Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Country/Region as subject
Publication year range
1.
BMC Microbiol ; 23(1): 185, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37438716

ABSTRACT

BACKGROUND: The nose and the throat are the most predominant colonizing sites of Staphylococcus aureus, and colonization is a risk factor for infection. Nasal colonization is well described; however, we have limited knowledge about S. aureus throat colonization. The main objective of this study was to explore differentially expressed genes (DEGs) in S. aureus throat isolate TR145 exposed to human tonsil epithelial cells (HTEpiC) by using RNA sequencing (RNA-seq) and pathway analysis. DEGs in S. aureus at 1 or 3 hours (h) interaction with its host were explored. RESULTS: S. aureus was co-cultured in absence and presence of tonsillar cells at 1 or 3 h. Over the 3 h time frame, the bacteria multiplied, but still caused only minor cytotoxicity. Upon exposure to tonsillar cell line, S. aureus changed its transcriptomic profile. A total of 508 DEGs were identified including unique (1 h, 160 DEGs and 3 h, 78 DEGs) and commonly shared genes (1 and 3 h, 270 DEGs). Among the DEGs, were genes encoding proteins involved in adhesion and immune evasion, as well as iron acquisition and transport. Reverse transcription qPCR was done on selected genes, and the results correlated with the RNA-seq data. CONCLUSION: We have shown the suitability of using HTEpiC as an in vitro model for investigating key determinants in S. aureus during co-incubation with host cells. Several DEGs were unique after 1 or 3 h exposure to host cells, while others were commonly expressed at both time points. As their expression is induced upon meeting with the host, they might be explored further for future targets for intervention to prevent either colonization or infection in the throat.


Subject(s)
Palatine Tonsil , Staphylococcal Infections , Humans , Staphylococcus aureus/genetics , Pharynx , Base Sequence
2.
Int J Mol Sci ; 24(22)2023 Nov 07.
Article in English | MEDLINE | ID: mdl-38003243

ABSTRACT

Enterococcus faecium is a leading cause of nosocomial infections, particularly in immunocompromised patients. The rise of multidrug-resistant E. faecium, including Vancomycin-Resistant Enterococci (VRE), is a major concern. Vaccines are promising alternatives to antibiotics, but there is currently no vaccine available against enterococci. In a previous study, we identified six protein vaccine candidates associated with extracellular membrane vesicles (MVs) produced by nosocomial E. faecium. In this study, we immunized rabbits with two different VRE-derived MV preparations and characterized the resulting immune sera. Both anti-MV sera exhibited high immunoreactivity towards the homologous strain, three additional VRE strains, and eight different unrelated E. faecium strains representing different sequence types (STs). Additionally, we demonstrated that the two anti-MV sera were able to mediate opsonophagocytic killing of not only the homologous strain but also three unrelated heterologous VRE strains. Altogether, our results indicate that E. faecium MVs, regardless of the purification method for obtaining them, are promising vaccine candidates against multidrug-resistant E. faecium and suggest that these naturally occurring MVs can be used as a multi-antigen platform to elicit protective immune responses against enterococcal infections.


Subject(s)
Enterococcus faecium , Gram-Positive Bacterial Infections , Vaccines , Vancomycin-Resistant Enterococci , Animals , Humans , Rabbits , Enterococcus faecalis , Anti-Bacterial Agents/therapeutic use , Gram-Positive Bacterial Infections/prevention & control , Gram-Positive Bacterial Infections/drug therapy , Microbial Sensitivity Tests
3.
Biomacromolecules ; 23(1): 303-315, 2022 01 10.
Article in English | MEDLINE | ID: mdl-34914360

ABSTRACT

To avert the poor bioavailability of antibiotics during S. aureus biofilm infections, a series of zwitterionic nanoparticles containing nucleic acid nanostructures were fabricated for the delivery of vancomycin. The nanoparticles were prepared with three main lipids: (i) neutral (soy phosphatidylcholine; P), (ii) positively charged ionizable (1,2-dioleyloxy-3-dimethylaminopropane; D), and (iii) anionic (1,2-dipalmitoyl-sn-glycero-3-phospho((ethyl-1',2',3'-triazole) triethylene glycolmannose; M) or (cholesteryl hemisuccinate; C) lipids. The ratio of the anionic lipid was tuned between 0 and 10 mol %, and its impact on surface charge, size, stability, toxicity, and biofilm sensitivity was evaluated. Under biofilm mimicking conditions, the enzyme degradability (via dynamic light scattering (DLS)), antitoxin (via DLS and spectrophotometry), and antibiotic release profile was assessed. Additionally, biofilm penetration, prevention (in vitro), and eradication (ex vivo) of the vancomycin loaded formulation was investigated. Compared with the unmodified nanoparticles which exhibited the smallest size (188 nm), all three surface modified formulations showed significantly larger sizes (i.e., 222-277 nm). Under simulations of biofilm pH conditions, the mannose modified nanoparticle (PDM 90/5/5) displayed ideal charge reversal from a neutral (+1.69 ± 1.83 mV) to a cationic surface potential (+17.18 ± 2.16 mV) to improve bacteria binding and biofilm penetration. In the presence of relevant bacterial enzymes, the carrier rapidly released the DNA nanoparticles to function as an antitoxin against α-hemolysin. Controlled release of vancomycin prevented biofilm attachment and significantly reduced early stage biofilm formations within 24 h. Enhanced biocompatibility and significant ex vivo potency of the PDM 90/5/5 formulation was also observed. Taken together, these results emphasize the benefit of these nanocarriers as potential therapies against biofilm infections and fills the gap for multifunctional nanocarriers that prevent biofilm infections.


Subject(s)
Anti-Infective Agents , Nanoparticles , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms , Microbial Sensitivity Tests , Nanoparticles/chemistry , Staphylococcus aureus
4.
Mar Drugs ; 19(5)2021 May 12.
Article in English | MEDLINE | ID: mdl-34065943

ABSTRACT

Burns and other skin injuries are growing concerns as well as challenges in an era of antimicrobial resistance. Novel treatment options to improve the prevention and eradication of infectious skin biofilm-producing pathogens, while enhancing wound healing, are urgently needed for the timely treatment of infection-prone injuries. Treatment of acute skin injuries requires tailoring of formulation to assure both proper skin retention and the appropriate release of incorporated antimicrobials. The challenge remains to formulate antimicrobials with low water solubility, which often requires carriers as the primary vehicle, followed by a secondary skin-friendly vehicle. We focused on widely used chlorhexidine formulated in the chitosan-infused nanocarriers, chitosomes, incorporated into chitosan hydrogel for improved treatment of skin injuries. To prove our hypothesis, lipid nanocarriers and chitosan-comprising nanocarriers (≈250 nm) with membrane-active antimicrobial chlorhexidine were optimized and incorporated into chitosan hydrogel. The biological and antibacterial effects of both vesicles and a vesicles-in-hydrogel system were evaluated. The chitosomes-in-chitosan hydrogel formulation demonstrated promising physical properties and were proven safe. Additionally, the chitosan-based systems, both chitosomes and chitosan hydrogel, showed an improved antimicrobial effect against S. aureus and S. epidermidis compared to the formulations without chitosan. The novel formulation could serve as a foundation for infection prevention and bacterial eradication in acute wounds.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Chitosan/pharmacology , Hydrogels/pharmacology , Skin Diseases, Infectious/prevention & control , Skin/drug effects , Skin/injuries , Anti-Infective Agents, Local/chemistry , Anti-Infective Agents, Local/toxicity , Cell Line , Chitosan/chemistry , Chitosan/toxicity , Chlorhexidine/pharmacology , Drug Delivery Systems/methods , Humans , Hydrogels/chemistry , Hydrogels/toxicity , Nanogels/chemistry , Nanogels/toxicity , Nanomedicine/methods , Skin/microbiology , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects , Wound Healing/drug effects
5.
Mar Drugs ; 18(2)2020 Jan 31.
Article in English | MEDLINE | ID: mdl-32023890

ABSTRACT

Topical administration of drugs into the vagina can provide local therapy of vaginal infections, preventing the possible systemic side effects of the drugs. The natural polysaccharide chitosan is known for its excellent mucoadhesive properties, safety profile, and antibacterial effects, and thus it can be utilized in improving localized vaginal therapy by prolonging the residence time of a drug at the vaginal site while acting as an antimicrobial in synergy. Therefore, we aimed to explore the potential of chitosan, namely chitosan-coated liposomes and chitosan hydrogel, as an excipient with intrinsic antimicrobial properties. Liposomes were prepared by the thin-film hydration method followed by vesicle size reduction by sonication to the desired size, approximately 200 nm, and coated with chitosan (0.01, 0.03, 0.1, and 0.3%, w/v, respectively). The mucoadhesive properties of chitosan-coated liposomes were determined through their binding efficiency to mucin compared to non-coated liposomes. Non-coated liposomal suspensions were incorporated in chitosan hydrogels forming the liposomes-in-hydrogel formulations, which were further assessed for their texture properties in the presence of biological fluid simulants. The antibacterial effect of chitosan-coated liposomes (0.03%, 0.1% and 0.3%, w/v) and chitosan hydrogels (0.1% and 0.3%, w/w) on Staphylococcus epidermidis and Staphylococcus aureus was successfully confirmed.


Subject(s)
Anti-Bacterial Agents/chemistry , Chitosan/chemistry , Excipients/chemistry , Adhesiveness , Administration, Intravaginal , Animals , Anti-Bacterial Agents/pharmacology , Chitosan/pharmacology , Female , Humans , Hydrogels , Liposomes , Particle Size , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects , Swine
6.
BMC Infect Dis ; 19(1): 607, 2019 Jul 10.
Article in English | MEDLINE | ID: mdl-31291897

ABSTRACT

BACKGROUND: Blood culture bottles (BCBs) provide a semiautomated method for culturing periprosthetic tissue specimens. A study evaluating BCBs for culturing clinical samples other than body fluids is needed before implementation into clinical practice. Our objective was to evaluate use of the BacT/Alert® Virtuo blood culture system for culturing periprosthetic tissue specimens. METHODS: The study was performed through the analysis of spiked (n = 36) and clinical (n = 158) periprosthetic tissue samples. Clinical samples were analyzed by the BCB method and the results were compared to the conventional microbiological culture-based method for time to detection and microorganisms identified. RESULTS: The BacT/Alert® Virtuo blood culture system detected relevant bacteria for prosthetic joint infection in both spiked and clinical samples. The BCB method was found to be as sensitive (79%) as the conventional method (76%) (p = 0.844) during the analyses of clinical samples. The BCB method yielded positive results much faster than the conventional method: 89% against 27% detection within 24 h, respectively. The median detection time was 11.1 h for the BCB method (12 h and 11 h for the aerobic and the anaerobic BCBs, correspondingly). CONCLUSION: We recommend using the BacT/Alert® Virtuo blood culture system for analyzing prosthetic joint tissue, since this detect efficiently and more rapidly a wider range of bacteria than the conventional microbiological method.


Subject(s)
Bacteria/isolation & purification , Blood Culture/methods , Joint Prosthesis/adverse effects , Prosthesis-Related Infections/microbiology , Blood Culture/instrumentation , Humans , Prosthesis-Related Infections/diagnosis , Prosthesis-Related Infections/pathology , Sensitivity and Specificity , Specimen Handling , Time Factors
7.
BMC Microbiol ; 18(1): 34, 2018 04 16.
Article in English | MEDLINE | ID: mdl-29661152

ABSTRACT

BACKGROUND: Staphylococcus aureus cell wall anchored Serine Aspartate repeat containing protein D (SdrD) is a member of the microbial surface component recognising adhesive matrix molecules (MSCRAMMs). It is involved in the bacterial adhesion and virulence. However the extent of genetic variation in S. aureus sdrD gene within isolates from healthy carriers are not known. The aim of this study was to evaluate allelic variation of the sdrD gene among S. aureus from healthy nasal carriers. RESULTS: The sdrD A region from 48 S. aureus isolates from healthy carriers were analysed and classified into seven variants. Variations in the sdrD A region were concentrated in the N2 and N3 subdomains. Sequence analysis of the entire sdrD gene of representative isolates revealed variations in the SD repeat and the EF motifs of the B repeat. In silico structural modelling indicates that there are no differences in the SdrD structure of the 7 variants. Variable amino acid residues mapped onto the 3D structure revealed that the variations are surface located, exist within the groove between the N2-N3 subdomains and distributed mainly on the N3 subdomain. Comparison of adhesion to keratinocytes in an in vitro cell adhesion assay, using NCTC 8325-4∆sdrD strains expressing the various sdrD gene variants, indicated a significant difference between only two complements while others showed no major difference in their adhesion. CONCLUSIONS: This study provides evidence of sequence variations across the different domains of SdrD from S. aureus isolated from healthy nasal carriers. Proper understanding of these variations is necessary in the study of S. aureus pathogenesis.


Subject(s)
Bacterial Proteins/genetics , Calcium-Binding Proteins/genetics , Genetic Variation , Nose/microbiology , Staphylococcus aureus/genetics , Amino Acid Sequence , Bacterial Adhesion , Bacterial Proteins/classification , Bacterial Proteins/isolation & purification , Calcium-Binding Proteins/classification , Calcium-Binding Proteins/isolation & purification , Cell Line , Humans , Keratinocytes/microbiology , Models, Molecular , Multilocus Sequence Typing , Phylogeny , Protein Conformation , Protein Domains , Staphylococcal Infections/microbiology , Staphylococcus aureus/isolation & purification , Virulence/genetics
8.
Infect Immun ; 85(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-27795358

ABSTRACT

Staphylococcus aureus expresses a panel of cell wall-anchored adhesins, including proteins belonging to the microbial surface components recognizing adhesive matrix molecule (MSCRAMM) family, exemplified by the serine-aspartate repeat protein D (SdrD), which serve key roles in colonization and infection. Deletion of sdrD from S. aureus subsp. aureus strain NCTC8325-4 attenuated bacterial survival in human whole blood ex vivo, which was associated with increased killing by human neutrophils. Remarkably, SdrD was able to inhibit innate immune-mediated bacterial killing independently of other S. aureus proteins, since addition of recombinant SdrD protein and heterologous expression of SdrD in Lactococcus lactis promoted bacterial survival in human blood. SdrD contributes to bacterial virulence in vivo, since fewer S. aureus subsp. aureus NCTC8325-4 ΔsdrD bacteria than bacteria of the parent strain were recovered from blood and several organs using a murine intravenous infection model. Collectively, our findings reveal a new property of SdrD as an important key contributor to S. aureus survival and the ability to escape the innate immune system in blood.


Subject(s)
Bacterial Proteins/metabolism , Calcium-Binding Proteins/metabolism , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Virulence/physiology , Adhesins, Bacterial/metabolism , Animals , Bacterial Adhesion/physiology , Female , Humans , Immunity, Innate/immunology , Lactococcus lactis/immunology , Lactococcus lactis/metabolism , Mice , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Recombinant Proteins/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Virulence/immunology
9.
BMC Microbiol ; 17(1): 89, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28381253

ABSTRACT

BACKGROUND: Colonization of the body is an important step in Staphylococcus aureus infection. S. aureus colonizes skin and mucous membranes in humans and several animal species. One important ecological niche of S. aureus is the anterior nares. More than 60% of the S. aureus in the nose are found in vestibulum nasi. Our aim was to describe the localization of S. aureus in nasal tissue from healthy carriers. METHODS: Punch skin biopsies were taken from vestibulum nasi from healthy volunteers (S. aureus carriers and non-/intermittent carriers, n = 39) attending the population-based Tromsø 6 study. The tissue samples were processed as frozen sections before immunostaining with a specific S. aureus antibody, and finally evaluated by a confocal laser-scanning microscope. RESULTS: Our results suggest that S. aureus colonize both the upper and lower layers of the epidermis within the nasal epithelium of healthy individuals. The number of S. aureus in epidermis was surprisingly low. Intracellular localization of S. aureus in nasal tissue from healthy individuals was also detected. CONCLUSIONS: Knowledge of the exact localization of S. aureus in nasal tissue is important for the understanding of the host responses against S. aureus. Our results may have consequences for the eradication strategy of S. aureus in carriers, and further work can provide us with tools for targeted prevention of S. aureus colonisation and infection.


Subject(s)
Carrier State/microbiology , Host-Parasite Interactions , Nasal Cavity/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Adult , Biopsy , Colony Count, Microbial , Cross-Sectional Studies , Epithelial Cells/microbiology , Epithelial Cells/pathology , Female , Humans , Male , Microscopy, Confocal , Nasal Mucosa/microbiology , Nasal Mucosa/pathology , Norway , Nose/microbiology , Skin/microbiology , Staphylococcal Infections/transmission , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology
10.
Virol J ; 12: 7, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25638270

ABSTRACT

BACKGROUND: The human polyomavirus BK expresses a 66 amino-acid peptide referred to as agnoprotein. Though mutants lacking agnoprotein are severely reduced in producing infectious virions, the exact function of this peptide remains incompletely understood. To elucidate the function of agnoprotein, we searched for novel cellular interaction partners. METHODS: Yeast-two hybrid assay was performed with agnoprotein as bait against human kidney and thymus libraries. The interaction between agnoprotein and putative partners was further examined by GST pull down, co-immunoprecipitation, and fluorescence resonance energy transfer studies. Biochemical and biological studies were performed to examine the functional implication of the interaction of agnoprotein with cellular target proteins. RESULTS: Proliferating cell nuclear antigen (PCNA), which acts as a processivity factor for DNA polymerase δ, was identified as an interaction partner. The interaction between agnoprotein and PCNA is direct and occurs also in human cells. Agnoprotein exerts an inhibitory effect on PCNA-dependent DNA synthesis in vitro and reduces cell proliferation when ectopically expressed. Overexpression of PCNA restores agnoprotein-mediated inhibition of cell proliferation. CONCLUSION: Our data suggest that PCNA is a genuine interaction partner of agnoprotein and the inhibitory effect on PCNA-dependent DNA synthesis by the agnoprotein may play a role in switching off (viral) DNA replication late in the viral replication cycle when assembly of replicated genomes and synthesized viral capsid proteins occurs.


Subject(s)
DNA Replication , Proliferating Cell Nuclear Antigen/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication , BK Virus/genetics , BK Virus/metabolism , Cell Line, Tumor , Cell Proliferation , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , Humans , Proliferating Cell Nuclear Antigen/genetics , Two-Hybrid System Techniques , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics
11.
mSystems ; 9(4): e0013024, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38470253

ABSTRACT

In the Staphylococcus aureus genome, a set of highly conserved two-component systems (TCSs) composed of histidine kinases (HKs) and their cognate response regulators (RRs) sense and respond to environmental stimuli, which drive the adaptation of the bacteria. This study investigates the complex interplay between TCSs in S. aureus USA300, a predominant methicillin-resistant S. aureus strain, revealing shared and unique virulence regulatory pathways and genetic variations mediating signal specificity within TCSs. Using TCS-related mutants from the Nebraska Transposon Mutant Library, we analyzed the effects of inactivated TCS HKs and RRs on the production of various virulence factors, in vitro infection abilities, and adhesion assays. We found that the TCSs' influence on virulence determinants was not associated with their phylogenetic relationship, indicating divergent functional evolution. Using the co-crystallized structure of the DesK-DesR from Bacillus subtilis and the modeled structures of the four NarL TCSs in S. aureus, we identified interacting residues, revealing specificity determinants and conservation within the same TCS, even from different strain backgrounds. The interacting residues were highly conserved within strains but varied between species due to selection pressures and the coevolution of cognate pairs. This study unveils the complex interplay and divergent functional evolution of TCSs, highlighting their potential for future experimental exploration of phosphotransfer between cognate and non-cognate recombinant HK and RRs.IMPORTANCEGiven the widespread conservation of two-component systems (TCSs) in bacteria and their pivotal role in regulating metabolic and virulence pathways, they present a compelling target for anti-microbial agents, especially in the face of rising multi-drug-resistant infections. Harnessing TCSs therapeutically necessitates a profound understanding of their evolutionary trajectory in signal transduction, as this underlies their unique or shared virulence regulatory pathways. Such insights are critical for effectively targeting TCS components, ensuring an optimized impact on bacterial virulence, and mitigating the risk of resistance emergence via the evolution of alternative pathways. Our research offers an in-depth exploration of virulence determinants controlled by TCSs in S. aureus, shedding light on the evolving specificity determinants that orchestrate interactions between their cognate pairs.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcus aureus , Staphylococcus aureus/genetics , Bacterial Proteins/genetics , Virulence/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , Phylogeny , Bacteria/metabolism
12.
mSystems ; 9(5): e0017924, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38656122

ABSTRACT

The utilization of ATP within cells plays a fundamental role in cellular processes that are essential for the regulation of host-pathogen dynamics and the subsequent immune response. This study focuses on ATP-binding proteins to dissect the complex interplay between Staphylococcus aureus and human cells, particularly macrophages (THP-1) and keratinocytes (HaCaT), during an intracellular infection. A snapshot of the various protein activity and function is provided using a desthiobiotin-ATP probe, which targets ATP-interacting proteins. In S. aureus, we observe enrichment in pathways required for nutrient acquisition, biosynthesis and metabolism of amino acids, and energy metabolism when located inside human cells. Additionally, the direct profiling of the protein activity revealed specific adaptations of S. aureus to the keratinocytes and macrophages. Mapping the differentially activated proteins to biochemical pathways in the human cells with intracellular bacteria revealed cell-type-specific adaptations to bacterial challenges where THP-1 cells prioritized immune defenses, autophagic cell death, and inflammation. In contrast, HaCaT cells emphasized barrier integrity and immune activation. We also observe bacterial modulation of host processes and metabolic shifts. These findings offer valuable insights into the dynamics of S. aureus-host cell interactions, shedding light on modulating host immune responses to S. aureus, which could involve developing immunomodulatory therapies. IMPORTANCE: This study uses a chemoproteomic approach to target active ATP-interacting proteins and examines the dynamic proteomic interactions between Staphylococcus aureus and human cell lines THP-1 and HaCaT. It uncovers the distinct responses of macrophages and keratinocytes during bacterial infection. S. aureus demonstrated a tailored response to the intracellular environment of each cell type and adaptation during exposure to professional and non-professional phagocytes. It also highlights strategies employed by S. aureus to persist within host cells. This study offers significant insights into the human cell response to S. aureus infection, illuminating the complex proteomic shifts that underlie the defense mechanisms of macrophages and keratinocytes. Notably, the study underscores the nuanced interplay between the host's metabolic reprogramming and immune strategy, suggesting potential therapeutic targets for enhancing host defense and inhibiting bacterial survival. The findings enhance our understanding of host-pathogen interactions and can inform the development of targeted therapies against S. aureus infections.


Subject(s)
Adenosine Triphosphate , Host-Pathogen Interactions , Keratinocytes , Macrophages , Staphylococcus aureus , Humans , Staphylococcus aureus/metabolism , Adenosine Triphosphate/metabolism , Host-Pathogen Interactions/immunology , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Keratinocytes/microbiology , Keratinocytes/metabolism , Keratinocytes/immunology , THP-1 Cells , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Proteomics/methods , Bacterial Proteins/metabolism , HaCaT Cells
13.
Front Cell Infect Microbiol ; 14: 1326730, 2024.
Article in English | MEDLINE | ID: mdl-38333035

ABSTRACT

Introduction: Improved understanding of Staphylococcus aureus throat colonization in the presence of other co-existing microbes is important for mapping S. aureus adaptation to the human throat, and recurrence of infection. Here, we explore the responses triggered by the encounter between two common throat bacteria, S. aureus and Streptococcus anginosus, to identify genes in S. aureus that are important for colonization in the presence of human tonsillar epithelial cells and S. anginosus, and further compare this transcriptome with the genes expressed in S. aureus as only bacterium. Methods: We performed an in vitro co-culture experiment followed by RNA sequencing to identify interaction-induced transcriptional alterations and differentially expressed genes (DEGs), followed by gene enrichment analysis. Results and discussion: A total of 332 and 279 significantly differentially expressed genes with p-value < 0.05 and log2 FoldChange (log2FC) ≥ |2| were identified in S. aureus after 1 h and 3 h co-culturing, respectively. Alterations in expression of various S. aureus survival factors were observed when co-cultured with S. anginosus and tonsillar cells. The serine-aspartate repeat-containing protein D (sdrD) involved in adhesion, was for example highly upregulated in S. aureus during co-culturing with S. anginosus compared to S. aureus grown in the absence of S. anginosus, especially at 3 h. Several virulence genes encoding secreted proteins were also highly upregulated only when S. aureus was co-cultured with S. anginosus and tonsillar cells, and iron does not appear to be a limiting factor in this environment. These findings may be useful for the development of interventions against S. aureus throat colonization and could be further investigated to decipher the roles of the identified genes in the host immune response in context of a throat commensal landscape.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Transcriptome , Streptococcus anginosus/genetics , Coculture Techniques , Staphylococcal Infections/microbiology
14.
Int J Pharm ; 657: 124136, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38642621

ABSTRACT

Increasing prevalence of infected and chronic wounds demands improved therapy options. In this work an electrospun nanofiber dressing with liposomes is suggested, focusing on the dressing's ability to support tissue regeneration and infection control. Chloramphenicol (CAM) was the chosen antibiotic, added to the nanofibers after first embedded in liposomes to maintain a sustained drug release. Nanofibers spun from five different polymer blends were tested, where pectin and polyethylene oxide (PEO) was identified as the most promising polymer blend, showing superior fiber formation and tensile strength. The wire-electrospinning setup (WES) was selected for its pilot-scale features, and water was applied as the only solvent for green electrospinning and to allow direct liposome incorporation. CAM-liposomes were added to Pectin-PEO nanofibers in the next step. Confocal imaging of rhodamine-labelled liposomes indicated intact liposomes in the fibers after electrospinning. This was supported by the observed in vitroCAM-release, showing that Pectin-PEO-nanofibers with CAM-liposomes had a delayed drug release compared to controls. Biological testing confirmed the antimicrobial efficacy of CAM and good biocompatibility of all CAM-nanofibers. The successful fiber formation and green production process with WES gives a promising outlook for industrial upscaling.


Subject(s)
Anti-Bacterial Agents , Bandages , Chloramphenicol , Drug Liberation , Liposomes , Nanofibers , Pectins , Polyethylene Glycols , Nanofibers/chemistry , Chloramphenicol/administration & dosage , Chloramphenicol/chemistry , Polyethylene Glycols/chemistry , Pectins/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Humans , Green Chemistry Technology/methods , Delayed-Action Preparations , Wound Healing/drug effects , Anti-Infective Agents/chemistry , Anti-Infective Agents/administration & dosage , Tensile Strength
15.
FEMS Microbes ; 5: xtae009, 2024.
Article in English | MEDLINE | ID: mdl-38606354

ABSTRACT

Enterococcus faecium (Efm) is a versatile pathogen, responsible for multidrug-resistant infections, especially in hospitalized immunocompromised patients. Its population structure has been characterized by diverse clades (A1, A2, and B (reclassified as E. lactis (Ela)), adapted to different environments, and distinguished by their resistomes and virulomes. These features only partially explain the predominance of clade A1 strains in nosocomial infections. We investigated in vitro interaction of 50 clinical isolates (clade A1 Efm) against 75 commensal faecal isolates from healthy humans (25 clade A2 Efm and 50 Ela). Only 36% of the commensal isolates inhibited clinical isolates, while 76% of the clinical isolates inhibited commensal isolates. The most apparent overall differences in inhibition patterns were presented between clades. The inhibitory activity was mainly mediated by secreted, proteinaceous, heat-stable compounds, likely indicating an involvement of bacteriocins. A custom-made database targeting 76 Bacillota bacteriocins was used to reveal bacteriocins in the genomes. Our systematic screening of the interactions between nosocomial and commensal Efm and Ela on a large scale suggests that, in a clinical setting, nosocomial strains not only have an advantage over commensal strains due to their possession of AMR genes, virulence factors, and resilience but also inhibit the growth of commensal strains.

16.
Microbiology (Reading) ; 159(Pt 10): 2001-2013, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23873783

ABSTRACT

The human body is constantly challenged by a variety of commensal and pathogenic micro-organisms that trigger the immune system. Central in the first line of defence is the pattern-recognition receptor (PRR)-induced stimulation of the NFκB pathway, leading to NFκB activation. The subsequent production of pro-inflammatory cytokines and/or antimicrobial peptides results in recruitment of professional phagocytes and bacterial clearance. To overcome this, bacteria have developed mechanisms for targeted interference in every single step in the PRR-NFκB pathway to dampen host inflammatory responses. This review aims to briefly overview the PRR-NFκB pathway in relation to the immune response and give examples of the diverse bacterial evasion mechanisms including changes in the bacterial surface, decoy production and injection of effector molecules. Targeted regulation of inflammatory responses is needed and bacterial molecules developed for immune evasion could provide future anti-inflammatory agents.


Subject(s)
Bacteria/immunology , Cytokines/metabolism , Host-Pathogen Interactions , Immune Evasion , Immunity, Innate , NF-kappa B/metabolism , Receptors, Immunologic/metabolism , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Signal Transduction
17.
J Mater Chem B ; 11(33): 7972-7985, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37505112

ABSTRACT

Nucleic acid-based materials showcase an increasing potential for antimicrobial drug delivery. Although numerous reports on drug-loaded DNA nanoparticles outline their pivotal antibacterial activities, their potential as drug delivery systems against bacterial biofilms awaits further studies. Among different oligonucleotide structures, micellar nanocarriers derived from amphiphilic DNA strands are of particular interest due to their spontaneous self-assembly and high biocompatibility. However, their clinical use is hampered by structural instability upon cation depletion. In this work, we used a cationic amphiphilic antibiotic (polymyxin B) to stabilize DNA micelles destined to penetrate P. aeruginosa biofilms and exhibit antibacterial/antibiofilm properties. Our study highlights how the strong affinity of this antibiotic enhances the stability of the micelles and confirms that antibacterial activity of the novel micelles remains intact. Additionally, we show that PMB micelles can penetrate P. aeruginosa biofilms and impact their metabolic activity. Finally, PMB micelles were highly safe and biocompatible, highlighting their possible application against P. aeruginosa biofilm-colonized skin wounds.


Subject(s)
Micelles , Polymyxin B , Pseudomonas aeruginosa , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Biofilms , DNA
18.
Cell Mol Life Sci ; 68(5): 847-62, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20734105

ABSTRACT

The mitogen-activated protein kinase-activated protein kinase-5 (MK5) resides predominantly in the nucleus of resting cells, but p38(MAPK), extracellular signal-regulated kinases-3 and -4 (ERK3 and ERK4), and protein kinase A (PKA) induce nucleocytoplasmic redistribution of MK5. The mechanism by which PKA causes nuclear export remains unsolved. In the study reported here we demonstrated that Ser-115 is an in vitro PKA phosphoacceptor site, and that PKA, but not p38(MAPK), ERK3 or ERK4, is unable to redistribute MK5 S115A to the cytoplasm. However, the phospho-mimicking MK5 S115D mutant resides in the cytoplasm in untreated cells. While p38(MAPK), ERK3 and ERK4 fail to trigger nuclear export of the kinase dead T182A and K51E MK5 mutants, S115D/T182A and K51E/S115D mutants were able to enter the cytoplasm of resting cells. Finally, we demonstrated that mutations in Ser-115 affect the biological properties of MK5. Taken together, our results suggest that Ser-115 plays an essential role in PKA-regulated nuclear export of MK5, and that it also may regulate the biological functions of MK5.


Subject(s)
Active Transport, Cell Nucleus , Cyclic AMP-Dependent Protein Kinases/physiology , Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Amino Acid Sequence , Animals , Extracellular Signal-Regulated MAP Kinases/physiology , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Molecular Sequence Data , Mutation , PC12 Cells , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Rats , Serine/chemistry , Signal Transduction , p38 Mitogen-Activated Protein Kinases/physiology
19.
Front Microbiol ; 13: 1023083, 2022.
Article in English | MEDLINE | ID: mdl-36246245

ABSTRACT

Infected chronic skin wounds and other skin infections are increasingly putting pressure on the health care providers and patients. The pressure is especially concerning due to the rise of antimicrobial resistance and biofilm-producing bacteria that further impair treatment success. Therefore, innovative strategies for wound healing and bacterial eradication are urgently needed; utilization of materials with inherent biological properties could offer a potential solution. Chitosan is one of the most frequently used polymers in delivery systems. This bioactive polymer is often regarded as an attractive constituent in delivery systems due to its inherent antimicrobial, anti-inflammatory, anti-oxidative, and wound healing properties. However, lipid-based vesicles and liposomes are generally considered more suitable as delivery systems for skin due to their ability to interact with the skin structure and provide prolonged release, protect the antimicrobial compound, and allow high local concentrations at the infected site. To take advantage of the beneficial attributes of the lipid-based vesicles and chitosan, these components can be combined into chitosan-containing liposomes or chitosomes and chitosan-coated liposomes. These systems have previously been investigated for use in wound therapy; however, their potential in infected wounds is not fully investigated. In this study, we aimed to investigate whether both the chitosan-containing and chitosan-coated liposomes tailored for infected wounds could improve the antimicrobial activity of the membrane-active antimicrobial chlorhexidine, while assuring both the anti-inflammatory activity and cell compatibility. Chlorhexidine was incorporated into three different vesicles, namely plain (chitosan-free), chitosan-containing and chitosan-coated liposomes that were optimized for skin wounds. Their release profile, antimicrobial activities, anti-inflammatory properties, and cell compatibility were assessed in vitro. The vesicles comprising chitosan demonstrated slower release rate of chlorhexidine and high cell compatibility. Additionally, the inflammatory responses in murine macrophages treated with these vesicles were reduced by about 60% compared to non-treated cells. Finally, liposomes containing both chitosan and chlorhexidine demonstrated the strongest antibacterial effect against Staphylococcus aureus. Both chitosan-containing and chitosan-coated liposomes comprising chlorhexidine could serve as excellent platforms for the delivery of membrane-active antimicrobials to infected wounds as confirmed by improved antimicrobial performance of chlorhexidine.

20.
J Surg Res ; 171(1): 58-65, 2011 Nov.
Article in English | MEDLINE | ID: mdl-20400116

ABSTRACT

BACKGROUND: Pretreatment with ß-glucan has been shown to protect against regional ischemia-reperfusion injury, through inhibition of myocardial NF-κB activation. The aim was to examine whether ß-glucan pretreatment could protect against the global ischemia-reperfusion injury, which is encountered in the clinical setting during open heart surgery. MATERIALS AND METHODS: Twenty-one pigs were randomized to pretreatment with oral ß-glucan (SBGo, n = 7), pretreatment with i.p. ß-glucan (SBGip, n = 7), and untreated controls (n = 7). The pigs were subjected to cardiopulmonary bypass (CPB) with 1 h of global cardioplegic ischemia followed by wean from CPB and reperfusion for 4 h. Cardiac function was determined by a conductance catheter, and troponin T was sampled from the coronary sinus. Atrial biopsies obtained at baseline, following 30 min, and 3 h of reperfusion were analyzed for phosphorylated NF-κB by Western blot. RESULTS: Following reperfusion, phosphorylated NF-κB increased by 210% in the control group, 197% in the SBGo group, but was reduced by 5% in the SBGip group (P < 0.01 versus control). After 4 h of reperfusion, preload recruitable stroke work dropped by 19% in the control group and 25% in the SBGo group compared with 60% in the SBGip group (P < 0.01 versus control). The area under the curve for troponin T was larger in the SBGip group compared with the control group (P < 0.05) and the SBGo group (P < 0.01). CONCLUSION: Inhibition of NF-κB activation by i.p. ß-glucan does not protect against ischemia-reperfusion injury in pigs subjected to global ischemia and reperfusion, and may be associated with aggravation of ischemia-reperfusion injury.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , NF-kappa B/antagonists & inhibitors , beta-Glucans/pharmacology , Animals , Coronary Sinus/metabolism , Disease Models, Animal , Heart Arrest, Induced/adverse effects , NF-kappa B/metabolism , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/metabolism , Sus scrofa , Treatment Failure , Troponin T/metabolism , Ventricular Function, Left/physiology
SELECTION OF CITATIONS
SEARCH DETAIL