3 disrupting PIP2-containing membranes.
Collapse
Affiliation(s)
- Liqun Zhang
- Department of Chemical Engineering, Tennessee Technological University, Cookeville, Tennessee 38505, United States
| |
Collapse
|
262
|
Yadav V, Misra R. A review emphasizing on utility of heptad repeat sequence as a tool to design pharmacologically safe peptide-based antibiotics. Biochimie 2021; 191:126-139. [PMID: 34492334 DOI: 10.1016/j.biochi.2021.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/25/2021] [Accepted: 09/03/2021] [Indexed: 12/31/2022]
Abstract
Extensive usage of antibiotics has created an unprecedented scenario of the rapid emergence of many drug-resistant bacteria, which has become an alarming public health concern around the globe. Search for better alternatives that are as efficacious as antibiotics led to the discovery of antimicrobial peptides (AMPs). These small cationic amphiphilic peptides have emerged as a promising option as antimicrobial agents, owing to their multifaceted implications against varied pathogens. Recent years have witnessed tremendous growth in research on AMPs resulting in them being tested in clinical trials of which six got approved for topical application. The relatively less successful outcome has been attributed to the poor cell selectivity shown by most of the naturally occurring AMPs. This drawback needs to be circumvented by identifying strategies to design safe and effective peptides. In the present review, we have emphasized the importance of heptad repeat sequence (leucine and/or phenylalanine zipper motif) as a tool that has shown great promise in remodeling the toxic AMPs to safe antimicrobial agents.
Collapse
Affiliation(s)
- Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden; Interdisciplinary Cluster for Applied Genoproteomics (GIGA), University of Liège (ULiège), Liège, Belgium.
| | - Richa Misra
- Department of Zoology, Sri Venkateswara College, University of Delhi, Delhi, India
| |
Collapse
|
263
|
Vishweshwaraiah YL, Acharya A, Hegde V, Prakash B. Rational design of hyperstable antibacterial peptides for food preservation. NPJ Sci Food 2021; 5:26. [PMID: 34471114 PMCID: PMC8410836 DOI: 10.1038/s41538-021-00109-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023] Open
Abstract
We describe the design of peptides with properties like thermostability, pH stability, and antibacterial activity against a few bacterial food pathogens. Insights obtained from classical structure-function analysis of natural peptides and their mutants through antimicrobial and enzymatic assays are used to rationally develop a set of peptides. pH and thermostability assays were performed to demonstrate robust antimicrobial activity post-treatment with high temperatures and at wide pH ranges. We have also investigated the mode of action of these hyperstable peptides using membrane permeability assays, electron microscopy, and molecular dynamics simulations. Notably, through mutational studies, we show that these peptides elicit their antibacterial action via both membrane destabilization and inhibition of intracellular trypsin-the two functions attributable to separate peptide segments. Finally, toxicity studies and food preservation assays demonstrate the safety and efficacy of the designed peptides for food preservation. Overall, the study provides a general 'blueprint' for the development of stable antimicrobial peptides (AMPs). Insights obtained from this work may also be combined with combinatorial methods in high-throughput studies for future development of antimicrobials for various applications.
Collapse
Affiliation(s)
- Yashavantha L. Vishweshwaraiah
- grid.417629.f0000 0004 0501 5711Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, India
| | - Abhishek Acharya
- grid.417629.f0000 0004 0501 5711Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, India
| | - Vinayak Hegde
- grid.417629.f0000 0004 0501 5711Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh India
| | - Balaji Prakash
- grid.417629.f0000 0004 0501 5711Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, India ,grid.448607.90000 0004 1781 3606Division of Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Ahmedabad, Gujarat India
| |
Collapse
|
264
|
The Emergence of Multidrug-Resistant Helicobacter pylori in Southeast Asia: A Systematic Review on the Trends and Intervention Strategies Using Antimicrobial Peptides. Antibiotics (Basel) 2021; 10:antibiotics10091061. [PMID: 34572643 PMCID: PMC8465560 DOI: 10.3390/antibiotics10091061] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
The emergence of multidrug-resistant H. pylori poses a public healthcare threat, particularly in low- and middle-income countries. Recently, the World Health Organization has classified clarithromycin-resistant H. pylori as high priority in the research and discovery of novel antibiotics. This study was aimed to systematically review the prevalence of primary antibiotic resistance in H. pylori in Southeast Asian countries (SEAC) and to review current studies of antimicrobial peptides against H. pylori. We systematically searched through electronic databases of studies conducted on antimicrobial resistance of H. pylori in SEA countries. Furthermore, we searched articles that conducted studies on antimicrobial peptides, naturally occurring host’s defense molecules, against H. pylori. After a series of screening processes, 15 studies were included in our systematic review. Our analysis revealed that primary resistance of H. pylori to metronidazole, clarithromycin, and levofloxacin were high in SEAC, although the primary resistance to amoxicillin and tetracycline remains low. Multidrug-resistant H. pylori are emerging in SE Asian countries. The antimicrobial peptides show promising antibacterial and antibiofilm activity against drug-resistant H. pylori. The research and discovery of antimicrobial peptides against H. pylori in SEAC will help in limiting the spread of antimicrobial resistance of H. pylori.
Collapse
|
265
|
He S, Stone TA, Deber CM. Uncoupling Amphipathicity and Hydrophobicity: Role of Charge Clustering in Membrane Interactions of Cationic Antimicrobial Peptides. Biochemistry 2021; 60:2586-2592. [PMID: 34423969 DOI: 10.1021/acs.biochem.1c00367] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Peptides with a combination of high positive charge and high hydrophobicity have high antimicrobial activity, as epitomized by peptide venoms, which are designed by nature as disruptors of host membranes yet also display significant efficacy against pathogens. To investigate this phenomenon systematically, here we focus on ponericin W1, a peptide venom isolated from Pachycondyla goeldii ants (WLGSALKIGAKLLPSVVGLFKKKKQ) to examine whether Lys positioning can be broadly applied to optimize the functional range of existing natural sequences. We prepared sets of ponericin W1 analogues, where Lys residues were either distributed in an amphipathic manner throughout the sequence (PonAmp), clustered at the N-terminus (PonN), or clustered at the C-terminus (PonC), along with their counterparts of reduced hydrophobicity through 2-4 Leu-to-Ala replacements. We found that wild-type ponericin W1 and all three variants displayed toxicity against human erythrocytes, but hemolysis was eliminated by the replacement of two or more Leu residues by Ala residues. As well, peptides containing up to 3 Leu-to-Ala replacements retained antimicrobial activity against E. coli bacteria. Biophysical analyses of peptide-membrane interaction patterns by circular dichroism spectroscopy revealed a novel mode of cluster-dependent peptide positioning vis-à-vis the water-membrane interface, where PonAmp and PonC peptides displayed full or partial helical structures, while PonN peptides were unstructured, likely due, in part, to dynamic interchange between aqueous and membrane surface environments. The overall findings suggest that the lower membrane penetration of N-terminal charge-clustered constructs coupled with moderate sequence hydrophobicity may be advantageous for conferring enhanced target selectivity for bacterial versus mammalian membranes.
Collapse
Affiliation(s)
- Shelley He
- Program in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tracy A Stone
- Program in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Charles M Deber
- Program in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
266
|
Khosravimelal S, Chizari M, Farhadihosseinabadi B, Moosazadeh Moghaddam M, Gholipourmalekabadi M. Fabrication and characterization of an antibacterial chitosan/silk fibroin electrospun nanofiber loaded with a cationic peptide for wound-dressing application. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:114. [PMID: 34455501 PMCID: PMC8403119 DOI: 10.1007/s10856-021-06542-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/30/2021] [Indexed: 05/03/2023]
Abstract
Wound infections are still problematic in many cases and demand new alternatives for current treatment strategies. In recent years, biomaterials-based wound dressings have received much attention due to their potentials and many studies have been performed based on them. Accordingly, in this study, we fabricated and optimized an antibacterial chitosan/silk fibroin (CS/SF) electrospun nanofiber bilayer containing different concentrations of a cationic antimicrobial peptide (AMP) for wound dressing applications. The fabricated CS/SF nanofiber was fully characterized and compared to the electrospun silk fibroin and electrospun chitosan alone in vitro. Then, the release rate of different concentrations of peptide (16, 32, and 64 µg/ml) from peptide-loaded CS/SF nanofiber was investigated. Finally, based on cytotoxic activity, the antibacterial activity of scaffolds containing 16 and 32 µg/ml of the peptide was evaluated against standard and multi-drug resistant strains of Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa isolated from burn patients. The peptide-loaded CS/SF nanofiber displayed appropriate mechanical properties, high water uptake, suitable biodegradation rate, a controlled release without cytotoxicity on Hu02 human foreskin fibroblast cells at the 16 and 32 µg/ml concentrations of peptide. The optimized CS/SF containing 32 μg/ml peptide showed strong antibacterial activity against all experimental strains from standard to resistance. The results showed that the fabricated antimicrobial nanofiber has the potential to be applied as a wound dressing for infected wound healing, although further studies are needed in vivo.
Collapse
Affiliation(s)
- Sadjad Khosravimelal
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Milad Chizari
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | | - Mazaher Gholipourmalekabadi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
267
|
Yang W, Yoon Y, Lee Y, Oh H, Choi J, Shin S, Lee S, Lee H, Lee Y, Seo J. Photosensitizer-peptoid conjugates for photoinactivation of Gram-negative bacteria: structure-activity relationship and mechanistic studies. Org Biomol Chem 2021; 19:6546-6557. [PMID: 34259297 DOI: 10.1039/d1ob00926e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Multitarget engagement is considered an effective strategy to overcome the threat of bacterial infection, and antimicrobials with multiple mechanisms of action have been successful as natural chemical weaponry. Here, we synthesized a library of photosensitizer-peptoid conjugates (PsPCs) as novel antimicrobial photodynamic therapy (aPDT) agents. The peptoids, linkers, and photosensitizers were varied, and their structure-antimicrobial activity relationships against Escherichia coli were evaluated; PsPC 9 was indicated to be the most promising photoresponsive antimicrobial agent among the synthesized PsPCs. Spectroscopic analyses indicated that 9 generated singlet oxygen upon absorption of visible light (420 nm) while maintaining the weakly helical conformation of the peptoid. Mechanistic studies suggested that damage to the bacterial membrane and cleavage of DNA upon light irradiation were the main causes of bactericidal activity, which was supported by flow cytometry and DNA gel electrophoresis experiments. We demonstrated that the optimal combination of membrane-active peptoids and photosensitizers can generate an efficient aPDT agent that targets multiple sites of bacterial components and kills bacteria by membrane disruption and reactive oxygen species generation.
Collapse
Affiliation(s)
- Woojin Yang
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Younggun Yoon
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Yunjee Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Hyeongyeol Oh
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Jieun Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Sujin Shin
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), 49 Dosicheomdansaneopro, Nam-gu, Gwangju 61751, South Korea
| | - Hohjai Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Yunho Lee
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Jiwon Seo
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| |
Collapse
|
268
|
Askari P, Namaei MH, Ghazvini K, Hosseini M. In vitro and in vivo toxicity and antibacterial efficacy of melittin against clinical extensively drug-resistant bacteria. BMC Pharmacol Toxicol 2021; 22:42. [PMID: 34261542 PMCID: PMC8281584 DOI: 10.1186/s40360-021-00503-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/21/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Melittin is one of the most studied antimicrobial peptides, and several in vitro experiments have demonstrated its antibacterial efficacy. However, there is evidence showing melittin has non-promising effects such as cytotoxicity and hemolysis. Therefore, concerns about unwanted collateral toxicity of melittin lie ahead in the path toward its clinical development. With these considerations, the present study aimed to fill the gap between in vitro and in vivo studies. METHODS In the first step, in vitro toxicity profile of melittin was assessed using cytotoxicity and hemolysis tests. Next, a maximum intraperitoneal (i.p.) sub-lethal dose was determined using BALB/c mice. Besides toxicity, antimicrobial efficacy of melittin against extensively drug-resistant (XDR) Acinetobacter baumannii, methicillin-resistant Staphylococcus aureus (MRSA), and KPC-producing Klebsiella pneumonia (KPC-KP) pathogens were tested using both in vitro and in vivo methods. RESULTS Melittin showed extensive hemolysis (HD50 = 0.44 µg/mL), and cytotoxicity (IC50 = 6.45 µg/mL) activities with i.p. LD50 value of 4.98 mg/kg in BALB/c mice. In vitro antimicrobial evaluation showed melittin MIC range from 8 to 32 µg/mL for the studied pathogens. Treatment of infected mice with repeated sub-lethal doses of melittin (2.4 mg/kg) displayed no beneficial effect on their survival and peritoneal bacterial loads. CONCLUSIONS These results indicate that melittin at its safe dose could not exhibit antimicrobial activity, which hinders its application in clinical practice.
Collapse
Affiliation(s)
- Parvin Askari
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Hasan Namaei
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Mashhad, Iran
| | - Kiarash Ghazvini
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mehran Hosseini
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
269
|
Gan BH, Gaynord J, Rowe SM, Deingruber T, Spring DR. The multifaceted nature of antimicrobial peptides: current synthetic chemistry approaches and future directions. Chem Soc Rev 2021; 50:7820-7880. [PMID: 34042120 PMCID: PMC8689412 DOI: 10.1039/d0cs00729c] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 12/13/2022]
Abstract
Bacterial infections caused by 'superbugs' are increasing globally, and conventional antibiotics are becoming less effective against these bacteria, such that we risk entering a post-antibiotic era. In recent years, antimicrobial peptides (AMPs) have gained significant attention for their clinical potential as a new class of antibiotics to combat antimicrobial resistance. In this review, we discuss several facets of AMPs including their diversity, physicochemical properties, mechanisms of action, and effects of environmental factors on these features. This review outlines various chemical synthetic strategies that have been applied to develop novel AMPs, including chemical modifications of existing peptides, semi-synthesis, and computer-aided design. We will also highlight novel AMP structures, including hybrids, antimicrobial dendrimers and polypeptides, peptidomimetics, and AMP-drug conjugates and consider recent developments in their chemical synthesis.
Collapse
Affiliation(s)
- Bee Ha Gan
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Josephine Gaynord
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Sam M Rowe
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Tomas Deingruber
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - David R Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| |
Collapse
|
270
|
Lima WG, Brito JCM, de Lima ME, Pizarro ACST, Vianna MAMDM, de Paiva MC, de Assis DCS, Cardoso VN, Fernandes SOA. A short synthetic peptide, based on LyeTx I from Lycosa erythrognatha venom, shows potential to treat pneumonia caused by carbapenem-resistant Acinetobacter baumannii without detectable resistance. J Antibiot (Tokyo) 2021; 74:425-434. [PMID: 33972716 DOI: 10.1038/s41429-021-00421-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/02/2021] [Accepted: 04/02/2021] [Indexed: 11/09/2022]
Abstract
The emergence of antibiotic-resistant bacteria, especially carbapenem-resistant Acinetobacter baumannii (CRAB), together with relative stagnation in the development of effective antibiotics, has led to enormous health and economic problems. In this study, we aimed to describe the antibacterial spectrum of LyeTx I mnΔK, a short synthetic peptide based on LyeTx I from Lycosa erythrognatha venom, against CRAB. LyeTx I mnΔK showed considerable antibacterial activity against extensively resistant A. baumannii, with minimum inhibitory and bactericidal concentrations ranging from 1 to 16 µM and 2 to 32 µM, respectively. This peptide significantly increased the release of 260 nm-absorbing intracellular material from CRAB, suggesting bacteriolysis. LyeTx I mnΔK was shown to act synergistically with meropenem and colistin against CRAB. The cytotoxic concentration of LyeTx I mnΔK against Vero cells (CC50 = 55.31 ± 5.00 µM) and its hemolytic activity (HC50 = 77.07 ± 4.00 µM) were considerably low; however, its antibacterial activity was significantly reduced in the presence of human and animal serum and trypsin. Nevertheless, the inhalation of this peptide was effective in reducing pulmonary bacterial load in a mouse model of CRAB infection. Altogether, these results demonstrate that the peptide LyeTx I mnΔK is a potential prototype for the development of new effective and safe antibacterial agents against CRAB.
Collapse
Affiliation(s)
- William Gustavo Lima
- Laboratório de Radioisótopos, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | | | - Maria Elena de Lima
- Faculdade Santa Casa de Belo Horizonte, Programa de Pós-Graduação Stricto Senso de Medicina e Biomedicina, Belo Horizonte, MG, Brazil
| | | | | | - Magna Cristina de Paiva
- Laboratório de Diagnóstico Laboratorial e Microbiologia Clínica, Universidade Federal de São João del-Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | | | - Valbert Nascimento Cardoso
- Laboratório de Radioisótopos, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Simone Odília Antunes Fernandes
- Laboratório de Radioisótopos, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
271
|
Frederiksen N, Louka S, Mudaliar C, Domraceva I, Kreicberga A, Pugovics O, Żabicka D, Tomczak M, Wygoda W, Björkling F, Franzyk H. Peptide/β-Peptoid Hybrids with Ultrashort PEG-Like Moieties: Effects on Hydrophobicity, Antibacterial Activity and Hemolytic Properties. Int J Mol Sci 2021; 22:ijms22137041. [PMID: 34208826 PMCID: PMC8268887 DOI: 10.3390/ijms22137041] [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: 06/01/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/01/2022] Open
Abstract
PEGylation of antimicrobial peptides as a shielding tool that increases stability toward proteolytic degradation typically leads to concomitant loss of activity, whereas incorporation of ultrashort PEG-like amino acids (sPEGs) remains essentially unexplored. Here, modification of a peptide/β-peptoid hybrid with sPEGs was examined with respect to influence on hydrophobicity, antibacterial activity and effect on viability of mammalian cells for a set of 18 oligomers. Intriguingly, the degree of sPEG modification did not significantly affect hydrophobicity as measured by retention in reverse-phase HPLC. Antibacterial activity against both wild-type and drug-resistant strains of Escherichia coli and Acinetobacter baumannii (both Gram-negative pathogens) was retained or slightly improved (MICs in the range 2–16 µg/mL equal to 0.7–5.2 µM). All compounds in the series exhibited less than 10% hemolysis at 400 µg/mL. While the number of sPEG moieties appeared not to be clearly correlated with hemolytic activity, a trend toward slightly increased hemolytic activity was observed for analogues displaying the longest sPEGs. In contrast, within a subseries the viability of HepG2 liver cells was least affected by analogues displaying the longer sPEGs (with IC50 values of ~1280 µg/mL) as compared to most other analogues and the parent peptidomimetic (IC50 values in the range 330–800 µg/mL).
Collapse
Affiliation(s)
- Nicki Frederiksen
- Center for Peptide-Based Antibiotics, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark; (N.F.); (S.L.); (C.M.); (F.B.)
| | - Stavroula Louka
- Center for Peptide-Based Antibiotics, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark; (N.F.); (S.L.); (C.M.); (F.B.)
| | - Chirag Mudaliar
- Center for Peptide-Based Antibiotics, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark; (N.F.); (S.L.); (C.M.); (F.B.)
| | - Ilona Domraceva
- Latvian Institute of Organic Synthesis, Aizkraukles 21, 1006 Riga, Latvia; (I.D.); (A.K.); (O.P.)
| | - Agrita Kreicberga
- Latvian Institute of Organic Synthesis, Aizkraukles 21, 1006 Riga, Latvia; (I.D.); (A.K.); (O.P.)
| | - Osvalds Pugovics
- Latvian Institute of Organic Synthesis, Aizkraukles 21, 1006 Riga, Latvia; (I.D.); (A.K.); (O.P.)
| | - Dorota Żabicka
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, ul. Chełmska 30/34, 00-725 Warsaw, Poland; (D.Ż.); (M.T.); (W.W.)
| | - Magdalena Tomczak
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, ul. Chełmska 30/34, 00-725 Warsaw, Poland; (D.Ż.); (M.T.); (W.W.)
| | - Weronika Wygoda
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, ul. Chełmska 30/34, 00-725 Warsaw, Poland; (D.Ż.); (M.T.); (W.W.)
| | - Fredrik Björkling
- Center for Peptide-Based Antibiotics, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark; (N.F.); (S.L.); (C.M.); (F.B.)
| | - Henrik Franzyk
- Center for Peptide-Based Antibiotics, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark; (N.F.); (S.L.); (C.M.); (F.B.)
- Correspondence:
| |
Collapse
|
272
|
Chai TT, Ee KY, Kumar DT, Manan FA, Wong FC. Plant Bioactive Peptides: Current Status and Prospects Towards Use on Human Health. Protein Pept Lett 2021; 28:623-642. [PMID: 33319654 DOI: 10.2174/0929866527999201211195936] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/28/2022]
Abstract
Large numbers of bioactive peptides with potential applications in protecting against human diseases have been identified from plant sources. In this review, we summarized recent progress in the research of plant-derived bioactive peptides, encompassing their production, biological effects, and mechanisms. This review focuses on antioxidant, antimicrobial, antidiabetic, and anticancer peptides, giving special attention to evidence derived from cellular and animal models. Studies investigating peptides with known sequences and well-characterized peptidic fractions or protein hydrolysates will be discussed. The use of molecular docking tools to elucidate inter-molecular interactions between bioactive peptides and target proteins is highlighted. In conclusion, the accumulating evidence from in silico, in vitro and in vivo studies to date supports the envisioned applications of plant peptides as natural antioxidants as well as health-promoting agents. Notwithstanding, much work is still required before the envisioned applications of plant peptides can be realized. To this end, future researches for addressing current gaps were proposed.
Collapse
Affiliation(s)
- Tsun-Thai Chai
- Department of Chemical Science, Faculty of Science, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
| | - Kah-Yaw Ee
- Center for Biodiversity Research, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
| | - D Thirumal Kumar
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602 105, India
| | - Fazilah Abd Manan
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Skudai 81310, Johor, Malaysia
| | - Fai-Chu Wong
- Department of Chemical Science, Faculty of Science, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
| |
Collapse
|
273
|
Kohut G, Juhász T, Quemé-Peña M, Bősze SE, Beke-Somfai T. Controlling Peptide Function by Directed Assembly Formation: Mechanistic Insights Using Multiscale Modeling on an Antimicrobial Peptide-Drug-Membrane System. ACS OMEGA 2021; 6:15756-15769. [PMID: 34179620 PMCID: PMC8223213 DOI: 10.1021/acsomega.1c01114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/01/2021] [Indexed: 05/16/2023]
Abstract
Owing to their potential applicability against multidrug-resistant bacteria, antimicrobial peptides (AMPs) or host defense peptides (HDPs) gain increased attention. Besides diverse immunomodulatory roles, their classical mechanism of action mostly involves membrane disruption of microbes. Notably, their unbalanced overexpression has also been associated with host cell cytotoxicity in various diseases. Relatedly, AMPs can be subject to aggregate formation, either via self-assembly or together with other compounds, which has demonstrated a modulation effect on their biological functions, thus highly relevant both for drug targeting projects and understanding their in vivo actions. However, the molecular aspects of the related assembly formation are not understood. Here, we focused in detail on an experimentally studied AMP-drug system, i.e., CM15-suramin, and performed all-atom and coarse-grain (CG) simulations. Results obtained for all systems were in close line with experimental observations and indicate that the CM15-suramin aggregation is an energetically favorable and dynamic process. In the presence of bilayers, the peptide-drug assembly formation was highly dependent on lipid composition, and peptide aggregates themselves were also capable of binding to the membranes. Interestingly, longer CG simulations with zwitterionic membranes indicated an intermediate state in the presence of both AMP-drug assemblies and monomeric peptides located on the membrane surface. In sharp contrast, larger AMP-drug aggregates could not be detected with a negatively charged membrane, rather the AMPs penetrated its surface in a monomeric form, in line with previous in vitro observations. Considering experimental and theoretical results, it is promoted that in biological systems, cationic AMPs may often form associates with anionic compounds in a reversible manner, resulting in lower bioactivity. This is only mildly affected by zwitterionic membranes; however, membranes with a negative charge strongly alter the energetic preference of AMP assemblies, resulting in the dissolution of the complexes into the membrane. The phenomenon observed here at a molecular level can be followed in several experimental systems studied recently, where peptides interact with food colors, drug molecules, or endogenous compounds, which strongly indicates that reversible associate formation is a general phenomenon for these complexes. These results are hoped to be exploited in novel therapeutic strategies aiming to use peptides as drug targets and control AMP bioactivity by directed assembly formation.
Collapse
Affiliation(s)
- Gergely Kohut
- Institute
of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
- Hevesy
György PhD School of Chemistry, ELTE
Eötvös Loránd University, Pázmány Péter sétány
1/A, H-1117 Budapest, Hungary
| | - Tünde Juhász
- Institute
of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Mayra Quemé-Peña
- Institute
of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
- Hevesy
György PhD School of Chemistry, ELTE
Eötvös Loránd University, Pázmány Péter sétány
1/A, H-1117 Budapest, Hungary
| | - Szilvia Erika Bősze
- ELKH
Research Group of Peptide Chemistry, Eötvös
Loránd University, Pázmány Péter sétány 1/A, H-1117 Budapest, Hungary
| | - Tamás Beke-Somfai
- Institute
of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| |
Collapse
|
274
|
Marrazzo P, Pizzuti V, Zia S, Sargenti A, Gazzola D, Roda B, Bonsi L, Alviano F. Microfluidic Tools for Enhanced Characterization of Therapeutic Stem Cells and Prediction of Their Potential Antimicrobial Secretome. Antibiotics (Basel) 2021; 10:750. [PMID: 34206190 PMCID: PMC8300685 DOI: 10.3390/antibiotics10070750] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Antibiotic resistance is creating enormous attention on the development of new antibiotic-free therapy strategies for bacterial diseases. Mesenchymal stromal stem cells (MSCs) are the most promising candidates in current clinical trials and included in several cell-therapy protocols. Together with the well-known immunomodulatory and regenerative potential of the MSC secretome, these cells have shown direct and indirect anti-bacterial effects. However, the low reproducibility and standardization of MSCs from different sources are the current limitations prior to the purification of cell-free secreted antimicrobial peptides and exosomes. In order to improve MSC characterization, novel label-free functional tests, evaluating the biophysical properties of the cells, will be advantageous for their cell profiling, population sorting, and quality control. We discuss the potential of emerging microfluidic technologies providing new insights into density, shape, and size of live cells, starting from heterogeneous or 3D cultured samples. The prospective application of these technologies to studying MSC populations may contribute to developing new biopharmaceutical strategies with a view to naturally overcoming bacterial defense mechanisms.
Collapse
Affiliation(s)
- Pasquale Marrazzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.P.); (L.B.); (F.A.)
| | - Valeria Pizzuti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.P.); (L.B.); (F.A.)
| | - Silvia Zia
- Stem Sel S.r.l., 40127 Bologna, Italy; (S.Z.); (B.R.)
| | | | - Daniele Gazzola
- Cell Dynamics i.S.r.l., 40129 Bologna, Italy; (A.S.); (D.G.)
| | - Barbara Roda
- Stem Sel S.r.l., 40127 Bologna, Italy; (S.Z.); (B.R.)
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy
| | - Laura Bonsi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.P.); (L.B.); (F.A.)
| | - Francesco Alviano
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.P.); (L.B.); (F.A.)
| |
Collapse
|
275
|
Schaefer S, Pham TTP, Brunke S, Hube B, Jung K, Lenardon MD, Boyer C. Rational Design of an Antifungal Polyacrylamide Library with Reduced Host-Cell Toxicity. ACS APPLIED MATERIALS & INTERFACES 2021; 13:27430-27444. [PMID: 34060800 DOI: 10.1021/acsami.1c05020] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Life-threatening invasive fungal infections represent an urgent threat to human health worldwide. The limited set of antifungal drugs has critical constraints such as resistance development and/or adverse side effects. One approach to overcome these limitations is to mimic naturally occurring antifungal peptides called defensins. Inspired by their advantageous amphiphilic properties, a library of 35 synthetic, linear, ternary polyacrylamides was prepared by controlled/living radical polymerization. The effect of the degree of polymerization (20, 40, and 100) and varying hydrophobic functionalities (branched, linear, cyclic, or aromatic differing in their number of carbons) on their antifungal activity was investigated. Short copolymers with a calculated log P of ∼1.5 revealed optimal activity against the major human fungal pathogen Candida albicans and other pathogenic fungal species with limited toxicity to mammalian host cells (red blood cells and fibroblasts). Remarkably, selected copolymers outperformed the commercial antifungal drug amphotericin B, with respect to the therapeutic index, highlighting their potential as novel antifungal compounds.
Collapse
Affiliation(s)
- Sebastian Schaefer
- School of Chemical Engineering, UNSW, Sydney, New South Wales 2052, Australia
- Australian Centre for Nanomedicine, UNSW, Sydney, New South Wales 2052, Australia
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, 07745 Jena, Germany
| | - Thi Thu Phuong Pham
- School of Chemical Engineering, UNSW, Sydney, New South Wales 2052, Australia
- Australian Centre for Nanomedicine, UNSW, Sydney, New South Wales 2052, Australia
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, 07745 Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| | - Kenward Jung
- School of Chemical Engineering, UNSW, Sydney, New South Wales 2052, Australia
- Australian Centre for Nanomedicine, UNSW, Sydney, New South Wales 2052, Australia
| | - Megan Denise Lenardon
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, New South Wales 2052, Australia
| | - Cyrille Boyer
- School of Chemical Engineering, UNSW, Sydney, New South Wales 2052, Australia
- Australian Centre for Nanomedicine, UNSW, Sydney, New South Wales 2052, Australia
| |
Collapse
|
276
|
Kumar V, Sinha N, Thakur AK. Necessity of regulatory guidelines for the development of amyloid based biomaterials. Biomater Sci 2021; 9:4410-4422. [PMID: 34018497 DOI: 10.1039/d1bm00059d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Amyloid diseases are caused due to protein homeostasis failure where incorrectly folded proteins/peptides form cross-β-sheet rich amyloid fibrillar structures. Besides proteins/peptides, small metabolite assemblies also exhibit amyloid-like features. These structures are linked to several human and animal diseases. In addition, non-toxic amyloids with diverse physiological roles are characterized as a new functional class. This finding, along with the unique properties of amyloid like stability and mechanical strength, led to a surge in the development of amyloid-based biomaterials. However, the usage of these materials by humans and animals may pose a health risk such as the development of amyloid diseases and toxicity. This is possible because amyloid-based biomaterials and their fragments may assist seeding and cross-seeding mechanisms of amyloid formation in the body. This review summarizes the potential uses of amyloids as biomaterials, the concerns regarding their usage, and a prescribed workflow to initiate a regulatory approach.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nabodita Sinha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, UP-208016, India.
| | - Ashwani Kumar Thakur
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, UP-208016, India.
| |
Collapse
|
277
|
Di Somma A, Canè C, Moretta A, Duilio A. Interaction of Temporin-L Analogues with the E. coli FtsZ Protein. Antibiotics (Basel) 2021; 10:antibiotics10060704. [PMID: 34208230 PMCID: PMC8230800 DOI: 10.3390/antibiotics10060704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 11/16/2022] Open
Abstract
The research of new therapeutic agents to fight bacterial infections has recently focused on the investigation of antimicrobial peptides (AMPs), the most common weapon that all organisms produce to prevent invasion by external pathogens. Among AMPs, the amphibian Temporins constitute a well-known family with high antibacterial properties against Gram-positive and Gram-negative bacteria. In particular, Temporin-L was shown to affect bacterial cell division by inhibiting FtsZ, a tubulin-like protein involved in the crucial step of Z-ring formation at the beginning of the division process. As FtsZ represents a leading target for new antibacterial compounds, in this paper we investigated in detail the interaction of Temporin L with Escherichia coli FtsZ and designed two TL analogues in an attempt to increase peptide-protein interactions and to better understand the structural determinants leading to FtsZ inhibition. The results demonstrated that the TL analogues improved their binding to FtsZ, originating stable protein-peptide complexes. Functional studies showed that both peptides were endowed with a high capability of inhibiting both the enzymatic and polymerization activities of the protein. Moreover, the TL analogues were able to inhibit bacterial growth at low micromolar concentrations. These observations may open up the way to the development of novel peptide or peptidomimetic drugs tailored to bind FtsZ, hampering a crucial process of bacterial life that might be proposed for future pharmaceutical applications.
Collapse
Affiliation(s)
- Angela Di Somma
- Department of Chemical Sciences, Università Federico II di Napoli, 80126 Napoli, Italy; (A.D.S.); (C.C.)
| | - Carolina Canè
- Department of Chemical Sciences, Università Federico II di Napoli, 80126 Napoli, Italy; (A.D.S.); (C.C.)
| | - Antonio Moretta
- Departiment of Science, Università degli Studi della Basilicata, 85100 Potenza, Italy;
| | - Angela Duilio
- Department of Chemical Sciences, Università Federico II di Napoli, 80126 Napoli, Italy; (A.D.S.); (C.C.)
- Correspondence:
| |
Collapse
|
278
|
Wani NA, Ben Hur D, Kapach G, Stolovicki E, Rotem E, Shai Y. Switching Bond: Generation of New Antimicrobial Peptides via the Incorporation of an Intramolecular Isopeptide Bond. ACS Infect Dis 2021; 7:1702-1712. [PMID: 34043312 PMCID: PMC8634383 DOI: 10.1021/acsinfecdis.1c00037] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Antimicrobial peptides (AMPs), which
can be modified to kill a
broad spectrum of microoganisms or a specific microorganism, are considered
as promising alternatives to combat the rapidly widespread, resistant
bacterial infections. However, there are still several obstacles to
overcome. These include toxicity, stability, and the ability to interfere
with the immune response and bacterial resistance. To overcome these
challenges, we herein replaced the regular peptide bonds with isopeptide
bonds to produce new AMPs based on the well-known synthetic peptides
Amp1L and MSI-78 (pexiganan). Two new peptides Amp1EP and MSIEP were
generated while retaining properties such as size, sequence, charge,
and molecular weight. These new peptides have reduced toxicity toward
murine macrophage (RAW 264.7) cells, human monocytic (THP-1) cells,
and human red blood cells (hRBCs) and enhanced the stability toward
proteolytic degradation. Importantly, the new peptides do not repress
the pro-inflammatory cytokine and hence should not modulate the immune
response. Structurally, the new peptides, Amp1EP and MSIEP, have a
structure of random coils in contrast to the helical structures of
the parental peptides as revealed by circular dichroism (CD) analysis.
Their mode of action, assessed by flow cytometry, includes permeabilization
of the bacterial membrane. Overall, we present here a new approach
to modulate AMPs to develop antimicrobial peptides for future therapeutic
purposes.
Collapse
Affiliation(s)
- Naiem Ahmad Wani
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Daniel Ben Hur
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gal Kapach
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elad Stolovicki
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Etai Rotem
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yechiel Shai
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
279
|
Recent strategies for inhibiting multidrug-resistant and β-lactamase producing bacteria: A review. Colloids Surf B Biointerfaces 2021; 205:111901. [PMID: 34116398 DOI: 10.1016/j.colsurfb.2021.111901] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 12/26/2022]
Abstract
β-lactam antibiotics are one of the most commonly used drugs for treating bacterial infections, but their clinical effectiveness has been severely affected with bacteria developing resistance against their action. Production of β-lactamase enzymes by bacteria that can degrade β-lactams is the most common mechanism of acquiring such resistance, leading to the emergence of multiple-drug resistance in them. Therefore, the development of efficient approaches to combat infections caused by β-lactamase producing and multidrug-resistant bacteria is the need of the hour. The present review attempts to understand such recent strategies that are in line for development as potential alternatives to conventional antibiotics. We find that apart from efforts being made to develop new antibiotics, several other approaches are being explored, which can help tackle infections caused by resistant bacteria. This includes the development of plant-based drugs, antimicrobial peptides, nano-formulations, bacteriophage therapy, use of CRISPR-Cas9, RNA silencing and antibiotic conjugates with nanoparticles of antimicrobial peptides. The mechanism of action of these novel approaches and potential issues limiting their translation from laboratory to clinics is also discussed. The review is important from an interesting knowledge base which can be useful for researchers working in this domain.
Collapse
|
280
|
Hayashida PY, da Silva Junior PI. Insights into Antimicrobial Peptides from Limacus flavus Mucus. Curr Microbiol 2021; 78:2970-2979. [PMID: 34086076 DOI: 10.1007/s00284-021-02552-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/24/2021] [Indexed: 11/27/2022]
Abstract
Antimicrobial peptides have been a major research subject since the rise of antimicrobial resistance as a major public health problem. These molecules are considered a potential therapeutic source of antibiotics with broad-spectrum activity against microorganisms. Two antimicrobial peptides were isolated from the mucus of the Limacus flavus slug. The mucus was obtained by thermal shock, lyophilized and extracted with acetic acid. The supernatant was prefractionated in Sep-Pak and shortly thereafter fractionated by reverse-phase high-performance liquid chromatography. The manually obtained fractions were used in antimicrobial and cytotoxic assays and finally subjected to mass spectrometry (MS/MS). Characterization was performed by bioinformatics analysis with the tool Peaks®X + and by comparison with the NCBI and UniProt-SwissProt databases. Additionally, the physicochemical parameters of the samples were evaluated with online programs. Two fractions comtained antimicrobial peptides with the ability to inhibit Micrococcus luteus A270; both samples, LFMP-001 and LFMP-002, were hydrophilic molecules consisting of fewer than 20 residues. Comparison of the SDS-PAGE and Peaks®X + data showed that both had Mw < 3 kDa. In summary, this study presents data on the isolation and characterization of antimicrobial peptides from a slug and shows their potential against gram-positive bacteria.
Collapse
Affiliation(s)
- Patricia Yumi Hayashida
- Laboratory for Applied Toxinology (LETA) - Center of Toxins, Immune-Response and Cell Signaling - CeTICS/CEPID, Butantan Institute, São Paulo, Brazil
- Interunit Postgraduate Program in Biotechnology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Pedro Ismael da Silva Junior
- Laboratory for Applied Toxinology (LETA) - Center of Toxins, Immune-Response and Cell Signaling - CeTICS/CEPID, Butantan Institute, São Paulo, Brazil.
- Interunit Postgraduate Program in Biotechnology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
281
|
Bevalian P, Pashaei F, Akbari R, Pooshang Bagheri K. Eradication of vancomycin-resistant Staphylococcus aureus on a mouse model of third-degree burn infection by melittin: An antimicrobial peptide from bee venom. Toxicon 2021; 199:49-59. [PMID: 34087287 DOI: 10.1016/j.toxicon.2021.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/18/2021] [Accepted: 05/29/2021] [Indexed: 01/10/2023]
Abstract
Third-degree burn infections caused by antibiotic-resistant bacteria are of high clinical concern. Chemical antibiotics are not promising in eradication of bacterial infections. In this challenging condition, antimicrobial peptides (AMPs) are recently introduced as novel promising agents to overcome the issue. Accordingly, our study aimed to evaluate the efficiency of 'melittin' as natural peptide in bee venom, in eradicating vancomycin resistant Staphylococcus aureus (VRSA) on a mouse model of third-degree burn infection. In vitro pharmacological value of melittin was determined by examining its inhibitory and killing activities on VRSA isolates at different doses and time periods. The action mechanism of 'melittin' was evaluated by fluorescent release assay and Field Emission Scanning Electron Microscopy (FE-SEM) analyses. In vivo activity and toxicity of melittin were also examined on a mouse model of third-degree burn infection. The Minimum Inhibitory Concentration (MIC) and the Minimum Bactericidal Concentration (MBC) of melittin on all isolates ranged from '0.125-2 μg/mL' and '0.125-4 μg/mL', respectively. Rapid antibacterial activity of melittin on VRSA isolates was demonstrated by killing kinetics assays. Fluorometric and FE-SEM analyses indicated the membranolytic effects of melittin on VRSA isolates. The colonized VRSA bacteria were eradicated by melittin at 16 μg, in a single dose. No dermal toxicity and in vivo hemolysis were observed in the examined mice. The lack of in vivo toxicity of melittin along with its potent antibacterial activity indicated its promising therapeutic value as a topical drug against S. aureus associated third-degree burn infections.
Collapse
Affiliation(s)
- Parvaneh Bevalian
- Venom and Biotherapeutics Molecules Lab., Biotechnology Dept., Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Pashaei
- Venom and Biotherapeutics Molecules Lab., Biotechnology Dept., Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Akbari
- Venom and Biotherapeutics Molecules Lab., Biotechnology Dept., Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran; Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, West Azerbaijan, Iran
| | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Lab., Biotechnology Dept., Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
282
|
Látrová K, Havlová N, Večeřová R, Pinkas D, Bogdanová K, Kolář M, Fišer R, Konopásek I, Do Pham DD, Rejman D, Mikušová G. Outer membrane and phospholipid composition of the target membrane affect the antimicrobial potential of first- and second-generation lipophosphonoxins. Sci Rep 2021; 11:10446. [PMID: 34001940 PMCID: PMC8129119 DOI: 10.1038/s41598-021-89883-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/27/2021] [Indexed: 02/03/2023] Open
Abstract
Lipophosphonoxins (LPPOs) are small modular synthetic antibacterial compounds that target the cytoplasmic membrane. First-generation LPPOs (LPPO I) exhibit an antimicrobial activity against Gram-positive bacteria; however they do not exhibit any activity against Gram-negatives. Second-generation LPPOs (LPPO II) also exhibit broadened activity against Gram-negatives. We investigated the reasons behind this different susceptibility of bacteria to the two generations of LPPOs using model membranes and the living model bacteria Bacillus subtilis and Escherichia coli. We show that both generations of LPPOs form oligomeric conductive pores and permeabilize the bacterial membrane of sensitive cells. LPPO activity is not affected by the value of the target membrane potential, and thus they are also active against persister cells. The insensitivity of Gram-negative bacteria to LPPO I is probably caused by the barrier function of the outer membrane with LPS. LPPO I is almost incapable of overcoming the outer membrane in living cells, and the presence of LPS in liposomes substantially reduces their activity. Further, the antimicrobial activity of LPPO is also influenced by the phospholipid composition of the target membrane. A higher proportion of phospholipids with neutral charge such as phosphatidylethanolamine or phosphatidylcholine reduces the LPPO permeabilizing potential.
Collapse
Affiliation(s)
- Klára Látrová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Noemi Havlová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Renata Večeřová
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15, Olomouc, Czech Republic
| | - Dominik Pinkas
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Kateřina Bogdanová
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15, Olomouc, Czech Republic
| | - Milan Kolář
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15, Olomouc, Czech Republic
| | - Radovan Fišer
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Ivo Konopásek
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic
| | - Duy Dinh Do Pham
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10, Prague 6, Czech Republic
| | - Dominik Rejman
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10, Prague 6, Czech Republic.
| | - Gabriela Mikušová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 00, Prague 2, Czech Republic.
| |
Collapse
|
283
|
Scorpion Venom Antimicrobial Peptides Induce Siderophore Biosynthesis and Oxidative Stress Responses in Escherichia coli. mSphere 2021; 6:6/3/e00267-21. [PMID: 33980680 PMCID: PMC8125054 DOI: 10.1128/msphere.00267-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of life-threatening resistance of pathogenic bacteria to the antibiotics typically in use in hospitals and the community today has led to an urgent need to discover novel antimicrobial agents with different mechanisms of action. As an ancient host defense mechanism of the innate immune system, antimicrobial peptides (AMPs) are attractive candidates to fill that role. The increasing development of microbial resistance to classical antimicrobial agents has led to the search for novel antimicrobials. Antimicrobial peptides (AMPs) derived from scorpion and snake venoms offer an attractive source for the development of novel therapeutics. Smp24 (24 amino acids [aa]) and Smp43 (43 aa) are broad-spectrum AMPs that have been identified from the venom gland of the Egyptian scorpion Scorpio mauruspalmatus and subsequently characterized. Using a DNA microarray approach, we examined the transcriptomic responses of Escherichia coli to subinhibitory concentrations of Smp24 and Smp43 peptides following 5 h of incubation. Seventy-two genes were downregulated by Smp24, and 79 genes were downregulated by Smp43. Of these genes, 14 genes were downregulated in common and were associated with bacterial respiration. Fifty-two genes were specifically upregulated by Smp24. These genes were predominantly related to cation transport, particularly iron transport. Three diverse genes were independently upregulated by Smp43. Strains with knockouts of differentially regulated genes were screened to assess the effect on susceptibility to Smp peptides. Ten mutants in the knockout library had increased levels of resistance to Smp24. These genes were predominantly associated with cation transport and binding. Two mutants increased resistance to Smp43. There was no cross-resistance in mutants resistant to Smp24 or Smp43. Five mutants showed increased susceptibility to Smp24, and seven mutants showed increased susceptibility to Smp43. Of these mutants, formate dehydrogenase knockout (fdnG) resulted in increased susceptibility to both peptides. While the electrostatic association between pore-forming AMPs and bacterial membranes followed by integration of the peptide into the membrane is the initial starting point, it is clear that there are numerous subsequent additional intracellular mechanisms that contribute to their overall antimicrobial effect. IMPORTANCE The development of life-threatening resistance of pathogenic bacteria to the antibiotics typically in use in hospitals and the community today has led to an urgent need to discover novel antimicrobial agents with different mechanisms of action. As an ancient host defense mechanism of the innate immune system, antimicrobial peptides (AMPs) are attractive candidates to fill that role. Scorpion venoms have proven to be a rich source of AMPs. Smp24 and Smp43 are new AMPs that have been identified from the venom gland of the Egyptian scorpion Scorpio maurus palmatus, and these peptides can kill a wide range of bacterial pathogens. By better understanding how these AMPs affect bacterial cells, we can modify their structure to make better drugs in the future.
Collapse
|
284
|
Ghimire A, Song J. Anti-Periprosthetic Infection Strategies: From Implant Surface Topographical Engineering to Smart Drug-Releasing Coatings. ACS APPLIED MATERIALS & INTERFACES 2021; 13:20921-20937. [PMID: 33914499 PMCID: PMC8130912 DOI: 10.1021/acsami.1c01389] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Despite advanced implant sterilization and aseptic surgical techniques, periprosthetic bacterial infection remains a major challenge for orthopedic and dental implants. Bacterial colonization/biofilm formation around implants and their invasion into the dense skeletal tissue matrices are difficult to treat and could lead to implant failure and osteomyelitis. These complications require major revision surgeries and extended antibiotic therapies that are associated with high treatment cost, morbidity, and even mortality. Effective preventative measures mitigating risks for implant-related infections are thus in dire need. This review focuses on recent developments of anti-periprosthetic infection strategies aimed at either reducing bacterial adhesion, colonization, and biofilm formation or killing bacteria directly in contact with and/or in the vicinity of implants. These goals are accomplished through antifouling, quorum-sensing interfering, or bactericidal implant surface topographical engineering or surface coatings through chemical modifications. Surface topographical engineering of lotus leaf mimicking super-hydrophobic antifouling features and cicada wing-mimicking, bacterium-piercing nanopillars are both presented. Conventional physical coating/passive release of bactericidal agents is contrasted with their covalent tethering to implant surfaces through either stable linkages or linkages labile to bacterial enzyme cleavage or environmental perturbations. Pros and cons of these emerging anti-periprosthetic infection approaches are discussed in terms of their safety, efficacy, and translational potentials.
Collapse
Affiliation(s)
- Ananta Ghimire
- Department of Orthopedics and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jie Song
- Department of Orthopedics and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
285
|
Manniello MD, Moretta A, Salvia R, Scieuzo C, Lucchetti D, Vogel H, Sgambato A, Falabella P. Insect antimicrobial peptides: potential weapons to counteract the antibiotic resistance. Cell Mol Life Sci 2021; 78:4259-4282. [PMID: 33595669 PMCID: PMC8164593 DOI: 10.1007/s00018-021-03784-z] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/19/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Misuse and overuse of antibiotics have contributed in the last decades to a phenomenon known as antibiotic resistance which is currently considered one of the principal threats to global public health by the World Health Organization. The aim to find alternative drugs has been demonstrated as a real challenge. Thanks to their biodiversity, insects represent the largest class of organisms in the animal kingdom. The humoral immune response includes the production of antimicrobial peptides (AMPs) that are released into the insect hemolymph after microbial infection. In this review, we have focused on insect immune responses, particularly on AMP characteristics, their mechanism of action and applications, especially in the biomedical field. Furthermore, we discuss the Toll, Imd, and JAK-STAT pathways that activate genes encoding for the expression of AMPs. Moreover, we focused on strategies to improve insect peptides stability against proteolytic susceptibility such as D-amino acid substitutions, N-terminus modification, cyclization and dimerization.
Collapse
Affiliation(s)
- M D Manniello
- Department of Sciences, University of Basilicata, Via dell'Ateneo Lucano 10, 85100, Potenza, Italy
| | - A Moretta
- Department of Sciences, University of Basilicata, Via dell'Ateneo Lucano 10, 85100, Potenza, Italy
| | - R Salvia
- Department of Sciences, University of Basilicata, Via dell'Ateneo Lucano 10, 85100, Potenza, Italy
- Spinoff XFlies S.R.L, University of Basilicata, Via dell'Ateneo Lucano 10, 85100, Potenza, Italy
| | - C Scieuzo
- Department of Sciences, University of Basilicata, Via dell'Ateneo Lucano 10, 85100, Potenza, Italy
- Spinoff XFlies S.R.L, University of Basilicata, Via dell'Ateneo Lucano 10, 85100, Potenza, Italy
| | - D Lucchetti
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - H Vogel
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knöll-Straße 8, 07745, Jena, Germany
| | - A Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro di Riferimento Oncologico Della Basilicata (IRCCS-CROB), Rionero in Vulture (PZ), Italy
| | - P Falabella
- Department of Sciences, University of Basilicata, Via dell'Ateneo Lucano 10, 85100, Potenza, Italy.
- Spinoff XFlies S.R.L, University of Basilicata, Via dell'Ateneo Lucano 10, 85100, Potenza, Italy.
| |
Collapse
|
286
|
Yu T, Xianyu Y. Array-Based Biosensors for Bacteria Detection: From the Perspective of Recognition. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006230. [PMID: 33870615 DOI: 10.1002/smll.202006230] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/18/2020] [Indexed: 05/24/2023]
Abstract
Array-based biosensors have shown as effective and powerful tools to distinguish intricate mixtures with infinitesimal differences among analytes such as nucleic acids, proteins, microorganisms, and other biomolecules. In array-based bacterial sensing, the recognition of bacteria is the initial step that can crucially influence the analytical performance of a biosensor array. Bacteria recognition as well as the signal readout and mathematical analysis are indispensable to ensure the discrimination ability of array-based biosensors. Strategies for bacteria recognition mainly include the specific interaction between biomolecules and the corresponding receptors on bacteria, the noncovalent interaction between materials and bacteria, and the specific targeting of bacterial metabolites. In this review, recent advances in array-based bacteria sensors are discussed from the perspective of bacteria recognition relying on the characteristics of different bacteria. Principles of bacteria recognition and signal readout for bacteria detection are highlighted as well as the discussion on future trends in array-based biosensors.
Collapse
Affiliation(s)
- Ting Yu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Yunlei Xianyu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
- Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Ningbo Research Institute, Zhejiang University, Ningbo, Zhejiang, 315100, China
| |
Collapse
|
287
|
Cao F, Ma G, Song M, Zhu G, Mei L, Qin Q. Evaluating the effects of hydrophobic and cationic residues on antimicrobial peptide self-assembly. SOFT MATTER 2021; 17:4445-4451. [PMID: 33908584 DOI: 10.1039/d1sm00096a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Antimicrobial peptides typically contain hydrophobic and cationic residues, which allow them to interact with microbial cells and induce cell death. In a previous study, we found that the hydrophobic and cationic residues could also help antimicrobial peptides self-assemble into hydrogels, and this could be used as a novel approach for the preparation of hydrogel wound dressings. Therefore, in this work, four PAF26 peptide derivatives with different hydrophobic and cationic residues were used to study the effects of hydrophobic and cationic residues on self-assembly behaviours. It was found that all the PAF26 peptide derivatives could self-assemble into hydrogels, but the storage moduli, microscopic structures, secondary structure transformations, and antimicrobial abilities varied. In particular, peptides with a greater number of hydrophobic and cationic residues tended to undergo an unordered coil transformation and form bent nanofibers, while peptides with a lower number of hydrophobic and cationic residues tended to undergo β-sheet transformation and form straight nanofibers. In addition, antimicrobial experiments demonstrated that a strong crosslinked structure may have negative effects on the antimicrobial activity.
Collapse
Affiliation(s)
- Fengyi Cao
- School of Materials and Chemical Engineering, Zhongyuan University of Technology, Zhengzhou 450007, P. R. China.
| | - Gangqing Ma
- School of Materials and Chemical Engineering, Zhongyuan University of Technology, Zhengzhou 450007, P. R. China.
| | - Meng Song
- School of Materials and Chemical Engineering, Zhongyuan University of Technology, Zhengzhou 450007, P. R. China.
| | - Genxing Zhu
- School of Materials and Chemical Engineering, Zhongyuan University of Technology, Zhengzhou 450007, P. R. China.
| | - Lin Mei
- School of Materials and Chemical Engineering, Zhongyuan University of Technology, Zhengzhou 450007, P. R. China.
| | - Qi Qin
- School of Materials and Chemical Engineering, Zhongyuan University of Technology, Zhengzhou 450007, P. R. China.
| |
Collapse
|
288
|
Michelet R, Ursino M, Boulet S, Franck S, Casilag F, Baldry M, Rolff J, van Dyk M, Wicha SG, Sirard JC, Comets E, Zohar S, Kloft C. The Use of Translational Modelling and Simulation to Develop Immunomodulatory Therapy as an Adjunct to Antibiotic Treatment in the Context of Pneumonia. Pharmaceutics 2021; 13:601. [PMID: 33922017 PMCID: PMC8143524 DOI: 10.3390/pharmaceutics13050601] [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: 02/28/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 11/16/2022] Open
Abstract
The treatment of respiratory tract infections is threatened by the emergence of bacterial resistance. Immunomodulatory drugs, which enhance airway innate immune defenses, may improve therapeutic outcome. In this concept paper, we aim to highlight the utility of pharmacometrics and Bayesian inference in the development of immunomodulatory therapeutic agents as an adjunct to antibiotics in the context of pneumonia. For this, two case studies of translational modelling and simulation frameworks are introduced for these types of drugs up to clinical use. First, we evaluate the pharmacokinetic/pharmacodynamic relationship of an experimental combination of amoxicillin and a TLR4 agonist, monophosphoryl lipid A, by developing a pharmacometric model accounting for interaction and potential translation to humans. Capitalizing on this knowledge and associating clinical trial extrapolation and statistical modelling approaches, we then investigate the TLR5 agonist flagellin. The resulting workflow combines expert and prior knowledge on the compound with the in vitro and in vivo data generated during exploratory studies in order to construct high-dimensional models considering the pharmacokinetics and pharmacodynamics of the compound. This workflow can be used to refine preclinical experiments, estimate the best doses for human studies, and create an adaptive knowledge-based design for the next phases of clinical development.
Collapse
Affiliation(s)
- Robin Michelet
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
| | - Moreno Ursino
- Unit of Clinical Epidemiology, Assistance Publique-Hôpitaux de Paris, CHU Robert Debré, Université de Paris, Sorbonne Paris-Cité, Inserm U1123 and CIC-EC 1426, F-75019 Paris, France;
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
| | - Sandrine Boulet
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
- HeKA, Inria, F-75006 Paris, France
| | - Sebastian Franck
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Fiordiligie Casilag
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Mara Baldry
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Jens Rolff
- Department of Evolutionary Biology, Institute of Biology, Freie Universitaet Berlin, 14195 Berlin, Germany;
| | - Madelé van Dyk
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia;
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Jean-Claude Sirard
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Emmanuelle Comets
- INSERM, University Rennes-1, CIC 1414, F-35000 Rennes, France;
- INSERM, IAME, Université de Paris, F-75006 Paris, France
| | - Sarah Zohar
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
- HeKA, Inria, F-75006 Paris, France
| | - Charlotte Kloft
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
| |
Collapse
|
289
|
Mousavi Maleki MS, Rostamian M, Madanchi H. Antimicrobial peptides and other peptide-like therapeutics as promising candidates to combat SARS-CoV-2. Expert Rev Anti Infect Ther 2021; 19:1205-1217. [PMID: 33844613 PMCID: PMC8054488 DOI: 10.1080/14787210.2021.1912593] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: There are currently no specific drugs and universal vaccines for Coronavirus disease 2019 (COVID-19), hence urgent effective measures are needed to discover and develop therapeutic agents. Applying peptide therapeutics and their related compounds is a promising strategy to achieve this goal. This review is written based on the literature search using several databases, previous studies, scientific reports, our current knowledge about the antimicrobial peptides (AMPs), and our personal analyses on the potential of the antiviral peptides for the treatment of COVID-19. Areas covered: In this review, we begin with a brief description of SARS-CoV2 followed by a comprehensive description of antiviral peptides (AVPs) including natural and synthetic AMPs or AVPs and peptidomimetics. Subsequently, the structural features, mechanisms of action, limitations, and therapeutic applications of these peptides are explained. Expert opinion: Regarding the lack and the limitations of drugs against COVID-19, AMPs, AVPs, and other peptide-like compounds such as peptidomimetics have captured the attention of researchers due to their potential antiviral activities. Some of these compounds comprise unique properties and have demonstrated the potential to fight SARS-CoV2, particularly melittin, lactoferrin, enfuvirtide, and rupintrivir that have the potential to enter animal and clinical trials for the treatment of COVID-19.
Collapse
Affiliation(s)
- Masoumeh Sadat Mousavi Maleki
- Department of Biotechnology and Biotechnology Research Center, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mosayeb Rostamian
- Infectious Diseases Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamid Madanchi
- Department of Biotechnology and Biotechnology Research Center, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.,Drug Design and Bioinformatics Unit, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
290
|
Chan LW, Hern KE, Ngambenjawong C, Lee K, Kwon EJ, Hung DT, Bhatia SN. Selective Permeabilization of Gram-Negative Bacterial Membranes Using Multivalent Peptide Constructs for Antibiotic Sensitization. ACS Infect Dis 2021; 7:721-732. [PMID: 33689277 PMCID: PMC8043124 DOI: 10.1021/acsinfecdis.0c00805] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The drug-impermeable bacterial membrane in Gram-negative pathogens limits antibiotic access to intracellular drug targets. To expand our rapidly waning antibiotic arsenal, one approach is to improve the intracellular delivery of drugs with historically poor accumulation in Gram-negative bacteria. To do so, we engineered macromolecular potentiators to permeabilize the Gram-negative membrane to facilitate drug influx. Potentiators, known as WD40, were synthesized by grafting multiple copies of a cationic α-helical antimicrobial peptide, WLBU2, onto a dextran polymer scaffold. WD40 enabled drug uptake in the model pathogen P. aeruginosa, a capability that was not observed with unmodified WLBU2 peptide. WD40 was able to reduce minimum inhibitory concentrations of a drug panel by up to 3 orders of magnitude. Hydrophobic and highly three-dimensional antibiotics exhibited the greatest potentiation. Antibiotic activity was potentiated in several clinical strains and resulted in sensitization of drug-resistant strains to rifampin, a drug not previously used for Gram-negative infections.
Collapse
Affiliation(s)
- Leslie W. Chan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Kelsey E. Hern
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Chayanon Ngambenjawong
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Katie Lee
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02139, United States
| | - Ester J. Kwon
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Deborah T. Hung
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02139, United States
- Department of Molecular Biology, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sangeeta N. Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02139, United States
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
291
|
Naphthalimide-Containing BP100 Leads to Higher Model Membranes Interactions and Antimicrobial Activity. Biomolecules 2021; 11:biom11040542. [PMID: 33917850 PMCID: PMC8068292 DOI: 10.3390/biom11040542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/26/2021] [Accepted: 04/06/2021] [Indexed: 11/17/2022] Open
Abstract
In a large variety of organisms, antimicrobial peptides (AMPs) are primary defenses against pathogens. BP100 (KKLFKKILKYL-NH2), a short, synthetic, cationic AMP, is active against bacteria and displays low toxicity towards eukaryotic cells. BP100 acquires a α-helical conformation upon interaction with membranes and increases membrane permeability. Despite the volume of information available, the action mechanism of BP100, the selectivity of its biological effects, and possible applications are far from consensual. Our group synthesized a fluorescent BP100 analogue containing naphthalimide linked to its N-terminal end, NAPHT-BP100 (Naphthalimide-AAKKLFKKILKYL-NH2). The fluorescence properties of naphthalimides, especially their spectral sensitivity to microenvironment changes, are well established, and their biological activities against transformed cells and bacteria are known. Naphthalimide derived compounds are known to interact with DNA disturbing related processes as replication and transcription, and used as anticancer agents due to this property. A wide variety of techniques were used to demonstrate that NAPHT-BP100 bound to and permeabilized zwitterionic POPC and negatively charged POPC:POPG liposomes and, upon interaction, acquired a α-helical structure. Membrane surface high peptide/lipid ratios triggered complete permeabilization of the liposomes in a detergent-like manner. Membrane disruption was driven by charge neutralization, lipid aggregation, and bilayer destabilization. NAPHT-BP100 also interacted with double-stranded DNA, indicating that this peptide could also affect other cellular processes besides causing membrane destabilization. NAPHT-BP100 showed increased antibacterial and hemolytic activities, compared to BP100, and may constitute an efficient antimicrobial agent for dermatological use. By conjugating BP100 and naphthalimide DNA binding properties, NAPHT-BP100 bound to a large extent to the bacterial membrane and could more efficiently destabilize it. We also speculate that peptide could enter the bacteria cell and interact with its DNA in the cytoplasm.
Collapse
|
292
|
Kim M, Son J, Kim Y. Structural and Mechanismic Studies of Lactophoricin Analog, Novel Antibacterial Peptide. Int J Mol Sci 2021; 22:ijms22073734. [PMID: 33918526 PMCID: PMC8038340 DOI: 10.3390/ijms22073734] [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: 03/02/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 11/30/2022] Open
Abstract
Naturally derived antibacterial peptides exhibit excellent pharmacological action without the risk of resistance, suggesting a potential role as biologicals. Lactophoricin-I (LPcin-I), found in the proteose peptone component-3 (PP3; lactophorin) of bovine milk, is known to exhibit antibiotic activity against Gram-positive and Gram-negative bacteria. Accordingly, we derived a new antibacterial peptide and investigated its structure–function relationship. This study was initiated by designing antibacterial peptide analogs with better antibacterial activity, less cytotoxicity, and shorter amino acid sequences based on LPcin-I. The structural properties of antibacterial peptide analogs were investigated via spectroscopic analysis, and the antibacterial activity was confirmed by measurement of the minimal inhibitory concentration (MIC). The structure and mechanism of the antibacterial peptide analog in the cell membrane were also studied via solution-state nuclear magnetic resonance (NMR) and solid-state NMR spectroscopy. Through 15N one-dimensional and two-dimensional NMR experiments and 31P NMR experiments, we suggest the 3D morphology and antibacterial mechanism in the phospholipid bilayer of the LPcin analog. This study is expected to establish a system for the development of novel antibacterial peptides and to establish a theoretical basis for research into antibiotic substitutes.
Collapse
Affiliation(s)
| | | | - Yongae Kim
- Correspondence: ; Tel.: +82-31-330-4604; Fax: +82-31-330-4566
| |
Collapse
|
293
|
Sharma R, Shrivastava S, Kumar Singh S, Kumar A, Saxena S, Kumar Singh R. Deep-ABPpred: identifying antibacterial peptides in protein sequences using bidirectional LSTM with word2vec. Brief Bioinform 2021; 22:6204762. [PMID: 33784381 DOI: 10.1093/bib/bbab065] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
The overuse of antibiotics has led to emergence of antimicrobial resistance, and as a result, antibacterial peptides (ABPs) are receiving significant attention as an alternative. Identification of effective ABPs in lab from natural sources is a cost-intensive and time-consuming process. Therefore, there is a need for the development of in silico models, which can identify novel ABPs in protein sequences for chemical synthesis and testing. In this study, we propose a deep learning classifier named Deep-ABPpred that can identify ABPs in protein sequences. We developed Deep-ABPpred using bidirectional long short-term memory algorithm with amino acid level features from word2vec. The results show that Deep-ABPpred outperforms other state-of-the-art ABP classifiers on both test and independent datasets. Our proposed model achieved the precision of approximately 97 and 94% on test dataset and independent dataset, respectively. The high precision suggests applicability of Deep-ABPpred in proposing novel ABPs for synthesis and experimentation. By utilizing Deep-ABPpred, we identified ABPs in the tail protein sequences of Streptococcus bacteriophages, chemically synthesized identified peptides in lab and tested their activity in vitro. These ABPs showed potent antibacterial activity against selected Gram-positive and Gram-negative bacteria, which confirms the capability of Deep-ABPpred in identifying novel ABPs in protein sequences. Based on the proposed approach, an online prediction server is also developed, which is freely accessible at https://abppred.anvil.app/. This web server takes the protein sequence as input and provides ABPs with high probability (>0.95) as output.
Collapse
Affiliation(s)
- Ritesh Sharma
- Department of Computer Science and Engineering at IIT (BHU), Varanasi, India
| | | | - Sanjay Kumar Singh
- Department of Computer Science and Engineering at IIT (BHU), Varanasi, India
| | - Abhinav Kumar
- Department of Computer Science and Engineering at IIT (BHU), Varanasi, India
| | - Sonal Saxena
- Division of Veterinary Biotechnology, IVRI, Izatnagar, India
| | | |
Collapse
|
294
|
D’Souza AR, Necelis MR, Kulesha A, Caputo GA, Makhlynets OV. Beneficial Impacts of Incorporating the Non-Natural Amino Acid Azulenyl-Alanine into the Trp-Rich Antimicrobial Peptide buCATHL4B. Biomolecules 2021; 11:421. [PMID: 33809374 PMCID: PMC8001250 DOI: 10.3390/biom11030421] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial peptides (AMPs) present a promising scaffold for the development of potent antimicrobial agents. Substitution of tryptophan by non-natural amino acid Azulenyl-Alanine (AzAla) would allow studying the mechanism of action of AMPs by using unique properties of this amino acid, such as ability to be excited separately from tryptophan in a multi-Trp AMPs and environmental insensitivity. In this work, we investigate the effect of Trp→AzAla substitution in antimicrobial peptide buCATHL4B (contains three Trp side chains). We found that antimicrobial and bactericidal activity of the original peptide was preserved, while cytocompatibility with human cells and proteolytic stability was improved. We envision that AzAla will find applications as a tool for studies of the mechanism of action of AMPs. In addition, incorporation of this non-natural amino acid into AMP sequences could enhance their application properties.
Collapse
Affiliation(s)
- Areetha R. D’Souza
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY 13244, USA; (A.R.D.); (A.K.)
| | - Matthew R. Necelis
- Department of Chemistry & Biochemistry, Rowan University, Glassboro, NJ 08028, USA; (M.R.N.); (G.A.C.)
| | - Alona Kulesha
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY 13244, USA; (A.R.D.); (A.K.)
| | - Gregory A. Caputo
- Department of Chemistry & Biochemistry, Rowan University, Glassboro, NJ 08028, USA; (M.R.N.); (G.A.C.)
- Department of Molecular & Cellular Biosciences, Rowan University, Glassboro, NJ 08028, USA
| | - Olga V. Makhlynets
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY 13244, USA; (A.R.D.); (A.K.)
| |
Collapse
|
295
|
Chauhan S, Dhawan DK, Saini A, Preet S. Antimicrobial peptides against colorectal cancer-a focused review. Pharmacol Res 2021; 167:105529. [PMID: 33675962 DOI: 10.1016/j.phrs.2021.105529] [Citation(s) in RCA: 7] [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: 11/05/2020] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 12/25/2022]
Abstract
Despite recent advances in the treatment of colorectal cancer (CRC), low patient survival rate due to emergence of drug resistant cancer cells, metastasis and multiple deleterious side effects of chemotherapy, is a cause of public concern globally. To negate these clinical conundrums, search for effective and harmless novel molecular entities for the treatment of CRC is an urgent necessity. Since antimicrobial peptides (AMPs) are part of innate immunity of living beings, it is quite imperative to look for essential attributes of these peptides which may contribute to their effectiveness against carcinogenesis. Once identified, those characteristics can be suitably modified using several synthetic and computational techniques to further enhance their selectivity and pharmacokinetic profiles. Hence, this review analyses scientific reports describing the antiproliferative action of AMPs derived from several sources, particularly focusing on various colon cancer in vitro/in vivo investigations. On perusal of the literature, it appears that AMPs based therapeutics would definitely find special place in CRC therapy in future either alone or as an adjunct to chemotherapy provided some necessary alterations are made in their natural structures to make them more compatible with modern clinical practice. In this context, further in-depth research is warranted in adequate in vivo models.
Collapse
Affiliation(s)
- Sonia Chauhan
- Department of Biophysics, Basic Medical Sciences, Panjab University, Block-II, South Campus, Sector-25, Chandigarh 160014, India.
| | - Devinder K Dhawan
- Department of Biophysics, Basic Medical Sciences, Panjab University, Block-II, South Campus, Sector-25, Chandigarh 160014, India.
| | - Avneet Saini
- Department of Biophysics, Basic Medical Sciences, Panjab University, Block-II, South Campus, Sector-25, Chandigarh 160014, India.
| | - Simran Preet
- Department of Biophysics, Basic Medical Sciences, Panjab University, Block-II, South Campus, Sector-25, Chandigarh 160014, India.
| |
Collapse
|
296
|
Legood S, Boneca IG, Buddelmeijer N. Mode of action of lipoprotein modification enzymes-Novel antibacterial targets. Mol Microbiol 2021; 115:356-365. [PMID: 32979868 PMCID: PMC8048626 DOI: 10.1111/mmi.14610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/10/2020] [Indexed: 01/04/2023]
Abstract
Lipoproteins are characterized by a fatty acid moiety at their amino-terminus through which they are anchored into membranes. They fulfill a variety of essential functions in bacterial cells, such as cell wall maintenance, virulence, efflux of toxic elements including antibiotics, and uptake of nutrients. The posttranslational modification process of lipoproteins involves the sequential action of integral membrane enzymes and phospholipids as acyl donors. In recent years, the structures of the lipoprotein modification enzymes have been solved by X-ray crystallography leading to a greater insight into their function and the molecular mechanism of the reactions. The catalytic domains of the enzymes are exposed to the periplasm or external milieu and are readily accessible to small molecules. Since the lipoprotein modification pathway is essential in proteobacteria, it is a potential target for the development of novel antibiotics. In this review, we discuss recent literature on the structural characterization of the enzymes, and the in vitro activity assays compatible with high-throughput screening for inhibitors, with perspectives on the development of new antimicrobial agents.
Collapse
Affiliation(s)
- Simon Legood
- Institut PasteurUnité Biologie et Génétique de la Paroi BactérienneParisFrance
- CNRS, UMR 2001 « Microbiologie intégrative et Moléculaire »ParisFrance
- INSERM Groupe AvenirParisFrance
- Université de ParisSorbonne Paris CitéParisFrance
| | - Ivo G. Boneca
- Institut PasteurUnité Biologie et Génétique de la Paroi BactérienneParisFrance
- CNRS, UMR 2001 « Microbiologie intégrative et Moléculaire »ParisFrance
- INSERM Groupe AvenirParisFrance
| | - Nienke Buddelmeijer
- Institut PasteurUnité Biologie et Génétique de la Paroi BactérienneParisFrance
- CNRS, UMR 2001 « Microbiologie intégrative et Moléculaire »ParisFrance
- INSERM Groupe AvenirParisFrance
| |
Collapse
|
297
|
Spirescu VA, Chircov C, Grumezescu AM, Andronescu E. Polymeric Nanoparticles for Antimicrobial Therapies: An Up-To-Date Overview. Polymers (Basel) 2021; 13:724. [PMID: 33673451 PMCID: PMC7956825 DOI: 10.3390/polym13050724] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Despite the many advancements in the pharmaceutical and medical fields and the development of numerous antimicrobial drugs aimed to suppress and destroy pathogenic microorganisms, infectious diseases still represent a major health threat affecting millions of lives daily. In addition to the limitations of antimicrobial drugs associated with low transportation rate, water solubility, oral bioavailability and stability, inefficient drug targeting, considerable toxicity, and limited patient compliance, the major cause for their inefficiency is the antimicrobial resistance of microorganisms. In this context, the risk of a pre-antibiotic era is a real possibility. For this reason, the research focus has shifted toward the discovery and development of novel and alternative antimicrobial agents that could overcome the challenges associated with conventional drugs. Nanotechnology is a possible alternative, as there is significant evidence of the broad-spectrum antimicrobial activity of nanomaterials and nanoparticles in particular. Moreover, owing to their considerable advantages regarding their efficient cargo dissolving, entrapment, encapsulation, or surface attachment, the possibility of forming antimicrobial groups for specific targeting and destruction, biocompatibility and biodegradability, low toxicity, and synergistic therapy, polymeric nanoparticles have received considerable attention as potential antimicrobial drug delivery agents. In this context, the aim of this paper is to provide an up-to-date overview of the most recent studies investigating polymeric nanoparticles designed for antimicrobial therapies, describing both their targeting strategies and their effects.
Collapse
Affiliation(s)
- Vera Alexandra Spirescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| | - Cristina Chircov
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| |
Collapse
|
298
|
Dijksteel GS, Ulrich MMW, Middelkoop E, Boekema BKHL. Review: Lessons Learned From Clinical Trials Using Antimicrobial Peptides (AMPs). Front Microbiol 2021; 12:616979. [PMID: 33692766 PMCID: PMC7937881 DOI: 10.3389/fmicb.2021.616979] [Citation(s) in RCA: 227] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial peptides (AMPs) or host defense peptides protect the host against various pathogens such as yeast, fungi, viruses and bacteria. AMPs also display immunomodulatory properties ranging from the modulation of inflammatory responses to the promotion of wound healing. More interestingly, AMPs cause cell disruption through non-specific interactions with the membrane surface of pathogens. This is most likely responsible for the low or limited emergence of bacterial resistance against many AMPs. Despite the increasing number of antibiotic-resistant bacteria and the potency of novel AMPs to combat such pathogens, only a few AMPs are in clinical use. Therefore, the current review describes (i) the potential of AMPs as alternatives to antibiotics, (ii) the challenges toward clinical implementation of AMPs and (iii) strategies to improve the success rate of AMPs in clinical trials, emphasizing the lessons we could learn from these trials.
Collapse
Affiliation(s)
- Gabrielle S Dijksteel
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Magda M W Ulrich
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther Middelkoop
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | |
Collapse
|
299
|
Tuning of a Membrane-Perforating Antimicrobial Peptide to Selectively Target Membranes of Different Lipid Composition. J Membr Biol 2021; 254:75-96. [PMID: 33564914 DOI: 10.1007/s00232-021-00174-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/21/2021] [Indexed: 12/16/2022]
Abstract
The use of designed antimicrobial peptides as drugs has been impeded by the absence of simple sequence-structure-function relationships and design rules. The likely cause is that many of these peptides permeabilize membranes via highly disordered, heterogeneous mechanisms, forming aggregates without well-defined tertiary or secondary structure. We suggest that the combination of high-throughput library screening with atomistic computer simulations can successfully address this challenge by tuning a previously developed general pore-forming peptide into a selective pore-former for different lipid types. A library of 2916 peptides was designed based on the LDKA template. The library peptides were synthesized and screened using a high-throughput orthogonal vesicle leakage assay. Dyes of different sizes were entrapped inside vesicles with varying lipid composition to simultaneously screen for both pore size and affinity for negatively charged and neutral lipid membranes. From this screen, nine different LDKA variants that have unique activity were selected, sequenced, synthesized, and characterized. Despite the minor sequence changes, each of these peptides has unique functional properties, forming either small or large pores and being selective for either neutral or anionic lipid bilayers. Long-scale, unbiased atomistic molecular dynamics (MD) simulations directly reveal that rather than rigid, well-defined pores, these peptides can form a large repertoire of functional dynamic and heterogeneous aggregates, strongly affected by single mutations. Predicting the propensity to aggregate and assemble in a given environment from sequence alone holds the key to functional prediction of membrane permeabilization.
Collapse
|
300
|
Bakovic A, Risner K, Bhalla N, Alem F, Chang TL, Weston WK, Harness JA, Narayanan A. Brilacidin Demonstrates Inhibition of SARS-CoV-2 in Cell Culture. Viruses 2021; 13:271. [PMID: 33572467 PMCID: PMC7916214 DOI: 10.3390/v13020271] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 12/17/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the newly emergent causative agent of coronavirus disease-19 (COVID-19), has resulted in more than two million deaths worldwide since it was first detected in 2019. There is a critical global need for therapeutic intervention strategies that can be deployed to safely treat COVID-19 disease and reduce associated morbidity and mortality. Increasing evidence shows that both natural and synthetic antimicrobial peptides (AMPs), also referred to as Host Defense Proteins/Peptides (HDPs), can inhibit SARS-CoV-2, paving the way for the potential clinical use of these molecules as therapeutic options. In this manuscript, we describe the potent antiviral activity exerted by brilacidin-a de novo designed synthetic small molecule that captures the biological properties of HDPs-on SARS-CoV-2 in a human lung cell line (Calu-3) and a monkey cell line (Vero). These data suggest that SARS-CoV-2 inhibition in these cell culture models is likely to be a result of the impact of brilacidin on viral entry and its disruption of viral integrity. Brilacidin demonstrated synergistic antiviral activity when combined with remdesivir. Collectively, our data demonstrate that brilacidin exerts potent inhibition of SARS-CoV-2 against different strains of the virus in cell culture.
Collapse
Affiliation(s)
- Allison Bakovic
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| | - Kenneth Risner
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| | - Nishank Bhalla
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| | - Farhang Alem
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| | - Theresa L. Chang
- Public Health Research Institute, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ 07103, USA;
| | - Warren K. Weston
- Innovation Pharmaceuticals Inc., Wakefield, MA 01880, USA; (W.K.W.); (J.A.H.)
| | - Jane A. Harness
- Innovation Pharmaceuticals Inc., Wakefield, MA 01880, USA; (W.K.W.); (J.A.H.)
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| |
Collapse
|