Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 169
Filter
1.
Methods Mol Biol ; 2775: 127-137, 2024.
Article in English | MEDLINE | ID: mdl-38758315

ABSTRACT

Proteomic profiling provides in-depth information about the regulation of diverse biological processes, activation of and communication across signaling networks, and alterations to protein production, modifications, and interactions. For infectious disease research, mass spectrometry-based proteomics enables detection of host defenses against infection and mechanisms used by the pathogen to evade such responses. In this chapter, we outline protein extraction from organs, tissues, and fluids collected following intranasal inoculation of a murine model with the human fungal pathogen Cryptococcus neoformans. We describe sample preparation, followed by purification, processing on the mass spectrometer, and a robust bioinformatics analysis. The information gleaned from proteomic profiling of fungal infections supports the detection of novel biomarkers for diagnostic and prognostic purposes.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Disease Models, Animal , Proteomics , Animals , Cryptococcus neoformans/metabolism , Cryptococcus neoformans/pathogenicity , Mice , Cryptococcosis/microbiology , Cryptococcosis/metabolism , Proteomics/methods , Computational Biology/methods , Proteome/metabolism , Biomarkers/metabolism , Mass Spectrometry/methods
2.
Methods Mol Biol ; 2775: 195-209, 2024.
Article in English | MEDLINE | ID: mdl-38758319

ABSTRACT

Cryptococcus neoformans, the predominant etiological agent of cryptococcosis, is an encapsulated fungal pathogen found ubiquitously in the environment that causes pneumonia and life-threatening infections of the central nervous system. Following inhalation of yeasts or desiccated basidiospores into the lung alveoli, resident pulmonary phagocytic cells aid in the identification and eradication of Cryptococcus yeast through their arsenal of pattern recognition receptors (PRRs). PRRs recognize conserved pathogen-associated molecular patterns (PAMPs), such as branched mannans, ß-glucans, and chitins that are the major components of the fungal cell wall. However, the key receptors/ligand interactions required for cryptococcal recognition and eventual fungal clearance have yet to be elucidated. Here we present an imaging flow cytometer (IFC) method that offers a novel quantitative cellular imaging and population statistics tool to accurately measure phagocytosis of fungal cells. It has the capacity to measure two distinct steps of phagocytosis: association/attachment and internalization in a high-throughput and quantitative manner that is difficult to achieve with other technologies. Results from these IFC studies allow for the potential to identify PRRs required for recognition, uptake, and subsequent activation of cytokine production, as well as other effector cell responses required for fungal clearance.


Subject(s)
Cryptococcus neoformans , Flow Cytometry , Phagocytosis , Flow Cytometry/methods , Cryptococcus neoformans/metabolism , Animals , Mice , Phagocytes/metabolism , Phagocytes/microbiology , Cryptococcosis/microbiology , Cryptococcosis/metabolism , Cryptococcosis/immunology , Cryptococcus/metabolism , Humans , Image Cytometry/methods , Receptors, Pattern Recognition/metabolism
3.
Cytokine ; 173: 156441, 2024 01.
Article in English | MEDLINE | ID: mdl-37995394

ABSTRACT

Macrophages have recently been discovered to assume a significant role in the progression of cryptococcosis. However, the potential involvement of macrophage-derived exosomes in the pathogenesis of cryptococcosis remains uncertain. In this study, we investigated the changes of microRNAs in macrophage exosomes (exo-miRNAs) in cryptococcal infections and the role of markedly altered exo-miRNAs in the modulation of Human Umbilical Vein Endothelial Cells (HUVEC) permeability and ROS accumulation and pyroptosis in Human Bronchial Epithelioid Cells (BEAS-2B). Techniques such as microarray analysis and real-time quantitative PCR were used to detect different exo-miRNAs and to screen for the most highly expressed exo-miRNAs. Then its mimics were transfected into HUVEC to study its effect on the monolayer permeability of HUVEC. Finally, the relationship between this exo-miRNAs and the ROS accumulation and pyroptosis was verified by bioinformatics analysis. The results showed that five exo-miRNAs were overexpressed and two exo-miRNAs were reduced, among which, exo-miR-4449 was expressed at the highest level. Exo-miR-4449 could be internalized by HUVEC and enhanced its monolayer permeability. Moreover, exo-miR-4449 was found to promote ROS accumulation and pyroptosis in BEAS-2B through HIC1 pathway. Thus, exo-miR-4449 plays an important role in the pathogenesis of cryptococcosis and holds promise as a significant biomarker for treatment.


Subject(s)
Cryptococcosis , Cryptococcus , MicroRNAs , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Pyroptosis/genetics , Cryptococcus/metabolism , Reactive Oxygen Species/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Macrophages/metabolism , Cryptococcosis/metabolism , Cryptococcosis/pathology , Kruppel-Like Transcription Factors
4.
Front Cell Infect Microbiol ; 13: 1331429, 2023.
Article in English | MEDLINE | ID: mdl-38149006

ABSTRACT

Cryptococcus neoformans can invade the central nervous system by crossing the blood-brain barrier via a transcellular mechanism that relies on multiple host factors. In this narrative, we review the evidence that a direct interplay between C. neoformans and brain endothelial cells forms the basis for invasion and transmigration across the brain endothelium. Adherence and internalization of C. neoformans is dependent on transmembrane proteins, including a hyaluronic acid receptor and an ephrin receptor tyrosine kinase. We consider the role of EphA2 in facilitating the invasion of the central nervous system by C. neoformans and highlight experimental evidence supporting macropinocytosis as a potential mechanism of internalization and transcytosis. How macropinocytosis might be conclusively demonstrated in the context of C. neoformans is also discussed.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Cryptococcus neoformans/metabolism , Endothelial Cells/metabolism , Cryptococcosis/metabolism , Brain/metabolism , Blood-Brain Barrier
5.
Immunity ; 55(9): 1591-1593, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36103858

ABSTRACT

The invasive fungal pathogen Cryptococcus neoformans promotes type 2 immunity to escape host defenses by unknown mechanisms. In a recent issue of Nature, Dang and colleagues identify a secreted fungal protein that triggers TLR4 signaling and supports a type 2 permissive environment and C. neoformans growth.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Cryptococcosis/metabolism , Cryptococcosis/microbiology , Cryptococcus neoformans/metabolism , Fungal Proteins/metabolism , Toll-Like Receptor 4/metabolism , Virulence
6.
J Biol Chem ; 298(10): 102453, 2022 10.
Article in English | MEDLINE | ID: mdl-36063996

ABSTRACT

The fungal pathogen Cryptococcus neoformans is a leading cause of meningoencephalitis in the immunocompromised. As current antifungal treatments are toxic to the host, costly, limited in their efficacy, and associated with drug resistance, there is an urgent need to identify vulnerabilities in fungal physiology to accelerate antifungal discovery efforts. Rational drug design was pioneered in de novo purine biosynthesis as the end products of the pathway, ATP and GTP, are essential for replication, transcription, and energy metabolism, and the same rationale applies when considering the pathway as an antifungal target. Here, we describe the identification and characterization of C. neoformans 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) transformylase/5'-inosine monophosphate cyclohydrolase (ATIC), a bifunctional enzyme that catalyzes the final two enzymatic steps in the formation of the first purine base inosine monophosphate. We demonstrate that mutants lacking the ATIC-encoding ADE16 gene are adenine and histidine auxotrophs that are unable to establish an infection in a murine model of virulence. In addition, our assays employing recombinantly expressed and purified C. neoformans ATIC enzyme revealed Km values for its substrates AICAR and 5-formyl-AICAR are 8-fold and 20-fold higher, respectively, than in the human ortholog. Subsequently, we performed crystallographic studies that enabled the determination of the first fungal ATIC protein structure, revealing a key serine-to-tyrosine substitution in the active site, which has the potential to assist the design of fungus-specific inhibitors. Overall, our results validate ATIC as a promising antifungal drug target.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Hydroxymethyl and Formyl Transferases , Phosphoribosylaminoimidazolecarboxamide Formyltransferase , Animals , Humans , Mice , Antifungal Agents , Cryptococcus neoformans/enzymology , Cryptococcus neoformans/genetics , Drug Discovery , Inosine Monophosphate , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/chemistry , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/genetics , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Purines , Cryptococcosis/metabolism
7.
Front Immunol ; 13: 931194, 2022.
Article in English | MEDLINE | ID: mdl-35967332

ABSTRACT

Lungs balance threat from primary viral infection, secondary infection, and inflammatory damage. Severe pulmonary inflammation induces vascular permeability, edema, and organ dysfunction. We previously demonstrated that poly(I:C) (pICLC) induced type 1 interferon (t1IFN) protected mice from Cryptococcus gattii (Cg) via local iron restriction. Here we show pICLC increased serum protein and intravenously injected FITC-dextran in the lung airspace suggesting pICLC induces vascular permeability. Interestingly, pICLC induced a pro-inflammatory signature with significant expression of IL-1 and IL-6 which depended on MDA5 and t1IFN. Vascular permeability depended on MDA5, t1IFN, IL-1, and IL-6. T1IFN also induced MDA5 and other MDA5 signaling components suggesting that positive feedback contributes to t1IFN dependent expression of the pro-inflammatory signature. Vascular permeability, induced by pICLC or another compound, inhibited Cg by limiting iron. These data suggest that pICLC induces t1IFN which potentiates pICLC-MDA5 signaling increasing IL-1 and IL-6 resulting in leakage of antimicrobial serum factors into lung airspace. Thus, induced vascular permeability may act as an innate defense mechanism against opportunistic fungal infection, such as cryptococcosis, and may be exploited as a host-directed therapeutic target.


Subject(s)
Cryptococcosis , Cryptococcus gattii , Interferon Type I , Opportunistic Infections , Animals , Capillary Permeability , Cryptococcosis/metabolism , Interferon Type I/metabolism , Interleukin-1/metabolism , Interleukin-6/metabolism , Iron/metabolism , Lung/metabolism , Mice , Opportunistic Infections/metabolism
8.
Microbes Infect ; 24(8): 105016, 2022.
Article in English | MEDLINE | ID: mdl-35640861

ABSTRACT

It is becoming increasingly obvious that glycophosphatidylinositol (GPI)-anchored proteins (GAPs) play a prominent role in fungi, a full understanding of GAPs is however lacking especially for the human opportunistic fungus Cryptococcus neoformans. Using online GPI prediction tools, GAPs were identified and subsequently a mutant library for these GAP-encoding genes was developed and a publicly available knock out (KO) mutant library was used. In total, 41 overexpression and 34 KO mutants, representing 47 unique genes, were analyzed. From the analysis of the two libraries, two main gene candidates, a mannoprotein 88 (MP88) (CNAG_00776) and an uncharacterized protein (CNAG_00137) were further investigated by constructing additional independent mutant strains. The CNAG_00776 mutant showed an impaired growth upon plasma membrane stress and significant decreased phagocytosis. The CNAG_00137 mutant showed impaired growth during cell wall stress or increased temperature and significant decreased phagocytosis. By performing a large genetic screen of GAPs in the genome of the human fungal pathogen C. neoformans, we identified two candidate GAP genes involved in C. neoformans/host interaction and stress response. Further research into these two genes could potentially result in new targets for antfungals, treatment strategies or vaccines to manage C. neoformans disease.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Humans , Glycosylphosphatidylinositols/metabolism , Cryptococcus neoformans/genetics , Cryptococcus neoformans/metabolism , Cell Wall/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Cell Membrane/metabolism , Cryptococcosis/metabolism
9.
Inflammation ; 45(3): 1269-1280, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35015189

ABSTRACT

Cryptococcosis (caused, for example, by Cryptococcus neoformans) and allergic asthma (caused, for example, by Dermatophagoides pteronyssinus) target the respiratory tract (the lung and bronchial epithelium). C. neoformans and D. pteronyssinus can coexist in the same indoor environment, and exposure to both can cause alterations in the local airway inflammatory milieu and exacerbation of airway inflammatory diseases. Here, we evaluated the effects of the association between C. neoformans and D. pteronyssinus in the modulation of airway inflammatory responses in an in vitro experimental model using human bronchial epithelial cells. BEAS-2B cells were cultivated and stimulated with D. pteronyssinus (10 µg/mL) and/or C. neoformans (MOI 100) for 24 h. No cytotoxic effect was observed in cells stimulated by C. neoformans and/or D. pteronyssinus. The production of IL-8, IL-6, and/or CCL2, but not IL-10, as well as the activation of NF-kB, STAT3, STAT6, and/or ERK1/2 were increased in cells stimulated by C. neoformans or D. pteronyssinus compared to controls. C. neoformans in association with D. pteronyssinus inhibited the CCL2­ERK1/2 signaling pathway in cells treated with both pathogens compared to cells stimulated by D. pteronyssinus alone. In addition, their association induced an additive effect on the IL-6/STAT3 signaling pathway in cells compared to cells stimulated with D. pteronyssinus or C. neoformans only. D. pteronyssinus increased the internalization and growth of C. neoformans in BEAS-2B cells. D. pteronyssinus in association with C. neoformans promoted pro- and anti-inflammatory responses, which can modulate cryptococcal infection and asthmaticus status.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Animals , Anti-Inflammatory Agents/pharmacology , Bronchi , Chemokine CCL2/metabolism , Cryptococcosis/metabolism , Cryptococcus neoformans/metabolism , Dermatophagoides pteronyssinus/metabolism , Down-Regulation , Epithelial Cells/metabolism , Humans , Interleukin-6/metabolism , MAP Kinase Signaling System , STAT3 Transcription Factor/metabolism
10.
Immunology ; 165(2): 143-157, 2022 02.
Article in English | MEDLINE | ID: mdl-34716931

ABSTRACT

Cryptococcus neoformans is an opportunistic fungal pathogen that can cause lethal cryptococcal meningitis in immunocompromised individuals such as those with HIV/AIDS. In addition, cryptococcal infections occasionally arise in immunocompetent individuals or those with previously undiagnosed immunodeficiencies. The course of cryptococcosis is highly variable in both patient groups, and there is rapidly growing evidence that genetic polymorphisms may have a significant impact on the trajectory of disease. Here, we review what is currently known about the nature of these polymorphisms and their impact on host response to C. neoformans infection. Thus far, polymorphisms in Fc gamma receptors, mannose-binding lectin, Dectin-2, Toll-like receptors and macrophage colony-stimulating factor have been associated with susceptibility to cryptococcal disease. Notably, however, in some cases the impact of these polymorphisms depends on the genetic background of the population; for example, the FCGR3A 158 F/V polymorphism was associated with an increased risk of cryptococcal disease in both HIV-positive and HIV-negative white populations, but not in Han Chinese patients. In most cases, the precise mechanism by which the identified polymorphisms influence disease progression remains unclear, although impaired fungal recognition and phagocytosis by innate immune cells appears to play a major role. Finally, we highlight outstanding questions in the field and emphasize the need for future research to include more diverse populations in their genetic association studies.


Subject(s)
Cryptococcosis/etiology , Cryptococcus neoformans/immunology , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Immunocompromised Host , Immunogenetic Phenomena , Polymorphism, Genetic , Adaptive Immunity , Animals , Biomarkers , Cryptococcosis/metabolism , Gene Expression Regulation , Genetic Variation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Signal Transduction
11.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830272

ABSTRACT

Fungal pathogens cause an array of diseases by targeting both immunocompromised and immunocompetent hosts. Fungi overcome our current arsenal of antifungals through the emergence and evolution of resistance. In particular, the human fungal pathogen, Cryptococcus neoformans is found ubiquitously within the environment and causes severe disease in immunocompromised individuals around the globe with limited treatment options available. To uncover fundamental knowledge about this fungal pathogen, as well as investigate new detection and treatment strategies, mass spectrometry-based proteomics provides a plethora of tools and applications, as well as bioinformatics platforms. In this review, we highlight proteomics approaches within the laboratory to investigate changes in the cellular proteome, secretome, and extracellular vesicles. We also explore regulation by post-translational modifications and the impact of protein-protein interactions. Further, we present the development and comprehensive assessment of murine models of cryptococcal infection, which provide valuable tools to define the dynamic relationship between the host and pathogen during disease. Finally, we explore recent quantitative proteomics studies that begin to extrapolate the findings from the bench to the clinic for improved methods of fungal detection and monitoring. Such studies support a framework for personalized medical approaches to eradicate diseases caused by C. neoformans.


Subject(s)
Cryptococcosis/metabolism , Cryptococcus neoformans/metabolism , Fungal Proteins/metabolism , Proteome/metabolism , Proteomics/methods , Animals , Antifungal Agents/therapeutic use , Cryptococcosis/drug therapy , Cryptococcosis/microbiology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Disease Models, Animal , Drug Resistance, Fungal/genetics , Extracellular Vesicles/metabolism , Fungal Proteins/genetics , Host-Parasite Interactions/genetics , Humans , Mice , Precision Medicine/methods , Protein Interaction Maps/genetics , Protein Processing, Post-Translational/genetics , Proteome/genetics , Secretome/metabolism , Transcriptome , Treatment Outcome , Virulence Factors/metabolism
12.
Front Immunol ; 12: 722500, 2021.
Article in English | MEDLINE | ID: mdl-34650554

ABSTRACT

With over 220,000 cases and 180,000 deaths annually, Cryptococcus neoformans is the most common cause of fungal meningitis and a leading cause of death in HIV/AIDS patients in Sub-Saharan Africa. Either C. neoformans can be killed by innate airway phagocytes, or it can survive intracellularly. Pulmonary murine macrophage and dendritic cell (DC) subsets have been identified in the naïve lung, and we hypothesize that each subset has different interactions with C. neoformans. For these studies, we purified murine pulmonary macrophage and DC subsets from naïve mice - alveolar macrophages, Ly6c- and Ly6c+ monocyte-like macrophages, interstitial macrophages, CD11b+ and CD103+ DCs. With each subset, we examined cryptococcal association (binding/internalization), fungicidal activity, intracellular fungal morphology, cytokine secretion and transcriptional profiling in an ex vivo model using these pulmonary phagocyte subsets. Results showed that all subsets associate with C. neoformans, but only female Ly6c- monocyte-like macrophages significantly inhibited growth, while male CD11b+ DCs significantly enhanced fungal growth. In addition, cytokine analysis revealed that some subsets from female mice produced increased amounts of cytokines compared to their counterparts in male mice following exposure to C. neoformans. In addition, although cells were analyzed ex vivo without the influence of the lung microenviroment, we did not find evidence of phagocyte polarization following incubation with C. neoformans. Imaging flow cytometry showed differing ratios of cryptococcal morphologies, c-shaped or budding, depending on phagocyte subset. RNA sequencing analysis revealed the up- and down-regulation of many genes, from immunological pathways (including differential regulation of MHC class I in the antigen processing pathway and the cell adhesion pathway) and pathways relating to relating to metabolic activity (genes in the Cytochrome P450 family, genes related to actin binding, calcium voltage channels, serine proteases, and phospholipases). Future studies gaining a more in-depth understanding on the functionality of individual genes and pathways specific to permissive and non-permissive pulmonary phagocytes will allow identification of key targets when developing therapeutic strategies to prevent cryptococcal meningitis.


Subject(s)
Cryptococcosis/etiology , Cryptococcus neoformans/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Phagocytes/immunology , Phagocytes/metabolism , Transcription, Genetic , Animals , Cell Plasticity , Cryptococcosis/metabolism , Cryptococcosis/pathology , Cytokines/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Female , Immunity, Innate , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Metabolic Networks and Pathways , Mice , Prognosis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
13.
Sci Rep ; 11(1): 18397, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526536

ABSTRACT

Cryptococcosis is an opportunistic disease caused by the fungus Cryptococcus neoformans and Cryptococcus gattii. It starts as a pulmonary infection that can spread to other organs, such as the brain, leading to the most serious occurrence of the disease, meningoencephalitis. The humoral response has already been described in limiting the progression of cryptococcosis where the B-1 cell seems to be responsible for producing natural IgM antibodies, crucial for combating fungal infections. The role of the B-1 cell in C. neoformans infection has been initially described, however the role of the humoral response of B-1 cells has not yet been evaluated during C. gattii infections. In the present study we tried to unravel this issue using XID mice, a murine model deficient in the Btk protein which compromises the development of B-1 lymphocytes. We use the XID mice compared to BALB/c mice that are sufficient for the B-1 population during C. gattii infection. Our model of chronic lung infection revealed that XID mice, unlike the sufficient group of B-1, had early mortality with significant weight loss, in addition to reduced levels of IgM and IgG specific to GXM isolated from the capsule of C. neoformans. In addition to this, we observed an increased fungal load in the blood and in the brain. We described an increase in the capsular size of C. gattii and the predominant presence of cytokines with a Th2 profile was also observed in these animals. Thus, the present study strongly points to a higher susceptibility of the XID mouse to C. gattii, which suggests that the presence of B-1 cells and anti-GXM antibodies is fundamental during the control of infection by C. gattii.


Subject(s)
Cryptococcosis/etiology , Cryptococcus gattii , Disease Susceptibility/immunology , Immunocompromised Host , X-Linked Combined Immunodeficiency Diseases/complications , Animals , Biomarkers , Colony Count, Microbial , Cryptococcosis/metabolism , Cryptococcus gattii/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Mice , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology
14.
Infect Immun ; 89(10): e0033021, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34251289

ABSTRACT

The cell walls and capsules of Cryptococcus neoformans, a yeast-type fungal pathogen, are rich in polysaccharides. Dectin-2 is a C-type lectin receptor (CLR) that recognizes high-mannose polysaccharides. Previously, we demonstrated that Dectin-2 is involved in cytokine production by bone marrow-derived dendritic cells (BM-DCs) in response to stimulation with C. neoformans. In the present study, we analyzed the role of Dectin-2 in the phagocytosis of C. neoformans by BM-DCs. The engulfment of this fungus by BM-DCs was significantly decreased in mice lacking Dectin-2 (Dectin-2 knockout [Dectin-2KO]) or caspase recruitment domain-containing protein 9 (CARD9KO), a common adapter molecule that delivers signals triggered by CLRs, compared to wild-type (WT) mice. Phagocytosis was likewise inhibited, to a similar degree, by the inhibition of Syk, a signaling molecule involved in CLR-triggered activation. A PI3K inhibitor, in contrast, completely abrogated the phagocytosis of C. neoformans. Actin polymerization, i.e., conformational changes in cytoskeletons detected at sites of contact with C. neoformans, was also decreased in BM-DCs of Dectin-2KO and CARD9KO mice. Finally, the engulfment of C. neoformans by macrophages was significantly decreased in the lungs of Dectin-2KO mice compared to WT mice. These results suggest that Dectin-2 may play an important role in the actin polymerization and phagocytosis of C. neoformans by DCs, possibly through signaling via CARD9 and a signaling pathway mediated by Syk and PI3K.


Subject(s)
Cryptococcosis/microbiology , Cryptococcus neoformans/pathogenicity , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Phagocytosis/physiology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/microbiology , CARD Signaling Adaptor Proteins/metabolism , Cryptococcosis/metabolism , Cytokines/metabolism , Dendritic Cells/microbiology , Female , Lung/metabolism , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism
15.
Infect Immun ; 89(10): e0012821, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34228504

ABSTRACT

Microbial penetration of the blood-brain barrier, a prerequisite for the development of central nervous system (CNS) infection, involves microbial invasion, intracellular traversal, and exocytosis. Microbial invasion of the blood-brain barrier has been investigated, but the molecular basis for microbial traversal and exit from the blood-brain barrier remains unknown. We performed transcriptome analysis of human brain microvascular endothelial cells (HBMEC) infected with Escherichia coli and Cryptococcus neoformans, representative bacterial and fungal pathogens common in CNS infections. Among the targets upregulated in response to E. coli and C. neoformans infection, PDLIM2 was knocked down by small hairpin RNA (shRNA) in HBMEC for further investigation. We demonstrated that Pdlim2 specifically regulated microbial traversal and exit from HBMEC by assessing microbial invasion, transcytosis, intracellular multiplication, and egression. Additionally, the defective exocytosis of internalized E. coli cells from the PDLIM2 shRNA knockdown cells was restored by treatment with a calcium ionophore (ionomycin). Moreover, we performed proximity-dependent biotin labeling with the biotin ligase BioID2 and identified 210 potential Pdlim2 interactors. Among the nine Pdlim2 interactors enriched in response to both E. coli and C. neoformans infection, we selected MPRIP and showed that HBMEC with knockdown of MPRIP mimicked the phenotype of PDLIM2 knockdown cells. These results suggest that the CNS-infecting microbes hijack Pdlim2 and Mprip for intracellular traversal and exocytosis in the blood-brain barrier.


Subject(s)
Blood-Brain Barrier/immunology , Central Nervous System Infections/immunology , Cryptococcosis/immunology , Cryptococcus neoformans/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Exocytosis/immunology , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , Biological Transport/immunology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/microbiology , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/microbiology , Central Nervous System Infections/metabolism , Central Nervous System Infections/microbiology , Cryptococcosis/metabolism , Cryptococcosis/microbiology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Humans , LIM Domain Proteins/immunology , Microfilament Proteins/immunology , Phosphorylation/immunology
16.
Molecules ; 26(11)2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34200464

ABSTRACT

Due to lifespan extension and changes in global climate, the increase in mycoses caused by primary and opportunistic fungal pathogens is now a global concern. Despite increasing attention, limited options are available for the treatment of systematic and invasive mycoses, owing to the evolutionary similarity between humans and fungi. Although plants produce a diversity of chemicals to protect themselves from pathogens, the molecular targets and modes of action of these plant-derived chemicals have not been well characterized. Using a reverse genetics approach, the present study revealed that thymol, a monoterpene alcohol from Thymus vulgaris L., (Lamiaceae), exhibits antifungal activity against Cryptococcus neoformans by regulating multiple signaling pathways including calcineurin, unfolded protein response, and HOG (high-osmolarity glycerol) MAPK (mitogen-activated protein kinase) pathways. Thymol treatment reduced the intracellular concentration of Ca2+ by controlling the expression levels of calcium transporter genes in a calcineurin-dependent manner. We demonstrated that thymol decreased N-glycosylation by regulating the expression levels of genes involved in glycan-mediated post-translational modifications. Furthermore, thymol treatment reduced endogenous ergosterol content by decreasing the expression of ergosterol biosynthesis genes in a HOG MAPK pathway-dependent manner. Collectively, this study sheds light on the antifungal mechanisms of thymol against C. neoformans.


Subject(s)
Antifungal Agents/pharmacology , Cryptococcosis/drug therapy , Cryptococcus neoformans/drug effects , Thymol/pharmacology , Calcineurin/metabolism , Cryptococcosis/metabolism , Cryptococcus neoformans/metabolism , Ergosterol/pharmacology , Fungal Proteins/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Monoterpenes/pharmacology , Signal Transduction/drug effects , Thymus Plant/chemistry
17.
Front Immunol ; 12: 670574, 2021.
Article in English | MEDLINE | ID: mdl-33995406

ABSTRACT

Neutrophils are critical as the first-line defense against fungal pathogens. Yet, previous studies indicate that neutrophil function is complex during Cryptococcus neoformans (Cn) infection. To better understand the role of neutrophils in acute pulmonary cryptococcosis, we analyzed neutrophil heterogeneity by single-cell transcriptional analysis of immune cells in the lung of Cn-infected mice from a published dataset. We identified neutrophils by reference-based annotation and identified two distinct neutrophil subsets generated during acute Cn infection: A subset with an oxidative stress signature (Ox-PMN) and another with enhanced cytokine gene expression (Cyt-PMN). Based on gene regulatory network and ligand-receptor analysis, we hypothesize that Ox-PMNs interact with the fungus and generate ROS, while Cyt-PMNs are longer-lived neutrophils that indirectly respond to Cn-derived ligands and cytokines to modulate cell-cell communication with dendritic cells and alveolar macrophages. Based on the data, we hypothesized that, during in vivo fungal infection, there is a division of labor in which each activated neutrophil becomes either Ox-PMN or Cyt-PMN.


Subject(s)
Cryptococcosis/immunology , Lung Diseases, Fungal/immunology , Neutrophil Activation/immunology , Neutrophils/immunology , Animals , Cryptococcosis/metabolism , Cryptococcus neoformans/immunology , Cytokines/immunology , Cytokines/metabolism , Lung Diseases, Fungal/metabolism , Mice , Oxidative Stress/immunology , Reactive Oxygen Species/immunology , Single-Cell Analysis
18.
Eur J Med Chem ; 221: 113524, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-33992927

ABSTRACT

Invasive fungal infections remain a challenge due to lack of effective antifungal agents and serious drug resistance. Discovery of antifungal agents with novel antifungal mechanism is important and urgent. Previously, we designed the first CYP51/HDAC dual inhibitors with potent activity against resistant Candida albicans infections. To better understand the antifungal spectrum and synergistic mechanism, herein new CYP51/HDAC dual inhibitors were designed which showed potent in vitro and in vivo antifungal activity against C. neoformans and C. tropicalis infections. Antifungal mechanism studies revealed that the CYP51/HDAC dual inhibitors acted by inhibiting various virulence factors of C. tropicalis and C. neoformans and down-regulating resistance-associated genes. This study highlights the potential of CYP51/HDAC dual inhibitors as a promising strategy for the discovery of novel broad-spectrum antifungal agents.


Subject(s)
14-alpha Demethylase Inhibitors/pharmacology , Antifungal Agents/pharmacology , Candidiasis, Cutaneous/drug therapy , Cryptococcosis/drug therapy , Histone Deacetylase Inhibitors/pharmacology , 14-alpha Demethylase Inhibitors/chemical synthesis , 14-alpha Demethylase Inhibitors/chemistry , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Candida tropicalis/drug effects , Candida tropicalis/metabolism , Candidiasis, Cutaneous/metabolism , Cryptococcosis/metabolism , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/metabolism , Cytochrome P450 Family 51/antagonists & inhibitors , Cytochrome P450 Family 51/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Fungal/drug effects , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
19.
Front Immunol ; 12: 654574, 2021.
Article in English | MEDLINE | ID: mdl-33796117

ABSTRACT

The small molecule (molecular mass <900 Daltons) composition of extracellular vesicles (EVs) produced by the pathogenic fungus Cryptococcus gattii is unknown, which limits the understanding of the functions of cryptococcal EVs. In this study, we analyzed the composition of small molecules in samples obtained from solid cultures of C. gattii by a combination of chromatographic and spectrometric approaches, and untargeted metabolomics. This analysis revealed previously unknown components of EVs, including small peptides with known biological functions in other models. The peptides found in C. gattii EVs had their chemical structure validated by chemical approaches and comparison with authentic standards, and their functions tested in a Galleria mellonella model of cryptococcal infection. One of the vesicular peptides (isoleucine-proline-isoleucine, Ile-Pro-Ile) improved the survival of G. mellonella lethally infected with C. gattii or C. neoformans. These results indicate that small molecules exported in EVs are biologically active in Cryptococcus. Our study is the first to characterize a fungal EV molecule inducing protection, pointing to an immunological potential of extracellular peptides produced by C. gattii.


Subject(s)
Cryptococcosis/metabolism , Cryptococcus gattii/physiology , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Host-Pathogen Interactions , Invertebrates , Animals , Computational Biology/methods , Cryptococcosis/microbiology , Extracellular Vesicles/ultrastructure , Metabolomics/methods , Molecular Structure , Peptides
20.
mBio ; 11(6)2020 11 24.
Article in English | MEDLINE | ID: mdl-33234684

ABSTRACT

Cryptococcus gattii is a major cause of life-threatening mycosis in immunocompetent individuals and responsible for the ongoing epidemic outbreak of cryptococcosis in the Pacific Northwest of North America. This deadly fungus is known to evade important host immune responses, including dendritic cell (DC) maturation and concomitant T cell immunity, via immune evasion mechanisms that remain unclear. Here, we demonstrate that primary human DCs phagocytose C. gattii but the maturation of phagosomes to phagolysosomes was blocked as a result of sustained filamentous actin (F-actin) that entrapped and concealed the phagosomes from recognition. Superresolution structured illumination microscopy (SR-SIM) revealed that the persistent phagosomal F-actin formed a cage-like structure that sterically hindered and functionally blocked the fusion of lysosomes. Blocking lysosome fusion was sufficient to inhibit phagosomal acidification and subsequent intracellular fungal killing by DCs. Retention of phagosomal F-actin by C. gattii also caused DC immunoparalysis. Disrupting the retained F-actin cage with cytochalasin D not only restored DC phagosomal maturation but also promoted DC costimulatory maturation and robust T cell activation and proliferation. Collectively, these results reveal a unique mechanism of DC immune evasion that enhances intracellular fungal pathogenicity and may explain suppressed cell-mediated immunity.IMPORTANCECryptococcus yeast species typically display characteristics of opportunistic pathogens, with the exception of C. gattii, which can cause life-threatening respiratory and disseminated brain infections in otherwise healthy people. The pathogenesis of C. gattii is not well understood, but an important characteristic is that C. gattii is capable of evading host cell-mediated immune defenses initiated by DCs. Here, we report that when virulent C. gattii becomes ingested by a DC, the intracellular compartment containing the fungi is covered by a persistent protein cage structure consisting of F-actin. This F-actin cage acts as a barrier to prevent interaction with other intracellular compartments, and as a result, the DC fails to kill the fungi and activate important cell-mediated immune responses. We propose that this unique immune evasion mechanism permits C. gattii to remain unchallenged within host cells, leading to persistent infection.


Subject(s)
Actins/metabolism , Cryptococcus gattii/immunology , Cryptococcus gattii/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Host-Pathogen Interactions/immunology , Phagosomes/metabolism , Biomarkers , Cell Communication/immunology , Cryptococcosis/immunology , Cryptococcosis/metabolism , Cryptococcosis/microbiology , Humans , Immunophenotyping , Lymphocyte Activation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL