48
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Evolving moldy murderers: Aspergillus section Fumigati as a model for studying the repeated evolution of fungal pathogenicity

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          “Biology is more like history than it is like physics. You have to know the past to understand the present.”–Carl Sagan (1980) Introduction Species in the genus Aspergillus are saprophytic filamentous fungi that are most commonly found in soil and litter environments of subtropical and warm temperate latitudes [1]. Inhalation of asexual spores produced by Aspergillus fumigatus and a few other species in the genus cause a group of diseases collectively referred to as aspergillosis [2]. The most severe form of aspergillosis is invasive aspergillosis, which primarily affects individuals with compromised immune systems or preexisting lung conditions [3]. Since drugs targeting invasive aspergillosis are not always effective due to our lack of understanding of how they function inside the human host [4] and the evolution of drug resistance [5, 6], infected individuals suffer high morbidity and mortality [7]. Collectively, Aspergillus fungi affect millions of patients and cause hundreds of thousands of life-threatening infections every year [8, 9]. Not all pathogenic Aspergillus species exhibit the same infection rates [10, 11]. Approximately 70% of all Aspergillus infections are caused by A. fumigatus, whereas the remaining 30% of infections stem from other species in the genus [12]. Some of these other pathogenic species–for example, Aspergillus flavus, Aspergillus niger, and Aspergillus terreus–are distantly related to A. fumigatus [12]; each of these three species belong to different Aspergillus sections (note “section” is a taxonomic rank in-between the genus and species ranks) and show extensive genomic divergence [13]. However, there are also pathogenic Aspergillus species, such as Aspergillus lentulus and Aspergillus udagawae, that belong to the same section as A. fumigatus (section Fumigati) and are much more closely related [14–16]. In contrast, the vast majority of Aspergillus species, including many close relatives of the pathogens mentioned above, are either not pathogenic or very rarely cause disease [16, 17]. Interestingly, A. fumigatus and a few other species in the genus can also cause opportunistic infections, mainly in other mammals and birds, and occasionally in other vertebrates and invertebrates [18]. Even though a great deal is known about some aspects of A. fumigatus pathogenicity [2, 19, 20], we have only recently begun to examine why pathogenicity varies so dramatically across the entire genus and the traits and genetic elements that contributed to this variation. Addressing this question requires that we consider the fact that pathogenic Aspergillus species are not dependent on their hosts for survival and their pathogenic effects are entirely accidental or opportunistic. Thus, understanding the evolution of pathogenicity in the genus requires that we understand how variation in the traits that enable Aspergillus species to survive in their natural soil and litter environments has rendered a few of these species capable to establish infections inside human hosts. In this pearl article, we focus on section Fumigati, a lineage of ~60 species that includes A. fumigatus and its close relatives [16, 21], to discuss the latest advances in our understanding of the evolution of pathogenicity in Aspergillus. More broadly, given that the ability to cause human disease has repeatedly evolved across the fungal tree of life, and that the vast majority of human fungal pathogens have non-pathogenic close relatives [22], understanding the evolution of Aspergillus pathogenicity can serve as a model for studying fungal pathogenicity in general. Pathogenicity in Aspergillus section Fumigati fungi has evolved multiple times independently Only a handful of species in section Fumigati are considered pathogenic [10, 16, 21], and the distribution of these pathogens on the section’s phylogeny [16] suggests that the ability to cause human disease has evolved at least 5 times independently ( Fig 1 ). For example, whereas A. fumigatus infects >300,000 humans per year [9], its close evolutionary relative A. fischeri, whose protein sequences exhibit, on average, 95% similarity to their A. fumigatus orthologs, has only rarely been reported to cause human disease and is not considered clinically relevant [23, 24]. Interestingly, evolutionary reconstruction shows that A. fumigatus pathogenicity likely evolved after the species diverged from either A. fischeri or from its even closer non-pathogenic relative A. oerlinghausenensis, suggesting that the last common ancestor of these three species was non-pathogenic ( Fig 1 ). The same is true for most other pathogens in the section. For example, the pathogenic A. udagawae accounts for a few thousand infections per year [25–27], but its close relatives (e.g., Aspergillus aureolus, Aspergillus acrensis, and Aspergillus wyomingensis) are not considered clinically relevant ( Fig 1 ), suggesting that the pathogenicity of A. udagawae evolved independently and that the common ancestor of A. udagawae and its close relatives was non-pathogenic. 10.1371/journal.ppat.1008315.g001 Fig 1 Reconstruction of the ability to cause human disease on the Aspergillus section Fumigati phylogeny suggests that pathogenicity evolved multiple times independently in the lineage. For the trait reconstruction inference, Biosafety Level (BSL) 2 organisms were considered pathogenic and BSL1 organisms or organisms that so far lack BSL labelling were considered non-pathogenic; these transitions to a pathogenic lifestyle (i.e., from BSL1 to BSL2) are labelled by red bars on the figure. Note that clinical isolates from humans or other mammals from a few additional species in the section have been identified [10, 28]; this handful includes relatively newly described species that some authors consider to have pathogenic potential (e.g., Aspergillus novofumigatus [29]) as well as organisms thought to be on the non-pathogenic end of the spectrum (e.g., A. fischeri [30, 31]). The phylogeny of the section was redrawn from Hubka et al. [16]. The observed spectrum of pathogenicity cannot be explained by ecology or ascertainment bias and is likely to have a genetic component Several ecological attributes, such as the global ubiquity of their small and readily airborne asexual spores, are thought to contribute to the pathogenicity of Aspergillus molds in general [11], and to the pathogenicity of A. fumigatus in particular [19, 32]. Although these ecological attributes are undoubtedly important for infecting and causing disease in a human host, the observed spectrum of pathogenicity among section Fumigati species cannot be solely explained by the known differences in species’ ecologies [1, 33–35]. For example, previous studies have shown that A. fischeri, one of the closest non-pathogenic relatives of A. fumigatus, can be frequently isolated from a variety of locales, including soils, fruits, and hospitals [33–35]. Case in point, approximately 2% of the fungi isolated from the Beijing Hospital environment were A. fischeri [34], but only a handful of infections caused by this fungus have ever been reported [10, 23, 24]. Another ecological attribute known to be associated with fungal pathogenicity is thermal tolerance [36]. However, all species in section Fumigati that have been tested can grow, dependent on the growth medium used, at 37°C [14]. These data suggest that pathogenicity in Aspergillus section Fumigati is not simply due to species’ abilities to grow at the human body temperature. Nevertheless, species in the section do show substantial differences in how well they can grow at 37°C [14], but these differences are likely to have a genetic basis (see below). It would be highly interesting for future studies to examine growth curves of closely related pathogenic and non-pathogenic species in specific stressful and human infection-relevant conditions (e.g., at 37°C, with limited nutrient availability, low levels of oxygen and high levels of oxidative stress). Another potential explanation for some of the observed differences in the spectrum of pathogenicity among species is ascertainment bias. In the context of Aspergillus pathogenicity, ascertainment bias is a term that describes systematic deviations from the true incidence of disease caused by a given species. These systematic deviations stem from the methods used to taxonomically identify (ascertain) clinical isolates and estimate how often they cause disease, i.e., from our failure to measure the true numbers of infections caused by so-called cryptic species, namely organisms that are morphologically similar to major pathogens, such as A. fumigatus, but genetically distinct from them [10]. The true burden some of these cryptic species, including species currently thought to not be clinically relevant, place on human health is unknown and may be in several cases underestimated [22]. However, numerous molecular typing studies of clinical isolates from diverse countries routinely identify the known pathogens in section Fumigati (Fig 1), but not the non-pathogens [25–27], indicating that the observed variation in pathogenicity is not solely an artifact of species misdiagnosis. These data suggest that the differences in pathogenicity observed across Aspergillus section Fumigati have, at least partially, a genetic basis. Support for the role of genetic differences in contributing to the observed spectrum of pathogenicity is provided by the numerous traits, and their underlying genes and pathways, that are required for pathogenicity in A. fumigatus [2, 19, 20, 37] and have been found to exhibit substantial genetic and phenotypic diversity among section Fumigati species. These traits include thermotolerance, the ability to respond to multiple environmental stresses, including antifungal drugs, and the capacity to biosynthesize a range of structurally diverse secondary metabolites [14, 30, 38, 39]. Two models for the evolution of pathogenicity in Aspergillus molds One useful approach for gaining insights into the genetic foundations of the multiple, independent origins of pathogenicity in Aspergillus section Fumigati is the development of conceptual models that describe the differences that we would expect to observe in genomic comparisons involving pathogenic and non-pathogenic species. We propose two alternative, although not necessarily mutually exclusive, models, which we have named the “conserved pathogenicity” model and the “species-specific pathogenicity” model ( Fig 2 ). The conserved pathogenicity model posits that A. fumigatus and other pathogenic species in section Fumigati share common pathogenicity traits and genetic elements (or shared differences in genetic elements) that are absent in non-pathogens (e.g., traits / elements E1 and E2 in Fig 2 ) or vice versa (e.g., trait / element E3 in Fig 2 ). In contrast, the species-specific model posits that each pathogen contains a unique suite of traits and genetic elements (or unique differences in genetic elements) that distinguish it from its non-pathogenic relatives; these traits / elements could be ones that are uniquely present in a given pathogen but absent in related pathogens and non-pathogens (e.g., traits / elements E4 and E5 in Fig 2 ) or vice versa (e.g., trait / element E6 in Fig 2 ). Note that these shared or species-specific genetic elements (or differences in genetic elements) are not limited to differences in gene content but to any type of genetic variation that alters pathogenicity trait values. These variants can range from, for example, differences in a single or in a handful of nucleotides within otherwise conserved protein-coding or non-coding (regulatory) regions to larger-scale differences concerning the presence of entire genetic pathways and networks. 10.1371/journal.ppat.1008315.g002 Fig 2 Graphic illustration of the “conserved pathogenicity” and the “species-specific” pathogenicity models. Discerning which model explains the repeated evolution of pathogenicity is key for developing research strategies to understand the underlying molecular mechanisms in the genus and, more broadly, in filamentous fungi and beyond. For example, the conserved pathogenicity model would predict that pathogenicity stems from the action of conserved genetic elements, suggesting that known genetic determinants of virulence in A. fumigatus [40] would be great candidates for involvement in virulence in other pathogenic Aspergillus species. It is the adoption of the conserved pathogenicity model that underlies recent examinations of the degree to which genetic elements known to contribute to A. fumigatus pathogenicity are conserved in other species [29, 30, 41]. In contrast, the species-specific model would predict the opposite, namely that the genetic determinants of virulence are unique to each pathogen, suggesting that extrapolations of knowledge on virulence mechanisms from one pathogenic species to another would be futile. Genomic comparisons between the major pathogen A. fumigatus and its close non-pathogenic relative A. fischeri [30, 31] as well as broad comparisons of select species across the genus [29, 41] have begun to shed light on the validity of, and provide support for, both of these models. Support for the conserved pathogenicity model Consistent with one of the predictions of the conserved pathogenicity model, examinations of dozens of A. fumigatus genes known to be associated with virulence [40] have shown that most of these genetic determinants of virulence are highly conserved in closely related species [29, 30]. For example, a recent genomic comparison of the major pathogen A. fumigatus with its close, non-pathogenic relative A. fischeri showed that 48 of 49 known genetic determinants of A. fumigatus virulence (e.g., CrzA, the C2H2-type zinc finger transcription factor involved in calcium ion homeostasis, or LaeA, a methyltransferase and master regulator of secondary metabolism) were highly conserved in A. fischeri [30]. However, these results also suggested that the differences in virulence among organisms spanning the pathogenicity spectrum may not be primarily due to differences in gene content, which virtually all genomic comparisons of fungal pathogens and non-pathogens in the genus [29–31, 41] and beyond [42–44] have heavily focused on. How these conserved genetic determinants of virulence function in other pathogenic, as well as in non-pathogenic, species is an interesting future direction of inquiry. Support for the species-specific pathogenicity model Comparisons of gene content between closely related Aspergillus species have also identified numerous genes that appear to be species-specific. For example, a broad scale comparison of A. fumigatus strains Af293 and A1163 against A. fischeri and A. clavatus found that approximately 8.5% of genes were unique to A. fumigatus and absent from the other two species [31]. These A. fumigatus-specific genes tend to reside near the ends of chromosomes (i.e., are subtelomeric) and have functions associated with metabolism (e.g., secondary metabolism, transport, and detoxification), raising the hypothesis that some of them may aid A. fumigatus survival inside the human host [31]. For example, more than two thirds of A. fumigatus biosynthetic gene clusters are absent from the closely related non-pathogen A. fischeri [30, 38]; however some of these biosynthetic gene clusters are found in other species in section Fumigati, suggesting that they were lost in A. fischeri rather than originated within A. fumigatus. Several other clusters and their secondary metabolites appear to have uniquely evolved in A. fumigatus, or their loss in species closely related to A. fumigatus was so widespread that they are now uniquely present in A. fumigatus. One recent, striking example in support of the species-specific model was the discovery of the gene hypoxia-responsive morphology factor A or hrmA (Afu5g14900) [45]. While homologs of hmrA are present in other distantly related fungi, this subtelomeric gene is polymorphic within A. fumigatus and is absent from the genomes of all sequenced Aspergillus section Fumigati species. Investigation of hrmA function shows that it likely regulates a cluster of genes, which also appear to be absent from the genomes of other Aspergillus section Fumigati species, that collectively contribute to the generation of a morphotype that facilitates adaptation to very low oxygen conditions encountered by the fungus inside human lungs [45]. Concluding remarks This pearl has focused on Aspergillus section Fumigati, outlining two general models for the repeated evolution of pathogenicity in the section and how they could aid in the design of experiments aimed at elucidating the underlying molecular changes responsible. The same approach could also be employed for developing strategies for the burgeoning problem of drug resistance (e.g., to what extent are mechanisms of drug resistance conserved across pathogenic species?). But the utility of studying the evolution of Aspergillus section Fumigati pathogenicity goes beyond, as Carl Sagan so aptly put it, understanding “the present”. By posing questions such as “are species that are currently considered non-pathogenic but contain conserved genetic determinants of virulence likely to emerge as new pathogens?”, we believe that an evolutionary approach–by identifying the presence of constellations of genes and traits associated with pathogenicity in non-pathogens–also holds promise for predicting the emergence of new pathogens in the future.

          Related collections

          Most cited references36

          • Record: found
          • Abstract: found
          • Article: not found

          Genomic Islands in the Pathogenic Filamentous Fungus Aspergillus fumigatus

          Introduction Aspergillus fumigatus is exceptional amongst the aspergilli in being both a primary and opportunistic pathogen as well as a major allergen associated with severe asthma and sinusitis [1]–[3]. It was first reported to cause opportunistic invasive infection about 50 years ago [4]. In immunocompromised patients, mycelial growth can proliferate throughout pulmonary or other tissues causing invasive aspergillosis. For these patients, the incidence of invasive aspergillosis can be as high as 50% and the mortality rate is often 50%, even with antifungal treatment. Since the late 1800's [2], A. fumigatus has been demonstrated to be a primary pathogen of the airways, sinuses, lungs, damaged skin and subcutaneous tissues. For example, it can cause post-operative infection in all human organs [5]. In most cases diagnosis remains problematic and can compromise effective medical treatment. A. fumigatus is thought to possess particular metabolic capabilities and genetic determinants that allow it to initiate and establish an in vivo infection. This conclusion is supported by the observation that the majority of invasive aspergillosis disease is caused by A. fumigatus, even though its conidia comprise only a small percentage of the total conidia found in air-sampling studies [6]. While the interaction of A. fumigatus spores with the human respiratory mucosa is understood to an extent, the basic biology of the organism has until recently received little attention. Recently we presented the genomic sequence of A. fumigatus strain Af293 (FGSC A1100) [7] isolated from a neutropenic patient, who died from invasive aspergillosis [8]. Its comparison with the genomes of two distantly related species, Aspergillus nidulans and Aspergillus oryzae, has led to many unexpected discoveries, including the possibility of a hidden sexual cycle in A. fumigatus and A. oryzae, and the detection of remarkable genetic variability of this genus [9],[10]. Although members of the same genus, these three species are approximately as evolutionarily distant from each other at the molecular level as humans and fish (Figures 1 and 2) [11]. This significant phylogenetic distance has hindered some aspects of comparative genomic analysis of the aspergilli such as identification of the genetic traits responsible for differences in virulence as well as in sexual and physiological properties. 10.1371/journal.pgen.1000046.g001 Figure 1 Molecular Divergence in Molds and Yeasts. A. fumigatus proteins are compared to their orthologs in N. fischeri, A. clavatus, A. terreus, A. oryzae, A. nidulans, and A. niger (mean values: 95%, 84%, 71%, 71%, 68%, and 69%, respectively). Saccharomyces paradoxus, Saccharomyces uvarum, Candida glabrata, and Kluyveromyces lactis are compared to Saccharomyces cerevisiae (adapted from [74,75]). Mean values for these species are 90%, 82%, 64%, and 60%, respectively. Median percent identity between pairs of orthologs from A. fumigatus and each successive genome in the tree is shown. Relative divergence of humans, mice, birds and fish are shown for reference. 10.1371/journal.pgen.1000046.g002 Figure 2 Three Closely Related Aspergilli. The three most closely related aspergilli, which constitute the Affc-core group (A. fumigatus, N. fischeri, and A. clavatus), are in bold black. The maximum-likelihood tree was constructed from an alignment of 90 proteins chosen on the basis of similar lengths and identical number of intron/exon structures in order to minimize the number of inconvenient or incongruent gene models (see Materials and Methods). To maximize the resolving power of whole-genome comparative analysis, we selected the environmental type strains of a very closely related sexual species, Neosartorya fischeri NRRL181 (A. fischerianus), and a more distantly related asexual species, A. clavatus NRRL1, for complete sequencing. These three species are referred to here as the Affc lineage for A. fumigatus, N. fischeri, and A. clavatus (Figure 2). In contrast to A. fumigatus, N. fischeri is only rarely identified as a human pathogen [12]–[15]; while A. clavatus is probably an important allergen and the causative agent of extrinsic allergic alveolitis known as malt worker's lung [16]. A. clavatus also produces a number of mycotoxins and has been associated with neurotoxicosis in sheep and cattle fed infected grain worldwide (e.g. [17]). Our phenotypic characterization (Table S1) has shown that both A. fumigatus and N. fischeri can grow at 42°C, which indicates that A. fumigatus may possess other genetic determinants besides thermotolerance that allow it to establish a successful in vivo infection. As determined by multilocus sequence comparison, most A. fumigatus isolates, including Af293 and A1163, lie within the main A. fumigatus clade and persist as a single, global phylogenetic population, presumably due to its small spore size [18]. Natural A. fumigatus isolates were described previously as having low genetic diversity in comparison to N. fischeri isolates [19]. However recent studies identified a number of strain-specific [7] and polymorphic [20],[21] genes. To further explore the extent of genetic variation within the A. fumigatus species, we included in this analysis the genome sequence of a second strain, A1163, made available through Merck & Co., Inc., Whitehouse Station, NJ. Our preliminary analysis has shown that Af293 and A1163 isolates vary greatly in their resistance to antifungals (Table S2). Results/Discussion A. fumigatus Af293 vs. A. fumigatus A1163 The genome of A. fumigatus strain A1163 was sequenced by the whole genome random sequencing method [22]. Its genome (29.2 Mb) is 1.4% larger than the genome of the first sequenced strain Af293 (28.8 Mb) (Table 1). About 98% of each genome can be aligned with high confidence. Alignment of the A1163 genome against the eight Af293 chromosomes has revealed 17 large syntenic blocks, which correspond roughly to the 16 Af293 chromosomal arms (Figure 3). The syntenic blocks were defined as regions containing at least five syntenic orthologs separated by no more than 20 genes without orthologs. 10.1371/journal.pgen.1000046.g003 Figure 3 Alignment of the A1163, N. fischeri, and A. clavatus Assemblies against the Eight Af293 Chromosomes. The first three tracks from the top for each reference chromosome show syntenic blocks (horizontal bars) identified in the target genomes, A. fumigatus A1163, N. fischeri, and A. clavatus. Each assembly from the target genomes is represented by a single color. Syntenic blocks are numbered based on the target genome assembly ID and the position of the block in the target genome assembly. Tracks 4 and 5 show Asp-core gene density and blocks (horizontal bars), respectively, in the Af293 genome. Tracks 6 and 7 show Afum-specific gene density and blocks (horizontal bars), respectively. Tracks 8 and 9 show the density of clustered secondary metabolite biosynthesis genes and transposable elements, respectively, found in Af293. Pink vertical bars represent putative centromeres, the purple vertical bar in chromosome 4 represents a region of ribosomal DNA, and horizontal black bars beneath each chromosome designate sequencing gaps. 10.1371/journal.pgen.1000046.t001 Table 1 Genome Statistics Sequenced organisms Af293 A1163 N. fischeri A. clavatus Length (Mp) 28.810 29.205 32.552 27.859 Assemblies >100 Kb 18 11 13 16 GC content 50% 49% 49% 49% No. of genes 9631 9906 10407 9125 No. of LS genes 818 1408 1151 Mean gene length (Bp) 1478 1455 1466 1483 % Genes with introns 79% 80% 80% 81% % Coding 49% 49% 47% 49% Most translocation events involving A. fumigatus chromosomes appear to have taken place within 300 Kb from the telomeres. The largest exchange involved a ∼500 Kb segment between Af293 chromosomes 1 and 6 and A1163, which contain regions aligning with A1163 assembly 1 (syntenic blocks 1.1 and 1.2 in Figure 3). This appears to be a recent event that happened in A293. In addition, Af293 chromosome 1 harbours a 400 Kb subtelomeric region that does not align well with A1163 assemblies. There is evidence of gene conversion between distal subtelomeric sequences encoding RecQ family helicases in A. fumigatus chromosomes 2, 4, and 7. Consistent with previous reports [19], the identity over the shared regions is very high (99.8% at the nucleotide level). This is higher than 99.3% and 99.5% identity between the two sequenced A. niger isolates (ATCC 1015 and CBS 513.88) [23] and between A. oryzae [10] and A. flavus [8], respectively. Unique regions represent 1.2% and 2.3% (and harbour 143 and 218 genes) in the Af293 and A1163 genomes, respectively. More than half of the Af293-specific genes are also absent in A. fumigatus isolates Af294 and Af71, according to the array-based comparative genome hybridization (aCGH) data [7]. The vast majority of Af293- and A1163-specific genes are clustered together in blocks ranging in size from 10 to 400 Kb, which seem to be the most variable segment of the species genome. A manual examination of these isolate-specific islands revealed that they contain numerous pseudogenes and repeat elements. One of the regions contains a putative secondary metabolism cluster (AFUA_3G02530-AFUA3G02670). The origin of 20% of Af293-specific genes can be attributed to two segmental duplication events. One of the duplicated regions (AFUA_1G16010- AFUA_1G16170) contains an arsenic detoxification cluster. The other (AFUA_1G00420-AFUA_1G00580) contains genes that may be involved in metabolism of betaine, which is often synthesized under osmotic and heavy metal stress. Interestingly the duplicated regions are also absent in Af294 and Af71 isolates, which suggests that the duplication event took place very recently. Segmental duplication events are thought to contribute to rapid adaptation of the species by increasing their expression. Since Af293 is a clinical isolate it is possible that these chromosomal aberrations were created due to selective pressures in the host. Highly Variable Loci in A. fumigatus Although most Af293 proteins are 100% identical to their A1163 orthologs, we have identified 41 orthologous pairs that share only 37% to 95% identity. To find out if these genes are also divergent in other A. fumigatus isolates, we identified Af293 genes that do not hybridize with DNA extracted from the Af294 and Af71 strains in aCGH experiments [7]. The comparison revealed that 27 out of 41 genes were possibly polymorphic (marked as absent or divergent) with respect to at least one other isolate (Table S3). Further analysis of three polymorphic loci in other A. fumigatus isolates has demonstrated that each of them harbours two or three alleles (Table S4). A PCR survey followed by Southern blot analysis and partial DNA sequencing has shown the presence of at least two alleles at each locus containing nearly identical sequences within each group of alleles (data not shown). In filamentous fungi, this high level of variability has been previously associated with heterokaryon incompatibility (het) genes involved in a programmed cell death (PCD) pathway triggered by hyphal fusion between two genetically incompatible individuals [24],[25]. So far several het loci have been described in A. nidulans [26], although none have been characterized at the molecular level. Incidentally, our results are consistent with previously identified vegetative incompatibility groups suggesting that some of these polymorphic genes may function in heterokaryon incompatibility in A. fumigatus. Thus, four clinical isolates from the same multi-member incompatibility group (WSA-270, WSA-1195, WSA-449, and WSA-172) contained the same alleles of the polymorphic genes (Table S4). Furthermore, at least five putative A. fumigatus het genes exhibit a pattern of trans-species (or trans-specific) polymorphism (Table S5), which has been previously associated with somatic and sexual incompatibility in fungi, self-incompatibility in plants, and the major histocompatibility complex (MHC) in vertebrates. These genes are more similar to their orthologs from other Aspergillus species than to those from A1163. We chose one putative het gene, rosA (AFUA_1G15910), and its close relative, nosA (AFUA_4G09710), whose orthologs encode two Zn2C6 transcriptional regulators of sexual development in A. nidulans [27],[28] for phylogenetic analysis (Figure 4). Unexpectedly, Af293 RosA clusters with its A. clavatus ortholog, while A1163 RosA clusters with N. fischeri. This is in contrast with the NosA tree, which perfectly mirrors the species tree (Figure 2), suggesting that these allelic classes may transcend species boundaries in the aspergilli. 10.1371/journal.pgen.1000046.g004 Figure 4 The Af293 RosA and NosA Proteins. Shown in bold red are RosA, NosA and Pro1 proteins that have been experimentally characterized are shown in bold black. Branches with a bootstrap of 75% or more are indicated in bold black. The trees are maximum-likelihood trees (see Materials and Methods). This is the first study that shows the diversity of het genes in aspergilli at the molecular level as well as patterns of trans-species polymorphism. These putative het genes are distinct from those identified in Neurospora crassa or Podospora anserina [24],[25], although many of them share the same domains such as the NACHT and NB-ARC domains of the STAND superfamily [29]. Coincidentally four of the A. fumigatus variable genes encoding STAND domain proteins have previously been predicted to function in heterokaryon incompatibility [30]. The discovery of putative het loci in the aspergilli may facilitate identification of downstream components of fungal PCD pathways or other drug targets. These loci may be also used as a basis for classification of natural and clinical isolates into different compatibility groups. A. fumigatus vs. N. fischeri vs. A. clavatus The genomes of N. fischeri and A. clavatus were sequenced by the whole genome sequencing method [22]. The N. fischeri genome (32.6 Mb) is 10–15% larger than the A. clavatus and A. fumigatus genomes (Table 1). There are 10,407 protein-coding genes and a large number of transposable elements, which may have contributed to its genome size expansion. The A. clavatus genome (27.9 Mb) is the smallest seen to date among the sequenced aspergilli (Table 1). There are currently 9,125 predicted protein-coding genes. This is consistent with past comparative studies that identified notable (up to 30%) genome size differences between distantly related aspergilli [7],[9],[10]. Despite this significant genome size variability, gene-level comparisons confirmed phylogenetic proximity of A. fumigatus, N. fischeri and A. clavatus (Figures 1 and 2). The three genomes also appear to be largely syntenic. Alignment of the N. fischeri and A. clavatus genomes against the eight Af293 chromosomes has revealed 20 and 55 syntenic blocks, respectively (Table 2). There is only one large-scale reciprocal translocation between chromosomes 2 and 5 in N. fischeri (blocks 8927.1, 8927.2, 9292.1 and 9292.2, in Figure 3). The A. clavatus supercontigs align with A. fumigatus chromosomes 2 and 5, suggesting that this was the ancestral topology. 10.1371/journal.pgen.1000046.t002 Table 2 Syntenic and Afum-specific Chromosomal Blocks in Af293 Af293 blocks Syntenic to A1163 Syntenic to N. fischeri Syntenic to A. clavatus Afum specific No. of original blocks 29 24 62 13 No. of merged blocks 17 20 55 13 Merged blocks length 28.4 Mb 27.6 Mb 26.0 Mb 1.7 Mb % Coding 50% 51% 52% 31% Repeata density 0.51% 0.50% 0.47% 1.83% TEb density 1.07% 0.96% 0.80% 4.17% Syntenic blocks for each pair of genomes were defined as areas containing a minimum of five orthologous genes in the Af293 and target genomes with a maximum of 20 adjacent non-matching genes. Afum-specific blocks were defined as Af293 areas containing at least ten Afum-specific genes and separated by no more than 5 other genes. Since most syntenic regions slightly overlap, the original blocks were merged to calculate repeat and TE density. Abbreviations: arepeat elements; btransposable elements. Repeat and TE densities were estimated as described in Materials and Methods. Core and Lineage-Specific Genes Features of Core and Lineage-Specific Genes Comparative genomic analysis has showed that the three Aspergillus genomes contain a large number of species-specific genes, which is consistent with previous comparative studies [7]. We have identified 7514 orthologous core and 818, 1402 and 1151 species-specific genes in the Af293, N. fischeri and A. clavatus genomes, respectively (Figure 5). Numbers of core- and species-specific genes, however, depend on selection of genomes from which they were derived. Thus, adding new genomes to this comparison resulted in fewer core and specific genes as shown for Af293 in Table S6. The availability of additional sequenced Aspergillus genomes allowed us to explore these patterns in a more systematic manner by comparing A. fumigatus Af293 genes with different lineage specificity (i.e. number of orthologs in other species). 10.1371/journal.pgen.1000046.g005 Figure 5 Proteins with Orthologs in the Three Most Closely Related Aspergilli (A. fumigatus, N. fischeri and A. clavatus). These proteins constitute the Affc-core group, and proteins with no orthologs in N. fischeri and A. clavatus constitute the A. fumigatus-specific group (Afum). The proteins in the Affc-core can be further divided into two groups, Aspergillus-core (Asp-core), which has orthologs in all of the other aspergilli, and the Affc-specific group, which is comprised of the rest of the Affc-core. To this end, we have selected four sets of genes based on the presence of orthologs in the six other sequenced aspergilli: N. fischeri, A. clavatus, A. terreus (CH476594), A. oryzae [10], A. nidulans [9] and A. niger CBS 513.88 [23] (Table S6; Figure 5). Genes with orthologs in the three most closely related aspergilli ( A. fumigatus, N. fischeri and A. clavatus) constitute the Affc-core group. The genes in the Affc-core can be further divided into two groups, the Aspergillus-core (Asp-core) with orthologs in all six other aspergilli and the Affc-specific group, which is comprised of the remaining Affc-core genes. Finally, the A. fumigatus-specific (Afum-specific) group contains Af293 genes that have orthologs in neither N. fischeri nor A. clavatus. One of the most striking observations to arise from this comparison was the marked differences in size and number of exons among genes from different lineage-specificity groups (Table 3). For example, Asp-core genes on average are almost twice as large as Afum-specific genes. The latter have on average only 1.35 introns and almost 31% lack introns completely. In contrast, Asp-core genes contain on average 2.16 introns, only 16% of them without introns. Consistent with previous reports of increased evolutionary rates in LS genes (e.g. [31]), Affc- and Afum-specific genes in A. fumigatus exhibit low sequence identity to their orthologs from more distantly related fungi (Table 3). 10.1371/journal.pgen.1000046.t003 Table 3 Comparison of Four Af293 Gene Sets with Different Lineage Specificity Lineage specificity group Asp-core Affc-core Affc-specific Afum-specific No of genes 5424 7514 2090 818 No of orthologs in 6 aspergilli 6 2–6 2–5 0–1 Mean gene length 1722 1579 1209 802 Mean No. of introns 2.16 2.02 1.66 1.35 %Genes without introns 15.9% 19.4% 28.5% 31.4% % Affc syntenic 98.3% 96.0% 89.8% n/a % Telomere-proximal 5.6% 9.1% 38.0% 36.5% % Expressed 42.5% 42.7% 43.3% 32.4% % Orthologs in A. clavatus 100% 100% 100% n/a % Orthologs in N. crassa 81.5% 70.7% 42.6% 4.5% % Orthologs in S. cerevisiae 49.9% 41.5% 19.9% 1.2% % Identity to A. clavatus orthologs 81.3% 78.6% 71.4% n/a % Identity to N. crassa orthologs 52.3% 51.6% 47.9% 43.3% % Identity to S. cerevisiae orthologs 43.1% 42.7% 40.4% 38.0% The numbers of Af293 genes in different categories are shown for Aspergillus-core (Asp-core), Affc-core, Affc-specific, and A. fumigatus-specific (Afum-specific) groups (see main text for definitions). Telomere-proximal genes are defined as genes located within 300 Kb from the chromosome end. Affc syntenic genes are defined as Af293 genes syntenic with respect to N. fischeri and A. clavatus (see the legend to Table 2). The ‘expressed’ genes are defined as Af293 genes that showed differential expression in at least one microarray study (W. Nierman, unpublished). These vast differences in gene features between core and specific genes are more likely to be explained by relaxed selective constraints (as discussed below) than by poor annotation quality of LS genes (due to misannotated gene models, gene fragments or random ORFs). We made significant improvements to Af294 gene models by leveraging the comparative genomic data (see Materials and Methods). In addition, all Affc-specific genes have orthologs in N. fischeri and A. clavatus and 43% of them are differentially expressed in various expression studies, which is similar to the A. fumigatus genome average (Table 3). On the other hand, many Afum-specific genes may be non-functional, since only 32% of them are differentially expressed in microarray studies (vs. the 43% genome average) and only 60% of them show sequence similarity to other fungal proteins (Table S7; Figure 6). Nonetheless, at least 20% of Afum-specific genes are supported by combined evidence (homology and expression data) and therefore are likely to be functional. Nonetheless, even these genes are still smaller in size than average Affc- and Asp-core genes. 10.1371/journal.pgen.1000046.g006 Figure 6 A. fumigatus-Species Specific Genes Supported by Homology and Expression Data. Genes with no orthologs in N. fischeri and A. clavatus constitute the A. fumigatus-specific group (Afum). Genes that have homologs in other fungal genomes constitute the Homology group. Genes differentially expressed in microarray studies represent the Expressed group. Biological Roles and Chromosomal Location of LS Genes Analysis of Gene Ontology (GO) terms [32] associated with core and lineage-specific groups has demonstrated that certain biological functions are unequally distributed among these groups (Table S8). The Afum-specific group is enriched for genes involved in carbohydrate transport and catabolism, secondary metabolite biosynthesis, and detoxification. In contrast, the invariable Asp-core genome encodes many functions associated with information processing and other cellular processes that contribute to the organism's fitness in most environments. Thus, a significant number of Asp-core genes (15%) are orthologous to yeast essential genes, which represents a two-fold enrichment in comparison to the rest of the proteome. Although most Af293 genes involved in carbohydrate transport and catabolism are found in the Asp-core group, only 10% of secondary metabolism genes have orthologs in all sequenced aspergilli including siderophore, pigment and Pes1-related clusters. These three conserved clusters are also found in Penicillium species and some more distantly related fungi. Similarly, only 30% of secondary metabolism Af293 genes are shared by N. fischeri and A. clavatus. The three species also vary considerably in the numbers of enzymes that control the first step in secondary metabolite biosynthesis such as nonribosomal peptide synthases (NRPS), polyketide synthases (PKS), and dimethylallyltryptophan synthases (DMATS) (Table S9). Interestingly, N. fischeri genome contains 46 enzymes, which is 35% more than A. clavatus (35) and A. fumigatus (34) genomes. Likewise, PFAM domains overrepresented among Affc- and Afum-specific genes have been shown to function in efflux or detoxification, secondary metabolite biosynthesis, resistance to antifungals, and other accessory metabolic pathways. They include MSF and ABC transporters, various oxidoreductases, cytochrome P450, glycosyl and alpha/beta fold hydrolases, polyketide synthases, glutathione transferases and methyltransferases (Table S10). On the other hand, core genes often contain AAA-superfamily ATPase, helicase, WD40, and SH3 domains associated with such important functions as cell organization and macromolecule biosynthesis. Lineage Specific Genomic Islands In addition to difference in size and function, lineage specific genes display a significant subtelomeric bias. As opposed to telomere-distal Asp- and Affc-core genes, Affc- and Afum-specific genes tend to be located within 300 Kb from chromosome ends (P value>0.01) (Table S11). About 38% of Affc-specific genes are telomere-proximal in comparison to 6% of Asp-core and 9% Affc-core genes (Table 3). Interestingly, 46% of Afum-specific genes with paralogs are telomere-proximal (Table S7), suggesting that they may have been recently duplicated and translocated to these regions. Our findings concur with previous reports of subtelomeric bias in LS genes in A. fumigatus [7], S. cerevisiae [33] and Pichia stipitis [34]. With the exception of one Af293 locus containing four P450 genes, the Aspergillus species do not have large variable subtelomeric arrays arising by a series of tandem duplications found in some protozoan parasites [35]. Almost 50% of the Afum-specific genes can be clustered together in 13 blocks containing more than 10 Afum-specific genes separated by no more than 5 genes outside this category (Table 2). Together these regions, referred to here as Afum-specific genomic islands, show an even more significant telomeric bias (68% of the clustered genes lay within 300 Kb from telomere ends) with larger blocks found almost exclusively at chromosome ends (Figure 3). In addition to non-syntenic genes, species-specific islands harbour a disproportionate number of transposons and other repeat elements in comparison with the syntenic areas of the Af293 genome (Table 2). Notably two A. fumigatus-specific blocks (2.2 and 3.1) contain gene clusters involved in biosynthesis of mycotoxin fumigaclavine and another unknown secondary metabolite [36]. Similar genomic islands have been described in the rice blast fungus Magnaporthe oryzae [37],[38] and in A. oryzae [10] suggesting that they may be shared across all filamentous ascomycota fungi. Unlike variable subtelomeric regions found in other eukaryotes [39],[40], these areas are often quite large (up to 400 Kb) and not always located near chromosome ends. Evolutionary Origins of Lineage-Specific Genes Most Affc- and Afum-specific genes have no orthologs in non-Aspergillus fungal species, which suggests that they were created de novo in the Affc lineage. To gain insight into the origin of the LS genes in aspergilli, we have performed phylogenetic analysis of two sets of A. fumigatus- and N. fischeri-specific genes. In Af293 and N. fischeri, Set 1 contains 790 and 1230 genes, respectively, that have an Aspergillus homolog as the best BLASTp hit; Set 2 contains 28 and 178 genes, respectively, that have a non-Aspergillus homolog as the closest relative. There is a significant difference in the numbers of trees including a non-Aspergillus species as the closest relative in N. fischeri and A. fumigatus (P value = 2.6e-08). This is indicative of major differences in retention and/or uptake of new genetic material in these two species, consistent with differences in their reproductive modes. The four repetitive scenarios identified by phylogenetic analysis are displayed in Figure 7. In both A. fumigatus and N. fischeri, most of the Set 1 genes exhibit topologies that do not strictly follow the Aspergillus species tree (Figure 2), although nested within the Aspergillus clade. Similarly, all 28 A. fumigatus Set 2 genes are nested within the Aspergillus genus. In contrast to the A. fumigatus genes, N. fischeri Set 2 genes sometimes cluster with a non-Aspergillus species with high bootstrap support. As shown in Figure 7B and 7C, both N. fischeri and non-Aspergillus species genes can be nested either in this non-Aspergillus clade or in the Aspergillus clade. At first sight, these repetitive topologies can be interpreted as supportive of a horizontal gene transfer (HGT) from a non-Aspergillus species into N. fischeri or visa versa. Further analysis, however, reveals that most of the conflicts involve sparsely populated trees, long branch attraction artifacts, and other situations, where phylogenetic methods tend to mislead (e.g. [41]). The last repetitive scenario includes genes that are only present in one other distant fungal genome (Figure 7D). The evolutionary origin of genes in this category cannot be resolved at this time. 10.1371/journal.pgen.1000046.g007 Figure 7 Four Common Topologies Detected by Phylogenetic Analysis of N. fischeri-Specific Proteins. The N. fischeri proteins under consideration are in bold red. The bootstrap supporting the clade containing the N. fischeri is also in bold red. Other N. fischeri proteins are shown in bold black. Blue species names correspond to the recipient genome when different from N. fischeri. Systematic gene names are indicated. Branches with a bootstrap of 75% or more are indicated in bold black. The trees are maximum-likelihood trees (see Materials and Methods). A. Set1 protein evolved by probable duplication, differentiation and differential loss in other Aspergillus species (DDL). B. Set 2 protein evolved by probable HGT from Sordaryomycetes into the N. fischeri lineage. C. Set 2 protein evolved by probable DDL and a Fusarium solani protein (in blue) evolved by probable HGT from the N. fischeri lineage into Sordaryomycetes. D. Set 2 protein showing similarity to a protein from the Sordaryomyce Chaetomium globosum. Our results are consistent with the well established role of gene duplication and divergence as the principal source of new genes [42]–[45]. They are however in conflict with previous studies that attributed the origin of LS genes in the aspergilli to gene acquisition through HGT from other fungal species [9],[10],[46]. This assumption was based on circumstantial evidence such as mosaic phyletic distribution, phylogenetic anomalies, and differences in gene content among A. fumigatus, A. nidulans and A. oryzae. Besides the absence of readily apparent HGT examples, the fact that LS genes tend to be smaller in size and have fewer exons is difficult to explain by HGT. These gene features are quite consistent across Aspergillus species, and it is therefore unclear what could be the donor organism for LS genes. The DDL scenario does not have this weakness, since these size differences can be a direct consequence of relaxed selective constraints operating on duplicate genes. According to the DDL hypothesis, the initial redundancy in gene function allows duplicate genes to quickly accumulate nonsynonymous mutations and even premature stop codons. Notably, over 20% of all Afum-specific genes can be linked to the two very recent segmental duplications events that occurred in Af293 but not in A1163. Both translocated segments are telomere-distal and contain genes that appear to be pseudogenized indicating that translocated gene copies may have evolved under relaxed selective constraints. Similarly in other species, accelerated evolution has been often associated with subtelomeric areas suggesting that the process is dependent on the local chromatin environment (e.g. [47]). The prevailing role of duplication in the origin of LS genes in the aspergilli is further underlined by their tendency to cluster in genomic islands. These regions may function as designated “gene dumps” and simultaneously as “gene factories”, since some LS genes appear to maintain their functional integrity or at least are differentially expressed in microarray studies as shown above. As shown above, 46% of Afum-specific genes with paralogs are telomere-proximal (Table S7), suggesting that they may have been recently duplicated and translocated to these regions. Evidence for gene duplication and/or transfer to evolutionarily labile regions is found in some protozoan parasites that have large variable subtelomeric arrays arising by a series of tandem duplications [35]. Conservation of Virulence-, Allergy-, and Sex-Associated Genes Previous studies however have shown a high level of evolutionary conservation and phyletic retention among known A. fumigatus virulence-associated genes [7]. Our analysis confirmed the low rate of protein evolution among these genes in four Aspergillus species (Table S12). Interestingly, four of the virulence-associated genes, pabaA (AFUA_6G04820), fos-1 (AFUA_6G10240), pes1 (AFUA_1G10380) and pksP (AFUA_2G17600), reveal evidence of accelerated evolution in the branch leading to the two A. fumigatus isolates. This pattern can affect only a few amino acid residues (e.g. PksP) or a significant proportion of the protein (e.g. Pes1). Such a pattern can be due to either relaxation of selection or selection for rapid diversification (positive selection). In the latter case specific amino acid substitutions may decrease susceptibility to specific environmental challenges and thus enhance A. fumigatus virulence. These four genes are involved in oxidative stress or nutrient availability, which is consistent with the positive selection scenario. Indeed, PabaA is involved in biosynthesis of folate, an essential co-factor for DNA synthesis. Since PABA is apparently limited in the mammalian lung, a functional pabaA gene is required for virulence [48]. Fos1, a putative two-component histidine kinase, may play a role in the regulation of cell-wall assembly [49]. Finally, PksP and Pes1 are enzymes, which catalyze the first steps in biosynthesis of the spore pigment and an unknown non-ribosomal peptide, have been shown to mediate resistance to oxidative stress in addition to their role in A. fumigatus virulence [50],[51]. The inclusion of additional taxa in the analyses might clarify the significance of the observed differences. This overall lack of variability among known virulence-associated factors suggests that yet unknown A. fumigatus-specific genes may contribute to its ability to survive in the human host. A recent microarray study demonstrated that the Affc-specific genes are over-represented among genes that are up-expressed in the neutropenic murine lung (Elaine Bignell submitted for publication). Many of them are found in chromosomal gene clusters associated with macromolecule catabolism and secondary metabolite biosynthesis. Similarly, clustered lineage-specific genes simultaneously induced in infected tissue have been observed in the ubiquitous maize pathogen Ustilago maydis [52] and some other species (for a recent review see [53]). Alternatively A. fumigatus virulence may be a combinatorial process, dependent on a pool of genes, which interact in various combinations in different genetic backgrounds as suggested previously [7]. Similar ‘ready-made’ virulence features have been described in other environmental pathogens such as Pseudomonas aeruginosa [54] and Cryptococcus neoformans [55],[56]. In addition to virulence factors, the A. fumigatus genome encodes 20 allergens (Table S13) and 25 proteins displaying significant sequence similarity to known fungal allergens (Table S14), some of which appear to contribute to its pathogenicity [57]. For example, A. fumigatus Asp f6 (AFUA_1G14550), also known as Mn2+-dependent superoxide dismutase (MnSOD), is specifically recognized by IgE from patients with allergic bronchopulmonary aspergillosis (ABPA) and is differentially expressed during germination [58]. The broad distribution of allergens among fungal taxa (Text S1) suggests that A. fumigatus possesses the same allergen complement as most other aspergilli and that its effect on hypersensitive individuals can be explained mostly by its ubiquity in the environment. Our analysis has demonstrated that, similar to known virulence-associated genes, most sexual development genes appear to be under negative (purifying) selection in both sexual and asexual Aspergillus species (Text S1 and Table S15). More detailed analysis has revealed four genes in the N. fisheri lineage that may be under positive selection. This suggests that a few amino acid changes may enable sexuality in N. fischeri. The conservation of sex genes in asexual species is due to a latent sexuality, a recent loss of sexuality, pleiotropy, or parasexual recombination following heterokaryon formation as suggested previously [59],[60]. Conclusions Lineage-specific (LS) genes (i.e. genes with limited phylogenetic distribution of orthologs in related species) have been the focal point of many comparative genomic studies, because of the assumption that they may be responsible for phenotypic differences among species and niche adaptation. Our analyses of the genomes of A. fumigatus and the two closely related species, N. fischeri and A. clavatus, demonstrates that A. fumigatus may possess genetic determinants that allow it to establish a successful in vivo infection. LS genes that have no orthologs in the other two species comprise 8,5% of the A. fumigatus genome and often have accessory functions such as carbohydrate and amino acid metabolism, transport, detoxification, or secondary metabolite biosynthesis. Further analysis showed that these genes have distinct features (e.g. the small gene length and number of introns) and tend to cluster in subtelomeric genomic islands, which may function as “gene dumps/factories”. The phylogenies of LS genes, their subtelomeric bias and size differences are consistent with the DDL hypothesis stating that duplication being the primary genetic mechanism responsible for the origin of species-specific genes. The presence of genomic islands indicates that A. fumigatus and may possess sophisticated genetic mechanisms that facilitate its adaptation to heterogeneous environments such as soil or a living host. Materials and Methods Fungal Isolates A. fumigatus Af293 (FGSC A1100) was isolated from patients with invasive aspergillosis [61]. A. fumigatus A1163 (FGSC A1163) is a derivative of A. fumigatus CEA17 converted to pyrG+ via the ectopic insertion of the A. niger pyrG gene [62],[63]. CEA17 is a uracil auxotroph of A. fumigatus clinical isolate CEA10 (CBS144.89). The type strains of A. clavatus (NRRL 1) and N. fischeri (NRRL 181) were used for sequencing and phenotypic characterization. Accession Numbers The genome sequences of A. clavatus, N. fischeri and A. fumigatus A1163 were deposited to the GenBank under the following accession numbers: AAKD00000000, AAKE00000000 and ABDB00000000, respectively. Whole Genome Sequencing A1163, A. clavatus and N. fischeri were sequenced using the whole genome shotgun method as previously described [22]. Random shotgun libraries of 2–3 Kb, 8–12 Kb and 50 Kb were constructed from genomic DNA from each strain, and DNA template was prepared for high-throughput sequencing using Big Dye Terminator chemistry (Applied Biosystems). Sequence data was assembled using Celera Assembler. For A. fumigatus A1163, scaffolds were compared to those of the first sequenced isolate, Af293 [7]. Sequence Identity at the Nucleotide Level A1163 assemblies larger than 5 Kb were aligned to the Af293 chromosomes using the MUMmer package (http://mummer.sourceforge.net/) [64]. Alignments longer than 100 Kb were used to determine average sequence identity to avoid highly repetitive and duplicated regions. The same approach was used to estimate sequence identity between A. flavus and A. oryzae and between the two sequenced A. niger strains. Gene Structure Annotation The JCVI eukaryotic annotation pipeline was applied to the A1163, A. clavatus and N. fischeri assemblies (supercontigs) larger than 2 Kb as described earlier [7]. We used PASA [65] and EvidenceModeler [66] to generate consensus gene models based on predictions from several types of genefinders including GlimmerHMM, Genezilla, SNAP, Genewise and Twinscan. Putative pseudogenes, small species-specific genes (less than 50 amino acids), and gene models overlapping with transposable elements (TE) shown in Table S16 were excluded from the final gene lists. Repetitive Elements Identification of repeat elements was performed using RepeatMasker (http://www.repeatmasker.org/), RepeatScout (http://repeatscout.bioprojects.org/), and Tandem Repeats Finder (http://tandem.bu.edu/trf/trf.html). Putative TEs (Table S16) were identified by Transposon-PSI (http://transposonpsi.sourceforge.net), a program that performs tBLASTn searches using a set of position specific scoring matrices (PSSMs) specific for different TE families. TE and repeat densities were calculated as the percentage of nucleotide bases in the regions of interest (i.e., syntenic or non-syntenic blocks) that overlap with a feature of the appropriate type (repeat or TE). A. fumigatus Annotation Improvements We leveraged the comparative genomic data to significantly improve annotation quality of the Af293 genome, which was previously annotated with relatively little supporting evidence [7]. The refinement of initial annotation was performed using the Sybil software package (http://sybil.sourceforge.net/), which allows for rapid identification of discrepancies in gene structure among orthologs. The comparison with orthologous N. fischeri and A. clavatus genes resulted in significant changes to the Af293 gene catalogue. Over 1100 gene models were updated and 130 new genes were identified. Initial A. fumigatus A1163 gene models were also improved using the PASA pipeline, initially developed to align expressed sequence tag (EST) data onto genomic sequences [65]. The pipeline was adapted to automatically update A1163 gene models by aligning them against Af293 coding sequences (CDSs). Functional Annotation We have performed transitive functional annotation from Af293 proteins to their A1163, N. fischeri and A. clavatus orthologs. Previously GO terms [32] were assigned to Af293 proteins based on sequence similarity to PFAM domains or experimentally characterized S. cerevisiae proteins [7]. Secondary metabolism gene clusters were identified using Secondary Metabolism Region Finder (SMURF) available at http://www.jcvi.org/smurf (Nora Khaldi, unpublished). The complete list of gene clusters can be downloaded at ftp://ftp.jcvi.org/pub/software/smurf/. Gene Ontology (GO) terms [32] were assigned as described in [7] Ortholog Identification After extensive computational and manual refinement, the improved protein datasets were used to generate the final set of orthologs. Orthologous groups in Aspergillus genomes were identified using a reciprocal-best-BLAST-hit (RBH) approach with a cut-off of 1e-05. In addition to the A1163, A. clavatus and N. fischeri genomes, the previously sequenced genomes of Af293 [7], A. terreus NIH2624 (http://www.broad.mit.edu), A. oryzae RIB40 [10], A. nidulans FGSC A4 [9] and A. niger CBS 513.55 [23] were included in the comparative analysis. The results of this analysis, as well as synteny visualisation and comparative analysis tools can be also found in the Aspergillus Comparative database at http://www.tigr.org/sybil/asp. Orthologous, unique and divergent genes in Af293 were identified based on alignments of Af293 CDSs against A1163 assemblies using gmap as implemented in PASA [65] using default parameters. Synteny Analysis Syntenic blocks for each pair of genomes (Af293 vs. A. clavatus and Af293 vs. N. fischeri) were defined as areas containing a minimum of five matching (orthologous) genes with a maximum of 20 adjacent non-matching genes (having no orthologs) in the reference and target genomes. Since most syntenic regions slightly overlapped, the original blocks were merged to calculate repeat and TE density. Af293 non-syntenic blocks were defined as areas excluded from the syntenic blocks and containing at least ten Af239 non-matching genes. Statistical Analysis Genes in four lineage-specificity groups were analyzed by the EASE module [67] in MEV within TM4 (http://TM4.org) [68] to identify overrepresented Gene Ontology (GO) terms, Pfam domains and Chromosomal Regions (telomere-proximal and central). Only categories with Fisher's exact test probabilities above with P>0.05 from the EASE analyses were reported for each gene set. Selective Constraints Selective constraints were estimated for sets of orthologous genes from the Af293, A1163, A. clavatus, N. fischeri and A. terreus genomes. The rate of substitution in synonymous (d S) and in non-synonymous (d N) sites, and their ratio (d N/d S) was calculated using the PAML package [69]. If a gene is very well conserved, d N/d S 1. The results are reported only for orthologous genes sets having unsaturated d S values, the same number of exons, and sequence alignment coverage >95%. For each gene, the average d N/d S ratio for five pairwise species comparisons was calculated. Phylogenetic Analyses We assembled a local database of protein sequences from the 28 publicly available fungal genome projects (Table S17). All phylogenetic analyses in this paper were carried out on protein sequences. The A. niger ATCC 1015, Nectria haematococca, Phanerochaete chrysosporium and Trichoderma reesei genomes projects was completed under the auspices of the US Department of Energy's Office of Science, Biological and Environmental Research Program and the by the University of California, Lawrence Livermore National Laboratory (Contract No. W-7405-Eng-48), Lawrence Berkeley National Laboratory (contract No. DE-AC03-76SF00098) and Los Alamos National Laboratory (contract No. W-7405-ENG-36). To produce a reference tree of species phylogeny we used the protein sequences of 90 likely orthologs from A. niger, A. nidulans, A. terreus, A. oryzae, A. clavatus, N. fischeri, A. fumigatus and Fusarium graminearum (teleomorph of Gibberella zeae) as an outgroup. To minimize the effect of incorrect or incongruent gene models, these proteins were chosen on the basis of having identical numbers of introns in each species and similar lengths. Sequences were aligned using MUSCLE [70] and columns of low conservation were removed manually. Maximum-likelihood trees were constructed using the PHYLIP package, applying the JTT substitution model with a gamma distribution (alpha = 0.5) of rates over four categories of variable sites. Phylogenetic analyses of individual Af293, A1163, and N. fischeri proteins were carried out on sets of homologs identified in BLASTP searches against our fungal database. The top 20 hits with E<10−4 were retained for analysis. Sequences were aligned using ClustalW [71]. Poorly aligned regions were removed using Gblocks [72]. Finally, a maximum likelihood tree was drawn using PHYML [73]. Southern Blot Analysis To detect polymorphisms in the rosA (AFUA_6G07010) gene, several hybridizations were performed using rosA gene as the probe and genomic DNA cleaved with EcoRI, ClaI, BamHI or EcoRV. For comparison, an invariable gene for all species (apg5; AFUA_6G07040) was used as the hybridization probe on genomic DNA digested with HpaI. Colony Radial Growth Rate Measurement Colony radial growth rate measurements were performed as described [74]. For each isolate, four (90 mm diameter) Petri dishes containing 25 ml agar medium were inoculated centrally with 2.5 µl of 1×106 spores/ml suspension in PBS/Tween 80. Plates were then incubated at temperatures ranging from 25°C to 50°C and colony edges were marked using a plate microscope. Colonies were marked twice daily for 4–5 days. For each colony, two diameters perpendicular to each other were measured. Eight replicates were measured for each isolate. The results reported here are the mean of two experiments. At least five time points during the log phase were used to calculate growth rate. The radius of the colonies was plotted against time using least-square regression analysis, and the slope of the regression line, which represents the growth rate, was calculated. Each replicate was analysed separately and the mean of the growth rate was then calculated. Supporting Information Text S1 Allergens and sexual development genes. (0.05 MB DOC) Click here for additional data file. Table S1 Growth rates of Af293, A1163, N. fischeri, and A. clavatus isolates at various temperatures. (0.02 MB XLS) Click here for additional data file. Table S2 Resistance to antifungals among A. fumigatus clinical isolates. (0.02 MB XLS) Click here for additional data file. Table S3 Divergent A. fumigatus Af293 genes with respect to Af294, Af71, and A1163. (0.03 MB XLS) Click here for additional data file. Table S4 Distribution of polymorphic alleles among A. fumigatus isolates. (0.02 MB XLS) Click here for additional data file. Table S5 Five A. fumigatus loci exhibiting trans-species polymorphism. (0.02 MB XLS) Click here for additional data file. Table S6 A. fumigatus core and species-specific genes. (0.02 MB XLS) Click here for additional data file. Table S7 Features of A. fumigatus-specific genes. (0.02 MB XLS) Click here for additional data file. Table S8 Top biological processes overrepresented among four lineage specificity groups. (0.02 MB XLS) Click here for additional data file. Table S9 Enzymes that control the first step in secondary metabolite biosynthesis. (0.02 MB XLS) Click here for additional data file. Table S10 Top PFAM domains overrepresented among four lineage specificity groups. (0.02 MB XLS) Click here for additional data file. Table S11 Lineage specificity and chromosomal location. (0.02 MB XLS) Click here for additional data file. Table S12 Selective constraints operating on virulence-associated genes. (0.02 MB XLS) Click here for additional data file. Table S13 Known A. fumigatus Af293 allergens. (0.02 MB XLS) Click here for additional data file. Table S14 Predicted A. fumigatus Af293 allergens. (0.02 MB XLS) Click here for additional data file. Table S15 Selective constraints operating on sex genes. (0.02 MB XLS) Click here for additional data file. Table S16 Families of transposable elements identified in the Affc genomes. (0.01 MB XLS) Click here for additional data file. Table S17 Fungal genomes used in phylogenetic analyses. (0.02 MB XLS) Click here for additional data file.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Aspergillosis.

              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              What makes Aspergillus fumigatus a successful pathogen? Genes and molecules involved in invasive aspergillosis.

              Aspergillus fumigatus is an opportunistic pathogen that causes 90% of invasive aspergillosis (IA) due to Aspergillus genus, with a 50-95% mortality rate. It has been postulated that certain virulence factors are characteristic of A. fumigatus, but the "non-classical" virulence factors seem to be highly variable. Overall, published studies have demonstrated that the virulence of this fungus is multifactorial, associated with its structure, its capacity for growth and adaptation to stress conditions, its mechanisms for evading the immune system and its ability to cause damage to the host. In this review we intend to give a general overview of the genes and molecules involved in the development of IA. The thermotolerance section focuses on five genes related with the capacity of the fungus to grow at temperatures above 30°C (thtA, cgrA, afpmt1, kre2/afmnt1, and hsp1/asp f 12). The following sections discuss molecules and genes related to interaction with the host and with the immune responses. These sections include β-glucan, α-glucan, chitin, galactomannan, galactomannoproteins (afmp1/asp f 17 and afmp2), hydrophobins (rodA/hyp1 and rodB), DHN-melanin, their respective synthases (fks1, rho1-4, ags1-3, chsA-G, och1-4, mnn9, van1, anp1, glfA, pksP/alb1, arp1, arp2, abr1, abr2, and ayg1), and modifying enzymes (gel1-7, bgt1, eng1, ecm33, afpigA, afpmt1-2, afpmt4, kre2/afmnt1, afmnt2-3, afcwh41 and pmi); several enzymes related to oxidative stress protection such as catalases (catA, cat1/catB, cat2/katG, catC, and catE), superoxide dismutases (sod1, sod2, sod3/asp f 6, and sod4), fatty acid oxygenases (ppoA-C), glutathione tranferases (gstA-E), and others (afyap1, skn7, and pes1); and efflux transporters (mdr1-4, atrF, abcA-E, and msfA-E). In addition, this review considers toxins and related genes, such as a diffusible toxic substance from conidia, gliotoxin (gliP and gliZ), mitogillin (res/mitF/asp f 1), hemolysin (aspHS), festuclavine and fumigaclavine A-C, fumitremorgin A-C, verruculogen, fumagillin, helvolic acid, aflatoxin B1 and G1, and laeA. Two sections cover genes and molecules related with nutrient uptake, signaling and metabolic regulations involved in virulence, including enzymes, such as serine proteases (alp/asp f 13, alp2, and asp f 18), metalloproteases (mep/asp f 5, mepB, and mep20), aspartic proteases (pep/asp f 10, pep2, and ctsD), dipeptidylpeptidases (dppIV and dppV), and phospholipases (plb1-3 and phospholipase C); siderophores and iron acquisition (sidA-G, sreA, ftrA, fetC, mirB-C, and amcA); zinc acquisition (zrfA-H, zafA, and pacC); amino acid biosynthesis, nitrogen uptake, and cross-pathways control (areA, rhbA, mcsA, lysF, cpcA/gcn4p, and cpcC/gcn2p); general biosynthetic pathway (pyrG, hcsA, and pabaA), trehalose biosynthesis (tpsA and tpsB), and other regulation pathways such as those of the MAP kinases (sakA/hogA, mpkA-C, ste7, pbs2, mkk2, steC/ste11, bck1, ssk2, and sho1), G-proteins (gpaA, sfaD, and cpgA), cAMP-PKA signaling (acyA, gpaB, pkaC1, and pkaR), His kinases (fos1 and tcsB), Ca(2+) signaling (calA/cnaA, crzA, gprC and gprD), and Ras family (rasA, rasB, and rhbA), and others (ace2, medA, and srbA). Finally, we also comment on the effect of A. fumigatus allergens (Asp f 1-Asp f 34) on IA. The data gathered generate a complex puzzle, the pieces representing virulence factors or the different activities of the fungus, and these need to be arranged to obtain a comprehensive vision of the virulence of A. fumigatus. The most recent gene expression studies using DNA-microarrays may be help us to understand this complex virulence, and to detect targets to develop rapid diagnostic methods and new antifungal agents. Copyright © 2010 Revista Iberoamericana de Micología. Published by Elsevier Espana. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Pathog
                PLoS Pathog
                plos
                plospath
                PLoS Pathogens
                Public Library of Science (San Francisco, CA USA )
                1553-7366
                1553-7374
                27 February 2020
                February 2020
                : 16
                : 2
                : e1008315
                Affiliations
                [1 ] Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
                [2 ] Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, United States of America
                [3 ] Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
                McGill University, CANADA
                Author notes

                The authors have declared that no competing interests exist.

                Author information
                http://orcid.org/0000-0002-7248-6551
                http://orcid.org/0000-0001-9195-7585
                http://orcid.org/0000-0002-8436-595X
                http://orcid.org/0000-0002-0354-8464
                http://orcid.org/0000-0002-2986-350X
                Article
                PPATHOGENS-D-19-02051
                10.1371/journal.ppat.1008315
                7046185
                32106242
                900ab349-a9de-4c52-9b4c-25e469015193
                © 2020 Rokas et al

                This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                Page count
                Figures: 2, Tables: 0, Pages: 9
                Funding
                This work was supported by a Discovery Grant from Vanderbilt University (to AR), by the Howard Hughes Medical Institute through the James H. Gilliam Fellowships for Advanced Study program (JLS and AR), and by the Fundação de Amparo a Pesquisa do Estado de São Paulo (FAPESP 2016/07870-9) and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), both from Brazil (to GHG). Research on bioactive fungal metabolites in NHO’s lab is supported by the National Cancer Institute (P01 CA125066). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Pearls
                Biology and Life Sciences
                Organisms
                Eukaryota
                Fungi
                Fungal Molds
                Aspergillus
                Aspergillus Fumigatus
                Biology and Life Sciences
                Microbiology
                Medical Microbiology
                Microbial Pathogens
                Fungal Pathogens
                Aspergillus Fumigatus
                Medicine and Health Sciences
                Pathology and Laboratory Medicine
                Pathogens
                Microbial Pathogens
                Fungal Pathogens
                Aspergillus Fumigatus
                Biology and Life Sciences
                Mycology
                Fungal Pathogens
                Aspergillus Fumigatus
                Medicine and Health Sciences
                Pathology and Laboratory Medicine
                Pathogenesis
                Biology and Life Sciences
                Organisms
                Eukaryota
                Fungi
                Fungal Molds
                Aspergillus
                Biology and Life Sciences
                Genetics
                Genetic Elements
                Biology and Life Sciences
                Mycology
                Fungal Evolution
                Biology and Life Sciences
                Microbiology
                Medical Microbiology
                Microbial Pathogens
                Fungal Pathogens
                Medicine and Health Sciences
                Pathology and Laboratory Medicine
                Pathogens
                Microbial Pathogens
                Fungal Pathogens
                Biology and Life Sciences
                Mycology
                Fungal Pathogens
                Biology and Life Sciences
                Organisms
                Eukaryota
                Fungi
                Medicine and Health Sciences
                Infectious Diseases
                Opportunistic Infections

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article