23 research outputs found

    Aspergillus fumigatus Trehalose-Regulatory Subunit Homolog Moonlights To Mediate Cell Wall Homeostasis through Modulation of Chitin Synthase Activity

    Get PDF
    Trehalose biosynthesis is found in fungi but not humans. Proteins involved in trehalose biosynthesis are essential for fungal pathogen virulence in humans and plants through multiple mechanisms. Loss of canonical trehalose biosynthesis genes in the human pathogen Aspergillus fumigatus significantly alters cell wall structure and integrity, though the mechanistic link between these virulence-associated pathways remains enigmatic. Here we characterize genes, called tslAand tslB, which encode proteins that contain domains similar to those corresponding to trehalose-6-phosphate phosphatase but lack critical catalytic residues for phosphatase activity. Loss of tslA reduces trehalose content in both conidia and mycelia, impairs cell wall integrity, and significantly alters cell wall structure. To gain mechanistic insights into the role that TslA plays in cell wall homeostasis, immunoprecipitation assays coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS) were used to reveal a direct interaction between TslA and CsmA, a type V chitin synthase enzyme. TslA regulates not only chitin synthase activity but also CsmA sub-cellular localization. Loss of TslA impacts the immunopathogenesis of murine invasive pulmonary aspergillosis through altering cytokine production and immune cell recruitment. In conclusion, our data provide a novel model whereby proteins in the trehalose pathway play a direct role in fungal cell wall homeostasis and consequently impact fungus-host interactions

    IL-1α Signaling Is Critical for Leukocyte Recruitment after Pulmonary Aspergillus fumigatus Challenge

    Get PDF
    Aspergillus fumigatus is a mold that causes severe pulmonary infections. Our knowledge of how A. fumigatus growth is controlled in the respiratory tract is developing, but still limited. Alveolar macrophages, lung resident macrophages, and airway epithelial cells constitute the first lines of defense against inhaled A. fumigatus conidia. Subsequently, neutrophils and inflammatory CCR2+ monocytes are recruited to the respiratory tract to prevent fungal growth. However, the mechanism of neutrophil and macrophage recruitment to the respiratory tract after A. fumigatus exposure remains an area of ongoing investigation. Here we show that A. fumigatus pulmonary challenge induces expression of the inflammasome-dependent cytokines IL-1β and IL-18 within the first 12 hours, while IL-1α expression continually increases over at least the first 48 hours. Strikingly, Il1r1-deficient mice are highly susceptible to pulmonary A. fumigatus challenge exemplified by robust fungal proliferation in the lung parenchyma. Enhanced susceptibility of Il1r1-deficient mice correlated with defects in leukocyte recruitment and anti-fungal activity. Importantly, IL-1α rather than IL-1β was crucial for optimal leukocyte recruitment. IL-1α signaling enhanced the production of CXCL1. Moreover, CCR2+ monocytes are required for optimal early IL-1α and CXCL1 expression in the lungs, as selective depletion of these cells resulted in their diminished expression, which in turn regulated the early accumulation of neutrophils in the lung after A. fumigatus challenge. Enhancement of pulmonary neutrophil recruitment and anti-fungal activity by CXCL1 treatment could limit fungal growth in the absence of IL-1α signaling. In contrast to the role of IL-1α in neutrophil recruitment, the inflammasome and IL-1β were only essential for optimal activation of anti-fungal activity of macrophages. As such, Pycard-deficient mice are mildly susceptible to A. fumigatus infection. Taken together, our data reveal central, non-redundant roles for IL-1α and IL-1β in controlling A. fumigatus infection in the murine lung

    IL-1α Signaling Is Critical for Leukocyte Recruitment after Pulmonary Aspergillus fumigatus Challenge

    Get PDF
    Aspergillus fumigatus is a mold that causes severe pulmonary infections. Our knowledge of how A. fumigatus growth is controlled in the respiratory tract is developing, but still limited. Alveolar macrophages, lung resident macrophages, and airway epithelial cells constitute the first lines of defense against inhaled A. fumigatus conidia. Subsequently, neutrophils and inflammatory CCR2+ monocytes are recruited to the respiratory tract to prevent fungal growth. However, the mechanism of neutrophil and macrophage recruitment to the respiratory tract after A. fumigatus exposure remains an area of ongoing investigation. Here we show that A. fumigatus pulmonary challenge induces expression of the inflammasome-dependent cytokines IL-1β and IL-18 within the first 12 hours, while IL-1α expression continually increases over at least the first 48 hours. Strikingly, Il1r1-deficient mice are highly susceptible to pulmonary A. fumigatus challenge exemplified by robust fungal proliferation in the lung parenchyma. Enhanced susceptibility of Il1r1-deficient mice correlated with defects in leukocyte recruitment and anti-fungal activity. Importantly, IL-1α rather than IL-1β was crucial for optimal leukocyte recruitment. IL-1α signaling enhanced the production of CXCL1. Moreover, CCR2+ monocytes are required for optimal early IL-1α and CXCL1 expression in the lungs, as selective depletion of these cells resulted in their diminished expression, which in turn regulated the early accumulation of neutrophils in the lung after A. fumigatus challenge. Enhancement of pulmonary neutrophil recruitment and anti-fungal activity by CXCL1 treatment could limit fungal growth in the absence of IL-1α signaling. In contrast to the role of IL-1α in neutrophil recruitment, the inflammasome and IL-1β were only essential for optimal activation of anti-fungal activity of macrophages. As such, Pycard-deficient mice are mildly susceptible to A. fumigatus infection. Taken together, our data reveal central, non-redundant roles for IL-1α and IL-1β in controlling A. fumigatus infection in the murine lung

    Host-Derived Leukotriene B4 Is Critical for Resistance against Invasive Pulmonary Aspergillosis

    No full text
    Aspergillus fumigatus is a mold that causes severe pulmonary infections. Our knowledge of how immune competent hosts maintain control of fungal infections while constantly being exposed to fungi is rapidly emerging. It is known that timely neutrophil recruitment to and activation in the lungs is critical to the host defense against development of invasive pulmonary aspergillosis, but the inflammatory sequelae necessary remains to be fully defined. Here, we show that 5-Lipoxygenase (5-LO) and Leukotriene B4 (LTB4) are critical for leukocyte recruitment and resistance to pulmonary A. fumigatus challenge in a fungal-strain-dependent manner. 5-LO activity was needed in radiosensitive cells for an optimal anti-fungal response and in vivo LTB4 production was at least partially dependent on myeloid-derived hypoxia inducible factor-1α. Overall, this study reveals a role for host-derived leukotriene synthesis in innate immunity to A. fumigatus

    Induction of Antiviral Immune Response through Recognition of the Repeating Subunit Pattern of Viral Capsids Is Toll-Like Receptor 2 Dependent

    No full text
    Although viruses and viral capsids induce rapid immune responses, little is known about viral pathogen-associated molecular patterns (PAMPs) that are exhibited on their surface. Here, we demonstrate that the repeating protein subunit pattern common to most virus capsids is a molecular pattern that induces a Toll-like-receptor-2 (TLR2)-dependent antiviral immune response. This early antiviral immune response regulates the clearance of subsequent bacterial superinfections, which are a primary cause of morbidities associated with influenza virus infections. Utilizing this altered susceptibility to subsequent bacterial challenge as an outcome, we determined that multiple unrelated, empty, and replication-deficient capsids initiated early TLR2-dependent immune responses, similar to intact influenza virus or murine pneumovirus. These TLR2-mediated responses driven by the capsid were not dependent upon the capsid’s shape, size, origin, or amino acid sequence. However, they were dependent upon the multisubunit arrangement of the capsid proteins, because unlike intact capsids, individual capsid subunits did not enhance bacterial clearance. Further, we demonstrated that even a linear microfilament protein built from repeating protein subunits (F-actin), but not its monomer (G-actin), induced similar kinetics of subsequent bacterial clearance as did virus capsid. However, although capsids and F-actin induced similar bacterial clearance, in macrophages they required distinct TLR2 heterodimers for this response (TLR2/6 or TLR2/1, respectively) and different phagocyte populations were involved in the execution of these responses in vivo. Our results demonstrate that TLR2 responds to invading viral particles that are composed of repeating protein subunits, indicating that this common architecture of virus capsids is a previously unrecognized molecular pattern

    Filamentous fungal carbon catabolite repression supports metabolic plasticity and stress responses essential for disease progression

    No full text
    Aspergillus fumigatus is responsible for a disproportionate number of invasive mycosis cases relative to other common filamentous fungi. While many fungal factors critical for infection establishment are known, genes essential for disease persistence and progression are ill defined. We propose that fungal factors that promote navigation of the rapidly changing nutrient and structural landscape characteristic of disease progression represent untapped clinically relevant therapeutic targets. To this end, we find that A. fumigatus requires a carbon catabolite repression (CCR) mediated genetic network to support in vivo fungal fitness and disease progression. While CCR as mediated by the transcriptional repressor CreA is not required for pulmonary infection establishment, loss of CCR inhibits fungal metabolic plasticity and the ability to thrive in the dynamic infection microenvironment. Our results suggest a model whereby CCR in an environmental filamentous fungus is dispensable for initiation of pulmonary infection but essential for infection maintenance and disease progression. Conceptually, we argue these data provide a foundation for additional studies on fungal factors required to support fungal fitness and disease progression and term such genes and factors, DPFs (disease progression factors)

    CCR2<sup>+</sup> monocyte regulate early IL-1α and CXCL1 expression.

    No full text
    <p>C57BL/6 or CCR2-depleter mice were treated i.p. with 250 ng of DT 24 h prior to challenge with 5×10<sup>7</sup> Af293 conidia. <b>(A)</b> Naïve C57BL/6 or CCR2-depleter mice or C57BL/6 or CCR2-depleter mice challenged eight hours prior were euthanized and the BALF and lung tissue collected for flow cytometric analysis to assess depletion of target cells by DT. Plots are gated on CD45<sup>+</sup> CD11b<sup>+</sup> cells and show Ly6c and Ly6g staining, which identify the CCR2<sup>+</sup> monocytes and neutrophils, respectively. <b>(B)</b> IL-1α and <b>(C)</b> CXCL1 protein levels in the lung parenchyma at 8 h post-challenge with 5×10<sup>7</sup> conidia of <i>A. fumigatus</i> strain Af293 were measured using ELISA assays. Bar graphs show the group means ± one SEM. <b>(D)</b> Eight hours post-challenge with 5×10<sup>7</sup> conidia of <i>A. fumigatus</i> strain Af293, neutrophils in the BALF were enumerated. Data are representative <b>(B-C)</b> or pooled <b>(D)</b> from two independent experiments consisting of 4 mice per group. Each symbol represents an individual mouse and the line represents the group mean. Statistically significant differences were determined using a one-way ANOVA with Bonferroni’s post-test compared C57BL/6 mice (*p < 0.05, **p < 0.01).</p

    CreA regulates CCR in <i>A</i>. <i>fumigatus</i>.

    No full text
    <p>A) Growth of wild type, Δ<i>creA</i>, and <i>creA</i><sup><i>R</i></sup> on 1% glucose minimal media with or without 0.1% allyl alcohol (AA) incubated for 48 hours. B) Growth on 1% glucose or 1% ethanol minimal media for 72 hours.</p

    Model for role of CreA in disease progression of invasive aspergillosis.

    No full text
    <p>Model of disease progression, where upon infection initiation, the presence of oxygen and alternative carbon sources allows for gluconeogenesis and oxidative phosphorylation. However increased fungal growth and influx of host immune cells results in depletion of local oxygen concentration, shifting metabolism towards glycolytic fermentative metabolism. Fungal metabolic adaptation is required for progression to invasive disease, and this requires CreA to mediate disease progression. We propose the concept of disease progression factors (DPFs) as factors required to navigate the dynamic microenvironments that occur during infection and disease progression.</p

    Treatment of <i>Il1r1</i>-deficient mice with CXCL1 partially increases resistance to <i>Aspergillus fumigatus</i> infection.

    No full text
    <p>C57BL/6 mice and <i>Il1r1</i>-deficient mice were challenged i.t. with 5×10<sup>7</sup> CEA10 conidia. Three hours post-challenge mice were given 0.5 μg CXCL1 i.t. or PBS alone. Twenty-four hours post-infection, mice were euthanized, BALF collected, and lungs saved for histological analysis. <b>(A)</b> Formalin-fixed lungs were paraffin embedded, sectioned and stained with H&E (top) or GMS (bottom) for analysis by microscopy. Representative lung sections from <i>Il1r1</i>-deficient mice challenged with CEA10 for 48 h and treated with either PBS or CXCL1 are shown using either the 4× (left) or 20× (right) objectives. <b>(B)</b><i>A. fumigatus</i> germination rates were assessed at 48 h of infection by microscopically counting both the number of conidia and number of germlings in GMS-stained section. Number of conidia and number of germlings were counted for each GMS-stained section to quantify the percent germination. <b>(C)</b> Macrophage and neutrophil recruitment in <i>Il1r1</i>-deficient mice 24 h post-challenge infected with <i>A. fumigatus</i> treated with PBS or CXCL1 given i.t. was determined via cytospins.<b>(D)</b> Bone marrow neutrophils from C57BL/6 and <i>Il1r1</i>-deficient mice were incubated with CEA10 germlings <i>in vitro</i> at a 10:1 ratio in normoxia for 2 h. The XTT assay was used to determine percent fungal damage. <b>(E)</b> Lung damage and <b>(F)</b> leakage were assessed by measuring LDH and albumin, respectively. Data is representative of at least two independent experiments consisting of three to five mice per group, except for the bone marrow neutrophil anti-hyphal XTT assay which is a single experiment which consisted of pooled bone marrow neutrophils from three mice done in triplicate. Each symbol represents an individual mouse or replicate and the line represents the group mean. Statistically significant differences were determined using a one-way ANOVA with Bonferroni’s post-test (*p < 0.05, **p < 0.01, ***p < 0.001, ns = not significant).</p
    corecore