Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.354
Filter
1.
PLoS One ; 19(5): e0300125, 2024.
Article in English | MEDLINE | ID: mdl-38722967

ABSTRACT

With the increasing problem of antimicrobial drug resistance, the search for new antimicrobial agents has become a crucial task in the field of medicine. Antimicrobial peptides, as a class of naturally occurring antimicrobial agents, possess broad-spectrum antimicrobial activity and lower risk of resistance development. However, traditional screening methods for antimicrobial peptides are inefficient, necessitating the development of an efficient screening model. In this study, we aimed to develop an ensemble learning model for the identification of antimicrobial peptides, named E-CLEAP, based on the Multilayer Perceptron Classifier (MLP Classifier). By considering multiple features, including amino acid composition (AAC) and pseudo amino acid composition (PseAAC) of antimicrobial peptides, we aimed to improve the accuracy and generalization ability of the identification process. To validate the superiority of our model, we employed five-fold cross-validation and compared it with other commonly used methods for antimicrobial peptide identification. In the experimental results on an independent test set, E-CLEAP achieved accuracies of 97.33% and 84% for the AAC and PseAAC features, respectively. The results demonstrated that our model outperformed other methods in all evaluation metrics. The findings of this study highlight the potential of the E-CLEAP model in enhancing the efficiency and accuracy of antimicrobial peptide screening, which holds significant implications for drug development, disease treatment, and biotechnology advancement. Future research can further optimize the model by incorporating additional features and information, as well as validating its reliability on larger datasets and in real-world environments. The source code and all datasets are publicly available at https://github.com/Wangsicheng52/E-CLEAP.


Subject(s)
Antimicrobial Peptides , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Machine Learning , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Amino Acids/chemistry
2.
Protein Sci ; 33(6): e5006, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723168

ABSTRACT

The emergence and spread of antibiotic-resistant bacteria pose a significant public health threat, necessitating the exploration of alternative antibacterial strategies. Antibacterial peptide (ABP) is a kind of antimicrobial peptide (AMP) that has the potential ability to fight against bacteria infection, offering a promising avenue for developing novel therapeutic interventions. This study introduces AMPActiPred, a three-stage computational framework designed to identify ABPs, characterize their activity against diverse bacterial species, and predict their activity levels. AMPActiPred employed multiple effective peptide descriptors to effectively capture the compositional features and physicochemical properties of peptides. AMPActiPred utilized deep forest architecture, a cascading architecture similar to deep neural networks, capable of effectively processing and exploring original features to enhance predictive performance. In the first stage, AMPActiPred focuses on ABP identification, achieving an Accuracy of 87.6% and an MCC of 0.742 on an elaborate dataset, demonstrating state-of-the-art performance. In the second stage, AMPActiPred achieved an average GMean at 82.8% in identifying ABPs targeting 10 bacterial species, indicating AMPActiPred can achieve balanced predictions regarding the functional activity of ABP across this set of species. In the third stage, AMPActiPred demonstrates robust predictive capabilities for ABP activity levels with an average PCC of 0.722. Furthermore, AMPActiPred exhibits excellent interpretability, elucidating crucial features associated with antibacterial activity. AMPActiPred is the first computational framework capable of predicting targets and activity levels of ABPs. Finally, to facilitate the utilization of AMPActiPred, we have established a user-friendly web interface deployed at https://awi.cuhk.edu.cn/∼AMPActiPred/.


Subject(s)
Anti-Bacterial Agents , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Bacteria/drug effects , Computational Biology/methods , Neural Networks, Computer , Microbial Sensitivity Tests
3.
PLoS One ; 19(5): e0302913, 2024.
Article in English | MEDLINE | ID: mdl-38728358

ABSTRACT

In the fight against antimicrobial resistance, host defense peptides (HDPs) are increasingly referred to as promising molecules for the design of new antimicrobial agents. In terms of their future clinical use, particularly small, synthetic HDPs offer several advantages, based on which their application as feed additives has aroused great interest in the poultry sector. However, given their complex mechanism of action and the limited data about the cellular effects in production animals, their investigation is of great importance in these species. The present study aimed to examine the immunomodulatory activity of the synthetic HDP Pap12-6 (PAP) solely and in inflammatory environments evoked by lipoteichoic acid (LTA) and polyinosinic-polycytidylic acid (Poly I:C), in a primary chicken hepatocyte-non-parenchymal cell co-culture. Based on the investigation of the extracellular lactate dehydrogenase (LDH) activity, PAP seemed to exert no cytotoxicity on hepatic cells, suggesting its safe application. Moreover, PAP was able to influence the immune response, reflected by the decreased production of interleukin (IL)-6, IL-8, and "regulated on activation, normal T cell expressed and secreted"(RANTES), as well as the reduced IL-6/IL-10 ratio in Poly I:C-induced inflammation. PAP also diminished the levels of extracellular H2O2 and nuclear factor erythroid 2-related factor 2 (Nrf2) when applied together with Poly I:C and in both inflammatory conditions, respectively. Consequently, PAP appeared to display potent immunomodulatory activity, preferring to act towards the cellular anti-inflammatory and antioxidant processes. These findings confirm that PAP might be a promising alternative for designing novel antimicrobial immunomodulatory agents for chickens, thereby contributing to the reduction of the use of conventional antibiotics.


Subject(s)
Chickens , Hepatocytes , Lipopolysaccharides , Poly I-C , Animals , Hepatocytes/drug effects , Hepatocytes/immunology , Hepatocytes/metabolism , Poly I-C/pharmacology , Lipopolysaccharides/pharmacology , Immunologic Factors/pharmacology , Teichoic Acids/pharmacology , Cells, Cultured , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry , Coculture Techniques , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Cytokines/metabolism , Antimicrobial Cationic Peptides/pharmacology
4.
Nat Commun ; 15(1): 3945, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730238

ABSTRACT

Proline-rich antimicrobial peptides (PrAMPs) inhibit bacterial protein biosynthesis by binding to the polypeptide exit tunnel (PET) near the peptidyl transferase center. Api137, an optimized derivative of honeybee PrAMP apidaecin, inhibits protein expression by trapping release factors (RFs), which interact with stop codons on ribosomes to terminate translation. This study uses cryo-EM, functional assays and molecular dynamic (MD) simulations to show that Api137 additionally occupies a second binding site near the exit of the PET and can repress translation independently of RF-trapping. Api88, a C-terminally amidated (-CONH2) analog of Api137 (-COOH), binds to the same sites, occupies a third binding pocket and interferes with the translation process presumably without RF-trapping. In conclusion, apidaecin-derived PrAMPs inhibit bacterial ribosomes by multimodal mechanisms caused by minor structural changes and thus represent a promising pool for drug development efforts.


Subject(s)
Antimicrobial Cationic Peptides , Molecular Dynamics Simulation , Ribosomes , Ribosomes/metabolism , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Protein Biosynthesis , Binding Sites , Cryoelectron Microscopy , Escherichia coli/metabolism , Escherichia coli/genetics , Escherichia coli/drug effects , Peptide Termination Factors/metabolism , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Protein Binding , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/metabolism , Antimicrobial Peptides/pharmacology
5.
J Med Microbiol ; 73(5)2024 May.
Article in English | MEDLINE | ID: mdl-38743468

ABSTRACT

Introduction. Innovative antifungal therapies are of crucial importance to combat the potentially life-threatening infections linked to the multidrug-resistant fungal pathogen Candida auris. Induction of regulated cell death, apoptosis, could provide an outline for future therapeutics. Human antimicrobial peptides (AMPs), well-known antifungal compounds, have shown the ability to induce apoptosis in pathogenic fungi.Hypothesis/Gap Statement . Although it is known that AMPs possess antifungal activity against C. auris, their ability to induce apoptosis requires further investigations.Aim. This study evaluated the effects of AMPs on the induction of apoptosis in C. auris.Methods. Human neutrophil peptide-1 (HNP-1), human ß-Defensins-3 (hBD-3) and human salivary histatin 5 (His 5) were assessed against two clinical C. auris isolates. Apoptosis hallmarks were examined using FITC-Annexin V/PI double labelling assay and terminal deoxynucleotidyl transferase deoxynucleotidyl transferase nick-end labelling (TUNEL) to detect phosphatidylserine externalization and DNA fragmentation, respectively. Then, several intracellular triggers were studied using JC-10 staining, spectrophotometric assay and 2',7'-dichlorofluorescin diacetate staining to measure the mitochondrial membrane potential, cytochrome-c release and reactive oxygen species (ROS) production, respectively.Results and conclusion. FITC-Annexin V/PI staining and TUNEL analysis revealed that exposure of C. auris cells to HNP-1 and hBD-3 triggered both early and late apoptosis, while His 5 caused significant necrosis. Furthermore, HNP-1 and hBD-3 induced significant mitochondrial membrane depolarization, which resulted in substantial cytochrome c release. In contrast to His 5, which showed minimal mitochondrial depolarization and no cytochrome c release. At last, all peptides significantly increased ROS production, which is related to both types of cell death. Therefore, these peptides represent promising and effective antifungal agents for treating invasive infections caused by multidrug-resistant C. auris.


Subject(s)
Antifungal Agents , Apoptosis , Candida auris , Histatins , Reactive Oxygen Species , Apoptosis/drug effects , Humans , Antifungal Agents/pharmacology , Histatins/pharmacology , Reactive Oxygen Species/metabolism , Candida auris/drug effects , beta-Defensins/pharmacology , Membrane Potential, Mitochondrial/drug effects , alpha-Defensins/pharmacology , Microbial Sensitivity Tests , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Candidiasis/drug therapy , Candidiasis/microbiology
6.
Eur J Med Chem ; 271: 116451, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38691892

ABSTRACT

The potent antibacterial activity and low resistance of antimicrobial peptides (AMPs) render them potential candidates for treating multidrug-resistant bacterial infections. Herein, a minimalist design strategy was proposed employing the "golden partner" combination of arginine (R) and tryptophan (W), along with a dendritic structure to design AMPs. By extension, the α/ε-amino group and the carboxyl group of lysine (K) were utilized to link R and W, forming dendritic peptide templates αRn(εRn)KWm-NH2 and αWn(εWn)KRm-NH2, respectively. The corresponding linear peptide templates R2nKWm-NH2 and W2nKRm-NH2 were used as controls. Their physicochemical properties, activity, toxicity, and stability were compared. Among these new peptides, the dendritic peptide R2(R2)KW4 was screened as a prospective candidate owing to its preferable antibacterial properties, biocompatibility, and stability. Additionally, R2(R2)KW4 not only effectively restrained the progression of antibiotic resistance, but also demonstrated synergistic utility when combined with conventional antibiotics due to its unique membrane-disruptive mechanism. Furthermore, R2(R2)KW4 possessed low toxicity (LD50 = 109.31 mg/kg) in vivo, while efficiently clearing E. coli in pulmonary-infected mice. In conclusion, R2(R2)KW4 has the potential to become an antimicrobial regent or adjuvant, and the minimalist design strategy of dendritic peptides provides innovative and encouraging thoughts in designing AMPs.


Subject(s)
Anti-Bacterial Agents , Arginine , Microbial Sensitivity Tests , Tryptophan , Tryptophan/chemistry , Tryptophan/pharmacology , Animals , Arginine/chemistry , Arginine/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Mice , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemical synthesis , Structure-Activity Relationship , Molecular Structure , Cell Membrane/drug effects , Dose-Response Relationship, Drug , Bacterial Infections/drug therapy , Humans , Escherichia coli/drug effects
7.
Front Cell Infect Microbiol ; 14: 1339285, 2024.
Article in English | MEDLINE | ID: mdl-38720961

ABSTRACT

Antimicrobial peptides (AMPs), often referred to as nature's antibiotics, are ubiquitous in living organisms, spanning from bacteria to humans. Their potency, versatility, and unique mechanisms of action have garnered significant research attention. Unlike conventional antibiotics, peptides are biodegradable, adding to their appeal as potential candidates to address bacterial resistance in livestock farming-a challenge that has been under scrutiny for decades. This issue is complex and multifactorial, influenced by a variety of components. The World Health Organization (WHO) has proposed a comprehensive approach known as One Health, emphasizing the interconnectedness of human-animal-environment relationships in tackling such challenges. This review explores the application of AMPs in livestock farming and how they can mitigate the impact of this practice within the One Health framework.


Subject(s)
Antimicrobial Peptides , Livestock , One Health , Livestock/microbiology , Animals , Humans , Antimicrobial Peptides/pharmacology , Drug Resistance, Bacterial , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects
8.
Appl Microbiol Biotechnol ; 108(1): 316, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38700735

ABSTRACT

Nowadays, it is very important to produce new-generation drugs with antimicrobial properties that will target biofilm-induced infections. The first target for combating these microorganisms, which are the source itself. Antimicrobial peptides, which are more effective than antibiotics due to their ability to kill microorganisms and use a different metabolic pathway, are among the new options today. The aim of this study is to develop new-generation antibiotics that inhibit both biofilm-producing bacteria and the biofilm itself. For this purpose, we designed four different peptides by combining two amino acid forms (D- and L-) with the same sequence having alpha helix structures. It was found that the combined use of these two forms can increase antimicrobial efficacy more than 30-fold. These results are supported by molecular modeling and scanning electron microscopy (SEM), at the same time cytotoxicity (IC50) and hemotoxicity (HC50) values remained within the safe range. Furthermore, antibiofilm activities of these peptides were investigated. Since the existing biofilm inhibition methods in the literature do not technically simulate the exact situation, in this study, we have developed a real-time observable biofilm model and a new detection method based on it, which we call the CoMIC method. Findings have shown that the NET1 peptide with D-leucine amino acid in its structure and the NET3 peptide with D-arginine amino acid in its structure are effective in inhibiting biofilm. As a conclusion, our peptides can be considered as potential next-generation broad-spectrum antibiotic molecule/drug candidates that might be used in biofilm and clinical important bacteria. KEY POINTS: • Antimicrobial peptides were developed to inhibit both biofilms producing bacteria and the biofilm itself. • CoMIC will fill a very crucial gap in understanding biofilms and conducting the necessary quantitative studies. • Molecular modelling studies, NET1 peptide molecules tends to move towards and adhere to the membrane within nanoseconds.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Peptides , Biofilms , Microbial Sensitivity Tests , Biofilms/drug effects , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Models, Molecular , Microscopy, Electron, Scanning , Bacteria/drug effects
9.
Front Immunol ; 15: 1384193, 2024.
Article in English | MEDLINE | ID: mdl-38694504

ABSTRACT

The common bed bug, Cimex lectularius, is an urban pest of global health significance, severely affecting the physical and mental health of humans. In contrast to most other blood-feeding arthropods, bed bugs are not major vectors of pathogens, but the underlying mechanisms for this phenomenon are largely unexplored. Here, we present the first transcriptomics study of bed bugs in response to immune challenges. To study transcriptional variations in bed bugs following ingestion of bacteria, we extracted and processed mRNA from body tissues of adult male bed bugs after ingestion of sterile blood or blood containing the Gram-positive (Gr+) bacterium Bacillus subtilis or the Gram-negative (Gr-) bacterium Escherichia coli. We analyzed mRNA from the bed bugs' midgut (the primary tissue involved in blood ingestion) and from the rest of their bodies (RoB; body minus head and midgut tissues). We show that the midgut exhibits a stronger immune response to ingestion of bacteria than the RoB, as indicated by the expression of genes encoding antimicrobial peptides (AMPs). Both the Toll and Imd signaling pathways, associated with immune responses, were highly activated by the ingestion of bacteria. Bacterial infection in bed bugs further provides evidence for metabolic reconfiguration and resource allocation in the bed bugs' midgut and RoB to promote production of AMPs. Our data suggest that infection with particular pathogens in bed bugs may be associated with altered metabolic pathways within the midgut and RoB that favors immune responses. We further show that multiple established cellular immune responses are preserved and are activated by the presence of specific pathogens. Our study provides a greater understanding of nuances in the immune responses of bed bugs towards pathogens that ultimately might contribute to novel bed bug control tactics.


Subject(s)
Bedbugs , Gene Expression Profiling , Transcriptome , Animals , Bedbugs/immunology , Bedbugs/genetics , Male , Escherichia coli/immunology , Bacillus subtilis/immunology , Bacillus subtilis/genetics , Signal Transduction/immunology , Antimicrobial Peptides/genetics , Antimicrobial Peptides/immunology
10.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732089

ABSTRACT

Antimicrobial peptides (AMPs) are molecules with an amphipathic structure that enables them to interact with bacterial membranes. This interaction can lead to membrane crossing and disruption with pore formation, culminating in cell death. They are produced naturally in various organisms, including humans, animals, plants and microorganisms. In higher animals, they are part of the innate immune system, where they counteract infection by bacteria, fungi, viruses and parasites. AMPs can also be designed de novo by bioinformatic approaches or selected from combinatorial libraries, and then produced by chemical or recombinant procedures. Since their discovery, AMPs have aroused interest as potential antibiotics, although few have reached the market due to stability limits or toxicity. Here, we describe the development phase and a number of clinical trials of antimicrobial peptides. We also provide an update on AMPs in the pharmaceutical industry and an overall view of their therapeutic market. Modifications to peptide structures to improve stability in vivo and bioavailability are also described.


Subject(s)
Antimicrobial Peptides , Humans , Animals , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Clinical Trials as Topic , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology
11.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732132

ABSTRACT

Insects possess an effective immune system, which has been extensively characterized in several model species, revealing a plethora of conserved genes involved in recognition, signaling, and responses to pathogens and parasites. However, some taxonomic groups, characterized by peculiar trophic niches, such as plant-sap feeders, which are often important pests of crops and forestry ecosystems, have been largely overlooked regarding their immune gene repertoire. Here we annotated the immune genes of soft scale insects (Hemiptera: Coccidae) for which omics data are publicly available. By using immune genes of aphids and Drosophila to query the genome of Ericerus pela, as well as the transcriptomes of Ceroplastes cirripediformis and Coccus sp., we highlight the lack of peptidoglycan recognition proteins, galectins, thaumatins, and antimicrobial peptides in Coccidae. This work contributes to expanding our knowledge about the evolutionary trajectories of immune genes and offers a list of promising candidates for developing new control strategies based on the suppression of pests' immunity through RNAi technologies.


Subject(s)
Hemiptera , Insect Proteins , Animals , Hemiptera/genetics , Hemiptera/immunology , Insect Proteins/genetics , Insect Proteins/immunology , Transcriptome/genetics , Phylogeny , Antimicrobial Peptides/genetics , Galectins/genetics , Galectins/metabolism , Carrier Proteins
12.
Int J Biol Macromol ; 267(Pt 1): 131480, 2024 May.
Article in English | MEDLINE | ID: mdl-38599427

ABSTRACT

Bone regeneration remains a major clinical challenge, especially when infection necessitates prolonged antibiotic treatment. This study presents a membrane composed of self-assembled and interpenetrating GL13K, an antimicrobial peptide (AMP) derived from a salivary protein, in a collagen membrane for antimicrobial activity and enhanced bone regeneration. Commercially available collagen membranes were immersed in GL13K solution, and self-assembly was initiated by raising the solution pH to synthesize the multifunctional membrane called COL-GL. COL-GL was composed of interpenetrating large collagen fibers and short GL13K nanofibrils, which increased hydrophobicity, reduced biodegradation from collagenase, and stiffened the matrix compared to control collagen membranes. Incorporation of GL13K led to antimicrobial and anti-fouling activity against early oral surface colonizer Streptococcus gordonii while not affecting fibroblast cytocompatibility or pre-osteoblast osteogenic differentiation. GL13K in solution also reduced macrophage inflammatory cytokine expression and increased pro-healing cytokine expression. Bone formation in a rat calvarial model was accelerated at eight weeks with COL-GL compared to the gold-standard collagen membrane based on microcomputed tomography and histology. Interpenetration of GL13K within collagen sidesteps challenges with antimicrobial coatings on bone regeneration scaffolds while increasing bone regeneration. This strength makes COL-GL a promising approach to reduce post-surgical infections and aid bone regeneration in dental and orthopedic applications. STATEMENT OF SIGNIFICANCE: The COL-GL membrane, incorporating the antimicrobial peptide GL13K within a collagen membrane, signifies a noteworthy breakthrough in bone regeneration strategies for dental and orthopedic applications. By integrating self-assembled GL13K nanofibers into the membrane, this study successfully addresses the challenges associated with antimicrobial coatings, exhibiting improved antimicrobial and anti-fouling activity while preserving compatibility with fibroblasts and pre-osteoblasts. The accelerated bone formation observed in a rat calvarial model emphasizes the potential of this innovative approach to minimize post-surgical infections and enhance bone regeneration outcomes. As a promising alternative for future therapeutic interventions, this material tackles the clinical challenges of extended antibiotic treatments and antibiotic resistance in bone regeneration scenarios.


Subject(s)
Antimicrobial Peptides , Bone Regeneration , Collagen , Membranes, Artificial , Nanofibers , Bone Regeneration/drug effects , Animals , Rats , Nanofibers/chemistry , Collagen/chemistry , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Osteogenesis/drug effects , Mice , Osteoblasts/drug effects , Streptococcus gordonii/drug effects , Male , Rats, Sprague-Dawley , Fibroblasts/drug effects
13.
Sci Total Environ ; 927: 172150, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38580107

ABSTRACT

The use of in-feed antibiotics has been widely restricted due to the significant environmental pollution and food safety concerns they have caused. Antimicrobial peptides (AMPs) have attracted widespread attention as potential future alternatives to in-feed antibiotics owing to their demonstrated antimicrobial activity and environment friendly characteristics. However, the challenges of weak bioactivity, immature stability, and low production yields of natural AMPs impede practical application in the feed industry. To address these problems, efforts have been made to develop strategies for approaching the AMPs with enhanced properties. Herein, we summarize approaches to improving the properties of AMPs as potential alternatives to in-feed antibiotics, mainly including optimization of structural parameters, sequence modification, selection of microbial hosts, fusion expression, and industrially fermentation control. Additionally, the potential for application of AMPs in animal husbandry is discussed. This comprehensive review lays a strong theoretical foundation for the development of in-feed AMPs to achieve the public health globally.


Subject(s)
Animal Feed , Anti-Bacterial Agents , Antimicrobial Peptides , Animal Husbandry/methods , Animals
14.
Cells ; 13(8)2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38667271

ABSTRACT

Even with the best infection control protocols in place, the risk of a hospital-acquired infection of the surface of an implanted device remains significant. A bacterial biofilm can form and has the potential to escape the host immune system and develop resistance to conventional antibiotics, ultimately causing the implant to fail, seriously impacting patient well-being. Here, we demonstrate a 4 log reduction in the infection rate by the common pathogen S. aureus of 3D-printed polyaryl ether ketone (PAEK) polymeric surfaces by covalently binding the antimicrobial peptide Mel4 to the surface using plasma immersion ion implantation (PIII) treatment. The surfaces with added texture created by 3D-printed processes such as fused deposition-modelled polyether ether ketone (PEEK) and selective laser-sintered polyether ketone (PEK) can be equally well protected as conventionally manufactured materials. Unbound Mel4 in solution at relevant concentrations is non-cytotoxic to osteoblastic cell line Saos-2. Mel4 in combination with PIII aids Saos-2 cells to attach to the surface, increasing the adhesion by 88% compared to untreated materials without Mel4. A reduction in mineralisation on the Mel4-containing surfaces relative to surfaces without peptide was found, attributed to the acellular portion of mineral deposition.


Subject(s)
Antimicrobial Peptides , Benzophenones , Polymers , Printing, Three-Dimensional , Prostheses and Implants , Staphylococcus aureus , Humans , Staphylococcus aureus/drug effects , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/metabolism , Prostheses and Implants/adverse effects , Polymers/chemistry , Polymers/pharmacology , Biofilms/drug effects , Ketones/chemistry , Ketones/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Surface Properties , Bone and Bones/drug effects , Bone and Bones/metabolism , Orthopedics
15.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38673985

ABSTRACT

Antimicrobial resistance is a silent pandemic harming human health, and Pseudomonas aeruginosa is the most common bacterium responsible for chronic pulmonary and eye infections. Antimicrobial peptides (AMPs) represent promising alternatives to conventional antibiotics. In this review, the in vitro/in vivo activities of the frog skin-derived AMP Esc(1-21) are shown. Esc(1-21) rapidly kills both the planktonic and sessile forms of P. aeruginosa and stimulates migration of epithelial cells, likely favoring repair of damaged tissue. However, to undertake preclinical studies, some drawbacks of AMPs (cytotoxicity, poor biostability, and limited delivery to the target site) must be overcome. For this purpose, the stereochemistry of two amino acids of Esc(1-21) was changed to obtain the diastereomer Esc(1-21)-1c, which is more stable, less cytotoxic, and more efficient in treating P. aeruginosa-induced lung and cornea infections in mouse models. Incorporation of these peptides (Esc peptides) into nanoparticles or immobilization to a medical device (contact lens) was revealed to be an effective strategy to ameliorate and/or to prolong the peptides' antimicrobial efficacy. Overall, these data make Esc peptides encouraging candidates for novel multifunctional drugs to treat lung pathology especially in patients with cystic fibrosis and eye dysfunctions, characterized by both tissue injury and bacterial infection.


Subject(s)
Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Pseudomonas aeruginosa/drug effects , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Humans , Anura , Skin/microbiology , Skin/drug effects , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/therapeutic use , Antimicrobial Cationic Peptides/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/chemistry
16.
Pak J Biol Sci ; 27(3): 152-159, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38686737

ABSTRACT

<b>Background and Objective:</b> Rabbit meat is a livestock product potentially viable as a protein source to obtain peptides. Antioxidant and antimicrobial peptides are ingredients extracted from various foods through enzymatic hydrolysis, chemical hydrolysis and fermentation to produce health-promoting foods. This research aims to investigate the potential of rabbit meat as a source of antioxidant and antimicrobial peptides through hydrolysis using trypsin and zingibain enzymes. <b>Materials and Methods:</b> This research conducted an explorative-descriptive approach, focusing on antioxidant and antimicrobial activity. Rabbit meat was extracted using trypsin, zingibain and a combination of trypsin and crude extract zingibain. The hydrolyzed rabbit meat extract was tested at intervals of 0, 2, 6, 16, 24, 40 and 48 hrs to determine the degree of hydrolysis and the profile of hydrolyzed proteins with electrophoresis SDS PAGE. The antioxidant activity was tested using the DPPH method and the antimicrobial activity using agar well diffusion method. <b>Results:</b> The degree of hydrolysis increased with the hydrolysis time. The highest protein content of rabbit meat extract hydrolyzed with trypsin was 287.65 mg/mL, observed during 12 hrs hydrolysis. The optimum conditions for the hydrolysis of rabbit meat protein were obtained at 24 hrs, with an IC<sub>50</sub> value of 52.45% hydrolyzed by trypsin. As per antimicrobial activities, <i>Escherichia coli</i> and <i>Salmonella</i> sp. were more effective in inhibiting rabbit meat hydrolysates compared to <i>Pseudomonas aeruginosa</i> and <i>Staphylococcus aureus</i>. The inhibition of all pathogen increased until 12 hrs hydrolysis but decreased in 24 hrs hydrolysis. <b>Conclusion:</b> The combination zingibain enzyme and trypsin is feasible for hydrolyzing rabbit meat and the optimum hydrolysis time was 24 hrs with IC<sub>50</sub> 52.45 ppm, although accompanied by reduction in antibacterial activities.


Subject(s)
Antioxidants , Meat , Trypsin , Animals , Rabbits , Antioxidants/pharmacology , Trypsin/metabolism , Hydrolysis , Protein Hydrolysates/pharmacology , Anti-Infective Agents/pharmacology , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Peptides/pharmacology , Peptides/chemistry , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology
17.
J Med Chem ; 67(9): 7487-7503, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38688020

ABSTRACT

Although bactericidal cationic antimicrobial peptides (AMPs) have been well characterized, less information is available about the antibacterial properties and mechanisms of action of nonbactericidal AMPs, especially nonbactericidal anionic AMPs. Herein, a novel anionic antimicrobial peptide (Gy-CATH) with a net charge of -4 was identified from the skin of the frog Glyphoglossus yunnanensis. Gy-CATH lacks direct antibacterial effects but exhibits significantly preventive and therapeutic capacities in mice that are infected with Staphylococcus aureus, Enterobacteriaceae coli, methicillin-resistant Staphylococcus aureus (MRSA), or carbapenem-resistant E. coli (CREC). In vitro and in vivo investigations proved the regulation of Gy-CATH on neutrophils and macrophages involved in the host immune defense against infection. Moreover, Gy-CATH significantly reduced the extent of pulmonary fibrin deposition and prevented thrombosis in mice, which was attributed to the regulatory role of Gy-CATH in physiological anticoagulants and platelet aggregation. These findings show that Gy-CATH is a potential candidate for the treatment of bacterial infection.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Peptides , Animals , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/therapeutic use , Anura , Bacterial Infections/drug therapy , Bacterial Infections/prevention & control , Escherichia coli/drug effects , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/therapeutic use , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunologic Factors/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Thrombosis/prevention & control , Thrombosis/drug therapy
18.
J Microbiol ; 62(3): 167-179, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38630349

ABSTRACT

The human gut houses a diverse and dynamic microbiome critical for digestion, metabolism, and immune development, exerting profound effects on human health. However, these microorganisms pose a potential threat by breaching the gut barrier, entering host tissues, and triggering infections, uncontrolled inflammation, and even sepsis. The intestinal epithelial cells form the primary defense, acting as a frontline barrier against microbial invasion. Antimicrobial proteins (AMPs), produced by these cells, serve as innate immune effectors that regulate the gut microbiome by directly killing or inhibiting microbes. Abnormal AMP production, whether insufficient or excessive, can disturb the microbiome equilibrium, contributing to various intestinal diseases. This review delves into the complex interactions between AMPs and the gut microbiota and sheds light on the role of AMPs in governing host-microbiota interactions. We discuss the function and mechanisms of action of AMPs, their regulation by the gut microbiota, microbial evasion strategies, and the consequences of AMP dysregulation in disease. Understanding these complex interactions between AMPs and the gut microbiota is crucial for developing strategies to enhance immune responses and combat infections within the gut microbiota. Ongoing research continues to uncover novel aspects of this intricate relationship, deepening our understanding of the factors shaping gut health. This knowledge has the potential to revolutionize therapeutic interventions, offering enhanced treatments for a wide range of gut-related diseases.


Subject(s)
Gastrointestinal Microbiome , Gastrointestinal Microbiome/physiology , Humans , Animals , Antimicrobial Peptides/metabolism , Immunity, Innate , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Bacteria/metabolism , Intestines/microbiology , Intestines/immunology
19.
Molecules ; 29(7)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38611812

ABSTRACT

Antibiotic resistance has emerged as a grave threat to global public health, leading to an increasing number of treatment failures. Antimicrobial peptides (AMPs) are widely regarded as potential substitutes for traditional antibiotics since they are less likely to induce resistance when used. A novel AMP named Brevinin-1BW (FLPLLAGLAASFLPTIFCKISRKC) was obtained by the Research Center of Molecular Medicine of Yunnan Province from the skin of the Pelophylax nigromaculatus. Brevinia-1BW had effective inhibitory effects on Gram-positive bacteria, with a minimum inhibitory concentration (MIC) of 3.125 µg/mL against Enterococcus faecalis (ATCC 29212) and 6.25 µg/mL against both Staphylococcus aureus (ATCC 25923) and multidrug-resistant Staphylococcus aureus (ATCC 29213) but had weaker inhibitory effects on Gram-negative bacteria, with a MIC of ≥100 µg/mL. Studies using scanning electron microscopy (SEM) and flow cytometry have revealed that it exerts its antibacterial activity by disrupting bacterial membranes. Additionally, it possesses strong biofilm inhibitory and eradication activities as well as significant lipopolysaccharide (LPS)-binding activity. Furthermore, Brevinin-1BW has shown a significant anti-inflammatory effect in LPS-treated RAW264.7 cells. In conclusion, Brevinin-1BW is anticipated to be a promising clinical agent with potent anti-Gram-positive bacterial and anti-inflammatory properties.


Subject(s)
Lipopolysaccharides , Methicillin-Resistant Staphylococcus aureus , China , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antimicrobial Peptides
20.
Int J Mol Sci ; 25(7)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38612644

ABSTRACT

Antimicrobial peptides (AMPs), as immune effectors synthesized by a variety of organisms, not only constitute a robust defense mechanism against a broad spectrum of pathogens in the host but also show promising applications as effective antimicrobial agents. Notably, insects are significant reservoirs of natural AMPs. However, the complex array of variations in types, quantities, antimicrobial activities, and production pathways of AMPs, as well as evolution of AMPs across insect species, presents a significant challenge for immunity system understanding and AMP applications. This review covers insect AMP discoveries, classification, common properties, and mechanisms of action. Additionally, the types, quantities, and activities of immune-related AMPs in each model insect are also summarized. We conducted the first comprehensive investigation into the diversity, distribution, and evolution of 20 types of AMPs in model insects, employing phylogenetic analysis to describe their evolutionary relationships and shed light on conserved and distinctive AMP families. Furthermore, we summarize the regulatory pathways of AMP production through classical signaling pathways and additional pathways associated with Nitric Oxide, insulin-like signaling, and hormones. This review advances our understanding of AMPs as guardians in insect immunity systems and unlocks a gateway to insect AMP resources, facilitating the use of AMPs to address food safety concerns.


Subject(s)
Antimicrobial Peptides , Food Safety , Humans , Animals , Phylogeny , Insecta , Nitric Oxide
SELECTION OF CITATIONS
SEARCH DETAIL
...