Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 20(5): 546-558, 2019 05.
Article in English | MEDLINE | ID: mdl-30911105

ABSTRACT

Neutrophils are essential first-line defense cells against invading pathogens, yet when inappropriately activated, their strong immune response can cause collateral tissue damage and contributes to immunological diseases. However, whether neutrophils can intrinsically titrate their immune response remains unknown. Here we conditionally deleted the Spi1 gene, which encodes the myeloid transcription factor PU.1, from neutrophils of mice undergoing fungal infection and then performed comprehensive epigenomic profiling. We found that as well as providing the transcriptional prerequisite for eradicating pathogens, the predominant function of PU.1 was to restrain the neutrophil defense by broadly inhibiting the accessibility of enhancers via the recruitment of histone deacetylase 1. Such epigenetic modifications impeded the immunostimulatory AP-1 transcription factor JUNB from entering chromatin and activating its targets. Thus, neutrophils rely on a PU.1-installed inhibitor program to safeguard their epigenome from undergoing uncontrolled activation, protecting the host against an exorbitant innate immune response.


Subject(s)
Epigenesis, Genetic/immunology , Epigenomics/methods , Neutrophils/immunology , Proto-Oncogene Proteins/immunology , Trans-Activators/immunology , Animals , Candida albicans/immunology , Candida albicans/physiology , Candidiasis/genetics , Candidiasis/immunology , Candidiasis/microbiology , Disease Resistance/genetics , Disease Resistance/immunology , Gene Expression Profiling/methods , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/metabolism , Neutrophils/microbiology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Survival Analysis , Trans-Activators/deficiency , Trans-Activators/genetics , Transcriptome/genetics , Transcriptome/immunology
2.
J Immunol ; 211(5): 804-815, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37436030

ABSTRACT

Because of the growing numbers of immunocompromised patients, the incidence of life-threatening fungal infections caused by Candida albicans and Aspergillus fumigatus is increasing. We have recently identified enolase 1 (Eno1) from A. fumigatus as an immune evasion protein. Eno1 is a fungal moonlighting protein that mediates adhesion and invasion of human cells and also immune evasion through complement inactivation. We now show that soluble Eno1 has immunostimulatory activity. We observed that Eno1 from both C. albicans and A. fumigatus directly binds to the surface of lymphocytes, preferentially human and mouse B cells. Functionally, Eno1 upregulated CD86 expression on B cells and induced proliferation. Although the receptor for fungal Eno1 on B lymphocytes is still unknown, the comparison of B cells from wild-type and MyD88-deficient mice showed that B cell activation by Eno1 required MyD88 signaling. With respect to infection biology, we noted that mouse B cells stimulated by Eno1 secreted IgM and IgG2b. These Igs bound C. albicans hyphae in vitro, suggesting that Eno1-induced Ab secretion might contribute to protection from invasive fungal disease in vivo. Eno1 also triggered the release of proinflammatory cytokines from monocytes, particularly IL-6, which is a potent activator of B cells. Together, our data shed new light on the role of secreted Eno1 in infections with C. albicans and A. fumigatus. Eno1 secretion by these pathogenic microbes appears to be a double-edged sword by supporting fungal pathogenicity while triggering (antifungal) immunity.


Subject(s)
Aspergillus fumigatus , Candida albicans , Phosphopyruvate Hydratase , Animals , Humans , Mice , Aspergillus fumigatus/enzymology , Aspergillus fumigatus/metabolism , Candida albicans/enzymology , Candida albicans/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Monocytes/metabolism , Monocytes/microbiology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phosphopyruvate Hydratase/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/microbiology
3.
PLoS Pathog ; 18(2): e1010283, 2022 02.
Article in English | MEDLINE | ID: mdl-35108336

ABSTRACT

Protein kinases play central roles in virtually all signaling pathways that enable organisms to adapt to their environment. Microbial pathogens must cope with severely restricted iron availability in mammalian hosts to invade and establish themselves within infected tissues. To uncover protein kinase signaling pathways that are involved in the adaptation of the pathogenic yeast Candida albicans to iron limitation, we generated a comprehensive protein kinase deletion mutant library of a wild-type strain. Screening of this library revealed that the protein kinase Ire1, which has a conserved role in the response of eukaryotic cells to endoplasmic reticulum stress, is essential for growth of C. albicans under iron-limiting conditions. Ire1 was not necessary for the activity of the transcription factor Sef1, which regulates the response of the fungus to iron limitation, and Sef1 target genes that are induced by iron depletion were normally upregulated in ire1Δ mutants. Instead, Ire1 was required for proper localization of the high-affinity iron permease Ftr1 to the cell membrane. Intriguingly, iron limitation did not cause increased endoplasmic reticulum stress, and the transcription factor Hac1, which is activated by Ire1-mediated removal of the non-canonical intron in the HAC1 mRNA, was dispensable for Ftr1 localization to the cell membrane and growth under iron-limiting conditions. Nevertheless, expression of a pre-spliced HAC1 copy in ire1Δ mutants restored Ftr1 localization and rescued the growth defects of the mutants. Both ire1Δ and hac1Δ mutants were avirulent in a mouse model of systemic candidiasis, indicating that an appropriate response to endoplasmic reticulum stress is important for the virulence of C. albicans. However, the specific requirement of Ire1 for the functionality of the high-affinity iron permease Ftr1, a well-established virulence factor, even in the absence of endoplasmic reticulum stress uncovers a novel Hac1-independent essential role of Ire1 in iron acquisition and virulence of C. albicans.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Candida albicans/metabolism , Candidiasis/microbiology , Iron/metabolism , Membrane Transport Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Candida albicans/genetics , Candida albicans/pathogenicity , DNA, Fungal , Endoplasmic Reticulum Stress , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Membrane Transport Proteins/genetics , Mice, Inbred BALB C , Protein Serine-Threonine Kinases/genetics , Sequence Deletion , Signal Transduction , Specific Pathogen-Free Organisms , Virulence
4.
Cell Microbiol ; 22(7): e13197, 2020 07.
Article in English | MEDLINE | ID: mdl-32083801

ABSTRACT

Biotin is an important cofactor for multiple enzymes in central metabolic processes. While many bacteria and most fungi are able to synthesise biotin de novo, Candida spp. are auxotrophic for this vitamin and thus require efficient uptake systems to facilitate biotin acquisition during infection. Here we show that Candida glabrata and Candida albicans use a largely conserved system for biotin uptake and regulation, consisting of the high-affinity biotin transporter Vht1 and the transcription factor Vhr1. Both species induce expression of biotin-metabolic genes upon in vitro biotin depletion and following phagocytosis by macrophages, indicating low biotin levels in the Candida-containing phagosome. In line with this, we observed reduced intracellular proliferation of both Candida cells pre-starved of biotin and deletion mutants lacking VHR1 or VHT1 genes. VHT1 was essential for the full virulence of C. albicans during systemic mouse infections, and the lack of VHT1 led to reduced fungal burden in C. glabrata-infected brains and C. albicans-infected brains and kidneys. Together, our data suggest a critical role of Vht1-mediated biotin acquisition for C. glabrata and C. albicans during intracellular growth in macrophages and systemic infections.


Subject(s)
Biotin/metabolism , Candida/metabolism , Homeostasis , Immune Evasion , Macrophages/microbiology , Phagocytosis/immunology , Animals , Biotin/genetics , Brain/microbiology , Candida/genetics , Candida/growth & development , Candida/pathogenicity , Candida albicans/genetics , Candida glabrata/genetics , Kidney/microbiology , Mice , Mice, Inbred BALB C , Phagosomes/microbiology , Symporters/genetics , Transcription Factors/metabolism , Virulence/genetics
5.
Curr Top Microbiol Immunol ; 422: 265-301, 2019.
Article in English | MEDLINE | ID: mdl-30062595

ABSTRACT

The human microbiota consists of bacteria, archaea, viruses, and fungi that build a highly complex network of interactions between each other and the host. While there are many examples for commensal bacterial influence on host health and immune modulation, little is known about the role of commensal fungi inside the gut community. Up until now, fungal research was concentrating on opportunistic diseases caused by fungal species, leaving the possible role of fungi as part of the microbiota largely unclear. Interestingly, fungal and bacterial abundance in the gut appear to be negatively correlated and disruption of the bacterial microbiota is a prerequisite for fungal overgrowth. The mechanisms behind bacterial colonization resistance are likely diverse, including direct antagonism as well as bacterial stimulation of host defense mechanisms. In this work, we will review the current knowledge of the development of the intestinal bacterial and fungal community, the influence of the microbiota on human health and disease, and the role of the opportunistic yeast C. albicans. We will furthermore discuss the possible benefits of commensal fungal colonization. Finally, we will summarize the recent findings on bacterial-fungal interactions.


Subject(s)
Bacteria , Fungi/physiology , Gastrointestinal Microbiome/physiology , Microbial Interactions , Bacterial Infections/microbiology , Candida albicans/pathogenicity , Candida albicans/physiology , Fungi/pathogenicity , Humans , Symbiosis
6.
Article in English | MEDLINE | ID: mdl-30420485

ABSTRACT

Candida glabrata can attach to various medical implants and forms thick biofilms despite its inability to switch from yeast to hyphae. The current in vivoC. glabrata biofilm models only provide limited information about colonization and infection and usually require animal sacrifice. To gain real-time information from individual BALB/c mice, we developed a noninvasive imaging technique to visualize C. glabrata biofilms in catheter fragments that were subcutaneously implanted on the back of mice. Bioluminescent C. glabrata reporter strains (lucOPT 7/2/4 and lucOPT 8/1/4), free of auxotrophic markers, expressing a codon-optimized firefly luciferase were generated. A murine subcutaneous model was used to follow real-time in vivo biofilm formation in the presence and absence of fluconazole and caspofungin. The fungal load in biofilms was quantified by CFU counts and by bioluminescence imaging (BLI). C. glabrata biofilms formed within the first 24 h, as documented by the increased number of device-associated cells and elevated bioluminescent signal compared with adhesion at the time of implant. The in vivo model allowed monitoring of the antibiofilm activity of caspofungin against C. glabrata biofilms through bioluminescent imaging from day four after the initiation of treatment. Contrarily, signals emitted from biofilms implanted in fluconazole-treated mice were similar to the light emitted from control-treated mice. This study gives insights into the real-time development of C. glabrata biofilms under in vivo conditions. BLI proved to be a dynamic, noninvasive, and sensitive tool to monitor continuous biofilm formation and activity of antifungal agents against C. glabrata biofilms formed on abiotic surfaces in vivo.


Subject(s)
Antifungal Agents/pharmacology , Caspofungin/pharmacology , Fluconazole/pharmacology , Animals , Biofilms/drug effects , Candida glabrata/drug effects , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests
7.
Cell Microbiol ; 20(12): e12943, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30112857

ABSTRACT

Candida glabrata is a common human fungal commensal and opportunistic pathogen. This fungus shows remarkable resilience as it can form recalcitrant biofilms on indwelling catheters, has intrinsic resistance against azole antifungals, and is causing vulvovaginal candidiasis. As a nosocomial pathogen, it can cause life-threatening bloodstream infections in immune-compromised patients. Here, we investigate the potential role of the high osmolarity glycerol response (HOG) MAP kinase pathway for C. glabrata virulence. The C. glabrata MAP kinase CgHog1 becomes activated by a variety of environmental stress conditions such as osmotic stress, low pH, and carboxylic acids and subsequently accumulates in the nucleus. We found that CgHog1 allows C. glabrata to persist within murine macrophages, but it is not required for systemic infection in a mouse model. C. glabrata and Lactobacilli co-colonise mucosal surfaces. Lactic acid at a concentration produced by vaginal Lactobacillus spp. causes CgHog1 phosphorylation and accumulation in the nucleus. In addition, CgHog1 enables C. glabrata to tolerate different Lactobacillus spp. and their metabolites when grown in co-culture. Using a phenotypic diverse set of clinical C. glabrata isolates, we find that the HOG pathway is likely the main quantitative determinant of lactic acid stress resistance. Taken together, our data indicate that CgHog1 has an important role in the confrontation of C. glabrata with the common vaginal flora.


Subject(s)
Antibiosis/physiology , Candida glabrata/physiology , Fungal Proteins/metabolism , Lactobacillus/physiology , Animals , Candida glabrata/drug effects , Candida glabrata/pathogenicity , Candidiasis/microbiology , Cell Nucleus/metabolism , Female , Fungal Proteins/genetics , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Lactic Acid/pharmacology , Macrophages/microbiology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Vagina/microbiology
9.
Mol Microbiol ; 103(4): 595-617, 2017 02.
Article in English | MEDLINE | ID: mdl-27623739

ABSTRACT

Morphogenesis in Candida albicans requires hyphal initiation and maintenance, and both processes are regulated by the fungal quorum sensing molecule (QSM) farnesol. We show that deletion of C. albicans EED1, which is crucial for hyphal extension and maintenance, led to a dramatically increased sensitivity to farnesol, and thus identified the first mutant hypersensitive to farnesol. Furthermore, farnesol decreased the transient filamentation of an eed1Δ strain without inducing cell death, indicating that two separate mechanisms mediate quorum sensing and cell lysis by farnesol. To analyze the cause of farnesol hypersensitivity we constructed either hyperactive or deletion mutants of factors involved in farnesol signaling, by introducing the hyperactive RAS1G13V or pADH1-CYR1CAT allele, or deleting CZF1 or NRG1 respectively. Neither of the constructs nor the exogenous addition of dB-cAMP was able to rescue the farnesol hypersensitivity, highlighting that farnesol mediates its effects not only via the cAMP pathway. Interestingly, the eed1Δ strain also displayed increased farnesol production. When eed1Δ was grown under continuous medium flow conditions, to remove accumulating QSMs from the supernatant, maintenance of eed1Δ filamentation, although not restored, was significantly prolonged, indicating a link between farnesol sensitivity, production, and the hyphal maintenance-defect in the eed1Δ mutant strain.


Subject(s)
Candida albicans/growth & development , Candida albicans/metabolism , Farnesol/metabolism , Fungal Proteins/genetics , Hyphae/growth & development , Quorum Sensing/physiology , Candida albicans/genetics , Cyclic AMP/metabolism , Gene Expression Regulation, Fungal , Hyphae/metabolism , Signal Transduction , Transcription Factors/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
10.
PLoS Pathog ; 12(9): e1005899, 2016 09.
Article in English | MEDLINE | ID: mdl-27649508

ABSTRACT

F-box proteins share the F-box domain to connect substrates of E3 SCF ubiquitin RING ligases through the adaptor Skp1/A to Cul1/A scaffolds. F-box protein Fbx15 is part of the general stress response of the human pathogenic mold Aspergillus fumigatus. Oxidative stress induces a transient peak of fbx15 expression, resulting in 3x elevated Fbx15 protein levels. During non-stress conditions Fbx15 is phosphorylated and F-box mediated interaction with SkpA preferentially happens in smaller subpopulations in the cytoplasm. The F-box of Fbx15 is required for an appropriate oxidative stress response, which results in rapid dephosphorylation of Fbx15 and a shift of the cellular interaction with SkpA to the nucleus. Fbx15 binds SsnF/Ssn6 as part of the RcoA/Tup1-SsnF/Ssn6 co-repressor and is required for its correct nuclear localization. Dephosphorylated Fbx15 prevents SsnF/Ssn6 nuclear localization and results in the derepression of gliotoxin gene expression. fbx15 deletion mutants are unable to infect immunocompromised mice in a model for invasive aspergillosis. Fbx15 has a novel dual molecular function by controlling transcriptional repression and being part of SCF E3 ubiquitin ligases, which is essential for stress response, gliotoxin production and virulence in the opportunistic human pathogen A. fumigatus.


Subject(s)
Aspergillosis/microbiology , Aspergillus fumigatus/enzymology , Gene Expression Regulation, Fungal , SKP Cullin F-Box Protein Ligases/metabolism , Amino Acid Sequence , Animals , Aspergillus fumigatus/genetics , Aspergillus fumigatus/growth & development , Aspergillus fumigatus/pathogenicity , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Cullin Proteins/genetics , Cullin Proteins/metabolism , Disease Models, Animal , F-Box Proteins/genetics , F-Box Proteins/metabolism , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gliotoxin/metabolism , Humans , Mice , Mutation , Oxidative Stress , Phosphorylation , Protein Transport , SKP Cullin F-Box Protein Ligases/genetics , Ubiquitins/metabolism , Virulence
11.
Crit Rev Microbiol ; 44(2): 230-243, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28609183

ABSTRACT

Candida albicans is a successful colonizer of the human host, which can, under certain circumstances cause a range of clinically diverse infections. Important virulence-associated traits of the fungus, such as the dimorphic switch and biofilm formation, are controlled by the quorum sensing molecule farnesol. Given the potential of farnesol as a novel antifungal drug, there has been increasing research into the mechanism underlying farnesol sensing and action in C. albicans. However, despite the identification of various factors involved in farnesol signalling, its exact mode of action remains largely unclear. This review provides an overview of the currently known aspects of farnesol production, sensing and action within C. albicans. We also illustrate the characteristic of C. albicans to simultaneously produce and tolerate high farnesol concentrations that are lethal to other microbes. Furthermore, we summarize new literature on the role of farnesol in the interaction of C. albicans with the human host and highlight its action as a potent immunomodulatory molecule.


Subject(s)
Anti-Infective Agents/metabolism , Candida albicans/physiology , Farnesol/metabolism , Quorum Sensing , Signal Transduction , Candida albicans/metabolism , Candidiasis/microbiology , Candidiasis/pathology , Host-Pathogen Interactions , Humans , Immunologic Factors/metabolism
12.
Med Mycol ; 56(suppl_1): 165-187, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29538732

ABSTRACT

The importance of fungal infections in both human and animals has increased over the last decades. This article represents an overview of the different categories of fungal infections that can be encountered in animals originating from environmental sources without transmission to humans. In addition, the endemic infections with indirect transmission from the environment, the zoophilic fungal pathogens with near-direct transmission, the zoonotic fungi that can be directly transmitted from animals to humans, mycotoxicoses and antifungal resistance in animals will also be discussed. Opportunistic mycoses are responsible for a wide range of diseases from localized infections to fatal disseminated diseases, such as aspergillosis, mucormycosis, candidiasis, cryptococcosis and infections caused by melanized fungi. The amphibian fungal disease chytridiomycosis and the Bat White-nose syndrome are due to obligatory fungal pathogens. Zoonotic agents are naturally transmitted from vertebrate animals to humans and vice versa. The list of zoonotic fungal agents is limited but some species, like Microsporum canis and Sporothrix brasiliensis from cats, have a strong public health impact. Mycotoxins are defined as the chemicals of fungal origin being toxic for warm-blooded vertebrates. Intoxications by aflatoxins and ochratoxins represent a threat for both human and animal health. Resistance to antifungals can occur in different animal species that receive these drugs, although the true epidemiology of resistance in animals is unknown, and options to treat infections caused by resistant infections are limited.


Subject(s)
Drug Resistance, Fungal , Mycoses/veterinary , Mycotoxicosis/veterinary , Animals , Antifungal Agents/therapeutic use , Endemic Diseases/veterinary , Humans , Mycoses/drug therapy , Mycoses/microbiology , Mycoses/transmission , Mycotoxins/toxicity , Opportunistic Infections/drug therapy , Opportunistic Infections/microbiology , Opportunistic Infections/transmission , Opportunistic Infections/veterinary , Zoonoses/drug therapy , Zoonoses/microbiology , Zoonoses/transmission
13.
Mol Microbiol ; 101(1): 92-108, 2016 07.
Article in English | MEDLINE | ID: mdl-26991818

ABSTRACT

Aspergillus fumigatus is the predominant airborne pathogenic fungus causing invasive aspergillosis in immunocompromised patients. During infection A. fumigatus has to adapt to oxygen-limiting conditions in inflammatory or necrotic tissue. Previously, we identified a mitochondrial protein to be highly up-regulated during hypoxic adaptation. Here, this protein was found to represent the novel oxidoreductase HorA. In Saccharomyces cerevisiae a homologue was shown to play a role in biosynthesis of coenzyme Q. Consistently, reduced coenzyme Q content in the generated ΔhorA mutant indicated a respective function in A. fumigatus. Since coenzyme Q is involved in cellular respiration and maintaining cellular redox homeostasis, the strain ΔhorA displayed an impaired response to both oxidative and reductive stress, a delay in germination and an accumulation of NADH. Moreover, an increased resistance against antifungal drugs was observed. All phenotypes were completely reversed by the addition of the synthetic electron carrier menadione. The deletion strain ΔhorA showed significantly attenuated virulence in two murine infection models of invasive pulmonary aspergillosis. Therefore, the biosynthesis of coenzyme Q and, particularly, the fungal-specific protein HorA play a crucial role in virulence of A. fumigatus. Due to its absence in mammals, HorA might represent a novel therapeutic target against fungal infections.


Subject(s)
Aspergillus fumigatus/metabolism , Azoles/pharmacology , Oxidoreductases/metabolism , Ubiquinone/analogs & derivatives , Animals , Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/enzymology , Aspergillus fumigatus/genetics , Cell Hypoxia/physiology , Disease Models, Animal , Female , Fungal Proteins/metabolism , Gene Deletion , Invasive Pulmonary Aspergillosis/microbiology , Mice , Oxidoreductases/genetics , Ubiquinone/biosynthesis , Virulence
14.
Curr Genet ; 63(5): 791-797, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28247023

ABSTRACT

Quorum sensing, a form of molecular communication in microbial communities, is relatively well studied in bacterial species, but poorly understood in fungi. Farnesol, a quorum sensing molecule secreted by the opportunistic human pathogenic fungus Candida albicans, was the first quorum sensing molecule described in a eukaryotic organism. However, despite considerable research efforts and advances in recent years, the mechanisms behind its action remain largely elusive. Only recently, we showed that deletion of the C. albicans gene EED1 (eed1Δ), which is essential for hyphal maintenance, resulted in both increased farnesol production and hypersensitivity to farnesol, providing a link between farnesol signaling and elongated hyphal growth. This finding raised several questions concerning farnesol signaling. In this short review we use the unique phenotype of the eed1Δ mutant to summarize current hypotheses and to speculate on possible mechanisms of quorum sensing in C. albicans and its implication in fungus-host interaction, by drawing comparisons to comparatively well-studied quorum sensing systems in bacteria.


Subject(s)
Bacterial Physiological Phenomena , Candida albicans/physiology , Farnesol/metabolism , Quorum Sensing , Host-Pathogen Interactions
15.
Cell Microbiol ; 18(12): 1709-1715, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27552083

ABSTRACT

Candida albicans is an important human opportunistic fungal pathogen which is frequently found as part of the normal human microbiota. It is well accepted that the fungus interacts with other components of the resident microbiota and that this impacts the commensal or pathogenic outcome of C. albicans colonization. Different types of interactions, including synergism or antagonism, contribute to a complex balance between the multitude of different species. Mixed biofilms of C. albicans and streptococci are a well-studied example of a mutualistic interaction often potentiating the virulence of the individual members. In contrast, other bacteria like lactobacilli are known to antagonize C. albicans, and research has just started elucidating the mechanisms behind these interactions. This scenario is even more complicated by a third player, the host. This review focuses on interactions between C. albicans and gram-positive bacteria whose investigation will without doubt ultimately help understanding C. albicans infections.


Subject(s)
Biofilms/growth & development , Candida albicans/pathogenicity , Candidiasis/immunology , Gram-Positive Bacteria/pathogenicity , Gram-Positive Bacterial Infections/immunology , Lactobacillaceae/pathogenicity , Antibiosis/physiology , Bacterial Adhesion , Candida albicans/genetics , Candida albicans/growth & development , Candidiasis/microbiology , Coinfection , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/growth & development , Gram-Positive Bacterial Infections/microbiology , Host-Pathogen Interactions , Humans , Lactobacillaceae/genetics , Lactobacillaceae/growth & development , Symbiosis/physiology , Virulence
16.
Vet Res ; 48(1): 23, 2017 04 12.
Article in English | MEDLINE | ID: mdl-28403908

ABSTRACT

Ruminants are the main source of human infections with the obligate intracellular bacterium Coxiella (C.) burnetii. Infected animals shed high numbers of C. burnetii by milk, feces, and birth products. In goats, shedding by the latter route coincides with C. burnetii replication in epithelial (trophoblast) cells of the placenta, which led us to hypothesize that epithelial cells are generally implicated in replication and shedding of C. burnetii. We therefore aimed at analyzing the interactions of C. burnetii with epithelial cells of the bovine host (1) at the entry site (lung epithelium) which govern host immune responses and (2) in epithelial cells of gut, udder and placenta decisive for the quantity of pathogen excretion. Epithelial cell lines [PS (udder), FKD-R 971 (small intestine), BCEC (maternal placenta), F3 (fetal placenta), BEL-26 (lung)] were inoculated with C. burnetii strains Nine Mile I (NMI) and NMII at different cultivation conditions. The cell lines exhibited different permissiveness for C. burnetii. While maintaining cell viability, udder cells allowed the highest replication rates with formation of large cell-filling Coxiella containing vacuoles. Intestinal cells showed an enhanced susceptibility to invasion but supported C. burnetii replication only at intermediate levels. Lung and placental cells also internalized the bacteria but in strikingly smaller numbers. In any of the epithelial cells, both Coxiella strains failed to trigger a substantial IL-1ß, IL-6 and TNF-α response. Epithelial cells, with mammary epithelial cells in particular, may therefore serve as a niche for C. burnetii replication in vivo without alerting the host's immune response.


Subject(s)
Cattle Diseases/microbiology , Coxiella burnetii/physiology , Epithelial Cells/microbiology , Intestinal Mucosa/microbiology , Lung/microbiology , Mammary Glands, Animal/microbiology , Placenta/microbiology , Q Fever/veterinary , Animals , Bacterial Shedding , Cattle/microbiology , Cell Line , Cytokines/physiology , Female , Flow Cytometry/veterinary , Host-Pathogen Interactions/physiology , Microscopy, Fluorescence/veterinary , Pregnancy , Q Fever/microbiology , Real-Time Polymerase Chain Reaction/veterinary
17.
PLoS Genet ; 10(8): e1004496, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25121733

ABSTRACT

Lichtheimia species are the second most important cause of mucormycosis in Europe. To provide broader insights into the molecular basis of the pathogenicity-associated traits of the basal Mucorales, we report the full genome sequence of L. corymbifera and compared it to the genome of Rhizopus oryzae, the most common cause of mucormycosis worldwide. The genome assembly encompasses 33.6 MB and 12,379 protein-coding genes. This study reveals four major differences of the L. corymbifera genome to R. oryzae: (i) the presence of an highly elevated number of gene duplications which are unlike R. oryzae not due to whole genome duplication (WGD), (ii) despite the relatively high incidence of introns, alternative splicing (AS) is not frequently observed for the generation of paralogs and in response to stress, (iii) the content of repetitive elements is strikingly low (<5%), (iv) L. corymbifera is typically haploid. Novel virulence factors were identified which may be involved in the regulation of the adaptation to iron-limitation, e.g. LCor01340.1 encoding a putative siderophore transporter and LCor00410.1 involved in the siderophore metabolism. Genes encoding the transcription factors LCor08192.1 and LCor01236.1, which are similar to GATA type regulators and to calcineurin regulated CRZ1, respectively, indicating an involvement of the calcineurin pathway in the adaption to iron limitation. Genes encoding MADS-box transcription factors are elevated up to 11 copies compared to the 1-4 copies usually found in other fungi. More findings are: (i) lower content of tRNAs, but unique codons in L. corymbifera, (ii) Over 25% of the proteins are apparently specific for L. corymbifera. (iii) L. corymbifera contains only 2/3 of the proteases (known to be essential virulence factors) in comparison to R. oryzae. On the other hand, the number of secreted proteases, however, is roughly twice as high as in R. oryzae.


Subject(s)
Evolution, Molecular , Genome, Fungal , Mucorales/genetics , Mucormycosis/genetics , Alternative Splicing/genetics , Gene Duplication , Genomics , Humans , Mucorales/pathogenicity , Mucormycosis/microbiology , Virulence Factors/genetics , Virulence Factors/isolation & purification
18.
PLoS Genet ; 10(12): e1004824, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25474009

ABSTRACT

Following antifungal treatment, Candida albicans, and other human pathogenic fungi can undergo microevolution, which leads to the emergence of drug resistance. However, the capacity for microevolutionary adaptation of fungi goes beyond the development of resistance against antifungals. Here we used an experimental microevolution approach to show that one of the central pathogenicity mechanisms of C. albicans, the yeast-to-hyphae transition, can be subject to experimental evolution. The C. albicans cph1Δ/efg1Δ mutant is nonfilamentous, as central signaling pathways linking environmental cues to hyphal formation are disrupted. We subjected this mutant to constant selection pressure in the hostile environment of the macrophage phagosome. In a comparatively short time-frame, the mutant evolved the ability to escape macrophages by filamentation. In addition, the evolved mutant exhibited hyper-virulence in a murine infection model and an altered cell wall composition compared to the cph1Δ/efg1Δ strain. Moreover, the transcriptional regulation of hyphae-associated, and other pathogenicity-related genes became re-responsive to environmental cues in the evolved strain. We went on to identify the causative missense mutation via whole genome- and transcriptome-sequencing: a single nucleotide exchange took place within SSN3 that encodes a component of the Cdk8 module of the Mediator complex, which links transcription factors with the general transcription machinery. This mutation was responsible for the reconnection of the hyphal growth program with environmental signals in the evolved strain and was sufficient to bypass Efg1/Cph1-dependent filamentation. These data demonstrate that even central transcriptional networks can be remodeled very quickly under appropriate selection pressure.


Subject(s)
Candida albicans/genetics , Candida albicans/pathogenicity , Hyphae/genetics , Macrophages/microbiology , Virulence/genetics , Animals , Candidiasis/microbiology , Candidiasis/mortality , Cell Wall/genetics , Cell Wall/metabolism , Cells, Cultured , Directed Molecular Evolution , Gene Expression Regulation, Fungal , Genetic Variation , Hyphae/pathogenicity , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Organisms, Genetically Modified
19.
PLoS Pathog ; 10(10): e1004478, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25356907

ABSTRACT

Candida glabrata is one of the most common causes of candidemia, a life-threatening, systemic fungal infection, and is surpassed in frequency only by Candida albicans. Major factors contributing to the success of this opportunistic pathogen include its ability to readily acquire resistance to antifungals and to colonize and adapt to many different niches in the human body. Here we addressed the flexibility and adaptability of C. glabrata during interaction with macrophages with a serial passage approach. Continuous co-incubation of C. glabrata with a murine macrophage cell line for over six months resulted in a striking alteration in fungal morphology: The growth form changed from typical spherical yeasts to pseudohyphae-like structures - a phenotype which was stable over several generations without any selective pressure. Transmission electron microscopy and FACS analyses showed that the filamentous-like morphology was accompanied by changes in cell wall architecture. This altered growth form permitted faster escape from macrophages and increased damage of macrophages. In addition, the evolved strain (Evo) showed transiently increased virulence in a systemic mouse infection model, which correlated with increased organ-specific fungal burden and inflammatory response (TNFα and IL-6) in the brain. Similarly, the Evo mutant significantly increased TNFα production in the brain on day 2, which is mirrored in macrophages confronted with the Evo mutant, but not with the parental wild type. Whole genome sequencing of the Evo strain, genetic analyses, targeted gene disruption and a reverse microevolution experiment revealed a single nucleotide exchange in the chitin synthase-encoding CHS2 gene as the sole basis for this phenotypic alteration. A targeted CHS2 mutant with the same SNP showed similar phenotypes as the Evo strain under all experimental conditions tested. These results indicate that microevolutionary processes in host-simulative conditions can elicit adaptations of C. glabrata to distinct host niches and even lead to hypervirulent strains.


Subject(s)
Adaptation, Physiological , Candida glabrata/genetics , Candidiasis/microbiology , Macrophages/microbiology , Polymorphism, Single Nucleotide , Animals , Candida glabrata/growth & development , Candida glabrata/pathogenicity , Cell Line , Chitin Synthase/genetics , Chitin Synthase/metabolism , Disease Models, Animal , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Host-Pathogen Interactions , Humans , Hyphae , Mice , Point Mutation , Serial Passage , Specific Pathogen-Free Organisms , Virulence
20.
PLoS Pathog ; 10(6): e1004211, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24945925

ABSTRACT

The opportunistic fungal pathogen Candida glabrata is a frequent cause of candidiasis, causing infections ranging from superficial to life-threatening disseminated disease. The inherent tolerance of C. glabrata to azole drugs makes this pathogen a serious clinical threat. To identify novel genes implicated in antifungal drug tolerance, we have constructed a large-scale C. glabrata deletion library consisting of 619 unique, individually bar-coded mutant strains, each lacking one specific gene, all together representing almost 12% of the genome. Functional analysis of this library in a series of phenotypic and fitness assays identified numerous genes required for growth of C. glabrata under normal or specific stress conditions, as well as a number of novel genes involved in tolerance to clinically important antifungal drugs such as azoles and echinocandins. We identified 38 deletion strains displaying strongly increased susceptibility to caspofungin, 28 of which encoding proteins that have not previously been linked to echinocandin tolerance. Our results demonstrate the potential of the C. glabrata mutant collection as a valuable resource in functional genomics studies of this important fungal pathogen of humans, and to facilitate the identification of putative novel antifungal drug target and virulence genes.


Subject(s)
Antifungal Agents/pharmacology , Azoles/pharmacology , Candida glabrata/genetics , Drug Resistance, Fungal/genetics , Echinocandins/pharmacology , Biofilms/growth & development , Candida glabrata/growth & development , Candidiasis/drug therapy , Caspofungin , Cell Wall/drug effects , Cell Wall/genetics , Fungal Proteins/genetics , Gene Deletion , Gene Knockout Techniques , Gene Library , Lipopeptides , Microbial Sensitivity Tests , Osmotic Pressure , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL