92 research outputs found

    New approaches in the development of a vaccine for mucosal candidiasis: progress and challenges

    Get PDF
    The commensal fungus Candida albicans causes mucosal candidiasis in the rapidly expanding number of immunocompromised patients. Mucosal candidiasis includes oropharyngeal, esophageal, gastrointestinal, and vaginal infections. Vulvovaginal candidiasis (VVC) and antimycotic-refractory recurrent VVC is a frequent problem in healthy childbearing women. Both these mucosal infections can affect the quality of life and finding new therapeutical and preventive approaches is a challenge. A vaccine against candidal infections would be a new important tool to prevent and/or cure mucosal candidiasis and would be of benefit to many patients. Several Candida antigens have been proposed as vaccine candidates including cell wall components and virulence factors. Here we discuss the recent progress and problems associated with vaccination against mucosal candidiasis

    Hyphal morphology, molecular genetics and phylogenetic relationships among commensal and pathogenic vulvovaginal isolates of Candida albicans

    Get PDF
    Vaginal candidiasis is a common disorder in women of childbearing age, caused primarily by Candida albicans. Since C. albicans is a commensal fungus of the vaginal mucosa, a long-standing question is how the fungus switches from being a harmless commensal to a virulent pathogen. Clinical studies and murine vaginitis models suggest that host inflammatory processes drive the onset of symptomatic infection. In previous work with fresh clinical samples, we found that the pro-inflammatory cell wall polysaccharide β-glucan is largely masked from immune recognition during vulvovaginal infection. Enhanced β-glucan availability was only found in hyphae from symptomatic patients with strong neutrophil infiltration. There was high variability in levels of β-glucan exposure and hyphal morphology among colonizing and infection-associated isolates, and we reasoned that this could be explained by fungal-intrinsic factors and/or host-associated traits. We assayed several aspects of C. albicans isolated from symptomatic and asymptomatic individuals to determine any associations between fungal-intrinsic traits and virulence: MLST analysis, sequencing of the gene encoding the candidalysin toxin, and propensity to form hyphal cells. Preliminary results suggest that none of these indicators correlates with isolates causing symptomatic infection, indicating that host-intrinsic mechanisms may play the most important role in the occurrence of symptomatic infections

    Induction of caspase-11 by aspartyl proteinases of Candida albicans and implication in promoting inflammatory response

    Get PDF
    We recently demonstrated that the secreted aspartyl proteinases (Saps), Sap2 and Sap6, of Candida albicans have the potential to induce the canonical activation of NLRP3-inflammasome leading to the secretion of IL-1β and IL-18 via caspase-1 activation. We also observed that the activation of caspase-1 is partially independent from the NLRP3 activation pathway. In this study, we examined whether Sap2 and Sap6 are also able to activate the noncanonical inflammasome pathway in murine macrophages. Our data show that both, Sap2 and Sap6, can activate caspase-11 through type I IFN production. Caspase-11 concurs to activate caspase-1 with subsequent increase of IL-1β secretion. Endocytosis and internalization of Saps are required for the induction of type I IFN production, that is essential for induction of noncanonical inflammasome activation. Our study indicates a sophisticated interplay between caspase-1 and caspase-11 that connects canonical and noncanonical pathways of inflammasome activation in response to C. albicans Saps

    In vivo induction of neutrophils chemotaxis by secretory aspartyl proteinases of Candida albicans

    Get PDF
    Secretory aspartyl proteinases (Saps) of Candida albicans are key virulence traits which cause inflammasome-dependent, aseptic inflammation in a mouse model of vaginitis. In this paper, neutrophil migration in response to Sap2, Sap6 and chemo-attractive products released from Sap-treated vaginal epithelium was measured in vitro, ex vivo and in vivo. Our results show that Sap2 and Sap6 induce neutrophil migration and production of potent chemoattractive chemokines such as IL-8 and MIP-2 by vaginal epithelial cells. Our data suggest that at least part of MIP-2 production depends upon IL-1β activity. The vaginal fluid of Candida-infected mice contained a heat-labile inhibitor of neutrophil candidacidal activity that was absent from the vaginal fluid of Sap-treated mice. Overall, our data provide additional information on the capacity of C. albicans Saps to cause aseptic vaginal inflammation and highlight the potential role of some chemokines released from vaginal epithelial cells in this phenomenon

    Lactobacillus (L.) acidophilus, L. plantarum, L. rhamnosus and L. reuteri Cell-Free-Supernatants inhibit Candida parapsilosis pathogenic potential upon infection of vaginal epithelial cells monolayer and in a transwell co-culture system in vitro.

    Get PDF
    Vulvovaginal candidiasis (VVC) is a common clinical condition with symptoms and signs of vaginal inflammation in the presence of Candida species. At least one episode of VVC is experienced in up to 75% of women in the reproductive age group during their lifetime, and 5-8% of such women suffer from the chronic form. Most cases of VVC are still caused by C. albicans; however, the incidence of VVC cases by non-albicans (NAC) species, such as C. parapsilosisis, is continuously increasing. Despite the prevalence of VVC from NAC, to date little is known on these species, and almost nothing on the mechanisms that trigger the VVC. Lactobacillus spp. are the most represented microorganisms in the vaginal microbiota of healthy women. Here, cell-free supernatants (CFS) obtained from L. acidophilus, L. plantarum, L. rhamnosus, L. reuteri were assessed for their effect on C. parapsilosis virulence traits. Moreover, we assessed if such effect persists even after removal of the CFS (CFS-preincubation effect). Moreover, a transwell co-culture system was employed, by which the relevant antifungal effect was shown to be attributable to the compounds released by Lactobacilli. Our results suggests that Lactobacilli can work: a) by reducing C. parapsilosis virulence traits, as indicated by the reduced fungal proliferation, viability and metabolic activity and b) by improving epithelial resistance to the fungus. Overall, these data suggest that, in the context of vaginal microbiota, the Lactobacilli may play a role in preventing the onset of mucosal C. parapsilosis infection

    Bioimmunological activities of Candida glabrata cellular mannan

    Get PDF
    Candida glabrata is a second most common human opportunistic pathogen which causes superficial but also life-threatening systemic candidiasis. According to the localization of mannans and mannoproteins in the outermost layer of the cell wall, mannan detection could be one of the first steps in the cell recognition of Candida cells by the host innate immune system. Mannans from the cell wall provide important immunomodulatory activities, compromising stimulation of cytokine production, induction of dendritic cells maturation and T-cell immunity. The model of DCs represents a promising tool to study immunomodulatory interventions throughout the vaccine development. Activated DCs induce, activate and polarize T-cell responses by expression of distinct maturation markers and cytokines regulating the adaptive immune responses. In addition, they are uniquely adept at decoding the fungus-associated information and translate it in qualitatively different T helper responses. We find out, that C. glabrata mannan is able to induce proliferation of splenocytes and to increase the production of TNF-α and IL-4. Next, increased the expression of co-stimulatory molecules CD80 and CD86 and the proportion of CD4+CD25+ and CD4+CD28+ T cells during in vitro stimulation of splenocytes

    Effects of benzydamine and mouthwashes containing benzydamine on Candida albicans adhesion, biofilm formation, regrowth, and persistence

    Get PDF
    Objectives To assess the effects of benzydamine and mouthwashes (MoWs) containing benzydamine on different stages of Candida albicans biofilm: adhesion, formation, persistence, and regrowth (if perturbed). Materialsandmethods C.albicansCA1398,carryingthebioluminescenceACT1p-gLUC59fusionproduct,wasemployed. Fungal cells were exposed for 1\u2032, 5\u2032, or 15\u2032 to 4 different benzydamine concentrations (0.075 to 0.6%) to 2 mouthwashes (MoWs) containing benzydamine and to a placebo MoW (without benzydamine). Treated cells were tested for adhesion (90 min) and biofilm formation (24-h assay). Next, 24- and 48-h-old biofilms were exposed to benzydamine and MoWs to assess regrowth and persistence, respectively. The effects of benzydamine, MoWs containing benzydamine, and placebo on different biofilm stages were quantified by bioluminescence assay and by the production of quorum sensing (QS) molecules. Results Benzydamine and MoWs containing benzydamine impaired C. albicans ability to adhere and form biofilm, counter- acted C. albicans persistence and regrowth, and impaired a 48-h-old biofilm. Some of these effects paralleled with alterations in QS molecule secretion. Conclusions Our results show for the first time that benzydamine and MoWs containing benzydamine impair C. albicans capacity to form biofilm and counteract biofilm persistence and regrowth. Clinical relevance Benzydamine and MoWs containing benzydamine capacity to affect C. albicans biofilm provides an interesting tool to prevent and treat oral candidiasis. Likely, restraining C. albicans colonization through daily oral hygiene may counteract colonization and persistence by other critical oral pathogens, such as Streptococcus mutans, whose increased virulence has been linked to the presence of C. albicans biofilm

    Pomegranate extract affects fungal biofilm production: consumption of phenolic compounds and alteration of fungal autoinducers release

    Get PDF
    Candida albicans expresses numerous virulence factors that contribute to pathogenesis, including its dimorphic transition and even biofilm formation, through the release of specific quorum sensing molecules, such as the autoinducers (AI) tyrosol and farnesol. In particular, once organized as biofilm, Candida cells can elude conventional antifungal therapies and the host’s immune defenses as well. Accordingly, biofilm-associated infections become a major clinical challenge underlining the need of innovative antimicrobial approaches. The aim of this in vitro study was to assess the effects of pomegranate peel extract (PomeGr) on C. albicans growth and biofilm formation; in addition, the release of tyrosol and farnesol was investigated. The phenolic profile of PomeGr was assessed by high-performance liquid chromatography coupled to electrospray ionization mass spectrometry (HPLC-ESI-MS) analysis before and after exposure to C. albicans. Here, we showed that fungal growth, biofilm formation and AI release were altered by PomeGr treatment. Moreover, the phenolic content of PomeGr was substantially hampered upon exposure to fungal cells; particularly pedunculagin, punicalin, punicalagin, granatin, di-(HHDP-galloyl-hexoside)-pentoside and their isomers as well as ellagic acid–hexoside appeared highly consumed, suggesting their role as bioactive molecules against Candida. Overall, these new insights on the anti-Candida properties of PomeGr and its potential mechanisms of action may represent a relevant step in the design of novel therapeutic approaches against fungal infections

    Saccharomyces cerevisiae CNCM I-3856 as a New Therapeutic Agent Against Oropharyngeal Candidiasis

    Get PDF
    Oropharyngeal candidiasis is a common opportunistic mucosal infection of the oral cavity, mainly caused by an overgrowth of Candida albicans. This infection can inhibit nutritional intakes and strongly affect quality of life. To date, standard therapeutic strategies involving the administration of antifungal drugs can bring several side effects, not least the emergence of drug-resistant strains. The purpose of this study is to investigate the effectiveness of Saccharomyces cerevisiae CNCM I-3856 (live or inactivated cells) against oropharyngeal candidiasis. Our results show that administration of S. cerevisiae CNCM I-3856 (live or inactivated cells) in the oral cavity of C57BL/6J mice resulted in a protective effect against oropharyngeal candidiasis. The strongest effect was obtained with live S. cerevisiae CNCM I-3856. This was related to: (1) a decrease in C. albicans load in the oral cavity, esophagus, stomach, and duodenum; (2) an early resolution of inflammatory process in the tongue; (3) a marked reduction in C. albicans virulence factors; and (4) a consistent increase in neutrophil antimicrobial capacity. These findings suggest that S. cerevisiae products are potentially beneficial in the treatment of oropharyngeal candidiasis
    • …
    corecore