Buscar en
Revista Iberoamericana de Micología
Toda la web
Inicio Revista Iberoamericana de Micología What makes Aspergillus fumigatus a successful pathogen? Genes and molecules invo...
Información de la revista
Vol. 27. Núm. 4.
Páginas 155-182 (Octubre - Diciembre 2010)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
29133
Vol. 27. Núm. 4.
Páginas 155-182 (Octubre - Diciembre 2010)
Review
Acceso a texto completo
What makes Aspergillus fumigatus a successful pathogen? Genes and molecules involved in invasive aspergillosis
¿Qué hace que Aspergillus fumigatus sea un patógeno de éxito? Genes y moléculas involucrados en la aspergilosis invasora
Visitas
29133
Ana Abada, Jimena Victoria Fernández-Molinaa, Joseba Bikandib, Andoni Ramíreza,b, Javier Margaretoc, Javier Sendinod, Fernando Luis Hernandoa, Jose Pontóne, Javier Garaizarb, Aitor Rementeriaa,
Autor para correspondencia
aitor.rementeria@ehu.es

Corresponding author.
a Departamento de Inmunología, Microbiología y Parasitología, Facultad de Ciencia y Tecnología, UPV/EHU, Campus de Bizkaia, Leioa, Spain
b Departamento de Inmunología, Microbiología y Parasitología, Facultad de Farmacia, UPV/EHU, Campus de Alava, Vitoria-Gasteiz, Spain
c Fundacion Leia, Parque Tecnológico de Alava, Miñano, Alava, Spain
d Departamento de Neurociencias, Facultad de Farmacia, UPV/EHU, Campus de Alava, Vitoria-Gasteiz, Spain
e Departamento de Inmunología, Microbiología y Parasitología, Facultad de Medicina y Odontología, UPV/EHU, Campus de Bizkaia, Leioa, Spain
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (3)
Mostrar másMostrar menos
Tablas (7)
Table 1. Aspergillus fumigatus thermotolerance genes and their relationship with virulence
Table 2. Genes and proteins related with cell wall structure and virulence
Table 3. Genes and molecules associated with resistance to host immune response
Table 4. Toxins related to the direct attack to the host organism
Table 5. Genes and molecules related with nutrient uptake in invasive growth
Table 6. Molecules and genes involved in signaling, metabolic regulation and response to stress conditions
Table 7. Allergens of A. fumigatus related with activation of Type I hypersensitivity
Mostrar másMostrar menos
Abstract

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), Ca2+ 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.

Keywords:
Aspergillus fumigatus
Virulence
Invasive aspergillosis
Genes and molecules
Pathogenesis
Thermotolerance
Immune response
Cell wall
Toxins
Nutrient uptake
Signaling and regulation
Allergens
Resumen

Aspergillus fumigatus es un patógeno oportunista que causa el 90% de las aspergilosis invasoras (AI) con un 50–95% de mortalidad. Se ha postulado la existencia de factores de virulencia característicos, pero en A. fumigatus existe una gran variabilidad de factores de virulencia «no clásicos». Todos los estudios han demostrado que la virulencia de este hongo es multifactorial, asociada a su estructura, su capacidad de crecimiento y adaptación a condiciones de estrés, sus mecanismos de evasión del sistema inmune y su capacidad de causar daños en un huésped. En esta revisión se pretende dar una visión general de los genes y moléculas que intervienen en el desarrollo de la AI. La sección de termotolerancia incluye cinco genes relacionados con la capacidad de que el hongo crezca a más de 30°C (thtA, cgrA, afpmt1, kre2/afmnt1 y hsp1/asp f 12). En las siguientes secciones se discuten las moléculas y los genes relacionados con la interacción con el huésped y con la respuesta inmune. Estas secciones incluyen el β-glucano, el α-glucano, la quitina, el galactomanano, galactomanoproteinas (afmp1/asp f 17 y afmp2), hidrofobinas (rodA/hyp1 y rodB), la DHN-melanina, sus respectivas enzimas sintasas (fks1, rho1-4, ags1-3, chsA-G, och1-4, mnn9, van1, anp1, glfA, pksP/alb1, arp1, arp2, abr1, abr2 y ayg1) y enzimas modificantes (gel1-7, bgt1, eng1, ecm33, afpigA, afpmt1-2, afpmt4, kre2/afmnt1, afmnt2-3, afcwh41 y pmi), varias enzimas relacionadas con la protección del estrés oxidativo como catalasas (catA, cat1/catB, cat2/katG, catC y catE), superóxido dismutasas (sod1-2, sod3/asp f 6 y sod4), oxigenasas de ácidos grasos (ppoA-C), glutatión transferasas (gstA-E) y otros (afyap1, skn7 y pes1), y los transportadores de moléculas (mdr1-4, atrF, abcA-E y msfA-E). Esta revisión también incluye las toxinas y los genes relacionados, como la sustancia difusible de conidios, la gliotoxina (gliP y gliZ), la mitogilina (asp f 1/mitF/res), la hemolisina (aspHS), la festuclavina y la fumigaclavina A-C, la fumitremorgina, el verruculógeno, la fumagilina, el ácido helvólico, las aflatoxinas B1 y G1, y laeA. Dos secciones incluyen los genes y moléculas relacionadas con la absorción de nutrientes, la señalización y las regulaciones metabólicas implicadas en la virulencia, incluyendo enzimas, como las serin-proteasas (alp/asp f 13, alp2 y asp f 18), metaloproteasas (mep/asp f 5, mepB y mep20), aspártico-proteasas (pep/asp f 10, pep2 y ctsD), dipeptidilpeptidasas (dppIV y dppV) y fosfolipasas (plb1-3 y fosfolipasa C); sideróforos y la adquisición de hierro (sidA-G sreA, ftrA, fetC, mirB-C y amcA); adquisición de zinc (zrfA-H, zafA, y pacC); biosíntesis de aminoácidos, absorción de nitrógeno, y regulación por Cross-pathway Control (areA, rhbA, mcsA, lysF, cpcA/gcn4p y cpcC/gcn2p); vías de biosíntesis generales (pyrG, hcsA, y pabaA) y biosíntesis de trehalosa (tpsA y tpsB); otras vías de regulación, como MAP quinasas (sakA/hogA, mpkA-C, ste7, pbs2, mkk2, steC/ste11, bck1, ssk2 y sho1), proteínas G (gpaA, sfaD y cpgA), AMPc-PKA (acyA, gpaB, pkaC1 y pkaR), histidin-quinasas (fos1 y tcsB), señalización de Ca2+(calA/cnaA, crzA, gprC y gprD), familia Ras (rasA, rasB y rhbA), y otros (ace2, medA, y srbA). Por último, también se comentan los efectos de los alérgenos de A. fumigatus (Asp f 1 a Asp f 34) en la AI. Los datos obtenidos generan un complejo rompecabezas, cuyas piezas serían factores de virulencia o diferentes actividades del hongo, que se deben reunir para obtener una visión conjunta de la virulencia de A. fumigatus. Los estudios de expresión mediante microarrays de ADN podrían ser útiles para entender esta compleja virulencia, y para detectar dianas para desarrollar métodos rápidos de diagnóstico y nuevos agentes antifúngicos.

Palabras clave:
Aspergillus fumigatus
Virulencia
Aspergilosis invasora
Genes y moléculas
Patogénesis
Termotolerancia
Respuesta inmune
Pared celular
Toxinas
Toma de nutrientes
Señalización y regulación
Alérgenos
Texto completo

Aspergillus fumigatus is a well adapted saprophytic mold that produces large number of small airborne spores that can survive a wide range of environmental conditions, and accordingly are abundant in soil and decaying organic matter. Due to the 10,000–15,000l of air we inhale each day, humans are continuously in contact with these asexual spores17 and it is estimated that an individual inhales several hundred conidia per day.154 It is remarkable that, despite this constant exposure, most humans do not develop any illness attributable to these spores. In immunocompetent hosts, these spores do not normally cause harm because they are eliminated by pulmonary defense mechanisms.17 However, in immunocompromised individuals, with altered or weakened immune responses, inhaled conidia are able to develop pulmonary mycoses known as aspergillosis. Aspergillosis can be regarded as a broad spectrum of diseases, each related to a spectrum of abnormal immune responses of the host.217 Among them invasive aspergillosis (IA) stands out, with mortality rates greater than 50%, reaching 95% in certain situations17,173 (Fig. 1). The higher mortality observed in the infections by A. fumigatus appears to be due to a weakened immune response, to the virulence of the microorganism itself and also, probably, to delays in establishing a diagnosis, which can prevent the success of treatments.72 About 40 of the 250 species of Aspergillus have been reported to be human pathogens,126 but although the spores of A. fumigatus are a small proportion of all the airborne spores within a hospital (0.3%), this fungus causes approximately 90% of the systemic infections due to Aspergillus.40 Given this, it has been postulated that A. fumigatus has characteristic virulence factors.

Fig. 1.

Model of invasive aspergillosis development. (A) First step of colonization and invasion of pulmonary epithelium. (B) Invasion of blood capillaries and haematogenous dissemination of hyphal fragments, galactomannan and other molecules. (C) Dissemination and first step of invasion of deep organs.

(1,01MB).

The genome sequence of A. fumigatus has been made available recently.201 About 9,630 predicted protein-coding genes have been described of which one-third have unknown functions.85 During infection, fungi encounter dynamic changes in host conditions to which they must continually adapt to survive. This adaptation often requires substantial metabolic reprogramming,91,263 with the simultaneous expression of virulence factors that mediate host cell damage.51 A successful invasion strategy can involve large-scale alterations in protein expression and/or cellular differentiation209. The degree of host immunosuppression needed to develop IA could reduce the requirement for adaptive responses for infection in A. fumigatus. The importance of A. fumigatus infections is reflected by a number of reviews that have been published in the last few years concerning its biology and pathology, and the great effort being made to identify virulence factors.13,19,21,40,41,69,72,112,133,153–155,183,193,208,232,272,285 All these studies have demonstrated that the virulence of this fungus is multifactorial and is due to a combination of biological characteristics of the fungus and the immune status of the patient. Some of these characteristics include the small size of its conidia (2–3μm in diameter) which allows them to reach the human pulmonary alveoli, its thermotolerance and resistance to oxidative stress, and its high growth rate and nutritional versatility, among others. Other fungi share some of these features, but A. fumigatus possibly possesses a unique combination of different traits that make it the primary pathogenic mold in the world.208

With the use of sequenced genomes, we begin to be able to dissect some complex networks of fungal gene interactions such as metabolic regulation, autophagy, and sexuality. Although it is currently unclear, some authors have suggested that these networks have certain effects on the adaptive response of A. fumigatus to infections. Autophagy helps organisms to survive periods of nutrient stress by providing a source of recycled intracellular nutrients to fuel essential cellular functions.209 In Saccharomyces cerevisiae, approximately 30 genes have been identified and collectively termed autophagy-related genes (ATGs)127,128 and many of these are highly conserved across eukaryotes. In A. fumigatus, autophagy is required for conidiation, hyphal foraging, and maintenance of metal-ion homeostasis (all related to nutrient deficiency).235,236 Nutrient deprivation is one of the antimicrobial mechanisms of the host, however, the number of secreted hydrolases encoded by the genome of A. fumigatus,201,239 may allow this fungus to obtain nutrients from mammalian tissues without activating the autophagic network209. The fact that other pathogenic fungi use this mechanism to adapt to the host makes autophagy a putative virulence factor that should be considered carefully in the future.

A. fumigatus, like many other clinically important fungal species, has traditionally been considered an asexual organism. However, the teleomorph phase of this fungus has been discovered and named Neosartorya fumigata.204 The genome sequence of this fungus has made it clear that it occurs in two idiomorphs, MAT1-1 and MAT1-2, and strains of the two opposite mating types occur at the same frequency and are found in close proximity to each other.204 Successful mating was obtained between unrelated, clinical isolates of A. fumigatus, and requires the presence of both mating-type idiomorphs.270 Others authors showed the need for the expression of MAT1-1 and MAT1-2, as well as the expression of genes that encode factors involved in this process, such as genes encoding for sex pheromones and pheromone receptors.80,212 For example, the nsdD gene, a conserved regulator of cleistothecium development, could be related to hyphal fusion and hence heterokaryon formation.270 Heterothallism has now been discovered in four species of Aspergillus that affect human health or have an economic impact, namely A. fumigatus, A. parasiticus, A. flavus, and N. udagawae, but these fungi appear to have relatively low levels of fertility compared to other heterothallic or homothallic species of Aspergillus and require unusually precise environmental parameters to complete their sexual cycle.142 There are different interpretations of this low fertility. Some authors favor the hypothesis that while fertility of these species is on the decline, this is compensated by their proficiency to reproduce asexually in a wider range of environmental conditions.142 Other authors believe that the maintenance of all the machinery required for sex and the limitation of their access to sexual reproduction, has enabled the pathogenic fungi to proliferate rapidly in their environmental niche, but also to undergo genetic exchange, via sexual reproduction, in response to stressful conditions, for example, new environments, different host organisms, or changes in the human host, such as antimicrobial therapy.200 Highly dynamic changes in A. fumigatus populations have been observed within a clinical setting, with new populations found in just a few months,11 and coinfections with different related species of the Aspergillus genus have already been reported.207 These data imply that there may be coinfections with different mating type strains, and surprisingly the possibility that mating could occur in hosts during fungal infection. The presence of a sexual cycle in A. fumigatus would have significant medical implications. Some data suggest a possible association between one idiomorph, the MAT1-1 mating type, and A. fumigatus invasiveness that might contribute to increased virulence and/or resistance to antifungal agents.7 The study of sexual reproduction of this fungus and its possible relationship with virulence will remain a topic of interest in the coming years.6

The intention in this review is to give a general overview of the genes and molecules which have been associated with fungal virulence in the literature, the activities which they can perform and the importance that they could have in the development of IA.

Genes and molecules related to A. fumigatus virulence

Virulence factors are defined as pathogen determinants that cause damage to the infected host.50 This definition includes genes the deletion of which reduces virulence of the reference strain without affecting normal growth, excluding therefore, genes encoding biosynthetic proteins.208 Other genes related to A. fumigatus virulence, like catalases or secreted proteases, do not fit with this definition due to the redundancy of their gene families, and the difficulty of developing disruption of all the genes of a family in a single strain. Nevertheless, all of the genes that help and promote the growth of A. fumigatus in its environmental niche are also implicated in the pathogenesis of aspergillosis in the human host, and hence have to be considered as possible targets for new antifungal agents.13

The genes and molecules related to A. fumigatus virulence can be classified according to the process they are involved in, e.g., thermotolerance; cell wall composition and maintenance; resistance to the immune response; toxins; nutrient uptake during invasive growth; signaling, metabolism regulation and response to stress conditions; and allergens.

Thermotolerance

A. fumigatus is a thermophilic fungus able to grow at 55°C and survive at more than 75°C,26,241 an essential ability to thrive in decaying organic matter and to infect mammalian hosts. Therefore, genes related to thermotolerance may also contribute to the virulence of this mold.31 Until now, only four genes studied have been found to be necessary for thermotolerance (Table 1). The thtA gene is essential for A. fumigatus growth at 48°C but does not contribute to the pathogenicity of the species.54 Similarly, the afpmt1 gene codes for an o-mannosyltransferase, necessary for growth over 37°C, but is not involved in virulence.311 A putative α-1,2-mannosyltransferase coded by afmnt1 was also shown to be necessary for growth at 48°C.289 These authors showed that the Δafmnt1 mutant grows normally at 37°C, and that the observed growth defect of the mutant at 48°C can be attributed to cell wall instability resulting in leakage at the hyphal tips. This Δafmnt1 mutant was attenuated in a mouse model of infection, and showed an increased sensitivity to azoles.289 Likewise the deletion of the cgrA gene, which is involved in ribosome biogenesis, produced a hypovirulent strain in a murine model of invasive aspergillosis but not in a fruit fly model, being so related to the growth at 37°C.32

Table 1.

Aspergillus fumigatus thermotolerance genes and their relationship with virulence

Genes  Gene product  Function  Pathogenesis related activities  Effect of deletiona  Uses  References 
thtA  Unknown  Function unknown; essential for growth at 48°C    Normal virulence    54 
afpmt1  Afpmt1 (o-mannosyl-transferase)  Necessary for growth > 37°C  Cell-wall assembly and morphogenesis  Normal virulence    311 
kre2 afmnt1  Kre2/Afmnt1 (α-1,2-mannosyltransferase)Necessary to growth at 48°C  Cell-wall assembly and morphogenesisHypovirulent  New antifungal target289 
  Cell wall integrity     
cgrA  CgrA (nucleolar protein)  Ribosome biogenesis at 37°C  Growth at 37°C  Hypovirulent    32 
hsp1/asp f 12  Hsp1/Asp f 12 (heat shock protein, Hsp90 family)Chaperone  Chaperone activity and protein transport in growth at 37°C      136 
    Stress response during inflammation       
    Autoimmunity       
    Type I hypersensitivity       
a

Virulence assayed in animal model.

Cells exposed to non-lethal high temperatures become transiently resistant to subsequent heat shock, producing proteins named heat shock proteins (HSPs). Thermotolerance development is paralleled by expression of these HSPs.203 HSPs have been identified as molecular chaperones conserved between organisms.46 It has also been reported that a protein, Hsp1/Asp f 12,136 classified as a member of the family of Hsp90 could be related to thermotolerance. In addition, the protein Hsp1/Asp f 12 may also play a role in protective immunity and autoimmunity, as it is one of the immunodominant antigens in allergic aspergillosis.136

Nierman et al.201 studied the differences in gene expression between 30 and 37°C and between 30 and 48°C, and detected some upregulated genes at 37°C, but to date none of the genes related to pathogenicity have been found to be more highly expressed at 37°C than that at 48°C. They concluded that host temperature alone is not sufficient to turn on many virulence-related genes. On the other hand, Do et al.76 proposed that the thermal tolerance of A. fumigatus might be due to the efficient regulation of metabolic genes by HSPs. These authors used a state space model to examine transcriptional regulation and found a negative association between many HSPs and the metabolic genes they regulate. Little is known about A. fumigatus proteome changes at different temperatures, but a recent study has described 64 proteins to be up or downregulated from 30 to 48°C.4 Of them, Hsp 30/Hsp 42 and Hsp 90 showed the highest increase in expression during the heat shock response of A. fumigatus. More studies of changes in the proteome and their relationship with transcriptome changes could enhance our understanding of the thermoregulation of this fungus, and would help identify new possible targets for IA treatment.

Cell wall composition and maintenance

The cell wall is the main line of defense of the fungus against a hostile environment providing structural integrity and physical protection to the cell. The fungal cell wall is also the structure responsible for the interaction with the host and their components are often the targets of the host immune system during fungal infections. In A. fumigatus, the cell wall is mainly composed of polysaccharides (at least 90%) and proteins.98 Among the polysaccharides there are linear β(1–3)-glucans (20–35%) branched with β(1–6) links (4%); linear β(1–3/1–4)-glucans (10%); α(1–3)-glucans (35–46%); chitins; and galactomannans (20–25%).98,152,156Fig. 2 shows a schematic drawing of the cell wall structure. The genes and molecules related to the cell wall and virulence included in this review are listed in Table 2. Additional layers in the outer part of the cell wall may be also present. A layer of hydrophobic components is detected on both hypha and conidia, and a melanin layer only on the conidia. The effect of melanin and hydrophobic components on the immune response is addressed in the next section. Further, the presence of sialic acids has been detected on the surface of conidia. These sialic acids are unsubstituted N-acetyl-neuraminic acids linked to galactose by α-2,6 bonds29 and could play an important role in their adhesion to the extracellular matrix.292

Fig. 2.

Scheme of Aspergillus fumigatus cell wall.

(1,01MB).
Table 2.

Genes and proteins related with cell wall structure and virulence

Molecules/genes  Gene product  Function  Pathogenesis related activities  Effect of deletiona  Uses  References 
β-(1-3)-glucan    Cell-wall integrity  Immunomodulator    Diagnosis  29,43,117 
      Recognition and cellular adhesion       
fks1  Fks1 (catalytic subunit of β-(1–3)-glucan synthase complex)  Synthesis of β-(1–3)-glucan  Essential for fungal growth    Echinocandin target  189 
      Cell-wall assembly and morphogenesis       
rho1–4  Rho1-4 (Regulatory subunit of β-(1–3)-glucan synthase complex)  Synthesis of β-(1–3)-glucan  Cell-wall integrityCytoskeleton control    New antifungal target?  75,114 
rom2  Activator of rho  No essential        98,114 
Glucanosyl-transferases         
gel1  Gel1 (Glucanosyl-transferase, GPI-anchored protein)  Elongation of β-(1–3)-glucan  Cell-wall assembly and morphogenesis    New antifungal target  187,190 
gel2  Gel2 (Glucanosyl-transferase, GPI-anchored protein)  Elongation of β-(1–3)-glucan  Cell-wall assembly and morphogenesis  Hypovirulent    191 
gel3–7  Gel3–7 (Glucanosyl-transferase, GPI-anchored protein)  Elongation of β-(1–3)-glucan  Cell-wall assembly and morphogenesis      98 
bgt1  Bgt1 (Glucanosyl-transferase)  β-(1–6) branching of β-(1–3)-glucan  Cell-wall assembly and morphogenesis      188 
eng1  Eng1 (β-(1–3)-endoglucanase)    Cell-wall assembly and morphogenesis      192 
ecm33  Ecm33 (GPI-anchored protein)    Cell-wall assembly and morphogenesis  Hypervirulent    52,240 
α-(1–3) glucan    Cell-wall integrity  Recognition and cellular adhesion       
ags1–2  Ags1–2(α-(1–3)-glucan synthases)  Sinthesis of α-(1–3) glucan  Cell-wall assembly and morphogenesis  Normal virulence    22 
ags3  Ags3 (α-(1–3)-glucan synthases)  Sinthesis of α-(1–3) glucan  Cell-wall assembly and morphogenesis  Hypervirulent    174 
Chitin    Cell-wall integrity  Antigen       
Chitin synthases  Cell-wall assembly and morphogenesisImmunomodulator (generation chito-oligosaccharides)?    New antifungal target?   
chsA  ChsA (Chitin synthase class I)  Synthesis of chitin        195 
chsB  ChsB (Chitin synthase class II)  Synthesis of chitin        195 
chsC  ChsC (Chitin synthase class III)  Synthesis of chitin    Normal virulence    177 
chsD  ChsD (Chitin synthase class VI)  Synthesis of chitin    Normal virulence    179 
chsE  ChsE (Chitin synthase class V)  Synthesis of chitin    Normal virulence    178 
chsF  ChsF (Chitin synthase class IV)  Synthesis of chitin         
chsG  ChsG (Chitin synthase class III)  Synthesis of chitin    Hypovirulent    177 
afpigA  AfpigA (N-acetyl-glucosaminyl-transferase catalytic subunit, GPI-anchored protein)  Synthesis of chitin  Cell wall assembly and morphogenesis  Hypovirulent    160 
Galactomannan    Cell-wall integrity  Extracellular antigen    Diagnosis  70,71 
      Immunomodulator       
      Recognition and cellular adhesion       
och1–4  Och1-4 (mannosyl-transferases)  Synthesis of mannan  Cell wall assembly and morphogenesis      98 
mnn9  Mnn9 (mannosyl-transferases)  Synthesis of mannan  Cell wall assembly and morphogenesis      98 
van1  Van1 (mannosyl-transferases)  Synthesis of mannan  Cell wall assembly and morphogenesis      98 
anp1  Anp1 (mannosyl-transferases)  Synthesis of mannan  Cell wall assembly and morphogenesis      98 
glfA  GlfA (UDP-gal-mutase)  Synthesis of galactofuran  Cell wall assembly and morphogenesis  Hypovirulent in low dose aspergillosis model  New antifungal target  98 
afmp1/asp f 17  Afmp1p/Asp f 17 (Galactomannoprotein)    Antigen, Type I hypersensitivity    Candidates for sero-diagnosis  306 
afmp2  Afmp2 (Mannoprotein)    Antigen    Candidates for sero-diagnosis  56 
pmi  Phosphomannose isomerase  Cell wall synthesis, morphology, conidiation, energy production  Cell wall assembly and morphogenesis      84 
Mannosyl-transferases      New antifungal target?   
afpmt1  Afpmt1 (o-mannosyl-transferase)  Glycosylation of proteinNecessary for growth >37°C  Cell-wall assembly and morphogenesis  Normal virulence    311 
afpmt2  Afpmt2 (o-mannosyl-transferase)  Glycosylation of proteinActin re-arrangement  Cell-wall assembly and morphogenesis  Normal virulence    83 
afpmt4  Afpmt4 (o-mannosyl-transferase)  Glycosylation of protein         
kre2/afmnt1  Kre2/Afmnt1 (α-1,2-mannosyltransferase)  Glycosylation of proteinNecessary to growth at 48°CCell wall integrity  Cell-wall assembly and morphogenesis  Hypovirulent  New antifungal target?  289 
afmnt2–3  Afmnt2-3 (α-1,2-mannosyltransferase)  Glycosylation of protein  Cell-wall assembly and morphogenesis       
afcwh41  Afcwh41 (α-glucosidase)  Cell wall integrity  Cell wall assembly and morphogenesis  Normal virulence    309 
a

Virulence assayed in animal model.

The cell wall consists in a polysaccharide-based three-dimensional network and is now seen as a dynamic structure that is continuously changing as a result of the modification of culture conditions and environmental stress.152 The maintenance of cell wall integrity and functionality as well as changes in cell wall composition to adapt to the environment of the host could be involved in pathogenicity. Genes participating in the biosynthesis of most of A. fumigatus cell wall components have been identified.98 The study of these genes has revealed that mutant strains for enzymes required to synthesize cell wall polysaccharides were at least as virulent as the reference strain on almost all occasions.

The major polysaccharides in the A. fumigatus cell wall are the α(1–3)-glucans, and these have been shown to contribute to the virulence of diverse fungal pathogens. In particular, three α(1–3)-glucan synthase genes, ags1, ags2, and ags3, have been identified and were found to be responsible for cell wall α(1–3)-glucan biosynthesis. The Δags1 and Δags2 strains were not defective in virulence,22 while the Δags3 mutant was hypervirulent in an experimental mouse model of aspergillosis.174 Hypervirulence was correlated with an increased melanin content of the conidial cell wall, which could protect the cells from oxidative stress, and a quicker germination rate, that could evade macrophage killing. These authors did not observe significant changes in cell wall composition of the mutants, probably because of the redundancy between ags1 and ags3.13

β(1–3)-glucan branched with β(1–6)-glucan form the skeleton of the wall, and these are covalently bound to chitin and β(1–3/1–4)-glucan. This component is an important fungal pathogen-associated molecular pattern (PAMP) being recognized by receptor dectin1 on immune cells,29,43 and has different types of biological activity, triggering the activation of complement and inflammatory responses through mediators such as leukotrienes and TNFα117. β-glucan is a compound that is present in almost all fungi and has been used for the diagnosis of invasive mycosis,117 its kinetics correlating very well with that of galactomannan in patients with IA.218 Several authors have reviewed the synthesis of this component.78,98 Briefly, β(1–3)-glucan synthase is a transmembrane complex formed by several different proteins.78,98 The fks1 gene encodes the catalytic subunit and some of the four rho genes (rho1–4) detected in A. fumigatus and may be the regulatory subunit of glucan synthase.78,189 Although it is not a real virulence factor, Fks1 is essential for the fungus and its interest lies in being the target for the antifungal echinocandins. Research has also indicated that Rho1 and Rho3 are involved in controlling cell wall integrity and the cytoskeleton, and these are localized in the hyphal tip.75 Therefore, in the future, Rho molecules could also be potential targets for developing new antifungal agents.

A. fumigatus has at least seven chitin synthase encoding genes, but just four of them have been assayed for virulence: chsC, chsD, chsE, and chsG.14,177–179 Only chsG seems to have an influence on virulence, with a ΔchsG mutant strain having been shown to produce lower mortality rates than the reference strain in a mice infection model.177 However, these results could also be explained by redundancy of this type of enzymes.

The galactomannans in the cell wall are composed of mannose chains (α-mannan), shorter than those of yeasts, with branches formed by small side chains of five molecules of β(1–5)-galactose linked to mannan.98 Galactomannan synthesis requires mannosyl- and galactosyl-transferases. In the A. fumigatus genome there are orthologs of the S. cerevisiae genes related to mannan synthesis, four of the OCH genes, that initiate the synthesis of mannan chains, and orthologs of MNN9, VAN1, and ANP1 genes, which encode for mannosyltransferases.98 The functional role of each gene remains unknown. Galactofuranose biosynthesis starts with the isomerization of UDP-galactopyranose to UDP galactofuranose by UDP galactomutase encoded by the glfA gene. ΔglfA strains displayed attenuated virulence in a low-dose mouse model of IA and showed an increased susceptibility to various antifungal agents.244 UDP-galactomutase thus appears to be an appealing target for adjuvant therapy due to its absence from mammalian cells.244 This galactomannan could be a PAMP of the fungus, and useful for adhesion to host components such as fibronectin and laminin, or to interact with pentraxin 3 and other surface receptors of macrophages, dendritic cells, and Langerhans cells.29,112 Galactomannan is the principal exoantigen released during tissue invasion154 and may activate the innate immune response away from the focus of the infection. At present, the galactomannan produced and released by A. fumigatus is used in a commercial test for the diagnosis of IA (Platelia®Aspergillus).70,71

Several proteins of the cell wall are also mannosylated. For example, afmp1 and afmp2 genes encode for a galactomannoprotein and a mannoprotein, respectively. Their role in virulence has not been investigated, but it is worth mentioning them as they are antigenic determinants and therefore possible candidates for serodiagnosis.56,232,306 The addition of N-linked and/or O-linked oligosaccharides is a common modification of cell wall proteins. Mannosyltransferases play a crucial role in this process and most likely are also engaged in the generation of other glycoconjugates. Mannosyltransferases are localized in intracellular compartments of the secretory pathway, e.g., the Golgi apparatus or the endoplasmic reticulum (ER)289 and initiate mannosylation of secretory proteins. In A. fumigatus three members of O-mannosyltransferases, orthologs of PMT family of S. cerevisiae, afpmt1, afpmt2 and afpmt4 have been detected. Two of these have been studied but were found not to be necessary for virulence. The Δafpmt1 mutant showed sensitivity to high temperatures, as mentioned above in the thermotolerance section, and also defects in growth and cell wall integrity, thereby affecting cell morphology, conidium formation, and germination in A. fumigatus.311 Reduced expression of the afpmt2 gene also led to delayed germination, retarded hyphal growth, reduced conidiation, and defects in cell wall integrity; but growth was not found to be temperature-sensitive.83 The reduced production of Afpmt2 also caused actin rearrangement to fail.83 The afpmt4 gene has not yet been studied. The genome of A. fumigatus also harbors three putative α-1,2-mannosyltransferases genes with homology with members of KTR family of S. cerevisiae. One of these, kre2/afmnt1, was studied by Wagener et al.289 and given the findings this has been discussed above in the thermotolerance section. The function of the other two genes still remains unknown. The importance of α-1,2-mannosyltransferases for the synthesis of O- and N-linked carbohydrates and their possible role in the generation of other glycoconjugates, as well as the fact that humans do not possess any homologous enzymes, make α-1,2-mannosyltransferases promising targets for novel antifungal therapies.289

Other proteins present in the cell wall and related to virulence are linked to glycosyl-phosphatidyl-inositol (GPI) motifs. The glucanosyltransferases are enzymes linked to the cell membrane and the cell wall by GPI motifs. Some of these enzymes are thought to participate in the elongation of β(1–3)-glucan side chains.13 For example, the Gel family which is composed of seven proteins coded for by gel1–7. One of these enzymes, encoded by the gel2 gene, was observed to be related to virulence in a coinfection study. Specifically, the presence of DNA from a Δgel2 mutant strain was lower than the DNA of the reference strain in the lungs of coinfected mice.191 Another gene, afpigA, encodes the catalytic subunit of a complex that catalyzes GPI anchor biosynthesis. The GPI anchor is not essential for viability, but does seem to be required for cell wall integrity, morphogenesis, and virulence in A. fumigatus, and accordingly disruption of this gene caused a hypovirulent strain in a model of infection.160 However, the deletion of the ecm33 gene, that codes for a GPI-linked protein, enhanced virulence and resulted in a higher rate of germination, with more resistant conidia but more susceptible hypha.52,240

The phosphomannose isomerase enzyme, Pmi1, is essential for viability and plays a central regulatory role in both cell wall synthesis and energy production in A. fumigatus. The deletion of this gene led to phenotypes showing defects in cell wall integrity, abnormal morphology, and reduced conidiation,84 but their effect on virulence was not tested.

Zhang et al.309 identified a gene in A. fumigatus encoding an α-glucosidase, afcwh41, involved in cell wall integrity, polarity, septation and conidiation, probably by affecting the proper function of the proteins required for cell wall synthesis. However, this gene was not essential for hyphal growth and virulence.

In addition, the polysaccharide matrix of the cell wall, mainly composed of α-glucans and galactomannans, can bind the hyphae of a colony to generate a biofilm. Such biofilms may have an impact on virulence increasing the resistance to antifungals, and concentrating the extracellular enzymes produced during growth, which are also necessary for tissue colonization and infection.25 They might also help fungi to resist the immune response, although more studies are needed.

Genes and molecules associated with resistance to immune response

As mentioned before, the small size of resting Aspergillus conidia means that some of the inhaled conidia are able to reach the respiratory zone of the lungs, beyond the ciliated epithelium. Various genes and molecules on surface structures of A. fumigatus form a set called PAMPs that interact with and activate the immune system. Host defense relies on soluble and cellular pattern recognition receptors; activation of the effector mechanisms of innate immunity, including the antimicrobial mechanisms of resident leukocytes in the lung, such as alveolar macrophages and dendritic cells; recruitment of other leukocytes; and activation of recruited leukocytes after their arrival at the site of infection. Several reviews focusing on immune response to A. fumigatus infections have been published in the recent years.17,55,69,112,185 With these defenses weakened, conidia are able to germinate and form hypha within 12–15h of arrival.217

In addition to the weakening of host immune response, A. fumigatus has a combination of characteristics that helps the fungus to evade or resist to immune response (Table 3). Pigmentation on A. fumigatus conidial surface has been shown to affect virulence by limiting C3 complement deposition and neutrophil activation.275 Further, A. fumigatus has demonstrated an ability to bind Factor H, FHL-1, and C4BP on their surface to down-regulate the complement cascade,27,180,288 and to produce a soluble complement-inhibitory factor, which may be lipid derived, that prevented the activation of the alternative pathways.293,294 Moreover the thick fungal cell wall is largely resistant to direct lysis by the terminal membrane attack complex of the complement system.132

Table 3.

Genes and molecules associated with resistance to host immune response

Molecules/genes  Gene product  Function  Pathogenesis related activities  Effect of deletiona  Uses  References 
rodA/hyp1 and rodB  RodA/Hyp1 and RodB (Rodlets)  Dispersion and fixation to soil surfaces  Oxidative stress (ROS) protection  Normal virulence    215,252,274 
      Adhesion       
DHN-melaninb    Conidium protection  ROS protection       
    Cell wall assembly  Reduction complement and neutrophil activation       
    Expression adhesins  Adhesion       
Gen cluster  DHN-melanin synthesis          278 
pksP/ alb1  PksP/Alb1 (Polyketide synthetase)DHN-melanin biosynthesis  ROS, phagocytosis and complement binding protection  Hypovirulent    42,120,150 
  cAMP signal transduction  Immunosuppression       
arp1  Arp1 (Scytalone dehydratase)  DHN-melanin biosynthesis  Reduction complement binding  Normal virulence    277,278 
arp2  Arp2 (Hydroxynaphthalenes reductase)  DHN-melanin biosynthesis  Reduction complement binding  Normal virulence    277 
abr1  Abr1 (Putative iron multicopper oxidase)  DHN-melanin biosynthesis    Normal virulence    277,278 
abr2  Abr2 (Putative laccase)  DHN-melanin biosynthesis    Normal virulence    277 
ayg1  Ayg1 (2-acetoacetil 1,3,6,8-tetrahydroxynapthalene hydrolase)  DHN-melanin biosynthesis    Normal virulence    93,276 
Catalases             
catA  CatA (Conidial-specific catalase)  H2O2 degradation  ROS protection  Normal virulence    216 
cat1/catB  Cat1/CatB (Mycelial catalase)  H2O2 degradation  ROS protection  Δcat1cat2 hypovirulent    48,216 
cat2/katG  Cat2/KatG (Mycelial catalase)  H2O2 degradation  ROS protection  Δcat1cat2 hypovirulent     
catC    H2O2 degradation  ROS protection?      208 
catE    H2O2 degradation  ROS protection?      208 
Superoxide dismutases (Sod)             
sod1  Sod1 (Cytoplasmic Cu,Zn-Sod)  O2 degradation  ROS protection  Δsod1sod2sod3 Normal virulence    113,147 
sod2  Sod2 (mitochondrial Mn-Sod)  O2 degradation  ROS protection  Δsod1sod2sod3 Normal virulence    147 
sod3/asp f 6  Sod3/Asp f 6 (cytoplasmic Mn-Sod, Cross-reactive pan-allergen)O2 degradation  ROS protection  Δsod1sod2sod3 Normal virulenceDiagnosis  63,64,147 
    Type I hypersensitivity  Confirm ABPAc   
    Autoimmunity     
    Antigen     
sod4  Sod4 (Mn-Sod)  O2 degradation  ROS protection      147 
afyap1  Afyap1 (transcription factor)  Mediation ROS response  ROS protection  Normal virulence    158,225 
skn7  Skn7 (transcription factor)  Mediation ROS response  ROS protection  Normal virulence    145 
pes1  Pes1 (nonribosomal peptide synthase)Peptide synthesis  ROS protection  Hypovirulent    227 
  Stress resistance         
Fatty acid oxygenases             
ppoA  PpoA (fatty acid oxygenase)  Prostaglandin synthesis related  Stress oxidative resistance  ΔppoAppoBppoC hypervirulent    279 
ppoB  PpoB (fatty acid oxygenase)  Prostaglandin synthesis related  Stress oxidative resistance  ΔppoAppoBppoC hypervirulent    279 
ppoC  PpoC (fatty acid oxygenase)  Prostaglandin synthesis related  Stress oxidative resistance  ΔppoAppoBppoC hypervirulent    279 
Glutathione transferases             
gstA-E  GstA–E    Stress oxidative resistance      47 
Efflux transporters             
mdr1, mdr 2, and mdr-4  Mdr1, Mdr 2, and Mdr4 (ABC transporters)Toxic molecule expulsion  Toxic molecules expulsion    196 
  Antifungal resistance       
mdr3  Mdr3 (major facilitator superfamily)Toxic molecule expulsion  Toxic molecules expulsion    196 
  Antifungal resistance       
atrF  AtrF (ABC transporter)  Toxic molecule expulsion  Toxic molecules expulsion      256 
    Antifungal resistance         
abcA-E  AbcA-E (ABC transporters)Toxic molecule expulsion  Toxic molecules expulsion    68 
  Antifungal resistance       
msfA-E  MsfA-E (major facilitator superfamily)Toxic molecule expulsion  Toxic molecules expulsion    68 
  Antifungal resistance       
a

Virulence assayed in animal model.

b

DHN-melanin: melanin-1,8 dihydroxynaphthalene.

c

Allergic bronchopulmonary aspergillosis.

Various different types of behaviour have been detected on activation of immune cells through Toll-like receptors (TLR) by conidia and hyphae of this fungus. A. fumigatus conidia induce signal transduction after their recognition by TLR2 and TLR4; during tissue invasion, the conidia germinate into hyphae with loss of TLR4 stimulation, leading to a less pronounced stimulation of proinflammatory cytokines.53,199 TLR4-mediated proinflammatory effects have been demonstrated to be important in the protection against IA.28 Hence the tissue-invasive hypha of A. fumigatus is able to tilt the balance towards a non-protective Th2 response by a predominant TLR2 activation.53 On the other hand, it has been demonstrated that A. fumigatus conidia can bind and become internalized by human epithelial cell lines,214 which also may limit the induced levels of protective proinflammatory cytokines. These endocytosed conidia remained viable for relatively longer periods of time compared to conidia within macrophages,296 and may eventually germinate and disseminate.295

Genes and molecules involved in resistance to immune response could be considered defensive virulence factors as proposed by Osherov.208 It is well known that A. fumigatus has certain hydrophobic proteins on the surface of its conidial and aerial hyphae which help conidial dispersion, fixation to soil surfaces,154,164 and conidial adherence to the respiratory epithelium,274 and are related to the protection against the oxidative stress produced by alveolar macrophages.215 These proteins are clustered in microfibrils called rodlets. A. fumigatus has at least six genes that code for hydrophobins, but only rodA/hyp1 and rodB have been studied for virulence implication. The rodA gene encodes a small hydrophobic cysteine-rich polypeptide and the mutant strains for this gene showed high sensitivity to destruction by alveolar macrophages but were as virulent as the wild strain.215,274 However, the ΔrodA strain produced smaller lung lesions and weaker inflammatory response than the reference strain.252 On the other hand, the ΔrodB mutant did not show high sensitivity to killing by alveolar macrophages and did not lose their virulence.215

Another surface component of the fungi that has been associated with virulence is melanin, a pigment that protects the integrity of the genome in conidia from ultraviolet light, enzymatic lysis, and oxidation. The conidia of A. fumigatus possess a greyish-green melanin layer, absent in hyphae,305 which contributes to their survival and longevity in the environment.297 Some reviews have focused on the synthesis of melanin in pathogenic fungus and its importance.15,151,221 This pigment appears adhered to the cell wall of the A. fumigatus conidia, coming into direct contact with the host immune system.119,150 The presence of melanin on the surface of the conidium appears to protect the fungus in three ways. Firstly, as described above, the pigmentation on A. fumigatus conidial surface has been shown to affect virulence by limiting the activation of the complement cascade and neutrophils, and through interference with intracellular trafficking of phagocytised conidia.29,275 Secondly, the wild pigmented strains have a 10- to 20-fold greater resistance against reactive oxygen species (ROS) than the white mutant strains, presumably due to their capacity to quench and detoxify these ROS.154 Finally, the melanin could be masking β-glucan. In fact, the absence of pigment produces white conidia, decreases their virulence and makes them more sensitive to the action of H2O2 and sodium hypochlorite, and more susceptible to phagocytosis and to damage by macrophages in vitro.118,150,275 Melanin synthesis seems to be produced in the synthesis route of melanin-1,8 dihydroxynaphthalene (DHN-melanin) and is regulated by a cluster of six genes, pksP/alb1, ayg1, arp1, arp2, abr1, and abr2.41,93,150,275,276,278 Of all these, the most interesting, from the point of view of virulence, is the pksP/alb1 gene which encodes a polyketide synthase and catalyses the first step of this pathway. The deletion in other genes of this pathway produces conidia with different coloration, and in some cases with less deposition of complement (arp1 and arp2), but they do not have any obvious effects on the virulence.277,278 However, the ΔpksP/alb1 mutant has been shown to produce a smooth white conidium, increased C3 deposition on the surface and increased phagocytosis and killing of conidia.42,120 Resting conidia of this mutant strain express β-glucan abundantly on their surface encouraging its recognition through dectin1 receptors. Moreover, a product of the pksP gene could act as an immunosuppressant due to the presence of a functioning pksP gene, which is associated with inhibition of phagosome–lysosome fusion following conidial phagocytosis,120 and may have a direct role in the virulence of the fungus in a murine infection model.150 Melanin is also a structural component of the conidial wall that is required for correct assembly of the cell wall layers and the expression at the conidial surface of adhesins and other virulence factors.221

A. fumigatus also has specific enzymes for detoxification of ROS produced by macrophages and neutrophils, such as five catalases (catA, cat1/catB, catC, catE, and cat2)48,208,215 and four superoxide dismutases (SODs): a cytoplasmic Cu/ZnSOD (Sod1), a mitochondrial MnSOD (Sod2), a cytoplasmic MnSOD (Sod3), and Sod4 displaying a MnSOD C-terminal domain.87,113,147 Deletion of catA, a conidial catalase, resulted in increased susceptibility of conidia to H2O2in vitro, but the virulence of the mutant strain did not change in a murine model.215. Disruptions of either cat1 or cat 2 genes, encoding the hyphal catalases, did not affect sensitivity to H2O2in vitro or the virulence of mutants in animal infection models.48,215 However, double mutant Δcat1cat2 exhibited reduced virulence in immunosuppressed rats.48,215 In any case, as noted above, the redundancy of these genes for detoxification of ROS makes it difficult to verify their relationship with the virulence of the fungus. Fungal SODs that detoxify superoxide anions could be putative virulence factors for this opportunistic pathogen. During growth, Sod1 and Sod2 were highly expressed in conidia whereas Sod3 was only strongly expressed in mycelium and Sod4 was weakly expressed compared to other SODs.147 The deletion of Sod4 was lethal. The Δsod1 and Δsod2 mutants showed an inhibition of growth at high temperatures and hypersensitivity to menadione, whereas the sod3 mutant had only slightly delayed growth at high temperatures. The triple sod1/sod2/sod3 mutant was characterized by a delay in conidial germination, lower rates of conidial survival over time during storage, the highest sensitivity to menadione and an increased sensitivity to killing by alveolar macrophages of immunocompetent mice. In spite of these phenotypes, no significant virulence difference was observed between the triple mutant and the parental strain in experimental murine aspergillosis models with immunocompromised animals.147 Recently, Lessing et al.158 investigated the enzymatic ROS detoxifying system by proteome analysis of A. fumigatus challenged by H2O2. These researchers discovered that many of the identified proteins and genes were apparently regulated by a putative S. cerevisiae YAP1 homologous gene. This gene codes for a bZip-type transcription factor that contributes to the response against oxidative stress. Deletion of this afyap1 homologous gene in A. fumigatus led to drastically increased sensitivity to H2O2, but this mutant strain did not show attenuated virulence in a murine model of Aspergillus infection.158 These data have been corroborated in another study by Qiao et al.225 Other researchers have suggested that catalase activity in the Δafyap1 strain could be sufficient or more than sufficient to provide protection during incubation with neutrophils or in vivo, than after exposure to H2O2in vitro.69 These authors also argued that this similarity in virulence could be due to the use of a severe immunosuppression model, which made it difficult to detect small variations in virulence between mutants and their reference strains. Another transcription factor that contributes to the response against oxidative stress in yeast is SKN7. The homolog of this gene in A. fumigatus showed a similar role to the YAP1 gene we have just discussed. The Δskn7 strain of A. fumigatus had an increased sensitivity to peroxides in vitro but this was not correlated with a modification of fungal virulence.145 These results suggest that reactive oxygen intermediates have a relatively low importance in the destruction of the hyphae and conidia of A. fumigatus. Other mechanisms, such as the production of nitric oxide by macrophages or lactoferrin by neutrophils, a molecule with an ability to sequester iron, could be more relevant in the immune response against this fungus.104,197,307 Therefore, the role of other genes and molecules of the fungus in combating stress should be studied.

Three glutathione transferases (GST) genes, termed gstAC in A. fumigatus, have also been described.47 The results from studying these genes suggested a role for these enzymes in the response of the organism to both oxidative stress and presence of xenobiotic compounds,47 but they have not been tested for virulence. It has also been suggested that the nonribosomal peptide synthetase gene, pes1, contribute to the resistance of A. fumigatus to oxidative stress. Disruption of this gene led to decreased fungal virulence in a moth model system, as well as an increased susceptibility to oxidative stress and neutrophil-mediated killing, in addition to altered conidial morphology and hydrophobicity.227

Three fatty acid oxygenases encoding genes (ppoA, ppoB, and ppoC) have also been tested for their role in the virulence of A. fumigatus. The triple mutant strain was found to be hypervirulent in an invasive murine model and showed increased tolerance to H2O2 stress relative to that of the wild type.279 These authors suggested that part of the increased virulence of the triple mutant strain might be due to the Ppo-generated prostaglandins, which could enhance host defense mechanisms, perhaps through initiation of inflammation responses involved in recruiting phagocytic cells.

Four genes that encode ATP-binding cassette (ABC)-type transporters (mdr1, mdr2, atrF, and mdr4), and one gene that codes for a protein of the major facilitator superfamily (MFS)(mdr3) related to azole resistance149,196,256 have been described in A. fumigatus. Other genes (abcA–E and mfsA–E) that encode for these types of transporter could be related to voriconazole resistance.68 These two classes of transporters or efflux pumps are associated with the membrane and could detoxify immune system components in a similar way to their involvement in resistance to antifungals.208 Today, thanks to genome sequencing of A. fumigatus, at least 327 genes that encode putative multidrug resistance efflux pumps have been reported, including 49 ABC-type genes, and 278 genes that encode MFS proteins.86,201

However, despite the varied capabilities possessed by the fungal pathogen to evade host detection, it should be emphasized that the normal host defense is generally effective against most fungal infections and the host has first to be in an immune suppressed state before it becomes susceptible to opportunistic pathogens.53

Toxins

Mycotoxins can be described as a chemically diverse group of low molecular weight organic substances produced by fungi. These substances are formed in the hyphae during growth, and may be actively expelled into the environment, or released after the death of the hyphae. The presence of preformed mycotoxins in conidia means that the toxins must be incorporated during conidiogenesis. However, these substances might be also produced during germination. Toxins are apparently produced by the fungus to protect itself from predators and competitors in its ecological niche,208 but they could also contribute to A. fumigatus pathogenesis, since they can directly attack the host (Table 4). Many of these toxins are secondary metabolites of these fungi. Depending on the mycotoxin, they can affect the synthesis of proteins, DNA and RNA, or alter the cell membrane, the consequences of which may be death or impairment of cellular functions.

Table 4.

Toxins related to the direct attack to the host organism

Molecules/genes  Gene product  Function  Pathogenesis related activities  Effect of deletiona  Uses  References 
Conidium toxin diffusible substance  Reversible macrophage inhibition      30,181 
Gliotoxin      Inhibition of macrophages, neutrophils, and T lymphocytes      10,81,206,213,262 
      Ciliostasis       
      Epithelial cells damage       
      Apoptosis       
Putative cluster gliotoxin synthesis (12 genes)         
gliP  GliP (nonribosomal peptide synthetase)Gliotoxin biosynthesis  Hypovirulent in cortisone treated non-neutropenic mice    61,137,261,269 
    Normal virulence in neutropenic mice     
gliZ  GliZ (transcriptional regulator)  Gliotoxin biosynthesis regulation    Normal virulence    37 
asp f 1/mitF/res  Asp f 1/MitF/Res (mitogillin, restrictocin)Ribotoxin  Protein biosynthesis inhibition  Normal virulenceDiagnosis  12,224,257 
    Cytotoxin     
    Apoptosis     
    Type I hypersensitivity     
aspHS  AspHS (hemolysin)  Hemolytic activity  Hemolytic and cytotoxic activity (erythrocytes, macrophages and endothelial cells)      135 
Festuclavine    Ergot alkaloid  Nervous and reproductive systems disfunction      59 
Fumigaclavine A–CErgot alkaloid  Nervous and reproductive systems disfunction      59 
Cluster ergot alkaloid genes        60 
dmaW/fgaPT2  DmaW/FgaPT2 (Dimethil-allyl-tryptophan synthase)  Ergot alkaloid biosynthesis        60 
fgaDH /fgaOx2  FgaDH /FgaOx2 (Dehydrogenase)  Ergot alkaloid biosynthesis        291 
fgaMT  FgaMT (4-dimethyl-allyl-tryptophan N-methyltransferase)  Ergot alkaloid biosynthesis        237 
aesA  AesA (Chanoclavine-I aldehyde alkene reductase)  Ergot alkaloid biosynthesis        58 
Fumitremorgin A–CNeurotropic toxins  Nervous system disfunctions       
Cluster fumitremorgin synthesis (nine genes)      171 
ftmA-I  FtmA-I  Fumitremorgin biosynthesis        106,107,125,171 
Verruculogen      Production on infections?       
ftmOx1  FtmOx1 (α-ketoglutarate-dependent dioxygenase)  Formation of verruculogen        264 
Fumagillin    Antitumor antibiotic (inhibition angiogenesis)Ciliostasis      45 
    Inhibition of endothelial cell proliferation       
Helvolic acid    Fusidanes (steroidal antibiotics)Inhibition macrophage (respiratory burst)      181 
    Ciliostasis and respiratory epithelium damage       
Cluster helvolic acid synthesis (nine genes)      166,182 
AfuOSC3  AfuOSC3 (oxidosqualene cyclase)  Helvolic acid biosynthesis        166 
Aflatoxin B1 and G1  Production on infections?      219 
laeA  LaeA (nuclear protein, Arg and His methyl-transferases homology)  Regulation of secondary metabolism and 10% genome expression    Hypovirulent    36,38,220 
a

Virulence assayed in animal model.

A diffusible, heat-stable substance, with a mass of less than 14kDa, can be rapidly extracted from the surface of the conidium. This diffusible substance has been shown to affect competent macrophages, inhibiting the respiratory burst, phagocytosis and the release of cytokines by macrophages,30,181 and its effect is reversible. This component has still not been identified, but may allow the fungus to remain in the lungs and express its pathogenic effects. In particular, it has been associated with the pathogenicity level of A. fumigatus strains, but not all strains produce it.30

Ergot alkaloids are a complex family of indole-derived mycotoxins that affect the nervous and reproductive systems of exposed individuals through interactions with monoamine receptors.59 The ergot alkaloids festuclavine and fumigaclavines A–C are present in or on conidia of A. fumigatus.59 An ergot alkaloid gene cluster in A. fumigatus genome has been described,60 of which the dmaW gene has been studied. This gene encodes a dimethylallyl tryptophan synthase that appears to control a determinant step in ergot alkaloid biosynthesis, as when dmaW was knocked out all known ergot alkaloids were eliminated from A. fumigatus.60 Another recently studied gene, easA encodes an enzyme which catalyzes the reduction of the chanoclavine-I aldehyde alkene to dihydrochanoclavine aldehyde, and facilitates an intramolecular reaction to generate the immediate precursor to festuclavine.58 Some other genes, like the 4-dimethylallyltryptophan N-methyltransferase encoded gene, fgaMT237 and a dehydrogenase gene, fgaDH/fgaOx2, that catalyzed the oxidation of chanoclavine-I to chanoclavine-I aldehyde,291 have also been reported. However, none of these genes have yet been tested for virulence.

Gliotoxin is the major and the most potent toxin produced by A. fumigatus.143 It belongs to the family of epipolythiodioxopiperazines, which are characterized by a disulfide bridge across a piperazine ring which is essential for their toxicity.97 Gliotoxin has several immunosuppressive roles including inhibition of macrophage phagocytosis, mitogen-activated T cell proliferation, mast cell activation, cytotoxic T-cell response, and monocyte apoptosis.81,194,262,301 It also inhibits the NADPH of neutrophils,280 suppresses ROS production and impairs neutrophil phagocytic capacity,206 reduces the ciliary movement of epithelial cells and leads to epithelial cells damage.10 It has also been reported that gliotoxin induces ROS-facilitated apoptotic cell death by activating the Bak gene of mice, a member of proapoptotic Bcl-2 family.213 It has been proven that this toxin is produced in experimental animal aspergillosis159,234 as well as in human IA, with serum concentrations of 166–785ng/ml in 80% of patients with IA.159 Although some studies have reported that a low proportion of strains produce this toxin,77,92 a recent study reported that gliotoxin is produced by more than 95% of A. fumigatus isolates from both clinical and environmental origins, while it is less often produced by other Aspergillus species.138 A putative cluster of 12 genes involved in gliotoxin biosynthesis was discovered.96 The gliZ gene controls expression of the remaining 11 genes in this cluster,37 while gliP encodes a multimodular nonribosomal peptide synthase that catalyzes the condensation of serine and phenylalanine, the first step of the pathway.16 In neutropenic models of IA, the mutant strains for these two genes were as virulent as the reference strain.37,61,137 Nevertheless, in non-neutropenic mice treated with cortisone, the virulence of gliP mutant strains was lower than the reference strains.261,269 These results suggest that gliotoxin induces neutrophil apoptosis261 and a direct role of gliotoxin in aspergillosis virulence in non-neutropenic immunocompromised individuals.

A. fumigatus is able to produce ribotoxins, proteins that have a highly specific activity against the sarcin/ricin domain universally preserved in 28S ribosomal RNA, inhibiting protein biosynthesis.123,124 One of these proteins is restrictocin, also known as mitogillin, encoded by the asp f 1/mitF/res gene. This toxin is related to the allergic process, since it is one of the immunodominant antigens of allergic aspergillosis.12 Mitogillin is secreted in vivo by A. fumigatus148 and has strong toxic effects that can cause cell death at low concentrations.224 Ok et al 205 showed in vitro that Asp f 1 is also able to induce cytokine release and apoptosis in human immature dendritic cells. This immunomodulator effect could be helping the immune evasion of A. fumigatus. However, the deletion of asp f 1/mitF/res did not affect fungal virulence in a neutropenic model of IA.257 The fungus also produces a hemolysin encoded by the aspHS gene. This molecule has hemolytic activity on rabbit and sheep erythrocytes, cytotoxic effects on macrophages and endothelial cells in vitro,135 and can be detected during infection in vivo304.

It is worth mentioning that in a recent study the levels of expression of certain of the genes discussed above (gliP, aspHS, asp f 1, and dmaW) were determined by real-time RT-PCR analysis, and higher expression was observed in vivo than in vitro.102 These results suggest an overexpression of these toxins during infection.

Other toxins produced by A. fumigatus are helvolic acid and fumagillin. Helvolic acid is part of a small family of steroidal antibiotics known as fusidanes. At high concentrations it can affect the oxidative burst of macrophages,181 the metabolism of low density lipoproteins254 and in vivo it induces ciliostasis and rupture of epithelial cells.10 On the other hand, fumagillin is an antitumor antibiotic that inhibits angiogenesis and in vitro directly inhibits endothelial cell proliferation and cilial movement in respiratory epithelium.45 The active concentrations of these toxins are considerably higher than those of gliotoxin, but it is still unknown in what concentrations are produced in vivo232. It has also been reported that fumitremorgin A302 fumitremorgin B165, and fumitremorgin C77, neurotropic toxins that cause tremors, seizures, and abnormal behavior in mice, are produced in a dose-dependent manner. Another toxin described to be produced by A. fumigatus that causes tremors is tryptoquivaline A.303 Further toxins, such as aflatoxin B1 and G1, and verruculogen, have been detected in culture filtrates of A. fumigatus, but their presence during infection has not yet been demonstrated.219 Other genes involved in the biosynthesis of these toxins have also been identified, such as ftmOx1, that encodes a non-heme Fe(II) α-ketoglutarate-dependent dioxygenase, which catalyses the endoperoxide formation of verruculogen in A. fumigatus.264 Fumitremorgin biosynthesis seems to be encoded by a cluster of nine genes, ftmA–I,171 and most of which have been described recently.106,107,125,171 A cluster of nine genes involved in helvolic acid biosynthesis has also been described.166,182 However, none of these genes have yet been tested for virulence.

The transcription factor leaA is a global regulator of secondary metabolite biosynthesis38 that modulates the expression of approximately a 10% of the genome of this fungus.220 The deletion of this gene in A. fumigatus blocked the production of almost all secondary metabolites, including gliotoxin,268 and a leaA mutant strain was hypovirulent after intranasal inoculation of neutropenic mice.36 These authors also showed that ΔlaeA mutants lost pigment production and their conidia were more susceptible than wild type A. fumigatus conidia to phagocytosis by macrophages.

Nutrient uptake in invasive growth

Mammalian organisms present a broad variety of microenvironments in which A. fumigatus must survive to cause disease, and these environmental conditions can rapidly change depending on the current stage of infection.298 Normal nutrient uptake systems, used in their ecological niche, might serve the fungus during infection, but it is possible that other systems could be activated by environmental conditions. Table 5 shows the major molecules and genes related to virulence covered in this section.

Table 5.

Genes and molecules related with nutrient uptake in invasive growth

Molecules/genes  Gene product  Function  Pathogenesis related activities  Effect of deletiona  Uses  References 
Enzymes    Nutrient obtention  Tissue destruction/invasion       
alp/asp f 13  Alp/Asp f 13, orycine (alkaline serine protease, Elastase)Protein degradation (elastin, collagen, fibrinogen and casein)Tissue destruction/invasion  Hypovirulent    121,130,228,271 
  Type I hypersensitivity       
alp2  Alp2 (cell wall alkaline serine protease)  Protein degradation (elastin)  Tissue destruction/invasion  Normal virulence    229 
asp f 18  Asp F 18 (vacuolar serine protease, related with Alp2?)Protein degradationTissue destruction/invasion  Normal virulence  249,251 
  Type I hypersensitivity     
mep/asp f 5  Mep/Asp f 5 (extracellular metalloprotease glycosylated)Protein degradation (collagen and elastin)Tissue destruction/invasion  Normal virulence  63,121,172,255 
  Type I hypersensitivity     
mepB  MepB (Intracellular metalloprotease)  Protein degradation    Normal virulence    115 
mep20  Mep20 (metalloprotease)  Protein degradation  Tissue destruction/invasion  Normal virulence    226 
pep/asp f 10  Pep/Asp f 10, aspergillopepsine F (aspatic endopeptidase)Protein degradation (collagen)Tissue destruction/invasion  Normal virulenceDiagnosis  63,157,231 
  Type I hypersensitivity     
pep2  Pep2 (aspatic protease cell wall-associated)  Protein degradation  Tissue destruction/invasion  Normal virulence  230 
ctsD  CtsD (extracellular aspartic protease)  Protein degradation (starvation condition produced)  Tissue destruction/invasion    287 
dppIV  DppIV (glycoprotein)Dipeptidyl-peptidase activity (Protein degradation)  Protein degradation (collagen, hormones and cytokine)      24 
    T lymphocyte activation       
dppV  DppV (glycoprotein)Dipeptidyl-peptidase activity (Protein degradation)Protein degradation (collagen, hormones and cytokine)    Diagnosis  23 
  T lymphocyte activation       
plb1-3  Plb1, Plb2 and Plb3 (phospholipases B)  Phospolipid degradationMembrane destruction  Tissue destruction/invasion      250 
Phospholipase C    Phospolipid degradationMembrane destruction  Tissue destruction/invasion      33 
Iron acquisition    Fe uptake         
sidA  SidA, L-ornithine hydroxilase  First common step of synthesis of siderophores  Iron acquisitionEssential to virulence  No virulent  New antifungal target  111,246 
sidC  SidC  Synthesis of ferrocrocin and hydroxyferricrocin  Intracellular iron storage  Hypovirulent  New antifungal target  247 
sidD  SidD  Synthesis of fusarinine C and triacetylfusarinine C  Extracelular Iron acquisition  Hypovirulent  New antifungal target?  247 
sidF  SidF  Synthesis of fusarinine C and triacetylfusarinine C  Extracelular Iron acquisition  Hypovirulent  New antifungal target?  247 
sidG  SidG  Synthesis of triacetylfusarinine C  Extracelular Iron acquisition  Hypovirulent  New antifungal target?  247 
sreA  SreA (transcription factor, GATA family protein)  Iron acquisition regulation    Normal virulence    248 
ftrA  FtrA (iron permease)  High affinity permease    Normal virulence    246 
fetC  FetC (Putative ferroxidase)          246 
mirB  MirB (Siderophore transport protein)  Siderophore transport gene        223 
mirC  MirC (Siderophore transport protein)  Siderophore transport gene        223 
amcA  AmcA (putative mitochondrial carrier for ornithine)  Putative mitochondrial carrier for siderophore precursor ornithine        223 
Zn acquisition             
zrfA  ZrfA (putative zinc transporter)  Zn acquisition in acidic pH  Zn acquisition      8 
zrfB  ZrfB (putative zinc transporter)  Zn acquisition in acidic pH  Zn acquisition      8 
zrfC  ZrfC (putative zinc transporter)  Zn acquisition in neutral or alkaline pH  Zn acquisition      9 
zafA  ZafA (zinc-responsive transcriptional activator)Induction of zrfC and asp f 2 expression in zinc-limiting mediaZn acquisitionEssential to virulence  No virulent  New antifungal target  9,186 
         
pacC  PacC (transcriptional regulator)  Repression of zrfC and asp f 2 at acidic pH        8,9 
N acquisition             
areA  AreA (transcriptional regulator)  Nitrate transport and processing regulator  N acquisition  Normal virulence    110 
rhbA  RhbA (GTPase, Ras-related protein)    N acquisition  Hypovirulent    211 
cpcA  CpcA (transcriptional activator)  Putative amino acid biosynthetic pathways regulator  N acquisition  Hypovirulent    134 
mcsA  McsA (methylcitrate synthase)      Hypovirulent  New antifungal target  116 
lysF  LysF (homoaconitase)  Lysine biosynthesis  Aminoacids biosynthesis and growth  Hypovirulent  New antifungal target?  162 
hcsA  HcsA (homocitrate synthase)First enzyme of α-aminoadipate pathway (lysine biosynthesis)Amino-acids biosynthesis and growthHypovirulent in pulmonary aspergillosis  New antifungal target?  245 
  Normal virulence in inoculation by intravenous via     
pyrG  PyrG (orotidine-5’-phosphate decarboxilase)  Pyrimidine biosynthesis  DNA biosynthesis and growth  Hypovirulent    74 
pabaA  PabaA (p-aminobenzoic acid synthase)  Folate biosynthesis  Folate biosynthesis and growth  Hypovirulent    44 
tpsA and tpsB  TpsA and TpsB  Trehalose biosynthesis  Response to heat shock  ΔtpsAtpsB hypervirulent    3 
a

Virulence assayed in animal model.

A. fumigatus can obtain important nutrients from destruction of host tissue. A. fumigatus secretes extracellular enzymes, most of them proteases, that degrade and recycle organic matter in the environment, but during infection they could serve to break down the structural barriers of the host and to obtain nutrients. As indicated above, one of the host antimicrobial mechanisms is nutrient deprivation, and the amount of secreted hydrolases encoded on the genome201,239 may allow A. fumigatus to obtain nutrients from mammalian tissues without the need to activate the autophagic network.209 Several articles have reviewed these proteases and their relationship with pathogenicity.112,133,208,232 Some of these proteases can degrade collagen and elastin, which are the main components of the lung matrix. Various researchers have demonstrated a clear link between elastase activity of A. fumigatus strains and their invasiveness,35,130 so the fungus seems to be able to adapt to the host environment increasing elastase activity.95 However, other authors found no statistical correlation between the existence of elastase or acid proteinase activity and the development of invasive disease.5 These enzymes include serine alkaline protease (Alp) from the family of subtilisins, which can degrade elastin, collagen, fibrinogen, and casein,130,228 and corresponds to the allergen Asp f 13; Alp2, a serine protease that is associated with the cell wall;229 and a vacuolar serine protease, the allergen Asp f 18.251 The extracellular metalloprotease Mep can degrade collagen and elastin,172,255 and is also known as Asp f 5 allergen. Other metalloproteases have been identified in A. fumigatus such as that encoded by the mep20 gene226 or the intracellular metalloproteinase encoded by the mepB gene, which appears to be associated with the cytoplasmic degradation of small peptides.115 Another group of extracellular enzymes produced by A. fumigatus are aspartic proteases, also called aspergillopepsins. Two aspergillopepsins have been identified, a secreted aspergillopepsin (Pep)157 which matches the known Asp f 10 allergen, and another one associated to the cell wall (Pep2).230,231 A novel aspartic protease, CtsD, has been described in culture supernants.287 The expression of the ctsD gene was absent under nutrient-rich conditions, but it was detected, in vivo, in a Galleria mellonella infection model.287 In culture supernatants of A. fumigatus two members of dipeptidylpeptidases family (Dpp) have also been detected, DppIV and DppV, which cut at the amino-terminal end of peptides and proteins. These enzymes can bind to collagen, and even to hormones and cytokines, and degrade them. Their role in T cell activation has also been described.23,24

Finally A. fumigatus also secretes phospholipases, which break the ester bond of phosphoglycerides and thus may destabilize the host cell membranes causing cell lysis.232 Activity of phospholipases A–D has also been detected in culture filtrates of A. fumigatus.34 The genes plb1, plb2, and plb3 of A. fumigatus encode for B phospholipases, which are characterized by their phospholipase, lysophospholipase, and lysophospholipase transacylase activity.250 Two of them, Plb1 and Plb3, are known to be secreted.250 The genome of A. fumigatus codes for at least another three putative secreted phospholipases.208 Although these enzymes have been considered virulence factors for other species such as C. albicans or C. neoformans, in clinical isolates of A. fumigatus the production of B phospholipases is lower than in environmental isolates, making unlikely, if not excluding, their involvement in the virulence of the fungus. This could be explained by the secretion of other phospholipases by A. fumigatus, such as phospholipase C which has not been detected in other species and is produced in a higher proportion in clinical than environmental isolates.33 It should however be noted that while high phospholipase production was found to be associated with development of invasive aspergillosis, not all isolates that caused invasive diseases have displayed high phospholipase activity.5

Different proteases may play unique or overlapping roles during pathogenesis, and is difficult to obtain evidence of them as individual virulence factors.298 Only one mutant strain of A. fumigatus in a 33-kDa protein, coded for by the alp/asp f 13 gene and which has elastase activity, has produces lower rates of mortality when neutropenic mice were infected by intranasal inoculation.130 However none of these extracellular enzymes, metalloproteases,115,121 alkaline proteases121,184,271 or aspartic proteases231 have demonstrated a direct role in virulence, probably due to their redundancy. It is worth noting that there are at least 99 putative secreted proteases for the A. fumigatus genome.168,201

Recently, the biosynthesis of trehalose has been linked to virulence in pathogenic fungi. Trehalose is a non-reducing disaccharide the expression of which increases during the life cycle of A. fumigatus. Its concentration also increases after heat shock but not in response to other types of stress and in this fungus it is related with reduction in pathogenicity.3 In A. fumigatus the proteins involved in threhalose biosynthesis are encoded by two genes, tpsA and tpsB. The deletion of both genes showed conidia with delayed germination at 37°C and susceptibility to oxidative stress. The double mutation was required to block the trehalose accumulation, and this double mutant was hypervirulent in murine model of IA and was also associated with alterations in the cell wall and resistance to macrophage phagocytosis.3

The uptake of certain components is essential for most organisms and the ability to acquire these components in limiting environments, such as in the human host, is a necessary requirement for virulence of human pathogens. One of these limiting components in the human host is iron. A. fumigatus can acquire iron in two different ways, by reductive iron assimilation and by siderophore-assisted iron uptake, both of which are induced upon iron starvation246. The reductive mechanism for iron assimilation consists in the reduction of ferric to ferrous iron and the subsequent uptake of ferrous iron by the FtrA/FetC complex.246 Inactivation of the high affinity iron permease FtrA did not produce a reduction in virulence in a murine infection model, suggesting that virulence of A. fumigatus does not depend on reductive iron assimilation.246 By contrast, the inactivation of the sidA gene, which catalyses the first step of the biosynthesis of all known siderophores, namely the hydroxylation of L-ornithine,111 was found to be absolutely essential for virulence.111,246 Siderophores are low-molecular weight proteins (Mr<1500), that act as ferric iron-specific, high-affinity chelators.198A. fumigatus possesses at least four siderophores: fusaricine C and triacetylfusaricine C are excreted for iron acquisition; and ferricrocin and hydroxyferricrocin are used for intracellular iron storage.247 The study and deletion of the four genes needed for the biosynthesis of these two types of siderophores, sidC, sidD, sidF, and sidG revealed that the nonribosomal peptide synthetase, sidC, is involved in intracellular siderophore biosynthesis and that this type of siderophore is required for germ tube formation, asexual sporulation, resistance to oxidative stress, catalase A activity, and virulence.247 The strains with deletion of sidD and sidF genes, which are involved in biosynthesis of extracellular siderophores, were found to have attenuated virulence in animal infections and partial sensitivity to oxidative stress.247 The acquisition of iron is also regulated by the protein SreA, of the GATA family, but as this gene acts as a repressor under high iron conditions, its genetic inactivation results in overaccumulation of iron. Although the ΔsreA strain showed increased sensitivity to iron and oxidative stress, it did not demonstrate a role in virulence in a murine infection model.248 Certain other genes, including amcA, a putative mitochondrial carrier for the siderophore precursor ornithine, and the siderophore transport gene mirB, have shown to be upregulated during iron starvation conditions,223 but have not yet been studied for their role in virulence. As humans do not produce siderophores, most of these genes, and particulary sidA and sidC, could be good targets for new antifungal therapies.

Zinc is another essential element for fungal growth. The genome of A. fumigatus contains three putative zinc transporter-encoding genes (zrfAC) whose expression is regulated by both pH and the environmental concentration of zinc.8,9 Two of these transporters, coded by genes zrfA and zrfB, are transcribed at higher levels and are required for fungal growth under acidic zinc-limiting conditions, while they are not required for growth in neutral or alkaline zinc-limiting media,286 the conditions found in lung tissues. It has recently been described that the zrfC gene encodes a transporter devoted to obtaining zinc from alkaline zinc-limiting media.9 This gene is adjacent to the asp f 2 gene, which encodes an allergen secreted by A. fumigatus. In alkaline and extreme zinc-limiting conditions, the transcriptional regulators ZafA and PacC induce the simultaneous transcription of zrfC and asp f 2 genes. Specifically, ZafA upregulates the expression of zrfC and Asp f 2 under zinc-limiting conditions regardless of the environmental pH, whereas PacC represses the expression of these genes under acidic growth conditions.9 The role in virulence of these transporters has not yet been studied. However, the deletion of the transcriptional regulator zafA gene impairs the germination and growth capacity of A. fumigatus in zinc-limiting media and the ΔzafA strain abrogated A. fumigatus virulence in a murine model of IA.186 The zafA gene may constitute a new target for the development of chemotherapeutic agents against Aspergillus, especially since no zafA orthologues have been found in mammals.186

Nitrogen metabolism has also been related to A. fumigatus virulence. Several sources of nitrogen may be used by A. fumigatus, such as nitrate or amino acids released during host tissue destruction or biosynthesized in their metabolism. The proteins that are involved in nitrate transport and processing are transcriptionally regulated by the areA gene.69 The study of an ΔareA mutant strain in a neutropenic model of IA showed similar virulence to the reference strain. However, this mutant strain presented a delayed-growth phenotype in the lung tissue.110 The expression of another gene, rhbA, was induced under nitrogen starvation conditions.210 This gene codes for a Ras-related protein and has been considered a virulence factor because ΔrhbA mutant strains displayed a significantly lower virulence in a murine infection model.211 Amino acids can be another source of nitrogen for microorganisms but not all amino acids are readily available in mammalian hosts during infections.298 The cpcA gene of the Cross-Pathway Control (CPC) system (also known as General Control of amino acid biosynthesis) is activated in amino acid-limiting conditions. It has been proposed that this system regulates the A. fumigatus amino acid biosynthetic pathways, and the deletion of this gene produced mutants with decreased virulence.134 The deletion of essential functional genes, such as lysF, which encodes a homoaconitase of lysine biosynthesis, produces mutants with decreased virulence in murine models of IA.162 The fungal α-aminoadipate pathway is also essential for lysine biosynthesis, and the first pathway specific enzyme, homocitrate synthase (HcsA), has recently been described.245 The hcsA deletion mutant was lysine auxotrophic, but although virulence of the mutant was strongly attenuated in murine models of bronchopulmonary aspergillosis, the mutant retained full virulence when injected intravenously.245 Therefore, inhibition of fungal lysine biosynthesis does not appear to provide a suitable target for new antifungals, at least not for disseminating invasive aspergillosis. The degradation of amino acids could be important in A. fumigatus pathogenesis, and during invasive growth the amino acid metabolism can produce propionyl-CoA accumulation, which is a toxic metabolite. The fungus metabolizes propionyl-CoA via the methylcitrate cycle.169 Recently the deletion of mcsA gene, which codes for the first enzyme of the methylcitrate cycle, a methylcitrate synthase, has been studied. This mutant strain displayed attenuated virulence in a murine model of IA, so that this activity does provide a suitable target for new antifungals.116

The genome of A. fumigatus contains some putative genes for the uptake of other essential elements such as magnesium or phosphate, but none of them have yet been studied for their role in virulence. That is the case, for example, of four putative inorganic phosphate transporters and six secreted acid phosphatases.208

Like other essential genes, strains with deletion of the pyrG gene that encodes an orotidine-5′-phosphate decarboxylase and catalyzes the last step of pyrimidine biosynthesis, has a reduced virulence and produced a low germination rate in murine models of IA.74 In the same way, mutant strains lacking the pabaA gene, that encodes for p-aminobenzoic acid synthase and is involved in folate biosynthesis, showed a severe reduction of virulence.44 In the case of these two latter genes, their involvement in virulence is attributed to a supposed low concentration of pyrimidine and p-amino benzoic acid in vivo.

Signaling, metabolic regulation and response to stress conditions

The environmental conditions found by pathogenic fungi in the colonization and infection of the host are different to those found in their normal environmental niche. The signals must be detected and transmitted through mechanisms of gene regulation and metabolism, enabling the fungus to adapt to them. Several regulatory mechanisms have been studied in A. fumigatus including mitogen-activated protein kinase (MAPK) pathways, signal transduction pathways activated by G-proteins, Ras proteins, histidine kinases, calcium signaling, and a CPC system, among others (Table 6).

Table 6.

Molecules and genes involved in signaling, metabolic regulation and response to stress conditions

Molecules/genes  Gene product  Function  Pathogenesis related activities  Effect of deletiona  Uses  References 
MAP kinasebpathways
sakA/hogA  SakA/HogA (MAP kinases)Stress regulation (osmotic, C and N starvation)  Response to stress      300 
  Regulation of conidium germination         
mpkA  MpkA (MAP kinases)Regulation of cell wall integrity signaling  Response to stress       
  Regulation of pyomelanin formation    Normal virulence    283,284 
mpkB  MpkB (MAP kinases)  Mating (putative pheromone)  Response to stress      175 
mpkC  MpkC (MAP kinases)  Regulation of conidium germination  Response to stress      233 
ste7  Ste7 (MAPK kinases)  Mating        175 
pbs2  Pbs2 (MAPK kinases)  Osmotic regulation        175 
mkk2  Mkk2 (MAPK kinases)  Cell wall integrity        175 
steC/ste11  SteC/Ste11 (MAPKK kinases)  Mating        175 
bck1  Bck1 (MAPKK kinases)  Cell-wall integrity        175 
ssk2  Ssk2 (MAPKK kinases)  Osmotic regulation        175 
sho1  Sho1(adaptor protein)  HOG–MAPK pathwayc  Response to stress  Normal virulence    167 
G-proteins             
gpaA  GpaA (G protein α subunit)  Regulation vegetative growth and conidium germination        170 
sfaD  SfaD (G protein β subunit)Regulation vegetative growth and conidium germination        253 
  Regulation metabolite production (gliotoxin, etc.)         
gpgA  GpgA (G protein γ subunit)Regulation vegetative growth and conidium germination        253 
  Regulation metabolite production (gliotoxin, etc.)         
cAMP-PKA signaling
acyA  AcyA (adenylate cyclase)  cAMP signal transduction        161 
gpaB  GpaB (G protein α subunit)  cAMP signal transduction, Stimulator of adenylate cyclase    Almost avirulent    163 
pkaC1  PkaC1 (cAMP-dependent PKA catalytic subunit)  cAMP signal transduction    Almost avirulent    163 
pkaR  PkaR (PKA regulatory subunit )  cAMP signal transduction    Hypovirulent    310 
His kinases    Osmolarity stress response        222 
    Dicarboximide fungicides resistance         
    Cell-wall assembly         
fos1  Fos1 (histidine kinase)    Stress response  Hypovirulent    57,222 
tcsB  TcsB (histidine kinase)           
Other kinases (Cross-Pathways Control)79 
cpcA/gcn4p  CpcA/Gcn4p (Transcriptional activator)  Putative amino acid biosynthetic pathways regulator  N acquisition  Hypovirulent    134 
cpcC/gcn2pC  CpcC/Gcn2p (eIF2a kinase)Sensor kinase, in amino acid starvation, down-regulation of general translations  N adquisition  Normal virulence    243 
  Derepress cpcA in nutritional stress conditions  Adaptation amino acid starvation       
Ca2+ signaling
calA/cnaA  CalA/CnaA (calcineurine catalytic subunit A)  Septum formation Conidiophore development  Stress response  Hypovirulent  Adjunct therapeutic target  265,266 
crzA  CrzA (zinc finger transcription factor)  Ca2+–Mn2+-tolerance  Stress response  Hypovirulent  New antifungal target  62,258 
gprC and gprD  GprC and GprD (putative G protein-coupled receptors)  Stress signals via modulation of the calcineurin pathway  Adaptation stress signaling  Hypovirulent    99 
Ras family             
rasA  RasA (GTPase)  Hyphal growth and asexual development        88 
    Cell wall integrity         
rasB  RasB (GTPase)  Germination and growth rates    Hypovirulent    90 
rhbA  RhbA (GTPase, Ras-related protein)    N acquisition  Hypovirulent    211 
ace2  Ace2 (transcription factor)  Pigment production and conidiation    Hypervirulent    82 
medA  MedA  Adherence  Host interactions (adherence to pulmonary epithelial cells, endothelial cells and fibronectin)Hypovirulent  New antifungal target103 
    Biofilm formation     
srbA  SrbA (related with SREBPd, homolog to Sre1)Ergosterol biosynthesis  Hypoxia adaptation  Hypovirulent    299 
  Maintenance of cell polarity  Azole resistance       
a

Virulence assayed in animal model.

b

Mitogen activated protein kinase (MAP kinase). MAP kinase kinase (MAPK kinase). MAPK kinase kinase (MAPKK kinase).

c

High osmolarity glycerol (HOG) MAPK signaling pathway.

d

Sterol regulatory element-binding proteins (SRBEP).

Fungi, like other eukaryotes, can regulate their cellular physiology in response to environmental changes via MAPK pathways. These environmental changes include conditions of stress (increased osmolarity, heat shock, high concentrations of heavy metals, and reactive oxygen species), nutrient limitation, disruption of cell wall integrity, and mating pheromones.176 For a better understanding of MAPK pathways in Aspergillus see the review of May.175 The MAPK pathways consist in three protein kinases that act subsequently by phosphorylation. The genome of A. fumigatus has four MAPK described genes, sakA/hogA, mpkA, mpkB, and mpkC, three putative MAPK kinases (MAPKK) and three MAPKK kinases (MAPKKK). The three MAPKKs are Ste7 like, Pbs2 like, and Mkk2 like, suggesting their possible roles in mating, osmotic regulation, and cell wall integrity, respectively. Similarly, the MAPKKKs are SteC/Ste11, Bck1, and Ssk2, with possible relations in mating, cell-wall integrity, and osmotic regulation, respectively.175 Of all of these genes, the sakA is the most intensively studied. This gene is necessary for the osmotic stress response, it negatively regulates conidial germination in response to less-preferred nitrogen sources; and is activated upon either carbon or nitrogen starvation during vegetative growth.300 On the other hand, the mpkA regulates cell wall integrity signaling and pyomelanin formation,284 and mpkC regulates conidial germination in response to the carbon source in the medium.233 The mpkA deletion has been carried out but no influence was observed on virulence of the mutant strain in a murine infection model,283 while the other genes have not been yet tested for virulence.

The high osmolarity–glycerol (HOG) MAPK (HOG–MAPK) signaling pathway plays an important role in regulating morphology, growth, and adaptation to stress and virulence in a number of fungal pathogens. The Sho1 adaptor protein is an important element of the two upstream branches of the HOG–MAPK pathway in S. cerevisiae. However, although the deletion of this gene in A. fumigatus produces a mutant sensitive to oxidative stress, it was still as virulent as the wild-type strain in an immunosuppressed mouse infection model.167

Many signal transduction pathways are activated by heterotrimeric G-proteins whose activation is frequently coupled to cell surface receptors. In fungi, G-proteins play integral roles in germination, vegetative growth, cell cycle control, mating, cell–cell fusion, morphogenesis, chemotaxis, pathogenicity, and secondary metabolism.253 The system consists of a membrane bound G-protein coupled receptor (GPCR), heterotrimeric G-protein α, β, γ subunits, and a diverse group of effectors. The G protein α subunit, GpaA, mediates signaling for vegetative growth and negative-regulation of conidiation in A. fumigatus,170 while the β subunit, SfaD, and γ subunit, GpgA, play crucial roles in proper control of vegetative growth, spore germination, asexual development and production of certain metabolites.253 The deletion of the sfaD and gpgA genes resulted in no or very low gliotoxin detection,253 suggesting a possible role of these proteins in gliotoxin biosynthesis.

The gpaB gene encodes a G protein α subunit involved in cAMP signal transduction that was found to be an upstream stimulator of adenylate cyclase, acyA. Deletion of these genes was studied and the mutant strains showed reduced conidiation, and also a slower growth rate in the ΔacyA mutant strain.161 The same effect was observed with the deletion of pkaC1 gene, which encodes the cAMP-dependent protein kinase A (PKA) catalytic subunit.163 The ΔgpaB and ΔpkaC1 strains were almost avirulent in an animal infection model of IA.163 The regulatory subunit of PKA is encoded by the pkaR gene. A ΔpkaR mutant had reduced growth and germination rates, increased susceptibility to oxidative stress, and reduced virulence in an immunosuppressed mouse model of IA.310 However, the reduced virulence of ΔpkaC1 and ΔpkaR observed in mice could be a general outcome of impaired growth.208 Recent studies have also related the cAMP–PKA signal transduction pathway with pigment formation105 and the nuclear duplication cycle.94 In fact, the sporulation and expression of the pksP/alb1 gene, which codes for the first enzyme of melanin production, is controlled by the cAMP signal transduction pathway, which includes a G protein α subunit, acenylate cyclase, and protein kinase A.42,161 Recently, two putative G protein-coupled receptors, GprC and GprD, have been characterized.99 Deletion of the corresponding genes resulted in drastic growth defects, including reduced hyphal extension, retarded germination and elevated levels of hyphae branching. Furthermore, compared with the wild type, the sensitivity of the mutant strains towards reactive oxygen intermediates was greater, and the mutants displayed attenuated virulence in a murine infection model. These authors concluded that the receptors are involved in integrating and processing stress signals via modulation of the calcineurin pathway.

Ras proteins are monomeric GTPases which act as molecular switches that transduce signals from the outside of the cell to signaling cascades inside the cell. In A. fumigatus, three of these proteins have been studied: RasA, RasB, and RhbA. The first, RasA, appears to have a crucial role in hyphal growth and asexual development, and its function is linked to cell wall integrity,88 while deletion of the A. fumigatus rasB gene caused decreased germination and growth rates as well as a diminished virulence in a mice infection model.90 The role of rhbA gene was discussed above in the nutrient uptake section.

In fungi, two-component histidine kinases are involved in response mechanisms to extracellular changes in osmolarity, resistance to dicarboximide fungicides, and cell-wall assembly.222 The A. fumigatus genome has at least 15 putative histidine kinase genes, of which only two have been studied, fos1 and tcsB.208 The Δfos1 mutant strain did not exhibit any detectable defects in either hyphal growth or morphology when grown on solid or liquid media222 but it had significantly lower virulence than the wild-type strain.57 The ΔtcsB mutant was similar to the wild type strain with regard to growth and morphology79 but its role in virulence has not been established.

Calcium signalling through the Ca2+-binding protein, calmodulin, and the Ca2+–calmodulin-dependent phosphatase, calcineurin, has been associated with a multitude of processes, including stress response, mating, budding, and actin-based processes66 as well astolerance to antifungal drugs.65,73,131,242,266,290 Notably, this pathway is highly conserved throughout the fungal kingdom.238 Calcineurin is a heterodimeric protein formed by a catalytic subunit A, and a calcium-dependent regulatory subunit B. Steinbach et al.265 demonstrated that the calA/cnaA gene, which codes for the calcineurin subunit A, is implicated in virulence. A ΔcnaA mutant strain exhibited decreased filamentation, morphological conidial defects and attenuation of pathogenicity compared to infection with the wild-type in several different animal models. In agreement with these results, Da Silva Ferreira et al.67 showed that the calA gene is not essential in A. fumigatus, but its deletion results in severe defects in branching and conidial architecture and limited growth. A recent study has also suggested that calcineurin is involved in septum formation and conidiophore development.122 Indeed, calcineurin may be an excellent target for adjuvant in combination with other cell wall inhibitors against A. fumigatus.266 A key target of calcineurin is the zinc finger transcription factor CrzA, a homologue of the S. cerevisiae transcription factor Crz1.258 The ΔcrzA mutant of A. fumigatus resulted in a strain with significant defects in conidial germination, polarized hyphal growth, cell wall structure, and asexual development62 and produced a significantly lower mortality rate in a neutropenic murine model of invasive pulmonary aspergillosis.62,258 Fortwendel et al.89 have obtained data suggesting that the Ras and calcineurin pathways act in parallel to regulate cell wall formation and hyphal growth, and additionally, that the calcineurin pathway elements cnaA and crzA play a major role in proper chitin and glucan incorporation into the A. fumigatus cell wall. Soriani et al.258 also demonstrated a role of crzA in the mediation of cellular tolerance to increased concentrations of calcium and manganese. Thus, crzA is an attractive fungus-specific antifungal target for the treatment of IA.62

A conserved signal transduction cascade linking environmental stress to amino acid homeostasis is the CPC system that acts via phosphorylation of the translation initiation factor eIF2 by a sensor kinase. As noted before, the cpcA gene encodes the transcriptional activator of the CPC-system of amino acid biosynthesis and ΔcpcA strains displayed attenuated virulence in a murine model of IA.134 On the other hand, the cpcC gene encodes the CPC eIF2a kinase. The ΔcpcC deletion mutant showed increased sensitivity towards amino acid starvation but it was not impaired in virulence in a murine model of pulmonary aspergillosis.243

The transcription factor Ace2 influences virulence in other fungi. A. fumigatus contains an ortholog of this gene, ace2, which governs pigment production, conidiation, and virulence82. Mice immunosuppressed with cortisone acetate and infected with the Δace2 mutant showed accelerated mortality, greater pulmonary fungal burden, and increased pulmonary inflammatory responses than mice infected with wild type strain. This hypervirulence of the Δace2 strain was related to reduced expression of ppoC, ecm33, and ags3 detected in this mutant. It is known that A. fumigatus mutants with null or reduced expression of these genes have increased virulence in mice.

MedA is a development regulated protein that governs adherence, host interactions, and virulence in A. fumigatus.103 These authors studied a ΔmedA strain and demonstrated a dramatic reduced conidiation, and impaired biofilm production and adherence to plastic, as well as adherence to pulmonary epithelial cells, endothelial cells, and fibronectin in vitro. This mutant also exhibited reduced virulence in both invertebrate and mammalian models of IA. These results suggest that MedA downstream targets mediate virulence and might provide novel therapeutic targets for IA.

The presence of A. fumigatus causes significant inflammation in the sites of infection. It is known that levels of oxygen are significantly lower at sites of inflammation.298 Accordingly, during infection, A. fumigatus may be exposed to rapid changes in oxygen concentration, even reaching extremely low levels, depending upon the tissue infected and current immune response. The mechanisms of hypoxic adaptation of the aerobic A. fumigatus are currently unknown. Willger et al.298,299 have hypothesized that a putative Sre1 homolog in A. fumigatus (SrbA), related to the sterol regulatory element-binding proteins (SREBPs), could also act as an indirect sensor of oxygen levels and could regulate the transcription of genes required for adaptation to hypoxic environments. These authors have demonstrated that the srbA gene plays a critical role in ergosterol biosynthesis, azole resistance, and the maintenance of cell polarity in A. fumigatus.299 The ΔsrbA strain was almost avirulent in mouse models of IA, and loss of this gene, affects the expression of 87 genes related sterol biosynthesis and hyphal morphology, as demonstrated by expression analysis using DNA microarrays.299 Hypoxia adaptation is likely an important virulence attribute of pathogenic molds.

Allergens

A. fumigatus produces a significant number of allergenic molecules which show reactions with IgE in asthmatic patients and patients with allergic bronchopulmonary aspergillosis (ABPA). Data concerning all known A. fumigatus allergens are collected by “Allergome, a platform for allergen knowledge”1 and the Allergen nomenclature website,2 and are summarized in Table 7 of this review. Only 23 molecules currently hold an official name of allergen, and have names in the range Asp f 1–Asp f 34. One of these, Asp f 15, has been proposed to be removed from the list due to it having been demonstrated that it is identical to Asp f 13, and the Asp f 6 allergen has shown a high degree of homology with Asp f 9.64 On the other hand, there are three candidates to be considered as allergens, Asp f 56kDa (a protease), Asp f AfCalAp, and Asp f GST (related to glutathione-S-transferase). However, the sequence of Asp f 56kDa is not predicted to be encoded in any of the sequenced Aspergillus genomes.64 Some of these allergens have known structural, toxic or enzymatic functions, and their relationship with virulence has been discussed in previous sections of this review. However, other allergenic components do not have virulence activities except as allergens. All Aspergillus allergens reacted with IgE in asthmatic patients and with ABPA.129 Some A. fumigatus allergens showed cross-reactivity with various conserved proteins including some human proteins. Among these, Asp f 6 (Mn-Sod), Asp f 8 (P2 acidic ribosomal protein), Asp f 11 and Asp f 27 (cyclophilins), and Asp f 28 and Asp f 29 (thioredoxins) have been shown to belong to families of cross-reactive pan-allergens.64 This fact could imply autoimmunity problems in human patients.

Table 7.

Allergens of A. fumigatus related with activation of Type I hypersensitivity

Genes  Gene product  Function  Pathogenesis related activities  Effect of deletiona  Uses  References 
asp f 1/mitF/res  Restrictocin, mitogillin  Ribotoxin  Protein biosynthesis inhibition  Normal virulence  Diagnosis  12,224,257 
      Cytotoxin       
      Apoptosis       
      Type I hypersensitivity       
asp f 2  Asp f 2  Fibrinogen binding protein  Adhesion    Diagnosis  9,18 
    Zn metabolisms?  Type I hypersensitivity       
asp f 3  Asp f 3, peroxisomal protein (PMP, redoxin)  Peroxisomal membrane Protein  Type I hypersensitivity    Diagnosis  109 
asp f 4      Type I hypersensitivity    Diagnosis  64,140 
          Confirm ABPA   
mep/asp f 5  Mep/Asp f 5 (extracellular metalloprotease glycosylated)Protein degradation (collagen and elastin)Tissue destruction/invasion      63,121,172,255 
  Type I hypersensitivity       
sod3/asp f 6  Sod3/Asp f 6 (cytoplasmic Mn superoxide dismutase, Cross-reactive pan-allergen)O2 degradation  ROS protection    Diagnosis  63,64,147 
    Type I hypersensitivity    Confirm ABPAb   
    Autoimmunity       
    Antigen       
           
asp f 7  Asp f 7    Type I hypersensitivity    Diagnosis  63 
asp f 8  Asp f 8, 60S acidic ribosomal protein P2 (cross-reactive pan-allergen)Protein synthesis (elongation step)Type I hypersensitivity    Diagnosis  141 
  Autoimmunity       
asp f 9/crf1  Asp f 9/Crf1 (cell wall glucanase)  Cell wall assembly  Type I hypersensitivity    Diagnosis  63 
pep/asp f 10  Pep/Asp f 10, aspergillopepsine F (aspatic endopeptidase)Protein degradation (collagen)Tissue destruction/invasion    Diagnosis  63,157,231 
  Type I hypersensitivity       
asp f 11  Asp f 11 (peptidyl-prolyl cis-trans isomerase, cyclophilin, cross-reactive pan-allergen)Peptide synthesis  Type I hypersensitivity    Diagnosis  87 
  Chaperone and cell signaling function  Autoimmunity       
hsp1/asp f 12  Hsp1/Asp f 2 (heat shock protein, Hsp90 family)Chaperone  Chaperone activity and protein transport in growth at 37°C      136 
    Stress response during inflammation       
    Autoimmunity       
    Type I hypersensitivity       
alp/asp f 13  Alp/Asp f 13, orycine (alkaline serine protease, Elastase)Protein degradation (elastin, collagen, fibrinogen and casein)Tissue destruction/invasion  Hypovirulent    121,130,228,271 
  Type I hypersensitivity       
asp f15  Asp f 15, (serine protease, homolog Asp f 13)  Tissue destruction/invasion    Diagnosis  64 
    Type I hypersensitivity       
asp f 16  Asp f 16  Putative glycosylhidrolase  Type I hypersensitivity      20 
mp1/asp f 17  Mp1/Asp f 17 (relation with Afmp1?)Cell wall galactomannoproteinAdhesion    Diagnosis  64 
  Type I hypersensitivity       
asp f 18  Asp f 18 (vacuolar serine protease, related with Alp2?)Protein degradation  Tissue destruction/invasion  Normal virulence    249,251 
    Type I hypersensitivity       
asp f 22  Asp f 22, Enolase  General metabolism  Type I hypersensitivity      144 
asp f 23  Asp f 23, 60S ribosomal protein L3  Protein synthesis  Type I hypersensitivity       
asp f 27  Asp f 27 (peptidyl-prolyl cis-trans isomerase, cyclophilin, cross-reactive pan-allergen)Peptide synthesis  Type I hypersensitivity    Diagnosis  101 
  Chaperone and cell signaling function  Autoimmunity       
asp f 28  Asp f 28 (thioredoxin, cross-reactive pan-allergen)Protein disulfide oxidoreductasesType I hypersensitivity    Diagnosis   
  Autoimmunity       
asp f 29  Asp f 29 (thioredoxin, cross-reactive pan-allergen)Protein disulfide oxidoreductasesType I hypersensitivity    Diagnosis   
  Autoimmunity       
asp f 34  Asp f 34, PhiA  Cell wall protein  Type I hypersensitivity    Diagnosis  100 
  Asp f 56kD  Protease  Type I hypersensitivity      202 
  Asp f AfCalAp    Type I hypersensitivity      282 
  Asp f GST (glutathione S-transferases)  Detoxification with glutathione  Type I hypersensitivity       
a

Virulence assayed in animal model.

b

Allergic bronchopulmonary aspergillosis.

Allergenic behaviour of the aforementioned molecules, due to their presence on conidia, their release by the destruction of the conidia by pulmonary phagocytes, or their production during the growth of fungus is unclear in IA. We were able to identify two different situations, namely, the infections caused by Aspergillus in immunocompetent or immunocompromised patients. In immunocompetent patients Aspergillus can produce several hypersensitivity diseases due to these allergens, such as ABPA, allergic rhinosinusitis, asthma, and aspergilloma. Inhalation of fungal spores, often considered the traditional route of exposure, has been associated with the induction or exacerbation of these respiratory diseases. Large numbers of inhaled fungal spores are removed from the lungs prior to germination,139 but a few conidia could escape phagocytosis and may begin to germinate. Dormant or nonviable A. fumigatus conidia uptake is associated with IFN-γ production and Th1 responses, while hyphae or swollen (germinating) conidia induce IL-4 production and eosinophil recruitment, a hallmark of allergic inflammation and Th2 responses.39,273 Therefore, successful germination is likely to contribute to the development of fungal allergy. Specific structures, factors secreted by fungi or released by killed conidia, can play an important role in allergic sensitization, but the environmental and patient-specific factors (such as the personal history of previous contact in early life immune development) are also critical to acquire tolerance or allergic sensitization in immunocompetent individuals. All Aspergillus allergens appear to activate a Type I hypersensitivity response in sensitized patients with production of high affinity IgG and IgE antibodies.232 In immunocompromised patients with debilitated innate immune responses, these allergenic compounds can increase the risk associated with aspergillosis because they may redirect the immune response to the fungus by the activation of Th2 lymphocytes, a response that does not seem to be efficient in eliminating this fungus.108 Some of these allergens have been studied for their usefulness for diagnosis (see Allergome1 and Table 7).

Gene expression assays

During the last years only a few studies have investigated A. fumigatus gene expression during infection. Zhang et al.308 analysed the expression of certain virulence factors in vivo and in vitro concluding that in vitro measurements of transcription compared to transcription in infected lung tissue demonstrated low levels of fos-1 and rhbA genes, and 20–40-fold increases in cpcA, lysF, and pabA genes, while the pksP gene was only detected in vivo. Gravelat et al.102 performed real-time reverse transcription-PCR analysis on lung samples from mice with invasive pulmonary aspergillosis to determine the expression of A. fumigatus genes that are expressed at specific stages of development. This study revealed that in established infections, A. fumigatus exhibited mRNA expression of specific genes to develop competent hyphae, such as stuA. The acquisition of competence is referred to the shift of hyphae from a state in which they cannot undergo asexual reproduction to one in which they can. In contrast, mRNA of genes expressed specifically by conidia and precompetent hyphae was not detected. Many genes required for mycotoxin synthesis, including aspHS, gliP, mitF, and metAP, were expressed at significantly higher levels during invasive infection than in vitro. On the other hand, the expression of gliP mRNA in vitro was found to be highly dependent on culture conditions. Furthermore, this expression was found to be dependent on the transcription factor StuA both in vitro and in vivo. These results highlight the importance of the evaluation of putative virulence factors expressed by competent hyphae and the analysis of gene expression levels during invasive infection rather than in vitro alone.

Gene expression assays have also been developed to analyse the function of various proteins, comparing the gene expression profiles of the mutant against those of the reference strain. For example, Soriani et al.259 searched the metabolic pathways influenced by A. fumigatus transcription factor AfCrzA after a short pulse of calcium, by determining the transcriptional profile of A. fumigatus wild type in comparison to ΔafcrzA mutant strains. Similarly, Twumasi-Boateng et al.281 described, on the basis of transcriptional profile studies, the role for BrlA in the response to nitrogen depletion and for StuA in the regulation of secondary metabolite clusters in A. fumigatus. Using transcriptomic analysis, other authors have investigated the exit from dormancy of A. fumigatus conidia146 and the genes differentially expressed in conidia and hyphae of this fungus upon exposure to human neutrophils.267 Gene expression assays with DNA microarrays are also being used to study the adaptation of A. fumigatus to different stress conditions such as hypoxia,299 heat shock,201 and antifungals activities such as voriconazole68. Finally, DNA microarray-based studies have also been used for the detection and identification of fungal pathogens, including A. fumigatus.49,260

Conclusions

A. fumigatus is an opportunistic pathogen whose ability to produce disease is inextricably linked to the host immune response. The most recent progress in research has revealed how components of the immune system are able to eliminate the fungus and that the weakness of immune system has a role in the development of aspergillosis. Likewise, some of the mechanisms that the fungus uses to evade immune responses, to obtain nutrients and to cause damage to the host and thus generate an IA, have been identified. If we consider only the classical definitions of virulence factors, i.e., a component of a pathogen that allows it to cause disease, we would probably have difficulties in deciding what is or is not a virulence factor in human fungal pathogenesis. In fact, that would exclude, for example, normal or adaptive mechanisms of the fungi to grow in different environmental niches, which are extensively used during the colonization of a human host. In A. fumigatus a great variability of “non-classical” virulence factors have been described, associated with its structure, its capacity to grow and adapt to stress conditions, its mechanisms for evading the immune system and its ability to cause host damage. As detailed in this review, a large number of genes and molecules have been identified and investigated in some depth as potential virulence factors. However, none of them have proven to be sufficiently important to fully explain the virulence of A. fumigatus. In most cases, the experiments based on the loss of gene/function by mutation have shown only small declines in virulence, unless the genes involved regulation of multiple activities of fungal adaptation and growth were eliminated. The pleiotropic effect of certain genes, the function of various genes associated with the virulence in the normal growth of A. fumigatus, and the redundancy due to the existance of several genes with the same activity, complicate the process of studying virulence factors of A. fumigatus with mutant strains. On the other hand, virulence studies use animal models with high levels of immunosuppression, which can also lead to failure to detect the effect on the virulence of the mutant strains. Likewise, the animal immunosuppression used, focusing mainly on causing neutropenia, only simulates the situation in neutropenic patients without providing any data for the other types of patients with aspergillosis. From all this data, the idea has emerged that the pathogenesis of diseases caused by this fungus in immunocompromised patients is very complex. As shown in Fig. 3, we could imagine a complex puzzle, the pieces of which would be virulence factors or the different activities of the fungus, and our task would then be to complete this puzzle to obtain a comprehensive vision of the virulence of A. fumigatus. We begin to understand the intricacies of its metabolism but much remains to be learned concerning the activity of this fungus in vivo. Furthermore, understanding changes in the host microenvironment, including hypoxia, pH, available nutrients, and immune responses, and how these signals are processed by the fungus, could be useful to determine the efficacy and effectiveness of particular antimicrobial strategies. The data so far have helped to improve diagnosis and identify new targets for antifungal development, which in combination with currently available therapies can improve the prognosis for IA patients. Expression studies using DNA microarrays of A. fumigatus during invasion or interaction with immune responses may help to provide a more rapid and profound understanding of the virulence capabilities of this fungus, as well as their adaptation mechanisms based on networks of complex metabolic and genetic regulation systems, in order to find new possible targets for detection and treatment of the disease. In particular, these expression studies using DNA microarrays are being applied to different stress conditions such as heat shock and antifungal activity.

Fig. 3.

Summary of genes and molecules associated with the virulence of Aspergillus fumigatus contained in this review.

(1,24MB).
Acknowledgements

This work was supported by General Grant to Research Groups (GIU08/20) from the UPV/EHU, and SAIOTEK Program Grant (S-PC09UN04) and Consolidated Research Group Grant (IT343-10) from the Basque Government. Jimena Victoria Fernandez Molina was supported with a “Beca de Investigación Predoctoral” from the UPV/EHU.

References
[1]
Allergome, a platform for allergen knowledge [page on Internet]. Latina, Italy: Allergy Data Laboratories S.C. [updated March 24, 2010; referenced May 12, 2010] Available at: ¿http://www.allergome.org¿.
[2]
Allergen nomenclature website [page on Internet]. The International Union of Immunological Societies (I.U.I.S.) Allergen Nomenclature Sub-committee, [updated May 5, 2010; referenced May 12, 2010]. Available at: ¿http://www.allergen.org¿.
[3]
N. Al-Bader, G. Vanier, H. Liu, F.N. Gravelat, M. Urb, C.M. Hoareau, et al.
The role of trehalose biosynthesis in Aspergillus fumigatus development, stress response and virulence.
Infect Immun, 78 (2010), pp. 3007-3018
[4]
D. Albrecht, R. Guthke, A.A. Brakhage, O. Kniemeyer.
Integrative analysis of the heat shock response in Aspergillus fumigatus.
BMC Genom, 11 (2010), pp. 32
[5]
S. Alp, S. Arikan.
Investigation of extracellular elastase, acid proteinase and phospholipase activities as putative virulence factors in clinical isolates of Aspergillus species.
J Basic Microbiol, 48 (2008), pp. 331-337
[6]
S. Álvarez-Pérez, J.L. Blanco, P. Alba, M.E. García.
Sexuality and pathogenicity in Aspergillus fumigatus: is there any relationship?.
Rev Iberoam Micol, 27 (2010), pp. 1-5
[7]
S. Álvarez-Pérez, J.L. Blanco, P. Alba, M.E. García.
Mating type and invasiveness are significantly associated in Aspergillus fumigatus.
Med Mycol, 48 (2009), pp. 273-277
[8]
J. Amich, F. Leal, J.A. Calera.
Repression of the acid ZrfA/ZrfB zinc-uptake system of Aspergillus fumigatus mediated by PacC under neutral, zinc-limiting conditions.
Int Microbiol, 12 (2009), pp. 39-47
[9]
J. Amich, R. Vicentefranqueira, F. Leal, J.A. Calera.
Aspergillus fumigatus survival in alkaline and extreme zinc-limiting environments relies on the induction of a zinc homeostasis system encoded by the zrfC and aspf2 genes.
Eukaryot Cell, 9 (2010), pp. 424-437
[10]
R. Amitani, G. Taylor, E.N. Elezis, C. Llewellyn-Jones, J. Mitchell, F. Kuze, et al.
Purification and characterization of factors produced by Aspergillus fumigatus which affect human ciliated respiratory epithelium.
Infect Immun, 63 (1995), pp. 3266-3271
[11]
R. Araujo, A. Amorim, L. Gusmao.
Genetic diversity of Aspergillus fumigatus in indoor hospital environments.
Med Mycol, 48 (2010), pp. 832-838
[12]
L.K. Arruda, T.A. Platts-Mills, J.W. Fox, M.D. Chapman.
Aspergillus fumigatus allergen I, a major IgE-binding protein, is a member of the mitogillin family of cytotoxins.
J Exp Med, 172 (1990), pp. 1529-1532
[13]
D.S. Askew.
Aspergillus fumigatus: virulence genes in a street-smart mold.
Curr Opin Microbiol, 11 (2008), pp. 331-337
[14]
A. Aufauvre-Brown, E. Mellado, N.A.R. Gow, D.W. Holden.
Aspergillus fumigatus chsE: a gene related to CHS3 of Saccharomyces cerevisiae and important for hyphal growth and conidiophore development but not pathogenicity.
Fungal Genet Biol, 21 (1997), pp. 141-152
[15]
S.E. Baker.
Aspergillus genomics and DHN-melanin conidial pigmentation.
Aspergillus in the genomic era, pp. 73-85
[16]
C.J. Balibar, C.T. Walsh.
GliP, a multimodular nonribosomal peptide synthetase in Aspergillus fumigatus, makes the diketopiperazine scaffold of gliotoxin.
Biochemistry, 45 (2006), pp. 15029-15038
[17]
V. Balloy, M. Chignard.
The innate immune response to Aspergillus fumigatus.
Microbes Infect, 11 (2009), pp. 919-927
[18]
B. Banerjee, P.A. Greenberger, J.N. Fink, V.P. Kurup.
Immunological characterization of Asp f 2, a major allergen from Aspergillus fumigatus associated with allergic bronchopulmonary aspergillosis.
Infect Immun, 66 (1998), pp. 5175-5182
[19]
B. Banerjee, V.P. Kurup.
Molecular biology of Aspergillus allergens.
Front Biosci, 8 (2003), pp. S128-S139
[20]
B. Banerjee, V.P. Kurup, P.A. Greenberger, B.D. Johnson, J.N. Fink.
Cloning and expression of Aspergillus fumigatus allergen Asp f 16 mediating both humoral and cell-mediated immunity in allergic bronchopulmonary aspergillosis (ABPA).
Clin Exp Allergy, 31 (2001), pp. 761-770
[21]
A. Beauvais, J.P. Latge.
Membrane and cell wall targets in Aspergillus fumigatus.
Drug Resist Updat, 4 (2001), pp. 38-49
[22]
A. Beauvais, D. Maubon, S. Park, W. Morelle, M. Tanguy, M. Huerre, et al.
Two alpha (1–3) glucan synthases with different functions in Aspergillus fumigatus.
Appl Environ Microbiol, 71 (2005), pp. 1531-1538
[23]
A. Beauvais, M. Monod, J.P. Debeaupuis, M. Diaquin, H. Kobayashi, J.P. Latge.
Biochemical and antigenic characterization of a new dipeptidyl-peptidase isolated from Aspergillus fumigatus.
J Biol Chem, 272 (1997), pp. 6238-6244
[24]
A. Beauvais, M. Monod, J. Wyniger, J.P. Debeaupuis, E. Grouzmann, N. Brakch, et al.
Dipeptidyl-peptidase IV secreted by Aspergillus fumigatus, a fungus pathogenic to humans.
Infect Immun, 65 (1997), pp. 3042-3047
[25]
A. Beauvais, C. Schmidt, S. Guadagnini, P. Roux, E. Perret, C. Henry, et al.
An extracellular matrix glues together the aerial-grown hyphae of Aspergillus fumigatus.
Cell Microbiol, 9 (2007), pp. 1588-1600
[26]
T. Beffa, F. Staib, J. Lott Fischer, P.F. Lyon, P. Gumowski, O.E. Marfenina, et al.
Mycological control and surveillance of biological waste and compost.
Med Mycol, 36 (1998), pp. 137-145
[27]
J. Behnsen, A. Hartmann, J. Schmaler, A. Gehrke, A.A. Brakhage, P.F. Zipfel.
The opportunistic human pathogenic fungus Aspergillus fumigatus evades the host complement system.
Infect Immun, 76 (2008), pp. 820-827
[28]
S. Bellocchio, C. Montagnoli, S. Bozza, R. Gaziano, G. Rossi, S.S. Mambula, et al.
The contribution of the Toll-like/IL-1 receptor superfamily to innate and adaptive immunity to fungal pathogens in vivo.
J Immunol, 172 (2004), pp. 3059-3069
[29]
R. Ben-Ami, D.P. Kontoyiannis.
Pathogenesis of invasive pulmonary aspergillosis.
Aspergillosis: from diagnosis to prevention, pp. 345-379
[30]
S. Bertout, C. Badoc, M. Mallie, J. Giaimis, J.M. Bastide.
Spore diffusate isolated from some strains of Aspergillus fumigatus inhibits phagocytosis by murine alveolar macrophages.
FEMS Immunol Med Microbiol, 33 (2002), pp. 101-106
[31]
R. Bhabhra, D.S. Askew.
Thermotolerance and virulence of Aspergillus fumigatus: role of the fungal nucleolus.
Med Mycol, 43 (2005), pp. S87-S93
[32]
R. Bhabhra, M.D. Miley, E. Mylonakis, D. Boettner, J. Fortwendel, J.C. Panepinto, et al.
Disruption of the Aspergillus fumigatus gene encoding nucleolar protein CgrA impairs thermotolerant growth and reduces virulence.
Infect Immun, 72 (2004), pp. 4731-4740
[33]
M. Birch, D.W. Denning, G.D. Robson.
Comparison of extracellular phospholipase activities in clinical and environmental Aspergillus fumigatus isolates.
Med Mycol, 42 (2004), pp. 81-86
[34]
M. Birch, G. Robson, D. Law, D.W. Denning.
Evidence of multiple extracellular phospholipase activities of Aspergillus fumigatus.
Infect Immun, 64 (1996), pp. 751-755
[35]
J.L. Blanco, R. Hontecillas, E. Bouza, I. Blanco, T. Peláez, P. Muñoz, et al.
Correlation between the elastase activity index and invasiveness of clinical isolates of Aspergillus fumigatus.
J Clin Microbiol, 40 (2002), pp. 1811-1813
[36]
J.W. Bok, S.A. Balajee, K.A. Marr, D. Andes, K.F. Nielsen, J.C. Frisvad, et al.
LaeA, a regulator of morphogenetic fungal virulence factors.
Eukaryot Cell, 4 (2005), pp. 1574-1582
[37]
J.W. Bok, D. Chung, S.A. Balajee, K.A. Marr, D. Andes, K.F. Nielsen, et al.
GliZ, a transcriptional regulator of gliotoxin biosynthesis, contributes to Aspergillus fumigatus virulence.
Infect Immun, 74 (2006), pp. 6761-6768
[38]
J.W. Bok, N.P. Keller.
LaeA, a regulator of secondary metabolism in Aspergillus spp.
Eukaryot Cell, 3 (2004), pp. 527-535
[39]
S. Bozza, R. Gaziano, A. Spreca, A. Bacci, C. Montagnoli, P. di Francesco, et al.
Dendritic cells transport conidia and hyphae of Aspergillus fumigatus from the airways to the draining lymph nodes and initiate disparate Th responses to the fungus.
J Immunol, 168 (2002), pp. 1362-1371
[40]
A.A. Brakhage, K. Langfelder.
Menacing mold: the molecular biology of Aspergillus fumigatus.
Annu Rev Microbiol, 56 (2002), pp. 433-455
[41]
A.A. Brakhage, K. Langfelder, G. Wanner, A. Schmidt, B. Jahn.
Pigment biosynthesis and virulence.
Contrib Microbiol, 2 (1999), pp. 205-215
[42]
A.A. Brakhage, B. Liebmann.
Aspergillus fumigatus conidial pigment and cAMP signal transduction: significance for virulence.
Med Mycol, 43 (2005), pp. S75-S82
[43]
G.D. Brown, P.R. Taylor, D.M. Reid, J.A. Willment, D.L. Williams, L. Martinez-Pomares, et al.
Dectin-1 is a major beta-glucan receptor on macrophages.
J Exp Med, 196 (2002), pp. 407-412
[44]
J.S. Brown, A. Aufauvre-Brown, J. Brown, J.M. Jennings, H. Arst Jr, D.W. Holden.
Signature-tagged and directed mutagenesis identify PABA synthetase as essential for Aspergillus fumigatus pathogenicity.
Mol Microbiol, 36 (2000), pp. 1371-1380
[45]
J. Bunger, G. Westphal, A. Monnich, B. Hinnendahl, E. Hallier, M. Muller.
Cytotoxicity of occupationally and environmentally relevant mycotoxins.
Toxicology, 202 (2004), pp. 199-211
[46]
J.P. Burnie, T.L. Carter, S.J. Hodgetts, R.C. Matthews.
Fungal heat-shock proteins in human disease.
FEMS Microbiol Rev, 30 (2006), pp. 53-88
[47]
C. Burns, R. Geraghty, C. Neville, A. Murphy, K. Kavanagh, S. Doyle.
Identification, cloning, and functional expression of three glutathione transferase genes from Aspergillus fumigatus.
Fungal Genet Biol, 42 (2005), pp. 319-327
[48]
J.A. Calera, S. Paris, M. Monod, A.J. Hamilton, J.P. Debeaupuis, M. Diaquin, et al.
Cloning and disruption of the antigenic catalase gene of Aspergillus fumigatus.
Infect Immun, 65 (1997), pp. 4718-4724
[49]
D. Campa, A. Tavanti, F. Gemignani, C.S. Mogavero, I. Bellini, F. Bottari, et al.
DNA microarray based on arrayed-primer extension technique for identification of pathogenic fungi responsible for invasive and superficial mycoses.
J Clin Microbiol, 46 (2008), pp. 909-915
[50]
A. Casadevall.
Fungal virulence, vertebrate endothermy, and dinosaur extinction: is there a connection?.
Fungal Genet Biol, 42 (2005), pp. 98-106
[51]
A. Casadevall, L.A. Pirofski.
The damage–response framework of microbial pathogenesis.
Nat Rev Microbiol, 1 (2003), pp. 17-24
[52]
S. Chabane, J. Sarfati, O. Ibrahim-Granet, C. Du, C. Schmidt, I. Mouyna, et al.
Glycosylphosphatidylinositol-anchored Ecm33p influences conidial cell wall biosynthesis in Aspergillus fumigatus.
Appl Environ Microbiol, 72 (2006), pp. 3259-3267
[53]
L.Y. Chai, M.G. Netea, A.G. Vonk, B.J. Kullberg.
Fungal strategies for overcoming host innate immune response.
Med Mycol, 47 (2009), pp. 227-236
[54]
Y.C. Chang, H.F. Tsai, M. Karos, K.J. Kwon-Chung.
THTA, a thermotolerance gene of Aspergillus fumigatus.
Fungal Genet Biol, 41 (2004), pp. 888-896
[55]
M. Chignard, V. Balloy, J.M. Sallenave, M. Si-Tahar.
Role of Toll-like receptors in lung innate defense against invasive aspergillosis. Distinct impact in immunocompetent and immunocompromized hosts.
Clin Immunol, 124 (2007), pp. 238-243
[56]
K.T. Chong, P.C. Woo, S.K. Lau, Y. Huang, K.Y. Yuen.
AFMP2 encodes a novel immunogenic protein of the antigenic mannoprotein superfamily in Aspergillus fumigatus.
J Clin Microbiol, 42 (2004), pp. 2287-2291
[57]
K.V. Clemons, T.K. Miller, C.P. Selitrennikoff, D.A. Stevens.
Fos-1, a putative histidine kinase as a virulence factor for systemic aspergillosis.
Med Mycol, 40 (2002), pp. 259-262
[58]
C.M. Coyle, J.Z. Cheng, S.E. O’Connor, D.G. Panaccione.
An old yellow enzyme gene that controls the branch point between Aspergillus fumigatus and Claviceps purpurea ergot alkaloid pathways.
Appl Environ Microbiol, 76 (2010), pp. 3898-3903
[59]
C.M. Coyle, S.C. Kenaley, W.R. Rittenour, D.G. Panaccione.
Association of ergot alkaloids with conidiation in Aspergillus fumigatus.
Mycologia, 99 (2007), pp. 804-811
[60]
C.M. Coyle, D.G. Panaccione.
An ergot alkaloid biosynthesis gene and clustered hypothetical genes from Aspergillus fumigatus.
Appl Environ Microbiol, 71 (2005), pp. 3112-3118
[61]
R.A. Cramer Jr, M.P. Gamcsik, R.M. Brooking, L.K. Najvar, W.R. Kirkpatrick, T.F. Patterson, et al.
Disruption of a nonribosomal peptide synthetase in Aspergillus fumigatus eliminates gliotoxin production.
Eukaryot Cell, 5 (2006), pp. 972-980
[62]
R.A. Cramer Jr, B.Z. Perfect, N. Pinchai, S. Park, D.S. Perlin, Y.G. Asfaw, et al.
Calcineurin target CrzA regulates conidial germination, hyphal growth, and pathogenesis of Aspergillus fumigatus.
Eukaryot Cell, 7 (2008), pp. 1085-1097
[63]
R. Crameri, A.G. Glaser, M. Vilhelmsson, S. Zeller, C. Rhyner.
Overview of Aspergillus allergens.
Aspergillosis: from diagnosis to prevention, pp. 655-669
[64]
R. Crameri.
Recombinant Aspergillus fumigatus allergens: from the nucleotide sequences to clinical applications.
Int Arch Allergy Immunol, 115 (1998), pp. 99-114
[65]
M.C. Cruz, A.L. Goldstein, J.R. Blankenship, M. Del Poeta, D. Davis, M.E. Cardenas, et al.
Calcineurin is essential for survival during membrane stress in Candida albicans.
EMBO J, 21 (2002), pp. 546-559
[66]
M.S. Cyert.
Genetic analysis of calmodulin and its targets in Saccharomyces cerevisiae.
Annu Rev Genet, 35 (2001), pp. 647-672
[67]
M.E. Da Silva Ferreira, T. Heinekamp, A. Hartl, A.A. Brakhage, C.P. Semighini, S.D. Harris, et al.
Functional characterization of the Aspergillus fumigatus calcineurin.
Fungal Genet Biol, 44 (2007), pp. 219-230
[68]
M.E. Da Silva Ferreira, I. Malavazi, M. Savoldi, A.A. Brakhage, M.H. Goldman, H.S. Kim, et al.
Transcriptome analysis of Aspergillus fumigatus exposed to voriconazole.
Curr Genet, 50 (2006), pp. 32-44
[69]
T.R. Dagenais, N.P. Keller.
Pathogenesis of Aspergillus fumigatus in invasive aspergillosis.
Clin Microbiol Rev, 22 (2009), pp. 447-465
[70]
A. Del Palacio, A. Alhambra, M.S. Cuétara, J. Ponton.
Early diagnosis of invasive fungal infections caused by Aspergillus and other emerging mycelial fungi.
Rev Iberoam Micol, 24 (2007), pp. 187-197
[71]
A. Del Palacio, M.S. Cuetara, A. Alhambra.
Detección de antígenos (ELISA: Platelia Aspergillus).
Aspergilosis invasora, pp. 87-101
[72]
A. Del Palacio, M.S. Cuetara, J. Ponton.
Invasive aspergillosis.
Rev Iberoam Micol, 20 (2003), pp. 77-78
[73]
M. Del Poeta, M.C. Cruz, M.E. Cárdenas, J.R. Perfect, J. Heitman.
Synergistic antifungal activities of bafilomycin A(1), fluconazole, and the pneumocandin MK-0991/caspofungin acetate (L-743,873) with calcineurin inhibitors FK506 and L-685,818 against Cryptococcus neoformans.
Antimicrob Agents Chemother, 44 (2000), pp. 739-746
[74]
C. D’Enfert, M. Diaquin, A. Delit, N. Wuscher, J.P. Debeaupuis, M. Huerre, et al.
Attenuated virulence of uridine-uracil auxotrophs of Aspergillus fumigatus.
Infect Immun, 64 (1996), pp. 4401-4405
[75]
K. Dichtl, F. Ebel, F. Dirr, F.H. Routier, J. Heesemann, J. Wagener.
Farnesol misplaces tip-localized Rho proteins and inhibits cell wall integrity signaling in Aspergillus fumigatus.
Mol Microbiol, 76 (2010), pp. 1191-1204
[76]
J.H. Do, R. Yamaguchi, S. Miyano.
Exploring temporal transcription regulation structure of Aspergillus fumigatus in heat shock by state space model.
BMC Genom, 10 (2009), pp. 306
[77]
V.M. Dos Santos, J.W. Dorner, F. Carreira.
Isolation and toxigenicity of Aspergillus fumigatus from moldy silage.
Mycopathologia, 156 (2003), pp. 133-138
[78]
C.M. Douglas.
Fungal beta(1,3)-D-glucan synthesis.
Med Mycol, 39 (2001), pp. 55-66
[79]
C. Du, J. Sarfati, J.P. Latge, R. Calderone.
The role of the sakA (Hog1) and tcsB (sln1) genes in the oxidant adaptation of Aspergillus fumigatus.
Med Mycol, 44 (2006), pp. 211-218
[80]
E. Duarte-Escalante, G. Zuniga, O.N. Ramírez, S. Córdoba, N. Refojo, R. Arenas, et al.
Population structure and diversity of the pathogenic fungus Aspergillus fumigatus isolated from different sources and geographic origins.
Mem Inst Oswaldo Cruz, 104 (2009), pp. 427-433
[81]
R.D. Eichner, M. Al Salami, P.R. Wood, A. Mullbacher.
The effect of gliotoxin upon macrophage function.
Int J Immunopharmacol, 8 (1986), pp. 789-797
[82]
D.E. Ejzykowicz, M.M. Cunha, S. Rozental, N.V. Solis, F.N. Gravelat, D.C. Sheppard, et al.
The Aspergillus fumigatus transcription factor Ace2 governs pigment production, conidiation and virulence.
Mol Microbiol, 72 (2009), pp. 155-169
[83]
W. Fang, W. Ding, B. Wang, H. Zhou, H. Ouyang, J. Ming, et al.
Reduced expression of the O-mannosyltransferase 2 (AfPmt2) leads to deficient cell wall and abnormal polarity in Aspergillus fumigatus.
Glycobiology, 20 (2010), pp. 542-552
[84]
W. Fang, X. Yu, B. Wang, H. Zhou, H. Ouyang, J. Ming, et al.
Characterization of the Aspergillus fumigatus phosphomannose isomerase Pmi1 and its impact on cell wall synthesis and morphogenesis.
Microbiology, 155 (2009), pp. 3281-3293
[85]
N.D. Fedorova, W.C. Nierman.
Comparative genomics of Aspergilli.
Aspergillus. Molecular biology and genomics, pp. 41-60
[86]
M.E. Ferreira, A.L. Colombo, I. Paulsen, Q. Ren, J. Wortman, J. Huang, et al.
The ergosterol biosynthesis pathway, transporter genes, and azole resistance in Aspergillus fumigatus.
Med Mycol, 43 (2005), pp. S313-S319
[87]
S. Fluckiger, P.R. Mittl, L. Scapozza, H. Fijten, G. Folkers, M.G. Grutter, et al.
Comparison of the crystal structures of the human manganese superoxide dismutase and the homologous Aspergillus fumigatus allergen at 2-A resolution.
J Immunol, 168 (2002), pp. 1267-1272
[88]
J.R. Fortwendel, K.K. Fuller, T.J. Stephens, W.C. Bacon, D.S. Askew, J.C. Rhodes.
Aspergillus fumigatus RasA regulates asexual development and cell wall integrity.
Eukaryot Cell, 7 (2008), pp. 1530-1539
[89]
J.R. Fortwendel, P.R. Juvvadi, N. Pinchai, B.Z. Perfect, J.A. Alspaugh, J.R. Perfect, et al.
Differential effects of inhibiting chitin and 1,3-{beta}-D-glucan synthesis in ras and calcineurin mutants of Aspergillus fumigatus.
Antimicrob Agents Chemother, 53 (2009), pp. 476-482
[90]
J.R. Fortwendel, W. Zhao, R. Bhabhra, S. Park, D.S. Perlin, D.S. Askew, et al.
A fungus-specific ras homolog contributes to the hyphal growth and virulence of Aspergillus fumigatus.
Eukaryot Cell, 4 (2005), pp. 1982-1989
[91]
C. Fradin, M. Kretschmar, T. Nichterlein, C. Gaillardin, C. d’Enfert, B. Hube.
Stage-specific gene expression of Candida albicans in human blood.
Mol Microbiol, 47 (2003), pp. 1523-1543
[92]
J.C. Frisvad, C. Rank, K.F. Nielsen, T.O. Larsen.
Metabolomics of Aspergillus fumigatus.
Med Mycol, 47 (2009), pp. S53-S71
[93]
I. Fujii, Y. Yasuoka, H.F. Tsai, Y.C. Chang, K.J. Kwon-Chung, Y. Ebizuka.
Hydrolytic polyketide shortening by ayg1p, a novel enzyme involved in fungal melanin biosynthesis.
J Biol Chem, 279 (2004), pp. 44613-44620
[94]
K.K. Fuller, W. Zhao, D.S. Askew, J.C. Rhodes.
Deletion of the protein kinase A regulatory subunit leads to deregulation of mitochondrial activation and nuclear duplication in Aspergillus fumigatus.
Eukaryot Cell, 8 (2009), pp. 271-277
[95]
M.E. García, J. Caballero, I. Blanco, M. Cruzado, E. Costas, J.L. Blanco.
Changes in the elastase activity and colonization ability of Aspergillus fumigatus after successive inoculations in mice.
Rev Iberoam Micol, 23 (2006), pp. 221-223
[96]
D.M. Gardiner, B.J. Howlett.
Bioinformatic and expression analysis of the putative gliotoxin biosynthetic gene cluster of Aspergillus fumigatus.
FEMS Microbiol Lett, 248 (2005), pp. 241-248
[97]
D.M. Gardiner, P. Waring, B.J. Howlett.
The epipolythiodioxopiperazine (ETP) class of fungal toxins: distribution, mode of action, functions and biosynthesis.
Microbiology, 151 (2005), pp. 1021-1032
[98]
A. Gastebois, C. Clavaud, V. Aimanianda, J.P. Latge.
Aspergillus fumigatus: cell wall polysaccharides, their biosynthesis and organization.
Future Microbiol, 4 (2009), pp. 583-595
[99]
A. Gehrke, T. Heinekamp, I.D. Jacobsen, A.A. Brakhage.
Heptahelical receptors GprC and GprD of Aspergillus fumigatus are essential regulators of colony growth, hyphal morphogenesis and virulence.
Appl Environ Microbiol, 76 (2010), pp. 3989-3998
[100]
A.G. Glaser, A.I. Kirsch, S. Zeller, G. Menz, C. Rhyner, R. Crameri.
Molecular and immunological characterization of Asp f 34, a novel major cell wall allergen of Aspergillus fumigatus.
[101]
A.G. Glaser, A. Limacher, S. Fluckiger, A. Scheynius, L. Scapozza, R. Crameri.
Analysis of the cross-reactivity and of the 1.5 A crystal structure of the Malassezia sympodialis Mala s 6 allergen, a member of the cyclophilin pan-allergen family.
Biochem J, 396 (2006), pp. 41-49
[102]
F.N. Gravelat, T. Doedt, L.Y. Chiang, H. Liu, S.G. Filler, T.F. Patterson, et al.
In vivo analysis of Aspergillus fumigatus developmental gene expression determined by real-time reverse transcription-PCR.
Infect Immun, 76 (2008), pp. 3632-3639
[103]
F.N. Gravelat, D.E. Ejzykowicz, L.Y. Chiang, J.C. Chabot, M. Urb, K.D. Macdonald, et al.
Aspergillus fumigatus MedA governs adherence, host cell interactions and virulence.
Cell Microbiol, 12 (2010), pp. 473-488
[104]
N.T. Gross, K. Nessa, P. Camner, C. Jarstrand.
Production of nitric oxide by rat alveolar macrophages stimulated by Cryptococcus neoformans or Aspergillus fumigatus.
Med Mycol, 37 (1999), pp. 151-157
[105]
C. Grosse, T. Heinekamp, O. Kniemeyer, A. Gehrke, A.A. Brakhage.
Protein kinase A regulates growth, sporulation, and pigment formation in Aspergillus fumigatus.
Appl Environ Microbiol, 74 (2008), pp. 4923-4933
[106]
A. Grundmann, T. Kuznetsova, S.S. Afiyatullov, S.M. Li.
FtmPT2, an N-prenyltransferase from Aspergillus fumigatus, catalyses the last step in the biosynthesis of fumitremorgin B.
Chembiochem, 9 (2008), pp. 2059-2063
[107]
A. Grundmann, S.M. Li.
Overproduction, purification and characterization of FtmPT1, a brevianamide F prenyltransferase from Aspergillus fumigatus.
Microbiology, 151 (2005), pp. 2199-2207
[108]
H. Hebart, C. Bollinger, P. Fisch, J. Sarfati, C. Meisner, M. Baur, et al.
Analysis of T-cell responses to Aspergillus fumigatus antigens in healthy individuals and patients with hematologic malignancies.
Blood, 100 (2002), pp. 4521-4528
[109]
S. Hemmann, K. Blaser, R. Crameri.
Allergens of Aspergillus fumigatus and Candida boidinii share IgE-binding epitopes.
Am J Respir Crit Care Med, 156 (1997), pp. 1956-1962
[110]
M. Hensel, H.N. Arst Jr, A. Aufauvre-Brown, D.W. Holden.
The role of the Aspergillus fumigatus areA gene in invasive pulmonary aspergillosis.
Mol Gen Genet, 258 (1998), pp. 553-557
[111]
A.H. Hissen, A.N. Wan, M.L. Warwas, L.J. Pinto, M.M. Moore.
The Aspergillus fumigatus siderophore biosynthetic gene sidA, encoding L-ornithine N5-oxygenase, is required for virulence.
Infect Immun, 73 (2005), pp. 5493-5503
[112]
T.M. Hohl, M. Feldmesser.
Aspergillus fumigatus: principles of pathogenesis and host defense.
Eukaryot Cell, 6 (2007), pp. 1953-1963
[113]
M.D. Holdom, B. Lechenne, R.J. Hay, A.J. Hamilton, M. Monod.
Production and characterization of recombinant Aspergillus fumigatus Cu,Zn superoxide dismutase and its recognition by immune human sera.
J Clin Microbiol, 38 (2000), pp. 558-562
[114]
W. Hu, S. Sillaots, S. Lemieux, J. Davison, S. Kauffman, A. Breton, et al.
Essential gene identification and drug target prioritization in Aspergillus fumigatus.
[115]
O. Ibrahim-Granet, C. D’Enfert.
The Aspergillus fumigatus mepB gene encodes an 82 kDa intracellular metalloproteinase structurally related to mammalian thimet oligopeptidases.
Microbiology, 143 (1997), pp. 2247-2253
[116]
O. Ibrahim-Granet, M. Dubourdeau, J.P. Latge, P. Ave, M. Huerre, A.A. Brakhage, et al.
Methylcitrate synthase from Aspergillus fumigatus is essential for manifestation of invasive aspergillosis.
Cell Microbiol, 10 (2008), pp. 134-148
[117]
K. Ishibashi, N.N. Miura, Y. Adachi, H. Tamura, S. Tanaka, N. Ohno.
The solubilization and biological activities of Aspergillus beta-(1→3)-D-glucan.
FEMS Immunol Med Microbiol, 42 (2004), pp. 155-166
[118]
B. Jahn, F. Boukhallouk, J. Lotz, K. Langfelder, G. Wanner, A.A. Brakhage.
Interaction of human phagocytes with pigmentless Aspergillus conidia.
Infect Immun, 68 (2000), pp. 3736-3739
[119]
B. Jahn, A. Koch, A. Schmidt, G. Wanner, H. Gehringer, S. Bhakdi, et al.
Isolation and characterization of a pigmentless-conidium mutant of Aspergillus fumigatus with altered conidial surface and reduced virulence.
Infect Immun, 65 (1997), pp. 5110-5117
[120]
B. Jahn, K. Langfelder, U. Schneider, C. Schindel, A.A. Brakhage.
PKSP-dependent reduction of phagolysosome fusion and intracellular kill of Aspergillus fumigatus conidia by human monocyte-derived macrophages.
Cell Microbiol, 4 (2002), pp. 793-803
[121]
K. Jaton-Ogay, S. Paris, M. Huerre, M. Quadroni, R. Falchetto, G. Togni, et al.
Cloning and disruption of the gene encoding an extracellular metalloprotease of Aspergillus fumigatus.
Mol Microbiol, 14 (1994), pp. 917-928
[122]
P.R. Juvvadi, J.R. Fortwendel, N. Pinchai, B.Z. Perfect, J. Heitman, W.J. Steinbach.
Calcineurin localizes to the hyphal septum in Aspergillus fumigatus: implications for septum formation and conidiophore development.
Eukaryot Cell, 7 (2008), pp. 1606-1610
[123]
R. Kao, J. Davies.
Molecular dissection of mitogillin reveals that the fungal ribotoxin are a family of natural genetically engineered ribonucleases.
J Biol Chem, 274 (1999), pp. 12576-12582
[124]
R. Kao, J. Davies.
Fungal ribotoxins: a family of naturally engineered targeted toxins?.
Biochem Cell Biol, 73 (1995), pp. 1151-1159
[125]
N. Kato, H. Suzuki, H. Takagi, Y. Asami, H. Kakeya, M. Uramoto, et al.
Identification of cytochrome P450s required for fumitremorgin biosynthesis in Aspergillus fumigatus.
Chembiochem, 10 (2009), pp. 920-928
[126]
M.A. Klich.
Identification of clinically relevant aspergilli.
Med Mycol, 44 (2006), pp. 127-131
[127]
D.J. Klionsky, H. Abeliovich, P. Agostinis, D.K. Agrawal, G. Aliev, D.S. Askew, et al.
Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes.
Autophagy, 4 (2008), pp. 151-175
[128]
D.J. Klionsky, J.M. Cregg, W.A. Dunn Jr, S.D. Emr, Y. Sakai, I.V. Sandoval, et al.
A unified nomenclature for yeast autophagy-related genes.
Dev Cell, 5 (2003), pp. 539-545
[129]
A.P. Knutsen, P.S. Hutchinson, G.M. Albers, J. Consolino, J. Smick, V.P. Kurup.
Increased sensitivity to IL-4 in cystic fibrosis patients with allergic bronchopulmonary aspergillosis.
Allergy, 59 (2004), pp. 81-87
[130]
P.E. Kolattukudy, J.D. Lee, L.M. Rogers, P. Zimmerman, S. Ceselski, B. Fox, et al.
Evidence for possible involvement of an elastolytic serine protease in aspergillosis.
Infect Immun, 61 (1993), pp. 2357-2368
[131]
D.P. Kontoyiannis, R.E. Lewis, N. Osherov, N.D. Albert, G.S. May.
Combination of caspofungin with inhibitors of the calcineurin pathway attenuates growth in vitro in Aspergillus species.
J Antimicrob Chemother, 51 (2003), pp. 313-316
[132]
T.R. Kozel.
Activation of the complement system by pathogenic fungi.
Clin Microbiol Rev, 9 (1996), pp. 34-46
[133]
S. Krappmann.
Pathogenicity determinants and allergens.
The aspergilli, pp. 377-400
[134]
S. Krappmann, E.M. Bignell, U. Reichard, T. Rogers, K. Haynes, G.H. Braus.
The Aspergillus fumigatus transcriptional activator CpcA contributes significantly to the virulence of this fungal pathogen.
Mol Microbiol, 52 (2004), pp. 785-799
[135]
T. Kumagai, T. Nagata, Y. Kudo, Y. Fukuchi, K. Ebina, K. Yokota.
Cytotoxic activity and cytokine gene induction of Asp-hemolysin to murine macrophages.
Nippon Ishinkin Gakkai Zasshi, 40 (1999), pp. 217-222
[136]
A. Kumar, L.V. Reddy, A. Sochanik, V.P. Kurup.
Isolation and characterization of a recombinant heat shock protein of Aspergillus fumigatus.
J Allergy Clin Immunol, 91 (1993), pp. 1024-1030
[137]
C. Kupfahl, T. Heinekamp, G. Geginat, T. Ruppert, A. Hartl, H. Hof, et al.
Deletion of the gliP gene of Aspergillus fumigatus results in loss of gliotoxin production but has no effect on virulence of the fungus in a low-dose mouse infection model.
Mol Microbiol, 62 (2006), pp. 292-302
[138]
C. Kupfahl, A. Michalka, C. Lass-Florl, G. Fischer, G. Haase, T. Ruppert, et al.
Gliotoxin production by clinical and environmental Aspergillus fumigatus strains.
Int J Med Microbiol, 298 (2008), pp. 319-327
[139]
V.P. Kurup.
Interaction of Aspergillus fumigatus spores and pulmonary alveolar macrophages of rabbits.
Immunobiology, 166 (1984), pp. 53-61
[140]
V.P. Kurup, B. Banerjee, S. Hemmann, P.A. Greenberger, K. Blaser, R. Crameri.
Selected recombinant Aspergillus fumigatus allergens bind specifically to IgE in ABPA.
Clin Exp Allergy, 30 (2000), pp. 988-993
[141]
V.P. Kurup, H.D. Shen, H. Vijay.
Immunobiology of fungal allergens.
Int Arch Allergy Immunol, 129 (2002), pp. 181-188
[142]
K.J. Kwon-Chung, J.A. Sugui.
Sexual reproduction in Aspergillus species of medical or economical importance: why so fastidious?.
Trends Microbiol, 17 (2009), pp. 481-487
[143]
K.J. Kwon-Chung, J.A. Sugui.
What do we know about the role of gliotoxin in the pathobiology of Aspergillus fumigatus?.
Med Mycol, 47 (2009), pp. S97-S103
[144]
H.Y. Lai, M.F. Tam, R.B. Tang, H. Chou, C.Y. Chang, J.J. Tsai, et al.
cDNA cloning and immunological characterization of a newly identified enolase allergen from Penicillium citrinum and Aspergillus fumigatus.
Int Arch Allergy Immunol, 127 (2002), pp. 181-190
[145]
C. Lamarre, O. Ibrahim-Granet, C. Du, R. Calderone, J.P. Latge.
Characterization of the SKN7 ortholog of Aspergillus fumigatus.
Fungal Genet Biol, 44 (2007), pp. 682-690
[146]
C. Lamarre, S. Sokol, J.P. Debeaupuis, C. Henry, C. Lacroix, P. Glaser, et al.
Transcriptomic analysis of the exit from dormancy of Aspergillus fumigatus conidia.
BMC Genom, 9 (2008), pp. 417
[147]
K. Lambou, C. Lamarre, R. Beau, N. Dufour, J.P. Latge.
Functional analysis of the superoxide dismutase family in Aspergillus fumigatus.
[148]
B. Lamy, M. Moutaouakil, J.P. Latge, J. Davies.
Secretion of a potential virulence factor, a fungal ribonucleotoxin, during human aspergillosis infections.
Mol Microbiol, 5 (1991), pp. 1811-1815
[149]
K. Langfelder, S. Gattung, A.A. Brakhage.
A novel method used to delete a new Aspergillus fumigatus ABC transporter-encoding gene.
Curr Genet, 41 (2002), pp. 268-274
[150]
K. Langfelder, B. Jahn, H. Gehringer, A. Schmidt, G. Wanner, A.A. Brakhage.
Identification of a polyketide synthase gene (pksP) of Aspergillus fumigatus involved in conidial pigment biosynthesis and virulence.
Med Microbiol Immunol, 187 (1998), pp. 79-89
[151]
K. Langfelder, M. Streibel, B. Jahn, G. Haase, A.A. Brakhage.
Biosynthesis of fungal melanins and their importance for human pathogenic fungi.
Fungal Genet Biol, 38 (2003), pp. 143-158
[152]
J.P. Latge.
The cell wall: a carbohydrate armour for the fungal cell.
Mol Microbiol, 66 (2007), pp. 279-290
[153]
J.P. Latge.
The pathobiology of Aspergillus fumigatus.
Trends Microbiol, 9 (2001), pp. 382-389
[154]
J.P. Latge.
Aspergillus fumigatus and aspergillosis.
Clin Microbiol Rev, 12 (1999), pp. 310-350
[155]
J.P. Latge, J.P. Debeaupuis, J. Sarfati, M. Diaquin, S. Paris.
Cell wall antigens in Aspergillus fumigatus.
Arch Med Res, 24 (1993), pp. 269-274
[156]
J.P. Latge, I. Mouyna, F. Tekaia, A. Beauvais, J.P. Debeaupuis, W. Nierman.
Specific molecular features in the organization and biosynthesis of the cell wall of Aspergillus fumigatus.
Med Mycol, 43 (2005), pp. S15-S22
[157]
J.D. Lee, P.E. Kolattukudy.
Molecular cloning of the cDNA and gene for an elastinolytic aspartic proteinase from Aspergillus fumigatus and evidence of its secretion by the fungus during invasion of the host lung.
Infect Immun, 63 (1995), pp. 3796-3803
[158]
F. Lessing, O. Kniemeyer, I. Wozniok, J. Loeffler, O. Kurzai, A. Haertl, et al.
The Aspergillus fumigatus transcriptional regulator AfYap1 represents the major regulator for defense against reactive oxygen intermediates but is dispensable for pathogenicity in an intranasal mouse infection model.
Eukaryot Cell, 6 (2007), pp. 2290-2302
[159]
R.E. Lewis, N.P. Wiederhold, J. Chi, X.Y. Han, K.V. Komanduri, D.P. Kontoyiannis, et al.
Detection of gliotoxin in experimental and human aspergillosis.
Infect Immun, 73 (2005), pp. 635-637
[160]
H. Li, H. Zhou, Y. Luo, H. Ouyang, H. Hu, C. Jin.
Glycosylphosphatidylinositol (GPI) anchor is required in Aspergillus fumigatus for morphogenesis and virulence.
Mol Microbiol, 64 (2007), pp. 1014-1027
[161]
B. Liebmann, S. Gattung, B. Jahn, A.A. Brakhage.
cAMP signaling in Aspergillus fumigatus is involved in the regulation of the virulence gene pksP and in defense against killing by macrophages.
Mol Genet Genom, 269 (2003), pp. 420-435
[162]
B. Liebmann, T.W. Muhleisen, M. Muller, M. Hecht, G. Weidner, A. Braun, et al.
Deletion of the Aspergillus fumigatus lysine biosynthesis gene lysF encoding homoaconitase leads to attenuated virulence in a low-dose mouse infection model of invasive aspergillosis.
Arch Microbiol, 181 (2004), pp. 378-383
[163]
B. Liebmann, M. Muller, A. Braun, A.A. Brakhage.
The cyclic AMP-dependent protein kinase a network regulates development and virulence in Aspergillus fumigatus.
Infect Immun, 72 (2004), pp. 5193-5203
[164]
M.B. Linder, G.R. Szilvay, T. Nakari-Setala, M.E. Penttila.
Hydrophobins: the protein-amphiphiles of filamentous fungi.
FEMS Microbiol Rev, 29 (2005), pp. 877-896
[165]
J. Liu, Z.J. Yang, Z.H. Meng.
The isolation, purification and identification of fumitremorgin B produced by Aspergillus fumigatus.
Biomed Environ Sci, 9 (1996), pp. 1-11
[166]
S. Lodeiro, Q. Xiong, W.K. Wilson, Y. Ivanova, M.L. Smith, G.S. May, et al.
Protostadienol biosynthesis and metabolism in the pathogenic fungus Aspergillus fumigatus.
Org Lett, 11 (2009), pp. 1241-1244
[167]
Y. Ma, J. Qiao, W. Liu, Z. Wan, X. Wang, R. Calderone, et al.
The sho1 sensor regulates growth, morphology, and oxidant adaptation in Aspergillus fumigatus but is not essential for development of invasive pulmonary aspergillosis.
Infect Immun, 76 (2008), pp. 1695-1701
[168]
M. Machida, K. Asai, M. Sano, T. Tanaka, T. Kumagai, G. Terai, et al.
Genome sequencing and analysis of Aspergillus oryzae.
Nature, 438 (2005), pp. 1157-1161
[169]
C. Maerker, M. Rohde, A.A. Brakhage, M. Brock.
Methylcitrate synthase from Aspergillus fumigatus. Propionyl-CoA affects polyketide synthesis, growth and morphology of conidia.
[170]
J.H. Mah, J.H. Yu.
Upstream and downstream regulation of asexual development in Aspergillus fumigatus.
Eukaryot Cell, 5 (2006), pp. 1585-1595
[171]
S. Maiya, A. Grundmann, S.M. Li, G. Turner.
The fumitremorgin gene cluster of Aspergillus fumigatus: identification of a gene encoding brevianamide F synthetase.
Chembiochem, 7 (2006), pp. 1062-1069
[172]
A. Markaryan, I. Morozova, H. Yu, P.E. Kolattukudy.
Purification and characterization of an elastinolytic metalloprotease from Aspergillus fumigatus and immunoelectron microscopic evidence of secretion of this enzyme by the fungus invading the murine lung.
Infect Immun, 62 (1994), pp. 2149-2157
[173]
G. Maschmeyer, A. Haas, O.A. Cornely.
Invasive aspergillosis: epidemiology, diagnosis and management in immunocompromised patients.
Drugs, 67 (2007), pp. 1567-1601
[174]
D. Maubon, S. Park, M. Tanguy, M. Huerre, C. Schmitt, M.C. Prevost, et al.
AGS3, an alpha(1–3)glucan synthase gene family member of Aspergillus fumigatus, modulates mycelium growth in the lung of experimentally infected mice.
Fungal Genet Biol, 43 (2006), pp. 366-375
[175]
G.S. May.
Mitogen-activated protein kinase pathways in Aspergilli.
The Aspergilli. Genomics, medical aspects, biotechnology, and research methods, pp. 121-127
[176]
G.S. May, T. Xue, D.P. Kontoyiannis, M.C. Gustin.
Mitogen activated protein kinases of Aspergillus fumigatus.
Med Mycol, 43 (2005), pp. S83-S86
[177]
E. Mellado, A. Aufauvre-Brown, N.A. Gow, D.W. Holden.
The Aspergillus fumigatus chsC and chsG genes encode class III chitin synthases with different functions.
Mol Microbiol, 20 (1996), pp. 667-679
[178]
E. Mellado, G. Dubreucq, P. Mol, J. Sarfati, S. Paris, M. Diaquin, et al.
Cell wall biogenesis in a double chitin synthase mutant (chsG-/chsE-) of Aspergillus fumigatus.
Fungal Genet Biol, 38 (2003), pp. 98-109
[179]
E. Mellado, C.A. Specht, P.W. Robbins, D.W. Holden.
Cloning and characterization of chsD, a chitin synthase-like gene of Aspergillus fumigatus.
FEMS Microbiol Lett, 143 (1996), pp. 69-76
[180]
T. Meri, A. Hartmann, D. Lenk, R. Eck, R. Wurzner, J. Hellwage, et al.
The yeast Candida albicans binds complement regulators factor H and FHL-1.
Infect Immun, 70 (2002), pp. 5185-5192
[181]
C.G. Mitchell, J. Slight, K. Donaldson.
Diffusible component from the spore surface of the fungus Aspergillus fumigatus which inhibits the macrophage oxidative burst is distinct from gliotoxin and other hyphal toxins.
Thorax, 52 (1997), pp. 796-801
[182]
H. Mitsuguchi, Y. Seshime, I. Fujii, M. Shibuya, Y. Ebizuka, T. Kushiro.
Biosynthesis of steroidal antibiotic fusidanes: functional analysis of oxidosqualene cyclase and subsequent tailoring enzymes from Aspergillus fumigatus.
J Am Chem Soc, 131 (2009), pp. 6402-6411
[183]
M. Monod, K. Jaton-Ogay, U. Reichard.
Aspergillus fumigatus-secreted proteases as antigenic molecules and virulence factors.
Contrib Microbiol, 2 (1999), pp. 182-192
[184]
M. Monod, S. Paris, J. Sarfati, K. Jaton-Ogay, P. Ave, J.P. Latge.
Virulence of alkaline protease-deficient mutants of Aspergillus fumigatus.
FEMS Microbiol Lett, 106 (1993), pp. 39-46
[185]
Montagnoli C, Bozza S, Gaziano R, Zelante T, Bonifazi P, Moretti S, et al. Immunity and tolerance to Aspergillus fumigatus. Novartis Found Symp 2006;279:66–77; discussion 77–79, 216–219.
[186]
M.A. Moreno, O. Ibrahim-Granet, R. Vicentefranqueira, J. Amich, P. Ave, F. Leal, et al.
The regulation of zinc homeostasis by the ZafA transcriptional activator is essential for Aspergillus fumigatus virulence.
Mol Microbiol, 64 (2007), pp. 1182-1197
[187]
I. Mouyna, T. Fontaine, M. Vai, M. Monod, W.A. Fonzi, M. Diaquin, et al.
Glycosylphosphatidylinositol-anchored glucanosyltransferases play an active role in the biosynthesis of the fungal cell wall.
J Biol Chem, 275 (2000), pp. 14882-14889
[188]
I. Mouyna, R.P. Hartland, T. Fontaine, M. Diaquin, C. Simenel, M. Delepierre, et al.
A 1,3-beta-glucanosyltransferase isolated from the cell wall of Aspergillus fumigatus is a homologue of the yeast Bgl2p.
Microbiology, 144 (1998), pp. 3171-3180
[189]
I. Mouyna, C. Henry, T.L. Doering, J.P. Latge.
Gene silencing with RNA interference in the human pathogenic fungus Aspergillus fumigatus.
FEMS Microbiol Lett, 237 (2004), pp. 317-324
[190]
I. Mouyna, M. Monod, T. Fontaine, B. Henrissat, B. Lechenne, J.P. Latge.
Identification of the catalytic residues of the first family of beta(1–3)glucanosyltransferases identified in fungi.
Biochem J, 347 (2000), pp. 741-747
[191]
I. Mouyna, W. Morelle, M. Vai, M. Monod, B. Lechenne, T. Fontaine, et al.
Deletion of GEL2 encoding for a beta(1–3)glucanosyltransferase affects morphogenesis and virulence in Aspergillus fumigatus.
Mol Microbiol, 56 (2005), pp. 1675-1688
[192]
I. Mouyna, J. Sarfati, P. Recco, T. Fontaine, B. Henrissatz, J.P. Latge.
Molecular characterization of a cell wall-associated beta(1–3)endoglucanase of Aspergillus fumigatus.
Med Mycol, 40 (2002), pp. 455-464
[193]
F. Muhlschlegal, W. Fonzi, L. Hoyer, T. Payne, F.M. Poulet, J. Clevenger, et al.
Molecular mechanisms of virulence in fungus–host interactions for Aspergillus fumigatus and Candida albicans.
Med Mycol, 36 (1998), pp. 238-248
[194]
A. Mullbacher, R.D. Eichner.
Immunosuppression in vitro by a metabolite of a human pathogenic fungus.
Proc Natl Acad Sci USA, 81 (1984), pp. 3835-3837
[195]
C.A. Munro, N.A. Gow.
Chitin synthesis in human pathogenic fungi.
Med Mycol, 39 (2001), pp. 41-53
[196]
A.M. Nascimento, G.H. Goldman, S. Park, S.A. Marras, G. Delmas, U. Oza, et al.
Multiple resistance mechanisms among Aspergillus fumigatus mutants with high-level resistance to itraconazole.
Antimicrob Agents Chemother, 47 (2003), pp. 1719-1726
[197]
C. Nathan, M.U. Shiloh.
Reactive oxygen and nitrogen intermediates in the relationship between mammalian hosts and microbial pathogens.
Proc Natl Acad Sci USA, 97 (2000), pp. 8841-8848
[198]
J.B. Neilands.
Siderophores.
Arch Biochem Biophys, 302 (1993), pp. 1-3
[199]
M.G. Netea, A. Warris, J.W. Van der Meer, M.J. Fenton, T.J. Verver-Janssen, L.E. Jacobs, et al.
Aspergillus fumigatus evades immune recognition during germination through loss of toll-like receptor-4-mediated signal transduction.
J Infect Dis, 188 (2003), pp. 320-326
[200]
K. Nielsen, J. Heitman.
Sex and virulence of human pathogenic fungi.
Adv Genet, 57 (2007), pp. 143-173
[201]
W.C. Nierman, A. Pain, M.J. Anderson, J.R. Wortman, H.S. Kim, J. Arroyo, et al.
Genomic sequence of the pathogenic and allergenic filamentous fungus Aspergillus fumigatus.
Nature, 438 (2005), pp. 1151-1156
[202]
S. Nigam, P.C. Ghosh, P.U. Sarma.
A new glycoprotein allergen/antigen with the protease activity from Aspergillus fumigatus.
Int Arch Allergy Immunol, 132 (2003), pp. 124-131
[203]
E.A. Nollen, J.F. Brunsting, H. Roelofsen, L.A. Weber, H.H. Kampinga.
In vivo chaperone activity of heat shock protein 70 and thermotolerance.
Mol Cell Biol, 19 (1999), pp. 2069-2079
[204]
C.M. O’Gorman, H.T. Fuller, P.S. Dyer.
Discovery of a sexual cycle in the opportunistic fungal pathogen Aspergillus fumigatus.
Nature, 457 (2009), pp. 471-474
[205]
M. Ok, J.P. Latge, C. Baeuerlein, F. Ebel, M. Mezger, M. Topp, et al.
Immune responses of human immature dendritic cells can be modulated by the recombinant Aspergillus fumigatus antigen Aspf1.
Clin Vaccine Immunol, 16 (2009), pp. 1485-1492
[206]
E. Orciuolo, M. Stanzani, M. Canestraro, S. Galimberti, G. Carulli, R. Lewis, et al.
Effects of Aspergillus fumigatus gliotoxin and methylprednisolone on human neutrophils: implications for the pathogenesis of invasive aspergillosis.
J Leukoc Biol, 82 (2007), pp. 839-848
[207]
M. Orzechowski Xavier, A.C. Pasqualotto, P. Uchoa Sales Mda, C. Bittencourt Severo, J.J. Peixoto Camargo, L.C. Severo.
Invasive pulmonary aspergillosis due to a mixed infection caused by Aspergillus flavus and Aspergillus fumigatus.
Rev Iberoam Micol, 25 (2008), pp. 176-178
[208]
N. Osherov.
The virulence of Aspergillus fumigatus.
New insights in medical mycology, pp. 185-212
[209]
G.E. Palmer, D.S. Askew, P.R. Williamson.
The diverse roles of autophagy in medically important fungi.
Autophagy, 4 (2008), pp. 982-988
[210]
J.C. Panepinto, B.G. Oliver, T.W. Amlung, D.S. Askew, J.C. Rhodes.
Expression of the Aspergillus fumigatus rheb homologue, rhbA, is induced by nitrogen starvation.
Fungal Genet Biol, 36 (2002), pp. 207-214
[211]
J.C. Panepinto, B.G. Oliver, J.R. Fortwendel, D.L. Smith, D.S. Askew, J.C. Rhodes.
Deletion of the Aspergillus fumigatus gene encoding the Ras-related protein RhbA reduces virulence in a model of invasive pulmonary aspergillosis.
Infect Immun, 71 (2003), pp. 2819-2826
[212]
M. Paoletti, C. Rydholm, E.U. Schwier, M.J. Anderson, G. Szakacs, F. Lutzoni, et al.
Evidence for sexuality in the opportunistic fungal pathogen Aspergillus fumigatus.
Curr Biol, 15 (2005), pp. 1242-1248
[213]
J. Pardo, C. Urban, E.M. Galvez, P.G. Ekert, U. Muller, J. Kwon-Chung, et al.
The mitochondrial protein Bak is pivotal for gliotoxin-induced apoptosis and a critical host factor of Aspergillus fumigatus virulence in mice.
J Cell Biol, 174 (2006), pp. 509-519
[214]
S. Paris, E. Boisvieux-Ulrich, B. Crestani, O. Houcine, D. Taramelli, L. Lombardi, et al.
Internalization of Aspergillus fumigatus conidia by epithelial and endothelial cells.
Infect Immun, 65 (1997), pp. 1510-1514
[215]
S. Paris, J.P. Debeaupuis, R. Crameri, M. Carey, F. Charles, M.C. Prevost, et al.
Conidial hydrophobins of Aspergillus fumigatus.
Appl Environ Microbiol, 69 (2003), pp. 1581-1588
[216]
S. Paris, D. Wysong, J.P. Debeaupuis, K. Shibuya, B. Philippe, R.D. Diamond, et al.
Catalases of Aspergillus fumigatus.
Infect Immun, 71 (2003), pp. 3551-3562
[217]
S.J. Park, B. Mehrad.
Innate immunity to Aspergillus species.
Clin Microbiol Rev, 22 (2009), pp. 535-551
[218]
C. Pazos, J. Ponton, A. Del Palacio.
Contribution of (1→3)-beta-D-glucan chromogenic assay to diagnosis and therapeutic monitoring of invasive aspergillosis in neutropenic adult patients: a comparison with serial screening for circulating galactomannan.
J Clin Microbiol, 43 (2005), pp. 299-305
[219]
S. Pepeljnjak, Z. Slobodnjak, M. Segvic, M. Peraica, M. Pavlovic.
The ability of fungal isolates from human lung aspergilloma to produce mycotoxins.
Hum Exp Toxicol, 23 (2004), pp. 15-19
[220]
R.M. Perrin, N.D. Fedorova, J.W. Bok, R.A. Cramer, J.R. Wortman, H.S. Kim, et al.
Transcriptional regulation of chemical diversity in Aspergillus fumigatus by LaeA.
[221]
M. Pihet, P. Vandeputte, G. Tronchin, G. Renier, P. Saulnier, S. Georgeault, et al.
Melanin is an essential component for the integrity of the cell wall of Aspergillus fumigatus conidia.
BMC Microbiol, 9 (2009), pp. 177
[222]
G.B. Pott, T.K. Miller, J.A. Bartlett, J.S. Palas, C.P. Selitrennikoff.
The isolation of FOS-1, a gene encoding a putative two-component histidine kinase from Aspergillus fumigatus.
Fungal Genet Biol, 31 (2000), pp. 55-67
[223]
T. Power, M. Ortoneda, J.P. Morrissey, A.D. Dobson.
Differential expression of genes involved in iron metabolism in Aspergillus fumigatus.
Int Microbiol, 9 (2006), pp. 281-287
[224]
P. Priyadarsiny, P.K. Swain, P.U. Sarma.
Expression and characterization of Asp fI, an immunodominant allergen/antigen of A. fumigatus in insect cell.
Mol Cell Biochem, 252 (2003), pp. 157-163
[225]
J. Qiao, D.P. Kontoyiannis, R. Calderone, D. Li, Y. Ma, Z. Wan, et al.
Afyap1, encoding a bZip transcriptional factor of Aspergillus fumigatus, contributes to oxidative stress response but is not essential to the virulence of this pathogen in mice immunosuppressed by cyclophosphamide and triamcinolone.
Med Mycol, 46 (2008), pp. 773-782
[226]
M.V. Ramesh, T.D. Sirakova, P.E. Kolattukudy.
Cloning and characterization of the cDNAs and genes (mep20) encoding homologous metalloproteinases from Aspergillus flavus and A. fumigatus.
Gene, 165 (1995), pp. 121-125
[227]
E.P. Reeves, K. Reiber, C. Neville, O. Scheibner, K. Kavanagh, S. Doyle.
A nonribosomal peptide synthetase (Pes1) confers protection against oxidative stress in Aspergillus fumigatus.
[228]
U. Reichard, S. Buttner, H. Eiffert, F. Staib, R. Ruchel.
Purification and characterisation of an extracellular serine proteinase from Aspergillus fumigatus and its detection in tissue.
J Med Microbiol, 33 (1990), pp. 243-251
[229]
U. Reichard, G.T. Cole, T.W. Hill, R. Ruchel, M. Monod.
Molecular characterization and influence on fungal development of ALP2, a novel serine proteinase from Aspergillus fumigatus.
Int J Med Microbiol, 290 (2000), pp. 549-558
[230]
U. Reichard, G.T. Cole, R. Ruchel, M. Monod.
Molecular cloning and targeted deletion of PEP2 which encodes a novel aspartic proteinase from Aspergillus fumigatus.
Int J Med Microbiol, 290 (2000), pp. 85-96
[231]
U. Reichard, M. Monod, F. Odds, R. Ruchel.
Virulence of an aspergillopepsin-deficient mutant of Aspergillus fumigatus and evidence for another aspartic proteinase linked to the fungal cell wall.
J Med Vet Mycol, 35 (1997), pp. 189-196
[232]
A. Rementeria, N. López-Molina, A. Ludwig, A.B. Vivanco, J. Bikandi, J. Ponton, et al.
Genes and molecules involved in Aspergillus fumigatus virulence.
Rev Iberoam Micol, 22 (2005), pp. 1-23
[233]
G. Reyes, A. Romans, C.K. Nguyen, G.S. May.
Novel mitogen-activated protein kinase MpkC of Aspergillus fumigatus is required for utilization of polyalcohol sugars.
Eukaryot Cell, 5 (2006), pp. 1934-1940
[234]
J.L. Richard, T.J. Dvorak, P.F. Ross.
Natural occurrence of gliotoxin in turkeys infected with Aspergillus fumigatus, Fresenius.
Mycopathologia, 134 (1996), pp. 167-170
[235]
D.L. Richie, D.S. Askew.
Autophagy in the filamentous fungus Aspergillus fumigatus.
Methods Enzymol, 451 (2008), pp. 241-250
[236]
D.L. Richie, K.K. Fuller, J. Fortwendel, M.D. Miley, J.W. McCarthy, M. Feldmesser, et al.
Unexpected link between metal ion deficiency and autophagy in Aspergillus fumigatus.
Eukaryot Cell, 6 (2007), pp. 2437-2447
[237]
O. Rigbers, S.M. Li.
Ergot alkaloid biosynthesis in Aspergillus fumigatus. Overproduction and biochemical characterization of a 4-dimethylallyltryptophan N-methyltransferase.
J Biol Chem, 283 (2008), pp. 26859-26868
[238]
N. Rispail, D.M. Soanes, C. Ant, R. Czajkowski, A. Grunler, R. Huguet, et al.
Comparative genomics of MAP kinase and calcium–calcineurin signalling components in plant and human pathogenic fungi.
Fungal Genet Biol, 46 (2009), pp. 287-298
[239]
G. Robson.
Hyphal cell biology.
Molecular fungal biology, pp. 164-184
[240]
J. Romano, G. Nimrod, N. Ben-Tal, Y. Shadkchan, K. Baruch, H. Sharon, et al.
Disruption of the Aspergillus fumigatus ECM33 homologue results in rapid conidial germination, antifungal resistance and hypervirulence.
Microbiology, 152 (2006), pp. 1919-1928
[241]
J. Ryckeboer, J. Mergaert, J. Coosemans, K. Deprins, J. Swings.
Microbiological aspects of biowaste during composting in a monitored compost bin.
J Appl Microbiol, 94 (2003), pp. 127-137
[242]
D. Sanglard, F. Ischer, O. Marchetti, J. Entenza, J. Bille.
Calcineurin A of Candida albicans: involvement in antifungal tolerance, cell morphogenesis and virulence.
Mol Microbiol, 48 (2003), pp. 959-976
[243]
C. Sasse, E.M. Bignell, M. Hasenberg, K. Haynes, M. Gunzer, G.H. Braus, et al.
Basal expression of the Aspergillus fumigatus transcriptional activator CpcA is sufficient to support pulmonary aspergillosis.
Fungal Genet Biol, 45 (2008), pp. 693-704
[244]
P.S. Schmalhorst, S. Krappmann, W. Vervecken, M. Rohde, M. Muller, G.H. Braus, et al.
Contribution of galactofuranose to the virulence of the opportunistic pathogen Aspergillus fumigatus.
Eukaryot Cell, 7 (2008), pp. 1268-1277
[245]
F. Schobel, I.D. Jacobsen, M. Brock.
Evaluation of lysine biosynthesis as antifungal drug target: biochemical characterization of Aspergillus fumigatus homocitrate synthase and virulence studies.
Eukaryot Cell, 9 (2010), pp. 878-893
[246]
M. Schrettl, E. Bignell, C. Kragl, C. Joechl, T. Rogers, H.N. Arst Jr, et al.
Siderophore biosynthesis but not reductive iron assimilation is essential for Aspergillus fumigatus virulence.
J Exp Med, 200 (2004), pp. 1213-1219
[247]
M. Schrettl, E. Bignell, C. Kragl, Y. Sabiha, O. Loss, M. Eisendle, et al.
Distinct roles for intra- and extracellular siderophores during Aspergillus fumigatus infection.
PLoS Pathog, 3 (2007), pp. 1195-1207
[248]
M. Schrettl, H.S. Kim, M. Eisendle, C. Kragl, W.C. Nierman, T. Heinekamp, et al.
SreA-mediated iron regulation in Aspergillus fumigatus.
Mol Microbiol, 70 (2008), pp. 27-43
[249]
H.D. Shen, W.L. Lin, M.F. Tam, H. Chou, C.W. Wang, J.J. Tsai, et al.
Identification of vacuolar serine proteinase as a major allergen of Aspergillus fumigatus by immunoblotting and N-terminal amino acid sequence analysis.
Clin Exp Allergy, 31 (2001), pp. 295-302
[250]
D.K. Shen, A.D. Noodeh, A. Kazemi, R. Grillot, G. Robson, J.F. Brugere.
Characterisation and expression of phospholipases B from the opportunistic fungus Aspergillus fumigatus.
FEMS Microbiol Lett, 239 (2004), pp. 87-93
[251]
H.D. Shen, M.F. Tam, H. Chou, S.H. Han.
The importance of serine proteinases as aeroallergens associated with asthma.
Int Arch Allergy Immunol, 119 (1999), pp. 259-264
[252]
K. Shibuya, M. Takaoka, K. Uchida, M. Wakayama, H. Yamaguchi, K. Takahashi, et al.
Histopathology of experimental invasive pulmonary aspergillosis in rats: pathological comparison of pulmonary lesions induced by specific virulent factor deficient mutants.
Microb Pathog, 27 (1999), pp. 123-131
[253]
K.S. Shin, N.J. Kwon, J.H. Yu.
Gbetagamma-mediated growth and developmental control in Aspergillus fumigatus.
Curr Genet, 55 (2009), pp. 631-641
[254]
C. Shinohara, K. Hasumi, A. Endo.
Inhibition of oxidized low-density lipoprotein metabolism in macrophage J774 by helvolic acid.
Biochim Biophys Acta, 1167 (1993), pp. 303-306
[255]
T.D. Sirakova, A. Markaryan, P.E. Kolattukudy.
Molecular cloning and sequencing of the cDNA and gene for a novel elastinolytic metalloproteinase from Aspergillus fumigatus and its expression in Escherichia coli.
Infect Immun, 62 (1994), pp. 4208-4218
[256]
J.W. Slaven, M.J. Anderson, D. Sanglard, G.K. Dixon, J. Bille, I.S. Roberts, et al.
Increased expression of a novel Aspergillus fumigatus ABC transporter gene, atrF, in the presence of itraconazole in an itraconazole resistant clinical isolate.
Fungal Genet Biol, 36 (2002), pp. 199-206
[257]
J.M. Smith, J.E. Davies, D.W. Holden.
Construction and pathogenicity of Aspergillus fumigatus mutants that do not produce the ribotoxin restrictocin.
Mol Microbiol, 9 (1993), pp. 1071-1077
[258]
F.M. Soriani, I. Malavazi, M.E. Da Silva Ferreira, M. Savoldi, M.R. Von Zeska Kress, M.H. de Souza Goldman, et al.
Functional characterization of the Aspergillus fumigatus CRZ1 homologue, CrzA.
Mol Microbiol, 67 (2008), pp. 1274-1291
[259]
F.M. Soriani, I. Malavazi, M. Savoldi, E. Espeso, T.M. Dinamarco, L.A. Bernardes, et al.
Identification of possible targets of the Aspergillus fumigatus CRZ1 homologue, CrzA.
BMC Microbiol, 10 (2010), pp. 12
[260]
B. Spiess, W. Seifarth, M. Hummel, O. Frank, A. Fabarius, C. Zheng, et al.
DNA microarray-based detection and identification of fungal pathogens in clinical samples from neutropenic patients.
J Clin Microbiol, 45 (2007), pp. 3743-3753
[261]
S. Spikes, R. Xu, C.K. Nguyen, G. Chamilos, D.P. Kontoyiannis, R.H. Jacobson, et al.
Gliotoxin production in Aspergillus fumigatus contributes to host-specific differences in virulence.
J Infect Dis, 197 (2008), pp. 479-486
[262]
M. Stanzani, E. Orciuolo, R. Lewis, D.P. Kontoyiannis, S.L. Martins, L.S. St John, et al.
Aspergillus fumigatus suppresses the human cellular immune response via gliotoxin-mediated apoptosis of monocytes.
Blood, 105 (2005), pp. 2258-2265
[263]
B.R. Steen, S. Zuyderduyn, D.L. Toffaletti, M. Marra, S.J. Jones, J.R. Perfect, et al.
Cryptococcus neoformans gene expression during experimental cryptococcal meningitis.
Eukaryot Cell, 2 (2003), pp. 1336-1349
[264]
N. Steffan, A. Grundmann, S. Afiyatullov, H. Ruan, S.M. Li.
FtmOx1, a non-heme Fe(II) and alpha-ketoglutarate-dependent dioxygenase, catalyses the endoperoxide formation of verruculogen in Aspergillus fumigatus.
Org Biomol Chem, 7 (2009), pp. 4082-4087
[265]
W.J. Steinbach, R.A. Cramer Jr, B.Z. Perfect, Y.G. Asfaw, T.C. Sauer, L.K. Najvar, et al.
Calcineurin controls growth, morphology, and pathogenicity in Aspergillus fumigatus.
Eukaryot Cell, 5 (2006), pp. 1091-1103
[266]
W.J. Steinbach, R.A. Cramer Jr, B.Z. Perfect, C. Henn, K. Nielsen, J. Heitman, et al.
Calcineurin inhibition or mutation enhances cell wall inhibitors against Aspergillus fumigatus.
Antimicrob Agents Chemother, 51 (2007), pp. 2979-2981
[267]
J.A. Sugui, H.S. Kim, K.A. Zarember, Y.C. Chang, J.I. Gallin, W.C. Nierman, et al.
Genes differentially expressed in conidia and hyphae of Aspergillus fumigatus upon exposure to human neutrophils.
[268]
J.A. Sugui, J. Pardo, Y.C. Chang, A. Mullbacher, K.A. Zarember, E.M. Galvez, et al.
Role of laeA in the regulation of alb1, gliP, conidial morphology, and virulence in Aspergillus fumigatus.
Eukaryot Cell, 6 (2007), pp. 1552-1561
[269]
J.A. Sugui, J. Pardo, Y.C. Chang, K.A. Zarember, G. Nardone, E.M. Galvez, et al.
Gliotoxin is a virulence factor of Aspergillus fumigatus: gliP deletion attenuates virulence in mice immunosuppressed with hydrocortisone.
Eukaryot Cell, 6 (2007), pp. 1562-1569
[270]
E. Szewczyk, S. Krappmann.
Conserved regulators of mating are essential for Aspergillus fumigatus cleistothecium formation.
Eukaryot Cell, 9 (2010), pp. 774-783
[271]
C.M. Tang, J. Cohen, T. Krausz, S. Van Noorden, D.W. Holden.
The alkaline protease of Aspergillus fumigatus is not a virulence determinant in two murine models of invasive pulmonary aspergillosis.
Infect Immun, 61 (1993), pp. 1650-1656
[272]
F. Tekaia, J.P. Latge.
Aspergillus fumigatus: saprophyte or pathogen?.
Curr Opin Microbiol, 8 (2005), pp. 385-392
[273]
S.P. Templeton, A.D. Buskirk, B.J. Green, D.H. Beezhold, D. Schmechel.
Murine models of airway fungal exposure and allergic sensitization.
Med Mycol, 48 (2010), pp. 217-228
[274]
N. Thau, M. Monod, B. Crestani, C. Rolland, G. Tronchin, J.P. Latge, et al.
Rodletless mutants of Aspergillus fumigatus.
Infect Immun, 62 (1994), pp. 4380-4388
[275]
H.F. Tsai, Y.C. Chang, R.G. Washburn, M.H. Wheeler, K.J. Kwon-Chung.
The developmentally regulated alb1 gene of Aspergillus fumigatus: its role in modulation of conidial morphology and virulence.
J Bacteriol, 180 (1998), pp. 3031-3038
[276]
H.F. Tsai, I. Fujii, A. Watanabe, M.H. Wheeler, Y.C. Chang, Y. Yasuoka, et al.
Pentaketide melanin biosynthesis in Aspergillus fumigatus requires chain-length shortening of a heptaketide precursor.
J Biol Chem, 276 (2001), pp. 29292-29298
[277]
H.F. Tsai, R.G. Washburn, Y.C. Chang, K.J. Kwon-Chung.
Aspergillus fumigatus arp1 modulates conidial pigmentation and complement deposition.
Mol Microbiol, 26 (1997), pp. 175-183
[278]
H.F. Tsai, M.H. Wheeler, Y.C. Chang, K.J. Kwon-Chung.
A developmentally regulated gene cluster involved in conidial pigment biosynthesis in Aspergillus fumigatus.
J Bacteriol, 181 (1999), pp. 6469-6477
[279]
D.I. Tsitsigiannis, J.W. Bok, D. Andes, K.F. Nielsen, J.C. Frisvad, N.P. Keller.
Aspergillus cyclooxygenase-like enzymes are associated with prostaglandin production and virulence.
Infect Immun, 73 (2005), pp. 4548-4559
[280]
S. Tsunawaki, L.S. Yoshida, S. Nishida, T. Kobayashi, T. Shimoyama.
Fungal metabolite gliotoxin inhibits assembly of the human respiratory burst NADPH oxidase.
Infect Immun, 72 (2004), pp. 3373-3382
[281]
K. Twumasi-Boateng, Y. Yu, D. Chen, F.N. Gravelat, W.C. Nierman, D.C. Sheppard.
Transcriptional profiling identifies a role for BrlA in the response to nitrogen depletion and for StuA in the regulation of secondary metabolite clusters in Aspergillus fumigatus.
Eukaryot Cell, 8 (2009), pp. 104-115
[282]
S.K. Upadhyay, L. Mahajan, S. Ramjee, Y. Singh, S.F. Basir, T. Madan.
Identification and characterization of a laminin-binding protein of Aspergillus fumigatus: extracellular thaumatin domain protein (AfCalAp).
J Med Microbiol, 58 (2009), pp. 714-722
[283]
V. Valiante, T. Heinekamp, R. Jain, A. Hartl, A.A. Brakhage.
The mitogen-activated protein kinase MpkA of Aspergillus fumigatus regulates cell wall signaling and oxidative stress response.
Fungal Genet Biol, 45 (2008), pp. 618-627
[284]
V. Valiante, R. Jain, T. Heinekamp, A.A. Brakhage.
The MpkA MAP kinase module regulates cell wall integrity signaling and pyomelanin formation in Aspergillus fumigatus.
Fungal Genet Biol, 46 (2009), pp. 909-918
[285]
J. Varga, B. Toth.
Genetic variability and reproductive mode of Aspergillus fumigatus.
Infect Genet Evol, 3 (2003), pp. 3-17
[286]
R. Vicentefranqueira, M.A. Moreno, F. Leal, J.A. Calera.
The zrfA and zrfB genes of Aspergillus fumigatus encode the zinc transporter proteins of a zinc uptake system induced in an acid, zinc-depleted environment.
Eukaryot Cell, 4 (2005), pp. 837-848
[287]
I. Vickers, E.P. Reeves, K.A. Kavanagh, S. Doyle.
Isolation, activity and immunological characterisation of a secreted aspartic protease, CtsD, from Aspergillus fumigatus.
Protein Expr Purif, 53 (2007), pp. 216-224
[288]
G. Vogl, I. Lesiak, D.B. Jensen, S. Perkhofer, R. Eck, C. Speth, et al.
Immune evasion by acquisition of complement inhibitors: the mould Aspergillus binds both factor H and C4b binding protein.
Mol Immunol, 45 (2008), pp. 1485-1493
[289]
J. Wagener, B. Echtenacher, M. Rohde, A. Kotz, S. Krappmann, J. Heesemann, et al.
The putative alpha-1,2-mannosyltransferase AfMnt1 of the opportunistic fungal pathogen Aspergillus fumigatus is required for cell wall stability and full virulence.
Eukaryot Cell, 7 (2008), pp. 1661-1673
[290]
L.A. Walker, C.A. Munro, I. de Bruijn, M.D. Lenardon, A. McKinnon, N.A. Gow.
Stimulation of chitin synthesis rescues Candida albicans from echinocandins.
PLoS Pathog, 4 (2008), pp. e1000040
[291]
C. Wallwey, M. Matuschek, S.M. Li.
Ergot alkaloid biosynthesis in Aspergillus fumigatus: conversion of chanoclavine-I to chanoclavine-I aldehyde catalyzed by a short-chain alcohol dehydrogenase FgaDH.
Arch Microbiol, 192 (2010), pp. 127-134
[292]
M.L. Warwas, J.N. Watson, A.J. Bennet, M.M. Moore.
Structure and role of sialic acids on the surface of Aspergillus fumigatus conidiospores.
Glycobiology, 17 (2007), pp. 401-410
[293]
R.G. Washburn, D.J. DeHart, D.E. Agwu, B.J. Bryant-Varela, N.C. Julian.
Aspergillus fumigatus complement inhibitor: production, characterization, and purification by hydrophobic interaction and thin-layer chromatography.
Infect Immun, 58 (1990), pp. 3508-3515
[294]
R.G. Washburn, C.H. Hammer, J.E. Bennett.
Inhibition of complement by culture supernatants of Aspergillus fumigatus.
J Infect Dis, 154 (1986), pp. 944-951
[295]
J.A. Wasylnka, M.M. Moore.
Aspergillus fumigatus conidia survive and germinate in acidic organelles of A549 epithelial cells.
J Cell Sci, 116 (2003), pp. 1579-1587
[296]
J.A. Wasylnka, M.M. Moore.
Uptake of Aspergillus fumigatus conidia by phagocytic and nonphagocytic cells in vitro: quantitation using strains expressing green fluorescent protein.
Infect Immun, 70 (2002), pp. 3156-3163
[297]
M.H. Wheeler, A.A. Bell.
Melanins and their importance in pathogenic fungi.
Curr Top Med Mycol, 2 (1988), pp. 338-387
[298]
S.D. Willger, N. Grahl, R.A. Cramer Jr.
Aspergillus fumigatus metabolism: clues to mechanisms of in vivo fungal growth and virulence.
Med Mycol, 47 (2009), pp. S72-S79
[299]
S.D. Willger, S. Puttikamonkul, K.H. Kim, J.B. Burritt, N. Grahl, L.J. Metzler, et al.
A sterol-regulatory element binding protein is required for cell polarity, hypoxia adaptation, azole drug resistance, and virulence in Aspergillus fumigatus.
PLoS Pathog, 4 (2008), pp. e1000200
[300]
T. Xue, C.K. Nguyen, A. Romans, G.S. May.
A mitogen-activated protein kinase that senses nitrogen regulates conidial germination and growth in Aspergillus fumigatus.
Eukaryot Cell, 3 (2004), pp. 557-560
[301]
A. Yamada, T. Kataoka, K. Nagai.
The fungal metabolite gliotoxin: immunosuppressive activity on CTL-mediated cytotoxicity.
Immunol Lett, 71 (2000), pp. 27-32
[302]
M. Yamazaki, H. Fujimoto, T. Kawasaki.
Chemistry of tremorogenic metabolites. I. Fumitremorgin A from Aspergillus fumigatus.
Chem Pharm Bull (Tokyo), 28 (1980), pp. 245-254
[303]
M. Yamazaki, E. Okuyama, Y. Maebayashi.
Isolation of some new tryptoquivaline-related metabolites from Aspergillus fumigatus.
Chem Pharm Bull (Tokyo), 27 (1979), pp. 1611-1617
[304]
K. Yokota, H. Shimada, A. Kamaguchi, O. Sakaguchi.
Studies on the toxin of Aspergillus fumigatus. VII. Purification and some properities of hemolytic toxin (asp-hemolysin) from culture filtrates and mycelia.
Microbiol Immunol, 21 (1977), pp. 11-22
[305]
S. Youngchim, R. Morris-Jones, R.J. Hay, A.J. Hamilton.
Production of melanin by Aspergillus fumigatus.
J Med Microbiol, 53 (2004), pp. 175-181
[306]
K.Y. Yuen, C.M. Chan, K.M. Chan, P.C. Woo, X.Y. Che, A.S. Leung, et al.
Characterization of AFMP1: a novel target for serodiagnosis of aspergillosis.
J Clin Microbiol, 39 (2001), pp. 3830-3837
[307]
K.A. Zarember, J.A. Sugui, Y.C. Chang, K.J. Kwon-Chung, J.I. Gallin.
Human polymorphonuclear leukocytes inhibit Aspergillus fumigatus conidial growth by lactoferrin-mediated iron depletion.
J Immunol, 178 (2007), pp. 6367-6373
[308]
L. Zhang, M. Wang, R. Li, R. Calderone.
Expression of Aspergillus fumigatus virulence-related genes detected in vitro and in vivo with competitive RT-PCR.
Mycopathologia, 160 (2005), pp. 201-206
[309]
L. Zhang, H. Zhou, H. Ouyang, Y. Li, C. Jin.
Afcwh41 is required for cell wall synthesis, conidiation, and polarity in Aspergillus fumigatus.
FEMS Microbiol Lett, 289 (2008), pp. 155-165
[310]
W. Zhao, J.C. Panepinto, J.R. Fortwendel, L. Fox, B.G. Oliver, D.S. Askew, et al.
Deletion of the regulatory subunit of protein kinase A in Aspergillus fumigatus alters morphology, sensitivity to oxidative damage, and virulence.
Infect Immun, 74 (2006), pp. 4865-4874
[311]
H. Zhou, H. Hu, L. Zhang, R. Li, H. Ouyang, J. Ming, et al.
O-Mannosyltransferase 1 in Aspergillus fumigatus (AfPmt1p) is crucial for cell wall integrity and conidium morphology, especially at an elevated temperature.
Eukaryot Cell, 6 (2007), pp. 2260-2268
Copyright © 2010. Revista Iberoamericana de Micología
Opciones de artículo
Herramientas
Quizás le interese:
10.1016/j.riam.2020.10.002
No mostrar más