Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.679
1.
Arch Microbiol ; 206(6): 251, 2024 May 10.
Article En | MEDLINE | ID: mdl-38727840

The prevalence of Candida albicans infection has increased during the past few years, which contributes to the need for new, effective treatments due to the increasing concerns regarding antifungal drug toxicity and multidrug resistance. Butyl isothiocyanate (butylITC) is a glucosinolate derivative, and has shown a significant antifungal effect contrary to Candida albicans. Additionally, how butylITC affects the virulence traits of C. albicans and molecular mode of actions are not well known. Present study shows that at 17.36 mM concentration butylITC inhibit planktonic growth. butylITC initially slowed the hyphal transition at 0.542 mM concentration. butylITC hampered biofilm development, and inhibits biofilm formation at 17.36 mM concentration which was analysed using metabolic assay (XTT assay) and Scanning Electron Microscopy (SEM). In addition, it was noted that butylITC inhibits ergosterol biosynthesis. The permeability of cell membranes was enhanced by butylITC treatment. Moreover, butylITC arrests cells at S-phase and induces intracellular Reactive Oxygen Species (ROS) accumulation in C. albicans. The results suggest that butylITC may have a dual mode of action, inhibit virulence factors and modulate cellular processes like inhibit ergosterol biosynthesis, cell cycle arrest, induces ROS production which leads to cell death in C. albicans.


Antifungal Agents , Biofilms , Candida albicans , Cell Membrane , Isothiocyanates , Oxidative Stress , Reactive Oxygen Species , Candida albicans/drug effects , Candida albicans/physiology , Biofilms/drug effects , Antifungal Agents/pharmacology , Isothiocyanates/pharmacology , Oxidative Stress/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Reactive Oxygen Species/metabolism , Microbial Sensitivity Tests , Cell Cycle/drug effects , Hyphae/drug effects , Hyphae/growth & development , Ergosterol/metabolism
2.
BMC Microbiol ; 24(1): 154, 2024 May 04.
Article En | MEDLINE | ID: mdl-38704559

BACKGROUND: Side effects associated with antimicrobial drugs, as well as their high cost, have prompted a search for low-cost herbal medicinal substances with fewer side effects. These substances can be used as supplements to medicine or to strengthen their effects. The current study investigated the effect of oleuropein on the inhibition of fungal and bacterial biofilm in-vitro and at the molecular level. MATERIALS AND METHODS: In this experimental study, antimicrobial properties were evaluated using microbroth dilution method. The effect of oleuropein on the formation and eradication of biofilm was assessed on 96-well flat bottom microtiter plates and their effects were observed through scanning electron microscopy (SEM). Its effect on key genes (Hwp1, Als3, Epa1, Epa6, LuxS, Pfs) involved in biofilm formation was investigated using the quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) method. RESULTS: The minimum inhibitory concentration (MIC) and minimum fungicidal/bactericidal concentration (MFC/MBC) for oleuropein were found to be 65 mg/ml and 130 mg/ml, respectively. Oleuropein significantly inhibited biofilm formation at MIC/2 (32.5 mg/ml), MIC/4 (16.25 mg/ml), MIC/8 (8.125 mg/ml) and MIC/16 (4.062 mg/ml) (p < 0.0001). The anti-biofilm effect of oleuropein was confirmed by SEM. RT-qPCR indicated significant down regulation of expression genes involved in biofilm formation in Candida albicans (Hwp1, Als3) and Candida glabrata (Epa1, Epa6) as well as Escherichia coli (LuxS, Pfs) genes after culture with a MIC/2 of oleuropein (p < 0.0001). CONCLUSIONS: The results indicate that oleuropein has antifungal and antibacterial properties that enable it to inhibit or destroy the formation of fungal and bacterial biofilm.


Antifungal Agents , Biofilms , Candida albicans , Candida glabrata , Escherichia coli , Fluconazole , Iridoid Glucosides , Iridoids , Microbial Sensitivity Tests , Biofilms/drug effects , Biofilms/growth & development , Iridoid Glucosides/pharmacology , Candida glabrata/drug effects , Candida glabrata/physiology , Candida glabrata/genetics , Candida albicans/drug effects , Candida albicans/genetics , Candida albicans/physiology , Escherichia coli/drug effects , Escherichia coli/genetics , Iridoids/pharmacology , Fluconazole/pharmacology , Antifungal Agents/pharmacology , Drug Resistance, Fungal , Anti-Bacterial Agents/pharmacology , Microscopy, Electron, Scanning
3.
Sci Rep ; 14(1): 9354, 2024 04 23.
Article En | MEDLINE | ID: mdl-38653744

Phage-antibiotic combinations to treat bacterial infections are gaining increased attention due to the synergistic effects often observed when applying both components together. Most studies however focus on a single pathogen, although in many clinical cases multiple species are present at the site of infection. The aim of this study was to investigate the anti-biofilm activity of phage-antibiotic/antifungal combinations on single- and dual-species biofilms formed by P. aeruginosa and the fungal pathogen Candida albicans. The Pseudomonas phage Motto in combination with ciprofloxacin had significant anti-biofilm activity. We then compared biofilms formed by P. aeruginosa alone with the dual-species biofilms formed by bacteria and C. albicans. Here, we found that the phage together with the antifungal fluconazole was active against 6-h-old dual-species biofilms but showed only negligible activity against 24-h-old biofilms. This study lays the first foundation for potential therapeutic approaches to treat co-infections caused by bacteria and fungi using phage-antibiotic combinations.


Anti-Bacterial Agents , Antifungal Agents , Biofilms , Candida albicans , Ciprofloxacin , Pseudomonas Phages , Pseudomonas aeruginosa , Biofilms/drug effects , Biofilms/growth & development , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/virology , Antifungal Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas Phages/physiology , Candida albicans/drug effects , Candida albicans/physiology , Ciprofloxacin/pharmacology , Fluconazole/pharmacology , Microbial Sensitivity Tests
4.
mBio ; 15(5): e0018424, 2024 May 08.
Article En | MEDLINE | ID: mdl-38624207

Microorganisms resist fluoride toxicity using fluoride export proteins from one of several different molecular families. Cariogenic species Streptococcus mutans and Candida albicans extrude intracellular fluoride using a CLCF F-/H+ antiporter and FEX fluoride channel, respectively, whereas oral commensal eubacteria, such as Streptococcus gordonii, export fluoride using a Fluc fluoride channel. In this work, we examine how genetic knockout of fluoride export impacts pathogen fitness in single-species and three-species dental biofilm models. For biofilms generated using S. mutans with the genetic knockout of the CLCF transporter, exposure to low fluoride concentrations decreased S. mutans counts, synergistically reduced the populations of C. albicans, increased the relative proportion of oral commensal S. gordonii, and reduced properties associated with biofilm pathogenicity, including acid production and hydroxyapatite dissolution. Biofilms prepared with C. albicans with genetic knockout of the FEX channel also exhibited reduced fitness in the presence of fluoride but to a lesser degree. Imaging studies indicate that S. mutans is highly sensitive to fluoride, with the knockout strain undergoing complete lysis when exposed to low fluoride for a moderate amount of time. Biochemical purification of the S. mutans CLCF transporter and functional reconstitution establishes that the functional protein is a dimer encoded by a single gene. Together, these findings suggest that fluoride export by oral pathogens can be targeted by specific inhibitors to restore biofilm symbiosis in dental biofilms and that S. mutans is especially susceptible to fluoride toxicity. IMPORTANCE: Dental caries is a globally prevalent condition that occurs when pathogenic species, including Streptococcus mutans and Candida albicans, outcompete beneficial species, such as Streptococcus gordonii, in the dental biofilm. Fluoride is routinely used in oral hygiene to prevent dental caries. Fluoride also has antimicrobial properties, although most microbes possess fluoride exporters to resist its toxicity. This work shows that sensitization of cariogenic species S. mutans and C. albicans to fluoride by genetic knockout of fluoride exporters alters the microbial composition and pathogenic properties of dental biofilms. These results suggest that the development of drugs that inhibit fluoride exporters could potentiate the anticaries effect of fluoride in over-the-counter products like toothpaste and mouth rinses. This is a novel strategy to treat dental caries.


Biofilms , Candida albicans , Fluorides , Streptococcus gordonii , Streptococcus mutans , Biofilms/drug effects , Biofilms/growth & development , Candida albicans/drug effects , Candida albicans/genetics , Candida albicans/physiology , Candida albicans/metabolism , Streptococcus mutans/genetics , Streptococcus mutans/drug effects , Streptococcus mutans/metabolism , Streptococcus mutans/physiology , Fluorides/pharmacology , Fluorides/metabolism , Streptococcus gordonii/drug effects , Streptococcus gordonii/genetics , Streptococcus gordonii/physiology , Streptococcus gordonii/metabolism , Gene Knockout Techniques , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Dental Caries/microbiology
5.
J Appl Oral Sci ; 32: e20230326, 2024.
Article En | MEDLINE | ID: mdl-38656049

OBJECTIVE: This study evaluated the surface roughness, wettability and adhesion of multispecies biofilms (Candida albicans, Staphylococcus aureus and Streptococcus mutans) on 3D-printed resins for complete denture bases and teeth compared to conventional resins (heat-polymerized acrylic resin; artificial pre-fabricated teeth). METHODOLOGY: Circular specimens (n=39; 6.0 mm Ø × 2.0 mm) of each group were subjected to roughness (n=30), wettability (n=30) and biofilm adhesion (n=9) tests. Three roughness measurements were taken by laser confocal microscopy and a mean value was calculated. Wettability was evaluated by the contact angle of sessile drop method, considering the mean of the three evaluations per specimen. In parallel, microorganism adhesion to resin surfaces was evaluated using a multispecies biofilm model. Microbial load was evaluated by determining the number of Colony Forming Units (CFU/mL) and by scanning electron microscopy (SEM). Data were subjected to the Wald test in a generalized linear model with multiple comparisons and Bonferroni adjustment, as well as two-way ANOVA (α=5%). RESULTS: The roughness of the conventional base resin (0.01±0.04) was lower than that of the conventional tooth (0.14±0.04) (p=0.023) and 3D-printed base (0.18±0.08) (p<0.001). For wettability, conventional resin (84.20±5.57) showed a higher contact angle than the 3D-printed resin (60.58±6.18) (p<0.001). Higher microbial loads of S. mutans (p=0.023) and S. aureus (p=0.010) were observed on the surface of the conventional resin (S. mutans: 5.48±1.55; S. aureus: 7.01±0.57) compared to the 3D-printed resin (S. mutans: 4.11±1.96; S. aureus: 6.42±0.78). The adhesion of C. albicans was not affected by surface characteristics. The conventional base resin showed less roughness than the conventional dental resin and the printed base resin. CONCLUSION: The 3D-printed resins for base and tooth showed less hydrophobicity and less adhesion of S. mutans and S. aureus than conventional resins.


Acrylic Resins , Bacterial Adhesion , Biofilms , Candida albicans , Denture Bases , Materials Testing , Microscopy, Confocal , Microscopy, Electron, Scanning , Printing, Three-Dimensional , Staphylococcus aureus , Streptococcus mutans , Surface Properties , Wettability , Streptococcus mutans/physiology , Staphylococcus aureus/physiology , Candida albicans/physiology , Denture Bases/microbiology , Acrylic Resins/chemistry , Analysis of Variance , Reproducibility of Results , Denture, Complete/microbiology , Reference Values , Colony Count, Microbial , Linear Models
6.
Methods Enzymol ; 696: 155-174, 2024.
Article En | MEDLINE | ID: mdl-38658078

The interactions between communities of microorganisms inhabiting the dental biofilm is a major determinant of oral health. These biofilms are periodically exposed to high concentrations of fluoride, which is present in almost all oral healthcare products. The microbes resist fluoride through the action of membrane export proteins. This chapter describes the culture, growth and harvest conditions of model three-species dental biofilm comprised of cariogenic pathogens Streptococcus mutans and Candida albicans and the commensal bacterium Streptococcus gordonii. In order to examine the role of fluoride export by S. mutans in model biofilms, procedures for generating a strain of S. mutans with a genetic knockout of the fluoride exporter are described. We present a case study examining the effects of this mutant strain on the biofilm mass, acid production and mineral dissolution under exposure to low levels of fluoride. These general approaches can be applied to study the effects of any gene of interest in physiologically realistic multispecies oral biofilms.


Biofilms , Candida albicans , Fluorides , Streptococcus gordonii , Streptococcus mutans , Biofilms/drug effects , Biofilms/growth & development , Streptococcus mutans/drug effects , Streptococcus mutans/genetics , Streptococcus mutans/physiology , Streptococcus mutans/metabolism , Streptococcus mutans/growth & development , Fluorides/pharmacology , Fluorides/metabolism , Candida albicans/drug effects , Candida albicans/genetics , Candida albicans/metabolism , Candida albicans/physiology , Streptococcus gordonii/drug effects , Streptococcus gordonii/genetics , Streptococcus gordonii/physiology , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Resistance, Bacterial/genetics , Dental Caries/microbiology
7.
Methods Enzymol ; 696: 3-24, 2024.
Article En | MEDLINE | ID: mdl-38658085

Tight regulation of molecules moving through the cell membrane is particularly important for free-living microorganisms because of their small cell volumes and frequent changes in the chemical composition of the extracellular environment. This is true for nutrients, but even more so for toxic molecules. Traditionally, the transport of these diverse molecules in microorganisms has been studied on cell populations rather than on single cells, mainly because of technical difficulties. The goal of this chapter is to make available a detailed method to prepare yeast spheroplasts to study the movement of fluoride ions across the plasma membrane of single cells by the patch-clamp technique. In this procedure, three steps are critical to achieve high resistance (GΩ) seals between the membrane and the glass electrode: (1) appropriate removal of the cell wall by enzymatic treatment; (2) balance between the osmotic strength of sealing solutions and cell membrane turgor; and (3) meticulous morphological inspection of spheroplasts suitable for gigaseal formation. We show now that this method, originally developed for Saccharomyces cerevisiae, can also be applied to Candida albicans, an opportunistic human pathogen.


Candida albicans , Fluorides , Patch-Clamp Techniques , Saccharomyces cerevisiae , Spheroplasts , Saccharomyces cerevisiae/metabolism , Candida albicans/metabolism , Candida albicans/physiology , Fluorides/chemistry , Patch-Clamp Techniques/methods , Spheroplasts/metabolism , Cell Membrane/metabolism , Ion Channels/metabolism
8.
FEMS Yeast Res ; 242024 Jan 09.
Article En | MEDLINE | ID: mdl-38592962

How mutations in mitochondrial electron transport chain (ETC) proteins impact the cell cycle of Candida albicans was investigated in this study. Using genetic null mutants targeting ETC complexes I (CI), III (CIII), and IV (CIV), the cell cycle stages (G0/G1, S phase, and G2/M) were analyzed via fluorescence-activated cell sorting (FACS). Four CI null mutants exhibited distinct alterations, including extended S phase, shortened G2/M population, and a reduction in cells size exceeding 10 µM. Conversely, CIII mutants showed an increased population in G1/G0 phase. Among four CI mutants, ndh51Δ/Δ and goa1Δ/Δ displayed aberrant cell cycle patterns correlated with previously reported cAMP/PKA downregulation. Specifically, nuo1Δ/Δ and nuo2Δ/Δ mutants exhibited increased transcription of RIM15, a central hub linking cell cycle with nutrient-dependent TOR1 and cAMP/PKA pathways and Snf1 aging pathway. These findings suggest that suppression of TOR1 and cAMP/PKA pathways or enhanced Snf1 disrupts cell cycle progression, influencing cell longevity and growth among CI mutants. Overall, our study highlights the intricate interplay between mitochondrial ETC, cell cycle, and signaling pathways.


Candida albicans , Mitochondria , Candida albicans/physiology , S Phase , Mitochondria/metabolism , Cell Cycle , Cell Division
9.
Virulence ; 15(1): 2334085, 2024 Dec.
Article En | MEDLINE | ID: mdl-38528835

Candida albicans is an immunogen for anti-Saccharomyces cerevisiae antibodies (ASCA), a serological marker of Crohn's disease. ASCA has also been reported in other autoimmune diseases, including coeliac disease (CeD). A strong antibody response against Hwp1, a protein associated with invasive hyphal form of C. albicans which presents peptide sequence homologies with gliadin, has also been described in CeD. This observation supports the hypothesis that C. albicans hyphal transition in C. albicans may trigger CeD onset through a mechanism of molecular/antigenic mimicry. In this study, we assessed whether the anti-C. albicans oligomannose and anti-Hwp1 protein responses may be linked despite their different pathophysiological significance. The measurement of ASCA levels in a cohort of patients involved in our previous Hwp1 study showed a significant correlation between the two biomarkers. This new observation further reinforces the link between C. albicans and CeD.


Celiac Disease , Crohn Disease , Humans , Candida albicans/physiology , Celiac Disease/microbiology , Antibodies, Fungal , Antibody Formation
10.
Microb Pathog ; 189: 106606, 2024 Apr.
Article En | MEDLINE | ID: mdl-38437994

Fungal keratitis (FK) is a highly blinding infectious corneal disease caused by pathogenic fungi. Candida albicans (C. albicans) is one of the main pathogens of fungal keratitis. Extracellular vesicles (EVs), lipid bilayer compartments released by almost all living cells, including fungi, have garnered attention for their role in pathogenic microbial infection and host immune responses in recent years. Studies have reported that pretreating the host with fungal EVs can reduce the inflammatory response of the host when attacked by fungi and reduce the lethality of fungal infection. However, there are no studies that have evaluated whether C. albicans EVs can modulate the inflammatory response associated with C. albicans keratitis. Our study revealed that C. albicans EVs could activate the polymorphonuclear cells (PMNs) and promote their secretion of proinflammatory cytokines and nitric oxide (NO), enhance their phagocytic and fungicidal abilities against C. albicans. C. albicans EVs also induced a proinflammatory response in RAW264.7 cells, which was characterized by increased production of inflammatory cytokines and elevated expression of the chemokine CCL2. Similarly, stimulation of C. albicans EVs to RAW264.7 cells also enhanced the phagocytosis and killing ability of cells against C. albicans. Besides, in our in vivo experiments, after receiving subconjunctival injection of C. albicans EVs, C57BL/6 mice were infected with C. albicans. The results demonstrated that pre-exposure to C. albicans EVs could effectively diminish the severity of keratitis, reduce fungal load and improve prognosis. Overall, we conclude that C. albicans EVs can modulate the function of immune cells and play a protective role in C. albicans keratitis.


Extracellular Vesicles , Keratitis , Animals , Mice , Candida albicans/physiology , Mice, Inbred C57BL , Keratitis/microbiology , Cytokines
11.
BMC Oral Health ; 24(1): 303, 2024 Mar 04.
Article En | MEDLINE | ID: mdl-38439020

BACKGROUND: The present systematic review and meta-analysis investigated the available evidence about the adherence of Candida Albicans to the digitally-fabricated acrylic resins (both milled and 3D-printed) compared to the conventional heat-polymerized acrylic resins. METHODS: This study followed the guidelines of the Preferred Reporting Items for Systematic Review and Meta-analyses (PRISMA). A comprehensive search of online databases/search tools (Web of Science, Scopus, PubMed, Ovid, and Google Scholar) was conducted for all relevant studies published up until May 29, 2023. Only in-vitro studies comparing the adherence of Candida albicans to the digital and conventional acrylic resins were included. The quantitative analyses were performed using RevMan v5.3 software. RESULTS: Fourteen studies were included, 11 of which were meta-analyzed based on Colony Forming Unit (CFU) and Optical Density (OD) outcome measures. The pooled data revealed significantly lower candida colonization on the milled digitally-fabricated compared to the heat-polymerized conventionally-fabricated acrylic resin materials (MD = - 0.36; 95%CI = - 0.69, - 0.03; P = 0.03 and MD = - 0.04; 95%CI = - 0.06, - 0.01; P = 0.0008; as measured by CFU and OD respectively). However, no differences were found in the adhesion of Candida albicans between the 3D-printed digitally-fabricated compared to the heat-polymerized conventionally-fabricated acrylic resin materials (CFU: P = 0.11, and OD: P = 0.20). CONCLUSION: The available evidence suggests that candida is less likely to adhere to the milled digitally-fabricated acrylic resins compared to the conventional ones.


Acrylic Resins , Candida albicans , Candida albicans/physiology , Databases, Factual
12.
Front Immunol ; 15: 1295168, 2024.
Article En | MEDLINE | ID: mdl-38384468

Candida albicans remains the predominant cause of fungal infections, where adhered microbial cells form biofilms - densely packed communities. The central feature of C. albicans biofilms is the production of an extracellular matrix (ECM) consisting of polymers and extracellular nucleic acids (eDNA, eRNA), which significantly impedes the infiltration of host cells. Neutrophils, as crucial players in the innate host defense, employ several mechanisms to eradicate the fungal infection, including NETosis, endocytosis, or the release of granules containing, among others, antimicrobial peptides (AMPs). The main representative of these is the positively charged peptide LL-37 formed from an inactive precursor (hCAP18). In addition to its antimicrobial functions, this peptide possesses a propensity to interact with negatively charged molecules, including nucleic acids. Our in vitro studies have demonstrated that LL-37 contacting with C. albicans nucleic acids, isolated from biofilm, are complexed by the peptide and its shorter derivatives, as confirmed by electrophoretic mobility shift assays. We indicated that the generation of the complexes induces discernible alterations in the neutrophil response to fungal nucleic acids compared to the effects of unconjugated molecules. Our analyses involving fluorescence microscopy, flow cytometry, and Western blotting revealed that stimulation of neutrophils with DNA:LL-37 or RNA:LL-37 complexes hamper the activation of pro-apoptotic caspases 3 and 7 and fosters increased activation of anti-apoptotic pathways mediated by the Mcl-1 protein. Furthermore, the formation of complexes elicits a dual effect on neutrophil immune response. Firstly, they facilitate increased nucleic acid uptake, as evidenced by microscopic observations, and enhance the pro-inflammatory response, promoting IL-8 production. Secondly, the complexes detection suppresses the production of reactive oxygen species and attenuates NETosis activation. In conclusion, these findings may imply that the neutrophil immune response shifts toward mobilizing the immune system as a whole, rather than inactivating the pathogen locally. Our findings shed new light on the intricate interplay between the constituents of the C. albicans biofilm and the host's immune response and indicate possible reasons for the elimination of NETosis from the arsenal of the neutrophil response during contact with the fungal biofilm.


Candida albicans , Nucleic Acids , Candida albicans/physiology , Neutrophils , Cathelicidins/pharmacology , Nucleic Acids/metabolism , Biofilms
13.
Future Microbiol ; 19: 307-316, 2024 Mar.
Article En | MEDLINE | ID: mdl-38358357

Background: The interaction between the host and Candida albicans is dynamic and intricate. We performed proteomic analysis to explore monocyte-C. albicans hyphae interaction. Materials & methods: Primary human monocytes were stimulated by heat-killed C. albicans hyphae and their proteins were profiled by tandem liquid chromatography with mass spectrometry (LC-MS/MS). Results: Based on the protein database of different species for analysis, we found that stimulation of monocytes by hyphae was accompanied by upregulation of histones and activation of extracellular traps (ETs) formation pathway. Meanwhile, monocyte ETs (MoETs) were evoked by synthesis or alteration of C. albicans cell wall proteins expression during the morphological switch to hyphal. Conclusion: MoETs formation is linked to cell wall proteins of C. albicans hyphae.


Candida albicans , Extracellular Traps , Humans , Candida albicans/physiology , Monocytes , Extracellular Traps/metabolism , Hyphae , Proteomics , Chromatography, Liquid , Tandem Mass Spectrometry , Fungal Proteins/metabolism
14.
Theranostics ; 14(4): 1781-1793, 2024.
Article En | MEDLINE | ID: mdl-38389835

Candida albicans and Porphyromonas gingivalis are prevalent in the subgingival area where the frequency of fungal colonization increases with periodontal disease. Candida's transition to a pathogenic state and its interaction with P. gingivalis exacerbate periodontal disease severity. However, current treatments for these infections differ, and combined therapy remains unexplored. This work is based on an antimicrobial peptide that is therapeutic and induces a color change in a nanoparticle reporter. Methods: We built and characterized two enzyme-activatable prodrugs to treat and detect C. albicans and P. gingivalis via the controlled release of the antimicrobial peptide. The zwitterionic prodrug quenches the antimicrobial peptide's activity until activation by a protease inherent to the pathogens (SAP9 for C. albicans and RgpB for P. gingivalis). The toxicity of the intact prodrugs was evaluated against fungal, bacterial, and mammalian cells. Therapeutic efficacy was assessed through microscopy, disk diffusion, and viability assays, comparing the prodrug to the antimicrobial peptide alone. Finally, we developed a colorimetric detection system based on the aggregation of plasmonic nanoparticles. Results: The intact prodrugs showed negligible toxicity to cells absent a protease trigger. The therapeutic impact of the prodrugs was comparable to that of the antimicrobial peptide alone, with a minimum inhibitory concentration of 3.1 - 16 µg/mL. The enzymatic detection system returned a detection limit of 10 nM with gold nanoparticles and 3 nM with silver nanoparticles. Conclusion: This approach offers a convenient and selective protease sensing and protease-induced treatment mechanism based on bioinspired antimicrobial peptides.


Metal Nanoparticles , Periodontal Diseases , Prodrugs , Animals , Porphyromonas gingivalis/physiology , Candida albicans/physiology , Peptide Hydrolases , Antimicrobial Peptides , Prodrugs/pharmacology , Delayed-Action Preparations , Gold , Silver , Endopeptidases , Mammals
15.
Int Endod J ; 57(6): 700-712, 2024 Jun.
Article En | MEDLINE | ID: mdl-38404175

AIM: To evaluate the influence of different preparation tapers on the reduction in planktonic bacteria and biofilms of Enterococcus faecalis and Candida albicans in the apical third (4 mm) of the mesial roots of mandibular molars, correlating decontamination with canal shape. METHODOLOGY: After microtomography analysis for morphological standardization of the canals, 48 mandibular molar roots, each containing two canals (96 canals), were contaminated with E. faecalis and C. albicans and divided into four groups (n = 11) for canal instrumentation using ProDesign Logic 2 files with different tapers G (.03): # 25.03; G (.04): # 25.04; G (.05): # 25.05; and G (.06): # 25.06 and irrigation with 2.5% sodium hypochlorite. Four roots were examined under a scanning electron microscope (SEM) to qualitatively assess biofilm formation. Eight roots were used as the negative control group (samples were not contaminated). Bacteriological samples were taken exclusively from the apical third of the roots before and after chemical-mechanical preparation and bacterial counts were determined (CFU/mL). The final micro-CT scan was used to quantify the volume variation and unprepared canal area in the apical third. Statistical analysis was performed using the Kruskal-Wallis, Student-Newman-Keuls and Wilcoxon tests for analysis of microbiological data. anova and the Tukey or Games-Howell test were used for analysis of micro-CT data and Spearman's test for correlations (α = 5%). RESULTS: All groups showed a significant reduction in bacteria (p < .05), with no statistically significant difference between groups. There was no significant difference in per cent volume increase between groups. The unprepared area (Δ%) was affected by the file used (p = .026) and was significantly lower for G (.06) compared to G (.03). There was no statistically significant correlation among bacterial reduction, volume and unprepared area (p > .05). CONCLUSION: The different preparation tapers influenced root canal shaping in the apical third but did not improve decontamination in this region.


Biofilms , Candida albicans , Dental Pulp Cavity , Enterococcus faecalis , Root Canal Preparation , X-Ray Microtomography , X-Ray Microtomography/methods , Humans , Root Canal Preparation/instrumentation , Root Canal Preparation/methods , Candida albicans/isolation & purification , Candida albicans/physiology , Dental Pulp Cavity/microbiology , Dental Pulp Cavity/diagnostic imaging , Sodium Hypochlorite/therapeutic use , Sodium Hypochlorite/pharmacology , Microscopy, Electron, Scanning , Molar/microbiology , Plankton , Root Canal Irrigants/administration & dosage , Root Canal Irrigants/therapeutic use , In Vitro Techniques , Tooth Apex/microbiology , Tooth Apex/diagnostic imaging
16.
Cells ; 13(2)2024 01 10.
Article En | MEDLINE | ID: mdl-38247818

Candida albicans is an opportunistic pathogenic yeast that can survive in both normoxic and hypoxic environments. The involvement of C. albicans secretome on host biological processes has been demonstrated. However, the immunoregulatory function of C. albicans secretome released under hypoxic condition remains unclear. This study demonstrated the differences in cytokine responses and protein profiles between secretomes prepared under normoxic and hypoxic conditions. Furthermore, the immunoregulatory effects of heat shock protein SSA1(Ssa1), a protein candidate enriched in the hypoxic secretome, were investigated. Stimulation of mouse bone marrow-derived macrophages (BMMs) with Ssa1 resulted in the significant production of interleukin (IL)-10, IL-6, and tumor necrosis factor (TNF)-α as well as the significant expression of M2b macrophage markers (CD86, CD274 and tumor necrosis factor superfamily member 14), suggesting that C. albicans Ssa1 may promote macrophage polarization towards an M2b-like phenotype. Proteomic analysis of Ssa1-treated BMMs also revealed that Ssa1 reduced inflammation-related factors (IL-18-binding protein, IL-1 receptor antagonist protein, OX-2 membrane glycoprotein and cis-aconitate decarboxylase) and enhanced the proteins involved in anti-inflammatory response (CMRF35-like molecule 3 and macrophage colony-stimulating factor 1 receptor). Based on these results, we investigated the effect of Ssa1 on C. albicans infection and showed that Ssa1 inhibited the uptake of C. albicans by BMMs. Taken together, our results suggest that C. albicans alters its secretome, particularly by promoting the release of Ssa1, to modulate host immune response and survive under hypoxic conditions.


Candida albicans , Heat-Shock Proteins , Macrophages , Animals , Mice , Candida albicans/metabolism , Candida albicans/physiology , Heat-Shock Proteins/metabolism , Hypoxia , Proteomics , Secretome , Tumor Necrosis Factors , Host-Parasite Interactions , Macrophages/immunology , Macrophages/metabolism
17.
Inflammation ; 47(1): 191-208, 2024 Feb.
Article En | MEDLINE | ID: mdl-37740789

Candida albicans is a common opportunistic pathogenic fungus. The innate immune system provides the first-line host defense against fungal infection. Innate immune receptors and downstream molecules have been shown to play various roles during fungal infection. The innate immune receptor MDA5, encoded by the gene Ifih1, enhances host resistance against viral and Aspergillus fumigatus infection by inducing the production of interferons (IFNs). However, the role of MDA5 in C. albicans infection is still unclear. Here, we found that the gene expression levels of IFIH1 were significantly increased in innate immune cells after C. albicans stimulation through human bioinformatics analysis or mouse experiments. Through in vivo study, MDA5 was shown to enhance host susceptibility to C. albicans infection independent of IFN production. Instead, MDA5 exerted its influence on macrophages and kidneys by modulating the expression of Noxa, Bcl2, and Bax, thereby promoting apoptosis. Additionally, MDA5 compromised killing capabilities of macrophage by inhibition iNOS expression. The introduction of the apoptosis inducer PAC1 further impaired macrophage functions, mimicking the enhancing effect of MDA5 on C. albicans infection. Furthermore, the administration of macrophage scavengers increased the susceptibility of Ifih1-/- mice to C. albicans. The founding suggests that MDA5 promote host susceptibility to invasive C. albicans by enhancing cell apoptosis and compromising macrophage functions, making MDA5 a target to treat candidiasis.


Candida albicans , Candidiasis , Animals , Humans , Mice , Apoptosis , Candida albicans/physiology , Interferon-Induced Helicase, IFIH1 , Macrophages , Phagocytosis
18.
PLoS Pathog ; 19(12): e1011833, 2023 Dec.
Article En | MEDLINE | ID: mdl-38091321

The ability of the fungus Candida albicans to filament and form biofilms contributes to its burden as a leading cause of hospital-acquired infections. Biofilm development involves an interconnected transcriptional regulatory network (TRN) consisting of nine transcription factors (TFs) that bind both to their own regulatory regions and to those of the other network TFs. Here, we show that seven of the nine TFs in the C. albicans biofilm network contain prion-like domains (PrLDs) that have been linked to the ability to form phase-separated condensates. Construction of PrLD mutants in four biofilm TFs reveals that these domains are essential for filamentation and biofilm formation in C. albicans. Moreover, biofilm PrLDs promote the formation of phase-separated condensates in the nuclei of live cells, and PrLD mutations that abolish phase separation (such as the removal of aromatic residues) also prevent biofilm formation. Biofilm TF condensates can selectively recruit other TFs through PrLD-PrLD interactions and can co-recruit RNA polymerase II, implicating condensate formation in the assembly of active transcriptional complexes. Finally, we show that PrLD mutations that block the phase separation of biofilm TFs also prevent filamentation in an in vivo model of gastrointestinal colonization. Together, these studies associate transcriptional condensates with the regulation of filamentation and biofilm formation in C. albicans, and highlight how targeting of PrLD-PrLD interactions could prevent pathogenesis by this species.


Candida albicans , Transcription Factors , Candida albicans/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Hyphae , Biofilms , Fungal Proteins/genetics , Fungal Proteins/metabolism
19.
Arch Razi Inst ; 78(3): 1005-1015, 2023 06.
Article En | MEDLINE | ID: mdl-38028848

This study aimed to investigate the antibacterial and antifungal activities of selenium nanoparticles (SeNPs) and berberine (BBR) despite antibiotic resistance against Klebsiella pneumoniae and Candida albicans. Cells of K. pneumoniae and C. albicans were treated with solutions of different concentrations of each bare SeNPs, BBR, and BBR-loaded SeNPs (BLS) using the disk diffusion method. The results indicated that the activities of SeNPs, BBR, and BLS were statistically significant (P<0.05) when the concentration of all agents increased. Moreover, it was found that BLS had a statistically significant effect against K. pneumoniae and C. albicans, compared to SeNPs and BBR alone (P<0.05). The largest zones of inhibition of SeNPs were 14 and 16 mm toward K. pneumoniae and C. albicans, respectively, at the concentration of 20 Mml, compared to the concentrations of 10 and 15 Mml. Furthermore, BBR showed a maximum zone of inhibition at the concentration of 1,200 mg (15 mm for K. pneumoniae and 18 mm for C. albicans) and it was statistically significant in comparison with other concentrations of 400 and 800 mg. In addition, the BLS underwent a statistically significant increase (P<0.05) when the concentration increased and it registered a large zone of inhibition of 22 and 25 mm against K. pneumoniae and C. albicans, respectively, at 20 Mml of SeNPs: 1,200 mg BBR, compared to 10 Mml of SeNPs: 400 mg BBR and 15 Mml of SeNPs: 800 mg BBR. Based on the results of the current study, there was a statistically synergistic effect of BBR-loaded SeNPs, compared to that of BBR and Se nanoparticles, only in the case of both K. pneumoniae and C. albicans. This study is promising as a blueprint for the enhancement of weak antimicrobial agents and their return to their previous role as antibiotics.


Berberine , Nanoparticles , Selenium , Animals , Candida albicans/physiology , Selenium/pharmacology , Berberine/pharmacology , Klebsiella pneumoniae , Microbial Sensitivity Tests , Drug Resistance, Microbial , Anti-Bacterial Agents/pharmacology
20.
Gut Microbes ; 15(2): 2287618, 2023 Dec.
Article En | MEDLINE | ID: mdl-38017705

Candida albicans is a commensal yeast present in the gut of most healthy individuals but with highly variable concentrations. However, little is known about the host factors that influence colonization densities. We investigated how microbiota, host lifestyle factors, and genetics could shape C. albicans intestinal carriage in 695 healthy individuals from the Milieu Intérieur cohort. C. albicans intestinal carriage was detected in 82.9% of the subjects using quantitative PCR. Using linear mixed models and multiway-ANOVA, we explored C. albicans intestinal levels with regard to gut microbiota composition and lifestyle factors including diet. By analyzing shotgun metagenomics data and C. albicans qPCR data, we showed that Intestinimonas butyriciproducens was the only gut microbiota species whose relative abundance was negatively correlated with C. albicans concentration. Diet is also linked to C. albicans growth, with eating between meals and a low-sodium diet being associated with higher C. albicans levels. Furthermore, by Genome-Wide Association Study, we identified 26 single nucleotide polymorphisms suggestively associated with C. albicans colonization. In addition, we found that the intestinal levels of C. albicans might influence the host immune response, specifically in response to fungal challenge. We analyzed the transcriptional levels of 546 immune genes and the concentration of 13 cytokines after whole blood stimulation with C. albicans cells and showed positive associations between the extent of C. albicans intestinal levels and NLRP3 expression, as well as secreted IL-2 and CXCL5 concentrations. Taken together, these findings open the way for potential new interventional strategies to curb C. albicans intestinal overgrowth.


Candida albicans , Gastrointestinal Microbiome , Humans , Candida albicans/physiology , Genome-Wide Association Study , Gastrointestinal Microbiome/physiology , Diet , Immunity
...