Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Dec 22.
Published in final edited form as: FEMS Microbiol Lett. 2008 Apr 16;283(2):129–139. doi: 10.1111/j.1574-6968.2008.01160.x

Animal models of mucosal Candida infection

Julian R Naglik 1, Paul L Fidel Jr 2, Frank C Odds 3
PMCID: PMC3244615  NIHMSID: NIHMS342119  PMID: 18422625

Abstract

Rodent models of oral, vaginal and gastrointestinal Candida infection are described and discussed in terms of their scientific merits. The common feature of all experimental mucosal Candida infections is the need for some level of host immunocompromise or exogenous treatment to ensure quantitatively reproducible disease. A growing literature describes the contributions of such candidiasis models to our understanding of certain aspects of fungal virulence and host response to mucosal Candida albicans challenge. Evidence to date shows that T-lymphocyte responses dominate host immune defences to oral and gastrointestinal challenge, while other, highly compartmentalized responses defend vaginal surfaces. By contrast the study of C. albicans virulence factors in mucosal infection models has only begun to unravel the complex of attributes required to define the difference between strongly and weakly muco-invasive strains.

Keywords: animal, mucosal, epithelium, oral, model, vaginal

Introduction

Candida species are ubiquitous fungal pathogens and are the most common cause of mucosal and invasive fungal infections in humans. Candida albicans, the leading cause of candidiasis, is a polymorphic fungus that reproduces by budding and commonly resides as a commensal in the mucosal tissues of approximately half the human population. When the balance of the normal flora is disrupted or the immune defences are compromised, Candida species can become pathogenic, often causing recurrent disease in susceptible individuals. As a result, Candida infections are recognized as a serious public health challenge with high medical and social-economical importance. Given the relatively limited number of suitable and effective antifungal drugs, the continuing increase in the incidence of Candida infections highlights the need to identify the fundamental pathogenic determinants of C. albicans and the reciprocal host protection mechanisms against this fungus at mucosal surfaces (Naglik et al., 2003).

The study of human infectious diseases requires the investigation of microorganisms in model systems. For pathogenic fungi, a number of invertebrate mini-host systems including Drosophila melanogaster, Acanthamoeba, Caenorhabditis elegans, Dictyostelium discoideum, and Galleria mellonella (Chamilos et al., 2007; Mylonakis et al., 2007) have been developed to study epithelial infections and cellular or humoral responses. Likewise, epithelial organotypic models have been developed for close analysis of the host-fungal interaction at mucosal surfaces (Dongari-Bagtzoglou & Kashleva, 2006; Schaller et al., 2006). These nonvertebrate models offer a number of advantages over mammalian vertebrate models, predominantly due to economy of size and ethical issues (Table 1). However, vertebrate models are invaluable systems for the molecular and cellular analysis of host-fungal interactions as the animals and their environment can be precisely controlled and manipulated to permit thorough investigations of fungal pathogenicity and immunity. Although many of the pioneering studies on mucosal candidiasis were performed in non-human primates (Samaranayake & Samaranayake, 2001), small mammals, including rats and mice, are the common choice for such studies for economical and ethical reasons and because of their relative anatomic and immunological similarity to humans (Samaranayake & Samaranayake, 2001; Chamilos et al., 2007).

Table 1.

Benefits and limitations of vertebrate and nonvertebrate mini-host models in studying the pathogenesis of Candida infections

Model system Benefits Limitations
Nonvertebrate mini-host Host and environment can be precisely controlled
Reproducibility
Low cost, simplicity, rapid results
High throughput preliminary screening for strain virulence and responses to antifungal agents
Well characterized genetics and immunity that can be manipulated to study host–Candida interactions
Many innate immune mechanisms conserved between invertebrates and mammals, permitting limited investigation of innate responses to Candida
Fewer ethical concerns
Precise inocula can be difficult
Molecular and cellular host mechanisms may not be evolutionarily conserved with mammalian species
Absence of most mammalian compartments and immune cells makes models over-simplistic (e.g. lack of adaptive immunity)
Mammalian specific pathogens and virulence determinants can not be investigated/identified
Do not provide a good substrate for modelling superficial infections
Effects of antifungal agents similar to in vitro tests
Many model hosts require experimentation at low temperatures (<30 °C)
Vertebrate (mouse/rat) Host and environment can be precisely controlled
Evolutionarily closer to humans, with anatomical and immunological similarity: host–Candida interactions more relevant to humans
Allow development of superficial and disseminated infection models
Amenable to genetic studies: knock-out and knock-in gene studies allow investigation of gene (product) contribution to the whole infectious process
Precise inocula via specific routes of inoculation
Large scale studies are time-consuming and prohibitive on cost
Severe ethical constraints
Physiologic and anatomic complexity
Quantitative outcome reproducibility a problem: usually overcome by using conditions and challenge doses that are not representative of natural human infections
Potentially difficult antifungal pharmacokinetics in some species mean responses may not be extrapolated to effects in humans
Long reproductive cycles

A number of clinically relevant rodent models of systemic and mucosal candidiasis have been established, predominantly to study host-pathogen interactions, anti-fungal drug efficacy and pharmacokinetics, and vaccine candidates. Systemic models rely on intravenous injection of C. albicans, which is generally an atypical route of infection and will not be covered in this review. Mucosal (oral) inoculation on the other hand more closely mimics the predominant gastrointestinal portal of entry leading to systemic dissemination. However, establishment of mucosal infection models (oral, gastrointestinal, vaginal) generally requires the use of immunosuppressive agents, antibiotic or oestrogen treatment, or the use of germ-free or transgenic animals (Fidel & Sobel, 1999; Samaranayake & Samaranayake, 2001; de Repentigny, 2004). Consequently, when interpreting data from such animal models, one needs to consider the severity of the manipulations before drawing conclusions.

One caveat to studying C. albicans–host interactions in rodents is that C. albicans is not a natural colonizer of mucosal surfaces in these animals - the rodent equivalent of ‘normal flora yeast’ is Candida pintolopesii (Savage &Dubos, 1967), which can sometimes cause infections in immuno-compromised rodents (Kurtzman et al., 2005). This has both benefits and limitations. The advantage is that any host response to C. albicans is not affected by pre-existing adaptive immune responses to the fungus. The disadvantage is that, as mentioned above, establishment of mucosal colonization or infection usually requires intervention with immunosuppressive agents, antibiotics or oestrogen. Nevertheless, experimental rodent models have been enormously valuable and our specific emphasis will be on the utility of published models for the study of host-pathogen interactions in superficial C. albicans infections.

General aspects of murine and rat mucosal models

The choice of rodent for experimental mucosal Candida infection needs to take into account many economic, scientific and ethical considerations (Table 2). In general, the larger the animal host the easier it is to collect repeated samples of fluids and some tissues - a factor which becomes important in experiments designed to monitor, for example, serological parameters over time. However, the higher cost of maintenance and the greater difficulties in handling associated with larger rodent species often favour the use of small rodents, particularly mice. To date, no rodent model of invasive mucosal Candida infection has been devised that does not depend on some form of predisposition of the animal by occlusion, immunosuppression, surgical alteration, or elimination of competing microbial flora: often more than one of these conditions. However, commensal C. albicans carriage can be induced in unmodified hosts (Samaranayake & Samaranayake, 2001).

Table 2.

Comparison of rat vs. mouse mucosal models in studying host–Candida interactions

Mouse
Rat
Advantage Disadvantage Advantage Disadvantage
General Cost
Species-specific reagents No adaptive immune response to Candida in naïve state Availability and ease of production of genetically modified strains
Not colonized with C. albicans like humans No adaptive immune response to Candida in naïve state Not colonized with C. albicans like humans
Lack of some species-specific reagents
Vaginal model Not mouse strain dependent Vaginal lavage to quantify fungal burden
No vaginal CD8 T cells Efficacy/predictability of drug studies
Parallel to human infection
Requirement for pseudoestrus Too robust with estrogen; not robust enough without pH (neutral)
Lack of symptoms
Short half-life of drugs
Repeat vaginal lavage during longitudinal studies
Spontaneous clearance of infection.
Longitudinal evaluation Vaginal CD8 T cells
Efficacy/predictability of drug studies
Requirement for pseudoestrus pH (neutral)
Lack of symptoms
Oophorectomy to establish pseudoestrus
Oral model Ease of inoculation (cotton swab) Need for immunosuppression to establish persistent infection
Poor for vaccine efficacy studies
Ease of inoculation (cotton swab) Need for immunosuppression to establish persistent infection
Poor for vaccine efficacy studies
GI model Fecal specimen quantification of fungal burden Lack of persistent infection as detected by fecal specimen Not characterized Not characterized

Rodent vaginitis models are highly dependent upon a prolonged state of pseudo-oestrus, which is usually induced and maintained with weekly administration of 17-β-estrodiol (Fidel & Sobel, 1999; Clemons et al., 2004) (Table 2). Additionally, rats must be oophorectomized to obtain a pseudo-oestrus state. Vaginitis models without oophorectomy or the use of oestrogen have been developed, but these are rare and are based on immunosuppression (Tan et al., 2005) or antibiotic treatment (Hamad et al., 2006). Oestrogen levels appear to be the main factor affecting host susceptibility to C. albicans and usually a relatively high dose of 100 µg per mouse per week is used to establish infection for 5 weeks or more, although an oestrogen range of 10–500 µg per mouse per week have been reported. In the absence of pseudo-oestrus vaginal infections are usually cleared by the second week (Fidel & Sobel, 1999). Oestrogen transforms the columnar epithelium into thicker stratified squamous epithelium and increases the glycogen content, pH and growth substrates, all of which facilitate C. albicans avidity for the tissue and growth (Fidel & Sobel, 1999). Oestrogen may also inhibit innate and/or adaptive immune defences, thus permitting an infection phenotype (Styrt & Sugarman, 1991). In addition, C. albicans is known to produce an oestrogen-binding protein (Skowronski & Feldman, 1989) and so addition of mammalian oestrogens might also directly affect fungal colonization or pathogenicity (O’Connor et al., 1998). Because most mouse strains (BALB/c, C57BL/6, DBA/2, CBA/J, C3H/HEN) are susceptible to oestrogen-induced C. albicans vaginitis, susceptibility to vaginal candidiasis appears to be independent of the major histocompatibility locus H-2 haplotype (Calderon et al., 2003). However, one exception is the CD-1 mouse, which is innately more resistant to candidal infection, probably due to its resistance to oestrodiol endocrine disruption (Spearow et al., 1999).

Many oral C. albicans infection models have been described, with persistent infections usually requiring some form of immunosuppression or intervention therapy (Samaranayake & Samaranayake, 2001)) (Table 2). Historically, the rat was used far more often than the mouse as a host for experimental oral Candida infections. Many early studies focused on models in which an acrylic device implanted in the animal’s mouth provided the occlusion necessary to establish an infection resembling denture stomatitis. Alternatively, antibacterial treatments predisposed the animals to infection. Without some form of immunosuppression or other manipulation, oral fungal burdens in mice and rats are variable and often decline rapidly. However, it has been demonstrated in both mice (Lacasse et al., 1993) and rats (Jones & Russell, 1973) that after an initial oral infection is established over c. 1 week, fungal burdens can reduce naturally to a carrier state for many weeks without the use of immunosuppressive agents. To establish oral candidiasis in mice, animals are usually treated with steroids (Deslauriers et al., 1995), often with the addition of antibiotics such as tetracycline (Kamai et al., 2001). Although fungal burdens can be initially high, burdens diminish to very low levels after cessation of drug treatment (Deslauriers et al., 1995).

One model that has been lacking, but which recently has been developed, is a model of concurrent oral and vaginal colonization in the same mouse, which has the added ethical benefit of reducing the required numbers of experimental mice by half (Rahman et al., 2007). This model appears successful due to the use of a fresh clinical oral strain of C. albicans (529L) and the administration of low doses of oestrogen (5 µg) via both intramuscular and subcutaneous routes. Colonization with C. albicans 529L was shown to be relatively high over a 5–6 week period and similar to that generally found during human mucosal colonization. Manipulation of this model to induce an infection state would be highly desirable, and would permit the detailed investigation of host-Candida interactions that exist during both the colonization and infection states at multiple sites.

Gastrointestinal candidiasis in humans often arises in patients treated with high dose immunosuppressants or cancer chemotherapy (Pappas et al., 2004). To mimic this condition, a number of gastrointestinal models have been developed in the mouse, many of which have been used to test the efficacy of antifungal drugs (Clemons et al., 2006; Capilla et al., 2007). Gastrointestinal infection models in the rat have occasionally been described, including models in which some animals developed measurable visceral burdens (Wong et al., 1990). The advantage of the larger rodent model in permitting repeated blood sampling for diagnostic purposes was well demonstrated in these studies. Establishing a model of disseminated candidiasis that arises from translocation of C. albicans across the gastrointestinal mucosa has been more challenging, but has been created in infant mice (aged 4–6 days), in which dissemination of C. albicans to the liver kidney and spleen could be detected up to 72 h after intra-gastric inoculation (Pope et al., 1979). However, the inconsistent visceral dissemination and low visceral fungal burdens arising in this model are typical of attempts to devise gut translocation systems in a variety of hosts. A mouse model recently developed using a specific regime of antibiotics and 5-fluorouracil appears more promising (Clemons et al., 2006), and seems useful not only for drug efficacy studies but also for fungal pathogenicity studies. Further developments in this area are necessary to advance our understanding of host-pathogen interactions at the gastrointestinal mucosa.

Germ-free or gnotobiotic mice appear uniquely suited for studies of mucosal candidiasis, because mucosal surfaces can be naturally and chronically colonized by C. albicans without the need for trauma, immunosuppressive or antimicrobial therapy. In addition, colonization can persist for the lifetime of the animal (Balish et al., 1990; Rahman & Challacombe, 1995). However, the germ-free/gnotobiotic model is not ideal for the study of fungal dissemination from mucosal sites or natural host-pathogen interactions, because the absence of the normal microbiota is likely to have a major effect on both fungal pathogenicity and the host response.

The utilization of transgenic or congenitally immunodeficient mice has been instrumental in advancing our understanding of the critical roles of CD4+ (helper) T cells, CD8+ (cytotoxic) T cells, γδT cells, polymorphonuclear leukocytes (PMNs), macrophages, and cytokines in host defence against mucosal candidiasis. These data indicate that protection against mucosal candidiasis involves the cooperation of several immune cell populations and molecules, which together can prevent invasion of mucosal surfaces by C. albicans in the normal host. It is thus evident that multiple, rather than single, defects in host defence mechanisms potentially underlie mucosal candidiasis.

Study of Candida pathogenesis in mucosal animal models

In humans, isolation of Candida species from mucosal tissues does not indicate a diseased state and over the past decade it has become clear that it is the physiological status of the host that is the primary factor governing the aetiology of candidiasis. However, it appears that the transition from harmless commensal to unrelenting pathogen is a fine line and one that is attributable to an extensive repertoire of virulence determinants selectively expressed under suitable predisposing conditions (Naglik et al., 2003). Animal models have been used to determine the importance and relative contribution of C. albicans pathogenic attributes to in vivo infections through a variety of experimental approaches, including gene expression (reverse-transcription PCR and in vivo expression technology), protein inhibition, and use of C. albicans mutant strains (gene disruption/knockouts) (Naglik et al., 2003; de Repentigny, 2004; Brown et al., 2007; Hoyer et al., 2007).

The majority of these studies have been performed using either in vitro models or systemic intravenous models of infection; significantly fewer have been undertaken in animal models of mucosal infection. This is likely due to the relative difficulty in establishing and maintaining mucosal models of infection in comparison with systemic models and underscores the absence of a clear, universally reproducible mucosal model to parallel the widely used disseminated model based on intravenous challenge. One of the most common and best-accepted approaches for identifying virulence genes in C. albicans is to create gene-disrupted mutant strains and to assess them in vivo. A large number of genes have been disrupted in C. albicans but only a minority have been investigated in mucosal infection models (Table 3): most of the genes listed are better known as putative virulence factors in systemic infection models. Therefore, many of the factors that have been concluded to be pathogenic determinants during systemic infections have yet to be confirmed as pathogenic determinants during mucosal infections. For example, we have previously demonstrated that several clinical mucosal isolates were less virulent than laboratory isolates in a systemic model but similarly virulent in a vaginal model of infection model (Taylor et al., 2000a). As such, our current understanding of mucosal Candida infections in vivo remains relatively elementary. More detailed investigations using animal mucosal models will be essential to confirm and to identify novel virulence factors during epithelial infections.

Table 3.

Animal mucosal models used to study Candida pathogenicity in ‘molecular Koch’s postulates’ experimental designs

Animal model Gene Biological function Result of gene disruption Reintegrant
tested?
References
Mouse oral ALS1 Surface glycoprotein Lower counts early in infection Y Kamai et al. (2002)
Mouse oral CKA1/2 Protein kinase Reduced virulence Y Chiang et al. (2007)
Mouse oral/gastric PLB1 Phospholipase B Reduced ability to cross gut wall Y Mukherjee et al. (2001)
Mouse oral/gastric PLD1 Phospholipase D Reduced virulence N Dolan et al. (2004)
Mouse gastrointestinal GPI7 Involved in GPI anchor attachment Reduced colonization N Richard et al. (2002)
Mouse gastrointestinal EFH1 Transcription factor involved in hypha formation Increased colonization Y White et al. (2007)
Rat oral ADE2 Nucleotide synthesis Reduced oral colonization N Cole et al. (1995)
Rat oral FAS2 Lipid biosynthesis Reduced oral colonization Y Zhao et al. (1996)
Rat oral URA3 Nucleotide synthesis Reduced oral colonization N Cole et al. (1995)
Piglet oral EFG1 Transcription factor involved in hypha formation Reduced virulence Y Riggle et al. (1999)
Mouse vaginal CSH1 Surface hydrophobicity Reduced colonization Y Singleton et al. (2005)
Mouse vaginal NOT5 Required for hypha formation No effect Y Cheng et al. (2005)
Rat vaginal HK1 Two-component histidine kinase No effect Y Calera et al. (1999)
Rat vaginal MNT1 Mannosyl transferase Reduced virulence N Buurman et al. (1998)
Rat vaginal PHR1 pH-regulated gene No effect Y DeBernardis et al. (1998)
Rat vaginal PHR2 pH-regulated gene Essential for virulence Y DeBernardis et al. (1998)
Rat vaginal SAP1 Secreted proteinase Reduced virulence N DeBernardis et al. (1999)
Rat vaginal SAP2 Secreted proteinase Reduced virulence Y DeBernardis et al. (1999)
Rat vaginal SAP3 Secreted proteinase Reduced virulence N DeBernardis et al. (1999)
Rat vaginal SAP4-6 Secreted proteinase No effect N DeBernardis et al. (1999)

One of the reasons why virulence studies in animal mucosal models are sparse is because of the use of C. albicans strain SC5314. Although almost universally utilized in systemic pathogenicity studies and other molecular-based investigations, SC5314 has limited use in mucosal pathogenicity studies because it is a very poor colonizer of epithelial surfaces in vivo, and particularly of mice (Taylor et al., 2000a; Rahman et al., 2007). Given that nearly all current C. albicans mutants are created in the CAF2-1, CAI4, BWP17 background, all of which derive from the wild type strain SC5314, potential errors are likely to be made in concluding a role (or lack of a role) for specific genes during mucosal C. albicans infections. To determine the role of putative virulence genes in mucosal infections more comprehensively, gene-disruption mutants would need to be created (or recreated) in a number of clinical wild-type strains known to colonize or infect mucosal surfaces, such as C. albicans strain 529L (Rahman et al., 2007). Utilizing these ‘mucosal’ C. albicans strains provides plenty of scope to identify novel genes, proteins, and pathways that might be specifically or preferentially required to colonize and infect epithelial cells in vivo. This is particularly relevant to oral and vaginal infections where stresses and environmental signals (e.g. temperature, pH, alternative nutrients, osmotic stress, oxygen concentration) are quite different to each other, let alone from those found during systemic infections.

Apart from the use of SC5314, other problems in mucosal animal experimentation are that the read-out is often based solely on fungal burdens. Very few studies include histological analysis, which would allow anatomical and morphological interactions to be assessed more closely (for example, a virulent strain may cause damage without multiplying extensively in the organ), or analysis of the host response, which would determine whether deficiencies in these putative virulence genes have a direct effect on how the host recognizes or responds to the fungus. This is a common problem of many studies in general which often target the fungus or the host, rather than investigating the two together. Other shortcomings include the effect of Candida preculture growth, the route of inoculation, innate differences between C. albicans strains, and the need to include both the disrupted mutant and the reintegrant strains, which is often lacking in animal pathogenicity studies (Table 3). Together, these data emphasize the need to develop current models and to establish more sophisticated models of C. albicans colonization and infection in order to advance our understanding of C. albicans pathogenicity at mucosal surfaces.

Study of Candida immunity in mucosal animal models

Host defence against C. albicans and most microorganisms fall into three categories: nonspecific (mucosal barrier, commensal bacteria, natural secretions), innate immunity based on PMN leukocytes and macrophages, and adaptive immune mechanisms based on T- and B-lymphocytes. Although mucosal animal models have not been utilized to their potential to study Candida pathogenesis, their use has been instrumental in deciphering the roles of specific immune components against Candida infections (Table 4). Animal studies have taught us that host defence against mucosal infections involves several arms of the immune response, that protective mechanisms differ in the vagina compared with oral/gastrointestinal, and that vaginal immune responses are compartmentalized and highly regulated.

Table 4.

Host defences against experimental mucosal Candida infections

Mucosal site (model)
Host defence mechanism
Innate
GI Oral Vaginal Reference (reviews)
PMNs +/− (ms*) +(ms) − −/++ (ms) Fidel & Finkel-Jimenez (2006); Fidel (2007)
Macrophage +/− (ms) +(ms/rat) − − (ms) Fidel & Finkel-Jimenez (2006); Fidel (2007)
Natural killer cells − − (ms) ? (ms) − − (ms) Fidel & Finkel-Jimenez (2006); Fidel (2007); Westwater et al. (2007)
Dendritic cells ++ ? ++§
++ (rat)
Fidel & Finkel-Jimenez (2006); Fidel (2007)
Epithelial cells ? ? ++ (ms/monkey) Fidel & Finkel-Jimenez (2006); Fidel (2007)
Humoral
 IgM − − (ms) ? + (ms) Fidel & Finkel-Jimenez (2006); Fidel (2007)
 IgG − − (ms) ? +/− (rat/ms) Fidel & Finkel-Jimenez (2006); Fidel (2007)
 IgA − − (ms) ? + (rat/ms) Fidel & Finkel-Jimenez (2006); Fidel (2007)
Cell-mediated
 CD4+ T cells (Th1) ++ (ms) +++(ms) +/− (rat/ms) Fidel & Finkel-Jimenez (2006); Fidel (2007)
 CD8+ T cells (cytotoxic) +(ms) ++ (ms) − − (rat/ms) Fidel & Finkel-Jimenez (2006); Fidel (2007)
 Treg/Th17 ? ? + (Treg) Fidel & Finkel-Jimenez (2006); Fidel (2007)

− −, fully negative; +/−, evidence for and against in the respective model in brackets (exception is ); +, ++, +++, different degrees of positive involvement; ?, unknown involvement at this stage.

*

Ms, mouse.

Activity is associated with susceptibility to infection rather than protection (unpublished, Fidel) parallel to the clinical condition (Fidel et al., 2004a).

Not based on the models, but from conventional research efforts.

§

Plamacytoid dendritic cells involved in immunoregulation.

Shown to be protective.

May be innate response rather than adaptive.

Strong evidence exists indicating that cellular immunity, mediated by CD4+ Th1 cells, protects against oral and gastrointestinal C. albicans infection. This was demonstrated experimentally in animals using congenitally immunodeficient, transgenic mice and virus-infected mouse models (Table 4) and clinically in humans, in which various immunocompromised population groups (e.g. AIDS, transplant and steroid therapy) are more susceptible to oropharyngeal candidiasis (Klein et al., 1984). Additionally, CD4+ T cell protection appears to work in combination with certain cytokines, including IL-12, IFN-γ, IL-4, TNF-α, and nitric oxide (Elahi et al., 2000, 2001; Farah et al., 2002a, 2002b). During murine vaginitis, a Th1-type Candida-specific systemic cell mediated immune (CMI) response can also be generated. However, unlike in the oral mucosa, the systemic CMI response does not appear to be the predominant protective defence mechanism against C. albicans vaginal infections, as no correlation was observed between vaginal T-helper, proinflammatory cytokines and fungal burden (Fidel, 2007). Rather, local mucosal immunity distinct from that in the peripheral circulation, together with immunoregulation by γδ T cells and dendritic cells (DCs), is now under consideration as the main immune mechanism governing at the vaginal mucosa (Fidel, 2007) (Table 4). The role of CD8+ T cells in protection against mucosal infection is more contentious than for CD4+ cells. In rats, CD8+ cells are more numerous than CD4+ cells in the vagina (similar to humans), whereas in mice CD4+ T cells are exclusive (Cassone et al., 2007). Irrespective, the current data indicate that CD8+ T cells may play protective roles orally and gastrointestinally, but not vaginally (Table 4).

Importantly, T cells alone do not appear to protect against systemic candidiasis of mucosal origin because neither ν/ν (T cell deficient) nor ν/+ (T cell competent) mice develop progressive systemic disease (Balish et al., 1990). Protection against systemic candidiasis of mucosal (oral/gastrointestinal) origin is predominantly mediated by phagocytic cells (PMNs and macrophages) (Cantorna & Balish, 1990). This is supported by a recent study which demonstrated protection from gastrointestinal transmission of C. albicans across the mucosa by bombesin (Algin et al., 2005), a 14 amino acid peptide from frogs that stimulates all steps of the phagocytic process in murine peritoneal macrophages. Interestingly, PMN do not play an obvious protective role in the vaginal lumen (Table 4), and indeed in humans PMNs appear to exacerbate the vaginal disease (Fidel et al., 2004b). The reason for this is as yet unknown. Nonetheless, animal data strongly indicate a fundamental role for PMNs in protection against systemic candidiasis of mucosal (oral/gastrointestinal) origin.

T regulatory (Treg) and Th17 cells (T cells that secrete IL-17) have recently emerged as important immune cells controlling immune responses to Candida infections (Netea et al., 2004; Yu & Gaffen, 2008). There is evidence for the presence of CD25+ CD4+ Treg cells in the vagina (Wormley et al., 2001) and the Th17 pathway is activated in response to disseminated C. albicans, which is thought to negatively regulate the Th1-mediated resistance pathway (Zelante et al., 2007; Huang et al., 2004). However, the precise functional roles of Treg and Th17 cells during mucosal fungal disease remain unclear. This is one area in which animal mucosal models can be specifically utilised to answer crucial questions concerning the roles of these two T cell types against superficial C. albicans infections.

Other immune cell types include DC’s, which represent the crucial link between innate and adaptive immunity and orchestrate cell mediated and humoral defence mechanisms. DC’s contribute to protection or immunoregulation against C. albicans infection in the gastrointestinal or vagina, respectively, but their protective role in oral tissues is undefined (Table 4) (De Bernardis et al., 2006). Conversely, natural killer cells are not thought to convey strong protection at any mucosal site (Table 4). Currently, there also is a lack of evidence concerning the protective role of naturally induced antibodies against mucosal Candida infection. However, although antibody levels against C. albicans appear unaltered irrespective of the disease state, there is good evidence from immunization studies that a limited subset of antibodies are able to protect against mucosal Candida infection (Table 4). This suggests that animal models have been useful in identifying immunotherapeutic and neutralizing antibodies, which could potentially be generated for clinical use. However, a more detailed analysis of the functional characteristics of these protective antibodies and their target epitopes is required.

More recently, epithelial cells and cytokines have emerged as potentially protecting and/or modulating the local immune response during Candida infections (Dongari-Bagtzoglou & Fidel, 2005; Fidel, 2007). Epithelial cells secrete high levels of the immunoregulatory cytokine TGF-β that promotes antibody class switching to IgA (the protective mucosal antibody) (Taylor et al., 2000b), numerous proinflammatory cytokines (Schaller et al., 2002; Dongari-Bagtzoglou & Fidel Jr., 2005) and antifungal peptides (Cole, 2006; Kazmi et al., 2006). Indeed, vaginal epithelial cells from mice and non-human primates are able to inhibit Candida growth in vitro (Table 4), and oral epithelial cells collected from saliva of humans have similar activity (Fidel, 2006). However, an oral epithelial-mediated inhibitory effect has not yet been demonstrated in animal models. In addition, in an in vitro model, human epithelial cells can also protect the oral mucosa from C. albicans infection directly through epithelial Toll-like receptor 4 (TLR4) via a process mediated by PMNs and TNFα (Schaller et al., 2004; Weindl et al., 2007). The protective role of TNFα at mucosal surfaces supports other clinical human data (Moen et al., 2005) but also animal data, in which oral administration of recombinant TNFα reduces fungal burdens (Ohta et al., 2007) and where TNFα knockout mice are more susceptible to oral C. albicans infection (Farah et al., 2006). The role of cytokines/chemokines in the protection against vaginal candidiasis is uncertain, but monocyte chemotactic protein-1(MCP-1) appears to play some role in reducing the fungal burden during vaginal infection as intravaginal neutralization of MCP-1 with antibodies results in a significant increase in vaginal fungal burden during infection (Saavedra et al., 1999). Given that the mucosal epithelium is the primary cell layer that initially encounters Candida, it is paramount that animal models are utilised to investigate the importance of epithelial cells and their associated innate factors in host defence.

Summary comments and future directions

Experimental animal models of mucosal candidiasis have been invaluable in assessing fungal pathogenicity and host immune defences. These studies have informed us that C. albicans is the most virulent Candida species in vivo and that the immune response to Candida is different at the various mucosal sites and is compartmentalized and highly immunoregulatory in the vagina. These findings, in turn, have highlighted the gaps in our knowledge of Candida pathogenicity and host immunity and provide clues as to how they might be addressed. To more accurately determine the role of the host in the host-pathogen interaction, a model is required in which specific host proteins or processes can be manipulated in order to permit either colonization or disease in the same animal. Likewise, to obtain a more global picture of why the immune response is distinct at different mucosal sites, various arms of the immune response (innate, CMI, humoral) need to be investigated at different mucosal sites (oral, vaginal and gastrointestinal) within the same animal strain. All too often only a single arm of the immune response is assessed and a single model utilized.

Specific areas for future research include the identification of surface recognition receptors on epithelial cells that bind Candida and initiate the innate immune response. Currently, only E-cadherin has been identified as an epithelial receptor for C. albicans (via Als3) permitting mucosal invasion (Phan et al., 2007). Although a number of receptors are known to recognize C. albicans on other immune cells [TLRs, dectin-1, dendritic cell-specific intercellular adhesion molecule 3 grabbing nonintegrin (DC-SIGN), mannose receptor, complement receptor 3, galectin-3], these have not yet been confirmed as genuine receptors on epithelial cells that can initiate immune responses. Experimental mucosal models using animals deficient in these receptors are necessary to determine their precise role in fungal recognition at epithelial surfaces. In addition, the signalling pathways in epithelial cells that orchestrate the subsequent immune response are undefined. Deciphering these pathways should provide vital clues as to how C. albicans is detected and whether the fungus manipulates epithelial signalling systems for its own benefit (e.g. to permit colonization). Moreover, little is known about the identity of the inhibitory proteins that prevent signalling activation in epithelial cells. This might provide an explanation for the considerable amount of immunoregulation that is observed at mucosal surfaces, and specifically in the vagina. Other areas that need to be explored are the precise roles of the Th1/Th2/Th17/Treg pathways in defence against mucosal diseases, and the interaction between epithelial cells and other cells of the immune system. This will be greatly facilitated by the use of transgenic or knockout mice, together with targeted investigation of the fungal cell wall and fundamental biological processes such as hypha formation.

Over the past two decades we have come far in our understanding of host-Candida interactions at mucosal surfaces. Over the next decade the fungal community can take advantage of advances in molecular, cellular, immunological and genomic/proteomic technologies, as well as mutant strain libraries and systems biology approaches in relevant animal models to more fully decipher the complex interactions that take place between this eukaryotic ‘opportunistic commensal pathogen’ and its mammalian host.

References

  1. Algin C, Sahin A, Kiraz N, Sahinturk V, Ihtiyar E. Effectiveness of bombesin and Saccharomyces boulardii against the translocation of Candida albicans in the digestive tract in immunosuppressed rats. Surg Today. 2005;35:869–873. doi: 10.1007/s00595-005-3049-9. [DOI] [PubMed] [Google Scholar]
  2. Balish E, Filutowicz H, Oberley TD. Correlates of cell-mediated immunity in Candida albicans-colonized gnotobiotic mice. Infect Immun. 1990;58:107–113. doi: 10.1128/iai.58.1.107-113.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Brown AJP, Odds FC, Gow NAR. Infection-related gene expression in Candida albicans. Curr Opin Microbiol. 2007;10:307–313. doi: 10.1016/j.mib.2007.04.001. [DOI] [PubMed] [Google Scholar]
  4. Buurman ET, Westwater C, Hube B, Brown AJ, Odds FC, Gow NAR. Molecular analysis of CaMnt1p, a mannosyl transferase important for adhesion and virulence of Candida albicans. Proc Natl Acad Sci USA. 1998;95:7670–7675. doi: 10.1073/pnas.95.13.7670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Calderon L, Williams R, Martinez M, Clemons KV, Stevens DA. Genetic susceptibility to vaginal candidiasis. Med Mycol. 2003;41:143–147. doi: 10.1080/mmy.41.2.143.147. [DOI] [PubMed] [Google Scholar]
  6. Calera JA, Zhao XJ, De Bernardis F, Sheridan M, Calderone R. Avirulence of Candida albicans CaHK1 mutants in a murine model of hematogenously disseminated candidiasis. Infect Immun. 1999;67:4280–4284. doi: 10.1128/iai.67.8.4280-4284.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Cantorna MT, Balish E. Mucosal and systemic candidiasis in congenitally immunodeficient mice. Infect Immun. 1990;58:1093–1100. doi: 10.1128/iai.58.4.1093-1100.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Capilla J, Clemons KV, Stevens DA. Animal models: an important tool in mycology. Med Mycol. 2007;45:657–684. doi: 10.1080/13693780701644140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Cassone A, De BF, Santoni G. Anticandidal immunity and vaginitis: novel opportunities for immune intervention. Infect Immun. 2007;75:4675–4686. doi: 10.1128/IAI.00083-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Chamilos G, Lionakis MS, Lewis RE, Kontoyiannis DP. Role of mini-host models in the study of medically important fungi. Lancet Infect Dis. 2007;7:42–55. doi: 10.1016/S1473-3099(06)70686-7. [DOI] [PubMed] [Google Scholar]
  11. Cheng S, Clancy CJ, Checkley MA, Zhang Z, Wozniak KL, Seshan KR, Jia HY, Fidel P, Jr, Cole G, Nguyen MH. The role of Candida albicans NOT5 in virulence depends upon diverse host factors in vivo. Infect Immun. 2005;73:7190–7197. doi: 10.1128/IAI.73.11.7190-7197.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chiang LY, Sheppard DC, Bruno VM, Mitchell AP, Edwards JE, Jr, Filler SG. Candida albicans protein kinase CK2 governs virulence during oropharyngeal candidiasis. Cell Microbiol. 2007;9:233–245. doi: 10.1111/j.1462-5822.2006.00784.x. [DOI] [PubMed] [Google Scholar]
  13. Clemons KV, Spearow JL, Parmar R, Espiritu M, Stevens DA. Genetic susceptibility of mice to Candida albicans vaginitis correlates with host estrogen sensitivity. Infect Immun. 2004;72:4878–4880. doi: 10.1128/IAI.72.8.4878-4880.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Clemons KV, Gonzalez GM, Singh G, Imai J, Espiritu M, Parmar R, Stevens DA. Development of an orogastrointestinal mucosal model of candidiasis with dissemination to visceral organs. Antimicrob Agents Chemother. 2006;50:2650–2657. doi: 10.1128/AAC.00530-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cole AM. Innate host defense of human vaginal and cervical mucosae. Curr Top Microbiol Immunol. 2006;306:199–230. [PubMed] [Google Scholar]
  16. Cole MF, Bowen WH, Zhao XJ, Cihlar RL. Avirulence of Candida albicans auxotrophic mutants in a rat model of oropharyngeal candidiasis. FEMS Microbiol Lett. 1995;126:177–180. doi: 10.1111/j.1574-6968.1995.tb07413.x. [DOI] [PubMed] [Google Scholar]
  17. De Bernardis F, Muhlschlegel FA, Cassone A, Fonzi WA. The pH of the host niche controls gene expression in and virulence of Candida albicans. Infect Immun. 1998;66:3317–3325. doi: 10.1128/iai.66.7.3317-3325.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. De Bernardis F, Arancia S, Morelli L, Hube B, Sanglard D, Schafer W, Cassone A. Evidence that members of the secretory aspartyl proteinase gene family, in particular SAP2, are virulence factors for Candida vaginitis. J Infect Dis. 1999;179:201–208. doi: 10.1086/314546. [DOI] [PubMed] [Google Scholar]
  19. De Bernardis F, Lucciarini R, Boccanera M, Amantini C, Arancia S, Morrone S, Mosca M, Cassone A, Santoni G. Phenotypic and functional characterization of vaginal dendritic cells in a rat model of Candida albicans vaginitis. Infect Immun. 2006;74:4282–4294. doi: 10.1128/IAI.01714-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. de Repentigny L. Animal models in the analysis of Candida host–pathogen interactions. Curr Opin Microbiol. 2004;7:324–329. doi: 10.1016/j.mib.2004.06.001. [DOI] [PubMed] [Google Scholar]
  21. Deslauriers N, Coulombe C, Carre B, Goulet JP. Topical application of a corticosteroid destabilizes the host–parasite relationship in an experimental model of the oral carrier state of Candida albicans. FEMS Immunol Med Microbiol. 1995;11:45–55. doi: 10.1111/j.1574-695X.1995.tb00077.x. [DOI] [PubMed] [Google Scholar]
  22. Dolan JW, Bell AC, Hube B, Schaller M, Warner TF, Balish E. Candida albicans PLD I activity is required for full virulence. Med Mycol. 2004;42:439–447. doi: 10.1080/13693780410001657162. [DOI] [PubMed] [Google Scholar]
  23. Dongari-Bagtzoglou A, Fidel PL., Jr The host cytokine responses and protective immunity in oropharyngeal candidiasis. J Dent Res. 2005;84:966–977. doi: 10.1177/154405910508401101. [DOI] [PubMed] [Google Scholar]
  24. Dongari-Bagtzoglou A, Kashleva H. Development of a novel three-dimensional in vitro model of oral Candida infection. Microbial Pathogen. 2006;40:271–278. doi: 10.1016/j.micpath.2006.02.004. [DOI] [PubMed] [Google Scholar]
  25. Elahi S, Pang G, Clancy R, Ashman RB. Cellular and cytokine correlates of mucosal protection in murine model of oral candidiasis. Infect Immun. 2000;68:5771–5777. doi: 10.1128/iai.68.10.5771-5777.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Elahi S, Pang G, Ashman RB, Clancy R. Nitric oxide-enhanced resistance to oral candidiasis. Immunology. 2001;104:447–454. doi: 10.1046/j.1365-2567.2001.01331.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Farah CS, Elahi S, Drysdale K, Pang G, Gotjamanos T, Seymour GJ, Clancy RL, Ashman RB. Primary role for CD4(+) T lymphocytes in recovery from oropharyngeal candidiasis. Infect Immun. 2002a;70:724–731. doi: 10.1128/iai.70.2.724-731.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Farah CS, Gotjamanos T, Seymour GJ, Ashman RB. Cytokines in the oral mucosa of mice infected with Candida albicans. Oral Microbiol Immunol. 2002b;17:375–378. doi: 10.1034/j.1399-302x.2002.170607.x. [DOI] [PubMed] [Google Scholar]
  29. Farah CS, Hu Y, Riminton S, Ashman RB. Distinct roles for interleukin-12p40 and tumour necrosis factor in resistance to oral candidiasis defined by gene-targeting. Oral Microbiol Immunol. 2006;21:252–255. doi: 10.1111/j.1399-302X.2006.00288.x. [DOI] [PubMed] [Google Scholar]
  30. Fidel PL., Jr Candida–host interactions in HIV disease: relationships in oropharyngeal candidiasis. Adv Dent Res. 2006;19:80–84. doi: 10.1177/154407370601900116. [DOI] [PubMed] [Google Scholar]
  31. Fidel PL. History and update on host defense against vaginal candidiasis. Am J Reprod Immunol. 2007;57:2–12. doi: 10.1111/j.1600-0897.2006.00450.x. [DOI] [PubMed] [Google Scholar]
  32. Fidel PL, Jr, Barousse M, Espinosa T, Camaratti C, Ficarra M, Martin DH, Quayle AJ, Dunlap K. A live intravaginal Candida challenge in humans reveals new hypotheses for the immunopathogenesis of vulvovaginal candidiasis. Infect Immun. 2004a;72:2939–2946. doi: 10.1128/IAI.72.5.2939-2946.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Fidel PL, Jr, Barousse M, Espinosa T, Ficarra M, Sturtevant J, Martin DH, Quayle AJ, Dunlap K. An intravaginal live Candida challenge in humans leads to new hypotheses for the immunopathogenesis of vulvovaginal candidiasis. Infect Immun. 2004b;72:2939–2946. doi: 10.1128/IAI.72.5.2939-2946.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Fidel PLJ, Finkel-Jimenez B. Site-specific host immunity to fungi: lessons learned from Candida albicans applied to other fungi. In: Heitman J, Mitchell AP, Filler SG, Edwards JE, editors. Molecular Principles of Fungal Pathogenesis. Washington, DC: ASM Press; 2006. pp. 505–526. [Google Scholar]
  35. Fidel PLJ, Sobel JD. Murine models of Candida vaginal infections. In: Sande M, Zak O, editors. Handbook of Animal Models of Infection. London: Academic Press; 1999. pp. 741–748. [Google Scholar]
  36. Hamad M, Muta’eb E, bu-Shaqra Q, Fraij A, bu-Elteen K, Yasin SR. Utility of the oestrogen-dependent vaginal candidosis murine model in evaluating the efficacy of various therapies against vaginal Candida albicans infection. Mycoses. 2006;49:104–108. doi: 10.1111/j.1439-0507.2006.01200.x. [DOI] [PubMed] [Google Scholar]
  37. Hoyer LL, Green CB, Oh SH, Zhao X. Discovering the secrets of the Candida albicans agglutinin-like sequence (ALS) gene family - a sticky pursuit. Med Mycol. 2007;46:1–15. doi: 10.1080/13693780701435317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Huang W, Na L, Fidel P, Schwarzenberger P. Requirement of interleukin-17A for systemic anti-Candida albicans host defense in mice. J Infect Dis. 2004;190:624–631. doi: 10.1086/422329. [DOI] [PubMed] [Google Scholar]
  39. Jones JH, Russell C. Experimental oral candidiasis in weanling rats. J Dent Res. 1973;52:182. doi: 10.1177/00220345730520012001. [DOI] [PubMed] [Google Scholar]
  40. Kamai Y, Kubota M, Kamai Y, Hosokawa T, Fukuoka T, Filler SG. New model of oropharyngeal candidiasis in mice. Antimicrob Agents Chemother. 2001;45:3195–3197. doi: 10.1128/AAC.45.11.3195-3197.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Kamai Y, Kubota M, Kamai Y, Hosokawa T, Fukuoka T, Filler SG. Contribution of Candida albicans ALS1 to the pathogenesis of experimental oropharyngeal candidiasis. Infect Immun. 2002;70:5256–5258. doi: 10.1128/IAI.70.9.5256-5258.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Kazmi SH, Naglik JR, Sweet SP, Evans RW, O’Shea S, Banatvala JE, Challacombe SJ. Comparison of human immunodeficiency virus type 1-specific inhibitory activities in saliva and other human mucosal fluids. Clin Vaccine Immunol. 2006;13:1111–1118. doi: 10.1128/CDLI.00426-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Klein RS, Harris CA, Small CB, Moll B, Lesser M, Friedland GH. Oral candidiasis in high-risk patients as the initial manifestation of the acquired immunodeficiency syndrome. NEJM. 1984;311:354–358. doi: 10.1056/NEJM198408093110602. [DOI] [PubMed] [Google Scholar]
  44. Kurtzman CP, Robnett CJ, Ward JM, Brayton C, Gorelick P, Walsh TJ. Multigene phylogenetic analysis of pathogenic Candida species in the Kazachstania (Arxiozyma) telluris complex and description of their ascosporic states as Kazachstania bovina sp. nov.,K. heterogenica sp. nov.,K. pintolopesii sp. nov., and K. slooffiae sp. nov. J Clin Microbiol. 2005;43:101–111. doi: 10.1128/JCM.43.1.101-111.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Lacasse M, Fortier C, Chakir J, Cote L, Deslauriers N. Acquired resistance and persistence of Candida albicans following oral candidiasis in the mouse: a model of the carrier state in humans. Oral Microbiol Immunol. 1993;8:313–318. doi: 10.1111/j.1399-302x.1993.tb00580.x. [DOI] [PubMed] [Google Scholar]
  46. Moen K, Kvalvik AG, Hellem S, Jonsson R, Brun JG. The long-term effect of anti TNF-[alpha] treatment on temporomandibular joints, oral mucosa, and salivary flow in patients with active rheumatoid arthritis: a pilot study. Oral Surg Oral Med Oral Pathol Oral Radiol Endodontol. 2005;100:433–440. doi: 10.1016/j.tripleo.2005.05.060. [DOI] [PubMed] [Google Scholar]
  47. Mukherjee PK, Seshan KR, Leidich SD, Chandra J, Cole GT, Ghannoum MA. Reintroduction of the PLB1 gene into Candida albicans restores virulence in vivo. Microbiology. 2001;147:2585–2597. doi: 10.1099/00221287-147-9-2585. [DOI] [PubMed] [Google Scholar]
  48. Mylonakis E, Casadevall A, Ausubel FM. Exploiting amoeboid and non-vertebrate animal model systems to study the virulence of human pathogenic fungi. PLoS Pathog. 2007;3:e101. doi: 10.1371/journal.ppat.0030101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Naglik JR, Challacombe SJ, Hube B. Candida albicans secreted aspartyl proteinases in virulence and pathogenesis. Microbiol Mol Biol Rev. 2003;67:400–428. doi: 10.1128/MMBR.67.3.400-428.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Netea MG, Sutmuller R, Hermann C, Van Der Graaf CA, Van Der Meer JW, van Krieken JH, Hartung T, Adema G, Kullberg BJ. Toll-like receptor 2 suppresses immunity against Candida albicans through induction of IL-10 and regulatory T cells. J Immunol. 2004;172:3712–3718. doi: 10.4049/jimmunol.172.6.3712. [DOI] [PubMed] [Google Scholar]
  51. O’Connor C, Essmann M, Larsen B. 17-beta-estradiol upregulates the stress response in Candida albicans: implications for microbial virulence. Infect Dis Obstet Gynecol. 1998;6:176–181. doi: 10.1002/(SICI)1098-0997(1998)6:4<176::AID-IDOG7>3.0.CO;2-V. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Ohta H, Tanimoto T, Taniai M, Taniguchi M, Ariyasu T, Arai S, Ohta T, Fukuda S. Regulation of Candida albicans morphogenesis by tumor necrosis factor-alpha and potential for treatment of oral candidiasis. In Vivo. 2007;21:25–32. [PubMed] [Google Scholar]
  53. Pappas PG, Rex JH, Sobel JD, Filler SG, Dismukes WE, Walsh TJ, Edwards JE. Guidelines for treatment of candidiasis. Clin Infect Dis. 2004;38:161–189. doi: 10.1086/380796. [DOI] [PubMed] [Google Scholar]
  54. Phan QT, Myers CL, Fu Y, Sheppard DC, Yeaman MR, Welch WH, Ibrahim AS, Edwards JE, Filler SG. Als3 is a Candida albicans invasin that binds to cadherins and induces endocytosis by host cells. PLoS Biol. 2007;5:e64. doi: 10.1371/journal.pbio.0050064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Pope LM, Cole GT, Guentzel MN, Berry LJ. Systemic and gastrointestinal candidiasis of infant mice after intragastric challenge. Infect Immun. 1979;25:702–707. doi: 10.1128/iai.25.2.702-707.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Rahman D, Challacombe SJ. Oral immunization against mucosal candidiasis in a mouse model. Adv Exp Med Biol. 1995;371B:1663–1666. [PubMed] [Google Scholar]
  57. Rahman D, Mistry M, Thavaraj S, Challacombe SJ, Naglik JR. Murine model of concurrent oral and vaginal Candida albicans colonization to study epithelial host–pathogen interactions. Microbes Infect. 2007;9:615–622. doi: 10.1016/j.micinf.2007.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Richard M, Ibata-Ombetta S, Dromer F, Bordon-Pallier F, Jouault T, Gaillardin C. Complete glycosylphosphatidylinositol anchors are required in Candida albicans for full morphogenesis, virulence and resistance to macrophages. Mol Microbiol. 2002;44:841–853. doi: 10.1046/j.1365-2958.2002.02926.x. [DOI] [PubMed] [Google Scholar]
  59. Riggle PJ, Andrutis KA, Chen X, Tzipori SR, Kumamoto CA. Invasive lesions containing filamentous forms produced by a Candida albicans mutant that is defective in filamentous growth in culture. Infect Immun. 1999;67:3649–3652. doi: 10.1128/iai.67.7.3649-3652.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Saavedra M, Taylor B, Lukacs N, Fidel PL., Jr Local production of chemokines during experimental vaginal candidiasis. Infect Immun. 1999;67:5820–5826. doi: 10.1128/iai.67.11.5820-5826.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Samaranayake YH, Samaranayake LP. Experimental oral candidiasis in animal models. Clin Microbiol Rev. 2001;14:398–429. doi: 10.1128/CMR.14.2.398-429.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Savage DC, Dubos RJ. Localization of indigenous yeast in the murine stomach. J Bacteriol. 1967;94:1811–1816. doi: 10.1128/jb.94.6.1811-1816.1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Schaller M, Mailhammer R, Grassl G, Sander CA, Hube B, Korting HC. Infection of human oral epithelia with Candida species induces cytokine expression correlated to the degree of virulence. J Invest Dermatol. 2002;118:652–657. doi: 10.1046/j.1523-1747.2002.01699.x. [DOI] [PubMed] [Google Scholar]
  64. Schaller M, Boeld U, Oberbauer S, Hamm G, Hube B, Korting HC. Polymorphonuclear leukocytes (PMNs) induce protective Th1-type cytokine epithelial responses in an in vitro model of oral candidosis. Microbiology. 2004;150:2807–2813. doi: 10.1099/mic.0.27169-0. [DOI] [PubMed] [Google Scholar]
  65. Schaller M, Zakikhany K, Naglik JR, Weindl G, Hube B. Models of oral and vaginal candidiasis based on in vitro reconstituted human epithelia. Nat Protoc. 2006;1:2767–2773. doi: 10.1038/nprot.2006.474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Singleton DR, Fidel PL, Jr, Wozniak KL, Hazen KC. Contribution of cell surface hydrophobicity protein 1 (Csh1p) to virulence of hydrophobic Candida albicans serotype A cells. FEMS Microbiol Lett. 2005;244:373–377. doi: 10.1016/j.femsle.2005.02.010. [DOI] [PubMed] [Google Scholar]
  67. Skowronski R, Feldman D. Characterization of an estrogen-binding protein in the yeast Candida albicans. Endocrinology. 1989;124:1965–1972. doi: 10.1210/endo-124-4-1965. [DOI] [PubMed] [Google Scholar]
  68. Spearow JL, Doemeny P, Sera R, Leffler R, Barkley M. Genetic variation in susceptibility to endocrine disruption by estrogen in mice. Science. 1999;285:1259–1261. doi: 10.1126/science.285.5431.1259. [DOI] [PubMed] [Google Scholar]
  69. Styrt B, Sugarman B. Estrogens and infection. Rev Infect Dis. 1991;13:1139–1150. doi: 10.1093/clinids/13.6.1139. [DOI] [PubMed] [Google Scholar]
  70. Tan J, Li J, Chen S, Wu Y, Qin FT, Ding J, Cao F, Zhang S. Susceptibility to vaginal candidiasis under different conditions in mice. J Huazhong Univ Sci Technolog Med Sci. 2005;25:744–746. doi: 10.1007/BF02896190. [DOI] [PubMed] [Google Scholar]
  71. Taylor BN, Fichtenbaum C, Saavedra M, Slavinsky III, Swoboda R, Wozniak K, Arribas A, Powderly W, Fidel JP. In vivo virulence of Candida albicans isolates causing mucosal infections in people infected with the human immunodeficiency virus. J Infect Dis. 2000a;182:955–959. doi: 10.1086/315768. [DOI] [PubMed] [Google Scholar]
  72. Taylor BN, Saavedra M, Fidel P., Jr Local Th1/Th2 cytokine production during experimental vaginal candidiasis: potential importance of transforming growth factor-beta. Med Mycol. 2000b;38:419–431. doi: 10.1080/mmy.38.6.419.431. [DOI] [PubMed] [Google Scholar]
  73. Weindl G, Naglik JR, Kaesler S, Biedermann T, Hube B, Korting HC, Schaller M. Human epithelial cells establish direct antifungal defense through TLR4-mediated signaling. J Clin Invest. 2007;117:3664–3672. doi: 10.1172/JCI28115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Westwater C, Balish E, Warner TF, Nicholas PJ, Paulling EE, Schofield DA. Susceptibility of gnotobiotic transgenic mice (Tg{epsilon}26) with combined deficiencies in natural killer cells and T cells to wild-type and hyphal signallingdefective mutants of Candida albicans. J Med Microbiol. 2007;56:1138–1144. doi: 10.1099/jmm.0.47110-0. [DOI] [PubMed] [Google Scholar]
  75. White SJ, Rosenbach A, Lephart P, Nguyen D, Benjamin A, Tzipori S, Whiteway M, Mecsas J, Kumamoto CA. Self-regulation of Candida albicans population size during GI colonization. PLoS Pathogen. 2007;3:1866–1878. doi: 10.1371/journal.ppat.0030184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Wong B, Brauer KL, Clemens JR, Beggs S. Effects of gastrointestinal candidiasis, antibiotics, dietary arabinitol, and cortisone acetate on levels of the Candida metabolite D-arabinitol in rat serum and urine. Infect Immun. 1990;58:283–288. doi: 10.1128/iai.58.2.283-288.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Wormley FL, Jr, Chaiban J, Fidel PL., Jr Cell adhesion molecule and lymphocyte activation marker expression during experimental vaginal candidiasis. Infect Immun. 2001;69:5072–5079. doi: 10.1128/IAI.69.8.5072-5079.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Yu JJ, Gaffen SL. Interleukin-17: a novel inflammatory cytokine that bridges innate and adaptive immunity. Front Biosci. 2008;13:170–177. doi: 10.2741/2667. [DOI] [PubMed] [Google Scholar]
  79. Zelante T, De LA, Bonifazi P, et al. IL-23 and the Th17 pathway promote inflammation and impair antifungal immune resistance. Eur J Immunol. 2007;37:2695–2706. doi: 10.1002/eji.200737409. [DOI] [PubMed] [Google Scholar]
  80. Zhao XJ, Elhaney-Feser GE, Bowen WH, Cole MF, Broedel SE, Jr, Cihlar RL. Requirement for the Candida albicans FAS2 gene for infection in a rat model of oropharyngeal candidiasis. Microbiology. 1996;142:2509–2514. doi: 10.1099/00221287-142-9-2509. [DOI] [PubMed] [Google Scholar]

RESOURCES