Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 18(12): e1011021, 2022 12.
Article in English | MEDLINE | ID: mdl-36476844

ABSTRACT

Toxoplasma gondii is an intracellular parasite that can infect many host species and is a cause of significant human morbidity worldwide. T. gondii secretes a diverse array of effector proteins into the host cell which are critical for infection. The vast majority of these secreted proteins have no predicted functional domains and remain uncharacterised. Here, we carried out a pooled CRISPR knockout screen in the T. gondii Prugniaud strain in vivo to identify secreted proteins that contribute to parasite immune evasion in the host. We demonstrate that ROP1, the first-identified rhoptry protein of T. gondii, is essential for virulence and has a previously unrecognised role in parasite resistance to interferon gamma-mediated innate immune restriction. This function is conserved in the highly virulent RH strain of T. gondii and contributes to parasite growth in both murine and human macrophages. While ROP1 affects the morphology of rhoptries, from where the protein is secreted, it does not affect rhoptry secretion. Finally, we show that ROP1 co-immunoprecipitates with the host cell protein C1QBP, an emerging regulator of innate immune signaling. In summary, we identify putative in vivo virulence factors in the T. gondii Prugniaud strain and show that ROP1 is an important and previously overlooked effector protein that counteracts both murine and human innate immunity.


Subject(s)
Immunity, Innate , Protozoan Proteins , Toxoplasma , Animals , Humans , Mice , Carrier Proteins , Mitochondrial Proteins/metabolism , Protozoan Proteins/metabolism , Virulence Factors
2.
Cell Surf ; 6: 100042, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33364531

ABSTRACT

We designed experiments to assess whether fungal cell wall mannans function as an immune shield or an immune agonist. Fungal cell wall ß-(1,3)-glucan normally plays a major and dominant role in immune activation. The outer mannan layer has been variously described as an immune shield, because it has the potential to mask the underlying ß-(1,3)-glucan, or an immune activator, as it also has the potential to engage with a wide range of mannose detecting PRRs. To resolve this conundrum we examined species-specific differences in host immune recognition in the och1Δ N-mannosylation-deficient mutant background in four species of yeast-like fungi. Irrespective of the fungal species, the cytokine response (TNFα and IL-6) induced by the och1Δ mutants in human monocytes was reduced compared to that of the wild type. In contrast, TNFα production induced by och1Δ was increased, relative to wild type, due to increased ß-glucan exposure, when mouse or human macrophages were used. These observations suggest that N-mannan is not a major PAMP for macrophages and that in these cells mannan does shield the fungus from recognition of the inner cell wall ß-glucan. However, N-mannan is a significant inducer of cytokine for monocytes. Therefore the metaphor of the fungal "mannan shield" can only be applied to some, but not all, myeloid cells used in immune profiling experiments of fungal species.

3.
mBio ; 11(4)2020 08 11.
Article in English | MEDLINE | ID: mdl-32788384

ABSTRACT

Amino acid metabolism is crucial for fungal growth and development. Ureohydrolases produce amines when acting on l-arginine, agmatine, and guanidinobutyrate (GB), and these enzymes generate ornithine (by arginase), putrescine (by agmatinase), or GABA (by 4-guanidinobutyrase or GBase). Candida albicans can metabolize and grow on arginine, agmatine, or guanidinobutyrate as the sole nitrogen source. Three related C. albicans genes whose sequences suggested that they were putative arginase or arginase-like genes were examined for their role in these metabolic pathways. Of these, Car1 encoded the only bona fide arginase, whereas we provide evidence that the other two open reading frames, orf19.5862 and orf19.3418, encode agmatinase and guanidinobutyrase (Gbase), respectively. Analysis of strains with single and multiple mutations suggested the presence of arginase-dependent and arginase-independent routes for polyamine production. CAR1 played a role in hyphal morphogenesis in response to arginine, and the virulence of a triple mutant was reduced in both Galleria mellonella and Mus musculus infection models. In the bloodstream, arginine is an essential amino acid that is required by phagocytes to synthesize nitric oxide (NO). However, none of the single or multiple mutants affected host NO production, suggesting that they did not influence the oxidative burst of phagocytes.IMPORTANCE We show that the C. albicans ureohydrolases arginase (Car1), agmatinase (Agt1), and guanidinobutyrase (Gbu1) can orchestrate an arginase-independent route for polyamine production and that this is important for C. albicans growth and survival in microenvironments of the mammalian host.


Subject(s)
Agmatine/metabolism , Arginine/metabolism , Candida albicans/enzymology , Candida albicans/pathogenicity , Fungal Proteins/metabolism , Ureohydrolases/metabolism , Amino Acids/metabolism , Animals , Arginase/genetics , Arginase/metabolism , Cloning, Molecular , Female , Larva/microbiology , Metabolic Networks and Pathways , Mice , Mice, Inbred BALB C , Moths/microbiology , RAW 264.7 Cells , Ureohydrolases/genetics , Virulence
4.
Elife ; 82019 10 02.
Article in English | MEDLINE | ID: mdl-31577230

ABSTRACT

Toxoplasma gondii contains a limited subset of actin binding proteins. Here we show that the putative actin regulator cyclase-associated protein (CAP) is present in two different isoforms and its deletion leads to significant defects in some but not all actin dependent processes. We observe defects in cell-cell communication, daughter cell orientation and the juxtanuclear accumulation of actin, but only modest defects in synchronicity of division and no defect in the replication of the apicoplast. 3D electron microscopy reveals that loss of CAP results in a defect in formation of a normal central residual body, but parasites remain connected within the vacuole. This dissociates synchronicity of division and parasite rosetting and reveals that establishment and maintenance of the residual body may be more complex than previously thought. These results highlight the different spatial requirements for F-actin regulation in Toxoplasma which appear to be achieved by partially overlapping functions of actin regulators.


Subject(s)
Actins/metabolism , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Cell Communication , Cell Division , Gene Deletion , Protein Isoforms/deficiency , Protein Isoforms/metabolism , Protozoan Proteins/genetics
5.
Nat Commun ; 10(1): 3963, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31481656

ABSTRACT

Genome-wide CRISPR screening is a powerful tool to identify genes required under selective conditions. However, the inherent scale of genome-wide libraries can limit their application in experimental settings where cell numbers are restricted, such as in vivo infections or single cell analysis. The use of small scale CRISPR libraries targeting gene subsets circumvents this problem. Here we develop a method for rapid generation of custom guide RNA (gRNA) libraries using arrayed single-stranded oligonucleotides for reproducible pooled cloning of CRISPR/Cas9 libraries. We use this system to generate mutant pools of different sizes in the protozoan parasite Toxoplasma gondi and describe optimised analysis methods for small scale libraries. An in vivo genetic screen in the murine host identifies novel and known virulence factors and we confirm results using cloned knock-out parasites. Our study also reveals a potential trans-rescue of individual knock-out parasites in pools of mutants compared to homogenous knock-out lines of the key virulence factor MYR1.


Subject(s)
CRISPR-Cas Systems , Toxoplasma/genetics , Virulence Factors/genetics , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Knockout Techniques/methods , Gene Library , Genome, Protozoan , Humans , Mice, Inbred C57BL , RNA, Guide, Kinetoplastida , Toxoplasma/pathogenicity , Toxoplasmosis/genetics , Toxoplasmosis/parasitology , Toxoplasmosis/pathology
6.
Cell Surf ; 1: 15-24, 2018 Mar.
Article in English | MEDLINE | ID: mdl-30123851

ABSTRACT

Cryptococcus neoformans is a human fungal pathogen that often causes infections in immunocompromised individuals. Upon inhalation into the lungs C. neoformans differentiates into cells with altered size and morphology, including production of large titan cells. Titan cells possess thickened cell wall and dense, cross-linked capsule when compared to in vitro grown cells. In addition, titan cells have increased cell wall chitin that is associated with a detrimental anti-inflammatory immune response. Here we examined the cell wall and capsule composition of in vitro, in vivo typical-sized and in vivo titan cells using High Performance Liquid Chromatography (HPLC). The monomer composition of cell wall polysaccharides showed that in vivo C. neoformans cells contained more glucosamine and less glucose than in vitro cells, suggesting alteration in abundance of both chitin and glucans, respectively. Low levels of galactosamine were also detected in carbohydrates from both in vivo and vitro cells. Within the in vivo cell population, differences in the proportions of cell wall and capsule monomers between typical and titan cells were also observed. Taken together, these results demonstrate that C. neoformans reshapes its cell wall and capsule composition during infection. These cell wall and capsule alterations likely help C. neoformans escape recognition by, and allow modulation of, the host immune system.

7.
PLoS One ; 12(10): e0184438, 2017.
Article in English | MEDLINE | ID: mdl-29023454

ABSTRACT

Candida albicans is an inhabitant of mucosal surfaces in healthy individuals but also the most common cause of fungal nosocomial blood stream infections, associated with high morbidity and mortality. As such life-threatening infections often disseminate from superficial mucosal infections we aimed to study the use of probiotic Lactobacillus rhamnosus GG (LGG) in prevention of mucosal C. albicans infections. Here, we demonstrate that LGG protects oral epithelial tissue from damage caused by C. albicans in our in vitro model of oral candidiasis. Furthermore, we provide insights into the mechanisms behind this protection and dissect direct and indirect effects of LGG on C. albicans pathogenicity. C. albicans viability was not affected by LGG. Instead, transcriptional profiling using RNA-Seq indicated dramatic metabolic reprogramming of C. albicans. Additionally, LGG had a significant impact on major virulence attributes, including adhesion, invasion, and hyphal extension, whose reduction, consequently, prevented epithelial damage. This was accompanied by glucose depletion and repression of ergosterol synthesis, caused by LGG, but also due to blocked adhesion sites. Therefore, LGG protects oral epithelia against C. albicans infection by preventing fungal adhesion, invasion and damage, driven, at least in parts, by metabolic reprogramming due to nutrient limitation caused by LGG.


Subject(s)
Bacterial Adhesion/drug effects , Candida albicans/pathogenicity , Candidiasis/immunology , Epithelial Cells/immunology , Glucose/deficiency , Lacticaseibacillus rhamnosus/physiology , Mouth/immunology , Probiotics/administration & dosage , Antifungal Agents/administration & dosage , Biofilms/drug effects , Biofilms/growth & development , Candidiasis/drug therapy , Candidiasis/microbiology , Cross Infection , Culture Media , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Humans , Mouth/drug effects , Mouth/microbiology
8.
mBio ; 8(1)2017 01 31.
Article in English | MEDLINE | ID: mdl-28143983

ABSTRACT

Compared to other fungal pathogens, Cryptococcus neoformans is particularly adept at avoiding detection by innate immune cells. To explore fungal cellular features involved in immune avoidance, we characterized cell surface changes of the C. neoformans rim101Δ mutant, a strain that fails to organize and shield immunogenic epitopes from host detection. These cell surface changes are associated with an exaggerated, detrimental inflammatory response in mouse models of infection. We determined that the disorganized strain rim101Δ cell wall increases macrophage detection in a contact-dependent manner. Using biochemical and microscopy methods, we demonstrated that the rim101Δ strain shows a modest increase in the levels of both cell wall chitin and chitosan but that it shows a more dramatic increase in chito-oligomer exposure, as measured by wheat germ agglutinin staining. We also created a series of mutants with various levels of cell wall wheat germ agglutinin staining, and we demonstrated that the staining intensity correlates with the degree of macrophage activation in response to each strain. To explore the host receptors responsible for recognizing the rim101Δ mutant, we determined that both the MyD88 and CARD9 innate immune signaling proteins are involved. Finally, we characterized the immune response to the rim101Δ mutant in vivo, documenting a dramatic and sustained increase in Th1 and Th17 cytokine responses. These results suggest that the Rim101 transcription factor actively regulates the C. neoformans cell wall to prevent the exposure of immune stimulatory molecules within the host. These studies further explored the ways in which immune cells detect C. neoformans and other fungal pathogens by mechanisms that include sensing N-acetylglucosamine-containing structures, such as chitin and chitosan. IMPORTANCE: Infectious microorganisms have developed many ways to avoid recognition by the host immune system. For example, pathogenic fungi alter their cell surfaces to mask immunogenic epitopes. We have created a fungal strain with a targeted mutation in a pH response pathway that is unable to properly organize its cell wall, resulting in a dramatic immune reaction during infection. This mutant cell wall is defective in hiding important cell wall components, such as the chito-oligomers chitin and chitosan. By creating a series of cell wall mutants, we demonstrated that the degree of chito-oligomer exposure correlates with the intensity of innate immune cell activation. This activation requires a combination of host receptors to recognize and respond to these infecting microorganisms. Therefore, these experiments explored host-pathogen interactions that determine the degree of the subsequent inflammatory response and the likely outcome of infection.


Subject(s)
Cell Wall/immunology , Cell Wall/metabolism , Cryptococcus neoformans/metabolism , Immune Evasion , Inflammation/pathology , Transcription Factors/metabolism , Animals , Cryptococcosis/microbiology , Cryptococcosis/pathology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/immunology , Cryptococcus neoformans/pathogenicity , Disease Models, Animal , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , Macrophages/immunology , Mice , Th1 Cells/immunology , Th17 Cells/immunology , Transcription Factors/genetics
9.
mBio ; 8(1)2017 01 24.
Article in English | MEDLINE | ID: mdl-28119468

ABSTRACT

The opportunistic human fungal pathogen Candida albicans can cause a variety of diseases, ranging from superficial mucosal infections to life-threatening systemic infections. Phagocytic cells of the innate immune response, such as neutrophils and macrophages, are important first-line responders to an infection and generate reactive oxygen and nitrogen species as part of their protective antimicrobial response. During an infection, host cells generate nitric oxide through the enzyme inducible nitric oxide synthase (iNOS) to kill the invading pathogen. Inside the phagocyte, iNOS competes with the enzyme arginase-1 for a common substrate, the amino acid l-arginine. Several pathogenic species, including bacteria and parasitic protozoans, actively modulate the production of nitric oxide by inducing their own arginases or the host's arginase activity to prevent the conversion of l-arginine to nitric oxide. We report here that C. albicans blocks nitric oxide production in human-monocyte-derived macrophages by induction of host arginase activity. We further determined that purified chitin (a fungal cell wall polysaccharide) and increased chitin exposure at the fungal cell wall surface induces this host arginase activity. Blocking the C. albicans-induced arginase activity with the arginase-specific substrate inhibitor Nω-hydroxy-nor-arginine (nor-NOHA) or the chitinase inhibitor bisdionin F restored nitric oxide production and increased the efficiency of fungal killing. Moreover, we determined that C. albicans influences macrophage polarization from a classically activated phenotype toward an alternatively activated phenotype, thereby reducing antimicrobial functions and mediating fungal survival. Therefore, C. albicans modulates l-arginine metabolism in macrophages during an infection, potentiating its own survival. IMPORTANCE: The availability and metabolism of amino acids are increasingly recognized as crucial regulators of immune functions. In acute infections, the conversion of the "conditionally essential" amino acid l-arginine by the inducible nitric oxide synthase to nitric oxide is a resistance factor that is produced by the host to fight pathogens. Manipulation of these host defense mechanisms by the pathogen can be key to successful host invasion. We show here that the human opportunistic fungal pathogen Candida albicans influences l-arginine availability for nitric oxide production by induction of the substrate-competing host enzyme arginase-1. This led to a reduced production of nitric oxide and, moreover, reduced eradication of the fungus by human macrophages. We demonstrate that blocking of host arginase-1 activity restored nitric oxide production and increased the killing potential of macrophages. These results highlight the therapeutic potential of l-arginine metabolism in fungal diseases.


Subject(s)
Arginase/metabolism , Candida albicans/chemistry , Chitin/metabolism , Host-Pathogen Interactions , Immune Evasion , Macrophages/enzymology , Macrophages/metabolism , Candida albicans/immunology , Cells, Cultured , Healthy Volunteers , Humans , Macrophages/immunology , Macrophages/microbiology , Nitric Oxide/metabolism
10.
Nat Microbiol ; 2: 16238, 2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27941860

ABSTRACT

As they proliferate, fungi expose antigens at their cell surface that are potent stimulators of the innate immune response, and yet the commensal fungus Candida albicans is able to colonize immuno competent individuals. We show that C. albicans may evade immune detection by presenting a moving immunological target. We report that the exposure of ß-glucan, a key pathogen-associated molecular pattern (PAMP) located at the cell surface of C. albicans and other pathogenic Candida species, is modulated in response to changes in the carbon source. Exposure to lactate induces ß-glucan masking in C. albicans via a signalling pathway that has recruited an evolutionarily conserved receptor (Gpr1) and transcriptional factor (Crz1) from other well-characterized pathways. In response to lactate, these regulators control the expression of cell-wall-related genes that contribute to ß-glucan masking. This represents the first description of active PAMP masking by a Candida species, a process that reduces the visibility of the fungus to the immune system.


Subject(s)
Candida albicans/immunology , Candida albicans/metabolism , Immune Evasion , Lactic Acid/metabolism , Membrane Proteins/metabolism , beta-Glucans/metabolism , Glycosylation
11.
Curr Opin Microbiol ; 34: 111-118, 2016 12.
Article in English | MEDLINE | ID: mdl-27689902

ABSTRACT

Candida albicans is a commensal coloniser of most people and a pathogen of the immunocompromised or patients in which barriers that prevent dissemination have been disrupted. Both the commensal and pathogenic states involve regulation and adaptation to the host microenvironment. The pathogenic potential can be downregulated to sustain commensalism or upregulated to damage host tissue and avoid and subvert immune surveillance. In either case it seems as though the cell biology of this fungus has evolved to enable the establishment of different types of relationships with the human host. Here we summarise latest advances in the analysis of mechanisms that enable C. albicans to occupy different body sites whilst avoiding being eliminated by the sentinel activities of the human immune system.


Subject(s)
Candida albicans/physiology , Host-Pathogen Interactions , Adaptation, Physiological , Animals , Candida albicans/immunology , Candida albicans/pathogenicity , Candidiasis/microbiology , Fungal Proteins/immunology , Fungal Proteins/metabolism , Humans , Immune Evasion , Mice , Symbiosis
12.
Eukaryot Cell ; 14(7): 684-97, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26002719

ABSTRACT

The yeast exocyst is a multiprotein complex comprised of eight subunits (Sec3, Sec5, Sec6, Sec8, Sec10, Sec15, Exo70, and Exo84) which orchestrates trafficking of exocytic vesicles to specific docking sites on the plasma membrane during polarized secretion. To study SEC6 function in Candida albicans, we generated a conditional mutant strain in which SEC6 was placed under the control of a tetracycline-regulated promoter. In the repressed state, the tetR-SEC6 mutant strain (denoted tSEC6) was viable for up to 27 h; thus, all phenotypic analyses were performed at 24 h or earlier. Strain tSEC6 under repressing conditions had readily apparent defects in cytokinesis and endocytosis and accumulated both post-Golgi apparatus secretory vesicles and structures suggestive of late endosomes. Strain tSEC6 was markedly defective in secretion of aspartyl proteases and lipases as well as filamentation under repressing conditions. Lack of SEC6 expression resulted in markedly reduced lateral hyphal branching, which requires the establishment of a new axis of polarized secretion. Aberrant localization of chitin at the septum and increased resistance to zymolyase activity were observed, suggesting that C. albicans Sec6 plays an important role in mediating trafficking and delivery of cell wall components. The tSEC6 mutant was also markedly defective in macrophage killing, indicating a role of SEC6 in C. albicans virulence. Taken together, these studies indicate that the late secretory protein Sec6 is required for polarized secretion, hyphal morphogenesis, and the pathogenesis of C. albicans.


Subject(s)
Candida albicans/growth & development , Candidiasis/microbiology , Cell Wall/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Hyphae/growth & development , Macrophages/pathology , Vesicular Transport Proteins/metabolism , Animals , Candida albicans/genetics , Candida albicans/metabolism , Candidiasis/genetics , Candidiasis/metabolism , Cell Membrane/metabolism , Cell Survival , Exocytosis/physiology , Fungal Proteins/genetics , Hyphae/genetics , Hyphae/metabolism , Macrophages/microbiology , Mice , Mutation/genetics , Protein Transport , Secretory Vesicles/metabolism , Vesicular Transport Proteins/genetics , Virulence
14.
PLoS Pathog ; 10(4): e1004050, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24722226

ABSTRACT

Chitin is an essential structural polysaccharide of fungal pathogens and parasites, but its role in human immune responses remains largely unknown. It is the second most abundant polysaccharide in nature after cellulose and its derivatives today are widely used for medical and industrial purposes. We analysed the immunological properties of purified chitin particles derived from the opportunistic human fungal pathogen Candida albicans, which led to the selective secretion of the anti-inflammatory cytokine IL-10. We identified NOD2, TLR9 and the mannose receptor as essential fungal chitin-recognition receptors for the induction of this response. Chitin reduced LPS-induced inflammation in vivo and may therefore contribute to the resolution of the immune response once the pathogen has been defeated. Fungal chitin also induced eosinophilia in vivo, underpinning its ability to induce asthma. Polymorphisms in the identified chitin receptors, NOD2 and TLR9, predispose individuals to inflammatory conditions and dysregulated expression of chitinases and chitinase-like binding proteins, whose activity is essential to generate IL-10-inducing fungal chitin particles in vitro, have also been linked to inflammatory conditions and asthma. Chitin recognition is therefore critical for immune homeostasis and is likely to have a significant role in infectious and allergic disease.


Subject(s)
Candida albicans/chemistry , Chitin/immunology , Interleukin-10/immunology , Nod2 Signaling Adaptor Protein/immunology , Toll-Like Receptor 9/immunology , Animals , Asthma/genetics , Asthma/immunology , Asthma/pathology , Candida albicans/immunology , Chitin/chemistry , Female , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Male , Mice , Nod2 Signaling Adaptor Protein/genetics , Toll-Like Receptor 9/genetics
15.
PLoS Pathog ; 9(4): e1003276, 2013.
Article in English | MEDLINE | ID: mdl-23633946

ABSTRACT

The fungal cell wall is the first point of interaction between an invading fungal pathogen and the host immune system. The outer layer of the cell wall is comprised of GPI anchored proteins, which are post-translationally modified by both N- and O-linked glycans. These glycans are important pathogen associated molecular patterns (PAMPs) recognised by the innate immune system. Glycan synthesis is mediated by a series of glycosyl transferases, located in the endoplasmic reticulum and Golgi apparatus. Mnn2 is responsible for the addition of the initial α1,2-mannose residue onto the α1,6-mannose backbone, forming the N-mannan outer chain branches. In Candida albicans, the MNN2 gene family is comprised of six members (MNN2, MNN21, MNN22, MNN23, MNN24 and MNN26). Using a series of single, double, triple, quintuple and sextuple mutants, we show, for the first time, that addition of α1,2-mannose is required for stabilisation of the α1,6-mannose backbone and hence regulates mannan fibril length. Sequential deletion of members of the MNN2 gene family resulted in the synthesis of lower molecular weight, less complex and more uniform N-glycans, with the sextuple mutant displaying only un-substituted α1,6-mannose. TEM images confirmed that the sextuple mutant was completely devoid of the outer mannan fibril layer, while deletion of two MNN2 orthologues resulted in short mannan fibrils. These changes in cell wall architecture correlated with decreased proinflammatory cytokine induction from monocytes and a decrease in fungal virulence in two animal models. Therefore, α1,2-mannose of N-mannan is important for both immune recognition and virulence of C. albicans.


Subject(s)
Candida albicans/immunology , Candida albicans/pathogenicity , Mannans/immunology , Mannose/metabolism , Mannosyltransferases/metabolism , Membrane Glycoproteins/immunology , Animals , Candida albicans/enzymology , Candidiasis/immunology , Cell Wall/chemistry , Cell Wall/immunology , Female , Fungal Proteins/genetics , Fungal Proteins/immunology , Fungal Proteins/metabolism , Humans , Mannans/chemistry , Mannose/chemistry , Mannosyltransferases/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Polysaccharides/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Sequence Alignment , Sequence Deletion
16.
J Invest Dermatol ; 133(1): 144-53, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22832495

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) has an important role not only in glycolysis but also in nonmetabolic processes, including transcription activation and apoptosis. We report the isolation of a human GAPDH (hGAPDH) (2-32) fragment peptide from human placental tissue exhibiting antimicrobial activity. The peptide was internalized by cells of the pathogenic yeast Candida albicans and initiated a rapid apoptotic mechanism, leading to killing of the fungus. Killing was dose-dependent, with 10 µg ml (3.1 µM) and 100 µg ml hGAPDH (2-32) depolarizing 45% and 90% of the fungal cells in a population, respectively. Experimental C. albicans infection induced epithelial hGAPDH (2-32) expression. Addition of the peptide significantly reduced the tissue damage as compared with untreated experimental infection. Secreted aspartic proteinase (Sap) activity of C. albicans was inhibited by the fragment at higher concentrations, with a median effective dose of 160 mg l(-1) (50 µM) for Sap1p and 200 mg l(-1) (63 µM) for Sap2p, whereas Sap3 was not inhibited at all. Interestingly, hGAPDH (2-32) induced significant epithelial IL-8 and GM-CSF secretion and stimulated Toll-like receptor 4 expression at low concentrations independently of the presence of C. albicans, without any toxic mucosal effects. In the future, the combination of different antifungal strategies, e.g., a conventional fungicidal with immunomodulatory effects and the inhibition of fungal virulence factors, might be a promising treatment option.


Subject(s)
Antifungal Agents/pharmacology , Epithelium/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Immunomodulation/drug effects , Peptide Fragments/pharmacology , Antifungal Agents/isolation & purification , Apoptosis/drug effects , Aspartic Acid Proteases/antagonists & inhibitors , Aspartic Acid Proteases/metabolism , Candida albicans/drug effects , Candida albicans/metabolism , Candidiasis/drug therapy , Candidiasis/immunology , Cell Line , Epithelium/immunology , Epithelium/microbiology , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-8/biosynthesis , Interleukin-8/immunology , Mouth Mucosa/drug effects , Mouth Mucosa/immunology , Mouth Mucosa/microbiology , Peptide Fragments/isolation & purification , Placenta/enzymology , Pregnancy , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/immunology
17.
PLoS One ; 7(11): e50518, 2012.
Article in English | MEDLINE | ID: mdl-23226301

ABSTRACT

C. albicans is one of the most common fungal pathogen of humans, causing local and superficial mucosal infections in immunocompromised individuals. Given that the key structure mediating host-C. albicans interactions is the fungal cell wall, we aimed to identify features of the cell wall inducing epithelial responses and be associated with fungal pathogenesis. We demonstrate here the importance of cell wall protein glycosylation in epithelial immune activation with a predominant role for the highly branched N-glycosylation residues. Moreover, these glycan moieties induce growth arrest and apoptosis of epithelial cells. Using an in vitro model of oral candidosis we demonstrate, that apoptosis induction by C. albicans wild-type occurs in early stage of infection and strongly depends on intact cell wall protein glycosylation. These novel findings demonstrate that glycosylation of the C. albicans cell wall proteins appears essential for modulation of epithelial immunity and apoptosis induction, both of which may promote fungal pathogenesis in vivo.


Subject(s)
Apoptosis/immunology , Candida albicans/cytology , Cell Wall/metabolism , Epithelial Cells/cytology , Epithelial Cells/microbiology , Fungal Proteins/metabolism , Immunity, Innate , Animals , Candida albicans/physiology , Cell Cycle Checkpoints/immunology , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Epithelial Cells/immunology , Fungal Proteins/immunology , Gene Expression Regulation/immunology , Glycosylation , Humans , Mice , Mice, Inbred C57BL , Polysaccharides/immunology , Polysaccharides/metabolism , Time Factors , Toll-Like Receptor 4/metabolism
18.
Methods Mol Biol ; 845: 333-44, 2012.
Article in English | MEDLINE | ID: mdl-22328385

ABSTRACT

In this protocol, we describe the application of commercially available three-dimensional organotypic tissues of human oral mucosa to study the interaction between Candida albicans and epithelial cells. Infection experiments show high reproducibility and can be used to analyse directly pathogen/epithelial cell interactions. However, the system is also very flexible. Using histological, biochemical, immunological, and molecular methods, it is possible to analyse several stages of infection by C. albicans wild type or mutant strains and demonstrate the consequence of disrupting genes encoding putative virulence factors required for host cell invasion and immune defence induction. This model provides information about host and pathogen protein and gene expression during direct interactions with each other. It can additionally be supplemented with other host factors, such as immune cells, saliva, and probiotic bacteria, which are relevant for host immune defence in the oral cavity.


Subject(s)
Candida albicans/immunology , Epithelial Cells/immunology , Models, Immunological , Mouth Mucosa/immunology , Humans , Mouth Mucosa/cytology
19.
Mycoses ; 55(1): 17-26, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21672043

ABSTRACT

The extensive use of immunosuppressive therapies in recent years has increased the number of patients prone to or actually suffering from localised candidosis. As Candida species gain increasing resistance towards common antifungal drugs, new strategies are needed to prevent and treat infections caused by these pathogens. Probiotic bacteria have been in vogue in the past two decades. More and more dairy products containing such organisms offer promising potential beneficial effects on human health and well-being. Because of the ability of probiotic bacteria to inhibit the growth of pathogens and to modulate human immune responses, these bacteria could provide new possibilities in antifungal therapy. We summarise the recent findings concerning the usefulness of probiotic treatment in localised candidosis, as well as discussing possible risks of probiotic treatment and highlighting the molecular mechanisms that are believed to contribute to probiotic effects.


Subject(s)
Candidiasis/prevention & control , Candidiasis/therapy , Probiotics/therapeutic use , Candida/pathogenicity , Candidiasis/microbiology , Clinical Trials as Topic , Humans , Intestines/microbiology , Microbial Interactions , Probiotics/administration & dosage , Probiotics/adverse effects , Risk Factors
20.
Int J Med Microbiol ; 301(5): 431-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21550846

ABSTRACT

The mucosal epithelium is of central importance in host defence and immune surveillance, as it is the primary cell layer that initially encounters environmental microorganisms. Induction of antifungal innate immune responses depends on recognition of fungal components by host pattern recognition receptors. Members of the Toll-like receptor family have emerged as key sensors that recognize fungal pathogens and trigger defence responses. During oral infection with the fungal pathogen Candida albicans, a large number of cytokines is secreted by oral epithelial cells, which in turn activate myeloid cells in the submucosal layers to clear the invading pathogen. Recent data provide novel insights into the complex molecular mechanisms of innate immune responses initiated by cooperation between epithelial cells and neutrophils. In this review, we discuss the role of epithelial TLRs and how the immunological crosstalk between C. albicans-infected oral epithelium and neutrophils protects the mucosal surface from fungal invasion and cell injury.


Subject(s)
Candida albicans/immunology , Candidiasis, Oral/immunology , Immunity, Mucosal , Cytokines/immunology , Cytokines/metabolism , Epithelial Cells/immunology , Humans , Neutrophils/immunology , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...