Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.109
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 86: 799-823, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28426241

ABSTRACT

Iron is essential for the survival of most bacteria but presents a significant challenge given its limited bioavailability. Furthermore, the toxicity of iron combined with the need to maintain physiological iron levels within a narrow concentration range requires sophisticated systems to sense, regulate, and transport iron. Most bacteria have evolved mechanisms to chelate and transport ferric iron (Fe3+) via siderophore receptor systems, and pathogenic bacteria have further lowered this barrier by employing mechanisms to utilize the host's hemoproteins. Once internalized, heme is cleaved by both oxidative and nonoxidative mechanisms to release iron. Heme, itself a lipophilic and toxic molecule, presents a significant challenge for transport into the cell. As such, pathogenic bacteria have evolved sophisticated cell surface signaling and transport systems to obtain heme from the host. In this review, we summarize the structure and function of the heme-sensing and transport systems of pathogenic bacteria and the potential of these systems as antimicrobial targets.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Cell Membrane/drug effects , Heme/antagonists & inhibitors , Iron/metabolism , Pseudomonas aeruginosa/drug effects , Receptors, Cell Surface/antagonists & inhibitors , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport/drug effects , Cell Membrane/metabolism , Cell Wall/drug effects , Cell Wall/metabolism , Gene Expression , Heme/metabolism , Metalloporphyrins/chemical synthesis , Metalloporphyrins/pharmacology , Models, Molecular , Protein Conformation , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Siderophores/antagonists & inhibitors , Siderophores/biosynthesis , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism
2.
Cell ; 151(5): 1005-16, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23178120

ABSTRACT

Phage G1 gp67 is a 23 kDa protein that binds to the Staphylococcus aureus (Sau) RNA polymerase (RNAP) σ(A) subunit and blocks cell growth by inhibiting transcription. We show that gp67 has little to no effect on transcription from most promoters but is a potent inhibitor of ribosomal RNA transcription. A 2.0-Å-resolution crystal structure of the complex between gp67 and Sau σ(A) domain 4 (σ(A)(4)) explains how gp67 joins the RNAP promoter complex through σ(A)(4) without significantly affecting σ(A)(4) function. Our results indicate that gp67 forms a complex with RNAP at most, if not all, σ(A)-dependent promoters, but selectively inhibits promoters that depend on an interaction between upstream DNA and the RNAP α-subunit C-terminal domain (αCTD). Thus, we reveal a promoter-specific transcription inhibition mechanism by which gp67 interacts with the RNAP promoter complex through one subunit (σ(A)), and selectively affects the function of another subunit (αCTD) depending on promoter usage.


Subject(s)
Growth Inhibitors/metabolism , Myoviridae/metabolism , Promoter Regions, Genetic , Staphylococcus aureus/growth & development , Staphylococcus aureus/virology , Viral Proteins/metabolism , Base Sequence , Crystallography, X-Ray , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Models, Molecular , Molecular Sequence Data , RNA, Bacterial/genetics , RNA, Ribosomal/genetics , Sigma Factor/metabolism , Staphylococcus aureus/genetics , Transcription, Genetic
3.
EMBO J ; 40(14): e107500, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34046916

ABSTRACT

The Staphylococcal Bap proteins sense environmental signals (such as pH, [Ca2+ ]) to build amyloid scaffold biofilm matrices via unknown mechanisms. We here report the crystal structure of the aggregation-prone region of Staphylococcus aureus Bap which adopts a dumbbell-shaped fold. The middle module (MM) connecting the N-terminal and C-terminal lobes consists of a tandem of novel double-Ca2+ -binding motifs involved in cooperative interaction networks, which undergoes Ca2+ -dependent order-disorder conformational switches. The N-terminal lobe is sufficient to mediate amyloid aggregation through liquid-liquid phase separation and maturation, and subsequent biofilm formation under acidic conditions. Such processes are promoted by disordered MM at low [Ca2+ ] but inhibited by ordered MM stabilized by Ca2+ binding, with inhibition efficiency depending on structural integrity of the interaction networks. These studies illustrate a novel protein switch in pathogenic bacteria and provide insights into the mechanistic understanding of Bap proteins in modulation of functional amyloid and biofilm formation, which could be implemented in the anti-biofilm drug design.


Subject(s)
Amyloid/metabolism , Bacterial Proteins/metabolism , Biofilms/growth & development , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Calcium/metabolism , Cell Aggregation/physiology
4.
BMC Microbiol ; 24(1): 212, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877418

ABSTRACT

BACKGROUND: Long-term treatment with trimethoprim-sulfamethoxazole (SXT) can lead to the formation of small-colony variants (SCVs) of Staphylococcus aureus. However, the mechanism behind SCVs formation remains poorly understood. In this study, we explored the phenotype and omics-based characterization of S. aureus SCVs induced by SXT and shed light on the potential causes of SCV formation. METHODS: Stable SCVs were obtained by continuously treating S. aureus isolates using 12/238 µg/ml of SXT, characterized by growth kinetics, antibiotic susceptibility testing, and auxotrophism test. Subsequently, a pair of representative strains (SCV and its parental strain) were selected for genomic, transcriptomic and metabolomic analysis. RESULTS: Three stable S. aureus SCVs were successfully screened and proven to be homologous to their corresponding parental strains. Phenotypic tests showed that all SCVs were non-classical mechanisms associated with impaired utilization of menadione, heme and thymine, and exhibited slower growth and higher antibiotic minimum inhibitory concentrations (MICs), compared to their corresponding parental strains. Genomic data revealed 15 missense mutations in 13 genes in the representative SCV, which were involved in adhesion, intramolecular phosphate transfer on ribose, transport pathways, and phage-encoded proteins. The combination analysis of transcriptome and metabolome identified 35 overlapping pathways possible associated with the phenotype switching of S. aureus. These pathways mainly included changes in metabolism, such as purine metabolism, pyruvate metabolism, amino acid metabolism, and ABC transporters, which could play a crucial role in promoting SCVs development by affecting nucleic acid synthesis and energy metabolism in bacteria. CONCLUSION: This study provides profound insights into the causes of S. aureus SCV formation induced by SXT. The findings may offer valuable clues for developing new strategies to combat S. aureus SCV infections.


Subject(s)
Anti-Bacterial Agents , Microbial Sensitivity Tests , Staphylococcus aureus , Trimethoprim, Sulfamethoxazole Drug Combination , Staphylococcus aureus/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Trimethoprim, Sulfamethoxazole Drug Combination/pharmacology , Anti-Bacterial Agents/pharmacology , Metabolomics , Humans , Genomics , Phenotype , Staphylococcal Infections/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Transcriptome , Gene Expression Profiling , Multiomics
5.
Arch Microbiol ; 206(8): 350, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38995446

ABSTRACT

KKL-35 is a new oxadiazole compound with potent broad-spectrum antibacterial activity against a number of gram-positive and gram-negative bacteria. However, its influences on bacterial growth are unclear. This study is to investigate phenotypic changes of Staphylococcus aureus (SA) caused by KKL-35 and evaluate antibacterial activity of combinations of KKL-35 with 7 class of antibiotics available in medical facilities. KKL-35-treated SA showed significantly lower survival under stresses of NaCl and H2O2 than DMSO (21.03 ± 2.60% vs. 68.21 ± 5.31% for NaCl, 4.91 ± 3.14% vs. 74.78 ± 2.88% for H2O2). UV exposure significantly decreased survival of SA treated with KKL-35 than DMSO-treated ones (23.91 ± 0.71% vs. 55.45 ± 4.70% for 4.2 J/m2, 12.80 ± 1.03% vs. 31.99 ± 5.99% for 7.0 J/m2, 1.52 ± 0.63% vs. 6.49 ± 0.51% for 14.0 J/m2). KKL-35 significantly decreased biofilm formation (0.47 ± 0.12 vs. 1.45 ± 0.21) and bacterial survival in the serum resistance assay (42.27 ± 2.77% vs. 78.31 ± 5.64%) than DMSO. KKL-35 significantly decreased ethidium bromide uptake and efflux, as well as the cell membrane integrity. KKL-35 had low cytotoxicity and low propensity for resistance. KKL-35 inhibited SA growth in concentration-independent and time-dependent manners, and showed additivity when combined with the majority class of available antibiotics. Antibiotic combinations of KKL-35 with ciprofloxacin, rifampicin, or linezolid significantly decreased bacterial loads than the most active antibiotic in the corresponding combination. Thus, KKL-35 inhibits growth of SA by decreasing bacterial environmental adaptations, biofilm formation, membrane uptake and efflux, as well as increasing antibiotic sensitivity. Its potent antibacterial activity, low cytotoxicity, low propensity for resistance, and wide choices in antibiotic combinations make KKL-35 a promising leading compound to design new antibiotics in monotherapies and combination therapies to treat bacterial infections.


Subject(s)
Anti-Bacterial Agents , Biofilms , Microbial Sensitivity Tests , Oxadiazoles , Staphylococcus aureus , Humans , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Oxadiazoles/pharmacology , Phenotype , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development
6.
J Immunol ; 208(2): 454-463, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34930781

ABSTRACT

Inflammation involves a delicate balance between pathogen clearance and limiting host tissue damage, and perturbations in this equilibrium promote disease. Patients suffering from autoimmune diseases, such as systemic lupus erythematosus (SLE), have higher levels of serum S100A9 protein and increased risk for infection. S100A9 is highly abundant within neutrophils and modulates antimicrobial activity in response to bacterial pathogens. We reasoned that increased serum S100A9 in SLE patients reflects accumulation of S100A9 protein in neutrophils and may indicate altered neutrophil function. In this study, we demonstrate elevated S100A9 protein within neutrophils from SLE patients, and MRL/lpr mice associates with lower mitochondrial superoxide, decreased suicidal neutrophil extracellular trap formation, and increased susceptibility to Staphylococcus aureus infection. Furthermore, increasing mitochondrial superoxide production restored the antibacterial activity of MRL/lpr neutrophils in response to S. aureus These results demonstrate that accumulation of intracellular S100A9 associates with impaired mitochondrial homeostasis, thereby rendering SLE neutrophils inherently less bactericidal.


Subject(s)
Calgranulin B/blood , Extracellular Traps/immunology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Mitochondria/metabolism , Staphylococcus aureus/immunology , Animals , Disease Susceptibility/immunology , Female , Homeostasis/physiology , Humans , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/growth & development , Superoxides/metabolism
7.
Antonie Van Leeuwenhoek ; 117(1): 95, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38967683

ABSTRACT

The decline of new antibiotics and the emergence of multidrug resistance in pathogens necessitates a revisit of strategies used for lead compound discovery. This study proposes to induce the production of bioactive compounds with sub-lethal concentrations of silver nanoparticles (Ag-NPs). A total of Forty-two Actinobacteria isolates from four Saudi soil samples were grown with and without sub-lethal concentration of Ag-NPs (50 µg ml-1). The spent broth grown with Ag-NPs, or without Ag-NPs were screened for antimicrobial activity against four bacteria. Interestingly, out of 42 strains, broths of three strains grown with sub-lethal concentration of Ag-NPs exhibit antimicrobial activity against Staphylococcus aureus and Micrococcus luteus. Among these, two strains S4-4 and S4-21 identified as Streptomyces labedae and Streptomyces tirandamycinicus based on 16S rRNA gene sequence were selected for detailed study. The change in the secondary metabolites profile in the presence of Ag-NPs was evaluated using GC-MS and LC-MS analyses. Butanol extracts of spent broth grown with Ag-NPs exhibit strong antimicrobial activity against M. luteus and S. aureus. While the extracts of the controls with the same concentration of Ag-NPs do not show any activity. GC-analysis revealed a clear change in the secondary metabolite profile when grown with Ag-NPs. Similarly, the LC-MS patterns also differ significantly. Results of this study, strongly suggest that sub-lethal concentrations of Ag-NPs influence the production of secondary metabolites by Streptomyces. Besides, LC-MS results identified possible secondary metabolites, associated with oxidative stress and antimicrobial activities. This strategy can be used to possibly induce cryptic biosynthetic gene clusters for the discovery of new lead compounds.


Subject(s)
Anti-Bacterial Agents , Metal Nanoparticles , Microbial Sensitivity Tests , RNA, Ribosomal, 16S , Silver , Staphylococcus aureus , Streptomyces , Streptomyces/metabolism , Streptomyces/genetics , Silver/pharmacology , Silver/chemistry , Silver/metabolism , Metal Nanoparticles/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , RNA, Ribosomal, 16S/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Soil Microbiology , Secondary Metabolism , Micrococcus luteus/drug effects , Micrococcus luteus/growth & development , Drug Discovery
8.
Food Microbiol ; 122: 104561, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38839224

ABSTRACT

The current study aimed to determine if the 3D-printing speed and temperature would impact the transferability of foodborne pathogens from the stainless-steel (SS) food cartridge to the 3D-printed food ink. Staphylococcus aureus and Escherichia coli were inoculated onto the interior surface of the SS food cartridges. Subsequently, a model food ink was extruded with a recommended macronutrient contribution of 55.8, 23.7, and 20.5% of carbohydrates, proteins, and fat, respectively. The impact of 3D-printing temperatures and speeds on transfer rates was analysed using a Two-Way ANOVA. S. aureus was transferred more from the cartridge to the food ink with a population of 3.39, 2.98, and 3.09 log CFU/g compared to 2.03, 2.06, and 2.00 log CFU/g for E. coli at 2000, 3000, and 4000 mm/s printing speed, respectively, at 25 °C. A Kruskal-Wallis Test was employed to investigate the effect of different speeds and temperatures on the transferability of S. aureus and E. coli. Speed was the main factor affecting S. aureus transferability, while temperature (25 and 50 °C) had the greatest impact on E. coli transferability. This research seeks to advance the understanding of 3D-printing parameters in pathogen transferability and help the food industry move towards this technology's quick and safe adoption.


Subject(s)
Escherichia coli , Food Microbiology , Printing, Three-Dimensional , Staphylococcus aureus , Temperature , Staphylococcus aureus/growth & development , Escherichia coli/growth & development , Stainless Steel , Food Handling/instrumentation , Food Handling/methods , Food Contamination/analysis , Colony Count, Microbial
9.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Article in English | MEDLINE | ID: mdl-34716264

ABSTRACT

Bacterial cell wall peptidoglycan is essential, maintaining both cellular integrity and morphology, in the face of internal turgor pressure. Peptidoglycan synthesis is important, as it is targeted by cell wall antibiotics, including methicillin and vancomycin. Here, we have used the major human pathogen Staphylococcus aureus to elucidate both the cell wall dynamic processes essential for growth (life) and the bactericidal effects of cell wall antibiotics (death) based on the principle of coordinated peptidoglycan synthesis and hydrolysis. The death of S. aureus due to depletion of the essential, two-component and positive regulatory system for peptidoglycan hydrolase activity (WalKR) is prevented by addition of otherwise bactericidal cell wall antibiotics, resulting in stasis. In contrast, cell wall antibiotics kill via the activity of peptidoglycan hydrolases in the absence of concomitant synthesis. Both methicillin and vancomycin treatment lead to the appearance of perforating holes throughout the cell wall due to peptidoglycan hydrolases. Methicillin alone also results in plasmolysis and misshapen septa with the involvement of the major peptidoglycan hydrolase Atl, a process that is inhibited by vancomycin. The bactericidal effect of vancomycin involves the peptidoglycan hydrolase SagB. In the presence of cell wall antibiotics, the inhibition of peptidoglycan hydrolase activity using the inhibitor complestatin results in reduced killing, while, conversely, the deregulation of hydrolase activity via loss of wall teichoic acids increases the death rate. For S. aureus, the independent regulation of cell wall synthesis and hydrolysis can lead to cell growth, death, or stasis, with implications for the development of new control regimes for this important pathogen.


Subject(s)
Cell Wall/physiology , Peptidoglycan/metabolism , Staphylococcus aureus/growth & development , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Bacterial Proteins/metabolism , Cell Wall/metabolism , Homeostasis , Methicillin/pharmacology , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Teichoic Acids/metabolism , Vancomycin/pharmacology
10.
Molecules ; 29(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38930981

ABSTRACT

ZnO nanorod nonwoven fabrics (ZNRN) were developed through hydrothermal synthesis to facilitate the prevention of the transmission of respiratory pathogens. The superhydrophobicity and antibacterial properties of ZNRN were improved through the response surface methodology. The synthesized material exhibited significant water repellency, indicated by a water contact angle of 163.9°, and thus demonstrated antibacterial rates of 91.8% for Escherichia coli (E. coli) and 79.75% for Staphylococcus aureus (S. aureus). This indicated that E. coli with thinner peptidoglycan may be more easily killed than S. aureus. This study identified significant effects of synthesis conditions on the antibacterial effectiveness, with comprehensive multivariate analyses elucidating the underlying correlations. In addition, the ZnO nanorod structure of ZNRN was characterized through SEM and XRD analyses. It endows the properties of superhydrophobicity (thus preventing bacteria from adhering to the ZNRN surface) and antibacterial capacity (thus damaging cells through the puncturing of these nanorods). Consequently, the alignment of two such features is desired to help support the development of personal protective equipment, which assists in avoiding the spread of respiratory infections.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Hydrophobic and Hydrophilic Interactions , Nanotubes , Staphylococcus aureus , Zinc Oxide , Zinc Oxide/chemistry , Zinc Oxide/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Nanotubes/chemistry , Escherichia coli/drug effects , Escherichia coli/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Textiles/microbiology , Microbial Sensitivity Tests , Surface Properties
11.
EMBO J ; 38(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30760492

ABSTRACT

Pathogenic bacteria must rapidly adapt to ever-changing environmental signals resulting in metabolism remodeling. The carbon catabolite repression, mediated by the catabolite control protein A (CcpA), is used to express genes involved in utilization and metabolism of the preferred carbon source. Here, we have identified RsaI as a CcpA-repressed small non-coding RNA that is inhibited by high glucose concentrations. When glucose is consumed, RsaI represses translation initiation of mRNAs encoding a permease of glucose uptake and the FN3K enzyme that protects proteins against damage caused by high glucose concentrations. RsaI also binds to the 3' untranslated region of icaR mRNA encoding the transcriptional repressor of exopolysaccharide production and to sRNAs induced by the uptake of glucose-6 phosphate or nitric oxide. Furthermore, RsaI expression is accompanied by a decreased transcription of genes involved in carbon catabolism pathway and an activation of genes involved in energy production, fermentation, and nitric oxide detoxification. This multifaceted RNA can be considered as a metabolic signature when glucose becomes scarce and growth is arrested.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Glucose/deficiency , RNA, Bacterial/genetics , RNA, Small Untranslated/genetics , Repressor Proteins/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/genetics , Binding Sites , Biofilms/drug effects , Gene Expression Regulation, Bacterial , Glucose/administration & dosage , Metabolic Networks and Pathways , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Ribosomes/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Sweetening Agents/administration & dosage , Transcriptome
12.
Nucleic Acids Res ; 49(18): 10644-10656, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34554192

ABSTRACT

Staphylococcus aureus is an opportunistic human and animal pathogen with an arsenal of virulence factors that are tightly regulated during bacterial infection. The latter is achieved through a sophisticated network of regulatory proteins and regulatory RNAs. Here, we describe the involvement of a novel prophage-carried small regulatory S. aureus RNA, SprY, in the control of virulence genes. An MS2-affinity purification assay reveals that SprY forms a complex in vivo with RNAIII, a major regulator of S. aureus virulence genes. SprY binds to the 13th stem-loop of RNAIII, a key functional region involved in the repression of multiple mRNA targets. mRNAs encoding the repressor of toxins Rot and the extracellular complement binding protein Ecb are among the targets whose expression is increased by SprY binding to RNAIII. Moreover, SprY decreases S. aureus hemolytic activity and virulence. Our results indicate that SprY titrates RNAIII activity by targeting a specific stem loop. Thus, we demonstrate that a prophage-encoded sRNA reduces the pathogenicity of S. aureus through RNA sponge activity.


Subject(s)
RNA, Bacterial/metabolism , RNA, Small Untranslated/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Animals , Female , Gene Expression Regulation, Bacterial , Hemolysis , Mice , RNA, Bacterial/chemistry , RNA, Small Untranslated/chemistry , RNA, Small Untranslated/genetics , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Virulence/genetics
13.
Mol Microbiol ; 116(1): 1-15, 2021 07.
Article in English | MEDLINE | ID: mdl-33576132

ABSTRACT

Pseudomonas aeruginosa (Pa) and Staphylococcus aureus (Sa) are opportunistic pathogens that are most commonly co-isolated from chronic wounds and the sputum of cystic fibrosis patients. Over the last few years, there have been plenty of contrasting results from studies involving P. aeruginosa and S. aureus co-cultures. The general concept that P. aeruginosa outcompetes S. aureus has been challenged and there is more evidence now that they can co-exist. Nevertheless, it still remains difficult to mimic polymicrobial infections in vitro and in vivo. In this review, we discuss recent advances in regard to Pa-Sa molecular interactions, their physical responses, and in vitro and in vivo models. We believe it is important to optimize growth conditions in the laboratory, determine appropriate bacterial starting ratios, and consider environmental factors to study the co-existence of these two pathogens. Ideally, optimized growth media should reflect host-mimicking conditions with or without host cells that allow both bacteria to co-exist. To further identify mechanisms that could help to treat these complex infections, we propose to use relevant polymicrobial animal models. Ultimately, we briefly discuss how polymicrobial infections can increase antibiotic tolerance.


Subject(s)
Biofilms/growth & development , Coinfection/microbiology , Microbial Interactions/physiology , Pseudomonas aeruginosa/growth & development , Staphylococcus aureus/growth & development , Anti-Bacterial Agents/pharmacology , Coculture Techniques , Cystic Fibrosis/microbiology , Fasciitis, Necrotizing/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/drug effects , Quorum Sensing/physiology , Staphylococcal Infections/pathology , Staphylococcus aureus/drug effects , Surgical Wound Infection/microbiology , Wound Infection/microbiology
14.
Cell Microbiol ; 23(3): e13301, 2021 03.
Article in English | MEDLINE | ID: mdl-33331054

ABSTRACT

Fungal spores are unique cells that mediate dispersal and survival in the environment. For pathogenic fungi encountering a susceptible host, these specialised structures may serve as infectious particles. The main causative agent of the opportunistic disease aspergillosis, Aspergillus fumigatus, produces asexual spores, the conidia, that become dissipated by air flows or water currents but also serve as propagules to infect a susceptible host. We demonstrate that the defX gene of this mould encodes putative antimicrobial peptides resembling cysteine-stabilised (CS)αß defensins that are expressed in a specific spatial and temporal manner in the course of asexual spore formation. Localisation studies on strains expressing a fluorescent proxy or tagged defX alleles expose that these antimicrobial peptides are secreted to coat the conidial surface. Deletion mutants reveal that the spore-associated defX gene products delay the growth of Gram-positive Staphylococcus aureus and demonstrate that the defX gene and presumably its encoded spore-associated defensins confer a growth advantage to the fungal opponent over bacterial competitors. These findings have implications with respect to the ecological niche of A. fumigatus that serves as a 'virulence school' for this human pathogenic mould; further relevance is given for the infectious process resulting in aspergillosis, considering competition with the host microbiome or co-infecting microorganisms to break colonisation resistance at host surfaces.


Subject(s)
Aspergillus fumigatus/pathogenicity , Defensins/metabolism , Fungal Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Aspergillosis/microbiology , Aspergillus fumigatus/genetics , Aspergillus fumigatus/physiology , Defensins/genetics , Escherichia coli/growth & development , Fungal Proteins/genetics , Genes, Fungal , Humans , Pore Forming Cytotoxic Proteins/genetics , Spores, Fungal/metabolism , Spores, Fungal/physiology , Staphylococcus aureus/growth & development , Virulence
15.
Nature ; 535(7613): 511-6, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27466123

ABSTRACT

The vast majority of systemic bacterial infections are caused by facultative, often antibiotic-resistant, pathogens colonizing human body surfaces. Nasal carriage of Staphylococcus aureus predisposes to invasive infection, but the mechanisms that permit or interfere with pathogen colonization are largely unknown. Whereas soil microbes are known to compete by production of antibiotics, such processes have rarely been reported for human microbiota. We show that nasal Staphylococcus lugdunensis strains produce lugdunin, a novel thiazolidine-containing cyclic peptide antibiotic that prohibits colonization by S. aureus, and a rare example of a non-ribosomally synthesized bioactive compound from human-associated bacteria. Lugdunin is bactericidal against major pathogens, effective in animal models, and not prone to causing development of resistance in S. aureus. Notably, human nasal colonization by S. lugdunensis was associated with a significantly reduced S. aureus carriage rate, suggesting that lugdunin or lugdunin-producing commensal bacteria could be valuable for preventing staphylococcal infections. Moreover, human microbiota should be considered as a source for new antibiotics.


Subject(s)
Anti-Bacterial Agents/metabolism , Peptides, Cyclic/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/growth & development , Staphylococcus lugdunensis/metabolism , Symbiosis , Thiazolidines/metabolism , Animals , Anti-Bacterial Agents/biosynthesis , Carrier State/microbiology , Disease Models, Animal , Drug Resistance, Microbial , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microbiota/physiology , Nose/microbiology , Sigmodontinae , Staphylococcal Infections/drug therapy , Staphylococcus aureus/pathogenicity
16.
Dermatology ; 238(1): 109-120, 2022.
Article in English | MEDLINE | ID: mdl-33887725

ABSTRACT

BACKGROUND: The pathophysiology in atopic dermatitis (AD) is not fully understood, but immune dysfunction, skin barrier defects, and alterations of the skin microbiota are thought to play important roles. AD skin is frequently colonized with Staphylococcus aureus (S. aureus) and microbial diversity on lesional skin (LS) is reduced compared to on healthy skin. Treatment with narrow-band ultraviolet B (nb-UVB) leads to clinical improvement of the eczema and reduced abundance of S. aureus. However, in-depth knowledge of the temporal dynamics of the skin microbiota in AD in response to nb-UVB treatment is lacking and could provide important clues to decipher whether the microbial changes are primary drivers of the disease, or secondary to the inflammatory process. OBJECTIVES: To map the temporal shifts in the microbiota of the skin, nose, and throat in adult AD patients after nb-UVB treatment. METHODS: Skin swabs were taken from lesional AD skin (n = 16) before and after 3 treatments of nb-UVB, and after 6-8 weeks of full-body treatment. We also obtained samples from non-lesional skin (NLS) and from the nose and throat. All samples were characterized by 16S rRNA gene sequencing. RESULTS: We observed shifts towards higher diversity in the microbiota of lesional AD skin after 6-8 weeks of treatment, while the microbiota of NLS and of the nose/throat remained unchanged. After only 3 treatments with nb-UVB, there were no significant changes in the microbiota. CONCLUSION: Nb-UVB induces changes in the skin microbiota towards higher diversity, but the microbiota of the nose and throat are not altered.


Subject(s)
Dermatitis, Atopic/microbiology , Dermatitis, Atopic/radiotherapy , Microbiota/radiation effects , Skin/microbiology , Ultraviolet Therapy , Adult , Aged , Biodiversity , Female , Humans , Male , Middle Aged , Nose/microbiology , Pharynx/microbiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/radiation effects , Treatment Outcome , Young Adult
17.
Mar Drugs ; 20(3)2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35323468

ABSTRACT

Volatile compounds from the marine cyanolichen Lichina pygmaea, collected from the Moroccan Atlantic coast, were extracted by hydrodistillation and their putative chemical composition was investigated by gas chromatography coupled to mass spectrometry (GC/MS). Based on the obtained results, Lichina pygmaea volatile compounds (LPVCs) were mainly dominated by sesquiterpenes compounds, where γ-himachalene, ß-himachalene, (2E,4E)-2,4 decadienal and α-himachalene were assumed to be the most abundant constituents, with percentage of 37.51%, 11.71%, 8.59% and 7.62%, respectively. LPVCs depicted significant antimicrobial activity against all tested strains (Staphylococcus aureus CCMM B3, Pseudomonas aeruginosa DSM 50090, Escherichia coli ATCC 8739 and Candida albicans CCMM-L4) with minimum inhibitory concentration (MIC) values within the range of 1.69-13.5 mg/mL. Moreover, this LPVC showed interesting scavenging effects on the 2,2-diphenyl-1-picrylhydrazyl radical with an IC50 of 0.21 mg/mL. LPVCs could be an approving resource with moderate antimicrobial potential and interesting antioxidant activity for cosmetics and pharmaceutical applications.


Subject(s)
Anti-Infective Agents , Antioxidants , Ascomycota/chemistry , Sesquiterpenes , Volatile Organic Compounds , Anti-Infective Agents/chemistry , Anti-Infective Agents/isolation & purification , Anti-Infective Agents/pharmacology , Antioxidants/chemistry , Antioxidants/isolation & purification , Antioxidants/pharmacology , Biphenyl Compounds/chemistry , Candida albicans/drug effects , Candida albicans/growth & development , Escherichia coli/drug effects , Escherichia coli/growth & development , Picrates/chemistry , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Sesquiterpenes/analysis , Sesquiterpenes/isolation & purification , Sesquiterpenes/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Volatile Organic Compounds/chemistry , Volatile Organic Compounds/isolation & purification , Volatile Organic Compounds/pharmacology
18.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163108

ABSTRACT

The biodiversity of microorganisms is maintained by intricate nets of interactions between competing species. Impaired functionality of human microbiomes correlates with their reduced biodiversity originating from aseptic environmental conditions and antibiotic use. Microbiomes of wild animals are free of these selective pressures. Microbiota provides a protecting shield from invasion by pathogens in the wild, outcompeting their growth in specific ecological niches. We applied ultrahigh-throughput microfluidic technologies for functional profiling of microbiomes of wild animals, including the skin beetle, Siberian lynx, common raccoon dog, and East Siberian brown bear. Single-cell screening of the most efficient killers of the common human pathogen Staphylococcus aureus resulted in repeated isolation of Bacillus pumilus strains. While isolated strains had different phenotypes, all of them displayed a similar set of biosynthetic gene clusters (BGCs) encoding antibiotic amicoumacin, siderophore bacillibactin, and putative analogs of antimicrobials including bacilysin, surfactin, desferrioxamine, and class IId cyclical bacteriocin. Amicoumacin A (Ami) was identified as a major antibacterial metabolite of these strains mediating their antagonistic activity. Genome mining indicates that Ami BGCs with this architecture subdivide into three distinct families, characteristic of the B. pumilus, B. subtilis, and Paenibacillus species. While Ami itself displays mediocre activity against the majority of Gram-negative bacteria, isolated B. pumilus strains efficiently inhibit the growth of both Gram-positive S. aureus and Gram-negative E. coli in coculture. We believe that the expanded antagonistic activity spectrum of Ami-producing B. pumilus can be attributed to the metabolomic profile predetermined by their biosynthetic fingerprint. Ultrahigh-throughput isolation of natural probiotic strains from wild animal microbiomes, as well as their metabolic reprogramming, opens up a new avenue for pathogen control and microbiome remodeling in the food industry, agriculture, and healthcare.


Subject(s)
Animals, Wild/microbiology , Anti-Bacterial Agents/administration & dosage , Bacillus pumilus/chemistry , Escherichia coli/growth & development , Microbiota , Probiotics/administration & dosage , Staphylococcus aureus/growth & development , Animals , Anti-Bacterial Agents/isolation & purification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli/drug effects , Genome, Bacterial , Metabolome , Multigene Family , Probiotics/isolation & purification , Staphylococcus aureus/drug effects
19.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163197

ABSTRACT

Problems connected with biofilm-related infections and antibiotic resistance necessitate the investigation and development of novel treatment strategies. Given their unique characteristics, one of the most promising alternatives to conventional antibiotics are bacteriophages. In the in vitro and in vivo larva model study, we demonstrate that phages vB_SauM-A, vB_SauM-C, and vB_SauM-D are effective antibiofilm agents. The exposure of biofilm to phages vB_SauM-A and vB_SauM-D led to 2-3 log reductions in the colony-forming unit number in most of the multidrug-resistant S. aureus strains. It was found that phage application reduced the formed biofilms independently of the used titer. Moreover, the study demonstrated that bacteriophages are more efficient in biofilm biomass removal and reduction in staphylococci count when compared to the antibiotics used. The scanning electron microscopy analysis results are in line with colony forming unit (CFU) counting but not entirely consistent with crystal violet (CV) staining. Additionally, phages vB_SauM-A, vB_SauM-C, and vB_SauM-D can significantly increase the survival rate and extend the survival time of Galleria mellonella larvae.


Subject(s)
Anti-Bacterial Agents/pharmacology , Staphylococcal Infections/therapy , Staphylococcus aureus/drug effects , Bacteriolysis/drug effects , Bacteriolysis/genetics , Bacteriophages/genetics , Bacteriophages/pathogenicity , Biofilms/drug effects , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/genetics , Genome, Viral/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Phage Therapy/methods , Staphylococcal Infections/drug therapy , Staphylococcus aureus/growth & development
20.
Int J Mol Sci ; 23(3)2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35163819

ABSTRACT

While blue LED (b-LED) light is increasingly being studied for its cytotoxic activity towards bacteria in therapy of skin-related infections, its effects on eukaryotic cells plasticity are less well characterized. Moreover, since different protocols are often used, comparing the effect of b-LED towards both microorganisms and epithelial surfaces may be difficult. The aim of this study was to analyze, in the same experimental setting, both the bactericidal activity and the effects on human keratinocytes. Exposure to b-LED induced an intense cytocidal activity against Gram-positive (i.e, Staphylococcus aureus) and Gram-negative (i.e., Pseudomonas aeruginosa) bacteria associated with catheter-related infections. Treatment with b-LED of a human keratinocyte cell line induced a transient cell cycle arrest. At the molecular level, exposure to b-LED induced a transient downregulation of Cyclin D1 and an upregulation of p21, but not signs of apoptosis. Interestingly, a transient induction of phosphor-histone γ-H2Ax, which is associated with genotoxic damages, was observed. At the same time, keratinocytes underwent a transient epithelial to mesenchymal transition (EMT)-like phenotype, characterized by E-cadherin downregulation and SNAIL/SLUG induction. As a functional readout of EMT induction, a scratch assay was performed. Surprisingly, b-LED treatment provoked a delay in the scratch closure. In conclusion, we demonstrated that b-LED microbicidal activity is associated with complex responses in keratinocytes that certainly deserve further analysis.


Subject(s)
Cell Cycle Checkpoints/radiation effects , Keratinocytes/cytology , Light/adverse effects , Pseudomonas aeruginosa/growth & development , Staphylococcus aureus/growth & development , Antigens, CD/metabolism , Cadherins/metabolism , Cell Proliferation , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down Syndrome , Epithelial-Mesenchymal Transition/radiation effects , Gene Expression Regulation/drug effects , HaCaT Cells , Humans , Keratinocytes/metabolism , Keratinocytes/radiation effects , Microbial Viability/radiation effects , Pseudomonas aeruginosa/radiation effects , Snail Family Transcription Factors/metabolism , Staphylococcus aureus/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL