Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 73
1.
Food Res Int ; 183: 114224, 2024 May.
Article En | MEDLINE | ID: mdl-38760143

During infant formula production, proteins are always heated, potentially affecting their digestibility and the bioactivities of resulting peptides. Although plant proteins are a promising dairy alternative for infant formula, they remain understudied, necessitating further investigations. Therefore, this research aimed to fill this gap by assessing the impact of different heating modes on soy protein (SP) and pea protein (PP), focusing on glycation levels, peptide formation during in vitro infant digestion, and immune protection potential (sRAGE-binding and antimicrobial activities) of the resulting peptides. Consequently, dry heating led to increased glycation and glycated peptide production, particularly with higher glycation in PP than SP. Moreover, PP exhibited an overall stronger sRAGE-binding capacity than SP, regardless of heating and digestion conditions. Regarding antimicrobial activity, both SP and PP-derived peptides displayed reduced effectiveness against Enterobacter cloacae after dry heating. Additionally, Staphylococcus epidermidis was differently inhibited, where PP-derived peptides showed inherent inhibition. The primary determinant of sRAGE-binding and antimicrobial potential in digestion-derived peptides was the protein source. Subsequent bioinformatics analysis predicted 519 and 133 potential antimicrobial peptides in SP and PP, respectively. This study emphasises the importance of protein source for infant formula to ensure infant health.


Digestion , Hot Temperature , Infant Formula , Pea Proteins , Soybean Proteins , Soybean Proteins/metabolism , Humans , Infant Formula/chemistry , Infant , Pea Proteins/metabolism , Pea Proteins/chemistry , Receptor for Advanced Glycation End Products/metabolism , Antimicrobial Peptides/metabolism , Anti-Infective Agents/pharmacology
2.
Nat Commun ; 15(1): 3945, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730238

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.


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
3.
Nat Commun ; 15(1): 4216, 2024 May 17.
Article En | MEDLINE | ID: mdl-38760394

Antimicrobial peptides (AMPs), ancient scavengers of bacteria, are very poorly induced in macrophages infected by Mycobacterium tuberculosis (M. tuberculosis), but the underlying mechanism remains unknown. Here, we report that L-alanine interacts with PRSS1 and unfreezes the inhibitory effect of PRSS1 on the activation of NF-κB pathway to induce the expression of AMPs, but mycobacterial alanine dehydrogenase (Ald) Rv2780 hydrolyzes L-alanine and reduces the level of L-alanine in macrophages, thereby suppressing the expression of AMPs to facilitate survival of mycobacteria. Mechanistically, PRSS1 associates with TAK1 and disruptes the formation of TAK1/TAB1 complex to inhibit TAK1-mediated activation of NF-κB pathway, but interaction of L-alanine with PRSS1, disables PRSS1-mediated impairment on TAK1/TAB1 complex formation, thereby triggering the activation of NF-κB pathway to induce expression of AMPs. Moreover, deletion of antimicrobial peptide gene ß-defensin 4 (Defb4) impairs the virulence by Rv2780 during infection in mice. Both L-alanine and the Rv2780 inhibitor, GWP-042, exhibits excellent inhibitory activity against M. tuberculosis infection in vivo. Our findings identify a previously unrecognized mechanism that M. tuberculosis uses its own alanine dehydrogenase to suppress host immunity, and provide insights relevant to the development of effective immunomodulators that target M. tuberculosis.


Alanine , Antimicrobial Peptides , Macrophages , Mycobacterium tuberculosis , NF-kappa B , Tuberculosis , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/metabolism , Animals , Mice , NF-kappa B/metabolism , Humans , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Alanine/metabolism , Antimicrobial Peptides/metabolism , Antimicrobial Peptides/genetics , Tuberculosis/microbiology , Tuberculosis/immunology , Alanine Dehydrogenase/metabolism , Alanine Dehydrogenase/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Signal Transduction , Mice, Inbred C57BL , RAW 264.7 Cells , Female
4.
Cells ; 13(8)2024 Apr 09.
Article En | MEDLINE | ID: mdl-38667271

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.


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
5.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38673813

We explored the metabolic integration of Blattella germanica and its obligate endosymbiont Blattabacterium cuenoti by the transcriptomic analysis of the fat body of quasi-aposymbiotic cockroaches, where the endosymbionts were almost entirely removed with rifampicin. Fat bodies from quasi-aposymbiotic insects displayed large differences in gene expression compared to controls. In quasi-aposymbionts, the metabolism of phenylalanine and tyrosine involved in cuticle sclerotization and pigmentation increased drastically to compensate for the deficiency in the biosynthesis of these amino acids by the endosymbionts. On the other hand, the uricolytic pathway and the biosynthesis of uric acid were severely decreased, probably because the reduced population of endosymbionts was unable to metabolize urea to ammonia. Metabolite transporters that could be involved in the endosymbiosis process were identified. Immune system and antimicrobial peptide (AMP) gene expression was also reduced in quasi-aposymbionts, genes encoding peptidoglycan-recognition proteins, which may provide clues for the maintenance of the symbiotic relationship, as well as three AMP genes whose involvement in the symbiotic relationship will require additional analysis. Finally, a search for AMP-like factors that could be involved in controlling the endosymbiont identified two orphan genes encoding proteins smaller than 200 amino acids underexpressed in quasi-aposymbionts, suggesting a role in the host-endosymbiont relationship.


Fat Body , Symbiosis , Transcriptome , Symbiosis/genetics , Animals , Fat Body/metabolism , Female , Gene Expression Profiling , Immune System/metabolism , Bacteroidetes/genetics , Bacteroidetes/metabolism , Antimicrobial Peptides/metabolism , Antimicrobial Peptides/genetics
6.
Front Immunol ; 15: 1191966, 2024.
Article En | MEDLINE | ID: mdl-38655253

NK-lysin is a potent antimicrobial peptide (AMP) with antimicrobial activity against bacteria, fungi, viruses, and parasites. NK-lysin is a type of granulysin, a member of the saposin-like proteins family first isolated from a pig's small intestine. In previous work, for the first time, we identified four variants of nk-lysin from Atlantic salmon (Salmo salar) using EST sequences. In the present study, we reported and characterized two additional transcripts of NK-lysin from S. salar. Besides, we evaluated the tissue distribution of three NK-lysins from S. salar and assessed the antimicrobial, hemolytic, and immunomodulatory activities and signaling pathways of three NK-lysin-derived peptides. The synthetic peptides displayed antimicrobial activity against Piscirickettsia salmonis (LF-89) and Flavobacterium psychrophilum. These peptides induced the expression of immune genes related to innate and adaptive immune responses in vitro and in vivo. The immunomodulatory activity of the peptides involves the mitogen-activated protein kinases-mediated signaling pathway, including p38, extracellular signal-regulated kinase 1/2, and/or c-Jun N-terminal kinases. Besides, the peptides modulated the immune response induced by pathogen-associated molecular patterns (PAMPs). Our findings show that NK-lysin could be a highly effective immunostimulant or vaccine adjuvant for use in fish aquaculture.


Antimicrobial Peptides , Fish Proteins , Proteolipids , Salmo salar , Animals , Antimicrobial Peptides/metabolism , Antimicrobial Peptides/pharmacology , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Proteins/metabolism , Fish Proteins/pharmacology , Immunity, Innate , Proteolipids/metabolism , Proteolipids/pharmacology , Salmo salar/immunology , Signal Transduction
7.
Biochem Biophys Res Commun ; 712-713: 149913, 2024 Jun 18.
Article En | MEDLINE | ID: mdl-38640738

Innate immunity of invertebrates offers potent antimicrobial peptides (AMPs) against drug-resistant infections. To identify new worm ß-hairpin AMPs, we explored the sequence diversity of proteins with a BRICHOS domain, which comprises worm AMP precursors. Strikingly, we discovered new BRICHOS AMPs not in worms, but in caecilians, the least studied clade of vertebrates. Two precursor proteins from Microcaecilia unicolor and Rhinatrema bivittatum resemble SP-C lung surfactants and bear worm AMP-like peptides at C-termini. The analysis of M. unicolor tissue transcriptomes shows that the AMP precursor is highly expressed in the lung along with regular SP-C, suggesting a different, protective function. The peptides form right-twisted ß-hairpins, change conformation upon lipid binding, and rapidly disrupt bacterial membranes. Both peptides exhibit broad-spectrum activity against multidrug-resistant ESKAPE pathogens with 1-4 µM MICs and remarkably low toxicity, giving 40-70-fold selectivity towards bacteria. These BRICHOS AMPs, previously unseen in vertebrates, reveal a novel lung innate immunity mechanism and offer a promising antibiotics template.


Antimicrobial Peptides , Lung , Animals , Amino Acid Sequence , Amphibians/immunology , Amphibians/metabolism , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/metabolism , Immunity, Innate , Lung/immunology , Lung/metabolism , Microbial Sensitivity Tests
8.
Biochemistry ; 63(10): 1257-1269, 2024 May 21.
Article En | MEDLINE | ID: mdl-38683758

Interactions between SJGAP (skipjack tuna GAPDH-related antimicrobial peptide) and four analogs thereof with model bacterial membranes were studied using Fourier-transform infrared spectroscopy (FTIR) and molecular dynamics (MD) simulations. MD trajectory analyses showed that the N-terminal segment of the peptide analogs has many contacts with the polar heads of membrane phospholipids, while the central α helix interacts strongly with the hydrophobic core of the membranes. The peptides also had a marked influence on the wave numbers associated with the phase transition of phospholipids organized as liposomes in both the interface and aliphatic chain regions of the infrared spectra, supporting the interactions observed in the MD trajectories. In addition, interesting links were found between peptide interactions with the aliphatic chains of membrane phospholipids, as determined by FTIR and from the MD trajectories, and the membrane permeabilization capacity of these peptide analogs, as previously demonstrated. To summarize, the combined experimental and computational efforts have provided insights into crucial aspects of the interactions between the investigated peptides and bacterial membranes. This work thus makes an original contribution to our understanding of the molecular interactions underlying the antimicrobial activity of these GAPDH-related antimicrobial peptides from Scombridae.


Molecular Dynamics Simulation , Animals , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Fish Proteins/chemistry , Fish Proteins/metabolism , Fish Proteins/pharmacology , Spectroscopy, Fourier Transform Infrared , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Amino Acid Sequence
9.
J Microbiol ; 62(3): 167-179, 2024 Mar.
Article En | MEDLINE | ID: mdl-38630349

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.


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
10.
Food Chem ; 448: 139119, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38547703

Buffalo colostrum is the initial mammary secretion after parturition, consisting of nutritional and bioactive components. In this study, we conducted a proteomic analysis of buffalo colostrum whey to identify bioactive proteins and peptides. A total of 107 differentially expressed proteins (DEPs) were identified in buffalo colostrum whey compared to those in mature milk. Gene Ontology analysis revealed that DEPs were primarily associated with immune response and tissue development. KEGG pathway enrichment suggested that colostrum actively enhances nascent immunity involved in interleukin and interferon signaling pathways. Furthermore, candidate antimicrobial peptides (AMPs) of whey protein hydrolysates from buffalo colostrum were characterized, which exhibits broad-spectrum activity against gram-positive and gram-negative pathogens. Overall, this study improves our understanding of protein variations in buffalo lactation, and contributes to the development of AMPs from buffalo colostrum.


Antimicrobial Peptides , Buffaloes , Colostrum , Milk , Proteomics , Whey Proteins , Animals , Colostrum/chemistry , Colostrum/metabolism , Female , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/analysis , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/metabolism , Milk/chemistry , Whey Proteins/chemistry , Whey Proteins/metabolism , Whey Proteins/analysis , Whey/chemistry , Whey/metabolism
11.
Insect Mol Biol ; 33(3): 270-282, 2024 Jun.
Article En | MEDLINE | ID: mdl-38329162

Insects rely on their innate immune system to eliminate pathogenic microbes. As a system component, cytokines transmit intercellular signals to control immune responses. Growth-blocking peptide (GBP) is a member of the stress-responsive peptide family of cytokines found in several orders of insects, including Drosophila. However, the physiological role of GBP in defence against pathogens is not thoroughly understood. In this study, we explored the functions of GBP in a lepidopteran pest, Ostrinia furnacalis. Injection of recombinant O. furnacalis GBP (OfGBP) precursor (proGBP) and chemically synthesised GBP significantly induced the transcription of antimicrobial peptides (AMPs) and other immunity-related genes including immune deficiency (IMD) and Dorsal. The level of OfGBP mRNA was upregulated after bacterial infection. Knockdown of OfGBP expression led to a decrease in IMD, Relish, MyD88 and Dorsal mRNA levels. OfGBP induced phenoloxidase activity and affected hemocyte behaviours in O. furnacalis larvae. In summary, GBP is a potent cytokine, effectively regulating AMP synthesis, melanization response and cellular immunity to eliminate invading pathogens.


Insect Proteins , Larva , Moths , Animals , Moths/immunology , Moths/genetics , Moths/growth & development , Insect Proteins/metabolism , Insect Proteins/genetics , Larva/growth & development , Larva/immunology , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/genetics , Antimicrobial Peptides/metabolism , Hemocytes/metabolism , Immunity, Innate
12.
Nature ; 626(7999): 617-625, 2024 Feb.
Article En | MEDLINE | ID: mdl-38081298

The outer membrane in Gram-negative bacteria consists of an asymmetric phospholipid-lipopolysaccharide bilayer that is densely packed with outer-membrane ß-barrel proteins (OMPs) and lipoproteins1. The architecture and composition of this bilayer is closely monitored and is essential to cell integrity and survival2-4. Here we find that SlyB, a lipoprotein in the PhoPQ stress regulon, forms stable stress-induced complexes with the outer-membrane proteome. SlyB comprises a 10 kDa periplasmic ß-sandwich domain and a glycine zipper domain that forms a transmembrane α-helical hairpin with discrete phospholipid- and lipopolysaccharide-binding sites. After loss in lipid asymmetry, SlyB oligomerizes into ring-shaped transmembrane complexes that encapsulate ß-barrel proteins into lipid nanodomains of variable size. We find that the formation of SlyB nanodomains is essential during lipopolysaccharide destabilization by antimicrobial peptides or acute cation shortage, conditions that result in a loss of OMPs and compromised outer-membrane barrier function in the absence of a functional SlyB. Our data reveal that SlyB is a compartmentalizing transmembrane guard protein that is involved in cell-envelope proteostasis and integrity, and suggest that SlyB represents a larger family of broadly conserved lipoproteins with 2TM glycine zipper domains with the ability to form lipid nanodomains.


Bacterial Outer Membrane Proteins , Cell Membrane , Gram-Negative Bacteria , Lipid Bilayers , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Glycine/metabolism , Lipopolysaccharides/metabolism , Lipoproteins/chemistry , Lipoproteins/metabolism , Phospholipids/metabolism , Binding Sites , Proteostasis , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Proteome/chemistry , Proteome/metabolism , Regulon , Protein Domains , Antimicrobial Peptides/metabolism , Gram-Negative Bacteria/chemistry , Gram-Negative Bacteria/cytology , Gram-Negative Bacteria/metabolism
13.
World J Microbiol Biotechnol ; 39(10): 276, 2023 Aug 12.
Article En | MEDLINE | ID: mdl-37567959

The increasing number of life-threatening infections caused by persister bacteria is associated with various issues, including antimicrobial resistance and biofilm formation. Infections due to persister cells are often difficult to suppress without the use of last-resort antibiotics. Throughout the world, bacterial persistence and resistance create an unmet clinical demand for the exploration of newly introduced therapeutic approaches. Mesenchymal stem / stromal cells (MSCs) have an antimicrobial activity to protect against bacterial infections, including those caused by bacterial persisters. MSCs have substantial potential to secrete antimicrobial peptides (AMPs), including cathelicidin, beta-defensins, lipocalin-2, hepcidin, indoleamine 2,3-dioxygenase (IDO), cysteine proteases, and inducible nitric oxide synthases (iNOS). MSCs possess the potential to contribute to innate immunity by regulating the immune response. Recently, MSCs and their secreted components have been reported to improve antimicrobial activity. Bactericidal activity by MSCs and their secretomes has been shown to be mediated in part by the secretion of AMPs. Even though they were discovered more than 80 years ago, therapeutic options for persisters are restricted, and there is an urgent need for alternative treatment regimens. Hence, this review intends to critically assess the current literature on the effects of MSCs and their secretomes on persister bacteria. MSCs and their secretome-based therapies could be preferred as an up-and-coming approach to reinforce the antimicrobial efficiency in persister infections.


Bacterial Infections , Cell- and Tissue-Based Therapy , Mesenchymal Stem Cells , Secretome , Stromal Cells , Antimicrobial Peptides/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Bacterial Infections/microbiology , Bacterial Infections/therapy , Humans , Animals , Stromal Cells/cytology , Stromal Cells/metabolism
14.
Science ; 381(6657): 502-508, 2023 08 04.
Article En | MEDLINE | ID: mdl-37535745

The mammalian gut secretes a family of multifunctional peptides that affect appetite, intestinal secretions, and motility whereas others regulate the microbiota. We have found that peptide YY (PYY1-36), but not endocrine PYY3-36, acts as an antimicrobial peptide (AMP) expressed by gut epithelial paneth cells (PC). PC-PYY is packaged into secretory granules and is secreted into and retained by surface mucus, which optimizes PC-PYY activity. Although PC-PYY shows some antibacterial activity, it displays selective antifungal activity against virulent Candida albicans hyphae-but not the yeast form. PC-PYY is a cationic molecule that interacts with the anionic surfaces of fungal hyphae to cause membrane disruption and transcriptional reprogramming that selects for the yeast phenotype. Hence, PC-PYY is an antifungal AMP that contributes to the maintenance of gut fungal commensalism.


Antifungal Agents , Antimicrobial Peptides , Candida , Paneth Cells , Peptide Fragments , Peptide YY , Animals , Antifungal Agents/metabolism , Antimicrobial Peptides/metabolism , Candida/drug effects , Candida/physiology , Paneth Cells/metabolism , Peptide Fragments/metabolism , Peptide YY/metabolism , Symbiosis , Humans , Mice
15.
Microb Pathog ; 182: 106234, 2023 Sep.
Article En | MEDLINE | ID: mdl-37442216

Growing antimicrobial resistance (AMR) is a threat to human and animal populations citing the limited available options. Alternative antimicrobial options or functional enhancement of currently available antimicrobials remains only options. One of the potential options seems stem cells especially the mesenchymal stem cells (MSCs) that show antimicrobial properties. These cells additionally have pro-healing effects that may plausibly improve healing outcomes. MSCs antimicrobial actions are mediated either through direct cell-cell contact or their secretome that enhances innate immune mediated antimicrobial activities. These cells synergistically enhance efficacy of currently available antimicrobials especially against the biofilms. Reciprocal action from antimicrobials on the MSCs functionality remains poorly understood. Currently, the main limitation with MSCs based therapy is their limited efficacy. This demands further understanding and can be enhanced through biotechnological interventions. One of the interventional options is the 'priming' to enhance MSCs resistance and specific expression potential. The available literature shows potential antimicrobial actions of MSCs both ex vivo as well as in vivo. The studies on veterinary species are very promising although limited by number and extensiveness in details for their utility as standard therapeutic agents. The current review aims to discuss the role of animals in AMR and the potential antimicrobial actions of MSCs in veterinary medicine. The review also discusses the limitations in their utilization as standard therapeutics.


Bacterial Infections , Cell- and Tissue-Based Therapy , Mesenchymal Stem Cells , Animals , Animals, Domestic , Bacterial Infections/therapy , Bacterial Infections/veterinary , Cell- and Tissue-Based Therapy/veterinary , Drug Resistance , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Virus Diseases/therapy , Virus Diseases/veterinary , Secretome , Antimicrobial Peptides/metabolism
16.
Science ; 381(6655): eadg5725, 2023 07 21.
Article En | MEDLINE | ID: mdl-37471548

Antimicrobial peptides are host-encoded immune effectors that combat pathogens and shape the microbiome in plants and animals. However, little is known about how the host antimicrobial peptide repertoire is adapted to its microbiome. Here, we characterized the function and evolution of the Diptericin antimicrobial peptide family of Diptera. Using mutations affecting the two Diptericins (Dpt) of Drosophila melanogaster, we reveal the specific role of DptA for the pathogen Providencia rettgeri and DptB for the gut mutualist Acetobacter. The presence of DptA- or DptB-like genes across Diptera correlates with the presence of Providencia and Acetobacter in their environment. Moreover, DptA- and DptB-like sequences predict host resistance against infection by these bacteria across the genus Drosophila. Our study explains the evolutionary logic behind the bursts of rapid evolution of an antimicrobial peptide family and reveals how the host immune repertoire adapts to changing microbial environments.


Acetobacter , Antimicrobial Peptides , Drosophila Proteins , Drosophila melanogaster , Host-Pathogen Interactions , Microbiota , Providencia , Animals , Antimicrobial Peptides/genetics , Antimicrobial Peptides/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/microbiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Evolution, Molecular , Host-Pathogen Interactions/immunology
17.
Fish Shellfish Immunol ; 132: 108454, 2023 Jan.
Article En | MEDLINE | ID: mdl-36442704

Ubiquitination and deubiquitination of target proteins is an important mechanism for cells to rapidly respond to changes in the external environment. The deubiquitinase, cylindromatosis (CYLD), is a tumor suppressor protein. CYLD from Drosophila melanogaster participates in the antimicrobial immune response. In vertebrates, CYLD also regulates bacterial-induced apoptosis. However, whether CYLD can regulate the bacterial-induced innate immune response in crustaceans is unknown. In the present study, we reported the identification and cloning of CYLD in Chinese mitten crab, Eriocheir sinensis. Quantitative real-time reverse transcription polymerase chain reaction analysis showed that EsCYLD was widely expressed in all the examined tissues and was upregulated in the hemolymph after Vibrio parahaemolyticus challenge. Knockdown of EsCYLD in hemocytes promoted the cytoplasm-to-nucleus translocation of transcription factor Relish under V. parahaemolyticus stimulation and increased the expression of corresponding antimicrobial peptides. In vivo, silencing of EsCYLD promoted the removal of bacteria from the crabs and enhanced their survival. In addition, interfering with EsCYLD expression inhibited apoptosis of crab hemocytes caused by V. parahaemolyticus stimulation. In summary, our findings revealed that EsCYLD negatively regulates the nuclear translocation of Relish to affect the expression of corresponding antimicrobial peptides and regulates the apoptosis of crab hemocytes, thus indirectly participating in the innate immunity of E. sinensis.


Apoptosis , Arthropod Proteins , Brachyura , Deubiquitinating Enzyme CYLD , Hemocytes , Immunity, Innate , Transcription Factors , Animals , Amino Acid Sequence , Antimicrobial Peptides/metabolism , Arthropod Proteins/classification , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Base Sequence , Brachyura/immunology , Brachyura/microbiology , Deubiquitinating Enzyme CYLD/classification , Deubiquitinating Enzyme CYLD/genetics , Deubiquitinating Enzyme CYLD/metabolism , Hemocytes/enzymology , Immunity, Innate/genetics , Phylogeny , Transcription Factors/metabolism , Vibrio parahaemolyticus , Active Transport, Cell Nucleus
18.
Am J Reprod Immunol ; 88(2): e13577, 2022 08.
Article En | MEDLINE | ID: mdl-35567598

PROBLEM: Cervical insufficiency (CI) is associated with intra-amniotic infection or inflammation. Antimicrobial peptides (AMPs) in amniotic fluid may protect the fetus against microbial invasion, giving their broad-spectrum microbiocidal properties. We evaluated changes in amniotic fluid AMP expression in women with CI and assessed whether these changes are related to their pregnancy outcomes. METHOD OF STUDY: We evaluated amniotic fluid human neutrophil peptide 3 (HNP-3), human ß-defensin-2 (hBD-2), and LL-37 levels in 66 women with CI and 25 normal controls at 16-24 weeks of gestation. The CI group was divided into short cervix and cervical dilation groups, and the cervical dilation group was further divided into preterm and full-term delivery groups according to the pregnancy outcomes, and AMP expression was analyzed in each group. RESULTS: HNP-3 and hBD-2 levels were higher in women with CI than in normal controls and in the cervical dilation as compared to the short cervix group. Among women with cervical dilation, 22 delivered at full-term, and 23 had spontaneous preterm births. The hBD-2 level in amniotic fluid mid-pregnancy was higher in the full-term delivery than in the preterm delivery groups. However, LL-37 levels in amniotic fluid were low in women with CI and normal controls. CONCLUSIONS: Amniotic fluid HNP-3 and hBD-2 levels increased in women with CI compared with normal controls. Moreover, increased amniotic fluid hBD-2 levels mid-pregnancy were associated with favorable pregnancy outcomes in women with CI. AMPs in the amniotic fluid may participate in host defense against ascending infection in women with CI.


Amniotic Fluid , Antimicrobial Peptides , Cervix Uteri , Female , Humans , Infant, Newborn , Pregnancy , Amniotic Fluid/metabolism , Antimicrobial Peptides/metabolism , Cervix Uteri/physiopathology , Inflammation/metabolism , Premature Birth/metabolism
19.
Proc Natl Acad Sci U S A ; 119(10): e2115669119, 2022 03 08.
Article En | MEDLINE | ID: mdl-35238667

SignificanceSimilar to mammalian TLR4/MD-2, the Toll9/MD-2-like protein complex in the silkworm, Bombyx mori, acts as an innate pattern-recognition receptor that recognizes lipopolysaccharide (LPS) and induces LPS-stimulated expression of antimicrobial peptides such as cecropins. Here, we report that papiliocin, a cecropin-like insect antimicrobial peptide from the swallowtail butterfly, competitively inhibits the LPS-TLR4/MD-2 interaction by directly binding to human TLR4/MD-2. Structural elements in papiliocin, which are important in inhibiting TLR4 signaling via direct binding, are highly conserved among insect cecropins, indicating that its TLR4-antagonistic activity may be related to insect Toll9-mediated immune response against microbial infection. This study highlights the potential of papiliocin as a potent TLR4 antagonist and safe peptide antibiotic for treating gram-negative sepsis.


Anti-Infective Agents, Local/pharmacology , Antimicrobial Peptides/pharmacology , Butterflies/immunology , Immunity, Innate/drug effects , Insect Proteins/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Anti-Infective Agents, Local/chemistry , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/metabolism , Escherichia coli Infections/drug therapy , Female , Insect Proteins/chemistry , Insect Proteins/metabolism , Lipopolysaccharides/metabolism , Mice , Mice, Inbred ICR , Molecular Docking Simulation , Protein Binding , Protein Conformation , Toll-Like Receptor 4/metabolism
20.
Sci Rep ; 12(1): 2576, 2022 02 16.
Article En | MEDLINE | ID: mdl-35173253

Antimicrobial peptides (AMPs) have been developed for the treatment of bacterial infections, but their applications are limited to topical infections since they are sequestered and inhibited in serum. Here we have discovered that the inhibition of AMPs by human serum was mediated through high-density lipoproteins (HDL) which are known to remove cholesterol from peripheral tissues. The susceptibility of AMPs to HDL varied depending on the degree of hydrophobicity of AMPs and their binding affinities to HDL. The phospholipids, such as phosphatidylcholine, of HDL were essential for AMP-binding. The dynamic binding interactions between AMPs and HDL were mediated through the hydrophobic interactions rather than by ionic strength. Interestingly, some AMPs, such as SMAP29, dissociated from the AMP-HDL complex and translocated to bacteria upon contact, while some AMPs, such as LL37, remained in complex with HDL. These results suggest that HDL binds AMPs and facilitates the translocation of them to the bacteria.


Anti-Bacterial Agents/metabolism , Antimicrobial Peptides/metabolism , Bacteria/metabolism , Blood Proteins/metabolism , Lipids/chemistry , Lipoproteins, HDL/metabolism , Serum/metabolism , Humans , Hydrophobic and Hydrophilic Interactions
...