13 Intriguingly, we found that treatment of BL cells

13 Intriguingly, we found that treatment of BL cells Idasanutlin with proteasome inhibitors partially restores their capacity to present the EBNA1 epitope, thereby suggesting that proteasomes from BL cells, although less active against prototype substrate peptides, which only partially indicate the in vivo proteasomal activities, degrade the HPV epitope during the processing of EBNA1. It

remains to be elucidated whether other EBNA1-derived CTL epitopes may be more efficiently generated and presented after partial inhibition of proteasomes or whether this effect is restricted to the HPV epitope. In conclusion, our study, together with previous reports, strongly supports the idea LY2109761 purchase that EBNA1-specific CTLs might be exploited therapeutically to target EBV-positive malignancies in combination with chemotherapy and protocols designed to restore antigen-presenting capacity in the tumour. In this context, it has been recently demonstrated that tubacin, a molecule that inhibits histone deacetylase 6, demonstrates a fairly selective capacity

to induce apoptosis in BL cells, but not in LCLs.37 Furthermore, the combination of tubacin with a proteasome inhibitor induced efficient killing of BL cells,37 which are known to be resistant to proteasome inhibitor-induced apoptosis.21,38 These findings, together with those reported in this study, suggest that the use of proteasome inhibitors, alone or in combination with other drugs such as tubacin, may represent a strategy Branched chain aminotransferase for the treatment of EBNA1-carrying

tumours, because proteasome inhibitors, in addition to their effect as pro-apoptotic drugs, may also increase the immunogenicity of EBNA1, thereby resulting in the efficient elimination of EBNA1-positive malignancies. This work was supported by grants from the University of Ferrara and Fondazione Cassa di Risparmio di Ferrara. We are grateful to A. Forster for editorial assistance and to Dr A. Balboni for HLA typing. The authors have no financial conflicts of interest. Table S1. MHC class I expression in lymphoblastoid cell line and in Burkitt’s lymphoma cells. “
“EAE, an animal model for multiple sclerosis, is a Th17- and Th1-cell-mediated auto-immune disease, but the mechanisms leading to priming of encephalitogenicTcells in autoimmune neuroinflammation are poorly understood. To investigate the role of dendritic cells (DCs) in the initiation of autoimmuneTh17- andTh1-cell responses andEAE, we used mice transgenic for a simian diphtheria toxin receptor (DTR) expressed under the control of the murineCD11c promoter (CD11c-DTRmice onC57BL/6 background).EAEwas induced by immunization with myelin oligodendrocyte glycoprotein (MOG) protein in CFA.

We previously identified

the adapter protein HS1 as a put

We previously identified

the adapter protein HS1 as a putative Nck-interacting protein. We now demonstrate that the SH2 domain of Nck specifically interacts with HS1 upon phosphorylation of its tyrosine residue 378. We report that in human T cells, ligation of the chemokine receptor CXCR4 by stromal cell-derived factor 1α (SDF1α) induces a rapid and transient phosphorylation drug discovery of tyrosine 378 of HS1 resulting in an increased association with Nck. Consequently, siRNA-mediated downregulation of HS1 and/or Nck impairs SDF1α-induced actin polymerization and T-cell migration. “
“The neonatal Fc receptor (FcRn) was first described as a receptor-mediating transplacental immunoglobulin (Ig)G transfer from mother to fetus, but it has other significant biological functions. It plays a key role in IgG and albumin homeostasis by efficient protection from catabolism [1]. It binds endocytosed IgG at acidic pH (< 6·5) within endosomes, FK506 mouse diverts it from degradation in lysosomes and instead transports the IgG–FcRn complexes back to the cell surface where, at neutral pH (> 7·0), IgG is released [1]. This process is highly efficient; FcRn recycles an equivalent amount of albumin and even four times as much IgG as can be produced

in a given time [2, 3]. Another notable function of FcRn is antigen delivery. FcRn was shown to be involved in the transcytosis of monomeric serum IgG from the basolateral to the apical side of the epithelium; immune complexes formed in the lumen are consequently delivered by FcRn to the lamina propria for antigen processing and triggering immune responses oxyclozanide [4]. Therefore, FcRn in the epithelium is probably able to sense luminal and epithelial infections and transmit evidence of these infections to the local and systemic immune apparatus. In the regulation of FCRN expression, polymorphism in the promoter region of the human gene consisting of a variable number of 37-base

pairs (bp) tandem repeats (VNTR) plays an important role. The allele with two tandem repeats (VNTR2) is associated with decreased promoter activity compared with the most common VNTR3 allele. VNTR2 carriers have been shown to have lower FCRN mRNA levels and decreased binding capacity of monocytes to immobilized IgG than VNTR3/3 homozygotes that predominate in general population [5]. We sought to determine whether FCRN expression influences intravenous immunoglobulin (IVIg) catabolism and clinical phenotype in patients with common variable immunodeficiency (CVID). This effect may be due not only to the role of FcRn in IgG protection from degradation, but also by influencing mucosal antigen presentation.

To increase our understanding of the mechanisms that play a role

To increase our understanding of the mechanisms that play a role in host immune responses, we investigated the effects of C. parvum antigens on the phenotype of mouse and human dendritic cells (DCs). Cryptosporidium parvum antigens induced DC activation as indicated by upregulation of the maturation marker CD209, as well as by the production of the cytokines interleukin-12 p70,

IL-2, IL-1beta, IL-6. In particular, significant increases in the expression of IL-12 p70 were observed from mouse DCs derived from bone marrow in response to solubilized sporozoite antigen and the recombinant cryptosporidial antigens, Cp40 and Cp23. We observed a small but Z-IETD-FMK price significant increase in IL-18 expression following the exposure to Cp40. We found that the induction of Th1 cytokines was MyD88 dependent (MyD88 knockout mouse DCs were unresponsive). Additionally, both sporozoite preparations (solubilized and live) significantly

induced IL-12 production by human monocytic dendritic cells (MoDCs). This finding indicates that solubilized as well as recombinant antigens can induce the maturation of DCs and subsequently initiate an innate immune response. Cryptosporidium CDK inhibitor parvum (C. parvum) is a zoonotic intracellular opportunistic protozoan parasite with a worldwide distribution. Infection is usually transmitted from one host to another through faecal contamination of drinking water or food or by contact with infected hosts (1). Following ingestion, C. parvum infection develops in the intestinal tract of the host, followed by symptoms of diarrhoea, low-grade fever, nausea and weight loss (2). In immunocompetent individuals, the disease is typically self-limiting. However, in individuals who are immunocompromised, such as adult patients infected oxyclozanide with HIV as well as HIV-positive children, diarrhoeal disease can be persistent and life-threatening. Chronic disease

in immunodeficient hosts is exacerbated because of the lack of effective treatment options (3). To date, no effective treatment regimen nor preventive intervention has been developed for immunocompromised individuals, partly due to the incomplete understanding of the host immune response to the parasite infection (4). Studies pertaining to host cell–mediated immune responses indicate the importance of T lymphocytes, specifically CD4+ T cells during recovery from cryptosporidial infections (5). The cytokine IFN-γ also plays an important role in adaptive as well as in innate immune responses to C. parvum infection in mice (6). Secretion of pro-inflammatory cytokines such as IL-12 p70 is a key in generating IFN-γ and can be induced through the activation of antigen-presenting cells (APCs) by various pathogens and their products. One type of antigen-presenting cell, dendritic cells (DCs), plays an important role in eliciting an immune response and is also the first line of defence against pathogens by activating an innate immune response.

In addition, systemic cytokine/chemokine responses can be identif

In addition, systemic cytokine/chemokine responses can be identified in patients with periodontitis [3–5]. Interleukin (IL)-1β, tumour necrosis factor (TNF)-α and IL-6 are principal pro-inflammatory cytokines with pleotropic biological activities on immune and non-immune cells, as well as in osteogenic pathways). IL-8 (CXCL8) is the major neutrophil chemokine, while macrophage chemotactic protein (MCP)-1 (CCL2), a major chemoattractant and maturation signal for macrophages, and regulated upon activation, normal T cell expressed and secreted (RANTES; CCL5) is a member of the IL-8 superfamily of cytokines.

It is a selective attractant for memory T lymphocytes and monocytes. These chemokines have all been detected in the serum of patients with microbial infections [6–10], including periodontitis [11–14]. Selleck Carfilzomib However, chronic stimulation of these biomolecules generally represents dysregulated responses, and is associated frequently with systemic disease sequelae [15–21]. In some cases, particularly with polymicrobial infections at mucosal surfaces, innate immune mechanisms may function exceptionally well to manage surface colonization by commensal opportunistic pathogens and maintain homeostasis [22–25]. Nevertheless, with respect to a number of chronic inflammatory diseases, the interaction between Pembrolizumab the challenge (e.g. bacteria) and

the inflammatory and innate immune response can result in collateral damage of the local tissues. Adverse pregnancy outcomes provide a potential example of these ramifications of a dysregulated

host response. Ascending vaginal infections trigger the local production of various inflammatory mediators and matrix metalloproteinases (MMP), resulting in amnionitis that impact placental functions negatively and lead potentially to fetal infection [26–32]. Reports described relationships between the presence of inflammatory mediators in amniotic fluid and uterine contractions and/or birth in humans and non-human primates. Proinflammatory cytokines/chemokines, immunomodulatory and immunosuppressive Org 27569 cytokines and prostanoids [e.g. prostaglandin E2 (PGE2)] are produced by the amniotic and decidual membranes and can be found in fetal circulation and amniotic fluid, often associated with premature delivery. Expanding literature supports that the levels of many of these cytokines/chemokines in serum are also reflective of, and potentially contribute to, the risk for premature rupture of membranes (PROM) with preterm labour and delivery [26,32–35]. Consequently, relationships between serum and local cytokine levels and their association with adverse pregnancy outcomes are possible. Periodontitis is a chronic oral infection with polymicrobial biofilms triggering a localized immunoinflammatory lesion.

After 7 days of culture, little difference was observed in CFSE p

After 7 days of culture, little difference was observed in CFSE profiles and per cent divided cells in SC-58125-treated B-cell cultures (Fig. 2b). Similar results were observed for B cells treated with NS-398, a different Cox-2 selective inhibitor (data not shown). The percentages of divided B cells following treatment with SC-58125 were averaged from three different donors (Fig. 2c). No significant change in the per cent divided B cells following

Cox-2 inhibitor treatment was detected, indicating that a decrease in proliferation does not account for the attenuation of antibody production. We next investigated whether attenuated antibody production was caused by a reduction in the selleck chemicals differentiation of human B cells to antibody-secreting cells. Human plasma cell precursors, defined by multiple investigators as CD38+ antibody-secreting cells,17–19 can be generated in vitro. On day 7 of culture, B cells were stained for surface expression of CD38 and CD19, as well as for intracellular IgM or IgG. Intracellular antibody gates were determined based upon unpermeabilized stained controls. Multiple blood donors were assessed via this method with similar results. Freshly isolated B cells express a relatively low frequency of CD38+ antibody-secreting cells, which is significantly selleckchem increased following 7 days of stimulation with CpG plus anti-IgM (Fig. 3a). A significant reduction in the

frequency of CD38+ antibody-secreting cells was observed following treatment with SC-58125 (Fig. 3a,c). In contrast there was no change in the frequency of CD38− Ig+-secreting cells (Fig. 3b). Generation of IgM-secreting, CD38+ B cells was significantly attenuated in a dose-dependent manner (Fig. 3c). These results mirrored the decrease in antibody production measured by ELISA (Fig. 1). Similarly, CD38+ IgG-secreting cells were also significantly decreased following treatment with the Cox-2 inhibitor (Fig. 3c). These new data demonstrate that the Cox-2 selective inhibitor, SC-58125 attenuated the ability of B cells to differentiate to CD38+ antibody-secreting plasma cell precursors. Cox-2 knockout

mice were next used to study the vital role of Cox-2 in B-cell differentiation to plasma cells. CD19+ B cells were cAMP isolated from the spleens of wild-type and Cox-2-deficient mice. Analysis of wild-type and Cox-2-deficient splenocytes revealed no significant differences in overall CD19+ cells or marginal zone B cells (CD19+ CD21+ CD23−), indicating that B-cell populations are similar. Following a 72-hr stimulation with LPS, Cox-2-deficient mice had a 60% reduction in the number of CD138+ plasma cells compared with wild-type controls (Fig. 4a,b). This indicates impairment in the differentiation of B cells to plasma cells in mice lacking Cox-2. Next, we tested whether expression of the essential plasma cell transcriptional regulator, Blimp-1, was regulated by Cox-2.

SD and VLG performed the experiments and drafted the manuscript,

SD and VLG performed the experiments and drafted the manuscript, NDS provided clinical samples, VLG and JG designed the study; all authors reviewed and approved the final manuscript. SD was supported by a University of Hull studentship. We would like to thank Mr Jose and other members of the head and neck surgical team in Hull for obtaining the patients’ consent and for collection of peripheral blood samples. The authors declare

BMN 673 ic50 no financial or commercial conflict of interest. “
“Toll-like receptor 4 (TLR4), a key member of the TLR family, has been well characterized by its function in the induction of inflammatory products of innate immunity. However, the involvement of TLR4 in a variety of apoptotic events by an unknown mechanism has been the focus of great interest. Our investigation found that TLR4 promoted apoptotic signalling by affecting the glycogen synthase kinase-3β (GSK-3β) pathway in a serum-deprivation-induced apoptotic paradigm. Serum deprivation induces GSK-3β activation in a pathway that leads to subsequent cell apoptosis. Intriguingly, this apoptotic cascade is amplified in presence of TLR4 but greatly attenuated by β-arrestin 2, another

critical molecule implicated in TLR4-mediated immune responses. Our data suggest that the association of β-arrestin 2 with GSK-3β contributes

Selleck Trametinib to the stabilization of phospho-GSK-3β, an inactive form of GSK-3β. It becomes a critical determinant for the attenuation of TLR4-initiated apoptosis by β-arrestin 2. Taken together, we demonstrate that the TLR4 possesses the capability of accelerating GSK-3β activation thereby deteriorating serum-deprivation-induced apoptosis; β-arrestin 2 represents an inhibitory effect on the TLR4-mediated apoptotic cascade, through controlling the homeostasis of activation and inactivation of GSK-3β. Toll-like receptor AMP deaminase 4 (TLR4), an extensively investigated member of the TLR family, represents the first line of defence against invading pathogens in the innate immune system.1 For conventional TLR4 signalling, it specifically recognizes lipopolysaccharide (LPS) from the outer membrane of Gram-negative bacteria and activates two major signalling pathways, nuclear factor-κB (NF-κB) pathway and mitogen-activated protein kinase pathway, both of which control the expression of key immunoregulatory genes.1 In addition to the widely accepted inflammatory response induced by exogenous infection, activation of TLR4 occurs as a result of non-infectious insults such as hypoxia, ischaemia,2,3 concomitantly with cell damage and apoptosis.

Following microscopic inspection, the 134 cases were assigned to

Following microscopic inspection, the 134 cases were assigned to one of the four pathological phenotypes according to the varying forms and distribution of Aβ deposition (as SP and/or CAA) within frontal, temporal and occipital lobes, and coded accordingly Fulvestrant (see methods for criteria) (Figure 1). However, there was often heterogeneity in phenotypic presentation across the three regions in individual cases. In some cases, all three regions showed

a similar histological phenotype, whereas in others there were regional variations with the frontal and temporal cortex closely resembling each other histologically though being dissimilar to occipital cortex, nearly always with respect to the presence/distribution of CAA. Hence, 35 cases (coded 111) showed type 1 pathology within all three regions (that is, Aβ deposition predominantly as SP with or without CAA, BEZ235 and involving only superficial (leptomeningeal) blood vessels) (red in Figure 2). Sixty-eight cases (coded 112, 122, 212 or 222) showed type 2 pathology with Aβ deposition as SP and CAA in leptomeningeal and deeper intracortical vessels,

in the occipital lobe: dyshoric change was often evident surrounding affected vessels (green in Figure 2). Sometimes, similar changes were also seen in frontal but not temporal cortex (where type 1 change was present, and coded 212 or 122 respectively), or type 1 changes were only seen in both regions (and coded 112). Twenty cases showed type 3 pathology in all three regions (and coded 333) with robust CAA predominantly within capillaries in the occipital lobe, and leptomeningeal and/or intracortical CAA in frontal and/or temporal region (and coded 113, 123, 213, 223 or 323) (blue in Figure 2). In these cases, within occipital lobe SP were absent or relatively few, though were usually much more numerous in frontal and temporal lobes. Four cases (coded 214,

224 or 444) showed type 4 pathology with a predominant CAA phenotype, where Aβ was heavily deposited in the leptomeningeal and cortical vessels, but not capillaries, within occipital lobe (and sometimes also in frontal and temporal Anidulafungin (LY303366) lobes): dyshoric change was always evident surrounding the vessels. Aβ deposition, as SP, in occipital lobe was absent or infrequent (orange in Figure 2). For group comparisons, cases were pooled according to the type of histological presentation within the occipital lobe, irrespective of whether changes in frontal and temporal lobe always followed suit. Nonetheless, there were seven cases (coded 121, 211 or 221) which formed an ‘outlier’ group within type 2 pathology (purple in Figure 2). These were differentiated from the other cases with type 2 pathology by virtue of the fact that there was intracortical CAA in frontal and/or temporal cortex but, in contrast to the other cases in that group, these were without occipital involvement.

Some studies have reported that PI3K inhibition reduces Th2 cytok

Some studies have reported that PI3K inhibition reduces Th2 cytokine production, pulmonary eosinophilia, airway inflammation, and bronchial hyperresponsiveness in a mouse asthma model 48, 49. In addition, PI3K is shown to be involved in HIF-1α activation induced by oxygen-dependent or oxygen-independent pathways 50, 51. Recently, p110δ and p110γ isoforms have sparked a great deal of interest, as there is increasing evidence that EGFR inhibitor these isoforms play key roles in immunity 52. We have also demonstrated that OVA-induced HIF-1α

activation is significantly reduced by administration of a PI3K-δ inhibitor and suggested that inhibition of the p110δ signaling pathway has therapeutic potential for allergic airway inflammation 33. Consistent with these observations, in the present study, levels of p-Akt protein and PI3K activity in lung tissues were increased after OVA inhalation. The increased levels of p-Akt and PI3K Selleckchem Navitoclax activity were significantly reduced after administration of IC87114, a PI3K-δ inhibitor. Moreover, the increased levels of HIF-1α after OVA inhalation were significantly reduced by IC87114 in primary tracheal epithelial cells isolated from OVA-treated mice. These findings suggest

that PI3K-δ regulates HIF-1α activation therewith inducing VEGF expression in a murine model of allergic airway disease. In summary, we have examined the roles of HIF-1α in allergen-induced airway inflammation and bronchial hyperresponsiveness using an HIF-1α inhibitor,

2ME2, and siRNA targeting HIF-1α and evaluated the role of PI3K-δ signaling in HIF-1α activation in allergic airway disease. The administration of 2ME2 was effective in reversing all pathophysiological symptoms examined. Our data have also revealed that HIF-1α inhibition substantially reduces the increase in VEGF expression as well as the activity of VEGF in lungs, especially in bronchial epithelial cells, of our murine model of allergic next airway disease. In addition, administration of IC87114 reduced the increase in HIF-1α activity in OVA-treated airway epithelial cells. Therefore, one likely mechanism for the roles of HIF-1α in pathobiology of allergen-induced airway inflammation and hyperresponsiveness is induction of vascular leakage via up-regulation of VEGF expression in lungs as well as bronchial epithelium. Thus, these findings provide a crucial molecular mechanism for the potential of HIF-1α inhibition in preventing and/or treating asthma and other airway inflammatory disorders. Female C57BL/6 mice, aged 8–10 wk and free of murine specific pathogens, were obtained from the Orientbio (Seoungnam, Korea) were housed throughout the experiments in a laminar flow cabinet and were maintained on standard laboratory chow ad libitum.

Indeed, IFN-β upregulated

T-bet expression to comparable

Indeed, IFN-β upregulated

T-bet expression to comparable levels as IL-12 by 48 h post-activation, indicating that type-I IFN signaling on activated CD8+ T cells directly regulates T-bet expression. Thus, under priming conditions with abundant type-I IFN levels, the initial differentiation of CD8+ T cells toward an SLEC phenotype is driven by T-bet that is directly induced by type-I IFN signaling. GDC-0449 research buy Finally, we addressed the ability WT and IFNAR−/− P14 cells to give rise to functional memory CD8+ T cells with recall potential in the context of LCMV8.7 and VVG2 co-infection. Analysis of the tissue distribution of memory WT and IFNAR−/− P14 cells at day 45 post-infection revealed that both WT and IFNAR−/− P14 cells could be found in the

spleen and lymph nodes but only WT P14 cells could be found in liver (Fig. 6A), as opposed to an equal tissue distribution of IFNAR−/− P14 cells seen in the spleen and liver on day 6 post-infection (data not shown and 19). To evaluate the quality of the generated memory cells, their ability to produce IFN-γ and their capacity to degranulate upon in vitro antigen recognition was determined. At day 45 post-priming, WT and IFNAR−/− memory P14 cells produced comparable levels of IFN-γ and WT P14 cells showed only slightly increased levels of CD107a compared with IFNAR−/− memory P14 cells (Fig. 6B). Thus, although the frequency of the IFNAR−/− memory P14 cells was strongly reduced, their per-cell functional properties did not differ from WT P14 cells. Selleckchem INK 128 In addition to equivalent ex vivo functional capacity, the proportion of P14 cells exhibiting a CD127high KLRG1low phenotype at day 60 post-infection was comparable between WT and IFNAR−/− P14 cells (Fig. 6C). To ascertain that the memory IFNAR−/− P14 cell population represented indeed memory cells and not naïve cells which had not

however been recruited into the primary response, we measured CD44 expression on the IFNAR−/− P14 cells. As all IFNAR−/− P14 cells uniformly expressed high levels of CD44, we conclude that these cells are indeed antigen-experienced memory cells (data not shown). To further validate the functionality of IFNAR−/− memory P14 cells, we determined their potential to re-expand and to produce effector cytokines upon viral re-challenge. We chose a challenge with VVG2 as it has been shown that CD8+ T-cell expansion is only marginally dependent on direct type-I IFN signaling during VVG2 infection 10, 17. Thus, memory WT and IFNAR−/− P14 cells were isolated from the spleen 45 days post-LCMV8.7 and VVG2 infection and transferred into naïve WT mice, which were subsequently challenged with VVG2. The fold expansion of both subsets 6 days post-challenge was calculated according to the frequency of cells before and after challenge.

Hoffmann et al investigated the association of diet with fungal

Hoffmann et al. investigated the association of diet with fungal populations, using fecal samples from 98 healthy individuals [158]. They characterized 62 fungal genera and 184 species by deep sequencing, and usually found that the presence of either the phyla Ascomycota or Basiodiomycota was mutually exclusive. The authors could not conclude which of those fungi are true gut residents selleck compound and which are passengers resulting from diet. We cannot exclude the possibility that the presence of Saccharomyces is due to the ingestion of yeast-containing foods such as bread and beer [82].

A recent study conducted on the Wayampi Amerindian community showed a high diversity among yeast species in the gut, with a prevalence of S. cerevisiae over Candida species [80], suggesting a role for

this fungus in gut immune homeostasis. Thus, integrating information on the repertoire of the gut mycobiota in the context of the broader microbiota and developing functional tests to measure its role in shaping immune function is necessary to better understand the role of the microbial communities in sustaining human health. Although we have described BIBW2992 research buy the composition of the fungal microbiota in various locations in the human body, we remain aware that these locations are not isolated and that DCs trained in the Peyer’s patches of the intestine (Fig. 1) can shape T-cell responses in other locations. A clear example of this crosstalk was recently shown in an elegant study by Kim et al. [159], who showed that antibiotic treatment of mice increases susceptibility to allergic airway disease by promoting varying degrees of fungal outgrowth in the intestine, ultimately resulting in the acquisition of an M2 phenotype by alveolar macrophages [159]. The authors isolated C. parapsilosis

from the feces of antibiotic-treated mice and showed that transferring this fungus to mice that did not carry this species increased their susceptibility to allergic airway inflammation induced by papain or house dust mite extract [159]. Oral treatment of mice with Candida species isolated from humans also led to fungal outgrowth in the gut and exacerbated allergic airway inflammation, increasing serum levels Mirabegron of prostaglandin E2, which promoted the development of M2 macrophages [159]. The mycobiota alteration mediated imbalance in alveolar macrophage function contributed to the increase in airway inflammation, as untreated animals receiving alveolar macrophages from antibiotic-treated mice developed more severe airway inflammation than animals that received alveolar macrophages from control mice. Based on this result, it appears that intestinal dysbiosis, particularly the altered ratio of fungi to bacteria, could be a causative factor in the development of allergic disease. Patients with severe asthma with fungal sensitization are often sensitized to C. albicans and benefit from antifungal drug therapy [160]. Colonization of mice with C.