Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 256
Filter
1.
Semin Cell Dev Biol ; 89: 91-98, 2019 05.
Article in English | MEDLINE | ID: mdl-29551572

ABSTRACT

The dimorphic fungal pathogen Histoplasma capsulatum takes advantage of the innate immune system, utilizing host macrophages as a proliferative niche while largely avoiding stimulation of signaling host receptors. As a result, innate immune cells are unable to control H. capsulatum on their own. Not all host phagocytes respond to H. capsulatum in the same way, with neutrophils and dendritic cells playing important roles in impeding fungal growth and initiating a protective TH1 response, respectively. Dendritic cells prime T-cell differentiation after internalization of yeasts via VLA-5 receptors and subsequent degradation of the yeasts. Dendritic cell-expressed TLR7 and TLR9 promote a type I interferon response for TH1 polarization. In contrast to dendritic cells, macrophages provide a hospitable intracellular environment. H. capsulatum yeasts enter macrophages via binding to phagocytic receptors. Simultaneously, α-glucan masks immunostimulatory cell wall ß-glucans and a secreted endoglucanase removes exposed ß-glucans to minimize recognition of yeasts by Dectin-1. This review highlights how phagocytes interact with H. capsulatum yeasts and the mechanisms H. capsulatum uses to limit the innate immune response.


Subject(s)
Histoplasma/immunology , Histoplasmosis/immunology , Immunity, Innate , Lymphocyte Activation/immunology , Animals , Cell Differentiation/immunology , Cell Wall/immunology , Cell Wall/microbiology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Histoplasma/pathogenicity , Histoplasmosis/microbiology , Humans , Macrophages/immunology , Macrophages/microbiology , Neutrophils/immunology , Neutrophils/microbiology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
2.
Curr Top Microbiol Immunol ; 422: 157-191, 2019.
Article in English | MEDLINE | ID: mdl-30043340

ABSTRACT

Histoplasmosis, caused by the dimorphic environmental fungus Histoplasma capsulatum, is a major mycosis on the global stage. Acquisition of the fungus by mammalian hosts can be clinically silent or it can lead to life-threatening systemic disease, which can occur in immunologically intact or deficient hosts, albeit severe disease is more likely in the setting of compromised cellular immunity. H. capsulatum yeast cells are highly adapted to the mammalian host as they can effectively survive within intracellular niches in select phagocytic cells. Understanding the biological response by both the host and H. capsulatum will facilitate improved approaches to prevent and/or modify disease. This review presents our current understanding of the major pathogenic mechanisms involved in histoplasmosis.


Subject(s)
Histoplasma/pathogenicity , Histoplasmosis/microbiology , Animals , Histoplasma/immunology , Histoplasmosis/immunology , Humans , Virulence
3.
Cell Microbiol ; 21(10): e13066, 2019 10.
Article in English | MEDLINE | ID: mdl-31173452

ABSTRACT

Free-living amoebae (FLAs) are major reservoirs for a variety of bacteria, viruses, and fungi. The most studied mycophagic FLA, Acanthamoeba castellanii (Ac), is a potential environmental host for endemic fungal pathogens such as Cryptococcus spp., Histoplasma capsulatum, Blastomyces dermatitides, and Sporothrix schenckii. However, the mechanisms involved in this interaction are poorly understood. The aim of this work was to characterize the molecular instances that enable Ac to interact with and ingest fungal pathogens, a process that could lead to selection and maintenance of possible virulence factors. The interaction of Ac with a variety of fungal pathogens was analysed in a multifactorial evaluation that included the role of multiplicity of infection over time. Fungal binding to Ac surface by living image consisted of a quick process, and fungal initial extrusion (vomocytosis) was detected from 15 to 80 min depending on the organism. When these fungi were cocultured with the amoeba, only Candida albicans and Cryptococcus neoformans were able to grow, whereas Paracoccidioides brasiliensis and Sporothrix brasiliensis displayed unchanged viability. Yeasts of H. capsulatum and Saccharomyces cerevisiae were rapidly killed by Ac; however, some cells remained viable after 48 hr. To evaluate changes in fungal virulence upon cocultivation with Ac, recovered yeasts were used to infect Galleria mellonella, and in all instances, they killed the larvae faster than control yeasts. Surface biotinylated extracts of Ac exhibited intense fungal binding by FACS and fluorescence microscopy. Binding was also intense to mannose, and mass spectrometry identified Ac proteins with affinity to fungal surfaces including two putative transmembrane mannose-binding proteins (MBP, L8WXW7 and MBP1, Q6J288). Consistent with interactions with such mannose-binding proteins, Ac-fungi interactions were inhibited by mannose. These MBPs may be involved in fungal recognition by amoeba and promotes interactions that allow the emergence and maintenance of fungal virulence for animals.


Subject(s)
Acanthamoeba castellanii/metabolism , Fungi/pathogenicity , Mannose-Binding Lectin/metabolism , Acanthamoeba castellanii/chemistry , Acanthamoeba castellanii/microbiology , Acanthamoeba castellanii/ultrastructure , Animals , Candida albicans/pathogenicity , Candida albicans/ultrastructure , Concanavalin A/metabolism , Cryptococcus neoformans/pathogenicity , Cryptococcus neoformans/ultrastructure , Histoplasma/pathogenicity , Histoplasma/ultrastructure , Host-Pathogen Interactions , Larva/microbiology , Lepidoptera/microbiology , Mannose/chemistry , Mannose/metabolism , Mannose-Binding Lectin/chemistry , Mass Spectrometry , Microscopy, Electron, Scanning , Paracoccidioides/pathogenicity , Paracoccidioides/ultrastructure , Saccharomyces cerevisiae/pathogenicity , Saccharomyces cerevisiae/ultrastructure , Time Factors , Time-Lapse Imaging , Virulence , Virulence Factors/metabolism
4.
Mycopathologia ; 185(5): 881-892, 2020 Oct.
Article in English | MEDLINE | ID: mdl-31845177

ABSTRACT

Histoplasmosis is considered the most common invasive opportunistic fungal disease in the Americas, with outbreaks and micro-epidemics reported for over 80 years. In Brazil, this disease has been described since 1946, reaching a remarkable incidence in the population, especially during the HIV-AIDS pandemic. In this study, published and unpublished outbreaks and micro-epidemics of histoplasmosis in Brazil were revisited by accessing different database sources and evaluating epidemiological and clinical features. We have found reports spanning 1946-2017, across 10 Brazilian states and with involvement of 370 humans and 2 dogs, and 13 disseminated cases and 3 deaths were reported. Rio de Janeiro had the largest number of outbreaks (n = 20/40; 50%) reported in this study. The majority of outbreaks and micro-epidemics was reported in caves (n = 21/40; 52.5%), followed by reports in abandoned/deactivated sites (n = 6/40; 15%), mines (n = 5/40; 12.5%), chicken coops (n = 4/40; 10%). Histoplasmosis is a serious health issue in Brazil considering the attractive and growing market of ecotourism throughout more than 7000 caves, and all levels of poultry farming activity are important to raise awareness about how dangerous this neglected disease can be and establish ways to decrease exposure to contaminated environmental sources through adequate preventive measures.


Subject(s)
Histoplasma , Histoplasmosis , AIDS-Related Opportunistic Infections/epidemiology , AIDS-Related Opportunistic Infections/microbiology , AIDS-Related Opportunistic Infections/prevention & control , Animals , Brazil/epidemiology , Caves/microbiology , Disease Outbreaks , Dogs , Histoplasma/classification , Histoplasma/isolation & purification , Histoplasma/pathogenicity , Histoplasmosis/epidemiology , Histoplasmosis/microbiology , Histoplasmosis/prevention & control , Histoplasmosis/veterinary , Humans , Incidence , Neglected Diseases/epidemiology , Neglected Diseases/microbiology , Neglected Diseases/prevention & control , Poultry Diseases/epidemiology , Poultry Diseases/microbiology , Zoonoses/epidemiology , Zoonoses/microbiology
5.
Clin Endocrinol (Oxf) ; 90(4): 534-541, 2019 04.
Article in English | MEDLINE | ID: mdl-30656706

ABSTRACT

OBJECTIVE: Detailed studies of Addison's disease resulting from disseminated adrenal histoplasmosis (AH) are not available. We describe the presentation and prognosis of AH and cortisol status before and after antifungal therapy. DESIGN: Single-centre retrospective hospital-based study of 40 consecutive adults with AH [39 males; age (mean ± SD) 53 ± 11 years] was conducted between 2006 and 2018. The median duration of follow-up was 2.5 years (range 0.2-12 years). PATIENTS AND METHODS: AH was diagnosed by bilateral adrenal enlargement on CT scan and presence of Histoplasma by histology and/or culture of biopsied adrenal tissue. All patients received oral itraconazole and, if required, amphotericin B as per guidelines. ACTH-stimulated serum cortisol (normal > 500 nmol/L) was measured in 38 patients at diagnosis and re-tested after one year of antifungal therapy in 21 patients. RESULTS: Seventy-three per cent of patients had primary adrenal insufficiency (PAI) and one-third had an adrenal crisis at presentation. HIV antibody was negative in all patients. Of the 29 patients who completed antifungal therapy, 25 (86%) were in remission at last follow-up. Overall, 8 (20%) patients died: three had a sudden death, four had severe histoplasmosis and one died due to adrenal crisis. No patient with PAI became eucortisolemic on re-testing after one year of antifungal therapy. Of the eight patients with normal cortisol at diagnosis, two developed adrenal insufficiency on follow-up. CONCLUSION: All patients with AH tested negative for HIV antibody. While patients achieved a high rate of clinical remission after antifungal therapy, overall mortality was significant. Cortisol insufficiency did not normalize despite treatment.


Subject(s)
Addison Disease/pathology , Histoplasma/pathogenicity , Histoplasmosis/metabolism , Histoplasmosis/pathology , Addison Disease/blood , Addison Disease/drug therapy , Addison Disease/metabolism , Adult , Amphotericin B/therapeutic use , Antifungal Agents/therapeutic use , Female , Follow-Up Studies , Histoplasma/drug effects , Histoplasmosis/drug therapy , Humans , Hydrocortisone/blood , Itraconazole/therapeutic use , Male , Middle Aged , Prognosis , Retrospective Studies
6.
J Biol Chem ; 292(12): 4801-4810, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28154008

ABSTRACT

Fungal cell walls contain ß-glucan polysaccharides that stimulate immune responses when recognized by host immune cells. The fungal pathogen Histoplasma capsulatum minimizes detection of ß-glucan by host cells through at least two mechanisms: concealment of ß-glucans beneath α-glucans and enzymatic removal of any exposed ß-glucan polysaccharides by the secreted glucanase Eng1. Histoplasma yeasts also secrete the putative glucanase Exg8, which may serve a similar role as Eng1 in removing exposed ß-glucans from the yeast cell surface. Here, we characterize the enzymatic specificity of the Eng1 and Exg8 proteins and show that Exg8 is an exo-ß1,3-glucanase and Eng1 is an endo-ß1,3-glucanase. Together, Eng1 and Exg8 account for nearly all of the total secreted glucanase activity of Histoplasma yeasts. Both Eng1 and Exg8 proteins are secreted through a conventional secretion signal and are modified post-translationally by O-linked glycosylation. Both glucanases have near maximal activity at temperature and pH conditions experienced during infection of host cells, supporting roles in Histoplasma pathogenesis. Exg8 has a higher specific activity than Eng1 for ß1,3-glucans; yet despite this, Exg8 does not reduce detection of yeasts by the host ß-glucan receptor Dectin-1. Exg8 is largely dispensable for virulence in vivo, in contrast to Eng1. These results show that Histoplasma yeasts secrete two ß1,3-glucanases and that Eng1 endoglucanase activity is the predominant factor responsible for removal of exposed cell wall ß-glucans to minimize host detection of Histoplasma yeasts.


Subject(s)
Glucan 1,3-beta-Glucosidase/metabolism , Glucan Endo-1,3-beta-D-Glucosidase/metabolism , Histoplasma/enzymology , Histoplasmosis/microbiology , Histoplasma/metabolism , Histoplasma/pathogenicity , Humans , Substrate Specificity , beta-Glucans/metabolism
7.
FEMS Yeast Res ; 18(8)2018 12 01.
Article in English | MEDLINE | ID: mdl-30101348

ABSTRACT

The fungal APSES protein family of transcription factors is characterized by a conserved DNA-binding motif facilitating regulation of gene expression in fungal development and other biological processes. However, their functions in the thermally dimorphic fungal pathogen Histoplasma capsulatum are unexplored. Histoplasma capsulatum switches between avirulent hyphae in the environment and virulent yeasts in mammalian hosts. We identified five APSES domain-containing proteins in H. capsulatum homologous to Swi6, Mbp1, Stu1 and Xbp1 proteins and one protein found in related Ascomycetes (APSES-family protein 1; Afp1). Through transcriptional analyses and RNA interference-based functional tests we explored their roles in fungal biology and virulence. Mbp1 serves an essential role and Swi6 contributes to full yeast cell growth. Stu1 is primarily expressed in mycelia and is necessary for aerial hyphae development and conidiation. Xbp1 is the only factor enriched specifically in yeast cells. The APSES proteins do not regulate conversion of conidia into yeast and hyphal morphologies. The APSES-family transcription factors are not individually required for H. capsulatum infection of cultured macrophages or murine infection, nor do any contribute significantly to resistance to cellular stresses including cell wall perturbation, osmotic stress, oxidative stress or antifungal treatment. Further studies of the downstream genes regulated by the individual APSES factors will be helpful in revealing their functional roles in H. capsulatum biology.


Subject(s)
Gene Expression Regulation, Fungal , Histoplasma/cytology , Histoplasma/growth & development , Hyphae/cytology , Hyphae/growth & development , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Adhesion , Cell Line , Gene Expression Profiling , Histoplasma/genetics , Histoplasma/pathogenicity , Histoplasmosis/microbiology , Histoplasmosis/pathology , Lung/pathology , Macrophages/microbiology , Mice, Inbred C57BL , RNA Interference , Virulence , Virulence Factors/metabolism
8.
PLoS Genet ; 11(7): e1005395, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26177267

ABSTRACT

Eukaryotic cells integrate layers of gene regulation to coordinate complex cellular processes; however, mechanisms of post-transcriptional gene regulation remain poorly studied. The human fungal pathogen Histoplasma capsulatum (Hc) responds to environmental or host temperature by initiating unique transcriptional programs to specify multicellular (hyphae) or unicellular (yeast) developmental states that function in infectivity or pathogenesis, respectively. Here we used recent advances in next-generation sequencing to uncover a novel re-programming of transcript length between Hc developmental cell types. We found that ~2% percent of Hc transcripts exhibit 5' leader sequences that differ markedly in length between morphogenetic states. Ribosome density and mRNA abundance measurements of differential leader transcripts revealed nuanced transcriptional and translational regulation. One such class of regulated longer leader transcripts exhibited tight transcriptional and translational repression. Further examination of these dually repressed genes revealed that some control Hc morphology and that their strict regulation is necessary for the pathogen to make appropriate developmental decisions in response to temperature.


Subject(s)
Gene Expression Regulation, Fungal , Histoplasma/genetics , Host-Pathogen Interactions/genetics , Transcription, Genetic , Fungal Proteins/biosynthesis , Histoplasma/pathogenicity , Humans , RNA, Messenger/genetics , Ribosomes/genetics , Temperature
10.
Mol Microbiol ; 98(5): 910-929, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26288377

ABSTRACT

Microbial pathogens induce or inhibit death of host cells during infection, with significant consequences for virulence and disease progression. Death of an infected host cell can either facilitate release and dissemination of intracellular pathogens or promote pathogen clearance. Histoplasma capsulatum is an intracellular fungal pathogen that replicates robustly within macrophages and triggers macrophage lysis by unknown means. To identify H. capsulatum effectors of macrophage lysis, we performed a genetic screen and discovered three mutants that grew to wild-type levels within macrophages but failed to elicit host-cell death. Each mutant was defective in production of the previously identified secreted protein Cbp1 (calcium-binding protein 1), whose role in intracellular growth had not been fully investigated. We found that Cbp1 was dispensable for high levels of intracellular growth but required to elicit a unique transcriptional signature in macrophages, including genes whose induction was previously associated with endoplasmic reticulum stress and host-cell death. Additionally, Cbp1 was required for activation of cell-death caspases-3/7, and macrophage death during H. capsulatum infection was dependent on the pro-apoptotic proteins Bax and Bak. Taken together, these findings strongly suggest that the ability of Cbp1 to actively program host-cell death is an essential step in H. capsulatum pathogenesis.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Death , Histoplasma/physiology , Histoplasmosis/microbiology , Macrophages/microbiology , Macrophages/physiology , Virulence Factors/metabolism , Animals , Calcium-Binding Proteins/genetics , Caspases/genetics , Caspases/metabolism , Cells, Cultured , Gene Expression Profiling , Genes, Fungal , Genome, Fungal , Histoplasma/growth & development , Histoplasma/pathogenicity , Mice , Molecular Sequence Data , Mutation , Virulence Factors/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
11.
PLoS Biol ; 11(7): e1001614, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23935449

ABSTRACT

Survival at host temperature is a critical trait for pathogenic microbes of humans. Thermally dimorphic fungal pathogens, including Histoplasma capsulatum, are soil fungi that undergo dramatic changes in cell shape and virulence gene expression in response to host temperature. How these organisms link changes in temperature to both morphologic development and expression of virulence traits is unknown. Here we elucidate a temperature-responsive transcriptional network in H. capsulatum, which switches from a filamentous form in the environment to a pathogenic yeast form at body temperature. The circuit is driven by three highly conserved factors, Ryp1, Ryp2, and Ryp3, that are required for yeast-phase growth at 37°C. Ryp factors belong to distinct families of proteins that control developmental transitions in fungi: Ryp1 is a member of the WOPR family of transcription factors, and Ryp2 and Ryp3 are both members of the Velvet family of proteins whose molecular function is unknown. Here we provide the first evidence that these WOPR and Velvet proteins interact, and that Velvet proteins associate with DNA to drive gene expression. Using genome-wide chromatin immunoprecipitation studies, we determine that Ryp1, Ryp2, and Ryp3 associate with a large common set of genomic loci that includes known virulence genes, indicating that the Ryp factors directly control genes required for pathogenicity in addition to their role in regulating cell morphology. We further dissect the Ryp regulatory circuit by determining that a fourth transcription factor, which we name Ryp4, is required for yeast-phase growth and gene expression, associates with DNA, and displays interdependent regulation with Ryp1, Ryp2, and Ryp3. Finally, we define cis-acting motifs that recruit the Ryp factors to their interwoven network of temperature-responsive target genes. Taken together, our results reveal a positive feedback circuit that directs a broad transcriptional switch between environmental and pathogenic states in response to temperature.


Subject(s)
Histoplasma/pathogenicity , Virulence/physiology , Chromatin Immunoprecipitation , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal/genetics , Gene Expression Regulation, Fungal/physiology , Histoplasma/genetics , Temperature , Virulence/genetics
12.
Mediators Inflamm ; 2016: 5813794, 2016.
Article in English | MEDLINE | ID: mdl-27698545

ABSTRACT

Histoplasma capsulatum is a dimorphic fungus that develops a yeast-like morphology in host's tissue, responsible for the pulmonary disease histoplasmosis. The recent increase in the incidence of histoplasmosis in immunocompromised patients highlights the need of understanding immunological controls of fungal infections. Here, we describe our discovery of the role of endogenous galectin-1 (Gal-1) in the immune pathophysiology of experimental histoplasmosis. All infected wild-type (WT) mice survived while only 1/3 of Lgals1-/- mice genetically deficient in Gal-1 survived 30 days after infection. Although infected Lgals1-/- mice had increased proinflammatory cytokines, nitric oxide (NO), and elevations in neutrophil pulmonary infiltration, they presented higher fungal load in lungs and spleen. Infected lung and infected macrophages from Lgals1-/- mice exhibited elevated levels of prostaglandin E2 (PGE2, a prostanoid regulator of macrophage activation) and prostaglandin E synthase 2 (Ptgs2) mRNA. Gal-1 did not bind to cell surface of yeast phase of H. capsulatum, in vitro, suggesting that Gal-1 contributed to phagocytes response to infection rather than directly killing the yeast. The data provides the first demonstration of endogenous Gal-1 in the protective immune response against H. capsulatum associated with NO and PGE2 as an important lipid mediator in the pathogenesis of histoplasmosis.


Subject(s)
Cytokines/metabolism , Dinoprostone/metabolism , Galectin 1/metabolism , Histoplasma/pathogenicity , Nitric Oxide/metabolism , Animals , Flow Cytometry , Galectin 1/genetics , Histoplasmosis/metabolism , Histoplasmosis/microbiology , Humans , Lung/metabolism , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
PLoS Genet ; 9(9): e1003799, 2013.
Article in English | MEDLINE | ID: mdl-24068964

ABSTRACT

The monosaccharide N-acetylglucosamine (GlcNAc) is a major component of microbial cell walls and is ubiquitous in the environment. GlcNAc stimulates developmental pathways in the fungal pathogen Candida albicans, which is a commensal organism that colonizes the mammalian gut and causes disease in the setting of host immunodeficiency. Here we investigate GlcNAc signaling in thermally dimorphic human fungal pathogens, a group of fungi that are highly evolutionarily diverged from C. albicans and cause disease even in healthy individuals. These soil organisms grow as polarized, multicellular hyphal filaments that transition into a unicellular, pathogenic yeast form when inhaled by a human host. Temperature is the primary environmental cue that promotes reversible cellular differentiation into either yeast or filaments; however, a shift to a lower temperature in vitro induces filamentous growth in an inefficient and asynchronous manner. We found GlcNAc to be a potent and specific inducer of the yeast-to-filament transition in two thermally dimorphic fungi, Histoplasma capsulatum and Blastomyces dermatitidis. In addition to increasing the rate of filamentous growth, micromolar concentrations of GlcNAc induced a robust morphological transition of H. capsulatum after temperature shift that was independent of GlcNAc catabolism, indicating that fungal cells sense GlcNAc to promote filamentation. Whole-genome expression profiling to identify candidate genes involved in establishing the filamentous growth program uncovered two genes encoding GlcNAc transporters, NGT1 and NGT2, that were necessary for H. capsulatum cells to robustly filament in response to GlcNAc. Unexpectedly, NGT1 and NGT2 were important for efficient H. capsulatum yeast-to-filament conversion in standard glucose medium, suggesting that Ngt1 and Ngt2 monitor endogenous levels of GlcNAc to control multicellular filamentous growth in response to temperature. Overall, our work indicates that GlcNAc functions as a highly conserved cue of morphogenesis in fungi, which further enhances the significance of this ubiquitous sugar in cellular signaling in eukaryotes.


Subject(s)
Acetylglucosamine/genetics , Blastomyces/genetics , Candida albicans/genetics , Histoplasma/genetics , Morphogenesis , Acetylglucosamine/metabolism , Blastomyces/pathogenicity , Candida albicans/pathogenicity , Cell Wall/metabolism , Fungi/genetics , Gene Expression Regulation, Fungal , Genome, Fungal , Histoplasma/pathogenicity , Humans , Signal Transduction , Soil Microbiology , Temperature
15.
Cytopathology ; 26(4): 244-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25123524

ABSTRACT

OBJECTIVE: The correct identification of fungal organisms is important for the appropriate clinical management of patients. It becomes difficult in necrotic smears when the tissue response is not clearly discernible. It is difficult to distinguish between histoplasma and cryptococcus in severely necrotic cases, where both appear as variably sized clear refractile haloes. METHODS: Four cases of adrenal necrotic histoplasma infection were studied and the morphology was compared with that of non-necrotic histoplasmosis and cases of cryptococcal infection. Eleven cases were analysed in fine needle aspiration cytology (FNAC) smears. Ziehl-Neelsen (ZN) stain was performed to exclude tuberculosis in necrotic smears. A clinical and serology correlation was performed where available. RESULTS: Necrotic cases of histoplasma infection revealed negative refractile clear haloes similar to those of cryptococcus. Histoplasma showed methylene blue-stained organisms in ZN stains, whereas the cryptococcus cases were negative. Similar methylene blue-stained organisms were seen in non-necrotic histoplasma infection. CONCLUSION: As a result of morphological overlap between cryptococcus and histoplasma, the distinction between the two fungi can be difficult in many cases. ZN staining appears to have a role in the differentiation of these fungi in severely necrotic cases. This observation needs to be validated on a larger number of cases with complete correlation with clinical, serology and treatment records.


Subject(s)
Cryptococcosis/diagnosis , Cryptococcosis/microbiology , Histoplasmosis/diagnosis , Histoplasmosis/microbiology , Adolescent , Adult , Aged , Biopsy, Fine-Needle/methods , Cryptococcosis/pathology , Cryptococcus/pathogenicity , Cytodiagnosis/methods , Female , Histoplasma/pathogenicity , Histoplasmosis/pathology , Humans , Male , Middle Aged
16.
PLoS Pathog ; 8(5): e1002713, 2012.
Article in English | MEDLINE | ID: mdl-22615571

ABSTRACT

In order to establish infections within the mammalian host, pathogens must protect themselves against toxic reactive oxygen species produced by phagocytes of the immune system. The fungal pathogen Histoplasma capsulatum infects both neutrophils and macrophages but the mechanisms enabling Histoplasma yeasts to survive in these phagocytes have not been fully elucidated. We show that Histoplasma yeasts produce a superoxide dismutase (Sod3) and direct it to the extracellular environment via N-terminal and C-terminal signals which promote its secretion and association with the yeast cell surface. This localization permits Sod3 to protect yeasts specifically from exogenous superoxide whereas amelioration of endogenous reactive oxygen depends on intracellular dismutases such as Sod1. While infection of resting macrophages by Histoplasma does not stimulate the phagocyte oxidative burst, interaction with polymorphonuclear leukocytes (PMNs) and cytokine-activated macrophages triggers production of reactive oxygen species (ROS). Histoplasma yeasts producing Sod3 survive co-incubation with these phagocytes but yeasts lacking Sod3 are rapidly eliminated through oxidative killing similar to the effect of phagocytes on Candida albicans yeasts. The protection provided by Sod3 against host-derived ROS extends in vivo. Without Sod3, Histoplasma yeasts are attenuated in their ability to establish respiratory infections and are rapidly cleared with the onset of adaptive immunity. The virulence of Sod3-deficient yeasts is restored in murine hosts unable to produce superoxide due to loss of the NADPH-oxidase function. These results demonstrate that phagocyte-produced ROS contributes to the immune response to Histoplasma and that Sod3 facilitates Histoplasma pathogenesis by detoxifying host-derived reactive oxygen thereby enabling Histoplasma survival.


Subject(s)
Histoplasma/enzymology , Histoplasma/pathogenicity , Histoplasmosis/immunology , Oxidative Stress , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Animals , Histoplasmosis/metabolism , Histoplasmosis/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/microbiology , Phagocytosis , RNA Interference , RNA, Small Interfering , Superoxide Dismutase/biosynthesis
17.
PLoS Pathog ; 8(7): e1002771, 2012.
Article in English | MEDLINE | ID: mdl-22829762

ABSTRACT

Vaccines may help reduce the growing incidence of fungal infections in immune-suppressed patients. We have found that, even in the absence of CD4(+) T-cell help, vaccine-induced CD8(+) T cells persist and confer resistance against Blastomyces dermatitidis and Histoplasma capsulatum. Type 1 cytokines contribute to that resistance, but they also are dispensable. Although the role of T helper 17 cells in immunity to fungi is debated, IL-17 producing CD8(+) T cells (Tc17 cells) have not been investigated. Here, we show that Tc17 cells are indispensable in antifungal vaccine immunity in hosts lacking CD4(+) T cells. Tc17 cells are induced upon vaccination, recruited to the lung on pulmonary infection, and act non-redundantly in mediating protection in a manner that requires neutrophils. Tc17 cells did not influence type I immunity, nor did the lack of IL-12 signaling augment Tc17 cells, indicating a distinct lineage and function. IL-6 was required for Tc17 differentiation and immunity, but IL-1R1 and Dectin-1 signaling was unexpectedly dispensable. Tc17 cells expressed surface CXCR3 and CCR6, but only the latter was essential in recruitment to the lung. Although IL-17 producing T cells are believed to be short-lived, effector Tc17 cells expressed low levels of KLRG1 and high levels of the transcription factor TCF-1, predicting their long-term survival and stem-cell like behavior. Our work has implications for designing vaccines against fungal infections in immune suppressed patients.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fungal Vaccines/immunology , Lung Diseases, Fungal/immunology , Pneumonia/immunology , Th17 Cells/immunology , Animals , Blastomyces/immunology , Blastomyces/pathogenicity , Blastomycosis/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Hepatocyte Nuclear Factor 1-alpha , Histoplasma/immunology , Histoplasma/pathogenicity , Histoplasmosis/immunology , Immunocompromised Host , Immunologic Deficiency Syndromes/immunology , Immunologic Memory/immunology , Interleukin-12/biosynthesis , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Lectins, C-Type/metabolism , Lung/immunology , Lung/microbiology , Lung Diseases, Fungal/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Pneumonia/microbiology , Receptors, CCR6/biosynthesis , Receptors, CCR6/metabolism , Receptors, CXCR3/biosynthesis , Receptors, Immunologic/biosynthesis , Signal Transduction , T Cell Transcription Factor 1/biosynthesis
18.
Eukaryot Cell ; 12(6): 828-52, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23563482

ABSTRACT

Histoplasma capsulatum is a fungal pathogen that infects both healthy and immunocompromised hosts. In regions where it is endemic, H. capsulatum grows in the soil and causes respiratory and systemic disease when inhaled by humans. An interesting aspect of H. capsulatum biology is that it adopts specialized developmental programs in response to its environment. In the soil, it grows as filamentous chains of cells (mycelia) that produce asexual spores (conidia). When the soil is disrupted, conidia aerosolize and are inhaled by mammalian hosts. Inside a host, conidia germinate into yeast-form cells that colonize immune cells and cause disease. Despite the ability of conidia to initiate infection and disease, they have not been explored on a molecular level. We developed methods to purify H. capsulatum conidia, and we show here that these cells germinate into filaments at room temperature and into yeast-form cells at 37°C. Conidia internalized by macrophages germinate into the yeast form and proliferate within macrophages, ultimately lysing the host cells. Similarly, infection of mice with purified conidia is sufficient to establish infection and yield viable yeast-form cells in vivo. To characterize conidia on a molecular level, we performed whole-genome expression profiling of conidia, yeast, and mycelia from two highly divergent H. capsulatum strains. In parallel, we used homology and protein domain analysis to manually annotate the predicted genes of both strains. Analyses of the resultant data defined sets of transcripts that reflect the unique molecular states of H. capsulatum conidia, yeast, and mycelia.


Subject(s)
Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Histoplasma/genetics , Mycelium/genetics , RNA, Fungal/genetics , Spores, Fungal/genetics , Transcriptome , Animals , Fungal Proteins/classification , Fungal Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Histoplasma/metabolism , Histoplasma/pathogenicity , Histoplasmosis/microbiology , Humans , Macrophages/microbiology , Mice , Molecular Sequence Annotation , Mycelium/metabolism , Mycelium/pathogenicity , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spores, Fungal/metabolism , Spores, Fungal/pathogenicity , Virulence
19.
Microbiol Mol Biol Rev ; 88(2): e0007623, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38819148

ABSTRACT

SUMMARYHistoplasmosis is arguably the most common fungal respiratory infection worldwide, with hundreds of thousands of new infections occurring annually in the United States alone. The infection can progress in the lung or disseminate to visceral organs and can be difficult to treat with antifungal drugs. Histoplasma, the causative agent of the disease, is a pathogenic fungus that causes life-threatening lung infections and is globally distributed. The fungus has the ability to germinate from conidia into either hyphal (mold) or yeast form, depending on the environmental temperature. This transition also regulates virulence. Histoplasma and histoplasmosis have been classified as being of emergent importance, and in 2022, the World Health Organization included Histoplasma as 1 of the 19 most concerning human fungal pathogens. In this review, we synthesize the current understanding of the ecological niche, evolutionary history, and virulence strategies of Histoplasma. We also describe general patterns of the symptomatology and epidemiology of histoplasmosis. We underscore areas where research is sorely needed and highlight research avenues that have been productive.


Subject(s)
Genetic Variation , Histoplasma , Histoplasmosis , Histoplasma/genetics , Histoplasma/pathogenicity , Histoplasmosis/microbiology , Humans , Virulence/genetics , Animals , Genotype
20.
mSphere ; 9(6): e0000924, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38771035

ABSTRACT

Histoplasmosis is an endemic mycosis that often presents as a respiratory infection in immunocompromised patients. Hundreds of thousands of new infections are reported annually around the world. The etiological agent of the disease, Histoplasma, is a dimorphic fungus commonly found in the soil where it grows as mycelia. Humans can become infected by Histoplasma through inhalation of its spores (conidia) or mycelial particles. The fungi transition into the yeast phase in the lungs at 37°C. Once in the lungs, yeast cells reside and proliferate inside alveolar macrophages. Genomic work has revealed that Histoplasma is composed of at least five cryptic phylogenetic species that differ genetically. Three of those lineages have received new names. Here, we evaluated multiple phenotypic characteristics (colony morphology, secreted proteolytic activity, yeast size, and growth rate) of strains from five of the phylogenetic species of Histoplasma to identify phenotypic traits that differentiate between these species: Histoplasma capsulatum sensu stricto, Histoplasma ohiense, Histoplasma mississippiense, Histoplasma suramericanum, and an African lineage. We report diagnostic traits for three species. The other two species can be identified by a combination of traits. Our results suggest that (i) there are significant phenotypic differences among the cryptic species of Histoplasma and (ii) those differences can be used to positively distinguish those species in a clinical setting and for further study of the evolution of this fungal pathogen.IMPORTANCEIdentifying species boundaries is a critical component of evolutionary biology. Genome sequencing and the use of molecular markers have advanced our understanding of the evolutionary history of fungal pathogens, including Histoplasma, and have allowed for the identification of new species. This is especially important in organisms where morphological characteristics have not been detected. In this study, we revised the taxonomic status of the four named species of the genus Histoplasma, H. capsulatum sensu stricto (ss), H. ohiense, H. mississippiense, and H. suramericanum, and propose the use of species-specific phenotypic traits to aid their identification when genome sequencing is not available. These results have implications not only for evolutionary study of Histoplasma but also for clinicians, as the Histoplasma species could determine the outcome of disease and treatment needed.


Subject(s)
Histoplasma , Histoplasmosis , Phenotype , Phylogeny , Histoplasma/genetics , Histoplasma/classification , Histoplasma/pathogenicity , Histoplasma/isolation & purification , Histoplasmosis/microbiology , Humans , Genome, Fungal
SELECTION OF CITATIONS
SEARCH DETAIL