Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cytokine ; 183: 156749, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39236431

RESUMEN

In humans and mice, the induction of interleukin (IL)-17 expression enhances epithelial barrier integrity through the secretion of antimicrobial peptides (AMP), thereby improving antibacterial defense. However, it is unclear whether IL-17 has similar antibacterial effects in chickens by modulating the expression of AMPs, such as avian beta-defensins (also known as gallinacins) and cathelicidins. This study evaluated the in vivo effects of inoculating 20-day-old broiler chickens with two doses of a plasmid encoding chicken IL-17 (pCDNA3.1/rchIL-17-V5-HIS TOPO plasmid [pCDNA3.1-IL-17]; 5 or 10 µg/bird). On day 23 of age, all broilers, except those in the negative control group, were orally challenged with a virulent Clostridium perfringens strain for three days. To investigate IL-17-mediated effects against C. perfringens infection, the expression of avian beta-defensin 1 (avBD1), avBD2, avBD4, avBD6, cathelicidins, and inducible nitric oxide synthase (iNOS) genes were quantified, and gross necrotic enteritis (NE) lesion scores were assessed in the small intestine. The results showed that broilers receiving the higher dose of pCDNA3.1-IL-17 (10 µg) had significantly lower NE lesion scores compared to those receiving the lower dose (5 µg), the vector control, and the positive control groups. Furthermore, the expression of all avian beta-defensins and cathelicidin genes was detectable across all groups, regardless of treatment and time points. IL-17 treatment led to significantly higher expression of avBD1, avBD2, avBD4, avBD6, cathelicidin, and iNOS in the duodenum, jejunum, and ileum compared to control chickens. In C. perfringens-infected chickens, the expression of avBD1, avBD2, avBD4, cathelicidin, and iNOS in the ileum was significantly higher than in control chickens. Pre-treatment with the higher dose of pCDNA3.1-IL-17 (10 µg) in infected chickens was associated with reduced NE lesion severity and increased expression of avBD1, avBD2, cathelicidin, and iNOS in the ileum, but not avBD4 and avBD6. These findings provide new insights into the potential effect of IL-17 and reduction in NE lesion severity by modulating AMP expression which may be involved in mediating protective immunity against intestinal infection with C. perfringens.


Asunto(s)
Pollos , Clostridium perfringens , Enteritis , Interleucina-17 , Intestino Delgado , beta-Defensinas , Animales , Pollos/microbiología , Interleucina-17/metabolismo , Interleucina-17/genética , Enteritis/microbiología , Enteritis/inmunología , Enteritis/veterinaria , Enteritis/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Intestino Delgado/inmunología , beta-Defensinas/metabolismo , beta-Defensinas/genética , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/metabolismo , Catelicidinas , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Necrosis , Modelos Animales de Enfermedad , Infecciones por Clostridium/veterinaria , Infecciones por Clostridium/inmunología , Péptidos Catiónicos Antimicrobianos/metabolismo , Péptidos Catiónicos Antimicrobianos/genética , Regulación de la Expresión Génica/efectos de los fármacos
2.
Gut Microbes ; 16(1): 2390680, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39244776

RESUMEN

The mucus serves as a protective barrier in the gastrointestinal tract against microbial attacks. While its role extends beyond merely being a physical barrier, the extent of its active bactericidal properties remains unclear, and the mechanisms regulating these properties are not yet understood. We propose that inflammation induces epithelial cells to secrete antimicrobial peptides, transforming mucus into an active bactericidal agent. To investigate the properties of mucus, we previously developed mucosoid culture models that mimic the healthy human stomach epithelium. Similar to organoids, mucosoids are stem cell-driven cultures; however, the cells are cultivated on transwells at air-liquid interface. The epithelial cells of mucosoids form a polarized monolayer, allowing differentiation into all stomach lineages, including mucus-secreting cells. This setup facilitates the secretion and accumulation of mucus on the apical side of the mucosoids, enabling analysis of its bactericidal effects and protein composition, including antimicrobial peptides. Our findings show that TNFα, IL1ß, and IFNγ induce the secretion of antimicrobials such as lactotransferrin, lipocalin2, complement component 3, and CXCL9 into the mucus. This antimicrobial-enriched mucus can partially eliminate Helicobacter pylori, a key stomach pathogen. The bactericidal activity depends on the concentration of each antimicrobial and their gene expression is higher in patients with inflammation and H.pylori-associated chronic gastritis. However, we also find that H. pylori infection can reduce the expression of antimicrobial encoding genes promoted by inflammation. These findings suggest that controlling antimicrobial secretion in the mucus is a critical component of epithelial immunity. However, pathogens like H. pylori can overcome these defenses and survive in the mucosa.


Asunto(s)
Péptidos Antimicrobianos , Mucosa Gástrica , Helicobacter pylori , Inflamación , Moco , Humanos , Moco/metabolismo , Moco/microbiología , Péptidos Antimicrobianos/metabolismo , Mucosa Gástrica/microbiología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/inmunología , Inflamación/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/metabolismo , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/inmunología , Estómago/microbiología , Organoides/metabolismo , Organoides/microbiología
3.
Dev Comp Immunol ; 161: 105242, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39128619

RESUMEN

Molting is a key biological process of crustaceans, which is mainly regulated by 20-hydroxyecdyone (20E). The molting cycle could be divided into three main stages including pre-molt, post-molt and inter-molt stages. The mechanism of immune regulation during molting process still requires further exploration. Yorkie (Yki) is a pivotal transcription factor in the Hippo signaling pathway, and it plays an essential role in regulating cell growth and immune response. In the present study, a Yki gene was identified from Eriocheir sinensis (designed as EsYki), and the regulatory role of EsYki in controlling the expression of antimicrobial peptide genes throughout the molting process was investigated. The mRNA expression level of EsYki was higher at the pre-molt stage compared to the post-molt stage and inter-molt stage. Following the injection of 20E, there was a notable and consistent rise in the EsYki mRNA expression in haemocytes. The increase was observed from 3 h to 48 h with the maximum level at 12 h. And the phosphorylation of Yki in the haemocytes was also significantly up-regulated at 3 h post 20E injection. Moreover, the levels of EsYki mRNA expression at three molting stages were significantly increased post Aeromonas hydrophila stimulation. The maximum level was detected at post-molt stage following A. hydrophila stimulation, while the lowest level was observed at inter-molt stage. The expression pattern of EsCrus was in contrast to EsCrus. After EsYki mRNA transcripts were inhibited by Yki inhibitor (CA3), the mRNA expression levels of EsCrus1 and EsCrus2 following A. hydrophila stimulation were significantly elevated. Furthermore, the phosphorylation level of NF-κB was also increased following the inhibition of Yki. Collectively, our findings indicated that EsYki could be induced by 20E and has a suppressive effect on the expression of EsCrus via inhibiting NF-κB during molting process. This research contributes to the understanding of the immunological regulation mechanism during molting process in crustaceans.


Asunto(s)
Aeromonas hydrophila , Proteínas de Artrópodos , Braquiuros , Hemocitos , Muda , Animales , Braquiuros/inmunología , Braquiuros/genética , Proteínas de Artrópodos/metabolismo , Proteínas de Artrópodos/genética , Hemocitos/metabolismo , Hemocitos/inmunología , Aeromonas hydrophila/fisiología , Aeromonas hydrophila/inmunología , Proteínas Señalizadoras YAP/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Transactivadores/genética , Péptidos Antimicrobianos/metabolismo , Péptidos Antimicrobianos/genética , Ecdisterona/metabolismo , Regulación del Desarrollo de la Expresión Génica , Inmunidad Innata
4.
Front Cell Infect Microbiol ; 14: 1450353, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39193502

RESUMEN

Ticks are obligate hematophagous arthropods that transmit a wide range of pathogens to humans as well as wild and domestic animals. They also harbor a non-pathogenic microbiota, although our previous study has shown that the diverse bacterial microbiome in the midgut of Ixodes ricinus is quantitatively poor and lacks a core. In artificial infections by capillary feeding of ticks with two model bacteria (Gram-positive Micrococcus luteus and Gram-negative Pantoea sp.), rapid clearance of these microbes from the midgut was observed, indicating the presence of active immune mechanisms in this organ. In the current study, RNA-seq analysis was performed on the midgut of I. ricinus females inoculated with either M. luteus or Pantoea sp. or with sterile water as a control. While no immune-related transcripts were upregulated by microbial inoculation compared to that of the sterile control, capillary feeding itself triggered dramatic transcriptional changes in the tick midgut. Manual curation of the transcriptome from the midgut of unfed I. ricinus females, complemented by the proteomic analysis, revealed the presence of several constitutively expressed putative antimicrobial peptides (AMPs) that are independent of microbial stimulation and are referred to here as 'guard' AMPs. These included two types of midgut-specific defensins, two different domesticated amidase effector 2 (Dae2), microplusin/ricinusin-related molecules, two lysozymes, and two gamma interferon-inducible lysosomal thiol reductases (GILTs). The in vitro antimicrobial activity assays of two synthetic mature defensins, defensin 1 and defensin 8, confirmed their specificity against Gram-positive bacteria showing exceptional potency to inhibit the growth of M. luteus at nanomolar concentrations. The antimicrobial activity of midgut defensins is likely part of a multicomponent system responsible for the rapid clearance of bacteria in the tick midgut. Further studies are needed to evaluate the role of other identified 'guard' AMPs in controlling microorganisms entering the tick midgut.


Asunto(s)
Ixodes , Animales , Ixodes/microbiología , Ixodes/inmunología , Femenino , Micrococcus luteus/inmunología , Péptidos Antimicrobianos/metabolismo , Transcriptoma , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/inmunología , Perfilación de la Expresión Génica , Proteómica
5.
PLoS Pathog ; 20(8): e1012447, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39133760

RESUMEN

Overnutrition with dietary sugar can worsen infection outcomes in diverse organisms including insects and humans, through generally unknown mechanisms. In the present study, we show that adult Drosophila melanogaster fed high-sugar diets became more susceptible to infection by the Gram-negative bacteria Providencia rettgeri and Serratia marcescens. We found that P. rettgeri and S. marcescens proliferate more rapidly in D. melanogaster fed a high-sugar diet, resulting in increased probability of host death. D. melanogaster become hyperglycemic on the high-sugar diet, and we find evidence that the extra carbon availability may promote S. marcescens growth within the host. However, we found no evidence that increased carbon availability directly supports greater P. rettgeri growth. D. melanogaster on both diets fully induce transcription of antimicrobial peptide (AMP) genes in response to infection, but D. melanogaster provided with high-sugar diets show reduced production of AMP protein. Thus, overnutrition with dietary sugar may impair host immunity at the level of AMP translation. Our results demonstrate that dietary sugar can shape infection dynamics by impacting both host and pathogen, depending on the nutritional requirements of the pathogen and by altering the physiological capacity of the host to sustain an immune response.


Asunto(s)
Drosophila melanogaster , Animales , Drosophila melanogaster/microbiología , Providencia , Serratia marcescens/patogenicidad , Azúcares de la Dieta/efectos adversos , Susceptibilidad a Enfermedades , Infecciones por Serratia/microbiología , Infecciones por Enterobacteriaceae/microbiología , Péptidos Antimicrobianos/metabolismo
6.
Funct Integr Genomics ; 24(5): 142, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39187716

RESUMEN

Parsley is a commonly cultivated Apiaceae species of culinary and medicinal importance. Parsley has several recognized health benefits and the species has been utilized in traditional medicine since ancient times. Although parsley is among the most commonly cultivated members of Apiaceae, no systematic genomic research has been conducted on parsley. In the present work, parsley genome was sequenced using the long-read HiFi (high fidelity) sequencing technology and a draft contig assembly of 1.57 Gb that represents 80.9% of the estimated genome size was produced. The assembly was highly repeat-rich with a repetitive DNA content of 81%. The assembly was phased into a primary and alternate assembly in order to minimize redundant contigs. Scaffolds were constructed with the primary assembly contigs, which were used for the identification of AMP (antimicrobial peptide) genes. Characteristic AMP domains and 3D structures were used to detect and verify antimicrobial peptides. As a result, 23 genes (PcAMP1-23) representing defensin, snakin, thionin, lipid transfer protein and vicilin-like AMP classes were identified. Bioinformatic analyses for the characterization of peptide physicochemical properties indicated that parsley AMPs are extracellular peptides, therefore, plausibly exert their antimicrobial effects through the most commonly described AMP action mechanism of membrane attack. AMPs are attracting increasing attention since they display their fast antimicrobial effects in small doses on both plant and animal pathogens with a significantly reduced risk of resistance development. Therefore, identification and characterization of AMPs is important for their incorporation into plant disease management protocols as well as medicinal research for the treatment of multi-drug resistant infections.


Asunto(s)
Petroselinum , Petroselinum/genética , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/metabolismo , Péptidos Antimicrobianos/química , Secuenciación Completa del Genoma , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Genoma de Planta
7.
Int J Biol Macromol ; 278(Pt 3): 134898, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39173793

RESUMEN

Edwardsiella tarda is an intracellular pathogenic bacteria that can imperil the health of farmed fish. However, the interactive networks of immune regulation and metabolic response in E. tarda-infected fish are still unclear. In this investigation, we aimed to explore immunometabolic interplay in crucian carp after E. tarda infection by utilizing multiomics analyses. Crucian carp (Carassius auratus) receiving E. tarda infection showed increased levels of tissue damage and oxidative injury in liver. Multiomics analyses suggested that carbon and amino acid metabolism may be considered as crucial metabolic pathways in liver of crucian carp following E. tarda infection, while spaglumic acid, isocitric acid and tetrahydrocortisone were the crucial liver biomarkers. After that, a potential antimicrobial peptide (AMP) sequence called apolipoprotein D (ApoD) was identified from omics study. Then, tissue-specific analysis indicated that liver CaApoD showed the highest expression among isolated tissues. After Aeromonas hydrophila stimulated, CaApoD expressions increased sharply in immune-related tissues. Moreover, CaApoD fusion protein could mediate the in vitro binding to A. hydrophila and E. tarda, attenuate bacterial growth as well as diminish bacterial biofilm forming activity. These findings may have a comprehensive implication for understanding immunometabolic response in crucian carp upon infection.


Asunto(s)
Apolipoproteínas D , Carpas , Edwardsiella tarda , Infecciones por Enterobacteriaceae , Enfermedades de los Peces , Hígado , Animales , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Apolipoproteínas D/metabolismo , Apolipoproteínas D/genética , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/veterinaria , Infecciones por Enterobacteriaceae/microbiología , Carpas/microbiología , Carpas/inmunología , Carpas/metabolismo , Hígado/metabolismo , Carpa Dorada/inmunología , Carpa Dorada/microbiología , Carpa Dorada/metabolismo , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/metabolismo , Péptidos Antimicrobianos/genética , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Proteínas de Peces/inmunología , Multiómica
8.
PLoS Pathog ; 20(8): e1012488, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39186812

RESUMEN

Antimicrobial peptides (AMPs) are a promising tool with which to fight rising antibiotic resistance. However, pathogenic bacteria are equipped with several AMP defense mechanisms, whose contributions to AMP resistance are often poorly defined. Here, we evaluate the genetic determinants of resistance to an insect AMP, cecropin B, in the opportunistic pathogen Enterobacter cloacae. Single-cell analysis of E. cloacae's response to cecropin revealed marked heterogeneity in cell survival, phenotypically reminiscent of heteroresistance (the ability of a subpopulation to grow in the presence of supra-MIC concentration of antimicrobial). The magnitude of this response was highly dependent on initial E. cloacae inoculum. We identified 3 genetic factors which collectively contribute to E. cloacae resistance in response to the AMP cecropin: The PhoPQ-two-component system, OmpT-mediated proteolytic cleavage of cecropin, and Rcs-mediated membrane stress response. Altogether, our data suggest that multiple, independent mechanisms contribute to AMP resistance in E. cloacae.


Asunto(s)
Péptidos Antimicrobianos , Enterobacter cloacae , Enterobacter cloacae/genética , Enterobacter cloacae/metabolismo , Enterobacter cloacae/efectos de los fármacos , Péptidos Antimicrobianos/metabolismo , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/genética , Farmacorresistencia Bacteriana , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Animales , Proteínas de Insectos/metabolismo , Proteínas de Insectos/genética , Infecciones por Enterobacteriaceae/microbiología
9.
Food Chem ; 460(Pt 2): 140646, 2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39089018

RESUMEN

The study aimed to mine and characterize novel antimicrobial peptides (AMPs) from the Shanxi aged vinegar microbiome. Utilizing machine learning techniques, AlphaFold2 structure prediction and molecular dynamics simulations, six novel AMPs were innovatively mined from 98,539 peptides based on metagenomic data, of which one peptide secreted by Lactobacillus (named La-AMP) was experimentally validated to have remarkable bactericidal effects against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) with high stability and no hemolytic activity. Scanning electron microscopy revealed that La-AMP caused irreversible damage to cell membranes of S. aureus and E. coli, a finding further confirmed by calcein-AM/propidium iodide staining. Additionally, La-AMP induced nucleic acid leakage and reactive oxygen species accumulation in bacterial cells. It was found to bind to DNA gyrase through salt bridges, hydrogen bonds, and hydrophobic interactions, ultimately inducing apoptosis. Thus, La-AMP exhibited encouraging promise as a valuable bioactive component for the development of natural preservatives.


Asunto(s)
Ácido Acético , Escherichia coli , Metagenómica , Simulación de Dinámica Molecular , Staphylococcus aureus , Staphylococcus aureus/efectos de los fármacos , Ácido Acético/química , Ácido Acético/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/metabolismo , Péptidos Antimicrobianos/genética , Antibacterianos/farmacología , Antibacterianos/química , Microbiota , Pruebas de Sensibilidad Microbiana , Humanos , Lactobacillus/química , Lactobacillus/metabolismo
10.
Nat Commun ; 15(1): 5778, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987259

RESUMEN

Antimicrobial proteins contribute to host-microbiota interactions and are associated with inflammatory bowel disease (IBD), but our understanding on antimicrobial protein diversity and functions remains incomplete. Ribonuclease 4 (Rnase4) is a potential antimicrobial protein with no known function in the intestines. Here we find that RNASE4 is expressed in intestinal epithelial cells (IEC) including Paneth and goblet cells, and is detectable in human and mouse stool. Results from Rnase4-deficient mice and recombinant protein suggest that Rnase4 kills Parasutterella to modulate intestinal microbiome, thereby enhancing indoleamine-2,3-dioxygenase 1 (IDO1) expression and subsequently kynurenic and xanthurenic acid production in IECs to reduce colitis susceptibility. Furthermore, deceased RNASE4 levels are observed in the intestinal tissues and stool from patients with IBD, correlating with increased stool Parasutterella. Our results thus implicate Rnase4 as an intestinal antimicrobial protein regulating gut microbiota and metabolite homeostasis, and as a potential diagnostic biomarker and therapeutic target for IBD.


Asunto(s)
Microbioma Gastrointestinal , Homeostasis , Enfermedades Inflamatorias del Intestino , Ratones Endogámicos C57BL , Animales , Femenino , Humanos , Masculino , Ratones , Péptidos Antimicrobianos/metabolismo , Colitis/microbiología , Colitis/metabolismo , Colitis/inducido químicamente , Heces/microbiología , Microbioma Gastrointestinal/fisiología , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestinos/microbiología , Ratones Noqueados , Ribonucleasas/metabolismo
11.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39000180

RESUMEN

The antimicrobial peptide LRGG (LLRLLRRGGRRLLRLL-NH2) was designed and chemically synthesized in a study conducted by Jia et al. Gram-negative bacteria were found to be sensitive to LRGG and exhibited a high therapeutic index. Genetic engineering methods were used to create the prokaryotic fusion expression vector pQE-GFP-LRGG, and the resulting corresponding fusion protein GFP-LRGG was subsequently expressed and purified. The precursor GFP was then removed by TEV proteolysis, and pure LRGG was obtained after another round of purification and endotoxin removal. The prokaryotic-expressed antimicrobial peptide LRGG displays a broad-spectrum antibacterial effect on Gram-negative bacteria, and its minimum inhibitory activity (MIC) against Escherichia coli can reach 2 µg/mL. Compared to the chemically synthesized LRGG, the prokaryotic-expressed LRGG exhibits similar temperature, pH, salt ion, serum stability, and cell selectivity. Furthermore, prokaryotic-expressed LRGG showed excellent therapeutic effects in both the infection model of cell selectivity and no embryotoxicity in a Galleria mellonella infection model. The mechanism by which LRGG causes bacterial death was found to be the disruption of the Gram-negative cell membrane.


Asunto(s)
Péptidos Antimicrobianos , Pruebas de Sensibilidad Microbiana , Animales , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , Bacterias Gramnegativas/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Catiónicos Antimicrobianos/genética , Humanos
12.
Chem Soc Rev ; 53(17): 8713-8763, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39041297

RESUMEN

Amyloid peptides (AMYs) and antimicrobial peptides (AMPs) are considered as the two distinct families of peptides, characterized by their unique sequences, structures, biological functions, and specific pathological targets. However, accumulating evidence has revealed intriguing pathological connections between these peptide families in the context of microbial infection and neurodegenerative diseases. Some AMYs and AMPs share certain structural and functional characteristics, including the ability to self-assemble, the presence of ß-sheet-rich structures, and membrane-disrupting mechanisms. These shared features enable AMYs to possess antimicrobial activity and AMPs to acquire amyloidogenic properties. Despite limited studies on AMYs-AMPs systems, the cross-seeding phenomenon between AMYs and AMPs has emerged as a crucial factor in the bidirectional communication between the pathogenesis of neurodegenerative diseases and host defense against microbial infections. In this review, we examine recent developments in the potential interplay between AMYs and AMPs, as well as their pathological implications for both infectious and neurodegenerative diseases. By discussing the current progress and challenges in this emerging field, this account aims to inspire further research and investments to enhance our understanding of the intricate molecular crosstalk between AMYs and AMPs. This knowledge holds great promise for the development of innovative therapies to combat both microbial infections and neurodegenerative disorders.


Asunto(s)
Péptidos Antimicrobianos , Enfermedades Neurodegenerativas , Humanos , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Proteínas Amiloidogénicas/metabolismo , Proteínas Amiloidogénicas/química , Proteínas Amiloidogénicas/antagonistas & inhibidores , Animales , Antiinfecciosos/química , Antiinfecciosos/farmacología
13.
Proc Natl Acad Sci U S A ; 121(25): e2401802121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38865264

RESUMEN

The spatial organization of gut microbiota is crucial for the functioning of the gut ecosystem, although the mechanisms that organize gut bacterial communities in microhabitats are only partially understood. The gut of the insect Riptortus pedestris has a characteristic microbiota biogeography with a multispecies community in the anterior midgut and a monospecific bacterial population in the posterior midgut. We show that the posterior midgut region produces massively hundreds of specific antimicrobial peptides (AMPs), the Crypt-specific Cysteine-Rich peptides (CCRs) that have membrane-damaging antimicrobial activity against diverse bacteria but posterior midgut symbionts have elevated resistance. We determined by transposon-sequencing the genetic repertoire in the symbiont Caballeronia insecticola to manage CCR stress, identifying different independent pathways, including AMP-resistance pathways unrelated to known membrane homeostasis functions as well as cell envelope functions. Mutants in the corresponding genes have reduced capacity to colonize the posterior midgut, demonstrating that CCRs create a selective barrier and resistance is crucial in gut symbionts. Moreover, once established in the gut, the bacteria differentiate into a CCR-sensitive state, suggesting a second function of the CCR peptide arsenal in protecting the gut epithelia or mediating metabolic exchanges between the host and the gut symbionts. Our study highlights the evolution of an extreme diverse AMP family that likely contributes to establish and control the gut microbiota.


Asunto(s)
Péptidos Antimicrobianos , Microbioma Gastrointestinal , Simbiosis , Animales , Péptidos Antimicrobianos/metabolismo , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/farmacología , Bacterias/genética , Bacterias/metabolismo , Bacterias/efectos de los fármacos , Tracto Gastrointestinal/microbiología
14.
Nat Commun ; 15(1): 4901, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38851779

RESUMEN

Antimicrobial resistance remains a significant global threat, driving up mortality rates worldwide. Ribosomally synthesized and post-translationally modified peptides have emerged as a promising source of novel peptide antibiotics due to their diverse chemical structures. Here, we report the discovery of new aminovinyl-(methyl)cysteine (Avi(Me)Cys)-containing peptide antibiotics through a synergistic approach combining biosynthetic rule-based omics mining and heterologous expression. We first bioinformatically identify 1172 RiPP biosynthetic gene clusters (BGCs) responsible for Avi(Me)Cys-containing peptides formation from a vast pool of over 50,000 bacterial genomes. Subsequently, we successfully establish the connection between three identified BGCs and the biosynthesis of five peptide antibiotics via biosynthetic rule-guided metabolic analysis. Notably, we discover a class V lanthipeptide, massatide A, which displays excellent activity against gram-positive pathogens, including drug-resistant clinical isolates like linezolid-resistant S. aureus and methicillin-resistant S. aureus, with a minimum inhibitory concentration of 0.25 µg/mL. The remarkable performance of massatide A in an animal infection model, coupled with a relatively low risk of resistance and favorable safety profile, positions it as a promising candidate for antibiotic development. Our study highlights the potential of Avi(Me)Cys-containing peptides in expanding the arsenal of antibiotics against multi-drug-resistant bacteria, offering promising drug leads in the ongoing battle against infectious diseases.


Asunto(s)
Antibacterianos , Staphylococcus aureus Resistente a Meticilina , Pruebas de Sensibilidad Microbiana , Animales , Antibacterianos/farmacología , Antibacterianos/química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/química , Humanos , Familia de Multigenes , Ratones , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Farmacorresistencia Bacteriana/genética , Farmacorresistencia Bacteriana/efectos de los fármacos , Genoma Bacteriano/genética , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Biología Computacional/métodos , Cisteína/metabolismo , Cisteína/química
15.
Sci Rep ; 14(1): 13818, 2024 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879638

RESUMEN

The hematophagous common bed bug, Cimex lectularius, is not known to transmit human pathogens outside laboratory settings, having evolved various immune defense mechanisms including the expression of antimicrobial peptides (AMPs). We unveil three novel prolixicin AMPs in bed bugs, exhibiting strong homology to the prolixicin of kissing bugs, Rhodnius prolixus, and to diptericin/attacin AMPs. We demonstrate for the first time sex-specific and immune mode-specific upregulation of these prolixicins in immune organs, the midgut and rest of body, following injection and ingestion of Gr+ (Bacillus subtilis) and Gr- (Escherichia coli) bacteria. Synthetic CL-prolixicin2 significantly inhibited growth of E. coli strains and killed or impeded Trypanosoma cruzi, the Chagas disease agent. Our findings suggest that prolixicins are regulated by both IMD and Toll immune pathways, supporting cross-talk and blurred functional differentiation between major immune pathways. The efficacy of CL-prolixicin2 against T. cruzi underscores the potential of AMPs in Chagas disease management.


Asunto(s)
Chinches , Escherichia coli , Trypanosoma cruzi , Animales , Trypanosoma cruzi/efectos de los fármacos , Chinches/microbiología , Chinches/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Bacillus subtilis/metabolismo , Bacillus subtilis/efectos de los fármacos , Femenino , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/metabolismo , Masculino , Enfermedad de Chagas/parasitología , Proteínas de Insectos/metabolismo , Secuencia de Aminoácidos
16.
Anim Sci J ; 95(1): e13972, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38923622

RESUMEN

High grain feeding or weaning, which could compromise the rumen epithelium by increasing ruminal short-chain fatty acid (SCFA) concentrations with pH reduction, is associated with high levels of ruminal toll-like receptor 5 (TLR5). This study aimed to determine the role of TLR5 in the rumen epithelium. Immunohistochemistry revealed that TLR5 was localized in cells on the basal side (i.e., basal and spinous layers) rather than in the granular layer in the rumen epithelium, where tight junctions are most potent, in pre- and post-weaning calves (n = 9). Primary bovine rumen epithelial cells (BRECs) obtained from Holstein cows (n = 3) were cultured to investigate the factors that upregulate TLR5; however, SCFA, low pH (pH 5.6), BHBA, L-lactate, D-lactate, and LPS did not upregulate TLR5 gene expression in BREC. Primary BREC treated with flagellin (TLR5 ligand) had higher expression of interleukin-1ß (IL-1ß) (P < 0.05) than BREC treated with vehicle. In addition, BREC treated with IL-1ß had higher expression of antimicrobial peptides and C-X-C motif chemokine ligand 8 than BREC treated with vehicle (P < 0.05). These results suggest that ruminal TLR5 may recognize epithelial disruption via flagellin and mediate the immune response via IL-1ß during high-grain feeding or weaning.


Asunto(s)
Células Epiteliales , Expresión Génica , Interleucina-1beta , Interleucina-8 , Rumen , Receptor Toll-Like 5 , Animales , Receptor Toll-Like 5/genética , Receptor Toll-Like 5/metabolismo , Rumen/metabolismo , Bovinos/metabolismo , Células Epiteliales/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Células Cultivadas , Interleucina-8/metabolismo , Interleucina-8/genética , Destete , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Flagelina/farmacología , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/farmacología , Ligandos , Regulación hacia Arriba
17.
Med Microbiol Immunol ; 213(1): 9, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38900248

RESUMEN

Endogenous antimicrobial peptides (AMPs) play a key role in the host defense against pathogens. AMPs attack pathogens preferentially at the site of entry to prevent invasive infection. Mycobacterium tuberculosis (Mtb) enters its host via the airways. AMPs released into the airways are therefore likely candidates to contribute to the clearance of Mtb immediately after infection. Since lysozyme is detectable in airway secretions, we evaluated its antimicrobial activity against Mtb. We demonstrate that lysozyme inhibits the growth of extracellular Mtb, including isoniazid-resistant strains. Lysozyme also inhibited the growth of non-tuberculous mycobacteria. Even though lysozyme entered Mtb-infected human macrophages and co-localized with the pathogen we did not observe antimicrobial activity. This observation was unlikely related to the large size of lysozyme (14.74 kDa) because a smaller lysozyme-derived peptide also co-localized with Mtb without affecting the viability. To evaluate whether the activity of lysozyme against extracellular Mtb could be relevant in vivo, we incubated Mtb with fractions of human serum and screened for antimicrobial activity. After several rounds of sub-fractionation, we identified a highly active fraction-component as lysozyme by mass spectrometry. In summary, our results identify lysozyme as an antimycobacterial protein that is detectable as an active compound in human serum. Our results demonstrate that the activity of AMPs against extracellular bacilli does not predict efficacy against intracellular pathogens despite co-localization within the macrophage. Ongoing experiments are designed to unravel peptide modifications that occur in the intracellular space and interfere with the deleterious activity of lysozyme in the extracellular environment.


Asunto(s)
Macrófagos , Muramidasa , Mycobacterium tuberculosis , Muramidasa/farmacología , Muramidasa/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/microbiología , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/metabolismo , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos
18.
Fish Shellfish Immunol ; 151: 109735, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38945414

RESUMEN

Antimicrobial peptide (AMP) is an important component of crustaceans' innate immune system. In this study, a short neuropeptide F (sNPF) gene (Pc-sNPF) and a Forkhead box O (FOXO) gene (PcFOXO) from Procambarus clarkii were identified. Analysis findings showed that the expression level of AMP genes differed between male and female P. clarkii. Furthermore, Pc-sNPF and PcFOXO were related to the sex dimorphism of AMP. Knockdown of Pc-sNPF in the eyestalk significantly upregulated the expression of PcFOXO and two anti-lipopolysaccharide factors (PcALF4 and PcALFL) in the intestine of P. clarkii. The expression of PcFOXO in the intestine of female P. clarkii was higher than in that of males. Results from RNA interference revealed that PcFOXO positively regulated the expression of PcALF4 and PcALFL in the intestine of male and female P. clarkii. In summary, our study showed that differences in Pc-sNPF expression in eyestalk of male and female P. clarkii leading to sex dimorphism of AMP expression in the intestine are mediated by the sNPF-FOXO-AMP signal pathway called the eyestalk-intestine axis.


Asunto(s)
Proteínas de Artrópodos , Regulación de la Expresión Génica , Neuropéptidos , Caracteres Sexuales , Animales , Masculino , Femenino , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/inmunología , Proteínas de Artrópodos/metabolismo , Regulación de la Expresión Génica/inmunología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Astacoidea/genética , Astacoidea/inmunología , Intestinos , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Inmunidad Innata/genética , Filogenia , Perfilación de la Expresión Génica , Secuencia de Aminoácidos , Alineación de Secuencia
19.
Mar Biotechnol (NY) ; 26(4): 696-715, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38922559

RESUMEN

Antimicrobial peptides (AMPs), including beta-defensin from fish, are a crucial class of peptide medicines. The focus of the current study is the molecular and functional attributes of CmDef, a 63-amino acid beta-defensin AMP from Malabar trevally, Carangoides malabaricus. This peptide demonstrated typical characteristics of AMPs, including hydrophobicity, amphipathic nature, and +2.8 net charge. The CmDef was recombinantly expressed and the recombinant peptide, rCmDef displayed a strong antimicrobial activity against bacterial fish pathogens with an MIC of 8 µM for V. proteolyticus and 32 µM for A. hydrophila. The E. tarda and V. harveyi showed an inhibition of 94% and 54%, respectively, at 32 µM concentration. No activity was observed against V. fluvialis and V. alginolyticus. The rCmDef has a multimode of action that exerts an antibacterial effect by membrane depolarization followed by membrane permeabilization and ROS production. rCmDef also exhibited anti-cancer activities in silico without causing hemolysis. The peptide demonstrated stability under various conditions, including different pH levels, temperatures, salts, and metal ions (KCl and CaCl2), and remained stable in the presence of proteases such as trypsin and proteinase K at concentrations up to 0.2 µg/100 µl. The strong antibacterial efficacy and non-cytotoxic nature suggest that rCmDef is a single-edged sword that can contribute significantly to aquaculture disease management.


Asunto(s)
Proteínas Recombinantes , beta-Defensinas , Animales , beta-Defensinas/farmacología , beta-Defensinas/genética , beta-Defensinas/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Proteínas de Peces/química , Proteínas de Peces/farmacología , Pruebas de Sensibilidad Microbiana , Vibrio/efectos de los fármacos , Secuencia de Aminoácidos , Humanos , Enfermedades de los Peces/microbiología , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Hemólisis/efectos de los fármacos
20.
Nat Commun ; 15(1): 3945, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730238

RESUMEN

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.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Simulación de Dinámica Molecular , Ribosomas , Ribosomas/metabolismo , Péptidos Catiónicos Antimicrobianos/metabolismo , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Biosíntesis de Proteínas , Sitios de Unión , Microscopía por Crioelectrón , Escherichia coli/metabolismo , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Factores de Terminación de Péptidos/metabolismo , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/genética , Unión Proteica , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/metabolismo , Péptidos Antimicrobianos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA