Next Article in Journal
Genetic Diversity of Salmonella Derby from the Poultry Sector in Europe
Next Article in Special Issue
The Contribution of Host Cells to Pneumocystis Immunity: An Update
Previous Article in Journal
Prevalence of Helicobacter pylori and Its Associated Factors among Healthy Asymptomatic Residents in the United Arab Emirates
Previous Article in Special Issue
Candida albicans at Host Barrier Sites: Pattern Recognition Receptors and Beyond
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Hidden Cost of Modern Medical Interventions: How Medical Advances Have Shaped the Prevalence of Human Fungal Disease

Institute of Immunology & Immunotherapy, Institute of Microbiology & Infection, University of Birmingham, Birmingham B15 2TT, UK
*
Author to whom correspondence should be addressed.
Pathogens 2019, 8(2), 45; https://doi.org/10.3390/pathogens8020045
Submission received: 8 February 2019 / Revised: 28 March 2019 / Accepted: 29 March 2019 / Published: 4 April 2019
(This article belongs to the Special Issue Immunology of Fungal Infections)

Abstract

:
Life expectancy in the West is the highest it has ever been, due to the introduction of better hygiene practices and sophisticated medical interventions for cancer, autoimmunity and infectious disease. With these modern advances, a rise in the prevalence of opportunistic infections has also been observed. These include several fungal infections, which present a particular clinical challenge due to the lack of fungal vaccines, limited diagnostics and increasing antifungal drug resistance. This mini-review outlines how modern-day clinical practices have shaped the recent increase in fungal diseases observed in the last few decades. We discuss new research that has implicated the use of immune-modulating drugs in the enhanced susceptibility of vulnerable patients to life-threatening fungal infections.

1. Introduction

Fuelled by a wealth of new technological and pharmaceutical advances, and an improved focus on the importance of hygiene and vaccination, human life expectancy has increased by over 50% over the last century [1]. Moreover, the advancement of recombinant DNA technology and the development of new biologics to generate safer vaccines and specific monoclonal antibodies has also become increasingly possible, leading to the development of more targeted therapies, such as small-molecule inhibitors (e.g. receptor tyrosine kinase inhibitors) and monoclonal antibody anti-cancer therapeutics. Alongside the introduction of these advances, the prevalence of life-threatening fungal infections has increased, which has largely coincided with significant progress in the treatment of other comorbidities. For example, the development of immunomodulatory agents for the control of cancer and treatment of autoimmunity has led to a significant increase in the number of immunosuppressed patients who are susceptible to opportunistic infections. The majority of fungal infections are opportunistic, since healthy individuals are able to mount successful immune responses against these microbes and rarely develop clinical infection. Deficiencies in the immune system (e.g., neutropenia), caused by chemotherapy conditioning regimens or immunomodulatory agents, render patients vulnerable to fungi, which they easily become exposed to, due to the large concentration of fungal spores in the environment and within the human commensal flora.
Invasive fungal infections kill over 1.5 million individuals annually [2]. Fungal infections may also be self-limiting mucosal/skin infections, such as oropharyngeal candidiasis, which, whilst non-lethal, can significantly reduce quality of life and impact the lives of >1 billion people [3]. Fungi are recognised by myeloid cells, such as neutrophils, macrophages and dendritic cells (DCs), via host pattern recognition receptors (PRRs), including the C-type lectin receptors (CLRs) and toll-like receptors (TLRs), which bind to pathogen-associated molecular patterns (PAMPs) embedded in the fungal cell wall [4]. The most critical antifungal immune molecule identified to date is CARD9, a signalling adaptor protein found downstream of several CLRs, including Dectin-1, which recognises the fungal cell wall carbohydrate β-glucan. Binding of CARD9-coupled CLRs initiates a signalling cascade via Syk kinase and the CBM (CARD9-BCL10-MALT1) signalosome to activate NF-κB, mitogen-activated protein kinase (MAPK) signalling and a calcium flux, resulting in pro-inflammatory cytokine release, phagocytosis and the killing of fungal cells (Figure 1). Dectin-1-CARD9 signalling is also thought to influence antigen presentation by DCs to shape adaptive immune responses, of which Th1 and Th17 responses are considered the most protective. Human mutations that result in disruption of these pathways cause enhanced susceptibility to fungal infections, underscoring their importance in antifungal immunity. For example, human CARD9 deficiency is associated with the spontaneous development of life-threatening Candida infections [5], while genetic mutations in Dectin-1 predispose individuals to chronic skin and nail fungal infections [6]. In this mini-review, we discuss how commonly prescribed drugs, such as steroids, antibiotics and biologic drugs, as well as emerging small-molecule inhibitors for anti-cancer therapies, affect these critical antifungal pathways that protect us against infection, and how their use has shaped the prevalence of human fungal diseases in modern times.

2. Corticosteroids

A major risk factor for invasive fungal infections are solid organ and stem cell transplants due to the immune-suppressing drug regimens used to prevent transplant rejection, which include the use of corticosteroids such as dexamethasone [7]. The most common fungal infections observed in transplant recipients are invasive aspergillosis and invasive candidiasis [7]. Invasive aspergillosis is most often caused by Aspergillus fumigatus, which invades the lung and central nervous system (CNS) following inhalation of spores, while invasive candidiasis is predominantly caused by Candida albicans, and mostly involves the kidney, liver and spleen, following dissemination of yeast via the bloodstream [8].
Corticosteroids activate the glucocorticoid receptor, which is directly antagonistic to NF-κB, a central transcription factor for pro-inflammatory cytokine production that is activated by Syk-CARD9 signalling during fungal infection [9]. Corticosteroid-induced disruption of NF-κB signalling can therefore significantly disrupt immune responses to fungal pathogens (Figure 1). Indeed, human mutations in IKK, the NF-κB activator, result in severe susceptibility to chronic mucocutaneous candidiasis, which is associated with an impairment in the production of cytokines that protect the mucosal barrier against fungal infection, including IL-17A, IL-22 and IFNγ [10]. Until recently, it was regarded that abrogation of NF-κB signalling was the predominant mode of corticosteroid-induced immune suppression. However, recent studies suggest that corticosteroids may have wider effects on different types of immune cells. For example, corticosteroids induce a state of hyporesponsiveness in DCs, impairing their ability to drive and shape T-cell immunity by limiting their capacity to secrete polarising cytokines IL-12 and IL-23, and by increasing their IL-10 secretion [11,12]. In other studies, steroids have been shown to cause CD4+ T-cell apoptosis in the gastrointestinal (GI) tract, which contributed towards the resulting immune dysregulation within the tissue [13]. CD4+ T-cells are particularly critical for protection against mucosal candidiasis and infections with the fungus Cryptococcus neoformans, as both of these fungal infections are prevalent in patients with dysregulated T-cell responses (e.g., chronic HIV infection). Indeed, these infections are also common in patients treated with corticosteroids [14]. The exact consequences of steroid-induced T-cell dysregulation for antifungal immunity have yet to be fully explored, but understanding how these drugs affect T-cell responses to pathogenic fungi will be important for the future development of therapies in patients receiving corticosteroid therapy, who are particularly at risk for dangerous fungal infections.
Corticosteroid treatment also has a profound effect on myeloid cells, including monocytes and polymorphonuclear neutrophils (PMN). Specifically, corticosteroids impair the generation of reactive oxygen species (ROS) in PMN in response to opsonised Escherichia coli, via decreased levels of NADPH activation. Other pathways were also thought to be affected in this study, but the exact mechanisms have yet to be fully defined, so this will be a key area for future investigation [15]. The generation of ROS forms a key part of several fungi-killing pathways, since patients with defective ROS generation (chronic granulomatous disease, CGD) have a decreased ability of host phagocytes to kill Aspergillus and Candida species, leading to an increased risk of infections with these fungi in CGD patients [10]. Corticosteroids have also been shown to block macrophage-activating factor (MAF) release and macrophage chemotaxis, and can inhibit neutrophil apoptosis pathways, leading to defects in their maturation and function [16]. Despite the key role for neutrophils in the control and elimination of Candida spp. and Aspergillus spp., there have been few studies performed to determine the specific effects of corticosteroid treatment on the antifungal functions of these cells. Yet, this will help our understanding of how to reduce the risk of invasive fungal infections currently associated with corticosteroid use.

3. Antibiotics

Broad-spectrum antibiotics are prescribed to multiple groups of patients at risk of bacterial infection, such as those in the intensive care unit (ICU), transplant patients and patients with acquired immunodeficiency (e.g., HIV). The increased usage of and dependence on antibiotics within our modern healthcare systems has led to a massive increase in bacterial resistance to these drugs. Antibiotic resistance is thus a major problem that threatens our ability to control even minor infections. It is therefore imperative that antibiotic stewardship is improved, and antibiotics are deployed in hospitals more effectively [17]. A further emerging issue with the use of antibiotics is an increased susceptibility to certain fungal infections. Antibiotics have been cited as an independent risk factor for the development of both mucosal and invasive Candida infections [18]. Mucosal candidiasis can occur as vulvovaginal candidiasis (VVC) or oropharyngeal candidiasis, and is associated specifically with the use of β-lactam antibiotics, whereas invasive candidiasis is mainly associated with the use of β-lactams, vancomycin and aminoglycosides [18,19]. VVC is thought to be linked to antibiotic use since these drugs deplete Lactobacillus species within the vagina. Lactobacilli are protective against Candida infection, as their secreted exopolysaccharides interfere with the growth, morphogenesis and adhesion of C. albicans to the vaginal epithelium—three vital components of disease [20]. Moreover, lactobacilli produce an aryl hydrocarbon receptor (AhR) ligand that induces AhR-dependent IL-22 transcription, a cytokine involved in barrier function and regulation of mucosal immune responses [21]. IL-22 is produced by innate lymphoid cells (ILCs) and is protective against VVC by regulating inappropriate neutrophil recruitment to the vaginal mucosa. This occurs via NLRC4-mediated production of the IL-1 receptor antagonist (IL-1Ra), which inhibits activation of the NLRP3 inflammasome and production of IL-1β, IL-1α and IL-18 (Figure 2) [22]. Thus, targeting excessive neutrophil responses in VVC can be therapeutic, since PMN depletion with an anti-Ly6G antibody or treatment with Anakinra (recombinant IL-1Ra) was found to be protective against VVC episodes, stressing the importance of IL-22 and neutrophils in the prevention of inflammation, dysbiosis and disease [22,23]. Indeed, human polymorphisms in IL22 or IDOL1 (the enzyme that catalyses IL-1Ra generation) are associated with VVC susceptibility, reinforcing the importance of this pathway in the protection against VVC [24].
For invasive candidiasis, antibiotics are thought to predispose individuals to this infection by significantly disrupting the microbiota, in particular by causing expansion of Proteobacteria and reductions in Bifidobacterium spp., Clostridial Firmicutes and Bacteroidetes [25], the latter of which are important for maintaining C. albicans colonisation resistance in mice by stimulating Hypoxia inducible factor 1α (HIF1α) activation and production of CRAMP, an antimicrobial peptide [26]. These antibiotic-induced perturbations in the microbiota create a niche availability for commensal C. albicans populations, and the resulting imbalance enhances the likelihood for invasive infection in immunocompromised individuals following disruption to the intestinal barrier caused by surgery, chemotherapy-induced mucositis and/or neutropenia [27]. Indeed, early studies showed that Candida isolated from the bloodstream of candidemic patients were the same as the isolates present in the patient’s GI tract, supporting the idea of an intestinal dysbiosis leading to a fungal bloom and subsequent dissemination via the bloodstream [28]. Due to the use of fluconazole prophylaxis in these at-risk patients, a shift in the causative agents of invasive candidiasis has occurred towards non-albicans species. For example, treatment with metronidazole, carbapenems, clindamycin and colistins have all been found to be strongly associated with disseminated Candida glabrata infections [19]. Unlike C. albicans, other non-albicans species have higher rates of fluconazole resistance [29]. Antibiotics may therefore favour colonisation by drug-resistant fungal isolates, or have mild antifungal effects, thus providing selection pressures for antifungal resistance to evolve [19].
In recent years, it has also been appreciated that antibiotics have additional consequences for the functioning of the mammalian immune system. For example, ciprofloxacin has been shown to suppress TLR expression, one of the key PRRs for anti-Candida defence [30], and its use in patients is significantly associated with invasive Candida infections [31]. Moreover, antibiotics alter the availability of adenosine monophosphate (AMP) within the intestine, which prevents AMPK activation of phagocytosis, and impairs the respiratory activity of macrophages, leading to an impairment of microbe killing [32,33]. A dysregulated metabolism can have significant consequences for antimicrobial immunity, since metabolic rewiring of immune cells is intimately linked with immune function. Upon LPS stimulation and fungal infection, macrophages switch to glycolysis for energy production, and the tricarboxylic acid (TCA) cycle subsequently stalls at two points, leading to the accumulation of the metabolites citrate and succinate. Citrate is used for membrane biogenesis, aiding lysosome formation and antigen presentation, while succinate accumulation leads to the sustained production of IL-1β and nitric oxide, two key components of antifungal immune defence [34]. Antibiotic-induced disturbance of immune-cell metabolism has now been described in several studies. For example, Kalghatgi and colleagues showed that quinolones, aminoglycosides and β-lactams disturbed the TCA cycle and electron transport chain within mitochondria, which led to the overproduction of ROS within immune cells, especially macrophages, leading to oxidative damage and hyper-responsiveness [35]. Indeed, intestinal macrophages within the gut are hyporesponsive and do not readily respond to PRR stimulation due to a high Treg presence, thus preventing unwarranted inflammation and intestinal injury. However, following antibiotic treatment, intestinal macrophages shift into a state of hyper-responsiveness associated with increased inflammatory cytokine production and a Th1 bias, which impacted the ability of antibiotic-treated mice to control infections requiring Th2 and Th17-mediated responses [36]. These antibiotic-induced defects could be reversed by dietary addition of short-chain fatty acids (SCFAs), a microbial metabolite that becomes depleted during antibiotic treatment. SCFAs modulate a variety of immune functions, including PMN recruitment, DC maturation and Treg differentiation [37,38]. In addition to depleting SCFA-producing bacteria, antibiotics have also been shown to impact other groups of bacteria important for mediating the Th17–Treg balance in the intestine. Early work by Ivanov et al. [39] showed that segmented filamentous bacteria (SFB) are responsible for the modulation of inflammation within the gut, and are required for the development of Th17 cells. SFB elimination by antibiotics results in enhanced Treg numbers, leading to an impaired Th17 response and enhanced susceptibility to Citrobacter infection [39,40]. Th17 responses are also important for antifungal defence at mucosal surfaces, since patients with IL17RA mutations are more susceptible to mucosal candidiasis [41]. Th17 cells promote neutrophil recruitment through the upregulation of CXCL1 and CXCL5, and also stimulate antimicrobial peptide release to help combat fungal invasion [42].
Lastly, it has become increasingly appreciated that the effects of antibiotics on the immune system are not limited to the GI tract and extend to peripheral systems. For example, antibiotic-induced alteration of the maternal microbiota in mice has been shown to impair neutrophil progenitor development within neonates, which compromises their ability to fight bacterial sepsis [43]. Antibiotics have also been shown to affect immunity within the CNS, since antibiotics affected microglia maturation and function by the depletion of SCFAs within the gut [44]. This indicates that antibiotics can cause localised defects in mucosal immunity, as well as systemic effects; as such, there are profound consequences for protection against pathogenic microbes, including fungi.

4. Monoclonal Antibodies

Monoclonal antibodies are a class of biological drugs that target specific antigens, such as cytokines and their receptors, in order to block the action of these molecules and modulate inflammation. Monoclonal antibodies targeting IL-17A (such as ixekizumab and secukinumab) or IL-17RA (such as brodalumab) have been used in the treatment of autoimmune diseases, such as rheumatoid arthritis and psoriasis. However, as mentioned above, the IL-17 pathway is critical for antifungal host defence at mucosal barriers. In line with this, an increased frequency of mucosal candidiasis is observed in patients treated with these drugs [45,46]. Similarly, blocking antibodies specific for TNFα (e.g., infiliximab), which has proven to be an effective therapeutic strategy for patients with inflammatory bowel disease, are also associated with an increased susceptibility to fungal infections. In particular, GI candidiasis seems to be particularly prevalent [47], while infections caused by Aspergillus and Histoplasma species have also been observed [48].

5. Anti-Cancer Therapies

The face of cancer treatment has changed significantly over the last few decades, with a greater number of more targeted approaches becoming available. Despite these advances, individuals undergoing cancer treatment are still predisposed to a wide range of infections, including several caused by pathogenic fungi, which impacts the effectiveness of our modern-day cancer treatment regimens and the clinical outcome for these patients. Mechanistic insights into how these modern anti-cancer approaches affect antifungal immunity is still limited, and is an exciting new area of emerging research. Here, we outline some of the recent advances reported in this field.
Ibrutinib: Ibrutinib is a recent addition to the arsenal of chronic lymphocytic leukaemia (CLL) treatment. Ibrutinib is an irreversible inhibitor of Bruton’s tyrosine kinase (Btk), which plays a key role in B-cell receptor signalling and induction of the NF-κB signalling pathway. Ibrutinib is an attractive treatment for cancer, as it has fewer infectious complications than other conventional chemotherapeutic agents, as it is not myelosuppressive and does not result in neutropenia [49,50]. However, several cases of fungal infections associated with the use of ibrutinib have now been reported in the literature. Specifically, ibrutinib appears to predispose individuals to invasive aspergillosis and Pneumocystis pneumonia within the first few months of treatment, while disseminated cryptococcosis has been reported as a late-onset infection [49,50]. These observations have led to the hypothesis that Btk may be a critical regulator of antifungal immunity. Recent studies support this, as ibrutinib treatment impairs the translocation of NFAT and NF-κB in human and murine macrophages stimulated with Aspergillus spores, thus disrupting Btk-mediated activation of NFAT signalling via a pathway that involved endosomal TLR9 [51]. Future work will need to determine the specific roles of Btk in antifungal immunity in response to different fungi and in different immune cell subsets, which may reveal new important pathways of protection and innate immune regulation that can be potentially utilised in the development of new antifungal treatments while preserving the benefits of ibrutinib.
CXCR1/2 Inhibitors: CXCR1 and CXCR2 are chemokine receptors that mediate neutrophil recruitment to the site of infection and activation of their antimicrobial activities. These receptors have also been linked to the survival and angiogenesis of melanoma cancers, hence they have become attractive targets for human melanoma therapy [52]. Indeed, CXCR1/2 inhibitors prevent metastasis of colonic cancer cells to the liver in a murine model, and have also been demonstrated to decrease the invasive potential of melanoma cells, indicating their effectiveness as therapeutics for metastatic cancers [52]. However, these chemokine receptors are also critical for the recruitment of neutrophils to fungal-infected organs. CXCR1 is highly induced in the Candida-infected kidney, and Cxcr1−/− mice are unable to control C. albicans infection due to impaired neutrophil killing [53]. In humans, the mutant CXCR1-T276 allele is associated with an increased likelihood of Candida infection, caused by defective neutrophil degranulation and killing [53]. In patients and mice deficient in CARD9, defective production of CXCR1/2 ligands in the brain leads to decreased neutrophil trafficking and an unchallenged invasive fungal infection in the CNS [54], further underscoring the importance of CXCR1/2 signalling in the control of systemic C. albicans infections. These studies therefore indicate that CXCR1/2 inhibition could be a potential risk factor for the development of invasive candidiasis, and warrants the careful observation of patients on CXCR1/2 inhibitors for any signs of disseminated fungal infection [53], especially since CXCR1/2 are thought to play other non-conventional roles in immunity, including cell survival, proliferation and differentiation [53].
Syk Inhibitors: In addition to its role in CLR signalling and antifungal immunity, Syk kinase is also involved in tumour biology, both as a tumour activator (in B-cell malignancies) and suppressor (in acute myeloid leukaemia and Epstein–Barr virus-derived tumours) through its ability to modulate apoptosis, cell migration and tumorigenesis, depending on cell type [55]. Syk inhibitors have therefore been trialled as a potential new therapy for chronic lymphoid leukaemia and other haematological malignancies [56]. Syk inhibitors are likely to have a detrimental effect on antifungal immunity due to the central role of this kinase in fungal detection pathways (Figure 1). Syk deletion in murine DCs causes an enhanced susceptibility to C. albicans infection, through impaired IL-23 release. The absence of IL-23 causes the abrogation of GM-CSF secretion by natural killer (NK) cells, resulting in reduced neutrophil-mediated killing [57]. Syk activation is also essential for inflammasome activation and IL-1β secretion, which plays an important protective role against several pathogenic fungi, including Aspergillus and Candida species [58,59]. Deletion of the cytokine receptor IL-1R results in an increased fungal burden within a murine pulmonary histoplasmosis model, due to an increased Th2 response and downregulation of IFNγ production [60]. As with CXCR1/2 inhibitors, caution will need to be exercised when Syk inhibitor trials begin, in order to determine their potential effects on human antifungal immunity.

6. Conclusions

As outlined in this review, there are several controllable risk factors for fungal infections, and it will be important to determine the mechanisms underlying how these iatrogenic interventions cause an enhanced susceptibility to fungal infections. In the meantime, treatment with new immune-modulating drugs, including those discussed here, should be combined with a high degree of clinical suspicion regarding potential fungal infections to ensure early diagnosis and initiation of antifungal therapy. Fungal infections currently remain a significant clinical challenge, impacted by the difficulties in generating new antifungal compounds, a lack of fungal vaccines and the rising threat of antifungal drug resistance. Early diagnosis is key for the successful treatment of most fungal infections; hence, we urgently need more rapid and reliable diagnostic tests for fungal infections. Although there are several new drugs that have contributed towards the rise of human fungal infections in modern medicine, there are some anti-cancer drugs that have been found to additionally protect against fungal diseases. Checkpoint inhibitors (CPIs) are a promising treatment for haematological malignancies and target immunosuppressive molecules on T-cells such as PD-1 and CTLA-4, thus enhancing anti-tumour immunity [61], but may also enhance T-cell responses to pathogenic fungi. Interestingly, CPIs have been shown to protect against numerous fungal infections in murine models, including invasive candidiasis, disseminated cryptococcosis and histoplasmosis. Moreover, CPIs have been successfully used to reverse refractory mucormycosis in a patient, indicating that CPIs may have beneficial antifungal functions in humans [61,62]. Therefore, studying how CPIs and other new therapeutics affect antifungal immunity can lead to a greater understanding of mechanisms of protection, which will underpin novel therapeutics and diagnostics for fungal infections, which are sorely needed.

Funding

We would like to thank the Wellcome Trust Antimicrobials and Antimicrobial Resistance Doctoral Training Programme, the University of Birmingham and the British Infection Association for funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Roser, M. Life Expectancy. Published online at OurWorldInData.org. Available online: https://ourworldindata.org/life-expectancy (accessed on 31 December 2018).
  2. Bongomin, F.; Gago, S.; Oladele, R.O.; Denning, D.W. Global and Multi-National Prevalence of Fungal Diseases-Estimate Precision. J. Fungi 2017, 3, 57. [Google Scholar] [CrossRef]
  3. Rodrigues, M.L.; Albuquerque, P.C. Searching for a change: The need for increased support for public health and research on fungal diseases. PLoS Negl. Trop. Dis. 2018, 12, e0006479. [Google Scholar] [CrossRef] [PubMed]
  4. Lionakis, M.S.; Iliev, I.D.; Hohl, T.M. Immunity against fungi. JCI Insight 2017, 2, e93156. [Google Scholar] [CrossRef] [PubMed]
  5. Drummond, R.A.; Lionakis, M.S. Mechanistic insights into the role of C-type lectin receptor/CARD9 signaling in human antifungal immunity. Front. Cell Infect. Microbiol. 2016, 6. [Google Scholar] [CrossRef] [PubMed]
  6. Ferwerda, B.; Ferwerda, G.; Plantinga, T.S.; Willment, J.A.; van Spriel, A.B.; Venselaar, H.; Elbers, C.C.; Johnson, M.D.; Cambi, A.; Huysamen, C.; et al. Human Dectin-1 Deficiency and Mucocutaneous Fungal Infections. N. Eng. J. Med. 2009, 361, 1760–1767. [Google Scholar] [CrossRef] [Green Version]
  7. Marr, K.A. Fungal infections in hematopoietic stem cell transplant recipients. Med. Mycol. 2008, 46, 293–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Brown, G.D.; Denning, D.W.; Gow, N.A.R.; Levitz, S.M.; Netea, M.G.; White, T.C. Hidden Killers: Human Fungal Infections. Sci. Transl. Med. 2012, 4, 165rv113. [Google Scholar] [CrossRef] [PubMed]
  9. McKay, L.I.; Cidlowski, J.A. Molecular Control of Immune/Inflammatory Responses: Interactions Between Nuclear Factor-κB and Steroid Receptor-Signaling Pathways. Endocr. Rev. 1999, 20, 435–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Lionakis, M.S.; Netea, M.G.; Holland, S.M. Mendelian genetics of human susceptibility to fungal infection. Cold Spring Harbor Perspect. Med. 2014, 4, a019638. [Google Scholar] [CrossRef] [PubMed]
  11. Richards, D.F.; Fernandez, M.; Caulfield, J.; Hawrylowicz, C.M. Glucocorticoids drive human CD8+ T cell differentiation towards a phenotype with high IL-10 and reduced IL-4, IL-5 and IL-13 production. Eur. J. Immunol. 2000, 30, 2344–2354. [Google Scholar] [CrossRef] [Green Version]
  12. Luther, C.; Adamopoulou, E.; Stoeckle, C.; Brucklacher-Waldert, V.; Rosenkranz, D.; Stoltze, L.; Lauer, S.; Poeschel, S.; Melms, A.; Tolosa, E. Prednisolone Treatment Induces Tolerogenic Dendritic Cells and a Regulatory Milieu in Myasthenia Gravis Patients. J. Immunol. 2009, 183, 841. [Google Scholar] [CrossRef] [PubMed]
  13. Hooper, K.M.; Barlow, P.G.; Stevens, C.; Henderson, P. Inflammatory Bowel Disease Drugs: A Focus on Autophagy. J. Crohns Colitis 2017, 11, 118–127. [Google Scholar] [CrossRef] [PubMed]
  14. Williamson, P.R.; Jarvis, J.N.; Panackal, A.A.; Fisher, M.C.; Molloy, S.F.; Loyse, A.; Harrison, T.S. Cryptococcal meningitis: Epidemiology, immunology, diagnosis and therapy. Nat. Rev. Neurol. 2017, 13, 13–24. [Google Scholar] [CrossRef]
  15. Johansson, Å.C.; Ohlsson, S.; Pettersson, Å.; Bengtsson, A.A.; Selga, D.; Hansson, M.; Hellmark, T. Impaired phagocytosis and reactive oxygen species production in phagocytes is associated with systemic vasculitis. Arthritis Res. Ther. 2016, 18, 92. [Google Scholar] [CrossRef] [PubMed]
  16. Saffar, A.S.; Ashdown, H.; Gounni, A.S. The molecular mechanisms of glucocorticoids-mediated neutrophil survival. Curr. Drug Targets 2011, 12, 556–562. [Google Scholar] [CrossRef]
  17. Timsit, J.-F.; Bassetti, M.; Cremer, O.; Daikos, G.; de Waele, J.; Kallil, A.; Kipnis, E.; Kollef, M.; Laupland, K.; Paiva, J.-A.; et al. Rationalizing antimicrobial therapy in the ICU: A narrative review. Intensiv. Care Med. 2019, 45, 172–189. [Google Scholar] [CrossRef]
  18. Liu, M.; Huang, S.; Guo, L.; Li, H.; Wang, F.; Zhang, Q.I.; Song, G. Clinical features and risk factors for blood stream infections of Candida in neonates. Exp. Ther. Med. 2015, 10, 1139–1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Ben-Ami, R.; Olshtain-Pops, K.; Krieger, M.; Oren, I.; Bishara, J.; Dan, M.; Wiener-Well, Y.; Weinberger, M.; Zimhony, O.; Chowers, M.; et al. Antibiotic Exposure as a Risk Factor for Fluconazole-Resistant Candida Bloodstream Infection. Antimicrob. Agents Chemother. 2012, 56, 2518–2523. [Google Scholar] [CrossRef] [Green Version]
  20. Allonsius, C.N.; van den Broek, M.F.L.; De Boeck, I.; Kiekens, S.; Oerlemans, E.F.M.; Kiekens, F.; Foubert, K.; Vandenheuvel, D.; Cos, P.; Delputte, P.; et al. Interplay between Lactobacillus rhamnosus GG and Candida and the involvement of exopolysaccharides. Microb. Biotech. 2017, 10, 1753–1763. [Google Scholar] [CrossRef] [Green Version]
  21. Zelante, T.; Iannitti, R.G.; Cunha, C.; De Luca, A.; Giovannini, G.; Pieraccini, G.; Zecchi, R.; D’Angelo, C.; Massi-Benedetti, C.; Fallarino, F.; et al. Tryptophan Catabolites from Microbiota Engage Aryl Hydrocarbon Receptor and Balance Mucosal Reactivity via Interleukin-22. Immunity 2013, 39, 372–385. [Google Scholar] [CrossRef] [Green Version]
  22. Borghi, M.; De Luca, A.; Puccetti, M.; Jaeger, M.; Mencacci, A.; Oikonomou, V.; Pariano, M.; Garlanda, C.; Moretti, S.; Bartoli, A.; et al. Pathogenic NLRP3 Inflammasome Activity during Candida Infection Is Negatively Regulated by IL-22 via Activation of NLRC4 and IL-1Ra. Cell Host Microbe 2015, 18, 198–209. [Google Scholar] [CrossRef] [Green Version]
  23. Peters, B.M.; Palmer, G.E.; Nash, A.K.; Lilly, E.A.; Fidel, P.L., Jr.; Noverr, M.C. Fungal morphogenetic pathways are required for the hallmark inflammatory response during Candida albicans vaginitis. Infect Immun. 2014, 82, 532–543. [Google Scholar] [CrossRef] [PubMed]
  24. De Luca, A.; Carvalho, A.; Cunha, C.; Iannitti, R.G.; Pitzurra, L.; Giovannini, G.; Mencacci, A.; Bartolommei, L.; Moretti, S.; Massi-Benedetti, C.; et al. IL-22 and IDO1 Affect Immunity and Tolerance to Murine and Human Vaginal Candidiasis. PLoS Pathog. 2013, 9, e1003486. [Google Scholar] [CrossRef]
  25. Francino, M.P. Antibiotics and the Human Gut Microbiome: Dysbioses and Accumulation of Resistances. Front. Microbiol. 2016, 6, 1543. [Google Scholar] [CrossRef]
  26. Fan, D.; Coughlin, L.A.; Neubauer, M.M.; Kim, J.; Kim, M.S.; Zhan, X.; Simms-Waldrip, T.R.; Xie, Y.; Hooper, L.V.; Koh, A.Y. Activation of HIF-1[alpha] and LL-37 by commensal bacteria inhibits Candida albicans colonization. Nat. Med. 2015, 21, 808. [Google Scholar] [CrossRef]
  27. Lionakis, M.S. New insights into innate immune control of systemic candidiasis. Med. Mycol. 2014, 52, 555–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Odds, F.C.; Davidson, A.D.; Jacobsen, M.D.; Tavanti, A.; Whyte, J.A.; Kibbler, C.C.; Ellis, D.H.; Maiden, M.C.J.; Shaw, D.J.; Gow, N.A.R. Candida albicans strain maintenance, replacement, and microvariation demonstrated by multilocus sequence typing. J. Clin. Microbiol. 2006, 44, 3647–3658. [Google Scholar] [CrossRef]
  29. Berkow, E.L.; Lockhart, S.R. Fluconazole resistance in Candida species: A current perspective. Infect. Drug Resist. 2017, 10, 237–245. [Google Scholar] [CrossRef] [PubMed]
  30. Katsuno, G.; Takahashi, H.K.; Iwagaki, H.; Sugita, S.; Mori, S.; Saito, S.; Yoshino, T.; Nishibori, M.; Tanaka, N. The Effect of Ciprofloxacin on CD14 and Toll-like Receptor-4 Expression on Human Monocytes. Shock 2006, 25, 247–253. [Google Scholar] [CrossRef] [PubMed]
  31. Jensen, J.-U.S.M.D.P.; Hein, L.M.D.; Lundgren, B.M.D.D.; Bestle, M.H.M.D.P.; Mohr, T.M.D.P.; Andersen, M.H.M.D.; Loken, J.M.D.; Tousi, H.M.D.; Soe-Jensen, P.M.D.; Lauritsen, A.O.M.D.; et al. Invasive Candida Infections and the Harm from Antibacterial Drugs in Critically Ill Patients: Data from a Randomized, Controlled Trial to Determine the Role of Ciprofloxacin, Piperacillin-Tazobactam, Meropenem, and Cefuroxime. Crit. Care Med. 2015, 43, 594–602. [Google Scholar] [CrossRef]
  32. Yang, J.H.; Bhargava, P.; McCloskey, D.; Mao, N.; Palsson, B.O.; Collins, J.J. Antibiotic-Induced Changes to the Host Metabolic Environment Inhibit Drug Efficacy and Alter Immune Function. Cell Host Microbe 2017, 22, 757–765.e753. [Google Scholar] [CrossRef] [PubMed]
  33. Bae, H.-B.; Zmijewski, J.W.; Deshane, J.S.; Tadie, J.-M.; Chaplin, D.D.; Takashima, S.; Abraham, E. AMP-activated protein kinase enhances the phagocytic ability of macrophages and neutrophils. FASEB J. 2011, 25, 4358–4368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. O’Neill, L.A.J.; Kishton, R.J.; Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 2016, 16, 553–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kalghatgi, S.; Spina, C.S.; Costello, J.C.; Liesa, M.; Morones-Ramirez, J.R.; Slomovic, S.; Molina, A.; Shirihai, O.S.; Collins, J.J. Bactericidal Antibiotics Induce Mitochondrial Dysfunction and Oxidative Damage in Mammalian Cells. Sci. Trans. Med. 2013, 5, 192ra185. [Google Scholar] [CrossRef] [PubMed]
  36. Scott, N.A.; Andrusaite, A.; Andersen, P.; Lawson, M.; Alcon-Giner, C.; Leclaire, C.; Caim, S.; Le Gall, G.; Shaw, T.; Connolly, J.P.R.; et al. Antibiotics induce sustained dysregulation of intestinal T cell immunity by perturbing macrophage homeostasis. Sci. Trans. Med. 2018, 10, eaao4755. [Google Scholar] [CrossRef] [PubMed]
  37. Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Yu, D.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D.; et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Corrêa-Oliveira, R.; Fachi, J.L.; Vieira, A.; Sato, F.T.; Vinolo, M.A.R. Regulation of immune cell function by short-chain fatty acids. Clin. Trans. Immunol. 2016, 5, e73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Ivanov, I.I.; Frutos, R.d.L.; Manel, N.; Yoshinaga, K.; Rifkin, D.B.; Sartor, R.B.; Finlay, B.B.; Littman, D.R. Specific Microbiota Direct the Differentiation of IL-17-Producing T-Helper Cells in the Mucosa of the Small Intestine. Cell Host Microbe 2008, 4, 337–349. [Google Scholar] [CrossRef]
  40. Fahey, J.R.; Lyons, B.L.; Olekszak, H.L.; Mourino, A.J.; Ratiu, J.J.; Racine, J.J.; Chapman, H.D.; Serreze, D.V.; Baker, D.L.; Hendrix, N.K. Antibiotic-associated Manipulation of the Gut Microbiota and Phenotypic Restoration in NOD Mice. Comp. Med. 2017, 67, 335–343. [Google Scholar]
  41. Puel, A.; Daffinger, R.; Natividad, A.; Chrabieh, M.; Barcenas-Morales, G.; Picard, C.; Cobat, A.; Ouachace-Chardin, M.; Toulon, A.; Bustamante, J.; et al. Autoantibodies against IL-17A, IL-17F, and IL-22 in patients with chronic mucocutaneous candidiasis and autoimmune polyendocrine syndrome type I. J. Exp. Med. 2010, 207, 291–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Conti, H.R.; Shen, F.; Nayyar, N.; Stocum, E.; Sun, J.N.; Lindemann, M.J.; Ho, A.W.; Hai, J.H.; Yu, J.J.; Jung, J.W.; et al. Th17 cells and IL-17 receptor signaling are essential for mucosal host defense against oral candidiasis. J. Exp. Med. 2009, 206, 299–311. [Google Scholar] [CrossRef] [Green Version]
  43. Deshmukh, H.S.; Liu, Y.; Menkiti, O.R.; Mei, J.; Dai, N.; O’Leary, C.E.; Oliver, P.M.; Kolls, J.K.; Weiser, J.N.; Worthen, G.S. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat. Med. 2014, 20, 528–534. [Google Scholar] [CrossRef] [PubMed]
  44. Erny, D.; Hrabe de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965. [Google Scholar] [CrossRef] [PubMed]
  45. Puig, L. Brodalumab: The first anti-IL-17 receptor agent for psoriasis. Drugs Today 2017, 53, 283–297. [Google Scholar] [CrossRef]
  46. Saunte, D.M.; Mrowietz, U.; Puig, L.; Zachariae, C. Candida infections in patients with psoriasis and psoriatic arthritis treated with interleukin-17 inhibitors and their practical management. Br. J. Dermatol. 2017, 177, 47–62. [Google Scholar] [CrossRef] [PubMed]
  47. Stamatiades, G.A.; Ioannou, P.; Petrikkos, G.; Tsioutis, C. Fungal infections in patients with inflammatory bowel disease: A systematic review. Mycoses 2018, 61, 366–376. [Google Scholar] [CrossRef] [PubMed]
  48. Sheppard, D.C.; Yeaman, M.R.; Filler, S.G. Tumor Necrosis Factor Inhibition and Invasive Fungal Infections. Clin. Infect. Dis. 2005, 41, S208–S212. [Google Scholar]
  49. Ghez, D.; Calleja, A.; Protin, C.; Baron, M.; Ledoux, M.-P.; Damaj, G.; Dupont, M.; Dreyfus, B.; Ferrant, E.; Herbaux, C.; et al. Early-onset invasive aspergillosis and other fungal infections in patients treated with ibrutinib. Blood 2018. [Google Scholar] [CrossRef]
  50. Baron, M.; Zini, J.M.; Challan Belval, T.; Vignon, M.; Denis, B.; Alanio, A.; Malphettes, M. Fungal infections in patients treated with ibrutinib: Two unusual cases of invasive aspergillosis and cryptococcal meningoencephalitis. Leuk. Lymphoma 2017, 58, 2981–2982. [Google Scholar] [CrossRef]
  51. Bercusson, A.; Colley, T.; Shah, A.; Warris, A.; Armstrong-James, D. Ibrutinib blocks Btk-dependent NF-ĸB and NFAT responses in human macrophages during Aspergillus fumigatus phagocytosis. Blood 2018. [Google Scholar] [CrossRef]
  52. Singh, S.; Sadanandam, A.; Nannuru, K.C.; Varney, M.L.; Mayer-Ezell, R.; Bond, R.; Singh, R.K. Small-Molecule Antagonists for CXCR2 and CXCR1 Inhibit Human Melanoma Growth by Decreasing Tumor Cell Proliferation, Survival, and Angiogenesis. Clin. Cancer Res. 2009, 15, 2380. [Google Scholar] [CrossRef] [PubMed]
  53. Swamydas, M.; Gao, J.-L.; Break, T.J.; Johnson, M.D.; Jaeger, M.; Rodriguez, C.A.; Lim, J.K.; Green, N.M.; Collar, A.L.; Fischer, B.G.; et al. CXCR1-mediated neutrophil degranulation and fungal killing promote Candida clearance and host survival. Sci. Trans. Med. 2016, 8, ra310–ra322. [Google Scholar] [CrossRef] [PubMed]
  54. Drummond, R.A.; Collar, A.L.; Swamydas, M.; Rodriguez, C.A.; Lim, J.K.; Mendez, L.M.; Fink, D.L.; Hsu, A.P.; Zhai, B.; Karauzum, H.; et al. CARD9-dependent neutrophil recruitment protects against fungal invasion of the central nervous system. PLoS Pathog. 2015, 11, e1005293. [Google Scholar] [CrossRef] [PubMed]
  55. Krisenko, M.O.; Geahlen, R.L. Calling in SYK: SYK’s dual role as a tumor promoter and tumor suppressor in cancer. Biochim. Biophys. Acta 2015, 1853, 254–263. [Google Scholar] [CrossRef]
  56. Friedberg, J.W.; Sharman, J.; Sweetenham, J.; Johnston, P.B.; Vose, J.M.; Lacasce, A.; Schaefer-Cutillo, J.; De Vos, S.; Sinha, R.; Leonard, J.P.; et al. Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia. Blood 2010, 115, 2578–2585. [Google Scholar] [CrossRef] [PubMed]
  57. Whitney, P.G.; Bär, E.; Osorio, F.; Rogers, N.C.; Schraml, B.U.; Deddouche, S.; LeibundGut-Landmann, S.; Reis e Sousa, C. Syk Signaling in Dendritic Cells Orchestrates Innate Resistance to Systemic Fungal Infection. PLoS Pathog. 2014, 10, e1004276. [Google Scholar] [CrossRef] [PubMed]
  58. Said-Sadier, N.; Padilla, E.; Langsley, G.; Ojcius, D.M. Aspergillus fumigatus Stimulates the NLRP3 Inflammasome through a Pathway Requiring ROS Production and the Syk Tyrosine Kinase. PLoS ONE 2010, 5. [Google Scholar] [CrossRef] [PubMed]
  59. Brossart, P.; Kotthoff, P. Dexamethasone Promotes Fungal Infection By Inhibition of APC Activation with Beta-Glucans Via STAT-3 and NF-κb. Blood 2016, 128, 3710. [Google Scholar]
  60. Deepe, G.S., Jr.; McGuinness, M. Interleukin-1 and host control of pulmonary histoplasmosis. J. Infect. Dis. 2006, 194, 855–864. [Google Scholar] [CrossRef]
  61. Daver, N.; Kontoyiannis, D.P. Checkpoint inhibitors and aspergillosis in AML: The double hit hypothesis. Lancet Oncol. 2017, 18, 1571–1573. [Google Scholar] [CrossRef]
  62. Grimaldi, D.; Pradier, O.; Hotchkiss, R.S.; Vincent, J.-L. Nivolumab plus interferon-beta in the treatment of intractable mucormycosis. Lancet Infect Dis. 2017, 17, 18. [Google Scholar] [CrossRef]
Figure 1. Iatrogenic interference with the initiation of antifungal immunity. Pathogen-associated molecular patterns (PAMPs) in the fungal cell wall (e.g., β-glucan) are detected by pattern recognition receptors (PRRs) on the plasma membranes of host phagocytes (e.g., Dectin-1). This initiates Syk-dependent assembly of the CBM (CARD9-BCL10-MALT1) signalosome, leading to NF-κB activation, mitogen-activated protein kinase (MAPK) signalling and a calcium flux, resulting in antifungal cellular responses. Treatments for comorbidities, such as anti-cancer therapies plus antibiotics, can interfere with this fungal detection pathway, as highlighted by red inhibitory arrows. Syk, spleen tyrosine kinase; CARD9, caspase recruitment domain-containing protein 9; MALT1, mucosa associated lymphoid tissue translocation protein 1; BCL10, B-cell lymphoma 10; IL, interleukin; Th, T helper; MAF, macrophage-activating factor; ROS, reactive oxygen species; NFκB, nuclear factor kappa B.
Figure 1. Iatrogenic interference with the initiation of antifungal immunity. Pathogen-associated molecular patterns (PAMPs) in the fungal cell wall (e.g., β-glucan) are detected by pattern recognition receptors (PRRs) on the plasma membranes of host phagocytes (e.g., Dectin-1). This initiates Syk-dependent assembly of the CBM (CARD9-BCL10-MALT1) signalosome, leading to NF-κB activation, mitogen-activated protein kinase (MAPK) signalling and a calcium flux, resulting in antifungal cellular responses. Treatments for comorbidities, such as anti-cancer therapies plus antibiotics, can interfere with this fungal detection pathway, as highlighted by red inhibitory arrows. Syk, spleen tyrosine kinase; CARD9, caspase recruitment domain-containing protein 9; MALT1, mucosa associated lymphoid tissue translocation protein 1; BCL10, B-cell lymphoma 10; IL, interleukin; Th, T helper; MAF, macrophage-activating factor; ROS, reactive oxygen species; NFκB, nuclear factor kappa B.
Pathogens 08 00045 g001
Figure 2. Antibiotics affect lactobacilli-mediated modulation of NLRP3 activation, predisposing to vulvovaginal candidiasis (VVC). Lactobacilli induce AhR (aryl hydrocarbon receptor)-dependent IL-22 (interleukin-22) transcription, promoting barrier function and regulation of mucosal immune responses through NLRC4-mediated production of the IL-1 receptor antagonist (IL1Ra), which inhibits activation of the NLRP3 inflammasome and production of IL-1β, IL-1α and IL-18, preventing neutrophil recruitment and C. albicans infection. Antibiotic treatment eradicates lactobacilli from the vaginal tract, leading to NLRP3 inflammasome activation and pro-inflammatory cytokine release, leading to dysbiosis and infection.
Figure 2. Antibiotics affect lactobacilli-mediated modulation of NLRP3 activation, predisposing to vulvovaginal candidiasis (VVC). Lactobacilli induce AhR (aryl hydrocarbon receptor)-dependent IL-22 (interleukin-22) transcription, promoting barrier function and regulation of mucosal immune responses through NLRC4-mediated production of the IL-1 receptor antagonist (IL1Ra), which inhibits activation of the NLRP3 inflammasome and production of IL-1β, IL-1α and IL-18, preventing neutrophil recruitment and C. albicans infection. Antibiotic treatment eradicates lactobacilli from the vaginal tract, leading to NLRP3 inflammasome activation and pro-inflammatory cytokine release, leading to dysbiosis and infection.
Pathogens 08 00045 g002

Share and Cite

MDPI and ACS Style

Clark, C.; Drummond, R.A. The Hidden Cost of Modern Medical Interventions: How Medical Advances Have Shaped the Prevalence of Human Fungal Disease. Pathogens 2019, 8, 45. https://doi.org/10.3390/pathogens8020045

AMA Style

Clark C, Drummond RA. The Hidden Cost of Modern Medical Interventions: How Medical Advances Have Shaped the Prevalence of Human Fungal Disease. Pathogens. 2019; 8(2):45. https://doi.org/10.3390/pathogens8020045

Chicago/Turabian Style

Clark, Callum, and Rebecca A. Drummond. 2019. "The Hidden Cost of Modern Medical Interventions: How Medical Advances Have Shaped the Prevalence of Human Fungal Disease" Pathogens 8, no. 2: 45. https://doi.org/10.3390/pathogens8020045

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