Abstract
Candida albicans is both a member of the human mucosal microbiota and a common agent of invasive fungal disease. Systems biology approaches allow for analysis of the interactions between this fungus and its mammalian host. Framing these studies by considering how C. albicans and its host construct the niche the other occupies provides insight into how these interactions shape the ecosystems, behavior, and evolution of each organism. Here, we discuss recent work on multiscale systems biology approaches for examining C. albicans in relation to the host ecosystem to identify the emergent properties of the interactions and new variables that can be targeted for development of therapeutic strategies.
Keywords: Candida albicans, Transcriptomics, functional genomics, microbial interactions, proteomics, screening, experimental evolution
Introduction
Systems biology is an integrative approach for studying biology that combines information from multiple levels to allow for the identification of emergent properties of the system [1,2]. A challenge of applying systems analysis to understanding biology is that organisms are not closed systems. Iterated interactions between organisms and their environments shape both the traits that organisms express in the short term and their evolutionary trajectories. When the environments that organisms respond to include other individuals or species, the potential for complex ecological and evolutionary feedback loops arises [3].
Microbe-host interactions provide a rich opportunity to study dynamics of eco-evolutionary feedback loops and their consequences for human health and disease. Candida albicans is an example of a eukaryotic microbe found in association with mammalian hosts at multiple body sites, and it is capable of both commensal colonization and invasive disease [4–7]. Fully understanding the complexity of C. albicans biology requires studying the interactions between this microbe and its complex ecosystem.
Niche construction is one framework for analyzing how the interactions among C. albicans, host, and microbiota co-determine the trajectory of the systems they all share. Niche construction is the idea that organisms both inhabit and modify their environments with consequences at ecological and evolutionary timescales [8] (Figure 1 and Table 1). Examples of niche construction include engineering of the abiotic environment, as in Candida alkalinization of extracellular pH during carbon limitation [9] or extrusion of extracellular matrix during biofilm formation [10,11], and biotic interactions, for example, the effects of C. albicans intestinal colonization on mediating immune regulation of the microbiota [4,12–14]. A key feature of niche construction is that changes produced in the environment are non-random and generate systematic selection pressures that lead to adaptation [8]. Systems biology approaches can uncover the diverse relationships between C. albicans and their hosts and provide a framework for analyzing these changing relationships across niches and time (Figure 1 and Table 1). Here we review recent literature elucidating Candida albicans interactions with host ecosystems at multiple scales with special attention to novel methods that will contribute to an understanding of the dynamic interactions, eco-evolutionary feedback loops, and emergent properties at work in these systems.
Figure 1: Schematic of Candida albicans and the host ecosystem.

Niche construction is the idea that organisms systemically remodel their environments by living in them, which can change the selection pressures operating in those environments. C. albicans (represented here by the purple yeast) is a eukaryotic microbe associated with mammalian hosts (large blue circle) that can inhabit many different body sites aka ecological niches in the host. Each of these niches has a characteristic set of abiotic conditions (e.g. pH, humidity, nutrients) and biotic conditions (e.g. microbial flora, immune cells) that can influence C. albicans growth and behavior. In response, C. albicans can shape these niches by altering local chemistry, competing with microbes, driving immune responses, and influencing immune surveillance. The effects of these niche modifications, whether that is the establishment of a persistent multispecies biofilm or training of host immunity, have the potential to outlast the life cycle of any single cell by many generations and generate systematic selection pressures on all the players in the niche. Thus, the interconnected feedback loops between C. albicans and the host demonstrate the importance of studying the interactions of the organism to understand their behavior and evolution.
Table 1: Multi-scale systems biology.
Systems biology approaches can capture a snapshot of the interactions between organisms. By using multi-scale approaches, it is possible to build a fuller picture of the relationships. Each set of tools can uncover different aspects of the interactions.
| Timescale | Evolutionary Time | Generation Time | Organismal Time |
|---|---|---|---|
| Unit of measure | Genomes | Communities | Molecules/Cells |
| Experimental Approaches | Experimental Evolution, Competition Assays, Natural variation | Microbial community composition, Competition assays, Functional Genomic Screens | Transcriptomics, Proteomics, Microscopy |
Microbial community dynamics
An integrative understanding of how C. albicans fits into its ecological context takes into account both the host ecosystem and the varied impacts of diverse microbes that C. albicans interacts with in situ [15]. These interactions between microbes can be antagonistic, mutualistic, or neutral, with varying impacts on host health. Better understanding of these interactions, and how these microbial players influence each other and the host, will be helpful in identifying variables that can be selectively perturbed to improve clinical outcomes.
One outcome of microbial interactions is synergism or mutualism between microbes. For example, C. albicans produces biofilms [16] that can offer surfaces for bacterial growth [17] and shelter from antibiotics [10]. Biofilms also form surfaces where cross-feeding interactions take place between Candida and bacteria [11]. In these examples, the host is damaged by microbial synergism. However, synergistic interactions may also offer new targets for clinical intervention when microbial communities depend on one another: targeting one member--the “weakest link”--could interfere with the entire relationship or community [18]. This strategy highlights the importance of understanding interdependent relationships in microbial communities.
Interactions among microbes can also antagonize pathogenicity. Bacterially-produced metabolites and peptides can interfere with C. albicans virulence [19–23]. For example, Enterococcus faecalis EntV peptide inhibits filamentation and biofilm development [19,24]. As E. faecalis and C. albicans co-occur in the GI, this interaction may promote C. albicans commensal growth and limit hyphal invasion [25]. Examining the interaction between C. albicans and additional gut commensal microbes identified Roseburia spp. and Bacteroides ovatus as organisms that can limit C. albicans growth [20]. Other microbial interactions can directly kill fungal cells, such as when Serratia marcescens deploys the Type VI secretion system [23], or Pseudomonas aeruginosa secretes phenazines [26,27]. Competition for resources or host niches may drive this antagonism, though bacterial use of fungal cell wall components as a nutrient [28–31] suggests the potential for parasitic or predatory interactions.
In vivo mouse studies provide an opportunity to understand microbial interaction in the context of a host, however, murine models do not always fully recapitulate human microbial communities. Lab mice do not natively possess C. albicans in their GI tracts and generally require antibiotic depletion or dietary changes to allow for stable fungal colonization [6,32], highlighting the role of lab-adapted microbiota in limiting C. albicans colonization. This colonization resistance has been attributed to Bacteroides regulation of HIF-1 [25,33]. A recent exciting development is that lab mice introduced to naturalistic contexts (“re-wilded”) acquire microbiomes that both include more fungi and induce immune states more closely resembling those of wild mice and humans [13]. Moving towards models like re-wilded mice [13] or more complex models of human tissues in culture with microbiota [22] that more faithfully reproduce the relevant systems will be an important source of new insight.
Transcriptomics
Profiling Candida transcriptomes in different environmental contexts and time points in colonization and infection has offered useful information on the key mechanisms this microbe uses to persist and thrive in diverse niches. Transcriptional analysis of fungi during infection can identify proteins that are specifically used to respond to and manipulate the host, for example the putative effector protein, Sel1, which is sufficient to induce an inflammatory response via signaling through TLR2 and TLR4 [34]. Dual RNA-seq, in which transcripts from the microbe and host are captured simultaneously, permits analysis of the interaction during infection. Dual RNA-seq during a time course of infection showed an upregulation of genes involved in glycolysis in both species with the ability of fungal cells to outcompete macrophages for glucose resulting in macrophage starvation and death [35]. Previous work on C. albicans growth in macrophages demonstrated the importance of the glyoxylate cycle and metabolic plasticity for virulence [36,37]; this work shows that fungal growth requires this pathway and that the competition impairs host cell survival. To further dissect the interplay between macrophage and C. albicans gene expression during infection, Munoz and colleagues [38] performed dual RNA-seq on host and C. albicans cells separated into subpopulations based on infection stage. Stratifying transcriptional signatures over the course of phagocytosis and infection allowed the authors to describe the trajectory of macrophage and microbe transcriptomes. Although the bulk of Candida-host experiments have been performed in macrophages, neutrophils also play a major role in anti-Candida responses. In neutrophils, the signature of infection is changes in cell signaling pathways [39], with enrichment in expression of genes related to the NLRP3 inflammasome. Current single-cell sequencing approaches to fungal infection have focused on host responses during gastrointestinal colonization [40] or at lymph nodes [41]. Future work using single-cell transcriptional analysis of both the fungus and the host during more realistic models will be vital for understanding the dynamic interactions that drive infection.
Proteomics
Proteomic studies complement transcriptomic studies by providing direct measures of the proteins present in a cell and can be used to measure protein abundance changes upon infection. In concordance with transcriptional data, the proteomic signature upon infection includes an increase in fatty acid beta oxidation and glyoxylate cycle proteins [42,43]. Proteomic analysis also revealed an increase in C. albicans phosphatidylethanolamine synthesis proteins [42] that are required for systemic infections [44]. Potentially, additional processes regulated upon infection can be targeted for novel antifungal development. A limitation of proteomic approaches is that lower abundance proteins are difficult to identify and quantify, although these proteins might be crucial players in mediating the interaction.
In addition to quantifying the proteins present under a particular condition, proteomic studies can be used to identify protein-protein interactions (PPIs), thus giving insight into functional relationships between proteins. One approach is to use affinity-purification mass spectrometry (AP-MS) to identify changes in abundance of an interaction upon an environmental stress, such as treatment with an antifungal drug [45], or during an infection. Another recently-developed approach for identifying protein complexes is to use non-denaturing co-fractionation followed by mass spectrometry to identify proteins that co-purify with each other [45,46]; when this is combined with stable isotope labeling, it is possible to obtain quantitative changes in interactions. Protein-protein interactions can also occur between microbial and host proteins (as reviewed in [47] for bacterial pathogens), and the development of C. albicans proteomic resources will soon allow for analysis of these interactions.
Functional genomics approaches
Functional genomics couples forward and reverse genetics by screening libraries of defined mutants for phenotypes of interest. In vivo functional genomics screens can reveal mutants that only display a phenotype during infection. The pioneering in vivo screen focused on C. albicans mutants with defects in systemic infection and identified that fungal sphingolipids are required during systemic disease but dispensable for in vitro growth, highlighting the utility of this approach [48]. However, hosts contain many microenvironments, and pinpointing which processes are required during a particular interaction can be challenging. This is particularly true for C. albicans, which can occupy many niches within a host and have a variety of relationships with that host.
To answer this challenge, researchers have designed screens to isolate particular components of in vivo fitness. For example, during invasive disease C. albicans must translocate across epithelial barriers. Using epithelial cells as a model of infection, Allert and colleagues screened fungal mutants for defects in translocation; by examining both host and fungus in parallel, they were able to show that translocation is a fungus-driven process [49]. The unbiased screen implicated multiple genes with unknown function in regulating this in vivo phenotype, suggesting new mechanisms of action. Additional models of in vivo infection will allow for a deeper understanding of how specific C. albicans genes contribute to fitness during systemic infection.
Candida albicans is also a colonizer of the gut, and the processes required for colonization are not necessarily the same processes required for causing invasive disease. Therefore, multiple studies have used mutant screens to identify genes required for colonization in the murine gut [50,51]. One surprising result was that ablating expression of several C. albicans genes conferred increased fitness over wild type during gut colonization, suggesting that C. albicans maintains genes whose fitness benefit in another environment trades off with fitness in the murine gut, or, possibly, that these genes function differently in the animal model than in the human GI tract. In deletion mutant screens, multiple yeast-locked mutants were hypercompetitive in the GI tract [32,50]; one hypothesis is that hyphal cells express proteases and other virulence factors that are a liability during colonization [50]. Alternately, bacteria could drive filamentation in the gut: C. albicans adopts a yeast form when colonizing gnotobiotic mice [32] but exists as a mixture of yeast and hyphae in antibiotic-treated mice [50,52]. Notably, ume6 and tec1 deletion mutants show morphogenesis defects during in vitro but not in vivo gut colonization, demonstrating that the signals that drive filamentation in the host are not fully recapitulated by in vitro conditions. Additionally, the GI tract comprises multiple environments potentially requiring differential fungal responses: overexpression of CRZ2 permits higher colonization of the stomach and upper GI presumably due to improved adaptation to acidic pH and bile salts [51]. These studies highlight the importance of multi-scale systems biology approaches that identify host environments and microbiota contributing to changes in fitness among mutant strains.
Experimental evolution
Experimental evolution approaches complement functional genomic experiments by identifying the genetic and phenotypic changes resulting from applying a selection pressure of interest over multiple generations. Selecting on naturally occurring variation in a population or among defined sets of genotypes permits unbiased identification of variants that increase fitness in a particular context. To examine adaptation to host environments, three recent studies described the genetic and phenotypic changes resulting from serial passaging of C. albicans through mice [7,53]. Consistent with the functional screen reported above, Tso and colleagues showed that evolving strains through antibiotic-treated mouse GI tracts for 10 weeks resulted in strains with defects in filamentation on agar and reduced virulence in systemic infection [7]. The authors propose that filamentation is costly but required for competition with gut-resident bacteria; the mechanism by which bacteria select for hyphae is currently unknown. In addition to mutations in transcription factors known to regulate filamentation, they observed multiple instances of loss of heterozygosity and aneuploidy, especially on Chromosome 7. While strains evolved in mice show up to 11x higher mutation rates than those evolved in vitro, a second study also recovered Chromosome 7 trisomies from multiple C. albicans genotypes passaged through mouse GI tracts in the presence of antibiotics. Direct competition assays showed that the trisomic strain exhibited increased fitness in antibiotic-treated mice [54]. Notably, passage of C. albicans through either mouse kidneys or the gut resulted in different suites of genomic changes, suggesting that different host niches may favor different evolved strategies and/or that different host environments could bias towards certain mutational possibilities. Consistent with this hypothesis, strains passaged through an oropharyngeal candidiasis model acquired trisomies on Chromosome 5 and 6 [53]. One striking observation is that global isolates of C. albicans collected from humans show less frequent aneuploidy than the experimentally-evolved lines [55], suggesting that selection over longer timescales may refine large effect mutations.
Microscopy-based approaches
Systems biology does not necessarily mean ‘omics approaches; instead, the goal is to use techniques where a range of properties can be examined to determine which will be the most informative. One such approach is detailed microscopy, which can reveal new aspects of the C. albicans-host interactions. A benefit of microscopy approaches is the single-cell resolution of features that are lost during bulk culture approaches, including confounding factors such as variation in phagocytosis rates, changes in morphology, or stochasticity in responses.
Recent work demonstrated that C. albicans interactions with host macrophages can result in a pyroptotic host cell death [56–59]. Time-lapse microscopy revealed that there are two stages of host cell death after phagocytosis with macrophages [57,60]; the first is NLRP3-dependent, and the second phase results from competition for glucose [35]. These microscopy-based approaches also revealed that hyphal formation is not sufficient for the induction of pyroptosis [58,60–62]. Without analyzing both the fungus and the host at single-interaction levels, it would not be possible to decouple the hyphal transition from the induction of host death. However, the transition from yeast to filamentous form within macrophages can drive additional processes that are linked with host cell death, such as the production of the cytolytic toxic peptide Candidalyin [63] or physical pressure driven by the elongating hyphae causing phagosomal membrane damage [64]. Microscopy approaches can also be coupled with functional genomic screens, such as with screens for mutants with defects in pyroptosis. Large-scale screens of a filamentous S. cerevisiae library or a tetracycline-repressible C. albicans library demonstrated that components of the fungal cell surface, such as ergosterol or fungal cell wall proteins, are required to drive the host cell death response [62,65,66]. The exposure of these components is regulated by interaction with host cells [66], highlighting the dynamic interactions between microbe and macrophage.
Recent work has used tissue clearing and light-sheet microscopy approaches for examining in vivo fungal infections and the influence of specific immune defects on fungal infections [67,68]. In Aspergillus fumigatus infections of murine lungs, these techniques showed that most fungal conidia did not reach the alveoli and were instead concentrated in the bronchioles [68], altering our understanding of which host cells and processes are important to these infections. Using this approach to examine C. albicans colonization and infection in situ at the whole-organ level will reveal new aspects of fungal interactions with the host.
Conclusions
Although it can cause life-threatening systemic infections, C. albicans is also an abundant fungal species colonizing mammalian mucosal surfaces. As part of this colonization, C. albicans may play a major role in continuously shaping immune surveillance and the host microbiome throughout the lifetime of the host. There is not a singular host-pathogen conflict, but multiple interfaces and interactions between these organisms at multiple levels. As the studies discussed above show, the development of technological advances allows for a deeper analysis of the eco-evolutionary feedback loops between Candida albicans and its host ecosystems. By studying both organisms in parallel during their interactions, it is possible to identify how they shape each other, both in the short term and in their evolutionary trajectories. Further studies to build more complex and representational models are an exciting direction for future research.
Highlights.
Interactions between Candida albicans and human hosts shape each other’s biology.
Systems biology provides tools to interrogate these interactions at multiple scales.
Microscopy, transcriptomics, and proteomics illustrate individual cell responses.
Functional genomics screens and experimental evolution reveal genotypes that impact fitness.
Microbial community interactions can modify each other and host health.
Acknowledgement:
We apologize to all the research that we could not include for space limitations. We thank the members of the O’Meara lab for helpful comments. TRO is funded by KAI137299 (NIAID).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declaration of interests
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References:
- 1.Westerhoff HV, Palsson BO: The evolution of molecular biology into systems biology. Nat Biotechnol 2004, 22:1249–1252. [DOI] [PubMed] [Google Scholar]
- 2.Aderem A: Systems biology: its practice and challenges. Cell 2005, 121:511–513. [DOI] [PubMed] [Google Scholar]
- 3.Post DM, Palkovacs EP: Eco-evolutionary feedbacks in community and ecosystem ecology: interactions between the ecological theatre and the evolutionary play. Philos Trans R Soc Lond B Biol Sci 2009, 364:1629–1640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. *.Shao T-Y, Ang WXG, Jiang TT, Huang FS, Andersen H, Kinder JM, Pham G, Burg AR, Ruff B, Gonzalez T, et al. : Commensal Candida albicans Positively Calibrates Systemic Th17 Immunological Responses. Cell Host Microbe 2019, 25:404–417.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper demonstrates the influence of C. albicans on modulating host immune development
- 5.Brown GD, Denning DW, Gow NAR, Levitz SM, Netea MG, White TC: Hidden killers: human fungal infections. Sci Transl Med 2012, 4:165rv13–165rv13. [DOI] [PubMed] [Google Scholar]
- 6.Mishra AA, Koh AY: Adaptation of Candida albicans during gastrointestinal tract colonization. Curr Clin Microbiol Rep 2018, 5:165–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tso GHW, Reales-Calderon JA, Tan ASM, Sem X, Le GTT, Tan TG, Lai GC, Srinivasan KG, Yurieva M, Liao W, et al. : Experimental evolution of a fungal pathogen into a gut symbiont. Science 2018, 362:589–595. [DOI] [PubMed] [Google Scholar]
- 8.Laland K, Matthews B, Feldman MW: An introduction to niche construction theory. Evol Ecol 2016, 30:191–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Vylkova S, Carman AJ, Danhof HA, Collette JR, Zhou H, Lorenz MC: The fungal pathogen Candida albicans autoinduces hyphal morphogenesis by raising extracellular pH. MBio 2011, 2:e00055–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kong EF, Tsui C, Kucharíková S, Andes D, Van Dijck P, Jabra-Rizk MA: Commensal Protection of Staphylococcus aureus against Antimicrobials by Candida albicans Biofilm Matrix. MBio 2016, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. *.Ellepola K, Truong T, Liu Y, Lin Q, Lim TK, Lee YM, Cao T, Koo H, Seneviratne CJ: Multi-omics Analyses Reveal Synergistic Carbohydrate Metabolism in Streptococcus mutans-Candida albicans Mixed-Species Biofilms. Infect Immun 2019, 87. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is a comprehensive analysis of a microbial interaction.
- 12.Markey L, Shaban L, Green ER, Lemon KP, Mecsas J, Kumamoto CA: Pre-colonization with the commensal fungus Candida albicans reduces murine susceptibility to Clostridium difficile infection. Gut Microbes 2018, 9:497–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. *.Yeung F, Chen Y-H, Lin J-D, Leung JM, McCauley C, Devlin JC, Hansen C, Cronkite A, Stephens Z, Drake-Dunn C, et al. : Altered Immunity of Laboratory Mice in the Natural Environment Is Associated with Fungal Colonization. Cell Host Microbe 2020, 0. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper highlights the importance and representation of fungi in the microbiome, and indicates aspects of laboratory mice that do not recapitulate human biology.
- 14.Bacher P, Hohnstein T, Beerbaum E, Röcker M, Blango MG, Kaufmann S, Röhmel J, Eschenhagen P, Grehn C, Seidel K, et al. : Human Anti-fungal Th17 Immunity and Pathology Rely on Cross-Reactivity against Candida albicans. Cell 2019, 176:1340–1355.e15. [DOI] [PubMed] [Google Scholar]
- 15.Casadevall A, Pirofski L-A: What is a host? Incorporating the microbiota into the damage-response framework. Infect Immun 2015, 83:2–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gulati M, Nobile CJ: Candida albicans biofilms: development, regulation, and molecular mechanisms. Microbes Infect 2016, 18:310–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sztukowska MN, Dutton LC, Delaney C, Ramsdale M, Ramage G, Jenkinson HF, Nobbs AH, Lamont RJ: Community Development between Porphyromonas gingivalis and Candida albicans Mediated by InlJ and Als3. MBio 2018, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Adamowicz EM, Flynn J, Hunter RC, Harcombe WR: Cross-feeding modulates antibiotic tolerance in bacterial communities. ISME J 2018, 12:2723–2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Graham CE, Cruz MR, Garsin DA, Lorenz MC: Enterococcus faecalis bacteriocin EntV inhibits hyphal morphogenesis, biofilm formation, and virulence of Candida albicans. Proc Natl Acad Sci U S A 2017, 114:4507–4512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.García C, Tebbji F, Daigneault M, Liu N-N, Köhler JR, Allen-Vercoe E, Sellam A: The Human Gut Microbial Metabolome Modulates Fungal Growth via the TOR Signaling Pathway. mSphere 2017, 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Fourie R, Pohl CH: Beyond Antagonism: The Interaction Between Candida Species and Pseudomonas aeruginosa. J Fungi (Basel) 2019, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. **.Graf K, Last A, Gratz R, Allert S, Linde S, Westermann M, Gröger M, Mosig AS, Gresnigt MS, Hube B: Keeping Candida commensal: how lactobacilli antagonize pathogenicity of Candida albicans in an in vitro gut model. Dis Model Mech 2019, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper includes the host as mediator of microbial interaction and builds more realistic models for the interactions.
- 23.Trunk K, Peltier J, Liu Y-C, Dill BD, Walker L, Gow NAR, Stark MJR, Quinn J, Strahl H, Trost M, et al. : The type VI secretion system deploys antifungal effectors against microbial competitors. Nat Microbiol 2018, 3:920–931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Brown AO, Graham CE, Cruz MR, Singh KV, Murray BE, Lorenz MC, Garsin DA: Antifungal Activity of the Enterococcus faecalis Peptide EntV Requires Protease Cleavage and Disulfide Bond Formation. MBio 2019, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mason KL, Erb Downward JR, Falkowski NR, Young VB, Kao JY, Huffnagle GB: Interplay between the gastric bacterial microbiota and Candida albicans during postantibiotic recolonization and gastritis. Infect Immun 2012, 80:150–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Morales DK, Jacobs NJ, Rajamani S, Krishnamurthy M, Cubillos-Ruiz JR, Hogan DA: Antifungal mechanisms by which a novel Pseudomonas aeruginosa phenazine toxin kills Candida albicans in biofilms. Mol Microbiol 2010, 78:1379–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Morales DK, Grahl N, Okegbe C, Dietrich LEP, Jacobs NJ, Hogan DA: Control of Candida albicans metabolism and biofilm formation by Pseudomonas aeruginosa phenazines. MBio 2013, 4:e00526–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Abbott DW, Martens EC, Gilbert HJ, Cuskin F, Lowe EC: Coevolution of yeast mannan digestion: Convergence of the civilized human diet, distal gut microbiome, and host immunity. Gut Microbes 2015, 6:334–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Cuskin F, Lowe EC, Temple MJ, Zhu Y, Cameron E, Pudlo NA, Porter NT, Urs K, Thompson AJ, Cartmell A, et al. : Human gut Bacteroidetes can utilize yeast mannan through a selfish mechanism. Nature 2015, 517:165–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cuskin F, Baslé A, Ladevèze S, Day AM, Gilbert HJ, Davies GJ, Potocki-Véronèse G, Lowe EC: The GH130 Family of Mannoside Phosphorylases Contains Glycoside Hydrolases That Target β−1,2-Mannosidic Linkages in Candida Mannan. J Biol Chem 2015, 290:25023–25033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Temple MJ, Cuskin F, Baslé A, Hickey N, Speciale G, Williams SJ, Gilbert HJ, Lowe EC: A Bacteroidetes locus dedicated to fungal 1,6-β-glucan degradation: Unique substrate conformation drives specificity of the key endo-1,6-β-glucanase. J Biol Chem 2017, 292:10639–10650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Böhm L, Torsin S, Tint SH, Eckstein MT, Ludwig T, Pérez JC: The yeast form of the fungus Candida albicans promotes persistence in the gut of gnotobiotic mice. PLoS Pathog 2017, 13:e1006699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Fan D, Coughlin LA, Neubauer MM, Kim J, Kim MS, Zhan X, Simms-Waldrip TR, Xie Y, Hooper LV, Koh AY: Activation of HIF-1α and LL-37 by commensal bacteria inhibits Candida albicans colonization. Nat Med 2015, 21:808–814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wang W, Deng Z, Wu H, Zhao Q, Li T, Zhu W, Wang X, Tang L, Wang C, Cui S-Z, et al. : A small secreted protein triggers a TLR2/4-dependent inflammatory response during invasive Candida albicans infection. Nat Commun 2019, 10:1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. **.Tucey TM, Verma J, Harrison PF, Snelgrove SL, Lo TL, Scherer AK, Barugahare AA, Powell DR, Wheeler RT, Hickey MJ, et al. : Glucose Homeostasis Is Important for Immune Cell Viability during Candida Challenge and Host Survival of Systemic Fungal Infection. Cell Metab 2018, 27:988–1006.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper effectively used dual-RNAseq to identify mechanisms driving microbial pathogenesis within host cells.
- 36.Lorenz MC, Fink GR: The glyoxylate cycle is required for fungal virulence. Nature 2001, 412:83–86. [DOI] [PubMed] [Google Scholar]
- 37.Williams RB, Lorenz MC: Multiple Alternative Carbon Pathways Combine To Promote Candida albicans Stress Resistance, Immune Interactions, and Virulence. MBio 2020, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. **.Muñoz JF, Delorey T, Ford CB, Li BY, Thompson DA, Rao RP, Cuomo CA: Coordinated host-pathogen transcriptional dynamics revealed using sorted subpopulations and single macrophages infected with Candida albicans. Nat Commun 2019, 10:1607. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper highlights the different stages of infection that can occur between C. albicans and host cells.
- 39.Niemiec MJ, Grumaz C, Ermert D, Desel C, Shankar M, Lopes JP, Mills IG, Stevens P, Sohn K, Urban CF: Dual transcriptome of the immediate neutrophil and Candida albicans interplay. BMC Genomics 2017, 18:696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Singh N, Kim HC, Song R, Dhinsa JK, Torres SR, De Jesus M: Candida albicans Elicits Pro-Inflammatory Differential Gene Expression in Intestinal Peyer’s Patches. Mycopathologia 2019, 184:461–478. [DOI] [PubMed] [Google Scholar]
- 41.Blecher-Gonen R, Bost P, Hilligan KL, David E, Salame TM, Roussel E, Connor LM, Mayer JU, Bahar Halpern K, Tóth B, et al. : Single-Cell Analysis of Diverse Pathogen Responses Defines a Molecular Roadmap for Generating Antigen-Specific Immunity. Cell Syst 2019, 8:109–121.e6. [DOI] [PubMed] [Google Scholar]
- 42.Kitahara N, Morisaka H, Aoki W, Takeda Y, Shibasaki S, Kuroda K, Ueda M: Description of the interaction between Candida albicans and macrophages by mixed and quantitative proteome analysis without isolation. AMB Express 2015, 5:127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Cabezón V, Vialás V, Gil-Bona A, Reales-Calderón JA, Martinez-Gomariz M, Gutiérrez-Blázquez D, Monteoliva L, Molero G, Ramsdale M, Gil C: Apoptosis of Candida albicans during the interaction with murine macrophages: proteomics and cell-death marker monitoring. J Proteome Res 2016, 15:1418–1434. [DOI] [PubMed] [Google Scholar]
- 44.Chen Y-L, Montedonico AE, Kauffman S, Dunlap JR, Menn F-M, Reynolds TB: Phosphatidylserine synthase and phosphatidylserine decarboxylase are essential for cell wall integrity and virulence in Candida albicans. Mol Microbiol 2010, 75:1112–1132. [DOI] [PubMed] [Google Scholar]
- 45. *.O’Meara TR, O’Meara MJ, Polvi EJ, Pourhaghighi MR, Liston SD, Lin Z-Y, Veri AO, Emili A, Gingras A-C, Cowen LE: Global proteomic analyses define an environmentally contingent Hsp90 interactome and reveal chaperone-dependent regulation of stress granule proteins and the R2TP complex in a fungal pathogen. PLoS Biol 2019, 17:e3000358. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper developed new methods for quantitative proteomic analysis in C. albicans.
- 46.Pourhaghighi R, O’Meara TR, Cowen LE, Emili A: Protein-Protein Interaction Profiling in Candida albicans Revealed by Biochemical Purification-Mass Spectrometry (BP/MS) In Yeast Systems Biology: Methods and Protocols. Edited by Oliver SG, Castrillo JI. Springer; New York; 2019:203–211. [DOI] [PubMed] [Google Scholar]
- 47.Nicod C, Banaei-Esfahani A, Collins BC: Elucidation of host-pathogen protein-protein interactions to uncover mechanisms of host cell rewiring. Curr Opin Microbiol 2017, 39:7–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Noble SM, French S, Kohn LA, Chen V, Johnson AD: Systematic screens of a Candida albicans homozygous deletion library decouple morphogenetic switching and pathogenicity. Nat Genet 2010, 42:590–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Allert S, Förster TM, Svensson C-M, Richardson JP, Pawlik T, Hebecker B, Rudolphi S, Juraschitz M, Schaller M, Blagojevic M, et al. : Candida albicans-Induced Epithelial Damage Mediates Translocation through Intestinal Barriers. MBio 2018, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. *.Witchley JN, Penumetcha P, Abon NV, Woolford CA, Mitchell AP, Noble SM: Candida albicans Morphogenesis Programs Control the Balance between Gut Commensalism and Invasive Infection. Cell Host Microbe 2019, 25:432–443.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper was one of the largest mutant screens performed for GI colonization, and the development of FISH probes was key for visualizing morphology of C. albicans during colonization.
- 51.Znaidi S, van Wijlick L, Hernández-Cervantes A, Sertour N, Desseyn J-L, Vincent F, Atanassova R, Gouyer V, Munro CA, Bachellier-Bassi S, et al. : Systematic gene overexpression in Candida albicans identifies a regulator of early adaptation to the mammalian gut. Cell Microbiol 2018, 20:e12890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Witchley JN, Penumetcha PM, Noble SM: Visualization of Candida albicans in the Murine Gastrointestinal Tract Using Fluorescent In Situ Hybridization. J Vis Exp 2019, doi: 10.3791/60283. [DOI] [PubMed] [Google Scholar]
- 53. *.Forche A, Cromie G, Gerstein AC, Solis NV, Pisithkul T, Srifa W, Jeffery E, Abbey D, Filler SG, Dudley AM, et al. : Rapid Phenotypic and Genotypic Diversification After Exposure to the Oral Host Niche in Candida albicans. Genetics 2018, 209:725–741. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study uses experimental evolution to show large changes in genome structure upon entry into a host.
- 54. *.Ene IV, Farrer RA, Hirakawa MP, Agwamba K, Cuomo CA, Bennett RJ: Global analysis of mutations driving microevolution of a heterozygous diploid fungal pathogen. Proc Natl Acad Sci U S A 2018, 115:E8688–E8697. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study uses experimental evolution to compare different host models and C. albicans genotypes.
- 55. **.Ropars J, Maufrais C, Diogo D, Marcet-Houben M, Perin A, Sertour N, Mosca K, Permal E, Laval G, Bouchier C, et al. : Gene flow contributes to diversification of the major fungal pathogen Candida albicans. Nat Commun 2018, 9:2253. [DOI] [PMC free article] [PubMed] [Google Scholar]; This was one of the largest genome sequencing efforts for C. albicans natural isolates and shows the range of genotypes that have been selected for in nature.
- 56.Gross O, Poeck H, Bscheider M, Dostert C, Hannesschläger N, Endres S, Hartmann G, Tardivel A, Schweighoffer E, Tybulewicz V, et al. : Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature 2009, 459:433–436. [DOI] [PubMed] [Google Scholar]
- 57.Uwamahoro N, Verma-Gaur J, Shen H-H, Qu Y, Lewis R, Lu J, Bambery K, Masters SL, Vince JE, Naderer T, et al. : The pathogen Candida albicans hijacks pyroptosis for escape from macrophages. MBio 2014, 5:e00003–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wellington M, Koselny K, Sutterwala FS, Krysan DJ: Candida albicans triggers NLRP3-mediated pyroptosis in macrophages. Eukaryot Cell 2014, 13:329–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Joly S, Ma N, Sadler JJ, Soll DR, Cassel SL, Sutterwala FS: Cutting edge: Candida albicans hyphae formation triggers activation of the Nlrp3 inflammasome. J Immunol 2009, 183:3578–3581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Tucey TM, Verma-Gaur J, Nguyen J, Hewitt VL, Lo TL, Shingu-Vazquez M, Robertson AAB, Hill JR, Pettolino FA, Beddoe T, et al. : The Endoplasmic Reticulum-Mitochondrion Tether ERMES Orchestrates Fungal Immune Evasion, Illuminating Inflammasome Responses to Hyphal Signals. mSphere 2016, 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wellington M, Koselny K, Krysan DJ: Candida albicans morphogenesis is not required for macrophage interleukin 1β production. MBio 2012, 4:e00433–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. **.O’Meara TR, Duah K, Guo CX, Maxson ME, Gaudet RG, Koselny K, Wellington M, Powers ME, MacAlpine J, O’Meara MJ, et al. : High-throughput screening identifies genes required for Candida albicans induction of macrophage pyroptosis. MBio 2018, 9:a019620. [DOI] [PMC free article] [PubMed] [Google Scholar]; This was the largest screen for fungal mutants that are defective in regulating host inflammasome activation.
- 63.Kasper L, König A, Koenig P-A, Gresnigt MS, Westman J, Drummond RA, Lionakis MS, Groß O, Ruland J, Naglik JR, et al. : The fungal peptide toxin Candidalysin activates the NLRP3 inflammasome and causes cytolysis in mononuclear phagocytes. Nat Commun 2018, 9:4260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Westman J, Moran G, Mogavero S, Hube B, Grinstein S: Candida albicans Hyphal Expansion Causes Phagosomal Membrane Damage and Luminal Alkalinization. MBio 2018, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Koselny K, Mutlu N, Minard AY, Kumar A, Krysan DJ, Wellington M: A Genome-Wide Screen of Deletion Mutants in the Filamentous Saccharomyces cerevisiae Background Identifies Ergosterol as a Direct Trigger of Macrophage Pyroptosis. MBio 2018, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.O’Meara TR, Veri AO, Ketela T, Jiang B, Roemer T, Cowen LE: Global analysis of fungal morphology exposes mechanisms of host cell escape. Nat Commun 2015, 6:6741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Brede C, Friedrich M, Jordán-Garrote A-L, Riedel SS, Bäuerlein CA, Heinze KG, Bopp T, Schulz S, Mottok A, Kiesel C, et al. : Mapping immune processes in intact tissues at cellular resolution. J Clin Invest 2012, 122:4439–4446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. *.Amich J, Mokhtari Z, Strobel M, Vialetto E, Sheta D, Yu Y, Hartweg J, Kalleda N, Jarick KJ, Brede C, et al. : Three-Dimensional Light Sheet Fluorescence Microscopy of Lungs To Dissect Local Host Immune-Aspergillus fumigatus Interactions. MBio 2020, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]; This microscopy approach will allow for new analysis of C. albicans infections at a whole-organ level.
