Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Biol ; 19(6): e3001247, 2021 06.
Article in English | MEDLINE | ID: mdl-34061822

ABSTRACT

Aspergillus fumigatus is a human fungal pathogen that can cause devastating pulmonary infections, termed "aspergilloses," in individuals suffering immune imbalances or underlying lung conditions. As rapid adaptation to stress is crucial for the outcome of the host-pathogen interplay, here we investigated the role of the versatile posttranslational modification (PTM) persulfidation for both fungal virulence and antifungal host defense. We show that an A. fumigatus mutant with low persulfidation levels is more susceptible to host-mediated killing and displays reduced virulence in murine models of infection. Additionally, we found that a single nucleotide polymorphism (SNP) in the human gene encoding cystathionine γ-lyase (CTH) causes a reduction in cellular persulfidation and correlates with a predisposition of hematopoietic stem cell transplant recipients to invasive pulmonary aspergillosis (IPA), as correct levels of persulfidation are required for optimal antifungal activity of recipients' lung resident host cells. Importantly, the levels of host persulfidation determine the levels of fungal persulfidation, ultimately reflecting a host-pathogen functional correlation and highlighting a potential new therapeutic target for the treatment of aspergillosis.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus fumigatus/pathogenicity , Fungal Proteins/metabolism , Host-Pathogen Interactions , Sulfides/metabolism , A549 Cells , Adult , Animals , Aspergillosis/epidemiology , Aspergillosis/genetics , Aspergillosis/microbiology , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/enzymology , Cystathionine gamma-Lyase/genetics , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Host-Pathogen Interactions/drug effects , Humans , Incidence , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/microbiology , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects , Polymorphism, Single Nucleotide/genetics , THP-1 Cells , Transplant Recipients , Virulence/drug effects , Young Adult
2.
mBio ; 11(1)2020 02 04.
Article in English | MEDLINE | ID: mdl-32019790

ABSTRACT

Aspergillus fumigatus is an opportunistic fungal pathogen that can cause life-threatening invasive lung infections in immunodeficient patients. The cellular and molecular processes of infection during onset, establishment, and progression of A. fumigatus infections are highly complex and depend on both fungal attributes and the immune status of the host. Therefore, preclinical animal models are of paramount importance to investigate and gain better insight into the infection process. Yet, despite their extensive use, commonly employed murine models of invasive pulmonary aspergillosis are not well understood due to analytical limitations. Here, we present quantitative light sheet fluorescence microscopy (LSFM) to describe fungal growth and the local immune response in whole lungs at cellular resolution within its anatomical context. We analyzed three very common murine models of pulmonary aspergillosis based on immunosuppression with corticosteroids, chemotherapy-induced leukopenia, or myeloablative irradiation. LSFM uncovered distinct architectures of fungal growth and degrees of tissue invasion in each model. Furthermore, LSFM revealed the spatial distribution, interaction, and activation of two key immune cell populations in antifungal defense: alveolar macrophages and polymorphonuclear neutrophils. Interestingly, the patterns of fungal growth correlated with the detected effects of the immunosuppressive regimens on the local immune cell populations. Moreover, LSFM demonstrates that the commonly used intranasal route of spore administration did not result in complete intra-alveolar deposition, as about 80% of fungal growth occurred outside the alveolar space. Hence, characterization by LSFM is more rigorous than by previously used methods employing murine models of invasive pulmonary aspergillosis and pinpoints their strengths and limitations.IMPORTANCE The use of animal models of infection is essential to advance our understanding of the complex host-pathogen interactions that take place during Aspergillus fumigatus lung infections. As in the case of humans, mice need to suffer an immune imbalance in order to become susceptible to invasive pulmonary aspergillosis (IPA), the most serious infection caused by A. fumigatus There are several immunosuppressive regimens that are routinely used to investigate fungal growth and/or immune responses in murine models of invasive pulmonary aspergillosis. However, the precise consequences of the use of each immunosuppressive model for the local immune populations and for fungal growth are not completely understood. Here, to pin down the scenarios involving commonly used IPA models, we employed light sheet fluorescence microscopy (LSFM) to analyze whole lungs at cellular resolution. Our results will be valuable to optimize and refine animal models to maximize their use in future research.


Subject(s)
Aspergillus fumigatus/immunology , Host-Pathogen Interactions/immunology , Invasive Pulmonary Aspergillosis/immunology , Lung/immunology , Lung/microbiology , Adrenal Cortex Hormones/administration & dosage , Animals , Aspergillus fumigatus/growth & development , Disease Models, Animal , Drug Therapy , Female , Imaging, Three-Dimensional , Immunosuppressive Agents/administration & dosage , Invasive Pulmonary Aspergillosis/pathology , Leukopenia/chemically induced , Lung/cytology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Neutrophils/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...