Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
mBio ; 14(1): e0318422, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36598192

ABSTRACT

Aspergillus fumigatus is a ubiquitous environmental mold that causes significant mortality particularly among immunocompromised patients. The detection of the Aspergillus-derived carbohydrate galactomannan in patient serum and bronchoalveolar lavage fluid is the major biomarker used to detect A. fumigatus infection in clinical medicine. Despite the clinical relevance of this carbohydrate, we lack a fundamental understanding of how galactomannan is recognized by the immune system and its consequences. Galactomannan is composed of a linear mannan backbone with galactofuranose sidechains and is found both attached to the cell surface of Aspergillus and as a soluble carbohydrate in the extracellular milieu. In this study, we utilized fungal-like particles composed of highly purified Aspergillus galactomannan to identify a C-type lectin host receptor for this fungal carbohydrate. We identified a novel and specific interaction between Aspergillus galactomannan and the C-type lectin receptor Dectin-2. We demonstrate that galactomannan bound to Dectin-2 and induced Dectin-2-dependent signaling, including activation of spleen tyrosine kinase, gene transcription, and tumor necrosis factor alpha (TNF-α) production. Deficiency of Dectin-2 increased immune cell recruitment to the lungs but was dispensable for survival in a mouse model of pulmonary aspergillosis. Our results identify a novel interaction between galactomannan and Dectin-2 and demonstrate that Dectin-2 is a receptor for galactomannan, which leads to a proinflammatory immune response in the lung. IMPORTANCE Aspergillus fumigatus is a fungal pathogen that causes serious and often fatal disease in humans. The surface of Aspergillus is composed of complex sugar molecules. Recognition of these carbohydrates by immune cells by carbohydrate lectin receptors can lead to clearance of the infection or, in some cases, benefit the fungus by dampening the host response. Galactomannan is a carbohydrate that is part of the cell surface of Aspergillus but is also released during infection and is found in patient lungs as well as their bloodstreams. The significance of our research is that we have identified Dectin-2 as a mammalian immune cell receptor that recognizes, binds, and signals in response to galactomannan. These results enhance our understanding of how this carbohydrate interacts with the immune system at the site of infection and will lead to broader understanding of how release of galactomannan by Aspergillus effects the immune response in infected patients.


Subject(s)
Aspergillus fumigatus , Mannans , Animals , Mice , Humans , Lectins, C-Type/metabolism , Mammals/metabolism
2.
iScience ; 25(9): 104934, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36060075

ABSTRACT

Memory T cells underpin vaccine-induced immunity but are not yet fully understood. To distinguish features of memory cells that confer protective immunity, we used single cell transcriptome analysis to compare antigen-specific CD4+T cells recalled to lungs of mice that received a protective or nonprotective subunit vaccine followed by challenge with a fungal pathogen. We unexpectedly found populations specific to protection that expressed a strong type I interferon response signature, whose distinctive transcriptional signature appeared unconventionally dependent on IFN-γ receptor. We also detected a unique population enriched in protection that highly expressed the gene for the natural killer cell marker NKG7. Lastly, we detected differences in TCR gene use and in Th1- and Th17-skewed responses after protective and nonprotective vaccine, respectively, reflecting heterogeneous Ifng- and Il17a-expressing populations. Our findings highlight key features of transcriptionally diverse and distinctive antigen-specific T cells associated with protective vaccine-induced immunity.

3.
J Immunol ; 208(6): 1417-1423, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35217584

ABSTRACT

Homotypic signaling lymphocyte activation molecule (SLAM) receptor-ligand cell surface interactions between myeloid and lymphoid cells regulate innate and adaptive immune responses. In this article, we report that SLAMF1 is indispensable for host resistance to primary and vaccine-induced protection against fungal infection. Because vaccine immunity is dependent on cell-mediated immunity, we investigated the development of Ag-specific T cells. We studied the T cell-intrinsic and -extrinsic role of SLAMF1. We generated SLAMF1-/- TCR transgenic mice and analyzed the responses of adoptively transferred T cells. We also tracked endogenous Ag-specific T cells by using a tetramer. Intrinsic and extrinsic SLAMF1 signaling was dispensable for the development of antifungal Th1 and Th17 cells, which are requisite for the acquisition of vaccine-induced immunity. Despite intact T cell development, vaccinated SLAMF1-/- mice failed to control fungal infection. Failed accumulation of Ag-specific T cells in the lung on infection of vaccinated mice was due to uncontrolled early infection and inflammation, revealing a role for SLAMF1 in innate host immunity.


Subject(s)
Mycoses , Vaccines , Animals , Cell Differentiation , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Signaling Lymphocytic Activation Molecule Family Member 1/genetics , Th17 Cells
4.
mBio ; 13(1): e0340021, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35089087

ABSTRACT

Host genetic determinants that underpin variation in susceptibility to systemic fungal infection are poorly understood. Genes responsible for complex traits can be identified by correlating variation in phenotype with allele in founder strains of wild mice with known genetic variation, assembled in genetic reference panels. In this work, we describe wide natural variation in both primary and acquired resistance to experimental pulmonary blastomycosis in eight founder strains, including 129, A/J, BL/6, CAST, NOD, NZO, PWK, and WSB of the Collaborative Cross collection, and the inbred DBA strain. These differences in susceptibility across strains were accompanied by sharp differences in the accumulation and function of immune cells in the lungs. Immune perturbations were mapped by identifying reagents that phenotypically mark immune cell populations in the distinct strains of mice. In particular, we uncovered marked differences between BL/6 and DBA/2 mouse strains in the development of acquired resistance. Our findings highlight the potential value in using genetic reference panels of mice, and particularly the BXD (recombinant inbred strains of mice from a cross of C57BL/6J and DBA/2J mice) collection harboring a cross between resistant BL/6 and susceptible DBA/2 mice, for unveiling genes linked with host resistance to fungal infection. IMPORTANCE Host genetic variation significantly impacts vulnerability to infectious diseases. While host variation in susceptibility to fungal infection with dimorphic fungi has long been recognized, genes that underpin this variation are poorly understood. We used a collection of seven mouse strains that represent nearly 90% of the genetic variation in mice to identify genetic variability among the strains in resistance to pulmonary infection with the dimorphic fungus Blastomyces dermatitidis. We analyzed differences between the strains in innate resistance by infecting naive mice and in acquired resistance by infecting vaccinated mice. We identified extreme variations in both innate and acquired resistance among the strains. In particular, we found sharp differences between C57BL/6 and DBA/2 strains in the ability to acquire vaccine-induced resistance. We also identified commercial reagents that allowed the phenotyping of immune cells from this strain collection of mice. Because there are additional mice harboring a genetic cross of the C57BL/6 and DBA/2 strains (BXD collection), such mice will permit future investigations to identify the genes that underlie differences in the ability to acquire resistance to infection.


Subject(s)
Blastomyces , Immunophenotyping , Mice, Inbred Strains , Animals , Mice , Blastomyces/genetics , Blastomyces/immunology , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Mice, Inbred Strains/genetics , Mice, Inbred Strains/immunology
5.
mBio ; 12(4): e0201821, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34399628

ABSTRACT

The development of effective vaccines against fungal infections requires the induction of protective, pathogen-specific cell-mediated immune responses. Here, we asked whether combination adjuvants based on delta inulin (Advax) formulated with Toll-like receptor (TLR) agonists could improve vaccine protection mediated by a fungal recombinant protein, Bl-Eng2 (i.e., Blastomyces endoglucanase 2), which itself harbors an immunodominant antigen and dectin-2 agonist/adjuvant. We found that Bl-Eng2 formulated with Advax3 containing TLR9 agonist or Advax8 containing TLR4 agonist provided the best protection against pulmonary infection with Blastomyces dermatitidis, being more effective than complete Freund's adjuvant or Adjuplex. Advax3 was most efficient in inducing gamma interferon (IFN-γ)- and interleukin-17 (IL-17)-producing antigen-specific T cells that migrated to the lung upon Blastomyces dermatitidis infection. Mechanistic studies revealed Bl-Eng2/Advax3 protection was tempered by neutralization of IL-17 and particularly IFN-γ. Likewise, greater numbers of lung-resident T cells producing IFN-γ, IL-17, or both IFN-γ and IL-17 correlated with fewer fungi recovered from lung. Protection was maintained after depletion of CD4+ T cells, partially reduced by depletion of CD8+ T cells, and completely eliminated after depletion of both CD4+ and CD8+ T cells. We conclude that Bl-Eng2 formulated with Advax3 is promising for eliciting vaccine-induced antifungal immunity, through a previously uncharacterized mechanism involving CD8+ and also CD4+ T cells producing IFN-γ and/or IL-17. Although no licensed vaccine exists as yet against any fungal disease, these findings indicate the importance of adjuvant selection for the development of effective fungal vaccines. IMPORTANCE Fungal disease remains a challenging clinical and public health problem. Despite medical advances, invasive fungal infections have skyrocketed over the last decade and pose a mounting health threat in immunocompetent and -deficient hosts, with worldwide mortality rates ranking 7th, even ahead of tuberculosis. The development of safe, effective vaccines remains a major hurdle for fungi. Critical barriers to progress include the lack of defined fungal antigens and suitable adjuvants. Our research is significant in identifying adjuvant combinations that elicit optimal vaccine-induced protection when formulated with a recombinant protective antigen and uncovering the mechanistic bases of the underlaying vaccine protection, which will foster the strategic development of antifungal vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Fungal Vaccines/genetics , Fungal Vaccines/immunology , Mycoses/prevention & control , Animals , Blastomyces/immunology , Blastomycosis/prevention & control , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Fungal Vaccines/administration & dosage , Immunity, Cellular , Interferon-gamma , Inulin/administration & dosage , Inulin/analogs & derivatives , Inulin/immunology , Male , Mice , Mice, Inbred C57BL , Mycoses/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
6.
PLoS Pathog ; 17(3): e1009324, 2021 03.
Article in English | MEDLINE | ID: mdl-33735218

ABSTRACT

The development of safe subunit vaccines requires adjuvants that augment immunogenicity of non-replicating protein-based antigens. Current vaccines against infectious diseases preferentially induce protective antibodies driven by adjuvants such as alum. However, the contribution of antibody to host defense is limited for certain classes of infectious diseases such as fungi, whereas animal studies and clinical observations implicate cellular immunity as an essential component of the resolution of fungal pathogens. Here, we decipher the structural bases of a newly identified glycoprotein ligand of Dectin-2 with potent adjuvancy, Blastomyces endoglucanase-2 (Bl-Eng2). We also pinpoint the developmental steps of antigen-specific CD4+ and CD8+ T responses augmented by Bl-Eng2 including expansion, differentiation and tissue residency. Dectin-2 ligation led to successful systemic and mucosal vaccination against invasive fungal infection and Influenza A infection, respectively. O-linked glycans on Bl-Eng2 applied at the skin and respiratory mucosa greatly augment vaccine subunit- induced protective immunity against lethal influenza and fungal pulmonary challenge.


Subject(s)
Antibodies, Viral/immunology , Blastomyces/immunology , Fungal Vaccines/immunology , Orthomyxoviridae Infections/immunology , Adjuvants, Immunologic , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cellulase/immunology , Influenza Vaccines/immunology
7.
PLoS Negl Trop Dis ; 14(6): e0008386, 2020 06.
Article in English | MEDLINE | ID: mdl-32542003

ABSTRACT

Chromoblastomycosis (CBM) is a chronic worldwide subcutaneous mycosis, caused by several dimorphic, pigmented dematiaceous fungi. It is difficult to treat patients with the disease, mainly because of its recalcitrant nature. The correct activation of host immune response is critical to avoid fungal persistence in the tissue and disease chronification. CD4+ T cells are crucial for the development of protective immunity to F. pedrosoi infection. Here, we investigated T helper cell response dynamics during experimental CBM. Following footpad injection with F. pedrosoi hyphae and conidia, T cells were skewed towards a Th17 and Th1 phenotype. The Th17 population was the main Th cell subset found in the infected area during the early stages of experimental murine CBM, followed by Th1 predominance in the later stages, coinciding with the remission phase of the disease in this experimental model. Depletion of CD25+ cells, which leads to a reduction of Treg cells in the draining lymph node, resulted in decline in fungal burden after 14 days of infection. However, fungal cells were not cleared in the later stages of the disease, prolonging CBM clinical features in those animals. IL-17A and IFN-γ neutralization hindered fungal cell elimination in the course of the disease. Similarly, in dectin-2 KO animals, Th17 contraction in the course of experimental CBM was accompanied by fungal burden decrease in the first 14 days of infection, although it did not affect disease resolution. In this study, we gained insight into T helper subsets' dynamics following footpad injections of F. pedrosoi propagules and uncovered their contribution to disease resolution. The Th17 population proved to be important in eliminating fungal cells in the early stages of infection. The Th1 population, in turn, closely assisted by Treg cells, proved to be relevant not only in the elimination of fungal cells at the beginning of infection but also essential for their complete elimination in later stages of the disease in a mouse experimental model of CBM.


Subject(s)
Ascomycota/immunology , Chromoblastomycosis/immunology , Lectins, C-Type/metabolism , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Chromoblastomycosis/microbiology , Chromoblastomycosis/pathology , Disease Models, Animal , Humans , Hyphae , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lectins, C-Type/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spores, Fungal
8.
J Immunol ; 204(12): 3296-3306, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32358020

ABSTRACT

Coccidioides species are fungal pathogens that can cause a widely varied clinical manifestation from mild pulmonary symptom to disseminated, life-threatening disease. We have previously created a subunit vaccine by encapsulating a recombinant coccidioidal Ag (rCpa1) in glucan-chitin particles (GCPs) as an adjuvant-delivery system. The GCP-rCpa1 vaccine has shown to elicit a mixed Th1 and Th17 response and confers protection against pulmonary coccidioidomycosis in mice. In this study, we further delineated the vaccine-induced protective mechanisms. Depletion of IL-17A in vaccinated C57BL/6 mice prior to challenge abrogated the protective efficacy of GCP-rCpa1 vaccine. Global transcriptome and Ingenuity Pathway Analysis of murine bone marrow-derived macrophages after exposure to this vaccine revealed the upregulation of proinflammatory cytokines (TNF-α, IL-6, and IL-1ß) that are associated with activation of C-type lectin receptors (CLR) Dectin-1- and Dectin-2-mediated CARD9 signaling pathway. The GCP formulation of rCpa1 bound soluble Dectin-1 and Dectin-2 and triggered ITAM signaling of corresponding CLR reporter cells. Furthermore, macrophages that were isolated from Dectin-1 -/-, Dectin-2 -/-, and CARD9 -/- mice significantly reduced production of inflammatory cytokines in response to the GCP-rCpa1 vaccine compared with those of wild-type mice. The GCP-rCpa1 vaccine had significantly reduced protective efficacy in Dectin-1 -/-, Dectin-2 -/-, and CARD9 -/- mice that showed decreased acquisition of Th cells in Coccidioides-infected lungs compared with vaccinated wild-type mice, especially Th17 cells. Collectively, we conclude that the GCP-rCpa1 vaccine stimulates a robust Th17 immunity against Coccidioides infection through activation of the CARD9-associated Dectin-1 and Dectin-2 signal pathways.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Coccidioides/immunology , Coccidioidomycosis/immunology , Fungal Vaccines/immunology , Lectins, C-Type/immunology , Vaccines, Combined/immunology , Animals , Coccidioidomycosis/microbiology , Coccidioidomycosis/prevention & control , Cytokines/immunology , Female , Lung/immunology , Lung/microbiology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Signal Transduction/immunology , Th17 Cells/immunology
9.
Mucosal Immunol ; 13(3): 518-529, 2020 05.
Article in English | MEDLINE | ID: mdl-31900406

ABSTRACT

Priming at the site of natural infection typically elicits a protective T cell response against subsequent pathogen encounter. Here, we report the identification of a novel fungal antigen that we harnessed for mucosal vaccination and tetramer generation to test whether we can elicit protective, antigen-specific tissue-resident memory (Trm) CD4+ T cells in the lung parenchyma. In contrast to expectations, CD69+, CXCR3+, CD103- Trm cells failed to protect against a lethal pulmonary fungal infection. Surprisingly, systemic vaccination induced a population of tetramer+ CD4+ T cells enriched within the pulmonary vasculature, and expressing CXCR3 and CX3CR1, that migrated to the lung tissue upon challenge and efficiently protected mice against infection. Mucosal vaccine priming of Trm may not reliably protect against mucosal pathogens.


Subject(s)
Antigens/immunology , Cell Movement/immunology , Disease Resistance/immunology , Fungi/immunology , Host-Pathogen Interactions/immunology , Immunologic Memory , Mycoses/immunology , Animals , Biomarkers , Epitopes, T-Lymphocyte/immunology , Immunization , Immunophenotyping , Interferon-gamma , Mice , Mycoses/microbiology , Mycoses/prevention & control , Receptors, CXCR3/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccines/immunology
10.
PLoS Pathog ; 15(6): e1007777, 2019 06.
Article in English | MEDLINE | ID: mdl-31247052

ABSTRACT

The majority of invasive human fungal pathogens gain access to their human hosts via the inhalation of spores from the environment into the lung, but relatively little is known about this infectious process. Among human fungal pathogens the most frequent cause of inhaled fatal fungal disease is Cryptococcus, which can disseminate from the lungs to other tissues, including the brain, where it causes meningoencephalitis. To determine the mechanisms by which distinct infectious particles of Cryptococcus cause disseminated disease, we evaluated two developmental cell types (spores and yeast) in mouse models of infection. We discovered that while both yeast and spores from several strains cause fatal disease, there was a consistently higher fungal burden in the brains of spore-infected mice. To determine the basis for this difference, we compared the pathogenesis of avirulent yeast strains with their spore progeny derived from sexual crosses. Strikingly, we discovered that spores produced by avirulent yeast caused uniformly fatal disease in the murine inhalation model of infection. We determined that this difference in outcome is associated with the preferential dissemination of spores to the lymph system. Specifically, mice infected with spores harbored Cryptococcus in their lung draining lymph nodes as early as one day after infection, whereas mice infected with yeast did not. Furthermore, phagocyte depletion experiments revealed this dissemination to the lymph nodes to be dependent on CD11c+ phagocytes, indicating a critical role for host immune cells in preferential spore trafficking. Taken together, these data support a model in which spores capitalize on phagocytosis by immune cells to escape the lung and gain access to other tissues, such as the central nervous system, to cause fatal disease. These previously unrealized insights into early interactions between pathogenic fungal spores and lung phagocytes provide new opportunities for understanding cryptococcosis and other spore-mediated fungal diseases.


Subject(s)
Cryptococcosis/immunology , Cryptococcus/immunology , Inhalation Exposure , Meningoencephalitis/immunology , Phagocytes/immunology , Spores, Fungal/immunology , Animals , Cryptococcosis/pathology , Cryptococcus/pathogenicity , Disease Models, Animal , Humans , Lung/immunology , Lung/pathology , Meningoencephalitis/pathology , Mice , Phagocytes/pathology , Phagocytosis , RAW 264.7 Cells , Spores, Fungal/pathogenicity
11.
Sci Rep ; 9(1): 6788, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31043669

ABSTRACT

White-nose syndrome (WNS) caused by the fungus, Pseudogymnoascus destructans (Pd) has killed millions of North American hibernating bats. Currently, methods to prevent the disease are limited. We conducted two trials to assess potential WNS vaccine candidates in wild-caught Myotis lucifugus. In a pilot study, we immunized bats with one of four vaccine treatments or phosphate-buffered saline (PBS) as a control and challenged them with Pd upon transfer into hibernation chambers. Bats in one vaccine-treated group, that received raccoon poxviruses (RCN) expressing Pd calnexin (CAL) and serine protease (SP), developed WNS at a lower rate (1/10) than other treatments combined (14/23), although samples sizes were small. The results of a second similar trial provided additional support for this observation. Bats vaccinated orally or by injection with RCN-CAL and RCN-SP survived Pd challenge at a significantly higher rate (P = 0.01) than controls. Using RT-PCR and flow cytometry, combined with fluorescent in situ hybridization, we determined that expression of IFN-γ transcripts and the number of CD4 + T-helper cells transcribing this gene were elevated (P < 0.10) in stimulated lymphocytes from surviving vaccinees (n = 15) compared to controls (n = 3). We conclude that vaccination with virally-vectored Pd antigens induced antifungal immunity that could potentially protect bats against WNS.


Subject(s)
Ascomycota/immunology , Chiroptera/immunology , Host-Pathogen Interactions , Immunization/veterinary , Mycoses/prevention & control , Poxviridae/genetics , Viral Vaccines/administration & dosage , Animals , Ascomycota/pathogenicity , Chiroptera/microbiology , Chiroptera/virology , Hibernation , Mycoses/epidemiology , Mycoses/veterinary , Nose Diseases/epidemiology , Nose Diseases/microbiology , Pilot Projects , Syndrome
13.
Eur Radiol ; 29(9): 4803-4811, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30741342

ABSTRACT

OBJECTIVES: To obtain an overview of the attitudes toward interdisciplinary further education of residents and consultants in radiology and nuclear medicine and preferences regarding a future joint training curriculum in Switzerland. METHODS: A 34-item questionnaire was sent electronically (SurveyMonkey online survey tool) to 1244 radiologists and nuclear physicians (residents and consultants) in Switzerland. The items asked about the motivation for further education in each other's specialty and preferences regarding a joint further education curriculum in radiology and nuclear medicine. RESULTS: Overall, 370 questionnaires were analyzed (370/1244, 30%). There were 280 (76%) board-certified physicians in either radiology (238/370, 64%) or nuclear medicine (42/370, 12%) and 65 (18%) residents (radiology 54/370, 15%; nuclear medicine 11/370, 3%). More than half of all residents (34/65, 52%) stated their conviction that a wide range of expertise in both disciplines could be fully guaranteed through adequate cross-curricular training. For responders already at a consultant level in radiology or nuclear medicine, the willingness to undergo further training in each other's specialty significantly increased with a shorter training period. The preferred option for a possible future joint training curriculum was a combination of a 5-year radiology training program with 2 years of further training in nuclear medicine. CONCLUSIONS: Both residents and board-certified physicians in Switzerland are highly interested in a cross-curricular training curriculum in radiology and nuclear medicine. KEY POINTS: • A systematic survey was conducted to obtain information on interest in cross-curricular training in radiology and nuclear medicine and preferences regarding a future joint training curriculum. • More than half of radiology and nuclear medicine residents would be interested in further training in the other specialty. • There is a strong desire for a shorter training program when combining training in both radiology and nuclear medicine.


Subject(s)
Curriculum , Nuclear Medicine/education , Radiology/education , Adult , Consultants , Education, Medical, Continuing , Female , Humans , Internship and Residency , Male , Middle Aged , Surveys and Questionnaires , Switzerland , Young Adult
14.
mBio ; 9(2)2018 04 03.
Article in English | MEDLINE | ID: mdl-29615501

ABSTRACT

Blastomyces dermatitidis is a human fungal pathogen of the lung that can lead to disseminated disease in healthy and immunocompromised individuals. Genetic analysis of this fungus is hampered by the relative inefficiency of traditional recombination-based gene-targeting approaches. Here, we demonstrate the feasibility of applying CRISPR/Cas9-mediated gene editing to Blastomyces, including to simultaneously target multiple genes. We created targeting plasmid vectors expressing Cas9 and either one or two single guide RNAs and introduced these plasmids into Blastomyces via Agrobacterium gene transfer. We succeeded in disrupting several fungal genes, including PRA1 and ZRT1, which are involved in scavenging and uptake of zinc from the extracellular environment. Single-gene-targeting efficiencies varied by locus (median, 60% across four loci) but were approximately 100-fold greater than traditional methods of Blastomyces gene disruption. Simultaneous dual-gene targeting proceeded with efficiencies similar to those of single-gene-targeting frequencies for the respective targets. CRISPR/Cas9 disruption of PRA1 or ZRT1 had a variable impact on growth under zinc-limiting conditions, showing reduced growth at early time points in low-passage-number cultures and growth similar to wild-type levels by later passage. Individual impairment of PRA1 or ZRT1 resulted in a reduction of the fungal burden in a mouse model of Blastomyces infection by a factor of ~1 log (range, up to 3 logs), and combined disruption of both genes had no additional impact on the fungal burden. These results underscore the utility of CRISPR/Cas9 for efficient gene disruption in dimorphic fungi and reveal a role for zinc metabolism in Blastomyces fitness in vivoIMPORTANCEBlastomyces is a human fungal pathogen that can cause serious, even fatal, lung infections. Genetic analysis of this fungus is possible but inefficient. We applied a recently developed gene editing technology, CRISPR/Cas9, to dramatically improve the efficiency with which gene disruptions are introduced into Blastomyces We used this system to disrupt genes involved in zinc uptake and found that this reduced the fitness of the fungus upon infection.


Subject(s)
Blastomyces/growth & development , Blastomyces/metabolism , Gene Editing/methods , Genetic Fitness , Zinc/metabolism , Animals , Blastomyces/genetics , Blastomycosis/microbiology , CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Colony Count, Microbial , Disease Models, Animal , Metabolic Networks and Pathways/genetics , Mice , RNA, Guide, Kinetoplastida/metabolism
15.
Cell Host Microbe ; 23(4): 511-522.e5, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29576482

ABSTRACT

Lung epithelial cells (LECs) are strategically positioned in the airway mucosa to provide barrier defense. LECs also express pattern recognition receptors and a myriad of immune genes, but their role in immunity is often concealed by the activities of "professional" immune cells, particularly in the context of fungal infection. Here, we demonstrate that NF-κB signaling in LECs is essential for immunity against the pulmonary fungal pathogen Blastomyces dermatitidis. LECs orchestrate innate antifungal immunity by augmenting the numbers of interleukin-17A (IL-17A)- and granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing innate lymphocytes, specifically "natural" Th17 (nTh17) cells. Innate lymphocyte-derived IL-17A and GM-CSF in turn enable phagocyte-driven fungal killing. LECs regulate the numbers of nTh17 cells via the production of chemokines such as CCL20, a process dependent on IL-1α-IL-1 receptor (IL-1R) signaling on LECs. Therefore, LECs orchestrate IL-17A- and GM-CSF-mediated immunity in an IL-1R-dependent manner and represent an essential component of innate immunity to pulmonary fungal pathogens.


Subject(s)
Blastomyces/immunology , Blastomycosis/immunology , Epithelial Cells/immunology , Immunity, Innate , Lung/immunology , Lymphocytes/immunology , Animals , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-17/metabolism , Interleukin-1alpha/metabolism , Mice, Inbred C57BL , Phagocytes/immunology , Receptors, Interleukin-1/metabolism
16.
mBio ; 9(1)2018 01 02.
Article in English | MEDLINE | ID: mdl-29295913

ABSTRACT

The ability to grow at mammalian body temperatures is critical for pathogen infection of humans. For the thermally dimorphic fungal pathogen Histoplasma capsulatum, elevated temperature is required for differentiation of mycelia or conidia into yeast cells, a step critical for invasion and replication within phagocytic immune cells. Posttranslational glycosylation of extracellular proteins characterizes factors produced by the pathogenic yeast cells but not those of avirulent mycelia, correlating glycosylation with infection. Histoplasma yeast cells lacking the Pmt1 and Pmt2 protein mannosyltransferases, which catalyze O-linked mannosylation of proteins, are severely attenuated during infection of mammalian hosts. Cells lacking Pmt2 have altered surface characteristics that increase recognition of yeast cells by the macrophage mannose receptor and reduce recognition by the ß-glucan receptor Dectin-1. Despite these changes, yeast cells lacking these factors still associate with and survive within phagocytes. Depletion of macrophages or neutrophils in vivo does not recover the virulence of the mutant yeast cells. We show that yeast cells lacking Pmt functions are more sensitive to thermal stress in vitro and consequently are unable to productively infect mice, even in the absence of fever. Treatment of mice with cyclophosphamide reduces the normal core body temperature of mice, and this decrease is sufficient to restore the infectivity of O-mannosylation-deficient yeast cells. These findings demonstrate that O-mannosylation of proteins increases the thermotolerance of Histoplasma yeast cells, which facilitates infection of mammalian hosts.IMPORTANCE For dimorphic fungal pathogens, mammalian body temperature can have contrasting roles. Mammalian body temperature induces differentiation of the fungal pathogen Histoplasma capsulatum into a pathogenic state characterized by infection of host phagocytes. On the other hand, elevated temperatures represent a significant barrier to infection by many microbes. By functionally characterizing cells lacking O-linked mannosylation enzymes, we show that protein mannosylation confers thermotolerance on H. capsulatum, enabling infection of mammalian hosts.


Subject(s)
Fungal Proteins/metabolism , Histoplasma/physiology , Histoplasma/radiation effects , Mannosyltransferases/metabolism , Microbial Viability/radiation effects , Protein Processing, Post-Translational , Animals , Disease Models, Animal , Histoplasma/metabolism , Histoplasmosis/microbiology , Histoplasmosis/pathology , Mice, Inbred C57BL , Virulence
17.
Am J Respir Cell Mol Biol ; 58(2): 232-240, 2018 02.
Article in English | MEDLINE | ID: mdl-28886250

ABSTRACT

Pneumocystis is an important fungal pathogen that causes life-threatening pneumonia in patients with AIDS and malignancy. Lung fungal pathogens are recognized by C-type lectin receptors (CLRs), which bind specific ligands and stimulate innate immune responses. The CLR Dectin-1 was previously shown to mediate immune responses to Pneumocystis spp. For this reason, we investigated a potential role for Dectin-2. Rats with Pneumocystis pneumonia (PCP) exhibited elevated Dectin-2 mRNA levels. Soluble Dectin-2 carbohydrate-recognition domain fusion protein showed binding to intact Pneumocystis carinii (Pc) and to native Pneumocystis major surface glycoprotein/glycoprotein A (Msg/gpA). RAW macrophage cells expressing V5-tagged Dectin-2 displayed enhanced binding to Pc and increased protein tyrosine phosphorylation. Furthermore, the binding of Pc to Dectin-2 resulted in Fc receptor-γ-mediated intracellular signaling. Alveolar macrophages from Dectin-2-deficient mice (Dectin-2-/-) showed significant decreases in phospho-Syk activation after challenge with Pc cell wall components. Stimulation of Dectin-2-/- alveolar macrophages with Pc components showed significant decreases in the proinflammatory cytokines IL-6 and TNF-α. Finally, during infection with Pneumocystis murina, Dectin-2-/- mice displayed downregulated mRNA expression profiles of other CLRs implicated in fungal immunity. Although Dectin-2-/- alveolar macrophages had reduced proinflammatory cytokine release in vitro, Dectin-2-/- deficiency did not reduce the overall resistance of these mice in the PCP model, and organism burdens were statistically similar in the long-term immunocompromised and short-term immunocompetent PCP models. These results suggest that Dectin-2 participates in the initial innate immune signaling response to Pneumocystis, but its deficiency does not impair resistance to the organism.


Subject(s)
Immunity, Innate/immunology , Lectins, C-Type/immunology , Macrophages, Alveolar/immunology , Pneumocystis carinii/immunology , Pneumonia, Pneumocystis/immunology , Animals , Cell Line , Glycoproteins/metabolism , Inflammation/immunology , Inflammation/pathology , Interleukin-6/metabolism , Lectins, C-Type/genetics , Mice , Mice, Knockout , Phosphorylation , Pneumonia, Pneumocystis/microbiology , Pneumonia, Pneumocystis/pathology , RNA, Messenger/genetics , Rats , Tumor Necrosis Factor-alpha/metabolism
18.
PLoS Pathog ; 13(8): e1006568, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28793349

ABSTRACT

The development of vaccines against fungi and other intracellular microbes is impeded in part by a lack of suitable adjuvants. While most current vaccines against infectious diseases preferentially induce production of antibodies, cellular immunity is essential for the resolution of fungal infections. Microbes such as fungi and Mycobacterium tuberculosis require Th17 and Th1 cells for resistance, and engage the C-type lectin receptors including Dectin-2. Herein, we discovered a novel Dectin-2 ligand, the glycoprotein Blastomyces Eng2 (Bl-Eng2). Bl-Eng2 triggers robust signaling in Dectin-2 reporter cells and induces IL-6 in human PBMC and BMDC from wild type but not Dectin-2-/- and Card9-/- mice. The addition of Bl-Eng2 to a pan-fungal subunit vaccine primed large numbers of Ag-specific Th17 and Th1 cells, augmented activation and killing of fungi by myeloid effector cells, and protected mice from lethal fungal challenge, revealing Bl-Eng2's potency as a vaccine adjuvant. Thus, ligation of Dectin-2 by Bl-Eng-2 could be harnessed as a novel adjuvant strategy to protect against infectious diseases requiring cellular immunity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Fungal Proteins/immunology , Fungal Vaccines/immunology , Lectins, C-Type/immunology , Adjuvants, Immunologic/chemistry , Animals , Blastomyces , Fungal Proteins/chemistry , Fungal Vaccines/chemistry , Humans , Lectins, C-Type/metabolism , Leukocytes, Mononuclear/immunology , Ligands , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mycoses/immunology , Mycoses/prevention & control
20.
Cell Rep ; 19(5): 1008-1021, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28467895

ABSTRACT

The Fenton-chemistry-generating properties of copper ions are considered a potent phagolysosome defense against pathogenic microbes, yet our understanding of underlying host/microbe dynamics remains unclear. We address this issue in invasive aspergillosis and demonstrate that host and fungal responses inextricably connect copper and reactive oxygen intermediate (ROI) mechanisms. Loss of the copper-binding transcription factor AceA yields an Aspergillus fumigatus strain displaying increased sensitivity to copper and ROI in vitro, increased intracellular copper concentrations, decreased survival in challenge with murine alveolar macrophages (AMΦs), and reduced virulence in a non-neutropenic murine model. ΔaceA survival is remediated by dampening of host ROI (chemically or genetically) or enhancement of copper-exporting activity (CrpA) in A. fumigatus. Our study exposes a complex host/microbe multifactorial interplay that highlights the importance of host immune status and reveals key targetable A. fumigatus counter-defenses.


Subject(s)
Aspergillus/metabolism , Copper/metabolism , Host-Pathogen Interactions , Reactive Oxygen Species/metabolism , Animals , Aspergillus/genetics , Aspergillus/pathogenicity , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Fungal Proteins/genetics , Fungal Proteins/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , P-type ATPases/genetics , P-type ATPases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...