INTRODUCTION
Fungal pathogen-related infections are now estimated to result in a higher number of human deaths than tuberculosis or malaria alone (1–3). The majority of systemic fungal infections are caused by Candida spp., Pneumocystis spp., Cryptococcus spp., and Aspergillus spp. (4, 5). Of the hundreds of known Aspergillus spp., only a few cause disease in animals, with the most prominent being
The primary route of infection of Aspergillus spp. is via the inhalation of conidia (asexual spores). In immunocompetent individuals, inhaled conidia are rapidly cleared by pulmonary resident and recruited neutrophils and macrophages, together preventing the onset of infection (8–10). However, disturbances to the immune system may render an individual susceptible to infection by Aspergillus spp. (11). The severity of infection largely depends on fungal species and genotype, the host immunological status, and host lung structure (6). Invasive aspergillosis (IA) is the most severe disease caused by Aspergillus spp. and is characterized by systemic host invasion, resulting in high mortality rates (30 to 95%) (2, 10, 11).
Patient populations with a highest risk of IA are (i) those with prolonged neutropenia from intensive myeloablative chemotherapy, (ii) cancer patients who are immunosuppressed due to chemotherapy and/or radiotherapy, (iii) those with cystic fibrosis, a hereditary disease that affects the lungs, (iv) and those with genetic disorders resulting in primary immune deficiencies, such as chronic granulomatous disease (CGD) (12, 13). CGD is a genetic disorder that affects 1 in 250,000 people, and in ∼80% of all cases subjects are of the male sex. CGD is caused by mutations in the genes encoding any of the five structural components of the NADPH-oxidase complex, an enzyme complex important for superoxide anion and downstream reactive oxygen species (ROS) production in phagocytic cells (14). As a result, immune cells are unable to efficiently kill microorganisms, and these microorganisms can then become pathogenic in such patients (13, 14)
Although
The aim of this work was to carry out a detailed molecular, phenotypic, and virulence characterization of two
RESULTS
Fungal metabolic plasticity, which allows growth in unique and diverse ambient and host microenvironments, has long been hypothesized to contribute to Aspergillus virulence, with carbon sources such as glucose (22), ethanol (23), and acetate (24) being predicted to be actively used during in vivo infection. In addition, fatty acids and lipids are also thought to serve as major nutrient sources during mammalian host colonization, as is evident by the importance of key glyoxylate cycle enzymes in fungal virulence (25). We therefore characterized growth by determining the fungal dry weight of the two
FIG 1
The
Metabolic profiles differ among the
To further investigate nutrient utilization in the
TABLE 1
Number and percentage of identified metabolite quantities that were significantly different in the
Carbon source (n)b | No. (%) of differentially produced metabolites | |
---|---|---|
MO80069 vs FGSC A4 | SP-2605-48 vs FGSC A4 | |
Glucose (40) | 18 (45) | 15 (38) |
Ethanol (40) | 22 (55) | 23 (58) |
Acetate (44) | 23 (52) | 30 (68) |
Mucin (44) | 24 (55) | 14 (32) |
a
Metabolite quantities in
b
n, number of metabolites tested.
When we further focused on metabolites that were significantly different in quantity between the CIs and the reference strain, we observed that in the presence of glucose and ethanol, the majority of identified metabolites were present in significantly lower quantities than in the reference strain whereas both CIs had significantly higher metabolite quantities in the presence of acetate than the reference strain (Fig. 2A to C). Furthermore, when the
FIG 2
The
When the CIs were grown in a glucose-rich MM, amino acids were found in lower quantities in both CIs than in the reference strain. In contrast, pentose phosphate pathway (PPP) intermediates, glycerol, glycerol derivatives, and aromatic amino acids were detected in significantly higher quantities in this carbon source (Fig. 2A). In an ethanol-rich MM, significantly lower quantities of various amino acids as well as of the citric acid cycle intermediate citrate were detected in the CIs whereas increased quantities of several amino acid pathway intermediates, the carbon compounds glycerol, mannitol, and trehalose, PPP intermediates, and lactate were detected in the CIs compared to levels in the reference strain grown in this carbon source (Fig. 2C). In acetate-rich MM, most identified metabolites, notably a variety of amino acids, were present in significantly larger amounts in the CIs than in the reference strain, with the exception of some amino acids, PPP intermediates, spermidine, rhamnose, and urea (Fig. 2B). When strains were grown in mucin-rich MM, differences in the quantities of a variety of amino acids were observed, whereas trehalose was present in significantly lower quantities and urea was present in significantly higher quantities in both CIs than in the reference strain (Fig. 2D). In summary, these results suggest significant differences in amino acid biosynthesis and degradation, carbon source storage compounds, and degradation among the different
To determine if any metabolic pathways were specifically enriched in the
TABLE 2
Significant metabolic pathway enrichments
Carbon source | Enriched pathways in: | |
---|---|---|
MO80069 | SP-2605-48 | |
Glucose | Aminoacyl-tRNA biosynthesis, arginine | Aminoacyl-tRNA biosynthesis, arginine |
Acetate | Aminoacyl-tRNA biosynthesis; alanine, | Aminoacyl-tRNA biosynthesis, beta- |
Ethanol | Aminoacyl-tRNA biosynthesis, arginine | Aminoacyl-tRNA biosynthesis, arginine |
The
Due to the significant metabolic differences observed between the CIs and the reference strain in the presence of physiologically relevant carbon sources and given that primary metabolism (carbon source utilization) has been shown to impact virulence factors in opportunistic pathogenic fungi (28, 29), we hypothesized that similar differences could be observed in the presence of physiologically relevant stress conditions. One such virulence factor is the fungal cell wall, which is crucial for protection, interaction with, and modulation or evasion of the host immune system (30). In addition, cell wall polysaccharide composition is dependent on carbon source primary metabolism (28, 29, 31).
The production of reactive oxygen species (ROS), such as H2O2, and subsequent augmentation of cellular oxidative stress are strategies employed by the mammalian immune system to combat potential invading pathogenic microorganisms (14). The
FIG 3
The
In summary, the aforementioned results suggest strain-specific differences in the response to different physiological stress conditions and imply that the two
The
Since both CIs showed increased susceptibility to caspofungin, an echinocandin that is being used as a second-line treatment for fungal infections (33), and to other cell wall-perturbing agents, we expanded our analyses to include two additional antifungal drugs classes. Specifically, we followed guidelines for the diagnosis and management of aspergillosis, which, in most cases, recommends treating aspergillosis with azoles and polyene drugs (11), both of which are known to interfere with the biosynthesis or physicochemical properties of fungal membrane sterols (10). Therefore, we determined the MICs of the azoles voriconazole and posaconazole and the polyene amphotericin B for all three strains. No differences in the MICs among all strains to these drugs was observed (Table 3).
TABLE 3
MICs of voriconazole, posaconazole, and amphotericin B for the
Strain | MIC (μg/ml) | ||
---|---|---|---|
Voriconazole | Posaconazole | Amphotericin B | |
FGSC A4 | 0.25 | 1.0 | 2.0 |
MO80069 | 0.25 | 1.0 | 2.0 |
SP260548 | 0.25 | 1.0 | 2.0 |
Cleistothecium formation is impaired in the
Strains were first crossed with themselves (self-crosses) at 30°C and 37°C, and cleistothecium formation was observed for all strains at both temperatures, except for strain SP-2605-48 at 37°C (Table 4). Density of cleistothecia (number of cleistothecia/square centimeter) also varied between strains in a temperature-dependent manner, with the clinical isolates forming fewer cleistothecia per square centimeter than the reference strain at 30°C and 37°C (Table 4). In addition, no difference in levels of ascospore viability was observed among strains (Table 4).
TABLE 4
Cleistothecium formation and density and ascospore viability resulting from diverse
Temp (°C) | Crossa | Cleistothecium | Cleistothecium density (no. | Ascospore viability (%) |
---|---|---|---|---|
30 | A4 × A4 | Yes | 15.0 ± 0.81 | 91.83 ± 3.53 |
MO × MO | Yes | 7.0 ± 1.35 | 92.83 ± 3.96 | |
SP × SP | Yes | 0.25 ± 0.25 | 89.83 ± 3.51 | |
MO × R21 | Yes | 1.25 ± 0.25 | 94.83 ± 3.85 | |
SP × R21 | No | |||
37 | A4 × A4 | Yes | 9.75 ± 1.43 | 90.67 ± 3.62 |
MO × MO | Yes | 5.25 ± 1.31 | 92.5 ± 2.76 | |
SP × SP | No | |||
MO × R21 | Yes | 5.0 ± 0.40 | 92.5 ± 1.28 | |
SP × R21 | No |
a
A4, FGSC A4 reference strain; MO, MO80069 clinical isolate; SP, SP-2605-48 clinical isolate; R21, R21XR135, paba-deficient strain.
Outcrosses were performed by crossing the pyrG (requirement for uridine and uracil) auxotrophic strains MO80069 and SP-2605-48 with the paba (requirement for para-aminobenzoic acid)-deficient strain R21XR135 (Table 5). Strain MO80069 produced cleistothecia at both 30 and 37°C whereas strain SP-2605-48 did not produce any cleistothecia under any of the tested conditions. Density of cleistothecia was very low at 30°C (1.25 cleistothecia/cm2) but increased to the same number observed for the self-crosses at 37°C, with high ascospore viability in all cases (Table 4).
TABLE 5
Strains used in this study
Strain | Genotype | Source | Reference |
---|---|---|---|
FGSC-A4 | Glasgow wild type (veA+) | Soil | 36 |
MO80069 | Wild type, clinical | Bronchoalveolar lavage | This study |
SP-2605-48 | Wild type, clinical | Patient with cystic fibrosis | This study |
R21XR135 | pabaA1 yA2 | NAa | This study |
MO80069 pyrG− | pyrG89 | This study | This study |
SP-2605-48 pyrG− | pyrG89 | This study | This study |
ΔmpkA strain | ΔakuB mpkA::ptrA PTR | NA | 75 |
a
NA, not applicable.
Identification of SNPs and copy number variations in the
The aforementioned phenotyping and metabolomics results indicate differences between the strains that affect traits such as nutrient source utilization and resistance to different stresses. These results are in agreement with studies in
The genomes of MO80069 and SP-2605-48 aligned at 98.3% and 97.4%, respectively, to the genome of the reference strain FGSC A4, with 99.8% nucleotide identity. On the other hand, 1.5% and 1.9% of the A4 assembled genome did not align to the MO80069 and SP-2605-48 genomes, respectively, indicating differences among the genomes of all three strains.
A total of 12,956 and 12,399 SNPs with respect to the A4 reference genome were detected in the genomes of MO80069 and SP-2605-48, respectively (Table 6; see also Table S2 at https://doi.org/10.6084/m9.figshare.11973936). When the genome of SP-260548 was compared to the genome of MO80069, 12,836 SNPs were detected (Table 6; see also Table S2). Each SNP mutation was classified as either high, moderate, or low according to its impact on the DNA codon frame and amino acid sequence. High-impact-type mutations encompass frameshift mutations and stop codon gain/loss, whereas missense mutations, resulting in amino acid changes, are considered moderate-impact-type mutations. Low-impact-type mutations contain all synonymous mutations and mutations within gene introns and untranslated regions (UTRs). The genome of MO80069 contained 501 high-impact mutations, 6,271 missense (moderate impact) mutations, and 6,184 synonymous (low impact) mutations in comparison to the sequence of the reference genome (Table 6; see also Table S2). In the genome of SP-2605-48, 465 high-impact mutations, 5,896 moderate-impact mutations, and 6,038 low-impact mutations were detected in comparison to the sequence of the reference genome (Table 6; see Table S2). When the genomes of both CIs were compared, 426 high-impact mutations, 6,288 missense mutations, and 6,122 synonymous mutations were detected (Table 6; see also Table S2 at the URL mentioned above). All nonsynonymous mutations were distributed throughout the genomes of both CIs, and no clear pattern in mutation accumulation could be observed for any of the 8 chromosomes (Fig. 4 and 5).
TABLE 6
Type and number of SNPs and long indels detected between the genomes of the
Mutation type | No. of mutations | ||
---|---|---|---|
MO80069 vs FGSC A4 | SP-2605-48 vs FGSC A4 | SP-2605-48 vs MO80069 | |
SNPs | |||
Stop codon gain/loss | 149 | 110 | 170 |
Frameshift | 352 | 355 | 256 |
Missense | 6,271 | 5,896 | 6,288 |
Synonymous | 6,184 | 6,038 | 6,122 |
Total | 12,956 | 12,399 | 12,836 |
Indels | |||
Insertions | 234 | 308 | 222 |
Deletion | 114 | 138 | 207 |
Total | 348 | 446 | 375 |
FIG 4
Diagram depicting the location of all detected nonsynonymous single nucleotide polymorphisms (SNPs) on the 8 chromosomes (Chr I to Chr VIII) of the
FIG 5
Diagram depicting the location of all detected small deletions on the 8 chromosomes (Chr I to Chr VIII) of the
In addition, the genomes of both CIs were screened for large-scale (>50 bp) insertions and deletions (indels). In total, 1,169 large-scale indels, consisting of anything between 3 bp to 23 kbp in size, were detected among the eight chromosomes of the CIs compared to the genome of the reference strain (see Table S3). Of these, 348 indels were specifically located in the genome of MO80069, 446 indels were found in the genome of SP-2605-48 only, and 375 indels were located in the genomes of both CIs (Table 6; see also Table S3). The majority of these indels were insertions (Table 6). Of the 375 indels found in the genomes of both CIs, 227 (60.5%) indels differed between the two strains, with the remaining 148 indels being identical for both strains (see Table S3 at the URL mentioned above).
The
As this work aimed to characterize metabolic utilization of physiologically relevant carbon and lipid sources in
Due to the absence of cleistothecium formation in strain SP-2605-48, we wondered whether this strain contained any mutations in genes encoding proteins required for
Last, as both CIs were sensitive to cell wall-perturbing agents, we screened for mutations in genes encoding enzymes involved in cell wall biosynthesis and degradation. Compared to the FGSC A4 reference genome, we found 159 and 90 mutations in 40 and 34 genes involved in cell wall biosynthesis, integrity, and signaling in the genomes of MO80069 and SP-2605-48, respectively (see Table S4 at https://doi.org/10.6084/m9.figshare.11973936). The majority of these mutations were moderate-impact missense mutations in genes that encode components required for 1,3-β- and α-glucan and chitin synthesis and degradation, including various types of glucanases, chitinases, and chitin synthases (see Table S4). However, 17 (MO80069) and 9 (SP-2605-48) mutations were high-impact-level mutations which occurred in genes AN0550 (putative glucan 1,3-beta-glucosidase), AN0509 (putative chitinase), AN0517 (putative chitinase), AN0549 (putative chitinase), AN9042 (putative alpha-1,3-glucanase), AN6324 (putative α-amylase), AN4504 (putative endo-mannanase), and AN0383 (putative endo-mannanase) (see Table S4). In addition, small frameshift mutations were detected in three genes encoding the mitogen-activated protein kinase (MAPK) kinase BckA (AN4887), the MAPK MpkA (AN5666), and the transcription factor RlmA (AN2984) (see Table S4). In
In order to determine whether the observed frameshift mutations had an impact on CWI signaling, we carried out Western blotting of phosphorylated MpkA in the presence of NaCl-induced cell wall stress in all three
FIG 6
MpkA is not phosphorylated in the
The
Due to the observed phenotypic and genotypic differences, we wondered whether the CIs were different in virulence from the reference strain. Virulence was first characterized under a variety of in vitro conditions. Macrophages play an essential role in clearing Aspergillus species conidia from the lung (8), whereas neutrophils are predicted to primarily be responsible for eliminating fungal hyphae (39). To determine whether any strain-specific differences exist in macrophage-mediated phagocytosis and killing, the respective assays were carried out for all three strains in the presence of murine wild-type and gp91phox knockout (CGD) macrophages. Macrophages from CGD patients are impaired in eliminating conidia from the lung environment, thus rendering the host more susceptible to fungal infections (20). Both types of macrophages phagocytosed a significantly higher number of conidia from both
FIG 7
The
When challenged with human polymorphonuclear (PMN) cells, fungal survival was reduced approximately 80% for all three
Virulence of the
We determined the virulence of both
FIG 8
DISCUSSION
The first CI (MO80069) was isolated from a patient with breast carcinoma and pneumonia whereas the second CI (SP-2605-48) was obtained from a patient with cystic fibrosis who underwent lung transplantation. Genome sequencing confirmed these strains to be
In addition, we also assessed the resistance of these strains to a variety of physiologically relevant stress conditions by growing them in the presence of oxidative stress and cell wall stress-inducing compounds, high temperature, iron limitation, and antifungal drugs. Some minor strain-specific differences were observed under these conditions, but the CIs were not significantly more resistant to these conditions, including exposure to azole- and polyene-type anti-fungal drugs, than the reference strain. It is possible that the patient-specific lung environment, biofilm formation, and/or interactions with other microorganisms may result in protection of these stresses, thus resulting in strains that do not have increased stress tolerance. In contrast to
In contrast, both
Last, this work examined the in vivo virulence of the
MATERIALS AND METHODS
Ethics statement.
The principles that guide our studies are based on the Declaration of Animal Rights ratified by the UNESCO on the 27 January 1978 in its 8th and 14th articles. All protocols used in this study were approved by the local ethics committee for animal experiments from Universidade de São Paulo, Campus Ribeirão Preto (permit number 08.1.1277.53.6). All adult and larval zebrafish procedures were in full compliance with NIH guidelines and approved by the University of Wisconsin—Madison Institutional Animal Care and Use Committee (no. M01570-0-02-13).
Strains, media, and growth conditions.
All strains used in this study are listed in Table 5.
Strains were grown in either complete medium (CM) or minimal medium as described previously (57). Iron-poor MM was devoid of all iron and supplemented with 200 μM concentrations of the iron chelators bathophenanthrolinedisulfonic acid (4,7-diphenyl-1,10-phenanthrolinedisulfonic acid [BPS]) and 300 μM 3-(2-pyridyl)-5,6-bis(4-phenylsulfonic acid)-1,2,4-triazine (ferrozine). All growth was carried out at 37°C for the indicated amounts of time, except where otherwise stated (Fig. 1 and 3; see also Fig. S3). Reagents were obtained from Sigma-Aldrich (St. Louis, MO) except where otherwise stated. Radial growth was determined by inoculating plates with 105 spores of each strain and incubation for 5 days before colony diameter was measured. Where required, the oxidative stress-inducing compound menadione or the cell wall-perturbing compounds Congo red (CR), caspofungin, and calcofluor white (CFW) were added in increasing concentrations. All radial growth was expressed as ratios, dividing colony radial diameter (in centimeters) of growth under the stress condition by colony radial diameter under the control (no stress) condition. To determine fungal dry weight, strains were grown from 3 × 106 spores in 30 ml of liquid MM supplemented with 1% (wt/vol) glucose, acetate, mucin, or Casamino Acids or 1% (vol/vol) ethanol, Tween 20 and 80, or olive oil for 48 h (glucose) or 72 h (others) at 37°C and 150 rpm. All liquid and solid growth experiments were carried out in biological triplicates.
Growth in the presence of H2O2 was carried out as serial dilutions (105 to 102 spores) in liquid CM in 24-well plates for 48 h in the presence of different concentrations of H2O2.
Metabolite analysis.
Metabolome analysis was performed as described previously (58). Briefly, metabolites were extracted from 5 mg of dry-frozen, mycelial powder of four biological replicates. The polar phase was dried, and the derivatized sample was analyzed on a Combi-PAL autosampler (Agilent Technologies GmbH, Waldbronn, Germany) coupled to an Agilent 7890 gas chromatograph coupled to a Leco Pegasus 2 time-of-flight mass spectrometer (LECO, St. Joseph, MI). Chromatograms were exported from the Leco ChromaTOF software, version 3.25, to the R software package (www.r-project.org). The Target Search R package was used for peak detection, retention time alignment, and library matching.
Metabolites were quantified by the peak intensity of a selective mass and normalized by dividing the value by the respective sample dry weight. Principal-component analysis was performed using the pcaMethods bioconductor package (59, 60). Pathway enrichment analysis was carried out using MetaboAnalyst (https://www.metaboanalyst.ca/faces/ModuleView.xhtml) (61).
Determination of MICs.
MICs of amphotericin B, voriconazole, and posaconazole were determined by growing 104 spores/well in 96-well plates containing 200 μl/well of RPMI medium and increasing concentrations of the aforementioned compounds, according to the protocol elaborated by the Clinical and Laboratory Standards Institute (62).
Induction of cleistothecium formation.
Cleistothecium formation through self-crossing was induced by growing the strains on glucose minimal medium (GMM) plates that were sealed airtight and incubated for 14 days at 30 or 37°C. Plates were scanned for the presence of cleistothecia under a light microscope. To assess ascospore viability, five cleistothecia of each strain were collected, cleaned on 4% (wt/vol) agar plates, and resuspended in 100 μl of water. Ascospores were counted, and 100 ascospores were plated on GMM before CFU counts were determined. Cleistothecium density was determined through counting the number of cleistothecia of a certain area and dividing the value by the area (in square centimeters).
Cleistothecium formation through outcrossing was carried out as described previously (57). To induce pyrG− auxotrophy in strains MO80069 and SP-2605-48 (Table 1), they were grown on GMM plates supplemented with 1.2 g/liter uridine and uracil (UU) and 0.75 mg/ml 5-fluoroorotic acid (FOA) in the form of a cross until single colonies appeared. Auxotrophy was confirmed by growing strains on GMM with and without UU before strains were crossed with strain R21XR135 (Table 1).
DNA extraction, genome sequencing, and detection of SNPs and indels.
DNA was extracted as described previously (57). Genomes were sequenced using 150-bp Illumina paired-end sequence reads at the Genomic Services Lab of Hudson Alpha (Huntsville, AL). Genomic libraries were constructed with the Illumina TruSeq library kit and sequenced on an Illumina HiSeq 2500 sequencer. Samples were sequenced at greater than 180× coverage or depth.
The Illumina reads were processed with the BBDuk and Tadpole programs of BBMap release 37.34 (https://sourceforge.net/projects/bbmap/files/BBMap_37.34.tar.gz/download) to remove sequencing adapters and phiX and to correct read errors.
Two different Illumina assemblies were performed with the trimmed reads, using platanus (63) and sparseAssembler (64). Nanopore reads were first filtered for quality using Nanofilt (quality of >7) and then were corrected using Canu (65). Once corrected, a subset of reads covering 30 times the estimated genome size of 30 Mb was selected, giving preference to the longest reads. DBG2OLC (66) was used with each of the two Illumina assemblies and the subset of nanopore reads to perform two hybrid assemblies. Independently, MaSuRCA (67) was used to perform a hybrid assembly using the raw nanopore and Illumina reads. The three hybrid assemblies were then corrected using Pilon (68) for three rounds on each assembly. Ragout (69) was then used to fuse the three assemblies into one final assembly using the assembly obtained with MaSuRCA as the base and the other two as references. This assembly was then corrected again for three rounds using Pilon. The mitochondrial genome was obtained from the discarded contigs of MaSuRCA.
The
Detection of large genome deletions and insertions.
Genome assemblies of the two clinical isolates were aligned to the FGSC A4 reference genome with nucmer (70). The alignments were filtered to keep only one-to-one matches. Strain-specific loci were detected by searching the alignment coordinates table for regions of the A4 genome with no match in the clinical isolate genome. Large insertions were detected by searching the alignment coordinate table for regions of the clinical isolate genomes with no match in the A4 genome.
Identification of transposon-like regions in the FGSC A4 reference genome.
Transposon-like regions were identified by running Pfam (71) on the six translation frames of the complete genome sequence. Regions containing any of the 14 domains typically known to be associated with transposable elements (see Table S1 at the URL mentioned above) were collected. Inverted repeats longer than 50 bp and separated by less than 5,000 bp were extracted and marked as potential miniature inverted-repeat transposable elements (MITE). The Pfam and MITE locations were combined to form the transposon track.
Figure generation.
DNAPlotter (72) was used to display the loci of all nonsynonymous SNPs and large deletions identified in the two clinical strains compared to the reference genome of FGSC A4. In addition, the locations of transposon-like regions in the A4 genome were also highlighted using DNAPlotter.
Western blotting.
Strains were grown from 1 × 107 spores at 37°C and 200 rpm in 50 ml of CM for 16 h before being exposed to 0.5 M NaCl for 0, 10, and 30 min. Total cellular proteins were extracted according to Fortwendel and colleagues (73) and quantified according to Hartree (74).
For each sample, 60 μg of total intracellular protein was run on a 12% (wt/vol) SDS-PAGE gel before being transferred to a polyvinylidene difluoride (PVDF) membrane (GE Healthcare). Phosphorylated MpkA or total MpkA was probed for by incubating the membrane with a 1:5,000 dilution of the anti-phospho-p44/42 MAPK (9101; Cell Signaling Technologies) antibody or with a 1:5,000 dilution of the p44-42 MAPK (Cell Signaling Technology) antibody overnight at 4°C, with shaking. Subsequently, membranes were washed three times with TBS-T (2.423 g/liter Tris, 8 g/liter NaCl, 1 ml/liter Tween 20) and incubated with a 1:5,000 dilution of an anti-rabbit IgG horseradish peroxidase (HRP) antibody (7074; Cell Signaling Technologies) for 1 h at room temperature. MpkA was detected by chemiluminescence using a Western ECL Prime (GE Healthcare) blot detection kit according to the manufacturer’s instructions. Films were submitted to densitometric analysis using ImageJ software (http://rsbweb.nih.gov/ij/index.html). The amount of phosphorylated MpkA was normalized by the amount of total MpkA. The
Isolation and differentiation of BMDM.
Bone marrow-derived macrophages (BMDMs) were isolated as described previously (76). Briefly, BMDMs were recovered from femurs of C57BL/6 wild-type and gp91phox knockout mice and were incubated in BMDM medium (RPMI medium [Gibco] supplemented with 30% [vol/vol] L929 growth-conditioning medium, 20% inactivated fetal bovine serum [FBS; Gibco], 2 mM glutamine, and 100 units/ml of penicillin-streptomycin [Life Technologies]). After 4 days, fresh medium was added for an additional 3 days before BMDMs were collected.
In vitro phagocytosis and killing assays.
Phagocytosis and killing assays of
To determine phagocytosis, macrophages were incubated with conidia for 1.5 h before the supernatant was removed, and 500 μl of PBS containing 3.7% formaldehyde was added for 15 min at room temperature (RT). Sample coverslips were washed with 1 ml of ultrapure water and incubated for 20 min with 500 μl of 0.1 mg/ml calcofluor white (CFW) to stain for the cell wall of nonphagocytized conidia. Samples were washed, and coverslips were viewed under a Zeiss Observer Z1 fluorescence microscope. In total, 100 conidia were counted per sample, and the phagocytosis index was calculated. Experiments were performed in biological triplicates.
To determine macrophage-induced killing of conidia, macrophages were incubated with conidia for 1.5 h before cell culture supernatants were collected and cytokine concentrations were determined. Macrophages were then washed twice with PBS to remove all nonadherent cells and subsequently lysed with 250 μl of 3% (vol/vol) Triton X-100 for 10 min at RT. Serial dilutions of lysed samples were performed in sterile PBS and plated onto CM and incubated at 37°C for 2 days before CFU counts were determined.
PMN cell isolation and spore germination assay.
Human polymorphonuclear (PMN) cells from fresh venous blood of healthy adult volunteers were isolated according to Drewniak et al. (78), with modifications. Cells were harvested by centrifugation in isotonic Percoll, lysed, and resuspended in HEPES-buffered saline solution.
In vivo infections in immunocompetent, CGD, and neutrophilic zebrafish.
We evaluated strain virulence in an established zebrafish-aspergillosis model. Seventy-two wild-type larvae were used as an immunocompetent model. Larvae with a dominant negative Rac2D57N mutation in neutrophils (mpx::rac2D57N) (39) were used as a model of leukocyte adhesion deficiency, where neutrophils do not reach the site of infection, and p22phox-deficient larvae [p22phox (sa11798)] were used as a chronic granulomatous disease (CGD) model (21).
Spore preparation and conidium microinjection into the hindbrain of 2-day postfertilization (dpf) larvae were performed as previously described (79). Briefly, after manual dechorionation of embryos, 3 nl of inoculum or PBS-control was injected into the hindbrain ventricle via the optic vesicle (∼50 conidia) in anesthetized larvae at approximately 36 h postfertilization.
In vivo infections in immunocompetent, CGD, and neutropenic mice.
Virulence of the
Mice were anesthetized and submitted to intratracheal (i.t.) infection as previously described (80) with some minor modifications. Briefly, after i.p. injection of ketamine and xylazine, animals were infected with 5.0 × 107 (immunocompetent) or 1 × 106 (CGD) conidia contained in 75 μl of PBS (81) by surgical i.t. inoculation, which allowed dispensing of the fungal conidia directly into the lungs. Neutropenic mice were infected by intranasal instillation of 1.0 × 104 conidia as described previously (70). Phosphate-buffered saline (PBS) was administered as a negative control for each murine model.
Mice were weighed every 24 h from the day of infection and visually inspected twice daily. The endpoint for survival experimentation was identified when a 20% reduction in body weight was recorded, at which time the mice were sacrificed.
Statistical analyses.
All statistical analyses were performed using GraphPad Prism, version 7.00 (GraphPad Software, San Diego, CA), with a P value of < 0.05 considered significantly different. A two-way analysis of variance (ANOVA) was carried out on all stress response tests whereas a one-way ANOVA with Tukey’s posttest was applied for growth in the presence of different carbon sources, for the phagocytosis index, and for the PMN cell killing assay. Survival curves were plotted by Kaplan-Meier analysis, and results were analyzed using a log rank test. All experiments were repeated at least twice.
Data availability.
Short-read sequences for these strains are available in the NCBI Sequence Read Archive (SRA) under accession numbers SRR10983230, SRR10983231, SRR10983232, and SRR10983233 and BioProject number PRJNA603646. Genomes were deposited in GenBank under accession numbers JAAFYM000000000 and JAAFYL000000000.
b Department of Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, Wisconsin, USA
c Department of Pediatrics, University of Wisconsin—Madison, Madison, Wisconsin, USA
d Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
e Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
f Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
g Life and Health Sciences Research Institute/3B’s Associate Laboratory, Guimarães, Portugal
h Laboratory of Clinical Bacteriology and Mycology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
i National Reference Center for Mycosis, University Hospitals Leuven, Leuven, Belgium
j Centre for Genomic Regulation, Barcelona, Spain
k Life Sciences Program, Barcelona Supercomputing Centre, Barcelona, Spain
l Mechanisms of Disease Program, Institute for Research in Biomedicine, Barcelona, Spain
m ICREA, Barcelona, Spain
n Centre for Structural and Functional Genomics, Concordia University, Montreal, Quebec, Canada
o Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
p Instituto de Ciência e Tecnologia, Universidade Federal de São Paulo, São José dos Campos, Brazil
q Department of Biological Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
University of Georgia
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2020 Bastos et al. This work is published under https://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
ABSTRACT
IMPORTANCE Immunocompromised patients are susceptible to infections with opportunistic filamentous fungi from the genus Aspergillus. Although
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer