Skip to main content

MINI REVIEW article

Front. Fungal Biol., 23 June 2022
Sec. Fungi-Animal Interactions
Volume 3 - 2022 | https://doi.org/10.3389/ffunb.2022.940884

Systemic Candidiasis in Mice: New Insights From an Old Model

  • 1Department of Cell Biology, Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
  • 2Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
  • 3Institute of Microbiology, Friedrich Schiller University, Jena, Germany
  • 4Center for Sepsis Control and Care, Jena, Germany

Animal models are essential to understand the pathophysiology of infections, to test novel antifungal compounds, and to determine the potential of adjunctive therapies, e.g. immune modulation. The murine model of systemic candidiasis induced by intravenous infection is technically straightforward, highly reproducible, and well-characterized. However, intravenous inoculation circumvents the necessity for the fungus to translocate across mucosal barriers, and the use of SPF mice that are immunologically naïve to Candida does not reflect the situation in human patients, in whom adaptive immune responses have been induced by mucosal colonization prior to infection. Therefore, mouse models that combine intestinal colonization and systemic infection have been developed, resulting in novel insights into host-fungal interactions and immunity. In this review, we summarize the main findings, current questions, and discuss how these might impact the translatability of results from mice to humans.

Introduction

Systemic candidiasis is a term used to describe a spectrum of infections caused by Candida species disseminating via the blood stream to a range of internal organs, including liver, brain, kidneys, heart and eyes. It includes candidemia, the presence of fungi in the blood, deep-seated candidiasis affecting one or few internal organs (e.g. hepatosplenic candidiasis), or disseminated infection affecting several organs in different parts of the body (McCarty et al., 2021). Systemic candidiasis is a life-threatening condition that despite antifungal treatment still results in relatively high mortality rates of 20-40% (Cornely et al., 2020). Although a significant proportion of infections are caused by non-albicans species such as C. glabrata, C. tropicalis, C. parapsilosis, and C. krusei, C. albicans is the most common cause of systemic candidiasis worldwide (Guinea, 2014). In contrast to C. tropicalis, C. parapsilosis, and C. krusei, which can be isolated from environmental samples, C. albicans is nearly exclusively found in association with warm-blooded animals (Odds, 1988). In humans, it is transmitted vertically from mother to child and colonizes the oral, urogenital, and intestinal mucosa (Ghannoum et al., 2010; Drell et al., 2013; Nash et al., 2017). C. albicans is one of the most abundant members of the intestinal mycobiome, and its prevalence in the Human Microbiome Project healthy cohort was >60% (Nash et al., 2017). Most cases of systemic candidiasis by C. albicans are caused by strains that colonized the mucosal surfaces of the individuals before disease onset, with the gut being one of the main reservoirs for the fungus (Miranda et al., 2009; Zhai et al., 2020).

Animal models are essential to understand the pathophysiology of infections, to test novel antifungal compounds, and to determine the potential of adjunctive therapies, e.g. immune modulation. Candidiasis occurs in a range of domestic and wild animals (Seyedmousavi et al., 2018), and can be easily induced in laboratory animals by injecting yeast cells into the blood stream. Due to practical considerations, such as ease of husbandry and handling, availability of genetically defined and genetically modified strains, and reagents for molecular analyses, mice are predominantly used as model host for systemic candidiasis (Szabo and MacCallum, 2011). The murine model of systemic candidiasis induced by intravenous infection is technically straightforward, highly reproducible, and well-characterized (Szabo and MacCallum, 2011; MacCallum, 2013). However, intravenous inoculation circumvents the necessity for the fungus to translocate across mucosal barriers (Koh et al., 2008). Furthermore, the host is challenged by a single bolus of pathogens, which might not reflect continuous translocation over a prolonged period of time. To address this, a murine model of gastrointestinal colonization with induced translocation has been developed by A. Y. Koh in the lab of G. B. Pier. The model requires antibiotic treatment to achieve colonization levels that are sufficient to sustain translocation rates leading to clinical disease (Koh et al., 2008). In addition, both intestinal barrier dysfunction and neutropenia are necessary to induce translocation, consistent with older reports (Umenai et al., 1979; Mellado et al., 2000). Finally, the number of fungi translocating as effective infectious dose cannot be defined (Koh et al., 2008; Koh, 2013). Thereby, this model is substantially more complex than the intravenous infection model, more difficult to standardize, and it does not allow to investigate candidiasis in immunocompetent hosts. While immunosuppression and gut barrier damage increase the risk of systemic candidiasis in humans, they are not essential. Thus, this model represents only one group of patients at risk of systemic candidiasis. Nonetheless, the colonization-dissemination model was essential for the identification of host defense systems preventing translocation and subsequent candidiasis. Furthermore, it elucidated the route of spread from the gut: While the kidneys are the main target organ in the murine intravenous infection model, induced dissemination from the gut results in fungi predominantly entering the liver via the hepatic portal vein (MacCallum and Odds, 2005; Koh et al., 2008; Koh, 2013).

Although the two-step colonization-dissemination model as developed by A. Y. Koh is not used routinely to study systemic candidiasis, it provided important basic information that, together with the efforts of others, e.g. the Huffnagle lab (Mason et al., 2012), led to the establishment of C. albicans intestinal colonization models. These models were used to investigate how the host immune system responds to Candida colonization, and recently were combined with intravenous infection to investigate if and how colonization affects subsequent systemic candidiasis.

Candida Colonization Affects Systemic Candidiasis

Why Is Fungal Colonization Important?

The profound impact of microbial colonization on the mammalian immune system has been well established since germ free mice were used on a larger scale in the 1960s [reviewed in (Smith et al., 2007)]. In the last decades multiple mechanisms by which bacteria shape the development of the immune system and homeostasis were identified (Hooper et al., 2012). In comparison, very little was known about the role of fungi as part of the microbiota, for which the term mycobiota was coined. One reason is the lower abundance of fungi compared to bacteria in the gut (Huffnagle and Noverr, 2013; Nash et al., 2017), and the necessity to develop specific methods to analyze mycobiota composition (reviewed in (Li et al., 2019). However, specific pattern recognition receptors (especially C-type lectins) evolved to recognize fungi (Ward and Vyas, 2020), suggesting that fungi could have specific effects on host immunity. The first evidence supporting this notion came from observations that the titers of antibodies reactive to S. cerevisiae mannan (ASCA) were elevated in patients with Crohn’s disease or other immune mediated diseases (Schaffer et al., 2007). More recently, C-type lectins were found to be important for controlling fungal-driven inflammation in colitis models (Iliev et al., 2012; Wang et al., 2016). Lack of CX3CR1+ mononuclear phagocytes affects gut mycobiome composition and results in colitis in mice, and CX3CR1 defects are found in some Crohn’s disease patients (Leonardi et al., 2018). In mice, mycobiota changes induced by antifungal treatment were found to exacerbate both colitis and allergic airway disease, which involves gut-resident CX3CR1+ mononuclear phagocytes (Wheeler et al., 2016; Li et al., 2018; Skalski et al., 2018). In the absence of bacteria, however, commensal fungi provide an essential stimulus for immune maturation (Jiang et al., 2017).

Immunological Responses to C. albicans Colonization

Most of the studies mentioned above used or included Candida, often C. albicans, as model fungi for intestinal colonization. C. albicans colonization furthermore induces robust Th17 responses in mice (Leonardi et al., 2018; Shao et al., 2019) and is the main inducer of Th17 cells in humans (Bacher et al., 2019). Importantly, these reactions are not limited to mucosal sites as reactive Th17 cells increase in the spleen (Shao et al., 2019). Th17 immunity has a crucial role in maintaining oral epithelial barriers and preventing oral candidiasis (Gaffen and Moutsopoulos, 2020) and IL-17 is important for defense against systemic candidiasis (Huang et al., 2004; Bär et al., 2014; Ramani et al., 2016). In addition, colonization induces antibody responses in mice and humans (Pitarch et al., 2006; Luo et al., 2016; Huertas et al., 2017; Doron et al., 2021), and antibodies occurring in human serum are able to increase interaction of phagocytes with C. albicans, and to reduce adhesion to and damage of epithelial cells by the fungus (Wich et al., 2021). In summary, mucosal colonization with C. albicans induces both local and systemic responses. Since most cases of systemic candidiasis by C. albicans are caused by endogenous strains that colonized the mucosal surfaces (Zhai et al., 2020), for humans it can be assumed that systemic infection with C. albicans occurs after some level of adaptive immunity has already been established. This is in stark contrast to the intravenous infection model using specific-pathogen free (SPF) mice. Laboratory mice bred and kept under strict hygiene regimens are usually not colonized with Candida albicans (Iliev et al., 2012; Wheeler et al., 2016; Skalski et al., 2018; Doron et al., 2019). Thus, SPF mice can be considered immunologically naïve to Candida.

Impact of Colonization on Systemic Candidiasis in Mice

Since both Th17 responses and the systemic antibodies induced affect host-fungal interactions in vivo and in vitro, respectively, one would expect that colonization-induced adaptive immunity could reduce susceptibility to systemic candidiasis. Indeed, earlier work by E. Balish’s group already provided evidence for a protective effect of intestinal colonization with C. albicans for subsequent systemic infection in mice (Cantorna and Balish, 1991; Wagner et al., 1996) which involved T cells. Huertas et al. (Huertas et al., 2017) confirmed these results and demonstrated a systemic antibody response following colonization, that could provide an explanation for the protective effect. Two recent studies using combined gastrointestinal colonization and tail vein infection not only confirmed these previous observations, but also identified both antibodies and Th17 responses induced by colonization as factors mediating colonization-induced protection against systemic candidiasis.

Shao et al. (Shao et al., 2019) used oral treatment with ampicillin to facilitate intestinal colonization of mice with C. albicans. Colonization alone did not result in clinical disease or detectable fungal dissemination into internal organs, but protected animals against systemic challenge. This could be linked to the induction and activation of fungal-specific Th17 CD4+ T cells that accumulated in both mucosal and systemic lymphatic tissue. These cells mediate protection by releasing IL-17 which in turn activates neutrophils, which are the most important effector cells for fungal clearance (d'Enfert et al., 2021). Importantly, these immunological changes also decreased susceptibility to the bacterial pathogen S. aureus, but increased susceptibility to allergic airway inflammation, highlighting the consequences of the enhanced Th17 response beyond antigen-specific effects. It is also notable that the effect was dependent on persistent fungal colonization. The same authors furthermore provide evidence that the level of C. albicans colonization correlates with the frequency of fungal-specific Th17 CD4+ T cells and neutrophil reactivity in humans, indicating that these mechanisms are conserved across mice and men.

Using antibody profiling Doron et al. showed that C. albicans is the major inducer of antifungal immunoglobulin G (IgG) present in the serum of humans and mice (Doron et al., 2021). Similar to the Th17 responses analyzed by Shao et al., the B cell responses in the study of Doron et al. originated in the spleen rather than mucosa-associated lymphoid tissue. The antibodies induced were protective against systemic candidiasis as demonstrated by transfer experiments into B cell-deficient mice.

In addition to adaptive immunity, Tsao et al. (Tso et al., 2018) reported that colonization seems to also induce trained immunity, which is a prolonged phenotypic adaptation of monocytes and macrophages in response to the fungal cell wall components chitin and β-glucan that mediates enhanced protection against systemic challenge (Cheng et al., 2014; Saeed et al., 2014; Rizzetto et al., 2016). Tsao et al. obtained gut-adapted C. albicans isolates by repeated long-term passaging through the murine gut. These isolates showed filamentation defects and strongly reduced virulence in the systemic infection model, but facilitated colonization-mediated protection against systemic challenge. As this was also observed in Rag1−/− mice lacking both B and T cells and was accompanied by increased innate cytokine responses, innate-like mechanisms contribute to the protective effect (Tso et al., 2018).

Discussion

As summarized above, Candida colonization is not only one of the risk factors for systemic candidiasis (Eggimann and Pittet, 2014) but also has profound effects on the host immune system that affect susceptibility to systemic candidiasis in mice. This raises further questions, related both to our basic understanding of host-Candida-interaction and possible consequences for preclinical animal models.

How are systemic responses induced by colonization? Low-grade dissemination of viable fungal cells is one possibility, and has been observed in C. albicans-colonized mice in the absence of clinical symptoms in some studies (Vautier et al., 2015). Instead of viable cells, components of the fungal cell wall could be transported by migrating CX3CR1+ mononuclear phagocytes to distant lymphoid organs (Doron et al., 2021) as described for some bacteria (Zeng et al., 2016). While this can explain the induction of both T and B cell responses, it remains unclear why C. albicans is the main inducer of Th17 cells in humans (Bacher et al., 2019) although other yeast such as Saccharomyces cerevisiae or Malassezia restricta are found even more frequently and with higher prevalence in the human gut (Nash et al., 2017). It is likely that specific features of C. albicans promote induction of systemic responses, but it is as of yet unknown what these features are. Intestinal IgA targets C. albicans hyphae over yeast (Ost et al., 2021), and hypha formation is an important virulence factor of C. albicans epithelial infection (Jacobsen et al., 2012). Comparison of the response to different C. albicans strains with different virulence potential could provide insights as to whether occasional, locally restricted epithelial invasion events promote the development of robust adaptive responses to colonization, possibly independent of trained innate immunity that is induced also by a filament-deficient strain (Tso et al., 2018).

Both antibody and Th17 responses require activation of CD4+ T cells, highlighting the importance of these cells for the overall adaptive response to C. albicans colonization. But what is the relative contribution of antibodies and IL-17, respectively? Can loss of one mechanism be compensated by the other? These questions cannot be answered yet, but would be clinically interesting regarding risk assessment of patients undergoing specific immunosuppressive therapies. Based on the available data, the beneficial effect of Th17 responses seems to rely on neutrophils (Shao et al., 2019), whereas antibody transfer protected mice treated with cyclophosphamide which induces neutropenia (Doron et al., 2021). This suggests that antibodies protect independent of neutrophils, and that B cell responses complement cellular Th17-driven protection.

How do the new insights on colonization and susceptibility to systemic candidiasis affect the design of infection experiments in mice? So far, immunocompetent SPF mice without any prior treatment infected via lateral tail vein injection have been used most commonly to study the pathogenesis of systemic candidiasis, to identify virulence factors, and evaluate efficacy of therapeutic approaches. In most cases, this simplistic model is likely to be sufficient for the comparison of fungal deletion mutants to parental strains, or to determine if an antifungal compound is effective in vivo. The recent findings are, however, relevant for immunological studies and evaluation of immunomodulatory therapy, because colonized mice and mice that are immunologically naïve to C. albicans are likely to respond differently. Shao et al. discuss that contradictory results regarding the role of IL-17A and IL-17 receptor for systemic candidiasis in mice might be the consequence of differences in the occurrence of C. albicans as commensal in each facility (Shao et al., 2019). This might also explain discordance of early studies on the role of antibodies for systemic candidiasis in mice (Wagner et al., 1996). Prior exposure to the fungus can even affect results of ex vivo models, such as the interaction of C. albicans with host cells in a whole-blood model (Machata et al., 2020). Given that human patients with systemic candidiasis are highly likely to have developed adaptive responses before infection, combining colonization with defined tail vein infection could refine this valuable model to better reflect the human situation and to increase the translatability of results from mice to men.

Author Contributions

Conceptualization and writing of the manuscript were performed by BJ and IDJ. All authors contributed to the article and approved the submitted version.

Funding

IJ and BJ are supported by the German Research Foundation (DFG) through the TRR 124 FungiNet, “Pathogenic fungi and their human host: Networks of Interaction,” DFG project number 210879364, project C5.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

We thank our friends and colleagues in the medical mycology and fungal immunology fields for many thought-provoking discussions.

References

Bacher P., Hohnstein T., Beerbaum E., Röcker M., Blango M. G., Kaufmann S., et al. (2019). Human Anti-Fungal Th17 Immunity and Pathology Rely on Cross-Reactivity Against Candida Albicans. Cell 176 (6), 1340–1355.e1315. doi: 10.1016/j.cell.2019.01.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Bär E., Whitney P. G., Moor K., Reis e Sousa C., LeibundGut-Landmann S. (2014). IL-17 Regulates Systemic Fungal Immunity by Controlling the Functional Competence of NK Cells. Immunity 40 (1), 117–127. doi: 10.1016/j.immuni.2013.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Cantorna M. T., Balish E. (1991). Acquired Immunity to Systemic Candidiasis in Immunodeficient Mice. J. Infect. Dis. 164 (5), 936–943. doi: 10.1093/infdis/164.5.936

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng S. C., Quintin J., Cramer R. A., Shepardson K. M., Saeed S., Kumar V., et al. (2014). mTOR- and HIF-1alpha-Mediated Aerobic Glycolysis as Metabolic Basis for Trained Immunity. Science 345 (6204), 1250684. doi: 10.1126/science.1250684

PubMed Abstract | CrossRef Full Text | Google Scholar

Cornely F. B., Cornely O. A., Salmanton-García J., Koehler F. C., Koehler P., Seifert H., et al. (2020). Attributable Mortality of Candidemia After Introduction of Echinocandins. Mycoses 63 (12), 1373–1381. doi: 10.1111/myc.13177

PubMed Abstract | CrossRef Full Text | Google Scholar

d'Enfert C., Kaune A. K., Alaban L. R., Chakraborty S., Cole N., Delavy M., et al. (2021). The Impact of the Fungus-Host-Microbiota Interplay Upon Candida Albicans Infections: Current Knowledge and New Perspectives. FEMS Microbiol. Rev. 45 (3), 1–55. doi: 10.1093/femsre/fuaa060

CrossRef Full Text | Google Scholar

Doron I., Leonardi I., Iliev I. D. (2019). Profound Mycobiome Differences Between Segregated Mouse Colonies Do Not Influence Th17 Responses to a Newly Introduced Gut Fungal Commensal. Fungal Genet. Biol. 127, 45–49. doi: 10.1016/j.fgb.2019.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Doron I., Leonardi I., Li X. V., Fiers W. D., Semon A., Bialt-DeCelie M., et al. (2021). Human Gut Mycobiota Tune Immunity via CARD9-Dependent Induction of Anti-Fungal IgG Antibodies. Cell 184 (4), 1017–1031.e1014. doi: 10.1016/j.cell.2021.01.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Drell T., Lillsaar T., Tummeleht L., Simm J., Aaspollu A., Vain E., et al. (2013). Characterization of the Vaginal Micro- and Mycobiome in Asymptomatic Reproductive-Age Estonian Women. PloS One 8 (1), e54379. doi: 10.1371/journal.pone.0054379

PubMed Abstract | CrossRef Full Text | Google Scholar

Eggimann P., Pittet D. (2014). Candida Colonization Index and Subsequent Infection in Critically Ill Surgical Patients: 20 Years Later. Intensive Care Med. 40 (10), 1429–1448. doi: 10.1007/s00134-014-3355-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Gaffen S. L., Moutsopoulos N. M. (2020). Regulation of Host-Microbe Interactions at Oral Mucosal Barriers by Type 17 Immunity. Sci. Immunol. 5 (43). doi: 10.1126/sciimmunol.aau4594

CrossRef Full Text | Google Scholar

Ghannoum M. A., Jurevic R. J., Mukherjee P. K., Cui F., Sikaroodi M., Naqvi A., et al. (2010). Characterization of the Oral Fungal Microbiome (Mycobiome) in Healthy Individuals. PloS Pathog. 6 (1), e1000713. doi: 10.1371/journal.ppat.1000713

PubMed Abstract | CrossRef Full Text | Google Scholar

Guinea J. (2014). Global Trends in the Distribution of Candida Species Causing Candidemia. Clin. Microbiol. Infect. 20 Suppl 6, 5–10. doi: 10.1111/1469-0691.12539

PubMed Abstract | CrossRef Full Text | Google Scholar

Hooper L. V., Littman D. R., Macpherson A. J. (2012). Interactions Between the Microbiota and the Immune System. Science 336 (6086), 1268–1273. doi: 10.1126/science.1223490

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang W., Na L., Fidel P. L., Schwarzenberger P. (2004). Requirement of Interleukin-17A for Systemic Anti-Candida Albicans Host Defense in Mice. J. Infect. Dis. 190 (3), 624–631. doi: 10.1086/422329

PubMed Abstract | CrossRef Full Text | Google Scholar

Huertas B., Prieto D., Pitarch A., Gil C., Pla J., Diez-Orejas R. (2017). Serum Antibody Profile During Colonization of the Mouse Gut by Candida Albicans: Relevance for Protection During Systemic Infection. J. Proteome Res. 16 (1), 335–345. doi: 10.1021/acs.jproteome.6b00383

PubMed Abstract | CrossRef Full Text | Google Scholar

Huffnagle G. B., Noverr M. C. (2013). The Emerging World of the Fungal Microbiome. Trends Microbiol. 21 (7), 334–341. doi: 10.1016/j.tim.2013.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Iliev I. D., Funari V. A., Taylor K. D., Nguyen Q., Reyes C. N., Strom S. P., et al. (2012). Interactions Between Commensal Fungi and the C-Type Lectin Receptor Dectin-1 Influence Colitis. Science 336 (6086), 1314–1317. doi: 10.1126/science.1221789

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacobsen I. D., Wilson D., Wachtler B., Brunke S., Naglik J. R., Hube B. (2012). Candida Albicans Dimorphism as a Therapeutic Target. Expert Rev. Anti Infect. Ther. 10 (1), 85–93. doi: 10.1586/eri.11.152

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang T. T., Shao T. Y., Ang W. X. G., Kinder J. M., Turner L. H., Pham G., et al. (2017). Commensal Fungi Recapitulate the Protective Benefits of Intestinal Bacteria. Cell Host Microbe 22 (6), 809–816.e804. doi: 10.1016/j.chom.2017.10.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Koh A. Y. (2013). Murine Models of Candida Gastrointestinal Colonization and Dissemination. Eukaryot. Cell 12 (11), 1416–1422. doi: 10.1128/ec.00196-13

PubMed Abstract | CrossRef Full Text | Google Scholar

Koh A. Y., Kohler J. R., Coggshall K. T., Van Rooijen N., Pier G. B. (2008). Mucosal Damage and Neutropenia are Required for Candida Albicans Dissemination. PloS Pathog. 4 (2), e35. doi: 10.1371/journal.ppat.0040035

PubMed Abstract | CrossRef Full Text | Google Scholar

Leonardi I., Li X., Semon A., Li D., Doron I., Putzel G., et al. (2018). CX3CR1(+) Mononuclear Phagocytes Control Immunity to Intestinal Fungi. Science 359 (6372), 232–236. doi: 10.1126/science.aao1503

PubMed Abstract | CrossRef Full Text | Google Scholar

Li X. V., Leonardi I., Iliev I. D. (2019). Gut Mycobiota in Immunity and Inflammatory Disease. Immunity 50 (6), 1365–1379. doi: 10.1016/j.immuni.2019.05.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Li X., Leonardi I., Semon A., Doron I., Gao I. H., Putzel G. G., et al. (2018). Response to Fungal Dysbiosis by Gut-Resident CX3CR1(+) Mononuclear Phagocytes Aggravates Allergic Airway Disease. Cell Host Microbe 24 (6), 847–856.e844. doi: 10.1016/j.chom.2018.11.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo T., Krüger T., Knüpfer U., Kasper L., Wielsch N., Hube B., et al. (2016). Immunoproteomic Analysis of Antibody Responses to Extracellular Proteins of Candida Albicans Revealing the Importance of Glycosylation for Antigen Recognition. J. Proteome Res. 15 (8), 2394–2406. doi: 10.1021/acs.jproteome.5b01065

PubMed Abstract | CrossRef Full Text | Google Scholar

MacCallum D. M. (2013). Mouse Model of Invasive Fungal Infection. Methods Mol. Biol. 1031, 145–153. doi: 10.1007/978-1-62703-481-4_17

PubMed Abstract | CrossRef Full Text | Google Scholar

MacCallum D. M., Odds F. C. (2005). Temporal Events in the Intravenous Challenge Model for Experimental Candida Albicans Infections in Female Mice. Mycoses 48 (3), 151–161. doi: 10.1111/j.1439-0507.2005.01121.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Machata S., Sreekantapuram S., Hünniger K., Kurzai O., Dunker C., Schubert K., et al. (2020). Significant Differences in Host-Pathogen Interactions Between Murine and Human Whole Blood. Front. Immunol. 11. doi: 10.3389/fimmu.2020.565869

PubMed Abstract | CrossRef Full Text | Google Scholar

Mason K. L., Erb Downward J. R., Mason K. D., Falkowski N. R., Eaton K. A., Kao J. Y., et al. (2012). Candida Albicans and Bacterial Microbiota Interactions in the Cecum During Recolonization Following Broad-Spectrum Antibiotic Therapy. Infect. Immun. 80 (10), 3371–3380. doi: 10.1128/iai.00449-12

PubMed Abstract | CrossRef Full Text | Google Scholar

McCarty T. P., White C. M., Pappas P. G. (2021). Candidemia and Invasive Candidiasis. Infect. Dis. Clin. North Am. 35 (2), 389–413. doi: 10.1016/j.idc.2021.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Mellado E., Cuenca-Estrella M., Regadera J., González M., Díaz-Guerra T. M., Rodríguez-Tudela J. L. (2000). Sustained Gastrointestinal Colonization and Systemic Dissemination by Candida Albicans, Candida Tropicalis and Candida Parapsilosis in Adult Mice. Diagn. Microbiol. Infect. Dis. 38 (1), 21–28. doi: 10.1016/s0732-8893(00)00165-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Miranda L. N., van der Heijden I. M., Costa S. F., Sousa A. P., Sienra R. A., Gobara S., et al. (2009). Candida Colonisation as a Source for Candidaemia. J. Hosp. Infect. 72 (1), 9–16. doi: 10.1016/j.jhin.2009.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Nash A. K., Auchtung T. A., Wong M. C., Smith D. P., Gesell J. R., Ross M. C., et al. (2017). The Gut Mycobiome of the Human Microbiome Project Healthy Cohort. Microbiome 5 (1), 153. doi: 10.1186/s40168-017-0373-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Odds F. C. (1988). Candida and Candidosis (London: Baillière Tindall).

Google Scholar

Ost K. S., O'Meara T. R., Stephens W. Z., Chiaro T., Zhou H., Penman J., et al. (2021). Adaptive Immunity Induces Mutualism Between Commensal Eukaryotes. Nature 596 (7870), 114–118. doi: 10.1038/s41586-021-03722-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Pitarch A., Jimenez A., Nombela C., Gil C. (2006). Decoding Serological Response to Candida Cell Wall Immunome Into Novel Diagnostic, Prognostic, and Therapeutic Candidates for Systemic Candidiasis by Proteomic and Bioinformatic Analyses. Mol. Cell Proteomics 5 (1), 79–96. doi: 10.1074/mcp.M500243-MCP200

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramani K., Garg A. V., Jawale C. V., Conti H. R., Whibley N., Jackson E. K., et al. (2016). The Kallikrein-Kinin System: A Novel Mediator of IL-17-Driven Anti-Candida Immunity in the Kidney. PloS Pathog. 12 (11), e1005952. doi: 10.1371/journal.ppat.1005952

PubMed Abstract | CrossRef Full Text | Google Scholar

Rizzetto L., Ifrim D. C., Moretti S., Tocci N., Cheng S. C., Quintin J., et al. (2016). Fungal Chitin Induces Trained Immunity in Human Monocytes During Cross-Talk of the Host With Saccharomyces Cerevisiae. J. Biol. Chem. 291 (15), 7961–7972. doi: 10.1074/jbc.M115.699645

PubMed Abstract | CrossRef Full Text | Google Scholar

Saeed S., Quintin J., Kerstens H. H., Rao N. A., Aghajanirefah A., Matarese F., et al. (2014). Epigenetic Programming of Monocyte-to-Macrophage Differentiation and Trained Innate Immunity. Science 345 (6204), 1251086. doi: 10.1126/science.1251086

PubMed Abstract | CrossRef Full Text | Google Scholar

Schaffer T., Müller S., Flogerzi B., Seibold-Schmid B., Schoepfer A. M., Seibold F. (2007). Anti-Saccharomyces Cerevisiae Mannan Antibodies (ASCA) of Crohn's Patients Crossreact With Mannan From Other Yeast Strains, and Murine ASCA IgM Can be Experimentally Induced With Candida Albicans. Inflammation Bowel. Dis. 13 (11), 1339–1346. doi: 10.1002/ibd.20228

CrossRef Full Text | Google Scholar

Seyedmousavi S., Bosco S. M. G., de Hoog S., Ebel F., Elad D., Gomes R. R., et al. (2018). Fungal Infections in Animals: A Patchwork of Different Situations. Med. Mycol. 56 (suppl_1), 165–187. doi: 10.1093/mmy/myx104

PubMed Abstract | CrossRef Full Text | Google Scholar

Shao T. Y., Ang W. X. G., Jiang T. T., Huang F. S., Andersen H., Kinder J. M., et al. (2019). Commensal Candida Albicans Positively Calibrates Systemic Th17 Immunological Responses. Cell Host Microbe 25 (3), 404–417.e406. doi: 10.1016/j.chom.2019.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Skalski J. H., Limon J. J., Sharma P., Gargus M. D., Nguyen C., Tang J., et al. (2018). Expansion of Commensal Fungus Wallemia Mellicola in the Gastrointestinal Mycobiota Enhances the Severity of Allergic Airway Disease in Mice. PloS Pathog. 14 (9), e1007260. doi: 10.1371/journal.ppat.1007260

PubMed Abstract | CrossRef Full Text | Google Scholar

Smith K., McCoy K. D., Macpherson A. J. (2007). Use of Axenic Animals in Studying the Adaptation of Mammals to Their Commensal Intestinal Microbiota. Semin. Immunol. 19 (2), 59–69. doi: 10.1016/j.smim.2006.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Szabo E. K., MacCallum D. M. (2011). The Contribution of Mouse Models to Our Understanding of Systemic Candidiasis. FEMS Microbiol. Lett. 320 (1), 1–8. doi: 10.1111/j.1574-6968.2011.02262.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Tso G. H. W., Reales-Calderon J. A., Tan A. S. M., Sem X., Le G. T. T., Tan T. G., et al. (2018). Experimental Evolution of a Fungal Pathogen Into a Gut Symbiont. Science 362 (6414), 589–595. doi: 10.1126/science.aat0537

PubMed Abstract | CrossRef Full Text | Google Scholar

Umenai T., Konno S., Ishida N. (1979). Systemic Candidiasis From Candida Albicans Colonizing the Gastrointestinal Tract of Mice. Experientia 35 (10), 1331–1332. doi: 10.1007/bf01963990

PubMed Abstract | CrossRef Full Text | Google Scholar

Vautier S., Drummond R. A., Chen K., Murray G. I., Kadosh D., Brown A. J., et al. (2015). Candida Albicans Colonization and Dissemination From the Murine Gastrointestinal Tract: The Influence of Morphology and Th17 Immunity. Cell Microbiol. 17 (4), 445–450. doi: 10.1111/cmi.12388

PubMed Abstract | CrossRef Full Text | Google Scholar

Wagner R. D., VazquezTorres A., JonesCarson J., Warner T., Balish E. (1996). B Cell Knockout Mice are Resistant to Mucosal and Systemic Candidiasis of Endogenous Origin But Susceptible to Experimental Systemic Candidiasis. J. Infect. Dis. 174 (3), 589–597. doi: 10.1093/infdis/174.3.589

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang T., Pan D., Zhou Z., You Y., Jiang C., Zhao X., et al. (2016). Dectin-3 Deficiency Promotes Colitis Development Due to Impaired Antifungal Innate Immune Responses in the Gut. PloS Pathog. 12 (6), e1005662. doi: 10.1371/journal.ppat.1005662

PubMed Abstract | CrossRef Full Text | Google Scholar

Ward R. A., Vyas J. M. (2020). The First Line of Defense: Effector Pathways of Anti-Fungal Innate Immunity. Curr. Opin. Microbiol. 58, 160–165. doi: 10.1016/j.mib.2020.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Wheeler M. L., Limon J. J., Bar A. S., Leal C. A., Gargus M., Tang J., et al. (2016). Immunological Consequences of Intestinal Fungal Dysbiosis. Cell Host Microbe 19 (6), 865–873. doi: 10.1016/j.chom.2016.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Wich M., Greim S., Ferreira-Gomes M., Krüger T., Kniemeyer O., Brakhage A. A., et al. (2021). Functionality of the Human Antibody Response to Candida Albicans. Virulence 12 (1), 3137–3148. doi: 10.1080/21505594.2021.2015116

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng M. Y., Cisalpino D., Varadarajan S., Hellman J., Warren H. S., Cascalho M., et al. (2016). Gut Microbiota-Induced Immunoglobulin G Controls Systemic Infection by Symbiotic Bacteria and Pathogens. Immunity 44 (3), 647–658. doi: 10.1016/j.immuni.2016.02.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhai B., Ola M., Rolling T., Tosini N. L., Joshowitz S., Littmann E. R., et al. (2020). High-Resolution Mycobiota Analysis Reveals Dynamic Intestinal Translocation Preceding Invasive Candidiasis. Nat. Med. 26 (1), 59–64. doi: 10.1038/s41591-019-0709-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: murine infection models, Candida albicans, filamentation, colonization, adaptive immunity

Citation: Jungnickel B and Jacobsen ID (2022) Systemic Candidiasis in Mice: New Insights From an Old Model. Front. Fungal Biol. 3:940884. doi: 10.3389/ffunb.2022.940884

Received: 10 May 2022; Accepted: 24 May 2022;
Published: 23 June 2022.

Edited by:

Chengshu Wang, Chinese Academy of Sciences (CAS), China

Reviewed by:

Yahui Guo, Memorial Sloan Kettering Cancer Center, United States

Copyright © 2022 Jungnickel and Jacobsen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ilse D. Jacobsen, ilse.jacobsen@leibniz-hki.de

Download