Next Article in Journal
Inner Leaf Gel of Aloe striata Induces Adhesion-Reducing Morphological Hyphal Aberrations
Next Article in Special Issue
Special Issue: Mucosal Fungal Infections
Previous Article in Journal
Experimental In Vivo Models of Candidiasis
Previous Article in Special Issue
Anti-Aspergillus Activities of the Respiratory Epithelium in Health and Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Candida–Epithelial Interactions

Mucosal & Salivary Biology Division, Dental Institute, King’s College London, London SE1 1UL, UK
*
Author to whom correspondence should be addressed.
J. Fungi 2018, 4(1), 22; https://doi.org/10.3390/jof4010022
Submission received: 8 December 2017 / Revised: 4 February 2018 / Accepted: 6 February 2018 / Published: 8 February 2018
(This article belongs to the Special Issue Mucosal Fungal Infections)

Abstract

:
A plethora of intricate and dynamic molecular interactions occur between microbes and the epithelial cells that form the mucosal surfaces of the human body. Fungi, particularly species of Candida, are commensal members of our microbiota, continuously interacting with epithelial cells. Transient and localised perturbations to the mucosal environment can facilitate the overgrowth of fungi, causing infection. This minireview will examine the direct and indirect mechanisms by which Candida species and epithelial cells interact with each other, and explore the factors involved in the central processes of adhesion, invasion, and destruction of host mucosal surfaces.

1. Introduction

The mucosal surfaces of the human body are colonised by a rich and diverse microbiota, placing them under relentless microbial scrutiny. Fungi are an important but often overlooked component of the microbiota [1,2,3] that colonise mucosal surfaces, and one of the most common of these species are the Candida spp. There are over 150 spp. of Candida fungi, of which approximately 20 are known to be pathogenic to humans. While Candida spp. are typically commensal organisms, several factors, including alterations in the host immune status, microbial dysbiosis, lifestyle choices (e.g., smoking), and genetic predisposition can facilitate fungal overgrowth and a switch to pathogenic behaviour. Although Candida albicans is regarded as the most common of the Candida spp, C. glabrata, C. tropicalis, C. parapsilosis, and others are increasingly isolated from mucosal surfaces [4]. Indeed, the prevalence of Candida spp. and their continually evolving threat to human health is underpinned by the recent emergence of C. auris [5].
In order to colonise a host, Candida spp. must first interact with a mucosal surface. Candida spp. use physical attributes and an array of proadhesive, proinvasive and damage-inducing factors to facilitate colonisation and infection, and in this minireview, we will discuss the role of these factors in Candida–epithelial interactions.

2. Adhesion of Candida Species to Epithelial Surfaces

Microbial adhesion to epithelial surfaces is an essential prerequisite for mucosal colonisation and infection. The contact with the epithelium can be mediated through direct physical attachment or indirectly through physical association with co-colonising microbes and/or abiotic substrates. The site of fungal attachment can serve as a staging post for superficial, deep-seated, and systemic infections with high mortality in susceptible individuals. The adhesion of Candida spp. to epithelial cells is a complex, dynamic, and multifactorial process defined by the intimate association between components of the fungal cell wall and epithelial surface proteins.
Initial interactions between C. albicans yeast and host epithelium occur through the passive processes of hydrophobic and electrostatic attraction [6]. Changes in cell surface hydrophobicity correlate with changes in external protein content [7] and the ability of C. albicans to adhere to epithelial cells [6,8]. Moreover, hydrophobic yeasts are more virulent than hydrophilic yeasts in mice [9].
Once in contact with the epithelium, some Candida spp. undergo morphological switching from yeast to hyphae, and this morphological transition is a major virulence trait of some, but not all, Candida spp. For example, while C. albicans, C. dubliniensis, and C. tropicalis form hyphae, which adhere more strongly to epithelial cells than yeast cells [10], other species such as C. glabrata and C. parapsilosis do not form hyphae but are still adherent.
C. albicans Hyphal Wall Protein 1 (Hwp1p) is a key hypha-associated adhesin that facilitates epithelial cell attachment and is highly expressed during colonisation and infection of the oral mucosa [11,12]. The N-terminus of Hwp1p is a substrate for mammalian transglutaminase enzymes, resulting in covalent cross-linking of C. albicans to the oral epithelium [13]. These interactions preferentially occur within the outermost, terminally differentiated layers of the stratified oral epithelium that display the differentiation markers keratin 13 and small proline-rich protein 3 (SPR3) [14]. Deletion of HWP1 results in reduced adhesion to oral epithelial cells and decreased virulence in a murine model of oropharyngeal candidiasis (OPC) [15]. In addition to Hwp1p, C. albicans Hwp2p has also been reported to be required for adhesion to HT-29 human epithelial cells in vitro [16].
The C. albicans Als (agglutinin-like sequence) proteins [17], comprise an eight-member family of cell surface adhesins (Als1–7p and Als9p) which are GPI-linked to β-1-6 glucans in the fungal cell wall. The Als protein family has conserved amyloid-forming regions, and Als1p, Als3p, and Als5p contain the same heptapeptide amyloid-forming sequence [18]. Als-dependent cellular adhesion is concomitant with increases in cell surface hydrophobicity [19], suggesting that the amyloid-forming regions of Als proteins may contribute to overall hydrophobicity. Indeed, it is now understood that the formation of amyloids is an intrinsic property of yeast cell adhesin proteins [20], and the activation of discrete amyloid nanodomains within adhesins is required for cellular attachment [21]. C. albicans Als3p (hypha-associated) is a major epithelial adhesin that is strongly upregulated during epithelial infection in vitro [12], and disruption of the ALS3 gene reduces epithelial adhesion in vitro. Likewise, decreasing the expression of the ALS2 gene also reduces adhesion [22,23]. Interestingly, deletion of the ALS5, ALS6, or ALS7 genes was observed to increase adhesion [24], indicating that the Als proteins can have opposing roles in mediating fungal attachment to mucosal surfaces. However, the role of certain Als proteins during epithelial interaction remains controversial, with conflicting reports regarding the contribution of Als1p, Als2p, and Als4–6p [22,25,26].
Putative homologues of Als proteins have also been identified in C. parapsilosis, C. dubliniensis, and C. tropicalis although there is significant sequence divergence between spp. [27,28] and limited experimental data regarding their role in epithelial adhesion. However, the deletion of a C. parapsilosis homologue of C. albicans Als3p (CPAR2_404800) resulted in reduced adhesion to buccal epithelial cells in vitro and diminished pathogenicity in a murine model of urinary candidiasis [29], suggesting that at least some of these homologues have similar roles.
While C. albicans and C. dubliniensis differ considerably in terms of their pathogenicity, their genomes are remarkably similar [30]. The similarity between genome sequences is broadly conserved across the ALS gene family with the exception of ALS3, which is absent from C. dubliniensis [30]. Indeed, it has been suggested that divergent evolutionary trajectories (gene deletions including ALS3 for C. dubliniensis and expansion of a telomere-associated (TLO) gene family of transcriptional regulators within C. albicans) may account for the differences in pathogenicity between species [30,31].
The role of C. albicans Hyphally Regulated protein (Hyr1p) in epithelial adhesion is less clear, although a role in biofilm formation has been established. Disruption of the HYR1 gene leads to reduced virulence in vivo [32]. Other proteins not associated with hypha formation are also involved in C. albicans adhesion. For example, Eap1p (Enhanced Adherence to Polystyrene) is required for biofilm formation and adhesion to epithelial and fungal cells [33,34,35], while the GPI-linked, cell wall-associated aspartic proteinases Sap9p and Sap10p have opposing roles in epithelial adhesion: deletion of the SAP9 gene from C. albicans increases adhesion, while deletion of the SAP10 gene reduces adhesion [36]. Overexpression of the GPI-anchored IPF Family F protein Iff4p increases adhesion of C. albicans to epithelial but not endothelial cells [37].
In contrast to C. albicans, the main adhesion proteins mediating attachment of the non-filamentous fungus C. glabrata are Epithelial Adhesin (Epa) and Epa-like proteins. Epa1p of C. glabrata is a 1034-amino acid GPI-anchored, glucan cross-linked, cell wall-associated lectin required for robust, calcium-dependent adhesion to laryngeal and hamster ovary epithelial cells in vitro [38]. Epa1p binds to asialo-lactosyl-containing carbohydrates on host cells. An EPA1 gene deletion mutant exhibited a 95% reduction in epithelial adhesion, identifying this factor as a major contributor to C. glabrata adherence. Indeed, Epa1p alone is sufficient to mediate adhesion to epithelial cells, as expression of EPA1 in Saccharomyces cerevisiae resulted in robust epithelial adhesion [38].
Intriguingly, Epa1p-mediated adhesion to epithelial cells appears to be strain-dependent [39]. Some strains of C. glabrata rely on Epa1p for the majority of their epithelial adhesion, while others have minimal dependence. C. glabrata contains a number of EPA1 gene paralogues located at the subtelomeric regions of its genome [40], and different strains of C. glabrata express differing repertoires of Epa adhesins [41], some of which (Epa6p and Epa7p) are regulated by subtelomeric silencing [42]. Given the large size of the EPA gene family in C. glabrata (>20 EPA-like members), it is likely that a certain degree of functional redundancy may account for the differences in experimental observations. Indeed, a degree of molecular promiscuity is apparent in the adhesin domain of Epa1p, Epa6p, and Epa7p that facilitates attachment to a number of different carbohydrate ligands, particularly those that contain terminal galactose residues [43,44]. Common adhesins used by Candida spp. to interact with epithelial cells are presented in Table 1.

3. Induced Endocytosis

Following colonisation, Candida spp. can invade the epithelium in order to establish an infection. C. albicans can enter epithelial cells by a passive host-mediated process called induced endocytosis [45,46]. Endocytosis of C. albicans hyphae and non-filamentous C. glabrata occurs within 4 h of initial contact with epithelial cells [47,48]. Induced endocytosis is triggered by the recognition of invasins expressed on the fungal cell surface. To date, two C. albicans invasins have been identified, Als3p and Ssa1p, both of which interact with the host epithelial receptor E-cadherin [49,50]. Interaction of E-cadherin with Als3p or Ssa1p promotes the accumulation and colocalisation of dynamin, clathrin, and cortactin at the site of hyphal contact, which coordinate the remodelling of the actin cytoskeleton required to endocytose the fungus [51]. C. albicans als3∆/∆ and ssa1∆/∆ gene deletion mutants exhibit reduced binding to, and invasion of epithelial cells and are attenuated for virulence in a murine model of OPC [49,50,52].
Induced endocytosis can also occur independently of epithelial E-cadherin through the interaction of fungal Als3p and Ssa1p with the epidermal growth factor receptor (EGFR/HER2) heterodimer expressed on epithelial cells [53] and through a mechanism involving host GTPases (Cdc42, Rac1, RhoA) and the tight junction protein ZO-1 [54]. The PDGF BB (platelet-derived growth factor BB) and NEDD9 (neural precursor cell-expressed developmentally downregulated protein 9) pathways are also implicated in epithelial uptake of C. albicans, as siRNA knockdown of PDGF receptor beta (PDGFRB) or NEDD9 causes a reduction in endocytosis [55]. More recently, the host epithelial aryl hydrocarbon receptor (AhR) has been identified as an important upstream component involved in fungal endocytosis [56]. Activation of AhR by C. albicans results in Src-mediated phosphorylation of EGFR and fungal internalisation, while inhibition of AhR was observed to reduce invasion and OPC severity [56].
Intriguingly, some Candida spp. exploit the process of induced endocytosis to avoid immune recognition. For instance, C. parapsilosis is efficiently endocytosed by endothelial cells which provide a means of escape from patrolling neutrophils [57]. However, it must be noted that this interaction occurs within the systemic compartment and its relevance to mucosal responses has yet to be established.

4. Active Penetration

The oral and vaginal mucosae are comprised of stratified layers, the outermost of which are terminally differentiated, nonproliferative, and therefore less likely to support fungal invasion through induced endocytosis. Candida spp. must therefore use an alternative mechanism to invade a tissue that does not readily support internalisation. This process is called active penetration. Active penetration of mucosal barriers requires a viable fungus and occurs by hyphal invasion through or between epithelial cells. Active penetration is dependent upon fungal attributes including hyphal turgor pressure, physical advancement of the hyphal tip, and secretion of factors such as hydrolytic enzymes that may facilitate the breaching of mucosal barriers by degrading host substrates. The C. albicans-secreted aspartic proteinase Sap5p can degrade E-cadherin, an important component of epithelial adherens junctions [58], while Sap2p can degrade mucins which coat all mucosal tissues [59]. Active penetration is mechanistically distinct from induced endocytosis, as blocking actin polymerisation does not prevent mucosal penetration from occurring [52,60]. While proteinases are thought to play a significant role in C. albicans invasion and penetration, secreted lipases and phospholipases appear to play a limited role in these processes [60,61]. C. parapsilosis has been observed to invade the connective tissue of human oral mucosa [62], although the mechanism of invasion has yet to be characterised. Clinical isolates of C. parapsilosis and C. tropicalis also colonise and invade reconstituted human oral epithelium [63,64] and both display strain-dependent variation in their ability to do so.
Active penetration of C. albicans hyphae occurs at all mucosal surfaces, but in the gut it is the sole driver of invasion across the epithelium [49,60]. Active penetration occurs at later time points during infection when compared with induced endocytosis. Although induced endocytosis is observed to occur prior to active penetration in vitro, active penetration is likely to be the initial mechanism that enables C. albicans to invade through the outermost layers of an epithelium in vivo. However, once the fungus has accessed the underlying proliferative layers of an epithelium, induced endocytosis may then occur to further facilitate invasion. While induced endocytosis and active penetration are therefore mechanistically distinct processes, both are nevertheless likely to be required to establish infection through stratified mucosal barriers in vivo.

5. Epithelial Interactions with Candida Species

The process of receptor-mediated epithelial recognition of Candida spp. is poorly understood, but recent studies have shed light on the signalling pathways activated by Candida, particularly C. albicans. C. albicans yeast cells weakly activate three key signalling pathways: all three MAPK (mitogen-activated protein kinase) pathways (namely, p38, JNK, ERK1/2), the NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) pathway, and the PI3K (phosphatidylinositide 3-kinase) pathway [65,66]. This drives the activation of the transcription factors NF-κB and c-Jun (via ERK1/2 and JNK) but is insufficient to induce immune activation. C. albicans hyphae activate the same signalling pathways, but there is a far stronger activation of MAPK signalling, with specific induction of the transcription factor c-Fos (via p38) [65]. C. albicans hyphae also activate MKP1 (MAPK phosphatase 1) via the ERK1/2 pathway, which is known to regulate MAPK-mediated immune responses [65]. This combination of c-Fos and MKP1 activation is specifically associated with hypha formation and correlates with immune activation. While the PI3K pathway is also involved in immune activation, its main function appears to be in protecting epithelial cells against hypha-induced damage [66].
The epithelial receptors that interact with C. albicans to initiate these signalling mechanisms and induce immune responses are unclear. Epithelial cells express numerous pattern recognition receptors (PRRs) (e.g., toll-like receptors (TLRs), C-type lectin receptors), including TLR2, TLR4, and dectin-1 [62,65,67,68], which can alter in expression after Candida challenge [62,66]. These PRRs recognise yeast and hyphal cells via conventional fungal pathogen-associated molecular patterns (PAMPs) (i.e., mannans, β-glucans) [69,70]. However, this conventional PAMP–PRR interaction does not appear to activate the c-Fos/MKP1 signalling pathway or the secretion of immune modulatory cytokines [65,67]. Notably, both c-Fos/MKP1 signalling and immune activation have recently been shown to be activated by a novel peptide toxin called Candidalysin that is secreted by C. albicans hyphae [71] (see below). However, recently it has been suggested that the β-glucan of C. glabrata, Rhizopus delemar, Aspergillus fumigatus, S. cerevisiae, and C. albicans yeast and hyphae can interact with the epithelial PRR Ephrin Type-A Receptor 2 (EphA2), activating signal transducer and activator of transcription 3 (STAT3) and MAPK signalling that culminates in proinflammatory antifungal responses [72]. This indicates that dectin-1 may not be the only PRR that recognises β-glucan and further suggests that epithelial cell recognition of fungal PAMPs may be different to myeloid cell recognition of fungal PAMPs. As such, while it is clear that dectin-1 plays a crucial role in systemic Candida infections [73,74,75,76], its role in mucosal infections and immune responses appears minimal [65]. This was elegantly demonstrated recently by Gaffen and colleagues who showed that protective innate Type 17 responses against oral candidiasis were independent of dectin-1 but dependent upon Candidalysin activity [77].

6. Secreted Factors, Nutrient Acquisition, and Damage

Numerous factors secreted by Candida spp. are capable of interacting with epithelial cells, including hydrolytic enzymes and toxins. Sap-like enzymes are present in several Candida spp., including C. dubliniensis, C. tropicalis, and C. parapsilosis [30,78,79,80,81]. The loss of these secreted factors can significantly impact on the interactions between these fungi and epithelial cells. For example, deletion of C. albicans SAP1, SAP2, or SAP3 genes has been observed to reduce adherence to buccal epithelial cells, while a sap4-6Δ/Δ triple mutant displayed increased adhesion [82]. Collectively, the SAP family exhibits broad substrate specificity [83] and can degrade epithelial cadherin [58] and gastrointestinal mucins [59], which may yield metabolically useful substrates during the course of an infection.
The secreted phospholipases of C. albicans have been implicated in adhesion to buccal epithelial cells, although their role is somewhat controversial [61,84,85]. Strain-dependent phospholipase and proteinase activity has also been detected in the emerging pathogen C. auris [86]. Deletion of the C. parapsilosis lipases CpLIP1 and CpLIP2 inhibits biofilm formation, reduces virulence on reconstituted human oral epithelium, and increases susceptibility to killing by immune cells [87], demonstrating a role for these enzymes in mucosal pathogenicity.
During mucosal infection, the hyphae of C. albicans secrete Candidalysin, a cytolytic peptide toxin derived from the hypha-associated protein Ece1p. The Candidalysin toxin is a dual-function molecule. As mentioned above, Candidalysin can induce mucosal immunity predominantly through the activation of MAPK signalling via c-Fos and MKP-1. In addition, Candidalysin also damages the plasma membrane of epithelial cells [71]. It is tempting to speculate that Candidalysin may facilitate fungal acquisition of nutrients by damaging epithelial membranes, causing the release of substrates that are metabolically useful to the invading fungus.
In this regard, it is interesting to note that Als3p expressed on C. albicans hyphae can sequester ferritin from epithelial cells [88]. The C. albicans CFEM (Common in Fungal Extracellular Membranes) proteins Rbt5p, Pga7p, and Csa1p also play a role in iron acquisition from host proteins [89,90]. Compared with C. albicans, the CFEM family is expanded in C. parapsilosis [91]. The CFEM2 and CFEM3 genes of C. parapsilosis are critical for haem utilisation, whereas the CFEM6 gene is only partially required [91].
The ability to acquire and assimilate host micronutrients including copper and zinc has a profound influence on fungal pathogenicity [92]. Indeed, the importance of micronutrients is such that host cells and tissues have evolved mechanisms to modulate the availability of these essential nutrients in a process collectively referred to as nutritional immunity [93]. Secreted factors such as C. albicans pH-regulated antigen 1 (Pra1p) is required to scavenge zinc from endothelial cells [94], while zinc limitation results in altered cellular morphology in several Candida spp. (C. albicans, C. dubliniensis, and C. tropicalis, but not C. parapsilosis or C. lusitaniae) and increased adhesion of C. albicans to abiotic substrates [95]. Factors that contribute to epithelial interactions are presented in Figure 1.

7. Heterotypic Interactions between Candida Species and Mucosal Bacteria

It is becoming increasingly clear that the microbial communities that colonise mucosal surfaces have a profound influence on Candida–epithelial interactions. Adhesion of Candida spp. to a mucosal surface can be direct (via classical adhesins for example) but can also occur indirectly through specific interactions with other fungi and bacteria [96,97].
The analysis of mixed fungal cultures using a magnetic bead-based adherence and aggregation assay revealed that C. albicans can form heterotypic aggregates with C. glabrata and C. parapsilosis in vitro [98]. Infection of tongue tissue by C. glabrata during murine OPC is enhanced when co-infected with C. albicans or when added to a pre-established C. albicans infection [99], strongly suggesting that an initial epithelial interaction with one spp. of Candida can have a dramatic influence on the pathogenicity of a different fungal spp. Such interactions are not limited to other fungi, however. Streptococcus gordonii is a primary commensal coloniser of the oral cavity, and C. albicans Als3p can interact with the SspB adhesin of S. gordonii [100], while Als5p is also capable of binding to S. gordonii [98]. Als3p also mediates binding between C. albicans and Staphylococcus aureus [101].
The interaction of Candida spp. with one another and with mucosal bacteria may serve to increase the likelihood of a successful Candida–epithelial interaction, although this is not always the case. For instance, the probiotic bacterium Lactobacillus rhamnosus GG induces a metabolic reprogramming of C. albicans causing reduced hyphal extension, adhesion to, and invasion of oral epithelial cells in vitro [102], demonstrating that the interactions between C. albicans and mucosal bacteria can have variable outcomes in the context of mucosal interaction.

8. Conclusions

Candida species employ a diverse range of direct and indirect mechanisms that enable the interaction with host epithelial cells. Under suitably predisposing conditions, commensal colonisation of mucosal surfaces is followed by pathogenic infiltration and secretion of hydrolytic enzymes and toxins that reduce mucosal barrier function, facilitating disease progression.
Importantly, variations between different Candida spp. and between fungal strains of the same species have an undoubted impact on Candida–epithelial interactions. For instance, while C. albicans triggers more epithelial cell damage than other Candida spp., the extent of damage varies between strains [103], leading to differences in alarmin production [83,104]. Likewise, differences in epithelial damage, invasion, and secretion of proinflammatory cytokines is also apparent between strains of C. glabrata [105], while different strains of C. albicans exhibit variable pathogenicity in animal models [103,106,107].
The physical contact between Candida spp. and members of the resident microbiota can influence the mucosal interactions and has direct consequences on disease outcome. In response, the epithelial receptor-mediated recognition of Candida spp. activates a number of dynamic host signalling pathways that enable appropriate proinflammatory and immune defence mechanisms to be implemented. The environmental conditions greatly affect the outcome of any experiment, including Candida–host interactions. The precise factors that influence these interactions are unquestionably variable and complex.
Numerous aspects of Candida–epithelial interactions remain to be explored in full. The majority of experimental data obtained thus far describes factors utilised by C. albicans, but there is an increasing need to identify the proteins used by other Candida spp. that influence epithelial interactions. It is likely that there are many more epithelial-specific receptors involved in Candida recognition that have yet to be identified and characterised. The signalling responses induced by fungal endocytosis have yet to be characterised in full, and the contribution of secreted factors to fungal pathogenicity remains an area of intense study. Indeed, while Candidalysin has been shown to contribute to both oral and vaginal infections in mice [71,108], the role of Candidalysin in the systemic compartment is not yet clear.
While considerable progress has increased our understanding of the complex relationship that exists between Candida fungi and mucosal surfaces, continued research will undoubtedly provide further clarity into the mechanisms that underpin this important host–pathogen interaction.

Acknowledgments

We thank David Moyes for critical reading of the manuscript. This work was supported by grants from the Biotechnology and Biological Sciences Research Council (BB/N014677/1), the Medical Research Council (MR/M011372/1), the National Institute for Health Research at Guys and St Thomas’s NHS Foundation Trust and King’s College London Biomedical Research Centre (IS-BRC-1215-20006), the Rosetrees Trust (M680), and the National Institutes of Health (R37-DE022550).

Author Contributions

Jonathan P. Richardson wrote the initial draft of the manuscript. Jemima Ho created the figure. Jonathan P. Richardson, Jemima Ho and Julian R. Naglik edited the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Huffnagle, G.B.; Noverr, M.C. The emerging world of the fungal microbiome. Trends Microbiol. 2013, 21, 334–341. [Google Scholar] [CrossRef] [PubMed]
  2. Baker, J.L.; Bor, B.; Agnello, M.; Shi, W.; He, X. Ecology of the oral microbiome: Beyond bacteria. Trends Microbiol. 2017, 25, 362–374. [Google Scholar] [CrossRef] [PubMed]
  3. Naglik, J.R.; Tang, S.X.; Moyes, D.L. Oral colonization of fungi. Curr. Fungal Infect. Rep. 2013, 7, 152–159. [Google Scholar] [CrossRef]
  4. Williams, D.; Lewis, M. Pathogenesis and treatment of oral candidosis. J. Oral Microbiol. 2011, 3. [Google Scholar] [CrossRef] [PubMed]
  5. Jeffery-Smith, A.; Taori, S.K.; Schelenz, S.; Jeffery, K.; Johnson, E.M.; Borman, A.; Candida auris Incident Management Team; Manuel, R.; Brown, C.S. Candida auris: A review of the literature. Clin. Microbiol. Rev. 2018, 31. [Google Scholar] [CrossRef]
  6. Hazen, K.C. Participation of yeast cell surface hydrophobicity in adherence of Candida albicans to human epithelial cells. Infect. Immun. 1989, 57, 1894–1900. [Google Scholar] [PubMed]
  7. Hazen, K.C.; Lay, J.G.; Hazen, B.W.; Fu, R.C.; Murthy, S. Partial biochemical characterization of cell surface hydrophobicity and hydrophilicity of Candida albicans. Infect. Immun. 1990, 58, 3469–3476. [Google Scholar] [PubMed]
  8. Ener, B.; Douglas, L.J. Correlation between cell-surface hydrophobicity of Candida albicans and adhesion to buccal epithelial cells. FEMS Microbiol. Lett. 1992, 78, 37–42. [Google Scholar] [CrossRef] [PubMed]
  9. Antley, P.P.; Hazen, K.C. Role of yeast cell growth temperature on Candida albicans virulence in mice. Infect. Immun. 1988, 56, 2884–2890. [Google Scholar] [PubMed]
  10. Sandin, R.L.; Rogers, A.L.; Patterson, R.J.; Beneke, E.S. Evidence for mannose-mediated adherence of Candida albicans to human buccal cells in vitro. Infect. Immun. 1982, 35, 79–85. [Google Scholar] [PubMed]
  11. Naglik, J.R.; Fostira, F.; Ruprai, J.; Staab, J.F.; Challacombe, S.J.; Sundstrom, P. Candida albicans HWP1 gene expression and host antibody responses in colonization and disease. J. Med. Microbiol. 2006, 55, 1323–1327. [Google Scholar] [CrossRef] [PubMed]
  12. Zakikhany, K.; Naglik, J.R.; Schmidt-Westhausen, A.; Holland, G.; Schaller, M.; Hube, B. In vivo transcript profiling of Candida albicans identifies a gene essential for interepithelial dissemination. Cell. Microbiol. 2007, 9, 2938–2954. [Google Scholar] [CrossRef] [PubMed]
  13. Staab, J.F.; Bradway, S.D.; Fidel, P.L.; Sundstrom, P. Adhesive and mammalian transglutaminase substrate properties of Candida albicans Hwp1. Science 1999, 283, 1535–1538. [Google Scholar] [CrossRef] [PubMed]
  14. Ponniah, G.; Rollenhagen, C.; Bahn, Y.S.; Staab, J.F.; Sundstrom, P. State of differentiation defines buccal epithelial cell affinity for cross-linking to Candida albicans Hwp1. J. Oral Pathol. Med. 2007, 36, 456–467. [Google Scholar] [CrossRef] [PubMed]
  15. Sundstrom, P.; Balish, E.; Allen, C.M. Essential role of the Candida albicans transglutaminase substrate, hyphal wall protein 1, in lethal oroesophageal candidiasis in immunodeficient mice. J. Infect. Dis. 2002, 185, 521–530. [Google Scholar] [CrossRef] [PubMed]
  16. Younes, S.; Bahnan, W.; Dimassi, H.I.; Khalaf, R.A. The Candida albicans Hwp2 is necessary for proper adhesion, biofilm formation and oxidative stress tolerance. Microbiol. Res. 2011, 166, 430–436. [Google Scholar] [CrossRef] [PubMed]
  17. Hoyer, L.L.; Cota, E. Candida albicans agglutinin-like sequence (Als) family vignettes: A review of Als protein structure and function. Front. Microbiol. 2016, 7, 280. [Google Scholar] [CrossRef] [PubMed]
  18. Otoo, H.N.; Lee, K.G.; Qiu, W.; Lipke, P.N. Candida albicans Als adhesins have conserved amyloid-forming sequences. Eukaryot. Cell 2008, 7, 776–782. [Google Scholar] [CrossRef] [PubMed]
  19. Rauceo, J.M.; Gaur, N.K.; Lee, K.G.; Edwards, J.E.; Klotz, S.A.; Lipke, P.N. Global cell surface conformational shift mediated by a Candida albicans adhesin. Infect. Immun. 2004, 72, 4948–4955. [Google Scholar] [CrossRef] [PubMed]
  20. Ramsook, C.B.; Tan, C.; Garcia, M.C.; Fung, R.; Soybelman, G.; Henry, R.; Litewka, A.; O’Meally, S.; Otoo, H.N.; Khalaf, R.A.; et al. Yeast cell adhesion molecules have functional amyloid-forming sequences. Eukaryot. Cell 2010, 9, 393–404. [Google Scholar] [CrossRef] [PubMed]
  21. Lipke, P.N.; Ramsook, C.; Garcia-Sherman, M.C.; Jackson, D.N.; Chan, C.X.; Bois, M.; Klotz, S.A. Between amyloids and aggregation lies a connection with strength and adhesion. New J. Sci. 2014, 2014, 815102. [Google Scholar] [CrossRef] [PubMed]
  22. Zhao, X.; Oh, S.H.; Cheng, G.; Green, C.B.; Nuessen, J.A.; Yeater, K.; Leng, R.P.; Brown, A.J.; Hoyer, L.L. ALS3 and ALS8 represent a single locus that encodes a Candida albicans adhesin; functional comparisons between Als3p and Als1p. Microbiology 2004, 150, 2415–2428. [Google Scholar] [CrossRef] [PubMed]
  23. Zhao, X.; Oh, S.H.; Yeater, K.M.; Hoyer, L.L. Analysis of the Candida albicans Als2p and Als4p adhesins suggests the potential for compensatory function within the Als family. Microbiology 2005, 151, 1619–1630. [Google Scholar] [CrossRef] [PubMed]
  24. Zhao, X.; Oh, S.H.; Hoyer, L.L. Deletion of ALS5, ALS6 or ALS7 increases adhesion of Candida albicans to human vascular endothelial and buccal epithelial cells. Med. Mycol. 2007, 45, 429–434. [Google Scholar] [CrossRef] [PubMed]
  25. Kamai, Y.; Kubota, M.; Kamai, Y.; Hosokawa, T.; Fukuoka, T.; Filler, S.G. Contribution of Candida albicans Als1 to the pathogenesis of experimental oropharyngeal candidiasis. Infect. Immun. 2002, 70, 5256–5258. [Google Scholar] [CrossRef] [PubMed]
  26. Murciano, C.; Moyes, D.L.; Runglall, M.; Tobouti, P.; Islam, A.; Hoyer, L.L.; Naglik, J.R. Evaluation of the role of Candida albicans agglutinin-like sequence (Als) proteins in human oral epithelial cell interactions. PLoS ONE 2012, 7, e33362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Butler, G.; Rasmussen, M.D.; Lin, M.F.; Santos, M.A.; Sakthikumar, S.; Munro, C.A.; Rheinbay, E.; Grabherr, M.; Forche, A.; Reedy, J.L.; et al. Evolution of pathogenicity and sexual reproduction in eight Candida genomes. Nature 2009, 459, 657–662. [Google Scholar] [CrossRef] [PubMed]
  28. Hoyer, L.L.; Fundyga, R.; Hecht, J.E.; Kapteyn, J.C.; Klis, F.M.; Arnold, J. Characterization of agglutinin-like sequence genes from non-albicans Candida and phylogenetic analysis of the Als family. Genetics 2001, 157, 1555–1567. [Google Scholar] [PubMed]
  29. Bertini, A.; Zoppo, M.; Lombardi, L.; Rizzato, C.; De Carolis, E.; Vella, A.; Torelli, R.; Sanguinetti, M.; Tavanti, A. Targeted gene disruption in Candida parapsilosis demonstrates a role for CPAR2_404800 in adhesion to a biotic surface and in a murine model of ascending urinary tract infection. Virulence 2016, 7, 85–97. [Google Scholar] [CrossRef] [PubMed]
  30. Jackson, A.P.; Gamble, J.A.; Yeomans, T.; Moran, G.P.; Saunders, D.; Harris, D.; Aslett, M.; Barrell, J.F.; Butler, G.; Citiulo, F.; et al. Comparative genomics of the fungal pathogens Candida dubliniensis and Candida albicans. Genome Res. 2009, 19, 2231–2244. [Google Scholar] [CrossRef] [PubMed]
  31. Sullivan, D.J.; Berman, J.; Myers, L.C.; Moran, G.P. Telomeric ORFS in Candida albicans: Does mediator tail wag the yeast? PLoS Pathog. 2015, 11, e1004614. [Google Scholar] [CrossRef] [PubMed]
  32. Dwivedi, P.; Thompson, A.; Xie, Z.; Kashleva, H.; Ganguly, S.; Mitchell, A.P.; Dongari-Bagtzoglou, A. Role of BCR1-activated genes HWP1 and HYR1 in Candida albicans oral mucosal biofilms and neutrophil evasion. PLoS ONE 2011, 6, e16218. [Google Scholar] [CrossRef] [PubMed]
  33. Li, F.; Palecek, S.P. EAP1, a Candida albicans gene involved in binding human epithelial cells. Eukaryot. Cell 2003, 2, 1266–1273. [Google Scholar] [CrossRef] [PubMed]
  34. Li, F.; Svarovsky, M.J.; Karlsson, A.J.; Wagner, J.P.; Marchillo, K.; Oshel, P.; Andes, D.; Palecek, S.P. Eap1p, an adhesin that mediates Candida albicans biofilm formation in vitro and in vivo. Eukaryot. Cell 2007, 6, 931–939. [Google Scholar] [CrossRef] [PubMed]
  35. Li, F.; Palecek, S.P. Distinct domains of the Candida albicans adhesin Eap1p mediate cell-cell and cell-substrate interactions. Microbiology 2008, 154, 1193–1203. [Google Scholar] [CrossRef] [PubMed]
  36. Albrecht, A.; Felk, A.; Pichova, I.; Naglik, J.R.; Schaller, M.; de Groot, P.; Maccallum, D.; Odds, F.C.; Schafer, W.; Klis, F.; et al. Glycosylphosphatidylinositol-anchored proteases of Candida albicans target proteins necessary for both cellular processes and host-pathogen interactions. J. Biol. Chem. 2006, 281, 688–694. [Google Scholar] [CrossRef] [PubMed]
  37. Fu, Y.; Luo, G.; Spellberg, B.J.; Edwards, J.E., Jr.; Ibrahim, A.S. Gene overexpression/suppression analysis of candidate virulence factors of Candida albicans. Eukaryot. Cell 2008, 7, 483–492. [Google Scholar] [CrossRef] [PubMed]
  38. Cormack, B.P.; Ghori, N.; Falkow, S. An adhesin of the yeast pathogen Candida glabrata mediating adherence to human epithelial cells. Science 1999, 285, 578–582. [Google Scholar] [CrossRef] [PubMed]
  39. Vale-Silva, L.A.; Moeckli, B.; Torelli, R.; Posteraro, B.; Sanguinetti, M.; Sanglard, D. Upregulation of the adhesin gene EPA1 mediated by PDR1 in Candida glabrata leads to enhanced host colonization. mSphere 2016, 1. [Google Scholar] [CrossRef] [PubMed]
  40. De Las Penas, A.; Pan, S.J.; Castano, I.; Alder, J.; Cregg, R.; Cormack, B.P. Virulence-related surface glycoproteins in the yeast pathogen Candida glabrata are encoded in subtelomeric clusters and subject to RAP1- and SIR-dependent transcriptional silencing. Genes Dev. 2003, 17, 2245–2258. [Google Scholar] [CrossRef] [PubMed]
  41. Gomez-Molero, E.; de Boer, A.D.; Dekker, H.L.; Moreno-Martinez, A.; Kraneveld, E.A.; Ichsan; Chauhan, N.; Weig, M.; de Soet, J.J.; de Koster, C.G.; et al. Proteomic analysis of hyperadhesive Candida glabrata clinical isolates reveals a core wall proteome and differential incorporation of adhesins. FEMS Yeast Res. 2015, 15. [Google Scholar] [CrossRef] [PubMed]
  42. Castano, I.; Pan, S.J.; Zupancic, M.; Hennequin, C.; Dujon, B.; Cormack, B.P. Telomere length control and transcriptional regulation of subtelomeric adhesins in Candida glabrata. Mol. Microbiol. 2005, 55, 1246–1258. [Google Scholar] [CrossRef] [PubMed]
  43. Maestre-Reyna, M.; Diderrich, R.; Veelders, M.S.; Eulenburg, G.; Kalugin, V.; Bruckner, S.; Keller, P.; Rupp, S.; Mosch, H.U.; Essen, L.O. Structural basis for promiscuity and specificity during Candida glabrata invasion of host epithelia. Proc. Natl. Acad. Sci. USA 2012, 109, 16864–16869. [Google Scholar] [CrossRef] [PubMed]
  44. Zupancic, M.L.; Frieman, M.; Smith, D.; Alvarez, R.A.; Cummings, R.D.; Cormack, B.P. Glycan microarray analysis of Candida glabrata adhesin ligand specificity. Mol. Microbiol. 2008, 68, 547–559. [Google Scholar] [CrossRef] [PubMed]
  45. Drago, L.; Mombelli, B.; De Vecchi, E.; Bonaccorso, C.; Fassina, M.C.; Gismondo, M.R. Candida albicans cellular internalization: A new pathogenic factor? Int. J. Antimicrob. Agents 2000, 16, 545–547. [Google Scholar] [CrossRef]
  46. Park, H.; Myers, C.L.; Sheppard, D.C.; Phan, Q.T.; Sanchez, A.A.; J, E.E.; Filler, S.G. Role of the fungal Ras-protein kinase a pathway in governing epithelial cell interactions during oropharyngeal candidiasis. Cell. Microbiol. 2005, 7, 499–510. [Google Scholar] [CrossRef] [PubMed]
  47. Villar, C.C.; Zhao, X.R. Candida albicans induces early apoptosis followed by secondary necrosis in oral epithelial cells. Mol. Oral Microbiol. 2010, 25, 215–225. [Google Scholar] [CrossRef] [PubMed]
  48. Li, L.; Dongari-Bagtzoglou, A. Epithelial GM-CSF induction by Candida glabrata. J. Dent. Res. 2009, 88, 746–751. [Google Scholar] [CrossRef] [PubMed]
  49. Phan, Q.T.; Myers, C.L.; Fu, Y.; Sheppard, D.C.; Yeaman, M.R.; Welch, W.H.; Ibrahim, A.S.; Edwards, J.E., Jr.; Filler, S.G. Als3 is a Candida albicans invasin that binds to cadherins and induces endocytosis by host cells. PLoS Biol. 2007, 5, e64. [Google Scholar] [CrossRef] [PubMed]
  50. Sun, J.N.; Solis, N.V.; Phan, Q.T.; Bajwa, J.S.; Kashleva, H.; Thompson, A.; Liu, Y.; Dongari-Bagtzoglou, A.; Edgerton, M.; Filler, S.G. Host cell invasion and virulence mediated by Candida albicans Ssa1. PLoS Pathog. 2010, 6, e1001181. [Google Scholar] [CrossRef] [PubMed]
  51. Moreno-Ruiz, E.; Galan-Diez, M.; Zhu, W.; Fernandez-Ruiz, E.; d’Enfert, C.; Filler, S.G.; Cossart, P.; Veiga, E. Candida albicans internalization by host cells is mediated by a clathrin-dependent mechanism. Cell. Microbiol. 2009, 11, 1179–1189. [Google Scholar] [CrossRef] [PubMed]
  52. Wachtler, B.; Citiulo, F.; Jablonowski, N.; Forster, S.; Dalle, F.; Schaller, M.; Wilson, D.; Hube, B. Candida albicans-epithelial interactions: Dissecting the roles of active penetration, induced endocytosis and host factors on the infection process. PLoS ONE 2012, 7, e36952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Zhu, W.; Phan, Q.T.; Boontheung, P.; Solis, N.V.; Loo, J.A.; Filler, S.G. EGFR and HER2 receptor kinase signaling mediate epithelial cell invasion by Candida albicans during oropharyngeal infection. Proc. Natl. Acad. Sci. USA 2012, 109, 14194–14199. [Google Scholar] [CrossRef] [PubMed]
  54. Atre, A.N.; Surve, S.V.; Shouche, Y.S.; Joseph, J.; Patole, M.S.; Deopurkar, R.L. Association of small Rho GTPases and actin ring formation in epithelial cells during the invasion by Candida albicans. FEMS Immunol. Med. Microbiol. 2009, 55, 74–84. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, Y.; Shetty, A.C.; Schwartz, J.A.; Bradford, L.L.; Xu, W.; Phan, Q.T.; Kumari, P.; Mahurkar, A.; Mitchell, A.P.; Ravel, J.; et al. New signaling pathways govern the host response to C. albicans infection in various niches. Genome Res. 2015, 25, 679–689. [Google Scholar] [CrossRef] [PubMed]
  56. Solis, N.V.; Swidergall, M.; Bruno, V.M.; Gaffen, S.L.; Filler, S.G. The aryl hydrocarbon receptor governs epithelial cell invasion during oropharyngeal candidiasis. mBio 2017, 8. [Google Scholar] [CrossRef] [PubMed]
  57. Glass, K.A.; Longley, S.J.; Bliss, J.M.; Shaw, S.K. Protection of Candida parapsilosis from neutrophil killing through internalization by human endothelial cells. Virulence 2015, 6, 504–514. [Google Scholar] [CrossRef] [PubMed]
  58. Villar, C.C.; Kashleva, H.; Nobile, C.J.; Mitchell, A.P.; Dongari-Bagtzoglou, A. Mucosal tissue invasion by Candida albicans is associated with E-cadherin degradation, mediated by transcription factor Rim101p and protease Sap5p. Infect. Immun. 2007, 75, 2126–2135. [Google Scholar] [CrossRef] [PubMed]
  59. Colina, A.R.; Aumont, F.; Deslauriers, N.; Belhumeur, P.; de Repentigny, L. Evidence for degradation of gastrointestinal mucin by Candida albicans secretory aspartyl proteinase. Infect. Immun. 1996, 64, 4514–4519. [Google Scholar] [PubMed]
  60. Dalle, F.; Wachtler, B.; L’Ollivier, C.; Holland, G.; Bannert, N.; Wilson, D.; Labruere, C.; Bonnin, A.; Hube, B. Cellular interactions of Candida albicans with human oral epithelial cells and enterocytes. Cell. Microbiol. 2010, 12, 248–271. [Google Scholar] [CrossRef] [PubMed]
  61. Wachtler, B.; Wilson, D.; Haedicke, K.; Dalle, F.; Hube, B. From attachment to damage: Defined genes of Candida albicans mediate adhesion, invasion and damage during interaction with oral epithelial cells. PLoS ONE 2011, 6, e17046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Bahri, R.; Curt, S.; Saidane-Mosbahi, D.; Rouabhia, M. Normal human gingival epithelial cells sense C. parapsilosis by toll-like receptors and module its pathogenesis through antimicrobial peptides and proinflammatory cytokines. Mediat. Inflamm. 2010, 2010, 940383. [Google Scholar] [CrossRef] [PubMed]
  63. Silva, S.; Henriques, M.; Oliveira, R.; Azeredo, J.; Malic, S.; Hooper, S.J.; Williams, D.W. Characterization of Candida parapsilosis infection of an in vitro reconstituted human oral epithelium. Eur. J. Oral Sci. 2009, 117, 669–675. [Google Scholar] [CrossRef] [PubMed]
  64. Silva, S.; Hooper, S.J.; Henriques, M.; Oliveira, R.; Azeredo, J.; Williams, D.W. The role of secreted aspartyl proteinases in Candida tropicalis invasion and damage of oral mucosa. Clin. Microbiol. Infect. 2011, 17, 264–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Moyes, D.L.; Runglall, M.; Murciano, C.; Shen, C.; Nayar, D.; Thavaraj, S.; Kohli, A.; Islam, A.; Mora-Montes, H.; Challacombe, S.J.; et al. A biphasic innate immune MAPK response discriminates between the yeast and hyphal forms of Candida albicans in epithelial cells. Cell Host Microbe 2010, 8, 225–235. [Google Scholar] [CrossRef] [PubMed]
  66. Moyes, D.L.; Shen, C.; Murciano, C.; Runglall, M.; Richardson, J.P.; Arno, M.; Aldecoa-Otalora, E.; Naglik, J.R. Protection against epithelial damage during Candida albicans infection is mediated by PI3K/AKT and mammalian target of rapamycin signaling. J. Infect. Dis. 2014, 209, 1816–1826. [Google Scholar] [CrossRef] [PubMed]
  67. Weindl, G.; Naglik, J.R.; Kaesler, S.; Biedermann, T.; Hube, B.; Korting, H.C.; Schaller, M. Human epithelial cells establish direct antifungal defense through TLR4-mediated signaling. J. Clin. Investig. 2007, 117, 3664–3672. [Google Scholar] [CrossRef] [PubMed]
  68. Decanis, N.; Savignac, K.; Rouabhia, M. Farnesol promotes epithelial cell defense against Candida albicans through toll-like receptor 2 expression, interleukin-6 and human β-defensin 2 production. Cytokine 2009, 45, 132–140. [Google Scholar] [CrossRef] [PubMed]
  69. Netea, M.G.; Brown, G.D.; Kullberg, B.J.; Gow, N.A. An integrated model of the recognition of Candida albicans by the innate immune system. Nat. Rev. Microbiol. 2008, 6, 67–78. [Google Scholar] [CrossRef] [PubMed]
  70. Netea, M.G.; Joosten, L.A.; van der Meer, J.W.; Kullberg, B.J.; van de Veerdonk, F.L. Immune defence against Candida fungal infections. Nat. Rev. Immunol. 2015, 15, 630–642. [Google Scholar] [CrossRef] [PubMed]
  71. Moyes, D.L.; Wilson, D.; Richardson, J.P.; Mogavero, S.; Tang, S.X.; Wernecke, J.; Hofs, S.; Gratacap, R.L.; Robbins, J.; Runglall, M.; et al. Candidalysin is a fungal peptide toxin critical for mucosal infection. Nature 2016, 532, 64–68. [Google Scholar] [CrossRef] [PubMed]
  72. Swidergall, M.; Solis, N.V.; Lionakis, M.S.; Filler, S.G. Epha2 is an epithelial cell pattern recognition receptor for fungal β-glucans. Nat. Microbiol. 2018, 3, 53–61. [Google Scholar] [CrossRef] [PubMed]
  73. Brown, G.D.; Gordon, S. Immune recognition. A new receptor for β-glucans. Nature 2001, 413, 36–37. [Google Scholar] [CrossRef] [PubMed]
  74. Taylor, P.R.; Tsoni, S.V.; Willment, J.A.; Dennehy, K.M.; Rosas, M.; Findon, H.; Haynes, K.; Steele, C.; Botto, M.; Gordon, S.; et al. Dectin-1 is required for β-glucan recognition and control of fungal infection. Nat. Immunol. 2007, 8, 31–38. [Google Scholar] [CrossRef] [PubMed]
  75. Reid, D.M.; Gow, N.A.; Brown, G.D. Pattern recognition: Recent insights from Dectin-1. Curr. Opin. Immunol. 2009, 21, 30–37. [Google Scholar] [CrossRef] [PubMed]
  76. Plato, A.; Willment, J.A.; Brown, G.D. C-type lectin-like receptors of the Dectin-1 cluster: Ligands and signaling pathways. Int. Rev. Immunol. 2013, 32, 134–156. [Google Scholar] [CrossRef] [PubMed]
  77. Verma, A.H.; Richardson, J.P.; Zhou, C.; Coleman, B.M.; Moyes, D.L.; Ho, J.; Huppler, A.R.; Ramani, K.; McGeachy, M.J.; Mufazalov, I.A.; et al. Oral epithelial cells orchestrate innate type 17 responses to Candida albicans through the virulence factor Candidalysin. Sci. Immunol. 2017, 2. [Google Scholar] [CrossRef] [PubMed]
  78. Zaugg, C.; Borg-Von Zepelin, M.; Reichard, U.; Sanglard, D.; Monod, M. Secreted aspartic proteinase family of Candida tropicalis. Infect. Immun. 2001, 69, 405–412. [Google Scholar] [CrossRef] [PubMed]
  79. De Viragh, P.A.; Sanglard, D.; Togni, G.; Falchetto, R.; Monod, M. Cloning and sequencing of two Candida parapsilosis genes encoding acid proteases. J. Gen. Microbiol. 1993, 139, 335–342. [Google Scholar] [CrossRef] [PubMed]
  80. Fusek, M.; Smith, E.A.; Monod, M.; Foundling, S.I. Candida parapsilosis expresses and secretes two aspartic proteinases. FEBS Lett. 1993, 327, 108–112. [Google Scholar] [CrossRef]
  81. Merkerova, M.; Dostal, J.; Hradilek, M.; Pichova, I.; Hruskova-Heidingsfeldova, O. Cloning and characterization of Sapp2p, the second aspartic proteinase isoenzyme from Candida parapsilosis. FEMS Yeast Res. 2006, 6, 1018–1026. [Google Scholar] [CrossRef] [PubMed]
  82. Watts, H.J.; Cheah, F.S.; Hube, B.; Sanglard, D.; Gow, N.A. Altered adherence in strains of Candida albicans harbouring null mutations in secreted aspartic proteinase genes. FEMS Microbiol. Lett. 1998, 159, 129–135. [Google Scholar] [CrossRef] [PubMed]
  83. Naglik, J.R.; Challacombe, S.J.; Hube, B. Candida albicans secreted aspartyl proteinases in virulence and pathogenesis. Microbiol. Mol. Biol. Rev. 2003, 67, 400–428. [Google Scholar] [CrossRef] [PubMed]
  84. Barrett-Bee, K.; Hayes, Y.; Wilson, R.G.; Ryley, J.F. A comparison of phospholipase activity, cellular adherence and pathogenicity of yeasts. J. Gen. Microbiol. 1985, 131, 1217–1221. [Google Scholar] [CrossRef] [PubMed]
  85. Ghannoum, M.A. Potential role of phospholipases in virulence and fungal pathogenesis. Clin. Microbiol. Rev. 2000, 13, 122–143. [Google Scholar] [CrossRef] [PubMed]
  86. Larkin, E.; Hager, C.; Chandra, J.; Mukherjee, P.K.; Retuerto, M.; Salem, I.; Long, L.; Isham, N.; Kovanda, L.; Borroto-Esoda, K.; et al. The emerging pathogen Candida auris: Growth phenotype, virulence factors, activity of antifungals, and effect of SCY-078, a novel glucan synthesis inhibitor, on growth morphology and biofilm formation. Antimicrob. Agents Chemother. 2017, 61. [Google Scholar] [CrossRef] [PubMed]
  87. Gacser, A.; Trofa, D.; Schafer, W.; Nosanchuk, J.D. Targeted gene deletion in Candida parapsilosis demonstrates the role of secreted lipase in virulence. J. Clin. Investig. 2007, 117, 3049–3058. [Google Scholar] [CrossRef] [PubMed]
  88. Almeida, R.S.; Brunke, S.; Albrecht, A.; Thewes, S.; Laue, M.; Edwards, J.E.; Filler, S.G.; Hube, B. The hyphal-associated adhesin and invasin Als3 of Candida albicans mediates iron acquisition from host ferritin. PLoS Pathog. 2008, 4, e1000217. [Google Scholar] [CrossRef] [PubMed]
  89. Weissman, Z.; Kornitzer, D. A family of Candida cell surface haem-binding proteins involved in haemin and haemoglobin-iron utilization. Mol. Microbiol. 2004, 53, 1209–1220. [Google Scholar] [CrossRef] [PubMed]
  90. Weissman, Z.; Shemer, R.; Conibear, E.; Kornitzer, D. An endocytic mechanism for haemoglobin-iron acquisition in Candida albicans. Mol. Microbiol. 2008, 69, 201–217. [Google Scholar] [CrossRef] [PubMed]
  91. Ding, C.; Vidanes, G.M.; Maguire, S.L.; Guida, A.; Synnott, J.M.; Andes, D.R.; Butler, G. Conserved and divergent roles of Bcr1 and CFEM proteins in Candida parapsilosis and Candida albicans. PLoS ONE 2011, 6, e28151. [Google Scholar] [CrossRef] [PubMed]
  92. Ballou, E.R.; Wilson, D. The roles of zinc and copper sensing in fungal pathogenesis. Curr. Opin. Microbiol. 2016, 32, 128–134. [Google Scholar] [CrossRef] [PubMed]
  93. Malavia, D.; Crawford, A.; Wilson, D. Nutritional immunity and fungal pathogenesis: The struggle for micronutrients at the host-pathogen interface. Adv. Microb. Physiol. 2017, 70, 85–103. [Google Scholar] [PubMed]
  94. Citiulo, F.; Jacobsen, I.D.; Miramon, P.; Schild, L.; Brunke, S.; Zipfel, P.; Brock, M.; Hube, B.; Wilson, D. Candida albicans scavenges host zinc via Pra1 during endothelial invasion. PLoS Pathog. 2012, 8, e1002777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Malavia, D.; Lehtovirta-Morley, L.E.; Alamir, O.; Weiss, E.; Gow, N.A.R.; Hube, B.; Wilson, D. Zinc limitation induces a hyper-adherent goliath phenotype in Candida albicans. Front. Microbiol. 2017, 8, 2238. [Google Scholar] [CrossRef] [PubMed]
  96. Allison, D.L.; Willems, H.M.; Jayatilake, J.A.; Bruno, V.M.; Peters, B.M.; Shirtliff, M.E. Candida-bacteria interactions: Their impact on human disease. Microbiol. Spectr. 2016, 4. [Google Scholar] [CrossRef]
  97. Forster, T.M.; Mogavero, S.; Drager, A.; Graf, K.; Polke, M.; Jacobsen, I.D.; Hube, B. Enemies and brothers in arms: Candida albicans and gram-positive bacteria. Cell. Microbiol. 2016, 18, 1709–1715. [Google Scholar] [CrossRef] [PubMed]
  98. Klotz, S.A.; Gaur, N.K.; De Armond, R.; Sheppard, D.; Khardori, N.; Edwards, J.E., Jr.; Lipke, P.N.; El-Azizi, M. Candida albicans Als proteins mediate aggregation with bacteria and yeasts. Med. Mycol. 2007, 45, 363–370. [Google Scholar] [CrossRef] [PubMed]
  99. Tati, S.; Davidow, P.; McCall, A.; Hwang-Wong, E.; Rojas, I.G.; Cormack, B.; Edgerton, M. Candida glabrata binding to Candida albicans hyphae enables its development in oropharyngeal candidiasis. PLoS Pathog. 2016, 12, e1005522. [Google Scholar] [CrossRef] [PubMed]
  100. Silverman, R.J.; Nobbs, A.H.; Vickerman, M.M.; Barbour, M.E.; Jenkinson, H.F. Interaction of Candida albicans cell wall Als3 protein with Streptococcus gordonii SspB adhesin promotes development of mixed-species communities. Infect. Immun. 2010, 78, 4644–4652. [Google Scholar] [CrossRef] [PubMed]
  101. Peters, B.M.; Ovchinnikova, E.S.; Krom, B.P.; Schlecht, L.M.; Zhou, H.; Hoyer, L.L.; Busscher, H.J.; van der Mei, H.C.; Jabra-Rizk, M.A.; Shirtliff, M.E. Staphylococcus aureus adherence to Candida albicans hyphae is mediated by the hyphal adhesin Als3p. Microbiology 2012, 158, 2975–2986. [Google Scholar] [CrossRef] [PubMed]
  102. Mailander-Sanchez, D.; Braunsdorf, C.; Grumaz, C.; Muller, C.; Lorenz, S.; Stevens, P.; Wagener, J.; Hebecker, B.; Hube, B.; Bracher, F.; et al. Antifungal defense of probiotic Lactobacillus rhamnosus GG is mediated by blocking adhesion and nutrient depletion. PLoS ONE 2017, 12, e0184438. [Google Scholar] [CrossRef] [PubMed]
  103. Schonherr, F.A.; Sparber, F.; Kirchner, F.R.; Guiducci, E.; Trautwein-Weidner, K.; Gladiator, A.; Sertour, N.; Hetzel, U.; Le, G.T.T.; Pavelka, N.; et al. The intraspecies diversity of Candida albicans triggers qualitatively and temporally distinct host responses that determine the balance between commensalism and pathogenicity. Mucosal Immunol. 2017, 10, 1335–1350. [Google Scholar] [CrossRef] [PubMed]
  104. Naglik, J.R.; Moyes, D.L.; Wachtler, B.; Hube, B. Candida albicans interactions with epithelial cells and mucosal immunity. Microbes Infect. 2011, 13, 963–976. [Google Scholar] [CrossRef] [PubMed]
  105. Li, L.; Kashleva, H.; Dongari-Bagtzoglou, A. Cytotoxic and cytokine-inducing properties of Candida glabrata in single and mixed oral infection models. Microb. Pathog. 2007, 42, 138–147. [Google Scholar] [CrossRef] [PubMed]
  106. Abu-Elteen, K.H.; Elkarmi, A.Z.; Hamad, M. Characterization of phenotype-based pathogenic determinants of various Candida albicans strains in Jordan. Jpn. J. Infect. Dis. 2001, 54, 229–236. [Google Scholar] [PubMed]
  107. De Bernardis, F.; Chiani, P.; Ciccozzi, M.; Pellegrini, G.; Ceddia, T.; D’Offizzi, G.; Quinti, I.; Sullivan, P.A.; Cassone, A. Elevated aspartic proteinase secretion and experimental pathogenicity of Candida albicans isolates from oral cavities of subjects infected with human immunodeficiency virus. Infect. Immun. 1996, 64, 466–471. [Google Scholar] [PubMed]
  108. Richardson, J.P.; Willems, H.M.E.; Moyes, D.L.; Shoaie, S.; Barker, K.S.; Tan, S.L.; Palmer, G.E.; Hube, B.; Naglik, J.R.; Peters, B.M. Candidalysin drives epithelial signaling, neutrophil recruitment, and immunopathology at the vaginal mucosa. Infect. Immun. 2017. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Summary of Candida and epithelial cell factors directly involved in attachment, entry, and damage to epithelial cells. (A) The prototypical Candida species C. albicans can attach to epithelial cells through numerous host cell receptors including EphA2 (via β-glucan) and E-cadherin (via Als3p). Host cell transglutaminases also cross-link C. albicans directly to the epithelial surface (via Hwp1p), whilst the non-filamentous C. glabrata can utilise Epa1p to bind host-cell glycans (asialo-lactosyl-containing carbohydrates). Additionally, electrostatic forces (dashed lines) contribute to the overall affinity between fungal and host cells; (B) C. albicans Als3p and Ssa1p interact with E-cadherin and EGFR/Her2 receptors to potentiate induced endocytosis; (C) several Candida species such as C. albicans, C. dubliniensis, and C. tropicalis secrete factors to actively penetrate mucosal tissues, including aspartic proteinases (Sap2p, Sap5p), lipases, phospholipases, and Candidalysin, predominantly from hyphae.
Figure 1. Summary of Candida and epithelial cell factors directly involved in attachment, entry, and damage to epithelial cells. (A) The prototypical Candida species C. albicans can attach to epithelial cells through numerous host cell receptors including EphA2 (via β-glucan) and E-cadherin (via Als3p). Host cell transglutaminases also cross-link C. albicans directly to the epithelial surface (via Hwp1p), whilst the non-filamentous C. glabrata can utilise Epa1p to bind host-cell glycans (asialo-lactosyl-containing carbohydrates). Additionally, electrostatic forces (dashed lines) contribute to the overall affinity between fungal and host cells; (B) C. albicans Als3p and Ssa1p interact with E-cadherin and EGFR/Her2 receptors to potentiate induced endocytosis; (C) several Candida species such as C. albicans, C. dubliniensis, and C. tropicalis secrete factors to actively penetrate mucosal tissues, including aspartic proteinases (Sap2p, Sap5p), lipases, phospholipases, and Candidalysin, predominantly from hyphae.
Jof 04 00022 g001
Table 1. Candida species adhesins and their role in epithelial attachment.
Table 1. Candida species adhesins and their role in epithelial attachment.
SpeciesGeneFunctionEpithelial Adhesion of Null MutantEpithelial Cell TypeReference
C. albicansALS1AdhesinDecreasedTongue[25]
ALS2AdhesinDecreased *Reconstituted human oral epithelium[23]
ALS3Adhesin (hypha-associated)DecreasedBuccal[22]
ALS5-7AdhesinIncreasedBuccal[24]
EAP1AdhesinDecreasedHEK293[34]
HWP1Cell wall protein (hypha-associated)DecreasedBuccal[13]
HWP2Cell wall proteinDecreasedHT-29[16]
SAP9Aspartic proteinaseIncreasedBuccal[36]
SAP10Aspartic proteinaseDecreasedBuccal[36]
C. glabrataEPA1AdhesinDecreasedLaryngeal, Hamster ovary[38]
EPA6AdhesinOverexpression in S. cerevisiae confers adhesionLec2[42]
EPA7AdhesinOverexpression in S. cerevisiae confers adhesionLec2[42]
C. parapsilosisCPAR2_404800AdhesinDecreasedBuccal[29]
* Heterozygous knockout only (als2Δ/ALS2).

Share and Cite

MDPI and ACS Style

Richardson, J.P.; Ho, J.; Naglik, J.R. Candida–Epithelial Interactions. J. Fungi 2018, 4, 22. https://doi.org/10.3390/jof4010022

AMA Style

Richardson JP, Ho J, Naglik JR. Candida–Epithelial Interactions. Journal of Fungi. 2018; 4(1):22. https://doi.org/10.3390/jof4010022

Chicago/Turabian Style

Richardson, Jonathan P., Jemima Ho, and Julian R. Naglik. 2018. "Candida–Epithelial Interactions" Journal of Fungi 4, no. 1: 22. https://doi.org/10.3390/jof4010022

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop