Newsletter February 2019

The above photo is my rendition of what a CDC alert may look like…

“The Spectrum of Fungi That Infects Humans”

Abstract

Few among the millions of fungal species fulfill four basic conditions necessary to infect humans: high temperature tolerance, ability to invade the human host, lysis and absorption of human tissue, and resistance to the human immune system. In previously healthy individuals, invasive fungal disease is rare because animals’ sophisticated immune systems evolved in constant response to fungal challenges. In contrast, fungal diseases occur frequently in immunocompromised patients. Paradoxically, successes of modern medicine have put increasing numbers of patients at risk for invasive fungal infections. Uncontrolled HIV infection additionally makes millions vulnerable to lethal fungal diseases. A concerted scientific and social effort is needed to meet these challenges.

Fungal infections today are among the most difficult diseases to manage in humans. Some fungi cause disease in healthy people, but most fungal infections occur in individuals already experiencing serious illness, and frequently jeopardize the success of the newest medical advances in cancer care, solid organ and hematopoietic stem cell transplantation, neonatal medicine, autoimmune disease therapies, trauma and intensive care, and sophisticated surgery. In fact, these medical advances themselves often make their beneficiaries vulnerable to fungal disease. Treatment can involve breaching normal immune functions, or susceptible patients, such as extremely premature newborns who survive long enough to become infected by a fungus.

The following discussion intends to touch on highlights of the evolutionary developments by which living humans became substrates for fungi. Given the tremendous wealth of recent findings on fungal evolution, phylogenetics, genomics, development, and pathogenesis, this overview will necessarily omit much work critical to our understanding of fungi, which the other articles in this collection will focus on in detail.

Among the estimated 1.5–5.0 million fungal species on planet Earth (O’Brien et al. 2005), only several hundred cause disease in humans, and very few are able to affect healthy people. Animals coevolved with fungi, and the sophisticated and potent human immune system arose from the constant challenge posed by the microbial world. Fungal pathogens likely adapted their pathogenic repertoire to other animal prey—mammals, insects, and even unicellular amoebae—before encountering and attacking humans. But unlike plants, insects, and ectothermic vertebrates (animals whose body temperature fluctuates with their surroundings), mammals are highly resistant to invasive fungal diseases, and evolution of endothermy and homeothermy enhanced antifungal immunity (Robert and Casadevall 2009Bergman and Casadevall 2010). The remarkable resistance of mammals to fungal pathogens has been hypothesized to be responsible for emergence of mammals as the dominant land species, when proliferation of fungi at the end of the Cretaceous era created a “fungal filter” that selected for this animal group (Casadevall 20052012).

For a fungus, parasitizing a human is a demanding strategy. Four criteria must be fulfilled. (1) It must be able to grow at a high temperature, at or above 37°C. (2) It must be able to reach the tissues it will parasitize, by penetrating host tissue barriers, or by circumventing them through small airborne cells that enter air-filled spaces of lungs and sinuses directly. (3) It must be able to digest and absorb components of human tissues. (4) Finally, it must be able to withstand the human immune system.

Virulence factors can be divided into aspects of physiology that allow a fungus to fulfill these four criteria. Growth at high temperature is a stringent criterion, because land-colonizing fungi likely evolved in association with plants (Humphreys et al. 2010), whose nighttime temperature must be cool enough to minimize the ratio of carbon-expending respiration to carbon-assimilating photosynthesis. Casadevall and colleagues have discussed how the stable elevated temperature of endothermic (warm-blooded) animals may have been one of the most potent developments in antifungal immunity (Robert and Casadevall 2009Bergman and Casadevall 2010Garcia-Solache and Casadevall 2010). The energy cost of the human fever response to infection, which is tightly regulated by cytokines like TNF-α, represents another testimony to the evolutionary importance of temperature as a major host defense against fungi.

The role of fungal heat intolerance is illustrated by the different clinical scenarios of disseminated Fusarium infection versus disseminated candidiasis. Fusarium spp. are important plant pathogens that can infect neutropenic patients. As filamentous Fusarium spp. produce cells resembling yeast in the human host, which readily float within the bloodstream, a hallmark of their dissemination is development of numerous metastatic foci in the skin, presumably because the skin is the coolest organ. The development of metastatic lesions in warm organs distant from the primary focus, like spleen and kidneys, is much more unusual in fusariosis. In contrast, invasive infections with human temperature-adapted Candida spp., which also use yeast to disseminate through the bloodstream, frequently lead to innumerable metastatic foci in deep organs.

Morphogenesis is an important virulence factor related to fungal locomotion. Although fungal hyphae can pierce tissue barriers owing to the turgor pressure at their tips, yeast can readily disseminate to distant sites in a large animal. Fungi that infect healthy humans do so almost exclusively in their yeast form. But for most fungi, the ability to assume various shapes is critical for infecting humans, because many enter the body in the form of small, round airborne dispersal propagules, sporangiospores or conidia, which are produced from hyphal cells. In the oceans, fungi originated as unicellular, oval cells propelled by flagella, so growth as a round or oval cell was a fundamental fungal trait. Lucking et al. (2009), in their comparison of fossil and molecular dating systems of fungal evolution, state that “all dating estimates show that the evolution of filamentous fungi occurred much later than the origin of the fungal lineage itself, suggesting that for a long time after their origin, fungi were heterotrophic, unicellular, flagellate, aquatic organisms.” An option for the hyphal form, whose adaptive utility is shown by convergent evolution of hyphae in fungi and oomycete water molds (Money et al. 2004), may have evolved when fungi colonized land as plant symbionts (Redecker et al. 2000Heckman et al. 2001Lucking et al. 2009). Morphogenesis also contributes to protection against amoeboid cell predation. Filamentous forms of Cryptococcus neoformansare resistant to amoeba (Neilson et al. 1978), and incubation of Histoplasma capsulatum and Blastomyces dermatitidis yeast with amoeba at 37°C triggers transition to filamentous growth (Steenbergen et al. 2004).

Tremendous variability of cell forms and the ability to switch between them continues in extant fungal phyla, and few lineages have completely lost a round or a filamentous cell form. A highly successful human-host-associated fungus, Candida albicans, is notable for the facility and frequency with which it switches between a broad spectrum of growth forms in the host, between round yeast, elongated pseudohyphae, and filamentous hyphae (Sudbery et al. 2004). Adhesion molecules are virulence factors related to fungal locomotion strategies because a hypha must anchor itself to host tissue to exert tissue-penetrating pressure at its tip, and because once a fungal cell has reached a favorable location, it needs to be able to stay at the site.

Secretion of digestive enzymes appropriate to dissolve host tissue is the first step for a fungus to use the host as a nutritious substrate. The next step is to absorb the small molecules released by digestion of host tissues; fungal cells have transporters of nitrogen and carbon sources as well as metal ions and other micronutrients. Iron acquisition plays a special role because of its importance in critical enzymatic processes including respiration, and because the human host actively sequesters iron during infection to withhold it from the pathogen (Boelaert et al. 1993Ibrahim et al. 2006Kronstad et al. 2013Noble 2013). The importance of iron sequestration in the host to defend against fungal growth was dramatically illustrated by disseminated Mucorales infections in patients receiving the iron chelator, deferoxamine (Windus et al. 1987).

Most fungi, even with these capabilities, cannot withstand the phagocytes unleashed against them by an immunocompetent human, which have help from opsonins such as complement and specific antibody, and from activating T lymphocytes. Fungi that infect healthy humans devote a large portion of their physiology to withstanding or evading the immune system. To this end they use an amazingly diverse array of strategies, which sometimes differ even between lineages of a single species. In contrast, many species that grow at 37°C can readily infect a severely immunocompromised patient.

Last, coordinating the processes to fulfill these four criteria of pathogenicity demands a complex network of sensing and signaling systems, which inform the fungal cell of external conditions and set in motion its appropriate responses. Loss of virulence was shown many times in many fungal pathogens, when just a single element of a sensing and signaling network was genetically or pharmacologically disturbed, such as mechanosensor, calcineurin, protein kinase A, MAP kinase, Tor, and high osmolarity signaling pathways (Rohde and Cardenas 2004Cramer et al. 2008Kumamoto 2008Bastidas and Heitman 2009Argimon et al. 2011Ernst and Pla 2011Hogan and Muhlschlegel 2011Shapiro et al. 2012Chen et al. 2013).Go to:

FUNGI THAT INFECT HEALTHY HUMANS

The capability to infect an immunocompetent human has arisen independently multiple times among three major fungal phyla: the Entomophthoromycota (Kwon-Chung 2012), the Ascomycota, and the Basidiomycota.Go to:

Entomophthoromycota

Entomophthoromycota contains effective pathogens of insects. They occur worldwide, but have been found as agents of invasive human infection only in subtropical and tropical regions. The species of genera pathogenic for humans, Basidiobolus and Conidiobolus, can be isolated from plant debris and soil especially during rainy months (Bittencourt 1988). As emphasized recently by Kwon-Chung (2012), these genera are evolutionarily distant from each other as well as from the Mucorales, with whom they traditionally were classified in Zygomycota.

Conidiobolus spp. cause submucosal disease of the nose, sinuses, and central face. In most cases, gradual progression causes swelling of submucosal and subcutaneous tissue, disfigurement, breathing difficulties, and chronic bacterial sinusitis caused by the blockage of ostia. Unlike the Mucorales causing sinus disease, Conidiobolus does not usually invade blood vessels nor penetrate into the central nervous system (CNS) (Prabhu and Patel 2004).

Two species, Conidiobolus coronatus and Conidiobolus incongruus, have been isolated from central facial disease. Presumably, the route of infection is inhalation of large ballistospores forcibly launched from the sporangiophores on which they are produced singly to distances up to 30 cm (Isa-Isa et al. 2012). Owing to their large size of 30–38 µm (Isa-Isa et al. 2012), spores presumably land on the mucosa of nasal air passages and fail to reach the distal airways (Bittencourt et al. 2006).

Conidiobolus spp. infect arthropods including mites, spiders, and insects (Isa-Isa et al. 2012). They elaborate lytic enzymes including elastases, collagenases, and lipases, likely to parasitize and kill arthropods (Comerio et al. 2008). These enzymes enable Conidiobolus to digest human tissue, growing by extension from nasal submucosa through the facial soft and bony tissue (Gugnani 1992). Metabolites toxic to insects have also been identified but because the fungus is highly lethal to arthropods and only of moderate virulence in mammals, it remains to be seen whether these metabolites play any role in human infection (Wieloch et al. 2011).

The host sends neutrophils, eosinophils, and histiocytes to contain the fungus, and granulomas (structured assemblies of histiocytes) develop. Histopathology can show dense eosinophilic material surrounding the hyphae, the Splendore-Hoeppli phenomenon, which has been suggested to consist of antigen–antibody precipitate (Isa-Isa et al. 2012).

Basidiobolus ranarum causes subcutaneous disease primarily in children of tropical and subtropical Africa, Asia, and the Americas. Nodules appear at sites of inoculation through insect bites, scratches, or small wounds (Ribes et al. 2000). In addition, Basidiobolus has caused gastrointestinal infections, presumably after ingestion of large inocula, and symptoms resemble inflammatory bowel disease (Zavasky et al. 1999Vikram et al. 2012). When insects infected by Basidiobolus are eaten by amphibians and reptiles, the fungus is shed in the feces (Ribes et al. 2000) and attaches to plant matter with which it is accidentally inoculated under the skin, for instance, by thorns, leaves used for cleansing, etc. As an insect pathogen, it also elaborates proteases and lipases (Echetebu and Ononogbu 1982Okafor et al. 1987Okafor and Gugnani 1990Okafor 1994). Its inability to invade deep organs is likely related to limited thermotolerance; 37°C may be its maximal growth temperature (Ribes et al. 2000).

Ascomycota

Pathogenic Onygenales

Several soil-inhabiting members of the ascomycete order Onygenales have evolved to parasitize mammals and cause systemic infection. They have been classified in the family Ajellomycetaceae (Untereiner et al. 2004Bagagli et al. 2008), which include B. dermatitidis (teleomorph, Ajellomyces dermatitidis), H. capsulatum (teleomorph, Ajellomyces capsulatus), Paracoccidioides brasiliensis, Paracoccidioides lutzii, Lacazia loboi, Coccidioides immitisCoccidioides posadasii, and related fungi with similar properties. The intracellular pathogenic strategy of some of these organisms is similar in protozoa and macrophages (Steenbergen et al. 20012003), raising the possibility that their capacity for virulence arose accidentally as a result of environmental pressures (Casadevall and Pirofski 2007).

Disease caused by these fungi begins asymptomatically in the lungs and progresses to an influenza-like illness or frank pneumonia. They can disseminate to distant sites, can persist and reactivate, with different organ predilections among different genera: Paracoccidioides in oral and respiratory mucous membranes (Queiroz-Telles and Escuissato 2011Marques 2012), Blastomyces in bones, joints, and skin (Bradsher et al. 2003), and Histoplasma in multiple organs including the gastrointestinal tract and adrenals, as well as bones and skin (Kauffman 2007).

Their primary lifestyle is saprobic. Most genera occur in defined geographic areas, which follow particular soil consistencies, for instance, dry and alkaline for the Coccidioides spp. and acidic for Histoplasma. It has not been ruled out that their geographic distribution follows that of specific host mammals. In the soil, they grow as a branched network of hyphal filaments, a mycelium, and saprobically use organic matter. They produce airborne dispersal cells: Conidia produced from specialized hyphae, as Histoplasma and Blastomyces do, or arthrospores from regulated fragmentation of hyphal compartments, as Coccidioidesand Paracoccidioides do. Paracoccidioides produces both arthroconidia and aleuriconidia (conidia directly produced along the hyphae). Once conidia or arthrospores are taken up by a mammal, usually by inhalation, they convert into parasitic yeast (or spherules, in the case of Coccidioides spp.) and initiate an infection. The principal signal for their conversion to the yeast form is the elevated mammalian body temperature, so that this switch can be induced in vitro when they are cultured at 37°C or above. For this reason they are grouped as thermal dimorphs. These fungi infect, persist, and cause disease in healthy hosts. In immunocompromised hosts, the infections are more clinically apparent, more severe, and more likely to disseminate to multiple organs.

Adaptive cellular immunity is required to control the infection with these fungi and can be detected by skin tests with purified antigens. For this reason, clinically apparent infections with some members of this group, such as H. capsulatum, became much more common with the rising prevalence of AIDS. These Onygenales (Ajellomycetaceae) are the only phylogenetically related group whose members all cause systemic disease in otherwise healthy people.

Other thermally dimorphic fungi are Sporothrix schenckii (order Ophiostomatales), which is primarily introduced into the host through skin injuries, and Talaromyces (Penicillium) marneffei (order Eurotiales), which does not infect healthy hosts but because of the AIDS epidemic has become a prevalent and important invasive pathogen in Southeast Asia. These fungi coevolved with higher vertebrates, as it is the adaptive immune system by which they are controlled. The fact that these evolutionarily distant fungi convert from saprobic hyphal to pathogenic yeast growth at the temperatures of endothermic animals suggests that an ability to access large nitrogen and other nutrient stores of an animal host is an advantageous lifestyle option for a soil dweller, and a result of convergent evolution in several phyla.

H. capsulatum will be discussed at greater length because it is well studied and also because it exemplifies important principles for other thermally dimorphic Onygenales. This fungus inhabits soil and other organic matter such as decaying wood enriched with bat and bird droppings. Its hyphae produce macro- and microconidia, and the latter are small enough, <5 µm, to penetrate into the alveoli of mammalian lungs. There they are phagocytized by alveolar macrophages and converted to yeast that is able to proliferate in the hostile environment of the phagolysosome.

Aspects of histoplasmosis resemble infections with a well-adapted bacterial intracellular pathogen, Mycobacterium tuberculosis. Depending on inoculum size, infection is most often inapparent or leads to a self-limited influenza-like illness. The pathogen can persist indefinitely until immune decline allows it to reactivate. Lung disease resembling TB can ensue immediately after infection, and dissemination to all organs occurs depending on host immune status and age. The response of an immunocompetent host is granuloma formation.

Histoplasma occurs on all continents, although it is rare in Eurasia and most common in North and South America. Two studies of H. capsulatum showed that its growth and clinical phenotypes, which had traditionally been the basis for its division into varieties (capsulatumduboisii, and farciminosum), did not align with the divisions of its seven phylogenetic clades (Kasuga et al. 19992003), and that the different clinical disease states attributed to these (now obsolete) varieties were caused by the same clades.

Phylogenetic analyses suggest that H. capsulatum arose in South America 3.2–13.0 million years ago and its present distribution reflects a period of rapid spread to all continents except Eurasia (where it was later introduced), followed by restriction to the warmest areas during a period of intense cold 1.8 million years ago (Kasuga et al. 19992003). From there, it spread to temperate regions as the earth warmed again. This idea is consistent with its low genetic diversity in temperate regions and high diversity in tropical areas (Kasuga et al. 1999), mirroring the diversity of plant species in areas of temperate and tropical forest, which are its geographic areas of distribution.

The two North American clades are not closely related and are thought to have been genetically isolated for the past ∼20 million years (Kasuga et al. 1999), despite overlapping territories. These two clades differ in virulence, with North American clade 1 having been isolated predominantly from AIDS patients, and clade 2 infecting immunocompetent hosts (Kasuga et al. 1999). A clade 2 isolate and a representative of the Panama clade have been used in genetic studies of virulence factors, and surprising differences were found, as discussed below.

The habitat of Histoplasma is soil enriched with bird and bat droppings, in specific geographic locations including the Ohio and Mississippi valleys of North America and moist regions of Central and South America. Whereas birds are very rarely infected with H. capsulatum (Quist et al. 2011), bats are frequently infected but respond with minimal inflammation (Taylor et al. 1999), suggesting active repression of the host immune response by the fungus. It was suggested that, “in at least some bat species natural exposure may result in a chronic, controlled infection, which allows the bat to excrete viable fungi over a long period” (Hoff and Bigler 1981). Experimentally infected bats also had a minimal inflammatory response (McMurray and Greer 1979).

A broad variety of other infected mammals such as cats (Aulakh et al. 2012), dogs (Brömel and Sykes 2005), sea otters (Morita et al. 2001), and badgers (Bauder et al. 2000) do respond with granulomatous inflammation to the presence of Histoplasma yeast. Perhaps H. capsulatum has evolved to infect bats, in which it avoids activation of host responses, by mechanisms only partially adapted to other immunocompetent mammals. If this is the case, the fungus has established an efficient amplification cycle by which its mycelial form grows in droppings under bat roosting places, for instance, in caves, and its microconidia are released into the air to be inhaled by the bats that fertilize its soil substrate (Taylor et al. 1999).

Several mechanisms by which H. capsulatum yeast successfully interacts with mammalian hosts have been identified. Surprisingly, different clades use different strategies to achieve an intracellular lifestyle in macrophages, and to suppress macrophage activation. For example, macrophages recognize the important pathogen associated molecular pattern (PAMP) β(1,3)-d-glucan with the receptor dectin-1 (Rappleye et al. 2007), and ligand binding triggers responses that enhance phagocyte activation responses (Underhill et al. 2005). β(1,3)-d-glucan is the main structural polymer of ascomycete cell walls, so Histoplasma cannot forgo its use to avoid activating their hosts’ macrophages. Instead, Histoplasma yeast of most clades hide it by coating their cell surface with α(1,3)-d-glucan. Perturbation of genes whose products are required for α(1,3)-d-glucan synthesis renders cells of the Panama clade avirulent (Rappleye et al. 2004). Notably, this α(1,3)-d-glucan coat is lacking in a North American clade 2 strain (Edwards et al. 2011), and an α(1,3)-d-glucan synthase mutant in this strain background is fully virulent (Edwards et al. 2011).

Another cell surface protein, Yps3, is expressed to a significant level only in the more virulent North American clade 2 (Bohse and Woods 2007). This protein, a homolog of B. dermatitidis BAD1, contributes to dissemination of H. capsulatum to extrapulmonary foci (Holbrook and Rappleye 2008). Its gene is present in most strains but substantial transcription is known to occur only in North American clade 2, the clade lacking α(1,3)-d-glucan. Yps3, like BAD1, may directly suppress TNF-α production, circumventing the need for this clade to use α(1,3)-d-glucan to hide the stimulatory PAMP β(1,3)-d-glucan (Holbrook and Rappleye 2008). The known variations in their pathogenic repertoires among clades of Histoplasma are based in differences of regulatory rather than coding regions between the sequenced strains (Edwards et al. 2013).

H. capsulatum yeast have evolved a repertoire of manipulative molecules to turn the host compartment specifically evolved to kill pathogens, the phagolysosome, into a habitat in which they thrive (Sebghati et al. 2000Youseff et al. 2009). Remarkably, within the teleomorph genus Ajellomyces, originally comprising the anamorph sister species H. capsulatum and B. dermatitidis, one species H. capsulatumchose this route of intracellular life, whereas the other, B. dermatitidis, is a successful extracellular pathogen.

Distinct H. capsulatum clades differ in specific details of the pathogenic program of their yeast, but they have in common that their suite of virulence factors is turned on simultaneously with the switch from hyphal to yeast growth. Mammalian body temperature of 37°C or above is the main signal to trigger conversion from the mycelium to yeast. This conversion entails differential transcription of ∼20% of the genome (Nguyen and Sil 2008), and is controlled by a network of transcriptional regulators apparently responding to a histidine kinase signal (Nemecek et al. 2006Webster and Sil 2008Cain et al. 2012Beyhan et al. 2013). Mutants in these regulators are incapable of the transition to yeast growth, and are avirulent (Nemecek et al. 2006Webster and Sil 2008Cain et al. 2012Beyhan et al. 2013).

Other members of the Ajellomycetaceae, all of which associate with mammals, have equally distinctive ecology and pathobiology. Mycelia of the Coccidioides spp., growing near rodent burrows in arid regions of the Americas (Nguyen et al. 2013), produce arthroconidia. When arthroconidia are inhaled by a mammal, they give rise to round cells—spherules—in which growth and mitosis occur to eventually fill the enlarging mother cell with hundreds of endospores. On maturity, the spherule ruptures and releases the endospores into host tissue. Newly released endospores repeat the growth cycle locally or disseminate hematogenously. Phylogenetic analysis shows that the fungus evolved in North America before that continent’s geologic connection with South America 2.5–3.5 million years ago (Fisher et al. 2001), and that the genetically homogeneous South American strains, derived from populations in present-day Texas, reached their current distribution 8940–134,000 years ago, possibly with the migration of humans from North into South America (Fisher et al. 2001). Another unique member of the Ajellomycetaceae, L. loboi, shares an important feature with a distant member of its phylum Ascomycota, Pneumocystis jirovecii. They are the only human fungal pathogens not grown in culture (Vilela et al. 2009). Lacazia, a sister genus to Paracoccidioides, has been found only in subcutaneous infections of previously healthy humans in the Amazon basin and in dolphins of the Atlantic ocean and of the Amazon (Vilela et al. 2009Theodoro et al. 2012), and (analogous to Mycobacterium leprae) can be amplified only in a mouse model. In contrast, Paracoccidioides spp., prevalent fungal pathogens that cause pneumonia, systemic disease of the monocyte–macrophage system, and destructive lesions of skin and oral mucous membranes (Fortes et al. 2011Queiroz-Telles and Escuissato 2011Marques 20122013), occur in Central and South America mainly outside the Amazon region (Theodoro et al. 2012). Paracoccidioides causes clinical disease in many more men than women, in a ratio of 13:1, possibly because estrogen blocks the conversion of inhaled arthroconidia to the tissue-invasive yeast form (Shankar et al. 2011). This fungus frequently infects armadillos, and its virulence factors such as the immunodominant adhesin glycoprotein 43 (Puccia et al. 199920082011Fortes et al. 2011) may have evolved in its coevolution with these ancient mammals (Bagagli et al. 2006Richini-Pereira et al. 2009).

Some members of the Onygenales infect healthy humans, but very rarely cause invasive disease. Dermatophytes of the genus Arthrodermataceae specialize in degrading keratins, important structural proteins in vertebrate skin whose extensive cross-linking by disulfide bonds makes them proteinase resistant (Gradisar et al. 2005). With the evolution of animals with substantial keratin appendages such as scales, feathers, nails, and hair, specializing in use of these nutrient sources must have been a worthwhile niche for the presumed ancestor of the dermatophytes (Weitzman and Summerbell 1995).

Reptilian amniotes had evolved keratin, so that the basis for this specialization was available as a fungal substrate 300 million years ago (Eckhart et al. 2008). Recent analysis of mitochondrial genomes of six dermatophytes comprising the three morphologically defined anamorphic genera, TrichophytonMicrosporum, and Epidermophyton, together with 29 other members of Ascomycota, confirmed that the dermatophytes are descended from a common ancestor (are monophyletic) and separated from other fungi only 32–50 million years ago (Wu et al. 2009). This study also found that classification of species by molecular characters frequently contradicted the traditional classifications based on morphologic traits (Wu et al. 2009), emphasizing the fact that fungal morphology is enormously flexible and convergent evolution is common, so that only limited predictions of their physiology can be made from the appearance of fungal structures. In contrast, molecular phylogenetic studies have confirmed the grouping of dermatophytes by their ecologic and clinical features, including the distinct sites of the human body they tend to infect (Cafarchia et al. 2013).

Anthropophilic dermatophytes travel over the world with their human hosts. Although many species are endemic to specific geographic regions, some species occur worldwide. Their prevalence varies with the lifestyle and socioeconomic conditions of their human hosts, and is undergoing continuous epidemiologic changes (Ameen 2010). Unlike their thermal dimorphic Onygenales relatives, they can reach a new host by person-to-person transmission, and their access to new human substrate is made even easier by the ability of their arthroconidia to persist for years in fomites (Weitzman and Summerbell 1995). Anthropophilic dermatophytes, relying on person-to-person access to new hosts, can forgo long-distance travel via airborne conidia, which their geophilic relatives produce in larger profusion to reach distant deposits of animal keratin (Weitzman and Summerbell 1995).

Coevolution with their animal hosts presumably allowed dermatophytes to evade or resist the evolutionarily more ancient innate immune mechanisms, so that today their control requires adaptive cellular immunity (Dahl 1993). By restricting their usual habitat to the most superficial keratinized layer of the skin and its appendages, dermatophytes reduce their contact with immune cells. Human-specific dermatophytes (anthropophiles) are able to down-regulate host inflammation to establish a chronic infection (Blake et al. 1991Shiraki et al. 2006). In contrast, soil-dwelling (geophilic) and animal-specific (zoophilic) dermatophytes are unable to manipulate host immunity and are eliminated by a vigorous inflammatory response, illustrating the principle that a well-adapted parasite carefully calibrates its virulence.

Basidiomycota

Cryptococci

Basidiomycete yeast with a worldwide distribution, cryptococci, in the past century, infected humans only rarely (Molez 1998). In the 1950s, increasing numbers of cryptococcal meningoencephalitis were reported from central Africa, viewed in retrospect as sentinels of the emergence of AIDS around the Congo River (Molez 1998). AIDS is still the setting in which the vast majority of cryptococcosis occurs (Mitchell and Perfect 1995Pukkila-Worley and Mylonakis 2008); for 2006, 957,900 cases of cryptococcal meningitis associated with AIDS were estimated, resulting in 624,700 deaths (Park et al. 2009). Furthermore, recent outbreaks of Cryptococcus gattii infections with significant mortality on Vancouver Island and the northwestern United States raise the concern that the fungus may be evolving to become virulent for healthy humans (Fraser et al. 2005Byrnes et al. 2010Springer et al. 2012).

The pathogenic cryptococci, C. neoformans and its sister species C. gattii, enter a human by inhalation of infectious cells: dried yeast or possibly basidiospores, the products of meiosis after mating. When these small airborne cells, <5 µm in size, are inhaled, pneumonia can ensue in susceptible hosts, and on gaining access to the bloodstream, the yeast can disseminate to all organs. Cryptococcus has a special predilection for the CNS and causes subacute meningoencephalitis, in which high intracranial pressure plays an especially deleterious role, which, if left untreated, is lethal. The fungus can persist for years in the lung or in sites of previous dissemination and reactivate only on weakened immune surveillance.

C. gattii causes invasive disease and death in wild and domestic land and ocean mammals and birds and pneumonia and meningoencephalitis in previously healthy people (Krockenberger et al. 2002Miller et al. 2002Raso et al. 2004Santos et al. 2008McGill et al. 2009). A wealth of analyses of pathogenic cryptococci over the past several decades have shown that C. gattii in many settings is a more common primary pathogen than C. neoformans (Speed and Dunt 1995Springer et al. 2012), but new reports continue to blur the boundaries between primary and opportunistic pathogens among the Cryptococcusspecies (Chen et al. 2008Chau et al. 2010Choi et al. 2010), illustrating the limited utility of these terms (Casadevall and Pirofski 2001). A recent report has indicated that the presence of anti-GM-CSF autoantibody is a risk factor for C. gattii CNS infection in otherwise healthy individuals (Saijo et al. 2014). It suggests that patients with cryptococcal CNS infection considered “immunocompetent” may carry immune defects that can yet not be identified by routine immunological screening. An important condition for a clearer understanding of ecology, epidemiology, and evolution of cryptococci is that more resources for microbiologic analysis become available in poor countries. Cultures of blood and cerebrospinal fluid currently cannot be performed in medical settings in which most patients with cryptococcosis will seek help.

The separation of the sister species C. gattii and C. neoformans is thought to have occurred 45 million years ago (Simwami et al. 2011). Molecular typing recapitulates traditional serologic classification of varieties into C. neoformans var. grubii (serotype A, VNI, VNII, VNB) and var. neoformans (serotype D, VNIV) and their hybrids (serotype AD, VNIII), as well as C. gattii (serotypes B and C) (Mitchell and Perfect 1995Meyer et al. 2003). C. gattii has been subdivided into molecular varieties or cryptic species VGI through VGIV, among which further subdivisions correlate with geographic location and virulence (Kidd et al. 2004Fraser et al. 2005Byrnes et al. 2010).

In the environment, C. gattii has been isolated from numerous tree species in tropical and more recently in temperate regions, particularly from sites of wood decay and insect consumption of vegetable matter (Ellis and Pfeiffer 1990Fortes et al. 2001Kidd et al. 2003); although it was first found on Eucalyptus trees and believed to follow their worldwide distribution (Ellis and Pfeiffer 1990), its isolation from the midst of pristine Amazon rainforest revises the interpretation of its origin (Fortes et al. 2001). Environmental sampling on Vancouver Island to define the reservoirs of the outbreak that began in 1999 found more than 10 species of trees to yield C. gattii, which comprised ∼10% of the trees sampled (Kidd et al. 2007), and it survives for at least 1 yr in fresh- and seawater, suggesting it could spread with ocean currents (Kidd et al. 2007).

Supporting the idea of an origin of pathogenic C. gattii in South America is the uniquely symmetric distribution of mating types in haploid isolates from that continent. Although clinical and environmental isolates of the α mating type vastly predominate in other parts of the world, and isolates of the a type are almost never encountered, the South American distribution is 0.8a–1.0α (Hagen et al. 2013).

C. neoformans var. grubii, which is responsible for ∼95% of cryptococcal infections worldwide and 98% of infections among AIDS patients (Simwami et al. 2011), is genotypically most diverse in southern Africa; and a collection of strains from Botswana contained 12% isolates of the a mating type, which in other sites is extremely rare (Litvintseva and Mitchell 2012). In that study, distinct genotypes were thought to have diverged 5000 years ago; the investigators speculate that pigeons contributed to the worldwide spread of one specific strain of C. neoformans var. grubii (Litvintseva and Mitchell 2012) because C. neoformans is enriched in bird droppings and has been shown to mate on pigeon droppings (Nielsen et al. 2007). A study from Thailand supports the concept of an origin of C. neoformans var. grubii in Africa (Simwami et al. 2011).

Like C. neoformans, with which it shares virulence traits, and unlike nonpathogenic cryptococcal species (Findley et al. 2009Araujo Gde et al. 2012), C. gattii possesses a thick polysaccharide capsule composed of glucurono- and galactoxylomannan, which, analogous to encapsulated bacteria like Streptococcus pneumoniae, blocks phagocytosis of the organism unless it is opsonized (Casadevall et al. 1998). Presumably, the dense capsule also provides some protection against hydrolytic enzymes exocytosed by neutrophils. Capsular glucuronoxylomannan and other macromolecules, as well as virulence-associated proteins, are carried through the cell wall in an amazing process involving vesicles that originated from the late endosome (multivesicular body) (Rodrigues et al. 20072008). This process, first discovered in Cryptococcus, subsequently was found in other pathogenic fungi like Paracoccidioides (Vallejo et al. 20112012), and in the model yeast Saccharomyces cerevisiae (Oliveira et al. 2010). Like other fungi, cryptococci can diversify morphologically in the host, producing a population of Titan cells, giant cells of up to 50 µm in diameter (Okagaki et al. 2010Zaragoza et al. 2010) created by DNA replication and growth not followed by mitosis (Okagaki et al. 2010). Other antiphagocytic mechanisms, independent of the capsule, are controlled by two GATA transcription factors and are likely to involve coordinated regulation of numerous physiologic processes because hundreds of genes are differentially expressed in their mutants (Liu et al. 2008Chun et al. 2011).

Once ingested by alveolar macrophages not activated by T lymphocytes, the fungus can survive and proliferate, and it is thought that some cryptococci may enter the brain within macrophages crossing the blood–brain barrier (a Trojan horse-like mechanism), whereas free yeast in the bloodstream cross the blood–brain barrier by transcytosis (Casadevall 2010Shi et al. 2010). To survive in phagocytes, it buffers oxidative and nitrosative stress with melanin deposited in its cell wall (Wang et al. 1995), and with neutralizing enzymes like superoxide dismutase, glutathione reductase, and thioredoxins (Kronstad et al. 2011). Both in macrophages and in the free-living amoeba Acanthamoeba castellanii, it disrupts phagolysosome membranes to prevent acidification of this compartment and to dilute the lytic enzymes it contains; it also sheds large amounts of capsular polysaccharide to fill the phagocytic cells’ cytoplasm (Steenbergen et al. 2001Tucker and Casadevall 2002). Virulent strains not only resist killing but proliferate in the amoebae (Steenbergen et al. 2001) so that the predator finds itself prey.

How did some cryptococci evolve from survivors of amoeba attacks to occasional parasites of humans? A recent study examined evolutionary relationships and virulence traits of C. gattii and C. neoformansstrains, and related fungi belonging to the order Tremellales, which do not infect mammals (Findley et al. 2009). Other Tremellales parasitize fungi by attaching to the host fungus with a specialized hypha and accessing its cytoplasm through a newly created pore (Zugmaier et al. 1994Millanes et al. 2014). An evolutionary trajectory from parasitizing another live fungus to parasitizing an animal host may have proceeded through association with and parasitism of insects, and through selection in soils by amoebae for traits that can also function in mammalian virulence. The human-pathogenic Cryptococcus species killed larvae of the wax moth Galleria mellonella in one study, whereas their Tremellales relatives partially or completely lacked virulence in this insect model (Findley et al. 2009). Of note, insect cellular immune defenses consist of amoeboid phagocytes (Williams 2007Browne et al. 2013), so that resistance to free-living amoebae may transfer well to resistance to insect plasmatocytes. However, in nature, cryptococci so far have only been found in association with insect frass (the excreta of plant-eating insects), not as insect parasites (unlike, e.g., the entomophthorales and the ascomycetous Cordyceps spp.). Further ecologic research will have to show whether insect parasitic cryptococcal relatives can be identified, perhaps in their ancestral tropical forest habitats. The role of its versatile mating system in virulence evolution of Cryptococcus will be discussed in the dedicated articles in this collection.Go to:

FUNGI THAT INFECT IMMUNOCOMPROMISED HUMANS

A sufficiently immunocompromised host can be infected by hundreds of environmental fungal species that grow at human core temperatures. However, a predictable set of actors is known to cause the most common invasive infections in immunocompromised individuals, and they will be discussed next according to their phylogenetic affiliations, which predict important parameters of their physiology.

Ascomycota

Candida

Candida species are a polyphyletic group of the order Saccharomycotina, which live as commensals on mammalian mucous membranes, particularly of the gastrointestinal tract (Wrobel et al. 2008). They have rarely been found in the soil (Marples and Di Menna 1952Skinner and Fletcher 1960). A limited number of species are commonly associated with humans as colonizers and opportunistic pathogens: C. albicansCandida glabrataCandida parapsilosisCandida tropicalisCandida lusitaniae, and Candida krusei. Of these, C. albicans is the most commonly isolated human commensal and pathogen (Odds 1988Kam and Xu 2002Krcmery and Barnes 2002). The frequency of colonization with nonalbicans Candida species shifts according to age of the host, with C. parapsilosis being more prevalent among children and C. glabrata among older adults (Soll 2002).

Invasive candidiasis takes many forms, depending on the setting in which a host became susceptible to this opportunist, and depletion of normally competing bacterial flora by antibiotics often plays a role. For example, in cancer patients receiving chemotherapy, candidiasis is often caused by fungal transmigration through disrupted intestinal epithelium, and the organs with the heaviest fungal burden are those connected by the portal circulation—liver and spleen—but not the brain. In contrast, premature newborns with candidiasis often have abscesses in the brain, because their blood–brain barrier may be immature and because their brain receives such a large fraction of cardiac blood output.

C. albicans is the human companion fungus par excellence. Many healthy humans carry C. albicans, which harmlessly colonizes mucous membranes to high numbers at different anatomic sites of a single individual (Odds 19841988Soll et al. 1991). It has evolved to flourish in a wide range of environmental conditions: high pH in the intestine versus low pH in the vagina, feast and famine nutritional conditions in the gastrointestinal tract according to its host’s mealtimes versus steady glycogen supply in the vagina, and aerobic conditions on oral surfaces versus anaerobic conditions in the intestine. Unlike the dermatophytes, in which different clades inhabit different anatomic sites, C. albicans biotypes are generalists and each can adapt to all sites (Odds 1984).

Not only can C. albicans survive and thrive in highly disparate microenvironments of the human host, but as part of the normal flora it has also honed its ability to avoid triggering human immune defenses, for instance, by covering its main structural cell wall component, β(1,3)-d-glucan, with glycoproteins to avoid engaging the macrophage dectin-1 receptor (Wheeler and Fink 2006).

Analysis of its populations, which comprise at least five clades, suggests that C. albicans migrated throughout the world with its human hosts (Lott et al. 2005). It is unknown whether the fungus accompanied the mammalian lineages that led to primates throughout evolution, or whether it made the jump to mammalian commensal at a more recent time before emergence of modern humans (Lott et al. 2005).

Perhaps in its long cohabitation with the human, which as to host numbers proved to be a winning strategy, C. albicans became so closely hostadapted that major genomic rearrangements, as facilitated by sex and meiotic recombination, became less advantageous (Goddard et al. 2005). In a stable and benign environment, such as the equilibrium between thriving hosts and commensals, the expenditure of time and energy required for sexual reproduction may cost more than it benefits. C. albicans appears to consist of stable clonal diploid populations (Bougnoux et al. 2008), and its mating and parasexual cycle seem to be such rare events that their discoveries were sensational paradigm shifts in the community of Candidaresearchers (Hull and Johnson 1999Magee and Magee 2000Miller and Johnson 2002Alby et al. 2009Hickman et al. 2013). Although the parasexual cycle occurs infrequently, it can generate progeny with extensive genetic diversity by shuffled combinations of eight chromosomes or by recombination between homologous chromosomes mediated by the conserved Spo11 protein integral to meiotic recombination (Forche et al. 2008). More discoveries are sure to follow on the role of mating and various ploidy states in Candida ecology today, and virulence associated with the mating loci (Lockhart et al. 2005Wu et al. 2007) may become more important as Candida habitat changes with current changes in human epidemiology.

In the millions of years of coevolution, human predecessors and humans survived only briefly with diseases during which C. albicans could significantly invade the host. Invasive candidiasis was very rare (Browne 1954Zimmerman 1955). C. albicans is an efficient invasive pathogen, causing mucous membrane infections in individuals with ineffective adaptive cellular immunity and fatal disseminated infections in patients lacking functional innate immune cells, neutrophils. For a microorganism that relies on person-to-person transmission and does not have a significant soil reservoir (or mode of locomotion back from the soil to its primary host), the benefit of multiplying to the point of host death seems mysterious. One idea might be that the Candida pathogenic repertoire evolved to take advantage of a constant large number of temporarily susceptible humans, infants, who develop oral candidiasis because of immature adaptive immunity. Human infants’ propensity to widely distribute their oral secretions (to slobber) may allow infecting Candida to launch its increased numbers from infected mucous membranes of these hosts and to colonize more of their contacts. If this is the case, Candida virulence factors evolved to efficiently infect mucous membranes, and invasive disease is for it an accidental dead end.

The pathogenic repertoire of Candida comprises all features needed for a human fungal pathogen. It is able to grow well at human febrile temperatures of 39°C–40°C. It can penetrate host tissues with hyphal cells and it has multiple adhesion molecules to facilitate the drilling action of the hyphal tip (Staab et al. 1999Sundstrom 2002Hoyer et al. 2008Liu and Filler 2011). Its hyphae constitutively produce yeast, constantly diversifying the population with more mobile cells (Shen et al. 2008). An array of lytic enzymes suitable for digesting human macromolecules is induced during tissue invasion (Ghannoum 2000Staib et al. 2000Calderone and Fonzi 2001Schaller et al. 2005Albrecht et al. 2006Trofa et al. 2011), and transporters are regulated coordinately to absorb the released monomers into the fungal cell, as reviewed by (Morschhäuser 2011), and to mobilize micronutrients from the host (Weissman et al. 2008Citiulo et al. 2012Noble 2013). Unbiased genetic screening yielded novel virulence factors like glucosylceramide synthesis (Noble et al. 2010) and is expected to reveal new aspects of Candida invasive lifestyle (Holland and Summers 2008).

Resistance of Candida to the human immune system is limited. Features increasing its virulence were likely selected against in its long coevolution with us as a commensal. Yet today we are saving, and improving the quality of, human lives that are vulnerable to this opportunist, and any line of investigation that might result in better control of invasive candidiasis has the potential worldwide to save hundreds of thousands of lives every year (Pfaller and Diekema 2007).

Aspergillus

Aspergillus fumigatus, the species responsible for ∼90% of invasive aspergillosis (Schmitt et al. 1990), causes relentless pneumonia, sinusitis progressing through tissue planes, and brain abscesses in neutropenic patients and those with phagocyte defects like chronic granulomatous disease. It is feared in immunosuppressed individuals because its susceptibility to antifungals is limited. This organism also causes disease at another extreme of immune function: allergic reactions.

Normal human innate immunity controls A. fumigatus, a versatile thermophilic plant saprobe. It is well adapted to the high temperatures that occur during bacterial decomposition of dead plants and tolerates thermophilic bacteria’s optimum of around 55°C (Latgé 1999). At human febrile temperatures of 39°C–40°C, it grows well. Its secretion of a very broad and redundant range of hydrolases (Kothary et al. 1984Kwon-Chung and Sugui 2013), evolved in the tumultuous competition for nutrients among the microbiome of decaying plants, allows it to easily access human tissues as sources of nutrition (Abad et al. 2010).

The mycelia of A. fumigatus give rise to conidiophores, specialized hyphae that produce small (∼3 µm) airborne dispersal cells, conidia. Their small size allows them to remain airborne for long periods and, incidentally, to enter human alveoli. On average, a person is estimated to inhale several hundred A. fumigatus conidia each day (Hospenthal et al. 1998); with exposure to grass cuttings, leaf litter, or compost, this number may be orders of magnitude higher (Mullins et al. 1976Poole and Wong 2013). Conidia are coated with hydrophobic proteins (Thau et al. 1994Paris et al. 2003) and with the chemoprotectant melanin (Pihet et al. 2009) to withstand harsh environmental stressors like freezing, sunlight, and desiccation (Kwon-Chung and Sugui 2013). They are recognized by innate immune cells of a human host only when they begin to germinate to produce a growing hypha (Levitz and Diamond 1985Aimanianda et al. 2009) after landing in an air-filled space like a paranasal sinus or lung. Aspergillus conidia can give rise to allergic lung disease, and the fungus can colonize the bronchiectasis of cystic fibrosis or chronic obstructive lung disease; but lethal invasive infection occurs when there is a dearth of functional neutrophils and macrophages to control germinating conidia. Hyphae proliferate in lung parenchyma or sinuses, and hyphal fragments occasionally are carried to highly perfused organs like the brain in which they initiate foci of metastatic infection. On encountering blood vessels, Aspergillus hyphae tend to enter and follow their course, clogging the vessel and causing infarction of downstream tissue. This angioinvasive behavior may be attributable to thigmotropism, the ability to sense and follow contours, which aspergilli have in common with fungi of diverse phyla (Perera et al. 1997Bowen et al. 2007).

Pathogenicity studies of A. fumigatus highlight general themes of innate immunity (Morton et al. 2012). For example, specialized alveolar epithelial cells, type II pneumocytes, phagocytose and kill conidia of A. fumigatus in their lysosomes, unless the conidia manage to germinate before their death (Wasylnka and Moore 2003). Neutrophils are attracted by the chemokines secreted by resident alveolar macrophages encountering A. fumigatus during their patrols of the alveolar space. Whereas small hyphae are engulfed and killed by neutrophils, larger hyphae are killed by their extracellular release of reactive oxygen and nitrogen species and antimicrobial peptides, in dependence on the pathogen recognition receptors (PRRs) TLR2 and 4, and dectin-1 (Netea et al. 2003Kennedy et al. 2007Werner et al. 2009). Neutrophil extracellular traps (NETs), critical to control polymicrobial infection in appendicitis (Brinkmann et al. 2004), nutritionally constrain A. fumigatus hyphae (McCormick et al. 2010) and down-regulate excessive inflammation elicited by their presence (Rohm et al. 2014).

Non-Fumigatus Aspergilli, Fusarium, Pseudoallescheria, and Other Opportunistic Ascomycetous Fungal Pathogens

Profound immunosuppression, for instance, during prolonged neutropenia, graft-versus-host disease, or severe rejection episodes of transplanted solid organs, permits invasive disease of many other types of environmental filamentous ascomycetes. Among them are the non-fumigatus species of Aspergillus (Torres et al. 2003), Fusarium solanumFusarium oxysporum, and other Fusarium spp., and their teleomorph Nectria spp. (Nucci and Anaissie 2007), Pseudoallescheria boydii, and its Scedosporium anamorphs (Quan and Spellberg 2010). Infections with these fungi are often lethal because the hosts they usually infect are incapable of mounting an effective immune response, and because they tend to be more resistant to currently available antifungals.

Fusarium spp. have evolved to infect plants, and the genomes of certain lineages contain one or more entire chromosomes encoding plant pathogenicity factors, which may be horizontally transferred within the genus (Ma et al. 2010). Encoded on pathogenicity chromosomes, and other genomic clusters reminiscent of bacterial pathogenicity islands, are secreted hydrolytic enzymes and signaling molecules expressed during early plant infection (Ma et al. 2010Rep and Kistler 2010). Whether possession of a pathogenicity chromosome corresponds to virulence for humans, for instance, in the F. oxysporum lineage identified to have caused 70% of invasive fusariosis in a San Antonio hospital (O’Donnell et al. 2004), is not yet known. In Fusarium, morphogenesis contributes to virulence for humans, because yeast-like cells are produced from hyphae in the host, and typically spread widely through the bloodstream to cause numerous foci of infection in the skin.

Pigmented filamentous ascomycetes, also called dematiaceous fungi, infect immunocompromised and, rarely, immunocompetent individuals to cause phaeohyphomycosis. Their pathobiology is diverse including brain abscesses (Cladophialphora bantianum, Ramichloridium spp., and Dactylaria gallopava), keratitis, sinus or soft tissue infections, ulcers, and cysts (Exophiala jeanselmeiExophiala dermatitidisCurvularia spp., Bipolaris, or Alternaria spp.). When directly inoculated into tissue, these fungi can cause devastating disease, as emphasized in a tragic outbreak caused by contaminated injectable steroids (Smith et al. 2013).

Another epidemiologically, biologically, and profoundly important ascomycete, P. jirovecii, is so distinctive in its biology, that only a reference to the related article in this collection can be made (Gigliotti et al. 2014).

Basidiomycota

In addition to the Cryptococcus spp. discussed above, other Basidiomycota growing predominantly as yeast in the host, Malassezia furfurTrichosporon asahii, and other members of human skin flora, are opportunists in patients with venous catheters and in immunosuppressed patients.

Mucorales

Profoundly immunocompromised patients at risk for the environmental fungi described above are also at risk for Mucorales infections. Additionally, Mucorales cause severe disease in diabetic patients especially at times of uncontrolled blood glucose, and patients with elevated serum iron (e.g., attributed to hemosiderosis), as described in the excellent series by Roden et al. (2005). Like the aspergilli, these fungi cause disease where airborne dispersal cells, sporangiospores, enter air-filled spaces (i.e., in sinuses and lungs). Infections with Mucorales progress rapidly as the fungus grows fast and relentlessly through tissue planes and bone, penetrating the eye and the brain when originating in sinuses, and causing widespread infarction because of its angioinvasive behavior. Depending on the underlying condition, such infections are often fatal (Roden et al. 2005), especially because these fungi tend toward resistance to current antifungals.

The Mucorales are an ubiquitous, ancient group mostly of saprotrophs, which, unlike the Ascomycota and Basidiomycota (the Dikarya), do not manifest sophisticated adaptations to diverse substrates, but rapidly use up available sugars and then move on via their sporangiospores (Hoffmann et al. 2013). Their evolutionary distance from the phyla of other human parasitic fungi is apparent in the structure of their hyphae, which are fragile, thin walled, and lack septa, and in the composition of their cell walls in which chitin and chitosan play the structural roles that glucans fulfill in the Dikarya (Dijksterhuis and Samson 2006). Some species like Mucor circillenoides are dimorphic and form yeast in the host (Dizbay et al. 2009Khan et al. 2009Lee et al. 2013), contributing to dissemination.

Among the Rhizopus spp., Rhizopus oryzae is responsible for ∼70% of human disease (Spellberg et al. 2005). Several of its virulence factors are areas of active investigation: its iron scavenging from the host (Fu et al. 2004Ibrahim et al. 20082010Spellberg et al. 2009Ibrahim 2011) and its ability to bind GRP78/BiP, a Kar2 homolog, on the surface of endothelial cells with its spore coat protein homolog CotH3 (Liu et al. 2010Gebremariam et al. 2014). Like a broad phylogenetic variety of fungi, Mucorales are angioinvasive. Thigmotropism, the proclivity to sense and follow curvatures of a surface, has been shown for one species, Mucor mucedo (Perera et al. 1997), and is likely to be common to all. Much remains to be learned about the biology of these evolutionarily old fungi and historically new human pathogens.Go to:

CONCLUSION

Immunologically intact humans manifest robust defenses against fungal diseases. Recently, human social evolution produced scientific medicine, whose progress has rendered a large population susceptible to infections with fungi not considered human pathogens as recently as a hundred years ago. Human social evolution may have reached a stage where prioritization of the resources needed for understanding fungal biology and for successful development of multiple new classes of antifungals is possible, as the example of AIDS shows. Effective antiretroviral therapy was developed with a massive research effort less than two decades ago, and if human society evolves to a point of valuing all human lives equally, near elimination of AIDS is as feasible as the quasi elimination of HIV mother-to-child transmission has been in wealthy countries (Siegfried et al. 2011Cohen et al. 2013Nicol et al. 2013). Unlike with HIV, which has no reservoir in nature, humans will always have to cope with fungal infections, because potential fungal pathogens are part of our normal flora and of soil, water, and air. But building on the tremendous efforts and findings of the past decades, new low-toxicity therapies and novel preventative measures of fungal infections can be within our reach.Go to:

ACKNOWLEDGMENTS

J.R.K. thanks Alison Clapp, Library Director of Boston Children’s Hospital, for ever-gracious and lightning-quick procurement of innumerable articles.Go to:

Footnotes

Editors: Arturo Casadevall, Aaron P. Mitchell, Judith Berman, Kyung J. Kwon-Chung, John R. Perfect, and Joseph Heitman

Additional Perspectives on Human Fungal Pathogens available at www.perspectivesinmedicine.orgGo to:

REFERENCES

*Reference is also in this collection.Abad A, Fernandez-Molina JV, Bikandi J, Ramirez A, Margareto J, Sendino J, Hernando FL, Ponton J, Garaizar J, Rementeria A 2010. What makes Aspergillus fumigatus a successful pathogen? Genes and molecules involved in invasive aspergillosis. Rev Iberoam Micol 27: 155–182. [PubMed]Aimanianda V, Bayry J, Bozza S, Kniemeyer O, Perruccio K, Elluru SR, Clavaud C, Paris S, Brakhage AA, Kaveri SV, et al. 2009. Surface hydrophobin prevents immune recognition of airborne fungal spores. Nature 460: 1117–1121. [PubMed]Albrecht A, Felk A, Pichova I, Naglik JR, Schaller M, de Groot P, Maccallum D, Odds FC, Schafer W, Klis F, et al. 2006. Glycosylphosphatidylinositol-anchored proteases of Candida albicans target proteins necessary for both cellular processes and host–pathogen interactions. J Biol Chem 281: 688–694. [PubMed]Alby K, Schaefer D, Bennett RJ 2009. Homothallic and heterothallic mating in the opportunistic pathogen Candida albicans. Nature 460: 890–893. [PMC free article] [PubMed]Ameen M 2010. Epidemiology of superficial fungal infections. Clin Dermatol 28: 197–201. [PubMed]Araujo Gde S, Fonseca FL, Pontes B, Torres A, Cordero RJ, Zancope-Oliveira RM, Casadevall A, Viana NB, Nimrichter L, Rodrigues ML, et al. 2012. Capsules from pathogenic and non-pathogenic Cryptococcusspp. manifest significant differences in structure and ability to protect against phagocytic cells. PLoS ONE7: e29561. [PMC free article] [PubMed]Argimon S, Fanning S, Blankenship JR, Mitchell AP 2011. Interaction between the Candida albicans high-osmolarity glycerol (HOG) pathway and the response to human β-defensins 2 and 3. Eukaryot Cell 10: 272–275. [PMC free article] [PubMed]Aulakh HK, Aulakh KS, Troy GC 2012. Feline histoplasmosis: A retrospective study of 22 cases (1986–2009). J Am Anim Hosp Assoc 48: 182–187. [PubMed]Bagagli E, Bosco SM, Theodoro RC, Franco M 2006. Phylogenetic and evolutionary aspects of Paracoccidioides brasiliensis reveal a long coexistence with animal hosts that explain several biological features of the pathogen. Infect Genet Evol 6: 344–351. [PubMed]Bagagli E, Theodoro RC, Bosco SM, McEwen JG 2008. Paracoccidioides brasiliensis: Phylogenetic and ecological aspects. Mycopathologia 165: 197–207. [PubMed]Bastidas RJ, Heitman J 2009. Trimorphic stepping stones pave the way to fungal virulence. Proc Natl Acad Sci 106: 351–352. [PMC free article] [PubMed]Bauder B, Kubber-Heiss A, Steineck T, Kuttin ES, Kaufman L 2000. Granulomatous skin lesions due to histoplasmosis in a badger (Meles meles) in Austria. Med Mycol 38: 249–253. [PubMed]Bergman A, Casadevall A 2010. Mammalian endothermy optimally restricts fungi and metabolic costs. mBio 1: e00212–10. [PMC free article] [PubMed]Beyhan S, Gutierrez M, Voorhies M, Sil A 2013. A temperature-responsive network links cell shape and virulence traits in a primary fungal pathogen. PLoS Biol 11: e1001614. [PMC free article] [PubMed]Bittencourt AL 1988. Entomophthoromycosis. Review. Med Cutan Ibero Lat Am 16: 93–100. [PubMed]Bittencourt AL, Marback R, Nossa LM 2006. Mucocutaneous entomophthoramycosis acquired by conjunctival inoculation of the fungus. Am J Trop Med Hyg 75: 936–938. [PubMed]Blake JS, Dahl MV, Herron MJ, Nelson RD 1991. An immunoinhibitory cell wall glycoprotein (Mannan) from Trichophyton rubrum. J Invest Dermatol 96: 657–661. [PubMed]Boelaert JR, de Locht M, Van Cutsem J, Kerrels V, Cantinieaux B, Verdonck A, Van Landuyt HW, Schneider YJ 1993. Mucormycosis during deferoxamine therapy is a siderophore-mediated infection. In vitro and in vivo animal studies. J Clin Invest 91: 1979–1986. [PMC free article] [PubMed]Bohse ML, Woods JP 2007. Expression and interstrain variability of the YPS3 gene of Histoplasma capsulatum. Eukaryot Cell 6: 609–615. [PMC free article] [PubMed]Bougnoux ME, Pujol C, Diogo D, Bouchier C, Soll DR, d’Enfert C 2008. Mating is rare within as well as between clades of the human pathogen Candida albicans. Fungal Genet Biol 45: 221–231. [PMC free article] [PubMed]Bowen AD, Davidson FA, Keatch R, Gadd GM 2007. Induction of contour sensing in Aspergillus niger by stress and its relevance to fungal growth mechanics and hyphal tip structure. Fungal Genet Biol 44: 484–491. [PubMed]Bradsher RW, Chapman SW, Pappas PG 2003. Blastomycosis. Infect Dis Clin North Am 17: 21–40. [PubMed]Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A 2004. Neutrophil extracellular traps kill bacteria. Science 303: 1532–1535. [PubMed]Brömel C, Sykes JE 2005. Histoplasmosis in dogs and cats. Clin Tech Small Anim Pract 20: 227–232. [PubMed]Browne SG 1954. Moniliasis following antibiotic therapy. Lancet 266: 393–395. [PubMed]Browne N, Heelan M, Kavanagh K 2013. An analysis of the structural and functional similarities of insect hemocytes and mammalian phagocytes. Virulence 4: 597–603. [PMC free article] [PubMed]Byrnes EJ III, Li W, Lewit Y, Ma H, Voelz K, Ren P, Carter DA, Chaturvedi V, Bildfell RJ, May RC, et al. 2010. Emergence and pathogenicity of highly virulent Cryptococcus gattii genotypes in the northwest United States. PLoS Pathog 6: e1000850. [PMC free article] [PubMed]Cafarchia C, Iatta R, Latrofa MS, Graser Y, Otranto D 2013. Molecular epidemiology, phylogeny and evolution of dermatophytes. Infect Genet Evol 20: 336–351. [PubMed]Cain CW, Lohse MB, Homann OR, Sil A, Johnson AD 2012. A conserved transcriptional regulator governs fungal morphology in widely diverged species. Genetics 190: 511–521. [PMC free article][PubMed]Calderone RA, Fonzi WA 2001. Virulence factors of Candida albicans. Trends Microbiol 9: 327–335. [PubMed]Casadevall A 2005. Fungal virulence, vertebrate endothermy, and dinosaur extinction: Is there a connection? Fungal Genet Biol 42: 98–106. [PubMed]Casadevall A 2010. Cryptococci at the brain gate: Break and enter or use a Trojan horse? J Clin Invest 120: 1389–1392. [PMC free article] [PubMed]Casadevall A 2012. Fungi and the rise of mammals. PLoS Pathog 8: e1002808. [PMC free article][PubMed]Casadevall A, Pirofski L 2001. Host–pathogen interactions: The attributes of virulence. J Infect Dis 184: 337–344. [PubMed]Casadevall A, Pirofski LA 2007. Accidental virulence, cryptic pathogenesis, Martians, lost hosts, and the pathogenicity of environmental microbes. Eukaryot Cell 6: 2169–2174. [PMC free article] [PubMed]Casadevall A, Cleare W, Feldmesser M, Glatman-Freedman A, Goldman DL, Kozel TR, Lendvai N, Mukherjee J, Pirofski LA, Rivera J, et al. 1998. Characterization of a murine monoclonal antibody to Cryptococcus neoformans polysaccharide that is a candidate for human therapeutic studies. Antimicrob Agents Chemother 42: 1437–1446. [PMC free article] [PubMed]Chau TT, Mai NH, Phu NH, Nghia HD, Chuong LV, Sinh DX, Duong VA, Diep PT, Campbell JI, Baker S, et al. 2010. A prospective descriptive study of cryptococcal meningitis in HIV uninfected patients in Vietnam—High prevalence of Cryptococcus neoformans var grubii in the absence of underlying disease. BMC Infect Dis 10: 199. [PMC free article] [PubMed]Chen J, Varma A, Diaz MR, Litvintseva AP, Wollenberg KK, Kwon-Chung KJ 2008. Cryptococcus neoformans strains and infection in apparently immunocompetent patients, China. Emerg Infect Dis 14: 755–762. [PMC free article] [PubMed]Chen YL, Lehman VN, Lewit Y, Averette AF, Heitman J 2013. Calcineurin governs thermotolerance and virulence of Cryptococcus gattii. G3 (Bethesda) 3: 527–539. [PMC free article] [PubMed]Choi YH, Ngamskulrungroj P, Varma A, Sionov E, Hwang SM, Carriconde F, Meyer W, Litvintseva AP, Lee WG, Shin JH, et al. 2010. Prevalence of the VNIc genotype of Cryptococcus neoformans in non-HIV-associated cryptococcosis in the Republic of Korea. FEMS Yeast Res 10: 769–778. [PMC free article][PubMed]Chun CD, Brown JC, Madhani HD 2011. A major role for capsule-independent phagocytosis-inhibitory mechanisms in mammalian infection by Cryptococcus neoformans. Cell Host Microbe 9: 243–251. [PMC free article] [PubMed]Citiulo F, Jacobsen ID, Miramon P, Schild L, Brunke S, Zipfel P, Brock M, Hube B, Wilson D 2012.Candida albicans scavenges host zinc via Pra1 during endothelial invasion. PLoS Pathog 8: e1002777. [PMC free article] [PubMed]Cohen MS, Smith MK, Muessig KE, Hallett TB, Powers KA, Kashuba AD 2013. Antiretroviral treatment of HIV-1 prevents transmission of HIV-1: Where do we go from here? Lancet 382: 1515–1524. [PMC free article] [PubMed]Comerio RM, Andorno AV, Botto EN 2008. Conidiobolus coronatus isolation from a pest aphids of chives (Allium schoenoprasum L.). Rev Iberoam Micol 25: 193–195. [PubMed]Cramer RA Jr, Perfect BZ, Pinchai N, Park S, Perlin DS, Asfaw YG, Heitman J, Perfect JR, Steinbach WJ 2008. Calcineurin target CrzA regulates conidial germination, hyphal growth, and pathogenesis of Aspergillus fumigatus. Eukaryot Cell 7: 1085–1097. [PMC free article] [PubMed]Dahl MV 1993. Suppression of immunity and inflammation by products produced by dermatophytes. J Am Acad Dermatol 28: S19–S23. [PubMed]Dijksterhuis J, Samson RA 2006. Zygomycetes. In Food spoilage microorganisms (ed. Blackburn CW), pp. 415–436 CRC, Boca Raton, FL.Dizbay M, Adisen E, Kustimur S, Sari N, Cengiz B, Yalcin B, Kalkanci A, Gonul, Sugita T 2009.Fungemia and cutaneous zygomycosis due to Mucor circinelloides in an intensive care unit patient: Case report and review of literature. Jpn J Infect Dis 62: 146–148. [PubMed]Echetebu CO, Ononogbu IC 1982. Extracellular lipase and proteinase of Basidiobolus haptosporus: Possible role in subcutaneous mycosis. Mycopathologia 80: 171–177. [PubMed]Eckhart L, Valle LD, Jaeger K, Ballaun C, Szabo S, Nardi A, Buchberger M, Hermann M, Alibardi L, Tschachler E 2008. Identification of reptilian genes encoding hair keratin-like proteins suggests a new scenario for the evolutionary origin of hair. Proc Natl Acad Sci 105: 18419–18423. [PMC free article][PubMed]Edwards JA, Alore EA, Rappleye CA 2011. The yeast–phase virulence requirement for α-glucan synthase differs among Histoplasma capsulatum chemotypes. Eukaryot Cell 10: 87–97. [PMC free article][PubMed]Edwards JA, Chen C, Kemski MM, Hu J, Mitchell TK, Rappleye CA 2013. Histoplasma yeast and mycelial transcriptomes reveal pathogenic-phase and lineage-specific gene expression profiles. BMC Genomics 14: 695. [PMC free article] [PubMed]Ellis DH, Pfeiffer TJ 1990. Natural habitat of Cryptococcus neoformans var. gattii. J Clin Microbiol 28: 1642–1644. [PMC free article] [PubMed]Ernst JF, Pla J 2011. Signaling the glycoshield: Maintenance of the Candida albicans cell wall. Int J Med Microbiol 301: 378–383. [PubMed]Findley K, Rodriguez-Carres M, Metin B, Kroiss J, Fonseca A, Vilgalys R, Heitman J 2009. Phylogeny and phenotypic characterization of pathogenic Cryptococcus species and closely related saprobic taxa in the Tremellales. Eukaryot Cell 8: 353–361. [PMC free article] [PubMed]Fisher MC, Koenig GL, White TJ, San-Blas G, Negroni R, Alvarez IG, Wanke B, Taylor JW 2001.Biogeographic range expansion into South America by Coccidioides immitis mirrors New World patterns of human migration. Proc Natl Acad Sci 98: 4558–4562. [PMC free article] [PubMed]Forche A, Alby K, Schaefer D, Johnson AD, Berman J, Bennett RJ 2008. The parasexual cycle in Candida albicans provides an alternative pathway to meiosis for the formation of recombinant strains. PLoS Biol 6: e110. [PMC free article] [PubMed]Fortes ST, Lazera MS, Nishikawa MM, Macedo RC, Wanke B 2001. First isolation of Cryptococcus neoformans var. gattii from a native jungle tree in the Brazilian Amazon rainforest. Mycoses 44: 137–140. [PubMed]Fortes MR, Miot HA, Kurokawa CS, Marques ME, Marques SA 2011. Immunology of paracoccidioidomycosis. An Bras Dermatol 86: 516–524. [PubMed]Fraser JA, Giles SS, Wenink EC, Geunes-Boyer SG, Wright JR, Diezmann S, Allen A, Stajich JE, Dietrich FS, Perfect JR, et al. 2005. Same-sex mating and the origin of the Vancouver Island Cryptococcus gattiioutbreak. Nature 437: 1360–1364. [PubMed]Fu Y, Lee H, Collins M, Tsai HF, Spellberg B, Edwards JE Jr, Kwon-Chung KJ, Ibrahim AS 2004. Cloning and functional characterization of the Rhizopus oryzae high affinity iron permease (rFTR1) gene. FEMS Microbiol Lett 235: 169–176. [PubMed]Garcia-Solache MA, Casadevall A 2010. Global warming will bring new fungal diseases for mammals. mBio 1: e00061–10. [PMC free article] [PubMed]Gebremariam T, Liu M, Luo G, Bruno V, Phan QT, Waring AJ, Edwards JE Jr, Filler SG, Yeaman MR, Ibrahim AS 2014. CotH3 mediates fungal invasion of host cells during mucormycosis. J Clin Invest 124: 237–250. [PMC free article] [PubMed]Ghannoum MA 2000. Potential role of phospholipases in virulence and fungal pathogenesis. Clin Microbiol Rev 13: 122–143. [PMC free article] [PubMed]* Gigliotti F, Limper AH, Wright T 2014. Pneumocystis. Cold Spring Harb Perspect Med. 10.1101/cshperspect.a019828. [PMC free article] [PubMed] [CrossRef]Goddard MR, Godfray HC, Burt A 2005. Sex increases the efficacy of natural selection in experimental yeast populations. Nature 434: 636–640. [PubMed]Gradisar H, Friedrich J, Krizaj I, Jerala R 2005. Similarities and specificities of fungal keratinolytic proteases: Comparison of keratinases of Paecilomyces marquandii and Doratomyces microsporus to some known proteases. Appl Environ Microbiol 71: 3420–3426. [PMC free article] [PubMed]Gugnani HC 1992. Entomophthoromycosis due to Conidiobolus. Eur J Epidemiol 8: 391–396. [PubMed]Hagen F, Ceresini PC, Polacheck I, Ma H, van Nieuwerburgh F, Gabaldon T, Kagan S, Pursall ER, Hoogveld HL, van Iersel LJ, et al. 2013. Ancient dispersal of the human fungal pathogen Cryptococcus gattii from the Amazon rainforest. PLoS ONE 8: e71148. [PMC free article] [PubMed]Heckman DS, Geiser DM, Eidell BR, Stauffer RL, Kardos NL, Hedges SB 2001. Molecular evidence for the early colonization of land by fungi and plants. Science 293: 1129–1133. [PubMed]Hickman MA, Zeng G, Forche A, Hirakawa MP, Abbey D, Harrison BD, Wang YM, Su CH, Bennett RJ, Wang Y, et al. 2013. The “obligate diploid” Candida albicans forms mating-competent haploids. Nature494: 55–59.Hoff GL, Bigler WJ 1981. The role of bats in the propagation and spread of histoplasmosis: A review. J Wildl Dis 17: 191–196. [PubMed]Hoffmann K, Pawlowska J, Walther G, Wrzosek M, de Hoog GS, Benny GL, Kirk PM, Voigt K 2013. The family structure of the Mucorales: A synoptic revision based on comprehensive multigene-genealogies. Persoonia 30: 57–76. [PMC free article] [PubMed]Hogan DA, Muhlschlegel FA 2011. Candida albicans developmental regulation: Adenylyl cyclase as a coincidence detector of parallel signals. Curr Opin Microbiol 14: 682–686. [PubMed]Holbrook ED, Rappleye CA 2008. Histoplasma capsulatum pathogenesis: Making a lifestyle switch. Curr Opin Microbiol 11: 318–324. [PubMed]Holland WL, Summers SA 2008. Sphingolipids, insulin resistance, and metabolic disease: New insights from in vivo manipulation of sphingolipid metabolism. Endocr Rev 29: 381–402. [PMC free article][PubMed]Hospenthal DR, Kwon-Chung KJ, Bennett JE 1998. Concentrations of airborne Aspergillus compared to the incidence of invasive aspergillosis: Lack of correlation. Med Mycol 36: 165–168. [PubMed]Hoyer LL, Green CB, Oh SH, Zhao X 2008. Discovering the secrets of the Candida albicans agglutinin-like sequence (ALS) gene family—A sticky pursuit. Med Mycol 46: 1–15. [PMC free article] [PubMed]Hull CM, Johnson AD 1999. Identification of a mating type-like locus in the asexual pathogenic yeast Candida albicans. Science 285: 1271–1275. [PubMed]Humphreys CP, Franks PJ, Rees M, Bidartondo MI, Leake JR, Beerling DJ 2010. Mutualistic mycorrhiza-like symbiosis in the most ancient group of land plants. Nat Commun 1: 103. [PubMed]Ibrahim AS 2011. Host cell invasion in mucormycosis: Role of iron. Curr Opin Microbiol 14: 406–411. [PMC free article] [PubMed]Ibrahim AS, Edwards JE Jr, Fu Y, Spellberg B 2006. Deferiprone iron chelation as a novel therapy for experimental mucormycosis. J Antimicrob Chemother 58: 1070–1073. [PubMed]Ibrahim AS, Spellberg B, Edwards J Jr 2008. Iron acquisition: A novel perspective on mucormycosis pathogenesis and treatment. Curr Opin Infect Dis 21: 620–625. [PMC free article] [PubMed]Ibrahim AS, Gebremariam T, Lin L, Luo G, Husseiny MI, Skory CD, Fu Y, French SW, Edwards JE Jr, Spellberg B 2010. The high affinity iron permease is a key virulence factor required for Rhizopus oryzaepathogenesis. Mol Microbiol 77: 587–604. [PMC free article] [PubMed]Isa-Isa R, Arenas R, Fernandez RF, Isa M 2012. Rhinofacial conidiobolomycosis (entomophthoramycosis). Clin Dermatol 30: 409–412. [PubMed]Kam AP, Xu J 2002. Diversity of commensal yeasts within and among healthy hosts. Diagn Microbiol Infect Dis 43: 19–28. [PubMed]Kasuga T, Taylor JW, White TJ 1999. Phylogenetic relationships of varieties and geographical groups of the human pathogenic fungus Histoplasma capsulatum Darling. J Clin Microbiol 37: 653–663. [PMC free article] [PubMed]Kasuga T, White TJ, Koenig G, McEwen J, Restrepo A, Castaneda E, Da Silva Lacaz C, Heins-Vaccari EM, De Freitas RS, Zancope-Oliveira RM, et al. 2003. Phylogeography of the fungal pathogen Histoplasma capsulatum. Mol Ecol 12: 3383–3401. [PubMed]Kauffman CA 2007. Histoplasmosis: A clinical and laboratory update. Clin Microbiol Rev 20: 115–132. [PMC free article] [PubMed]Kennedy AD, Willment JA, Dorward DW, Williams DL, Brown GD, DeLeo FR 2007. Dectin-1 promotes fungicidal activity of human neutrophils. Eur J Immunol 37: 467–478. [PubMed]Khan ZU, Ahmad S, Brazda A, Chandy R 2009. Mucor circinelloides as a cause of invasive maxillofacial zygomycosis: An emerging dimorphic pathogen with reduced susceptibility to posaconazole. J Clin Microbiol 47: 1244–1248. [PMC free article] [PubMed]Kidd SE, Sorrell TC, Meyer W 2003. Isolation of two molecular types of Cryptococcus neoformans var. gattii from insect frass. Med Mycol 41: 171–176.Kidd SE, Hagen F, Tscharke RL, Huynh M, Bartlett KH, Fyfe M, Macdougall L, Boekhout T, Kwon-Chung KJ, Meyer W 2004. A rare genotype of Cryptococcus gattii caused the cryptococcosis outbreak on Vancouver Island (British Columbia, Canada). Proc Natl Acad Sci 101: 17258–17263. [PMC free article][PubMed]Kidd SE, Bach PJ, Hingston AO, Mak S, Chow Y, MacDougall L, Kronstad JW, Bartlett KH 2007.Cryptococcus gattii dispersal mechanisms, British Columbia, Canada. Emerg Infect Dis 13: 51–57. [PMC free article] [PubMed]Kothary MH, Chase T Jr, Macmillan JD 1984. Correlation of elastase production by some strains of Aspergillus fumigatus with ability to cause pulmonary invasive aspergillosis in mice. Infect Immun 43: 320–325. [PMC free article] [PubMed]Krcmery V, Barnes AJ 2002. Non-albicans Candidaspp. causing fungaemia: Pathogenicity and antifungal resistance. J Hosp Infect 50: 243–260. [PubMed]Krockenberger MB, Canfield PJ, Malik R 2002. Cryptococcus neoformans in the koala (Phascolarctos cinereus): Colonization by C. n. var. gattii and investigation of environmental sources. Med Mycol 40: 263–272. [PubMed]Kronstad JW, Attarian R, Cadieux B, Choi J, D’Souza CA, Griffiths EJ, Geddes JM, Hu G, Jung WH, Kretschmer M, et al. 2011. Expanding fungal pathogenesis: Cryptococcus breaks out of the opportunistic box. Nat Rev Microbiol 9: 193–203. [PMC free article] [PubMed]Kronstad JW, Hu G, Jung WH 2013. An encapsulation of iron homeostasis and virulence in Cryptococcus neoformans. Trends Microbiol 21: 457–465. [PMC free article] [PubMed]Kumamoto CA 2008. Molecular mechanisms of mechanosensing and their roles in fungal contact sensing. Nat Rev Microbiol 6: 667–673. [PMC free article] [PubMed]Kwon-Chung KJ 2012. Taxonomy of fungi causing mucormycosis and entomophthoramycosis (Zygomycosis) and nomenclature of the disease: Molecular mycologic perspectives. Clin Infect Dis 54: S8–S15. [PMC free article] [PubMed]Kwon-Chung KJ, Sugui JA 2013. Aspergillus fumigatus—What makes the species a ubiquitous human fungal pathogen? PLoS Pathog 9: e1003743. [PMC free article] [PubMed]Latgé JP 1999. Aspergillus fumigatus and aspergillosis. Clin Microbiol Rev 12: 310–350. [PMC free article] [PubMed]Lee SC, Li A, Calo S, Heitman J 2013. Calcineurin plays key roles in the dimorphic transition and virulence of the human pathogenic zygomycete Mucor circinelloides. PLoS Pathog 9: e1003625. [PMC free article] [PubMed]Levitz SM, Diamond RD 1985. Mechanisms of resistance of Aspergillus fumigatus Conidia to killing by neutrophils in vitro. J Infect Dis 152: 33–42. [PubMed]Lin JS, Huang JH, Hung LY, Wu SY, Wu-Hsieh BA 2010. Distinct roles of complement receptor 3, Dectin-1, and sialic acids in murine macrophage interaction with Histoplasma yeast. J Leukoc Biol 88: 95–106. [PubMed]Litvintseva AP, Mitchell TG 2012. Population genetic analyses reveal the African origin and strain variation of Cryptococcus neoformans var. grubii. PLoS Pathog 8: e1002495. [PMC free article] [PubMed]Liu Y, Filler SG 2011. Candida albicans Als3, a multifunctional adhesin and invasin. Eukaryot Cell 10: 168–173. [PMC free article] [PubMed]Liu OW, Chun CD, Chow ED, Chen C, Madhani HD, Noble SM 2008. Systematic genetic analysis of virulence in the human fungal pathogen Cryptococcus neoformans. Cell 135: 174–188. [PMC free article][PubMed]Liu M, Spellberg B, Phan QT, Fu Y, Lee AS, Edwards JE Jr, Filler SG, Ibrahim AS 2010. The endothelial cell receptor GRP78 is required for mucormycosis pathogenesis in diabetic mice. J Clin Invest 120: 1914–1924. [PMC free article] [PubMed]Lockhart SR, Wu W, Radke JB, Zhao R, Soll DR 2005. Increased virulence and competitive advantage of a/α over a/a or α/α offspring conserves the mating system of Candida albicans. Genetics 169: 1883–1890. [PMC free article] [PubMed]Lott TJ, Fundyga RE, Kuykendall RJ, Arnold J 2005. The human commensal yeast, Candida albicans, has an ancient origin. Fungal Genet Biol 42: 444–451. [PubMed]Lucking R, Huhndorf S, Pfister DH, Plata ER, Lumbsch HT 2009. Fungi evolved right on track. Mycologia101: 810–822. [PubMed]Ma LJ, van der Does HC, Borkovich KA, Coleman JJ, Daboussi MJ, Di Pietro A, Dufresne M, Freitag M, Grabherr M, Henrissat B, et al. 2010. Comparative genomics reveals mobile pathogenicity chromosomes in Fusarium. Nature 464: 367–373. [PMC free article] [PubMed]Magee BB, Magee PT 2000. Induction of mating in Candida albicans by construction of MTLa and MTLα strains. Science 289: 310–313. [PubMed]Marples MJ, Di Menna ME 1952. The incidence of Candida albicans in Dunedin, New Zealand. J Pathol Bacteriol 64: 497–502. [PubMed]Marques SA 2012. Paracoccidioidomycosis. Clin Dermatol 30: 610–615. [PubMed]Marques SA 2013. Paracoccidioidomycosis: Epidemiological, clinical, diagnostic and treatment up-dating. An Bras Dermatol 88: 700–711. [PMC free article] [PubMed]McCormick A, Heesemann L, Wagener J, Marcos V, Hartl D, Loeffler J, Heesemann J, Ebel F 2010. NETs formed by human neutrophils inhibit growth of the pathogenic mold Aspergillus fumigatus. Microbes Infect 12: 928–936. [PubMed]McGill S, Malik R, Saul N, Beetson S, Secombe C, Robertson I, Irwin P 2009. Cryptococcosis in domestic animals in Western Australia: A retrospective study from 1995–2006. Med Mycol 47: 625–639. [PubMed]McMurray DN, Greer DL 1979. Immune responses in bats following intranasal infection with Histoplasma capsulatum. Am J Trop Med Hyg 28: 1036–1039. [PubMed]Meyer W, Castaneda A, Jackson S, Huynh M, Castaneda E 2003. Molecular typing of IberoAmerican Cryptococcus neoformans isolates. Emerg Infect Dis 9: 189–195. [PMC free article] [PubMed]Millanes AM, Truong C, Westberg M, Diederich P, Wedin M 2014. Host switching promotes diversity in host-specialized mycoparasitic fungi: Uncoupled evolution in the Biatoropsis-Usnea system. Evolution10.1111/evo.12374. [PubMed] [CrossRef]Miller MG, Johnson AD 2002. White-opaque switching in Candida albicans is controlled by mating-type locus homeodomain proteins and allows efficient mating. Cell 110: 293–302. [PubMed]Miller WG, Padhye AA, van Bonn W, Jensen E, Brandt ME, Ridgway SH 2002. Cryptococcosis in a bottlenose dolphin (Tursiops truncatus) caused by Cryptococcus neoformans var. gattii. J Clin Microbiol40: 721–724. [PMC free article] [PubMed]Mitchell TG, Perfect JR 1995. Cryptococcosis in the era of AIDS—100 years after the discovery of Cryptococcus neoformans. Clin Microbiol Rev 8: 515–548. [PMC free article] [PubMed]Molez JF 1998. The historical question of acquired immunodeficiency syndrome in the 1960s in the Congo River basin area in relation to cryptococcal meningitis. Am J Trop Med Hyg 58: 273–276. [PubMed]Money NP, Davis CM, Ravishankar JP 2004. Biomechanical evidence for convergent evolution of the invasive growth process among fungi and oomycete water molds. Fungal Genet Biol 41: 872–876. [PubMed]Morita T, Kishimoto M, Shimada A, Matsumoto Y, Shindo J 2001. Disseminated histoplasmosis in a sea otter (Enhydra lutris). J Comp Pathol 125: 219–223. [PubMed]Morschhäuser J 2011. Nitrogen regulation of morphogenesis and protease secretion in Candida albicans. Int J Med Microbiol 301: 390–394. [PubMed]Morton CO, Bouzani M, Loeffler J, Rogers TR 2012. Direct interaction studies between Aspergillus fumigatus and human immune cells: What have we learned about pathogenicity and host immunity? Front Microbiol 3: 413. [PMC free article] [PubMed]Mullins J, Harvey R, Seaton A 1976. Sources and incidence of airborne Aspergillus fumigatus (Fres). Clin Allergy 6: 209–217. [PubMed]Neilson JB, Ivey MH, Bulmer GS 1978. Cryptococcus neoformans: Pseudohyphal forms surviving culture with Acanthamoeba polyphaga. Infect Immun 20: 262–266. [PMC free article] [PubMed]Nemecek JC, Wuthrich M, Klein BS 2006. Global control of dimorphism and virulence in fungi. Science312: 583–588. [PubMed]Netea MG, Warris A, Van der Meer JW, Fenton MJ, Verver-Janssen TJ, Jacobs LE, Andresen T, Verweij PE, Kullberg BJ 2003. Aspergillus fumigatus evades immune recognition during germination through loss of toll-like receptor-4-mediated signal transduction. J Infect Dis 188: 320–326. [PubMed]Nguyen VQ, Sil A 2008. Temperature-induced switch to the pathogenic yeast form of Histoplasma capsulatum requires Ryp1, a conserved transcriptional regulator. Proc Natl Acad Sci 105: 4880–4885. [PMC free article] [PubMed]Nguyen C, Barker BM, Hoover S, Nix DE, Ampel NM, Frelinger JA, Orbach MJ, Galgiani JN 2013.Recent advances in our understanding of the environmental, epidemiological, immunological, and clinical dimensions of coccidioidomycosis. Clin Microbiol Rev 26: 505–525. [PMC free article] [PubMed]Nicol MR, Adams JL, Kashuba AD 2013. HIV PrEP trials: The road to success. Clin Investig (Lond) 3: 295–308. [PMC free article] [PubMed]Nielsen K, De Obaldia AL, Heitman J 2007. Cryptococcus neoformans mates on pigeon guano: Implications for the realized ecological niche and globalization. Eukaryot Cell 6: 949–959. [PMC free article] [PubMed]Noble SM 2013. Candida albicans specializations for iron homeostasis: From commensalism to virulence. Curr Opin Microbiol 16: 708–715. [PMC free article] [PubMed]Noble SM, French S, Kohn LA, Chen V, Johnson AD 2010. Systematic screens of a Candida albicanshomozygous deletion library decouple morphogenetic switching and pathogenicity. Nat Genet 42: 590–598. [PMC free article] [PubMed]Nucci M, Anaissie E 2007. Fusarium infections in immunocompromised patients. Clin Microbiol Rev 20: 695–704. [PMC free article] [PubMed]O’Brien HE, Parrent JL, Jackson JA, Moncalvo JM, Vilgalys R 2005. Fungal community analysis by large-scale sequencing of environmental samples. Appl Environ Microbiol 71: 5544–5550. [PMC free article][PubMed]Odds FC 1984. Ecology and epidemiology of Candida species. Zentralbl Bakteriol Mikrobiol Hyg A 257: 207–212. [PubMed]Odds F 1988. Candida and candidosis: A review and bibliography. Bailliere and Tindall, London.O’Donnell K, Sutton DA, Rinaldi MG, Magnon KC, Cox PA, Revankar SG, Sanche S, Geiser DM, Juba JH, van Burik JA, et al. 2004. Genetic diversity of human pathogenic members of the Fusarium oxysporumcomplex inferred from multilocus DNA sequence data and amplified fragment length polymorphism analyses: Evidence for the recent dispersion of a geographically widespread clonal lineage and nosocomial origin. J Clin Microbiol 42: 5109–5120. [PMC free article] [PubMed]Okafor JI 1994. Purification and characterization of protease enzymes of Basidiobolus and Conidiobolusspecies. Mycoses 37: 265–269. [PubMed]Okafor JI, Gugnani HC 1990. Lipase activity of Basidiobolus and Conidiobolus species. Mycoses 33: 81–85. [PubMed]Okafor JI, Gugnani HC, Testratke D, Yangoo BG 1987. Extracellular enzyme activities by Basidiobolusand Conidiobolus isolates on solid media. Mykosen 30: 404–407. [PubMed]Okagaki LH, Strain AK, Nielsen JN, Charlier C, Baltes NJ, Chretien F, Heitman J, Dromer F, Nielsen K 2010. Cryptococcal cell morphology affects host cell interactions and pathogenicity. PLoS Pathog 6: e1000953. [PMC free article] [PubMed]Oliveira DL, Nakayasu ES, Joffe LS, Guimaraes AJ, Sobreira TJ, Nosanchuk JD, Cordero RJ, Frases S, Casadevall A, Almeida IC, et al. 2010. Characterization of yeast extracellular vesicles: Evidence for the participation of different pathways of cellular traffic in vesicle biogenesis. PLoS ONE 5: e11113. [PMC free article] [PubMed]Paris S, Debeaupuis JP, Crameri R, Carey M, Charles F, Prevost MC, Schmitt C, Philippe B, Latgé JP 2003. Conidial hydrophobins of Aspergillus fumigatus. Appl Environ Microbiol 69: 1581–1588. [PMC free article] [PubMed]Park BJ, Wannemuehler KA, Marston BJ, Govender N, Pappas PG, Chiller TM 2009. Estimation of the current global burden of cryptococcal meningitis among persons living with HIV/AIDS. AIDS 23: 525–530. [PubMed]Perera TH, Gregory DW, Marshall D, Gow NA 1997. Contact-sensing by hyphae of dermatophytic and saprophytic fungi. J Med Vet Mycol 35: 289–293. [PubMed]Pfaller MA, Diekema DJ 2007. Epidemiology of invasive candidiasis: A persistent public health problem. Clin Microbiol Rev 20: 133–163. [PMC free article] [PubMed]Pihet M, Vandeputte P, Tronchin G, Renier G, Saulnier P, Georgeault S, Mallet R, Chabasse D, Symoens F, Bouchara JP 2009. Melanin is an essential component for the integrity of the cell wall of Aspergillus fumigatus conidia. BMC Microbiol 9: 177. [PMC free article] [PubMed]Poole CJ, Wong M 2013. Allergic bronchopulmonary aspergillosis in garden waste (compost) collectors—Occupational implications. Occup Med (Lond) 63: 517–519. [PubMed]Prabhu RM, Patel R 2004. Mucormycosis and entomophthoramycosis: A review of the clinical manifestations, diagnosis and treatment. Clin Microbiol Infect 10: 31–47. [PubMed]Puccia R, Juliano MA, Juliano L, Travassos LR, Carmona AK 1999. Detection of the basement membrane-degrading proteolytic activity of Paracoccidioides brasiliensis after SDS-PAGE using agarose overlays containing Abz-MKALTLQ-EDDnp. Braz J Med Biol Res 32: 645–649. [PubMed]Puccia R, McEwen JG, Cisalpino PS 2008. Diversity in Paracoccidioides brasiliensis. The PbGP43 gene as a genetic marker. Mycopathologia 165: 275–287. [PubMed]Puccia R, Vallejo MC, Matsuo AL, Longo LV 2011. The paracoccidioides cell wall: Past and present layers toward understanding interaction with the host. Front Microbiol 2: 257. [PMC free article][PubMed]Pukkila-Worley R, Mylonakis E 2008. Epidemiology and management of cryptococcal meningitis: Developments and challenges. Expert Opin Pharmacother 9: 551–560. [PubMed]Quan C, Spellberg B 2010. Mucormycosis, pseudallescheriasis, and other uncommon mold infections. Proc Am Thorac Soc 7: 210–215. [PMC free article] [PubMed]Queiroz-Telles F, Escuissato DL 2011. Pulmonary paracoccidioidomycosis. Semin Respir Crit Care Med32: 764–774. [PubMed]Quist EM, Belcher C, Levine G, Johnson M, Heatley JJ, Kiupel M, Giri D 2011. Disseminated histoplasmosis with concurrent oral candidiasis in an Eclectus parrot (Eclectus roratus). Avian Pathol 40: 207–211. [PubMed]Rappleye CA, Engle JT, Goldman WE 2004. RNA interference in Histoplasma capsulatum demonstrates a role for α(1,3)-glucan in virulence. Mol Microbiol 53: 153–165. [PubMed]Rappleye CA, Eissenberg LG, Goldman WE 2007. Histoplasma capsulatum α(1,3)-glucan blocks innate immune recognition by the β-glucan receptor. Proc Natl Acad Sci 104: 1366–1370. [PMC free article][PubMed]Raso TF, Werther K, Miranda ET, Mendes-Giannini MJ 2004. Cryptococcosis outbreak in psittacine birds in Brazil. Med Mycol 42: 355–362. [PubMed]Redecker D, Kodner R, Graham LE 2000. Glomalean fungi from the Ordovician. Science 289: 1920–1921. [PubMed]Rep M, Kistler HC 2010. The genomic organization of plant pathogenicity in Fusarium species. Curr Opin Plant Biol 13: 420–426. [PubMed]Ribes JA, Vanover-Sams CL, Baker DJ 2000. Zygomycetes in human disease. Clin Microbiol Rev 13: 236–301. [PMC free article] [PubMed]Richini-Pereira VB, Bosco Sde M, Theodoro RC, Macoris SA, Bagagli E 2009. Molecular approaches for eco-epidemiological studies of Paracoccidioides brasiliensis. Mem Inst Oswaldo Cruz 104: 636–643. [PubMed]Robert VA, Casadevall A 2009. Vertebrate endothermy restricts most fungi as potential pathogens. J Infect Dis 200: 1623–1626. [PubMed]Roden MM, Zaoutis TE, Buchanan WL, Knudsen TA, Sarkisova TA, Schaufele RL, Sein M, Sein T, Chiou CC, Chu JH, et al. 2005. Epidemiology and outcome of zygomycosis: A review of 929 reported cases. Clin Infect Dis 41: 634–653. [PubMed]Rodrigues ML, Nimrichter L, Oliveira DL, Frases S, Miranda K, Zaragoza O, Alvarez M, Nakouzi A, Feldmesser M, Casadevall A 2007. Vesicular polysaccharide export in Cryptococcus neoformans is a eukaryotic solution to the problem of fungal trans-cell wall transport. Eukaryot Cell 6: 48–59. [PMC free article] [PubMed]Rodrigues ML, Nakayasu ES, Oliveira DL, Nimrichter L, Nosanchuk JD, Almeida IC, Casadevall A 2008.Extracellular vesicles produced by Cryptococcus neoformans contain protein components associated with virulence. Eukaryot Cell 7: 58–67. [PMC free article] [PubMed]Rohde JR, Cardenas ME 2004. Nutrient signaling through TOR kinases controls gene expression and cellular differentiation in fungi. Curr Top Microbiol Immunol 279: 53–72. [PubMed]Rohm M, Grimm MJ, D’Auria AC, Almyroudis NG, Segal BH, Urban CF 2014. NADPH oxidase promotes neutrophil extracellular trap formation in pulmonary aspergillosis. Infect Immun 82: 1766–1777. [PMC free article] [PubMed]Saijo T, Chen J, Chen SC, Rosen LB, Yi J, Sorrell TC, Bennett JE, Holland SM, Browne SK, Kwon-Chung KJ 2014. Anti-granulocyte-macrophage colony-stimulating factor autoantibodies are a risk factor for central nervous system infection by Cryptococcus gattii in otherwise immunocompetent patients. mBio 5: e00912–14. [PMC free article] [PubMed]Santos WR, Meyer W, Wanke B, Costa SP, Trilles L, Nascimento JL, Medeiros R, Morales BP, Bezerra Cde C, Macedo RC, et al. 2008. Primary endemic Cryptococcosis gattii by molecular type VGII in the state of Para, Brazil. Mem Inst Oswaldo Cruz 103: 813–818. [PubMed]Schaller M, Borelli C, Korting HC, Hube B 2005. Hydrolytic enzymes as virulence factors of Candida albicans. Mycoses 48: 365–377. [PubMed]Schmitt HJ, Blevins A, Sobeck K, Armstrong D 1990. Aspergillus species from hospital air and from patients. Mycoses 33: 539–541. [PubMed]Sebghati TS, Engle JT, Goldman WE 2000. Intracellular parasitism by Histoplasma capsulatum: Fungal virulence and calcium dependence. Science 290: 1368–1372. [PubMed]Shankar J, Restrepo A, Clemons KV, Stevens DA 2011. Hormones and the resistance of women to paracoccidioidomycosis. Clin Microbiol Rev 24: 296–313. [PMC free article] [PubMed]Shapiro RS, Ryan O, Boone C, Cowen LE 2012. Regulatory circuitry governing morphogenesis in Saccharomyces cerevisiae and Candida albicans. Cell Cycle 11: 4294–4295. [PMC free article] [PubMed]Shen J, Cowen LE, Griffin AM, Chan L, Köhler JR 2008. The Candida albicans pescadillo homolog is required for normal hypha-to-yeast morphogenesis and yeast proliferation. Proc Natl Acad Sci 105: 20918–20923. [PMC free article] [PubMed]Shi M, Li SS, Zheng C, Jones GJ, Kim KS, Zhou H, Kubes P, Mody CH 2010. Real-time imaging of trapping and urease-dependent transmigration of Cryptococcus neoformans in mouse brain. J Clin Invest120: 1683–1693. [PMC free article] [PubMed]Shiraki Y, Ishibashi Y, Hiruma M, Nishikawa A, Ikeda S 2006. Cytokine secretion profiles of human keratinocytes during Trichophyton tonsurans and Arthroderma benhamiae infections. J Med Microbiol 55: 1175–1185. [PubMed]Siegfried N, van der Merwe L, Brocklehurst P, Sint TT 2011. Antiretrovirals for reducing the risk of mother-to-child transmission of HIV infection. Cochrane Database Syst Rev CD003510. [PubMed]Simwami SP, Khayhan K, Henk DA, Aanensen DM, Boekhout T, Hagen F, Brouwer AE, Harrison TS, Donnelly CA, Fisher MC 2011. Low diversity Cryptococcus neoformans variety grubii multilocus sequence types from Thailand are consistent with an ancestral African origin. PLoS Pathog 7: e1001343. [PMC free article] [PubMed]Skinner CE, Fletcher DW 1960. A review of the genus Candida. Bacteriol Rev 24: 397–416. [PMC free article] [PubMed]Smith RM, Schaefer MK, Kainer MA, Wise M, Finks J, Duwve J, Fontaine E, Chu A, Carothers B, Reilly A, et al. 2013. Fungal infections associated with contaminated methylprednisolone injections. N Engl J Med 369: 1598–1609. [PubMed]Soll DR 2002. Candida commensalism and virulence: The evolution of phenotypic plasticity. Acta Trop81: 101–110. [PubMed]Soll DR, Galask R, Schmid J, Hanna C, Mac K, Morrow B 1991. Genetic dissimilarity of commensal strains of Candida spp. carried in different anatomical locations of the same healthy women. J Clin Microbiol 29: 1702–1710. [PMC free article] [PubMed]Speed B, Dunt D 1995. Clinical and host differences between infections with the two varieties of Cryptococcus neoformans. Clin Infect Dis 21: 28–34; discussion 35–36. [PubMed]Spellberg B, Edwards J Jr, Ibrahim A 2005. Novel perspectives on mucormycosis: Pathophysiology, presentation, and management. Clin Microbiol Rev 18: 556–569. [PMC free article] [PubMed]Spellberg B, Andes D, Perez M, Anglim A, Bonilla H, Mathisen GE, Walsh TJ, Ibrahim AS 2009. Safety and outcomes of open-label deferasirox iron chelation therapy for mucormycosis. Antimicrob Agents Chemother 53: 3122–3125. [PMC free article] [PubMed]Springer DJ, Phadke S, Billmyre B, Heitman J 2012. Cryptococcus gattii, no longer an accidental pathogen? Curr Fungal Infect Rep 6: 245–256. [PMC free article] [PubMed]Staab JF, Bradway SD, Fidel PL, Sundstrom P 1999. Adhesive and mammalian transglutaminase substrate properties of Candida albicans Hwp1. Science 283: 1535–1538. [PubMed]Staib P, Kretschmar M, Nichterlein T, Hof H, Morschhäuser J 2000. Differential activation of a Candida albicans virulence gene family during infection. Proc Natl Acad Sci 97: 6102–6107. [PMC free article][PubMed]Steenbergen JN, Shuman HA, Casadevall A 2001. Cryptococcus neoformans interactions with amoebae suggest an explanation for its virulence and intracellular pathogenic strategy in macrophages. Proc Natl Acad Sci 98: 15245–15250. [PMC free article] [PubMed]Steenbergen JN, Nosanchuk JD, Malliaris SD, Casadevall A 2003. Cryptococcus neoformans virulence is enhanced after growth in the genetically malleable host Dictyostelium discoideum. Infect Immun 71: 4862–4872. [PMC free article] [PubMed]Steenbergen JN, Nosanchuk JD, Malliaris SD, Casadevall A 2004. Interaction of Blastomyces dermatitidis, Sporothrix schenckii, and Histoplasma capsulatum with Acanthamoeba castellanii. Infect Immun 72: 3478–3488. [PMC free article] [PubMed]Sudbery P, Gow N, Berman J 2004. The distinct morphogenic states of Candida albicans. Trends Microbiol 12: 317–324. [PubMed]Sundstrom P 2002. Adhesion in Candida spp. Cell Microbiol 4: 461–469. [PubMed]Taylor ML, Chavez-Tapia CB, Vargas-Yanez R, Rodriguez-Arellanes G, Pena-Sandoval GR, Toriello C, Perez A, Reyes-Montes MR 1999. Environmental conditions favoring bat infection with Histoplasma capsulatum in Mexican shelters. Am J Trop Med Hyg 61: 914–919. [PubMed]Thau N, Monod M, Crestani B, Rolland C, Tronchin G, Latgé JP, Paris S 1994. rodletless mutants of Aspergillus fumigatus. Infect Immun 62: 4380–4388. [PMC free article] [PubMed]Theodoro RC, Teixeira Mde M, Felipe MS, Paduan Kdos S, Ribolla PM, San-Blas G, Bagagli E 2012.Genus paracoccidioides: Species recognition and biogeographic aspects. PLoS ONE 7: e37694. [PMC free article] [PubMed]Torres HA, Rivero GA, Lewis RE, Hachem R, Raad, Kontoyiannis DP 2003. Aspergillosis caused by non-fumigatus Aspergillus species: Risk factors and in vitro susceptibility compared with Aspergillus fumigatus. Diagn Microbiol Infect Dis 46: 25–28. [PubMed]Trofa D, Soghier L, Long C, Nosanchuk JD, Gacser A, Goldman DL 2011. A rat model of neonatal candidiasis demonstrates the importance of lipases as virulence factors for Candida albicans and Candida parapsilosis. Mycopathologia 172: 169–178. [PubMed]Tucker SC, Casadevall A 2002. Replication of Cryptococcus neoformans in macrophages is accompanied by phagosomal permeabilization and accumulation of vesicles containing polysaccharide in the cytoplasm. Proc Natl Acad Sci 99: 3165–3170. [PMC free article] [PubMed]Underhill DM, Rossnagle E, Lowell CA, Simmons RM 2005. Dectin-1 activates Syk tyrosine kinase in a dynamic subset of macrophages for reactive oxygen production. Blood 106: 2543–2550. [PMC free article][PubMed]Untereiner WA, Scott JA, Naveau FA, Sigler L, Bachewich J, Angus A 2004. The Ajellomycetaceae, a new family of vertebrate-associated Onygenales. Mycologia 96: 812–821. [PubMed]Vallejo MC, Matsuo AL, Ganiko L, Medeiros LC, Miranda K, Silva LS, Freymuller-Haapalainen E, Sinigaglia-Coimbra R, Almeida IC, Puccia R 2011. The pathogenic fungus Paracoccidioides brasiliensisexports extracellular vesicles containing highly immunogenic α-galactosyl epitopes. Eukaryot Cell 10: 343–351. [PMC free article] [PubMed]Vallejo MC, Nakayasu ES, Matsuo AL, Sobreira TJ, Longo LV, Ganiko L, Almeida IC, Puccia R 2012.Vesicle and vesicle-free extracellular proteome of Paracoccidioides brasiliensis: Comparative analysis with other pathogenic fungi. J Proteome Res 11: 1676–1685. [PMC free article] [PubMed]Vikram HR, Smilack JD, Leighton JA, Crowell MD, De Petris G 2012. Emergence of gastrointestinal basidiobolomycosis in the United States, with a review of worldwide cases. Clin Infect Dis 54: 1685–1691. [PubMed]Vilela R, Rosa PS, Belone AF, Taylor JW, Diorio SM, Mendoza L 2009. Molecular phylogeny of animal pathogen Lacazia loboi inferred from rDNA and DNA coding sequences. Mycol Res 113: 851–857. [PubMed]Wang Y, Aisen P, Casadevall A 1995. Cryptococcus neoformans melanin and virulence: Mechanism of action. Infect Iimmun 63: 3131–3136. [PMC free article] [PubMed]Wasylnka JA, Moore MM 2003. Aspergillus fumigatus conidia survive and germinate in acidic organelles of A549 epithelial cells. J Cell Sci 116: 1579–1587. [PubMed]Webster RH, Sil A 2008. Conserved factors Ryp2 and Ryp3 control cell morphology and infectious spore formation in the fungal pathogen Histoplasma capsulatum. Proc Natl Acad Sci 105: 14573–14578. [PMC free article] [PubMed]Weissman Z, Shemer R, Conibear E, Kornitzer D 2008. An endocytic mechanism for haemoglobin-iron acquisition in Candida albicans. Mol Microbiol 69: 201–217. [PubMed]Weitzman I, Summerbell RC 1995. The dermatophytes. Clin Microbiol Rev 8: 240–259. [PMC free article][PubMed]Werner JL, Metz AE, Horn D, Schoeb TR, Hewitt MM, Schwiebert LM, Faro-Trindade I, Brown GD, Steele C 2009. Requisite role for the dectin-1 β-glucan receptor in pulmonary defense against Aspergillus fumigatus. J Immunol 182: 4938–4946. [PMC free article] [PubMed]Wheeler RT, Fink GR 2006. A drug-sensitive genetic network masks fungi from the immune system. PLoS Pathog 2: e35. [PMC free article] [PubMed]Wieloch W, Bogus MI, Ligeza M, Koszela-Piotrowska I, Szewczyk A 2011. Coronatin-1 isolated from entomopathogenic fungus Conidiobolus coronatus kills Galleria mellonella hemocytes in vitro and forms potassium channels in planar lipid membrane. Toxicon 58: 369–379. [PubMed]Williams MJ 2007. Drosophila hemopoiesis and cellular immunity. J Immunol 178: 4711–4716. [PubMed]Windus DW, Stokes TJ, Julian BA, Fenves AZ 1987. Fatal Rhizopus infections in hemodialysis patients receiving deferoxamine. Ann Intern Med 107: 678–680. [PubMed]Wrobel L, Whittington JK, Pujol C, Oh SH, Ruiz MO, Pfaller MA, Diekema DJ, Soll DR, Hoyer LL 2008.Molecular phylogenetic analysis of a geographically and temporally matched set of Candida albicansisolates from humans and nonmigratory wildlife in central Illinois. Eukaryot Cell 7: 1475–1486. [PMC free article] [PubMed]Wu W, Lockhart SR, Pujol C, Srikantha T, Soll DR 2007. Heterozygosity of genes on the sex chromosome regulates Candida albicans virulence. Mol Microbiol 64: 1587–1604. [PubMed]Wu Y, Yang J, Yang F, Liu T, Leng W, Chu Y, Jin Q 2009. Recent dermatophyte divergence revealed by comparative and phylogenetic analysis of mitochondrial genomes. BMC Genomics 10: 238. [PMC free article] [PubMed]Youseff BH, Dougherty JA, Rappleye CA 2009. Reverse genetics through random mutagenesis in Histoplasma capsulatum. BMC Microbiol 9: 236. [PMC free article] [PubMed]Zaragoza O, Garcia-Rodas R, Nosanchuk JD, Cuenca-Estrella M, Rodriguez-Tudela JL, Casadevall A 2010. Fungal cell gigantism during mammalian infection. PLoS Pathog 6: e1000945. [PMC free article][PubMed]Zavasky DM, Samowitz W, Loftus T, Segal H, Carroll K 1999. Gastrointestinal zygomycotic infection caused by Basidiobolus ranarum: Case report and review. Clin Infect Dis 28: 1244–1248. [PubMed]Zimmerman LE 1955. Fatal fungus infections complicating other diseases. Am J Clin Pathol 25: 46–65. [PubMed]Zugmaier W, Bauer R, Oberwinkler F 1994. Mycoparasitism of some Tremella species. Mycologia 86: 49–56.