Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 11: 592826, 2021.
Article in English | MEDLINE | ID: mdl-34055661

ABSTRACT

Coccidioides is a soil-borne fungal pathogen and causative agent of a human respiratory disease (coccidioidomycosis) endemic to semi-desert regions of southwestern United States, Mexico, Central and South America. Aerosolized arthroconidia inhaled by the mammalian host first undergo conversion to large parasitic cells (spherules, 80-100 µm diameter) followed by endosporulation, a process by which the contents of spherules give rise to multiple endospores. The latter are released upon rupture of the maternal spherules and establish new foci of lung infection. A novel feature of spherule maturation prior to endosporulation is the secretion of a lipid-rich, membranous cell surface layer shed in vivo during growth of the parasitic cells and secretion into liquid culture medium during in vitro growth. Chemical analysis of the culture derived spherule outer wall (SOW) fraction showed that it is composed largely of phospholipids and is enriched with saturated fatty acids, including myristic, palmitic, elaidic, oleic, and stearic acid. NMR revealed the presence of monosaccharide- and disaccharide-linked acylglycerols and sphingolipids. The major sphingolipid components are sphingosine and ceramide. Primary neutrophils derived from healthy C57BL/6 and DBA/2 mice incubated with SOW lipids revealed a significant reduction in fungicidal activity against viable Coccidioides arthroconidia compared to incubation of neutrophils with arthroconidia alone. Host cell exposure to SOW lipids had no effect on neutrophil viability. Furthermore, C57BL/6 mice that were challenged subcutaneously with Coccidioides arthroconidia in the presence of the isolated SOW fraction developed disseminated disease, while control mice challenged with arthroconidia alone by the same route showed no dissemination of infection. We hypothesize that SOW lipids contribute to suppression of inflammatory response to Coccidioides infection. Studies are underway to characterize the immunosuppressive mechanism(s) of SOW lipids.


Subject(s)
Coccidioides , Coccidioidomycosis , Lipids , Animals , Mice , Mice, Inbred C57BL , Mice, Inbred DBA
2.
Infect Immun ; 86(11)2018 11.
Article in English | MEDLINE | ID: mdl-30104216

ABSTRACT

Developing an effective and safe recombinant vaccine requires microbe-specific antigens combined with an adjuvant/delivery system to strengthen protective immunity. In this study, we designed and expressed a multivalent recombinant Coccidioides polypeptide antigen (rCpa1) that consists of three previously identified antigens (i.e., Ag2/Pra, Cs-Ag, and Pmp1) and five pathogen-derived peptides with high affinity for human major histocompatibility complex class II (MHC-II) molecules. The purified rCpa1 was encapsulated into four types of yeast cell wall particles containing ß-glucan, mannan, and chitin in various proportions or was mixed with an oligonucleotide (ODN) containing two methylated dinucleotide CpG motifs. This multivalent antigen encapsulated into glucan-chitin particles (GCP-rCpa1) showed significantly greater reduction of fungal burden for human HLA-DR4 transgenic mice than the other adjuvant-rCpa1 formulations tested. Among the adjuvants tested, both GCPs and ß-glucan particles (GPs) were capable of stimulating a mixed Th1 and Th17 response. Mice vaccinated with GCP-rCpa1 showed higher levels of interleukin 17 (IL-17) production in T-cell recall assays and earlier lung infiltration by activated Th1 and Th17 cells than GP-rCpa1-vaccinated mice. Both C57BL/6 and HLA-DR4 transgenic mice that were vaccinated with the GCP-rCpa1 vaccine showed higher survival rates than mice that received GCPs alone. Concurrently, the GCP-rCpa1 vaccine stimulated greater infiltration of the injection sites by macrophages, which engulf and process the vaccine for antigen presentation, than the GP-rCpa1 vaccine. This is the first attempt to systematically characterize the presentation of a multivalent coccidioidomycosis vaccine encapsulated with selected adjuvants that enhance the protective cellular immune response to infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Chitin/administration & dosage , Coccidioides/immunology , Coccidioidomycosis/prevention & control , Glucans/administration & dosage , Protozoan Vaccines/immunology , Th17 Cells/immunology , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Disease Models, Animal , Drug Delivery Systems , HLA-DR4 Antigen/genetics , HLA-DR4 Antigen/metabolism , Humans , Mice, Inbred C57BL , Mice, Transgenic , Nanoparticles/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Protein Binding , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Survival Analysis , Th1 Cells/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
3.
Vaccine ; 34(44): 5336-5343, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27622300

ABSTRACT

There is an emerging interest to develop human vaccines against medically-important fungal pathogens and a need for a preclinical animal model to assess vaccine efficacies and protective correlates. HLA-DR4 (DRB1∗0401 allele) transgenic mice express a human major histocompatibility complex class II (MHC II) receptor in such a way that CD4+ T-cell response is solely restricted by this human molecule. In this study HLA-DR4 transgenic mice were immunized with a live-attenuated vaccine (ΔT) and challenged by the intranasal route with 50-70 Coccidioides posadasii spores, a potentially lethal dose. The same vaccination regimen offers 100% survival for C57BL/6 mice. Conversely, ΔT-vaccinated HLA-DR4 mice displayed 3 distinct manifestations of Coccidioides infection including 40% fatal acute (FAD), 30% disseminated (DD) and 30% pulmonary disease (PD). The latter 2 groups of mice had reduced loss of body weight and survived to at least 50days postchallenge (dpc). These results suggest that ΔT vaccinated HLA-DR4 mice activated heterogeneous immunity against pulmonary Coccidioides infection. Vaccinated HLA-DR4 mice displayed early expansion of Th1 and Th17 cells and recruitment of inflammatory innate cells into Coccidioides-infected lungs during the first 9dpc. While contraction rates of Th cells and the inflammatory response during 14-35dpc significantly differed among the 3 groups of vaccinated HLA-DR4 mice. The FAD group displayed a sharply reduced Th1 and Th17 response, while overwhelmingly recruiting neutrophils into lungs during 9-14days. The FAD group approached moribund by 14dpc. In contrast, vaccinated HLA-DR4 survivors gradually contracted Th cells and inflammatory response with the greatest rate in the PD group. While vaccinated HLA-DR4 mice are susceptible to Coccidioides infection, they are useful for evaluation of vaccine efficacy and identification of immunological correlates against this mycosis.


Subject(s)
Coccidioides/isolation & purification , Coccidioidomycosis/immunology , Coccidioidomycosis/prevention & control , Fungal Vaccines/immunology , HLA-DR4 Antigen/immunology , Adaptive Immunity , Animals , Coccidioides/immunology , Coccidioidomycosis/microbiology , Cytokines/biosynthesis , Fungal Vaccines/administration & dosage , HLA-DR4 Antigen/genetics , Humans , Immunity, Innate , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spores, Fungal , Th17 Cells/immunology , Vaccines, Attenuated
4.
Methods Mol Biol ; 1403: 551-66, 2016.
Article in English | MEDLINE | ID: mdl-27076153

ABSTRACT

The incidence of systemic fungal infections has increased throughout the world, spurring much interest in developing effective vaccines. Coccidioidomycosis, also known as San Joaquin Valley fever, is a potentially life-threatening respiratory mycosis. A vaccine against Coccidioides infection would contribute significantly to the well-being of the approx. 30 million residents in the Southwestern USA as well as the multitude of travelers who annually visit the endemic regions. We have applied a live, attenuated vaccine (∆T) to explore the nature of vaccine immunity in mice after intranasal challenge with a potentially lethal dose of Coccidioides spores. Coccidioides spores are airborne and highly infectious for mammalian hosts and classified as a biosafety level 3 agent. T cells are critical in the development of protective immunity against a variety of microorganisms as well as the development of autoimmune disease and allergic responses. Profiles of cytokines detected in lung homogenates of ∆T-vaccinated mice were indicative of a mixed Th1, Th2, and Th17 immune response. We have developed an intracellular cytokine staining and flow cytometric (ICS) technique to measure activated CD4(+) and CD8(+) T cells and IFN-γ-, IL-4-, IL-5-, and IL-17A-producing T cells in the lungs of mice that are challenged with a potentially lethal dose of Coccidioides spores. The numbers of pulmonary Th1 and Th17 cells during the first 2 weeks post-challenge showed a progressive increase in vaccinated mice and corresponded with reduction of fungal burden. In this protocol, we describe the methodology for culture and isolation of the live, attenuated ΔT spores of Coccidioides used to vaccinate mice, preparation of pulmonary cells, and staining protocol for cell surface markers and intracellular cytokines. This is the most reliable and robust procedure to measure frequencies and numbers of each selected T-cell subsets in lungs of vaccinated versus control mice and can be readily applied to evaluate T-cell response against other microbial infections.


Subject(s)
Coccidioides/immunology , Coccidioidomycosis/prevention & control , Flow Cytometry/methods , Lung/microbiology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Biomarkers/metabolism , Coccidioides/physiology , Cytokines/metabolism , Fungal Vaccines/immunology , Intracellular Space/metabolism , Lung/immunology , Lymphocyte Activation/immunology , Mice , Vaccination , Vaccines, Attenuated/immunology
5.
Infect Immun ; 84(4): 1166-75, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26857574

ABSTRACT

Coccidioidomycosis is a potentially life-threatening respiratory disease which is endemic to the southwestern United States and arid regions of Central and South America. It is responsible for approximately 150,000 infections annually in the United States alone. Almost every human organ has been reported to harbor parasitic cells of Coccidioides spp. in collective cases of the disseminated form of this mycosis. Current understanding of the mechanisms of protective immunity against lung infection has been largely derived from murine models of pulmonary coccidioidomycosis. However, little is known about the nature of the host response to Coccidioides in extrapulmonary tissue. Primary subcutaneous coccidioidal infection is rare but has been reported to result in disseminated disease. Here, we show that activation of MyD88 and Card9 signal pathways are required for resistance to Coccidioides infection following subcutaneous challenge of C57BL/6 mice, which correlates with earlier findings of the protective response to pulmonary infection. MyD88(-/-) andCard9(-/-) mice recruited reduced numbers of T cells, B cells, and neutrophils to the Coccidioides-infected hypodermis com pared to wild-type mice; however, neutrophils were dispensable for resistance to skin infection. Further studies have shown that gamma interferon (IFN-γ) production and activation of Th1 cells characterize resistance to subcutaneous infection. Furthermore, activation of a phagosomal enzyme, inducible nitric oxide synthase, which is necessary for NO production, is a requisite for fungal clearance in the hypodermis. Collectively, our data demonstrate that MyD88- and Card9-mediated IFN-γ and nitric oxide production is essential for protection against subcutaneous Coccidioides infection.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Coccidioidomycosis/microbiology , Interferon-gamma/metabolism , Myeloid Differentiation Factor 88/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Animals , B-Lymphocytes , CARD Signaling Adaptor Proteins/genetics , Coccidioides/physiology , Coccidioidomycosis/immunology , Coccidioidomycosis/metabolism , Gene Expression Regulation/physiology , Interferon-gamma/genetics , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukins/genetics , Interleukins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neutrophils , Nitric Oxide Synthase Type II/genetics , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , T-Lymphocytes/physiology , Interferon gamma Receptor
6.
Infect Immun ; 82(5): 2106-14, 2014 May.
Article in English | MEDLINE | ID: mdl-24614655

ABSTRACT

Interleukin-17A (IL-17A)-producing CD4(+) T helper (Th17) cells have been shown to be essential for defense against pulmonary infection with Coccidioides species. However, we have just begun to identify the required pattern recognition receptors and understand the signal pathways that lead to Th17 cell activation after fungal infection. We previously reported that Card9(-/-) mice vaccinated with formalin-killed spherules failed to acquire resistance to Coccidioides infection. Here, we report that both MyD88(-/-) and Card9(-/-) mice immunized with a live, attenuated vaccine also fail to acquire protective immunity to this respiratory disease. Like Card9(-/-) mice, vaccinated MyD88(-/-) mice revealed a significant reduction in numbers of both Th17 and Th1 cells in their lungs after Coccidioides infection. Both Toll-like receptor 2 (TLR2) and IL-1 receptor type 1 (IL-1r1) upstream of MyD88 have been implicated in Th17 cell differentiation. Surprisingly, vaccinated TLR2(-/-) and wild-type (WT) mice showed similar outcomes after pulmonary infection with Coccidioides, while vaccinated IL-1r1(-/-) mice revealed a significant reduction in the number of Th17 cells in their infected lungs compared to WT mice. Thus, activation of both IL-1r1/MyD88- and Card9-mediated Th17 immunity is essential for protection against Coccidioides infection. Our data also reveal that the numbers of Th17 cells were reduced in IL-1r1(-/-) mice to a lesser extent than in MyD88(-/-) mice, raising the possibility that other TLRs are involved in MyD88-dependent Th17 immunity to coccidioidomycosis. An antimicrobial action of Th17 cells is to promote early recruitment of neutrophils to infection sites. Our data revealed that neutrophils are required for vaccine immunity to this respiratory disease.


Subject(s)
Coccidioides , Coccidioidomycosis/immunology , Myeloid Differentiation Factor 88/metabolism , Receptors, Interleukin-1 Type I/metabolism , Toll-Like Receptor 2/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CARD Signaling Adaptor Proteins , Fungal Vaccines/immunology , Gene Expression Regulation/immunology , Mice , Myeloid Differentiation Factor 88/genetics , Receptors, Interleukin-1 Type I/genetics , Th17 Cells , Toll-Like Receptor 2/genetics
7.
J Immunol ; 192(3): 1107-1119, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24391211

ABSTRACT

Vaccine immunity to the endemic mycoses of North America requires Th17 cells, but the pattern recognition receptors and signaling pathways that drive these protective responses have not been defined. We show that C-type lectin receptors exert divergent contributions to the development of antifungal Th17 cells and vaccine resistance against Blastomyces dermatitidis, Histoplasma capsulatum, and Coccidioides posadasii. Acquired immunity to B. dermatitidis requires Dectin-2, whereas vaccination against H. capsulatum and C. posadasii infection depends on innate sensing by Dectin-1 and Dectin-2, but not Mincle. Tracking Ag-specific T cells in vivo established that the Card9 signaling pathway acts indispensably and exclusively on differentiation of Th17 cells, while leaving intact their activation, proliferation, survival, and migration. Whereas Card9 signaling is essential, C-type lectin receptors offer distinct and divergent contributions to vaccine immunity against these endemic fungal pathogens. Our work provides new insight into innate immune mechanisms that drive vaccine immunity and Th17 cells.


Subject(s)
Blastomyces/immunology , Blastomycosis/prevention & control , Coccidioides/immunology , Coccidioidomycosis/prevention & control , Endemic Diseases , Fungal Vaccines/immunology , Histoplasma/immunology , Histoplasmosis/prevention & control , Lectins, C-Type/immunology , Th17 Cells/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/immunology , Adoptive Transfer , Animals , Animals, Congenic , Antibodies, Fungal/biosynthesis , Antibodies, Fungal/immunology , Blastomycosis/epidemiology , CARD Signaling Adaptor Proteins , Coccidioidomycosis/epidemiology , Cytokines/biosynthesis , Dendritic Cells/immunology , Histoplasmosis/epidemiology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Radiation Chimera , Signal Transduction/immunology , Vaccination
8.
Infect Immun ; 82(2): 903-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478103

ABSTRACT

High concentrations of lung tissue-associated interleukin-10 (IL-10), an anti-inflammatory and immunosuppressive cytokine, correlate with susceptibility of mice to Coccidioides spp. infection. In this study, we found that macrophages, dendritic cells, neutrophils, and both CD8(+) and CD4(+) T cells recruited to Coccidioides posadasii-infected lungs of nonvaccinated and vaccinated mice contributed to the production of IL-10. The major IL-10-producing leukocytes were CD8(+) T cells, neutrophils, and macrophages in lungs of nonvaccinated mice, while both Foxp3(+) and Foxp3(-) subsets of IL-10(+) CD4(+) T cells were significantly elevated in vaccinated mice. Profiles of the recruited leukocytes in lungs revealed that only CD4(+) T cells were significantly increased in IL-10(-/-) knockout mice compared to their wild-type counterparts. Furthermore, ex vivo recall assays showed that CD4(+) T cells isolated from vaccinated IL-10(-/-) mice compared to vaccinated wild-type mice produced significantly higher amounts of IL-2, gamma interferon (IFN-γ), IL-4, IL-6, and IL-17A in the presence of a coccidioidal antigen, indicating that IL-10 suppresses Th1, Th2, and Th17 immunity to Coccidioides infection. Analysis of absolute numbers of CD44(+) CD62L(-) CD4(+) T effector memory T cells (TEM) and IFN-γ- and IL-17A-producing CD4(+) T cells in the lungs of Coccidioides-infected mice correlated with better fungal clearance in nonvaccinated IL-10(-/-) mice than in nonvaccinated wild-type mice. Our results suggest that IL-10 suppresses CD4(+) T-cell immunity in nonvaccinated mice during Coccidioides infection but does not impede the development of a memory response nor exacerbate immunopathology of vaccinated mice over at least a 4-month period after the last immunization.


Subject(s)
Coccidioides/immunology , Coccidioidomycosis/immunology , Coccidioidomycosis/prevention & control , Fungal Vaccines/immunology , Immunologic Memory , Interleukin-10/immunology , T-Lymphocytes/immunology , Animals , Female , Fungal Vaccines/administration & dosage , Lung/immunology , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/immunology
10.
Microb Pathog ; 59-60: 19-28, 2013.
Article in English | MEDLINE | ID: mdl-23583291

ABSTRACT

Coccidioides is the causative agent of a potentially life-threatening respiratory disease of humans. A feature of this mycosis is that pH measurements of the microenvironment of pulmonary abscesses are consistently alkaline due to ammonia production during the parasitic cycle. We previously showed that enzymatically active urease is partly responsible for elevated concentrations of extracellular ammonia at sites of lung infection and contributes to both localized host tissue damage and exacerbation of the respiratory disease in BALB/c mice. Disruption of the urease gene (URE) of Coccidioides posadasii only partially reduced the amount of ammonia detected during in vitro growth of the parasitic phase, suggesting that other ammonia-producing pathways exist that may also contribute to the virulence of this pathogen. Ureidoglycolate hydrolase (Ugh) expressed by bacteria, fungi and higher plants catalyzes the hydrolysis of ureidoglycolate to yield glyoxylate and the release CO2 and ammonia. This enzymatic pathway is absent in mice and humans. Ureidoglycolate hydrolase gene deletions were conducted in a wild type (WT) isolate of C. posadasii as well as the previously generated Δure knock-out strain. Restorations of UGH in the mutant stains were performed to generate and evaluate the respective revertants. The double mutant revealed a marked decrease in the amount of extracellular ammonia without loss of reproductive competence in vitro compared to both the WT and Δure parental strains. BALB/c mice challenged intranasally with the Δugh/Δure mutant showed 90% survival after 30 days, decreased fungal burden, and well-organized pulmonary granulomas. We conclude that loss of both Ugh and Ure activity significantly reduced the virulence of this fungal pathogen.


Subject(s)
Amidine-Lyases/metabolism , Ammonia/metabolism , Coccidioides/metabolism , Coccidioides/pathogenicity , Coccidioidomycosis/pathology , Lung Diseases, Fungal/pathology , Urease/metabolism , Amidine-Lyases/genetics , Animals , Coccidioides/enzymology , Coccidioides/genetics , Coccidioidomycosis/microbiology , Disease Models, Animal , Gene Knockout Techniques , Humans , Lung Diseases, Fungal/microbiology , Mice , Mice, Inbred BALB C , Survival Analysis , Urease/genetics , Virulence
11.
Infect Immun ; 80(11): 3960-74, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22949556

ABSTRACT

Clinical and animal studies of coccidioidomycosis have demonstrated that activated CD4(+) T lymphocytes are essential for protection against this fungal respiratory disease. We previously reported a vaccine against Coccidioides infection which contained three recombinant CD4(+) T cell-reactive proteins and induced a robust, protective immune response in mice. Due to the anticipated high cost of production and clinical assessment of this multivalent vaccine, we generated a single protein which contained immunodominant T cell epitopes of the three polypeptides. Epitopes were initially identified by computational prediction of their ability to bind promiscuously to human major histocompatibility complex class II (MHC II) molecules. Cellular immunoassays confirmed the immunogenicity of the synthesized epitope peptides, while in vitro binding assays revealed a range of peptide affinity for MHC II. A DNA construct was synthesized for bacterial expression of a recombinant protein vaccine which contained five epitopes with the highest affinity for human MHC II, each fused with leader and spacer peptides proposed to optimize epitope processing and presentation to T cell receptors. Recall assays of immune T lymphocytes obtained from human MHC II-expressing HLA-DR4 transgenic mice confirmed that 4 of the 5 epitope peptides were processed. Mice immunized with the epitope-based vaccine admixed with a synthetic oligodeoxynucleotide adjuvant or loaded into yeast glucan particles and then challenged intranasally with Coccidioides showed early lung infiltration of activated T helper-1 (Th1), Th2, and Th17 cells, elevated gamma interferon (IFN-γ) and interleukin (IL)-17 production, significant reduction of fungal burden, and prolongation of survival compared to nonvaccinated mice. This is the first report of an epitope-based vaccine against coccidioidomycosis.


Subject(s)
Coccidioides/immunology , Coccidioidomycosis/immunology , Epitopes, T-Lymphocyte/immunology , Fungal Vaccines/immunology , Lung Diseases, Fungal/prevention & control , Animals , Coccidioidomycosis/prevention & control , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Lung/immunology , Lung/microbiology , Lung Diseases, Fungal/immunology , Mice , Mice, Inbred Strains , Recombinant Proteins/immunology , Vaccines, Synthetic/immunology
12.
PLoS One ; 7(7): e41034, 2012.
Article in English | MEDLINE | ID: mdl-22911737

ABSTRACT

Coccidioides immitis and C. posadasii, the causative agents of coccidioidomycosis, are dimorphic fungal pathogens, which grow as hyphae in the saprobic phase in the environment and as spherules in the parasitic phase in the mammalian host. In this study, we use comparative transcriptomics to identify gene expression differences between the saprobic and parasitic growth phases. We prepared Illumina mRNA sequencing libraries for saprobic-phase hyphae and parasitic-phase spherules in vitro for C. immitis isolate RS and C. posadasii isolate C735 in biological triplicate. Of 9,910 total predicted genes in Coccidioides, we observed 1,298 genes up-regulated in the saprobic phase of both C. immitis and C. posadasii and 1,880 genes up-regulated in the parasitic phase of both species. Comparing the saprobic and parasitic growth phases, we observed considerable differential expression of cell surface-associated genes, particularly chitin-related genes. We also observed differential expression of several virulence factors previously identified in Coccidioides and other dimorphic fungal pathogens. These included alpha (1,3) glucan synthase, SOWgp, and several genes in the urease pathway. Furthermore, we observed differential expression in many genes predicted to be under positive selection in two recent Coccidioides comparative genomics studies. These results highlight a number of genes that may be crucial to dimorphic phase-switching and virulence in Coccidioides. These observations will impact priorities for future genetics-based studies in Coccidioides and provide context for studies in other fungal pathogens.


Subject(s)
Coccidioides/genetics , Gene Expression Regulation, Fungal , Transcriptome , Coccidioides/growth & development , Gene Expression Profiling , Virulence Factors/genetics
13.
J Immunol ; 189(2): 606-15, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22711891

ABSTRACT

Influenza A virus (flu) is a respiratory tract pathogen causing high morbidity and mortality among the human population. NO is a cellular mediator involved in tissue damage through its apoptosis of target cells and resulting enhancement of local inflammation. Inducible NO synthase (iNOS) is involved in the production of NO following infection. Although NO is a key player in the development of exaggerated lung disease during flu infection, the underlying mechanism, including the role of NO in apoptosis during infection, has not been reported. Similarly, the mechanism of iNOS gene induction during flu infection is not well defined in terms of the host transactivator(s) required for iNOS gene expression. In the current study, we identified Kruppel-like factor 6 (KLF6) as a critical transcription factor essential for iNOS gene expression during flu infection. We also underscored the requirement for iNOS in inducing apoptosis during infection. KLF6 gene silencing in human lung epithelial cells resulted in the drastic loss of NO production, iNOS promoter-specific luciferase activity, and expression of iNOS mRNA following flu infection. Chromatin immunoprecipitation assay revealed a direct interaction of KLF6 with iNOS promoter during in vitro and in vivo flu infection of human lung cells and mouse respiratory tract, respectively. A significant reduction in flu-mediated apoptosis was noted in KLF6-silenced cells, cells treated with iNOS inhibitor, and primary murine macrophages derived from iNOS knockout mice. A similar reduction in apoptosis was noted in the lungs following intratracheal flu infection of iNOS knockout mice.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Kruppel-Like Transcription Factors/physiology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Orthomyxoviridae Infections/immunology , Proto-Oncogene Proteins/physiology , Transcriptional Activation/immunology , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Cell Line , Gene Silencing/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/deficiency , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Transcriptional Activation/genetics
14.
Vaccine ; 30(31): 4681-90, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22575167

ABSTRACT

Coccidioides is a fungal pathogen and causative agent of a human respiratory disease against which no clinical vaccine exists. In this study we evaluated a novel vaccine adjuvant referred to as EP67, which is a peptide agonist of the biologically active C-terminal region of human complement component C5a. The EP67 peptide was conjugated to live spores of an attenuated vaccine strain (ΔT) of Coccidioides posadasii. The non-conjugated ΔT vaccine provided partial protection to BALB/c mice against coccidioidomycosis. In this report we compared the protective efficacy of the ΔT-EP67 conjugate to the ΔT vaccine in BALB/c mice. Animals immunized subcutaneously with the ΔT-EP67 vaccine showed significant increase in survival and decrease in fungal burden over 75 days postchallenge. Increased pulmonary infiltration of dendritic cells and macrophages was observed on day 7 postchallenge but marked decrease in neutrophil numbers had occurred by 11 days. The reduced influx of neutrophils may have contributed to the observed reduction of inflammatory pathology. Mice immunized with the ΔT-EP67 vaccine also revealed enhanced expression of MHC II molecules on the surface of antigen presenting cells, and in vitro recall assays of immune splenocytes showed elevated Th1- and Th17-type cytokine production. The latter correlated with a marked increase in lung infiltration of IFN-γ- and IL-17-producing CD4(+) T cells. Elevated expression of T-bet and RORc transcription factors in ΔT-EP67-vaccinated mice indicated the promotion of Th1 and Th17 cell differentiation. Higher titers of Coccidioides antigen-specific IgG1 and IgG2a were detected in mice immunized with the EP67-conjugated versus the non-conjugated vaccine. These combined results suggest that the EP67 adjuvant enhances protective efficacy of the live vaccine by augmentation of T-cell immunity, especially through Th1- and Th17-mediated responses to Coccidioides infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Coccidioidomycosis/prevention & control , Complement C5a/agonists , Fungal Vaccines/immunology , Animals , Antibodies, Fungal/blood , Antigen-Presenting Cells/immunology , Coccidioides/immunology , Coccidioidomycosis/immunology , Female , Humans , Immunity, Cellular , Interferon-gamma/immunology , Interleukin-17/immunology , Mice , Mice, Inbred BALB C , Peptides/administration & dosage , Th1 Cells/immunology , Th17 Cells/immunology , Vaccines, Attenuated/immunology
15.
Methods Mol Biol ; 845: 131-47, 2012.
Article in English | MEDLINE | ID: mdl-22328372

ABSTRACT

The following transformation protocol is based on homologous recombination that occurs between a gene disruption or gene replacement construct and a target gene of Coccidioides. The DNA constructs employed contain either the gene that encodes for hygromycin B or phleomycin resistance, which are present in the pAN7.1 or pAN8.1 plasmid vectors, respectively. Hygromycin B or phleomycin are used to select for transformants at concentrations that inhibit growth of the parental strain. Coccidioides protoplasts generated from germinated arthroconidia are used for the transformation experiments. The plasmid DNA constructs are taken up by the protoplasts in the presence of calcium and polyethylene glycol. Twenty to 100 transformants/µg DNA can be obtained in each transformation experiment. Approximately 5-10% of the transformation events are homologous recombinations. Coccidioides cells in all developmental stages, including arthroconidia, are multinucleate. Since all Coccidioides nuclei are haploid, only one run of transformation is sufficient to create a mutant strain. However, the transformed protoplasts develop into heterokaryotic cells that typically contain both the parental and mutated nuclei. To isolate a homokaryotic strain, we perform multiple subcultures of the single colonies which contain heterokaryotic cells on selection plates with hygromycin B or phleomycin to enrich for the mutated nuclei. Homokaryotic mutants can be obtained after three to four subcultures of isolated colonies. In this protocol, we describe the methodology for preparation of Coccidioides protoplasts, transformation and isolation of homokaryotic mutants.


Subject(s)
Cinnamates/pharmacology , Coccidioides/drug effects , Coccidioides/genetics , Drug Resistance, Fungal/genetics , Gene Deletion , Genetic Markers/genetics , Hygromycin B/analogs & derivatives , Phleomycins/pharmacology , Drug Resistance, Fungal/drug effects , Genetic Markers/drug effects , Hygromycin B/pharmacology , Mutagenesis/genetics
16.
Curr Fungal Infect Rep ; 6(4): 235-244, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23585916

ABSTRACT

Coccidioidomycosis (San Joaquin Valley fever) is a human respiratory disease caused by a soil-borne mold, and is recognized as an intransigent microbial infection by physicians who treat patients with the potentially life-threatening, disseminated form of this mycosis. Epidemiological studies based on surveys of skin-test reactivity of people who reside in the endemic regions of the Southwestern US have shown that at least 150,000 new infections occur annually. The clinical spectrum of coccidioidomycosis ranges from an asymptomatic insult to a severe pulmonary disease in which the pathogen may spread from the lungs to the skin, bones, brain and other body organs. Escalation of symptomatic infections and increased cost of long-term antifungal treatment warrant a concerted effort to develop a vaccine against coccidioidomycosis. This review examines recently reported strategies used to generate such a vaccine and summarizes current understanding of the nature of protective immunity to this formidable disease.

17.
BMC Microbiol ; 11: 245, 2011 Nov 02.
Article in English | MEDLINE | ID: mdl-22047041

ABSTRACT

BACKGROUND: Streptococcus pneumoniae (the pneumococcus) is the leading cause of otitis media, community-acquired pneumonia (CAP), sepsis, and meningitis. It is now evident that S. pneumoniae forms biofilms during nasopharyngeal colonization; the former which facilitates persistence, the latter, a prerequisite for subsequent development of invasive disease. Proteomic evaluation of S. pneumoniae suggests the antigen profile available for host-recognition is altered as a consequence of biofilm growth. This has potentially meaningful implications in regards to adaptive immunity and protection from disseminated disease. We therefore examined the antigen profile of biofilm and planktonic pneumococcal cell lysates, tested their reactivity with human convalescent sera and that generated against biofilm pneumococci, and examined whether immunization with biofilm pneumococci protected mice against infectious challenge. RESULTS: Biofilm pneumococci have dramatically altered protein profiles versus their planktonic counterparts. During invasive disease the humoral immune response is skewed towards the planktonic protein profile. Immunization with biofilm bacteria does not elicit a strong-cross-reactive humoral response against planktonic bacteria nor confer resistance against challenge with a virulent isolate from another serotype. We identified numerous proteins, including Pneumococcal serine-rich repeat protein (PsrP), which may serve as a protective antigens against both colonization and invasive disease. CONCLUSION: Differential protein production by planktonic and biofilm pneumococci provides a potential explanation for why individuals remain susceptible to invasive disease despite previous colonization events. These findings also strongly suggest that differential protein production during colonization and disease be considered during the selection of antigens for any future protein vaccine.


Subject(s)
Antigens, Bacterial/immunology , Biofilms/growth & development , Nasopharynx/microbiology , Pneumococcal Infections/microbiology , Proteome/analysis , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/metabolism , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Female , Humans , Immunity, Humoral , Mice , Mice, Inbred BALB C , Phenotype , Streptococcus pneumoniae/growth & development
18.
Microb Pathog ; 51(6): 432-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21896326

ABSTRACT

Production of reactive oxygen species (ROS) resulting from phagocytic NADPH oxidase (NOX2) activity has been reported to contribute to host defense against numerous microbial pathogens. In this study we explored the role of NOX2 production in experimental coccidioidomycosis, a human respiratory disease caused by a soil-borne fungal pathogen. Activated and non-activated macrophages isolated from either NOX2(-/-) knock-out or wild type (WT) mice showed comparable ROS production and killing efficiency in vitro when infected with parasitic cells of Coccidioides. Both mouse strains also revealed similar fungal burden in their lungs and spleen at 7 and 11 days after intranasal challenge with Coccidioides spores, although the NOX2(-/-) mice died earlier than the WT strain. Immunization of the NOX2(-/-) and WT mice with a live, attenuated vaccine strain of Coccidioides also resulted in comparable reduction of the fungal burden in both lungs and spleen. These combined results initially suggested that NOX2 activity and ROS production are not essential for protection against Coccidioides infection. However, the reduced survival of non-vaccinated NOX2(-/-) mice correlated with high, sustained numbers of lung-infiltrated neutrophils on days 7 and 11 postchallenge, an expansion of the regulatory T cell population in infected lungs in the knock-out mice, and elevated concentrations of pro-inflammatory cytokines and chemokines in lung homogenates compared to infected WT mice. Although NOX2-derived ROS appeared to be dispensable for both innate and acquired immunity to pulmonary Coccidioides infection, evidence is presented that NOX2 production plays a role in limiting pathogenic inflammation in this murine model of coccidioidomycosis.


Subject(s)
Coccidioides/pathogenicity , Coccidioidomycosis/immunology , Inflammation/pathology , Lung/immunology , Membrane Glycoproteins/immunology , NADPH Oxidases/immunology , Phagocytes/enzymology , Animals , Coccidioides/immunology , Colony Count, Microbial , Cytokines/metabolism , Disease Models, Animal , Fungal Vaccines/administration & dosage , Fungal Vaccines/immunology , Lung/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/genetics , Phagocytes/immunology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Spleen/microbiology , Survival Analysis , T-Lymphocytes, Regulatory/immunology , Time Factors
19.
Infect Immun ; 79(11): 4511-22, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21859851

ABSTRACT

We have previously reported that C57BL/6 mice vaccinated with a live, attenuated mutant of Coccidioides posadasii, referred to as the ΔT vaccine, are fully protected against pulmonary coccidioidomycosis. This model was used here to explore the nature of vaccine immunity during the initial 2-week period after intranasal challenge. Elevated neutrophil and eosinophil infiltration into the lungs of nonvaccinated mice contrasted with markedly reduced recruitment of these cells in vaccinated animals. The numbers of lung-infiltrated macrophages and dendritic cells showed a progressive increase in vaccinated mice and corresponded with reduction of the lung infection. Concentrations of selected inflammatory cytokines and chemokines were initially higher in lung homogenates of vaccinated mice but then generally decreased at 14 days postchallenge in correlation with containment of the organism and apparent dampening of the inflammation of host tissue. Profiles of cytokines detected in lung homogenates of ΔT-vaccinated mice were indicative of a mixed T helper 1 (Th1)-, Th2-, and Th17-type immune response, a conclusion which was supported by detection of lung infiltration of activated T cells with the respective CD4(+) gamma interferon (IFN-γ)(+), CD4(+) interleukin-5 (IL-5)(+), and CD4(+) IL-17A(+) phenotypes. While Th1 and Th2 immunity was separately dispensed of by genetic manipulation without loss of ΔT vaccine-mediated protection, loss of functional Th17 cells resulted in increased susceptibility to infection in immunized mice. Characterization of the early events of protective immunity to Coccidioides infection in vaccinated mice contributes to the identification of surrogates of immune defense and provides potential insights into the design of immunotherapeutic protocols for treatment of coccidioidomycosis.


Subject(s)
Coccidioidomycosis/prevention & control , Fungal Vaccines/immunology , Signal Transduction/immunology , Th1 Cells/physiology , Th17 Cells/physiology , Th2 Cells/physiology , Animals , Coccidioidomycosis/immunology , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation/immunology , Immunity, Innate , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , Vaccines, Attenuated
20.
J Immunol ; 187(3): 1421-31, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21705621

ABSTRACT

Dimorphic fungi collectively account for 5-10 million new infections annually worldwide. Ongoing efforts seek to clarify mechanisms of cellular resistance to these agents and develop vaccines. A major limitation in studying the development of protective T cells in this group of organisms is the lack of tools to detect, enumerate, and characterize fungus-specific T cells during vaccination and infection. We generated a TCR transgenic mouse (Bd 1807) whose CD4(+) T cells respond to a native epitope in Blastomyces dermatitidis and also in Histoplasma capsulatum. In this study, we characterize the mouse, reveal its applications, and extend our analysis showing that 1807 cells also respond to the related dimorphic fungi Coccidioides posadasii and Paracoccidioides lutzii. On adoptive transfer into vaccinated wild-type mice, 1807 cells become activated, proliferate, and expand in the draining lymph nodes, and they differentiate into T1 effectors after trafficking to the lung upon lethal experimental challenge. Bd 1807 cells confer vaccine-induced resistance against B. dermatitidis, H. capsulatum, and C. posadasii. Transfer of naive 1807 cells at serial intervals postvaccination uncovered the prolonged duration of fungal Ag presentation. Using 1807 cells, we also found that the administration of vaccine only once induced a maximal pool of effector/memory CD4(+) cells and protective immunity by 4 wk after vaccination. The autologous adoptive transfer system described in this study reveals novel features of antifungal immunity and offers a powerful approach to study the differentiation of Ag-specific T cells responsive to multiple dimorphic fungi and the development of CD4(+) T cell memory needed to protect against fungal infection.


Subject(s)
Blastomyces/genetics , Blastomyces/immunology , Fungal Vaccines/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Coccidioidomycosis/genetics , Coccidioidomycosis/immunology , Coccidioidomycosis/pathology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Fungal Vaccines/administration & dosage , Fungal Vaccines/genetics , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Immunodominant Epitopes/metabolism , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Paracoccidioidomycosis/genetics , Paracoccidioidomycosis/immunology , Paracoccidioidomycosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...