Abstract
IL-17 (IL-17A) has emerged as a key mediator of protection against extracellular microbes, but this cytokine also drives pathology in various autoimmune diseases. Overwhelming data in both humans and mice reveal a clear and surprisingly specific role for IL-17 in protection against the fungus Candida albicans, a commensal of the human oral cavity, gastrointestinal tract and reproductive mucosa. The IL-17 pathway regulates antifungal immunity through upregulation of pro-inflammatory cytokines including IL-6, neutrophil-recruiting chemokines such as CXCL1 and CXCL5 and antimicrobial peptides such as the defensins, which act in concert to limit fungal overgrowth. This review will focus on diseases caused by C. albicans, the role of IL-17-mediated immunity in candidiasis, and the implications for clinical therapies for both autoimmune conditions and fungal infections.
Introduction
Extensive research effort has centered on the role of the bacterial flora in human health and disease. Less well understood is the pathogenesis of the fungal species that inhabit our bodies. Fungi of the species Candida, dominantly C. albicans, are commensal microbes of the mouth, gastrointestinal tract, skin and vagina of healthy individuals (1). When host immunity is compromised, either through antibiotic use, barrier breach or immunodeficiency, pathogenic infection by C. albicans is a frequent consequence (2). There are no effective vaccines for C. albicans, or indeed for any fungi, and the development of Candida strains resistant to antifungal therapy is an increasing problem (3). In recent years the identification of genetic defects in mice and humans that impact the Th17/IL-17 axis has revealed the central importance of this pathway in controlling C. albicans infections, which is the subject of this review.
1. Infections caused by C. albicans
Several species of Candida cause candidiasis, though C. albicans is the most frequently isolated and is by far the best characterized. The other major disease-causing non-albicans species include C. glabrata, C. tropicalis, C. krusei, C. dubliniensis and C. parapsilosis (4). Most pathogenic Candida species are dimorphic, existing as yeast or pseudohyphal and hyphal forms. For these species, dimorphism is a key feature of virulence, and the tissue-invasive hyphal form is generally the most pathogenic (5). The recognition of different morphotypes by the host permits discrimination between commensal and pathogenic disease-causing forms of C. albicans (6–9) (see section 2 below).
1A. Mucocutaneous Candidiasis
There are multiple manifestations of candidiasis, differing in the immune response invoked. Mucocutaneous candidiasis broadly encompasses infections of the mucosae, nail and skin surfaces. C. albicans infection in the oral cavity is termed oropharyngeal candidiasis (OPC) or thrush, and is often mild and self-limiting. OPC is one of the first clinical signs of HIV, and OPC is common in neonates, the elderly, patients with xerostomia (dry-mouth) and individuals undergoing chemotherapy and radiotherapy for head-neck cancers. Severe cases in infants can lead to malnutrition and a failure to thrive. OPC is also a risk factor for esophageal cancer (10).
Chronic mucocutaneous candidiasis (CMC) presents as OPC and superficial lesions on the mucosa or thickened skin and nails, and is typically refractory to treatment. CMC occurs in patients with underlying genetic defects in IL-17-related immunity (10, 11). Although not life threatening, significant morbidity is associated with OPC and CMC due to pain, weight loss and decreased nutritional intake.
1B. Vaginal Candidiasis
C. albicans colonizes the reproductive tract in most women without pathological consequence, but at least one episode of vulvovaginal candidiasis (VVC) is diagnosed in 75% of women of reproductive age (12). Recurrent VVC, though infrequent, is associated with significant treatment costs and decreased quality of life. Consequently, experimental vaccines against Candida are being evaluated in the context of chronic VVC (13, 14).
1C. Disseminated Candidiasis
Systemic candidiasis is the most severe form of Candida infection. Candida species represent the fourth most common cause of bloodstream infections in U.S. hospitals, with a mortality rate of 40–60% (10). Systemic candidiasis is typically caused by medical intervention, including indwelling catheters, antibiotics or abdominal surgery. Notably, mucocutaneous overgrowth of C. albicans is not usually associated with invasive disease, indicating tissue-specific compartmentalization of responses to Candida (15, 16).
1D. Candida in the intestinal tract
Although C. albicans does not usually invade intestinal tissue to cause disease, Candida species colonize the GI tract and can translocate to the bloodstream during intestinal barrier breaches (10, 17). While GI translocation of C. albicans into circulation is not common, systemic invasion resulting from damage to the GI tract, as during abdominal surgery, is a significant problem (18).
As will be described throughout this review, there are numerous immune mechanisms that participate in anti-Candida immunity, the dominance of which varies among tissues. Oral and dermal candidiasis are strongly IL-17-dependent, whereas immunity to vaginal candidiasis relies more on extrinsic factors such as microbial flora and changes in pH (14, 19). Although systemic candidiasis has an IL-17 component, IFNγ from Th1 and NK cells seems to play a relatively more important role (1, 20). Studies using an intragastric colonization model indicated that Th1 cells and IL-22 were the dominant protective factors, whereas Th17 cells and IL-17 promote tissue destruction in this setting (21, 22). Murine models of disseminated, OPC, vaginal and cutaneous candidiasis are established that recapitulate human candidiasis with reasonable fidelity (23–26), and thus offer a cost-effective platform to study the immune response to Candida and to facilitate development of new therapeutics.
2. Pattern Recognition of Candida albicans
Although excellent reviews of the pattern recognition receptors (PRRs) involved in recognition of C. albicans are available (27–29), a brief discussion of this topic is in order. C-type lectin receptors (CLRs), particularly Dectin-1, are the main sensors of Candida species, although there are also significant contributions from TLRs and Nod-like receptors. The Candida cell wall consists of an outer mannoprotein layer that conceals an inner layer composed of β-glucan derivatives and chitin. CLRs recognize carbohydrate moieties found in the fungal cell wall, including mannans (Dectin-2, −3, Mincle, the mannose receptor, among others), chitin (receptor unknown), and β-glucan (Dectin-1). Activation of these PRRs triggers NF-κB and other downstream signals, triggering an inflammatory response. The dimorphic nature of C. albicans is part of its immune evasion strategy, as mannans in the external cell wall largely shield the β-glucans from exposure and thereby limit Dectin-1 signaling (30). The host response is activated during budding or transition to hyphae, when mannan reconfiguration exposes the glucan layer. Interestingly, anti-fungal drugs such as caspofungin may act by unmasking β-glucans and activating the immune system (31). Activation of certain TLRs is also anti-inflammatory, which helps to maintain homeostasis in the face of commensalism (30).
3. IL-17 in candidiasis
In 2005, the discovery of the “Th17” cell population fundamentally altered how CD4-dependent immunity was understood (32). Th17 cells arise from naïve precursors through signals from IL-1β, IL-6, TGFβ and IL-23. These cells express IL-17 (IL-17A) as well as IL-17F, IL-21 IL-22 and GM-CSF, and express characteristic factors including CCR6 and RORγt. Historically, one of the first indications that CD4+ T cells were vital in protection against C. albicans infection came from HIV/AIDS patients, nearly all of whom exhibited OPC (33, 34). Subsequently, human T cells with reactivity to C. albicans were found to be predominantly of the Th17 subset (35). In this regard, studies of human T cells ex vivo showed that C. albicans primes Th17 cells that produce IL-17 and IFN-γ, but not IL-10. Interestingly, this property was not generalizable, as Staphylococcus aureus-primed Th17 cells produce IL-10, which may constrain immune pathology. The differences in the response induced by distinct pathogens at the priming and effector stage is due to the different cytokine environments induced by each microbe, with IL-1β and IL-2 being important for the pro- and anti-inflammatory effects of C. albicans-specific Th17 cells respectively (36).
Studies conducted prior to the recognition of Th17 cells reported that IL-12 and Th1 cells were protective in mucosal candidiasis. This conclusion was based partly on the susceptibility of IL-12p40−/− mice to OPC (2, 37) and was reasonable given the Th1-Th2 paradigm that prevailed at the time (38). Studies in a GI candidiasis model also indicated that Th1 cells were protective, whereas IL-17 activity was detrimental (22). However, mice lacking IFN-γ were resistant to OPC (39) and IL-17RA-deficient mice were susceptible to systemic candidiasis (20). Moreover, mice deficient in IL-17RA or IL-23p19, but not IL-12p35, are susceptible to oral infection (40). Consistently, defects anywhere along the IL-17 signaling pathway predispose to oral candidiasis, including IL-17RA, IL-17RC and Act1 (40–42). Strikingly, parallel defects are also seen in humans (see section 6) (43). Correlating with susceptibility in these settings is defective neutrophil recruitment and impaired antimicrobial peptide (AMP) production (40–42).
In VVC the role of IL-17-mediated immune responses is especially controversial. In this setting, neutrophils are more damaging than host-protective. Rather, resistance to VVC centers on maintenance of the epithelial layer and a balanced vaginal microbial flora (44). Whereas one report demonstrated a protective role for IL-17 cells in an estrogen-induced model of VVC, another concluded that the neutrophil response was not linked to Th17 cells (14, 45). Notably, humans with mutations in the IL-17 axis are not particularly susceptible to VVC (46). Thus, additional studies to elucidate the role of IL-17 in VVC are needed.
In systemic candidiasis, IL-17 is also important, as IL-17A−/− and IL-17RA−/− mice are more susceptible to infection than WT mice (20, 47, 48). In addition, there are key roles for IFNγ and TNFα in driving neutrophil recruitment to affected organs as well as enhancing the fungicidal activity of phagocytes (49–54). Experimental vaccines targeting C. albicans that are protective in systemic candidiasis, and notably generate both Th1 and Th17 responses; probably probably both cell subsets are needed for effective immunity (55). A recent report suggests that IL-17-dependent signaling in candidiasis does not occur locally, but instead targets bone marrow to stimulate NK cell production of GM-CSF; this cytokine in turn induces the candidacidal activity of neutrophils in the kidney (56).
Surprisingly little is known about IL-17-dependent immunity to non-albicans Candida species. Bar et al. identified an epitope from the C. albicans ALS1/3 adhesin molecule that is recognized by Th17 cells. This epitope is conserved among Candida species, including C. dubliniensis, C. tropicalis, C. krusei and C. glabrata (57). In addition, a patient with C. dubliniensis meningitis as a result of CARD9 deficiency exhibited a reduced Th17 cell frequency (58). C. tropicalis is part of the commensal mycobiome in the mouse intestine, and its proportion increases in colitis-prone Dectin-1−/− mice, correlating with increased levels of IL-17 as well as IFNγ and TNFα (59). Our recent analysis of C. tropicalis systemic infection in mice revealed, unexpectedly, that IL-17R/Act1 signaling does not contribute to immunity, whereas CARD9 and TNFα in neutrophils are essential (60). Clearly more analysis will be essential to understand immune responses to other fungi.
4. Sources of IL-17 in Candidiasis
Conventional CD4+ Th17 cells differentiate upon exposure to IL-6, TGF-β and IL-1β and the transcription factors (TFs) RORγt and STAT3 (Figure 1). IL-23, though not required for Th17 development, promotes the maintenance and function of Th17 cells in a STAT3-dependent manner. In addition, various innate cell types express IL-17 in an IL-23- and RORγt-dependent manner, referred to broadly as ‘Type 17’ cells (61). Type 17 lymphoid subsets include iNKT, LTi γδ-T and natural Th17 (nTh17) cells. IL-17-expressing cells lacking an antigen receptor are classified as Group 3 innate lymphoid cells (ILC3s) (62). In some settings, neutrophils have been reported to express IL-17, though so far this does not seem to be the case in candidiasis (63–65). Innate Type 17 cells tend to reside in non-lymphoid tissues, where they are poised to be activated rapidly in an antigen-independent manner (61).
The awareness that IL-17 is produced by innate cell types as well as conventional Th17 cells stimulated studies to define the cellular sources of IL-17 during Candida infections (61). Newborns are highly prone to thrush, yet the disease is generally self-limiting, indicating that innate mechanisms effectively control oral C. albicans (2). Unlike humans, mice do not harbor C. albicans as a commensal microbe, and studies in a re-challenge model of OPC demonstrated that there is no pre-existing cross-reactive immunity to components in commensal microbiota or food (57, 66). Consistently, CD4-deficient mice are resistant to acute OPC (66), and the standard model of OPC used in the field (where clearance occurs 5 days) (23) reflects the innate response (59, 67) (Figure 1). Consistently, IL-17 mRNA is detected within 24 hours of infection (40, 66), pointing to an innate rather than adaptive origin of IL-17. Using a fate tracking reporter mouse system (68), we identified oral-resident IL-17+ γδ-T cells and nTh17 cells following encounter with C. lbicans (65). Work by Gladiator et al. alternatively suggested a role for ILC3s in OPC (69), though the concept is inconsistent with the high susceptibility of Rag1−/− mice (which have ILCs) to OPC (66, 70). An innate source of IL-17 in OPC agrees with data in dermal candidiasis demonstrating IL-17 production by γδ-T cells (68, 71, 72).
In adults, defects in conventional Th17 cells are consistently associated with CMC (73, 74), indicating that the adaptive response is dominant in anti-Candida immunity in humans. While innate responses to mucosal candidiasis apparently predominate in infants and naïve mice (Figure 1), generation of adaptive Th17 cells in murine OPC does confer additional protection (66, 75). The relative importance of the innate response in mice compared to humans may reflect important species differences. It is plausible, for example, that mice may require more robust oral innate immunity, since they are coprophagic.
Specific morphologies of C. albicans direct Th17 differentiation. The Kaplan group recently reported that the yeast form of C. albicans promotes development of a protective Th17 response through Dectin-1. Filamentous hyphae, independently of dectin-1, induce a Th1 response that is protective in a subsequent systemic challenge (6). Mucosal surfaces provide the interface where the transition from health to pathogenic state occurs, so understanding what provokes immune defenses at this intersection is essential. The switch between commensal and pathogenic C. albicans has been attributed to signaling differences between yeast and hyphae in oral epithelial cells, with distinct differences in downstream MAPK signaling seen between morphotypes (76).
5. IL-17-mediated Mechanisms of fungal immunity
How does IL-17 mediate antifungal immunity? The IL-17 family consists of 6 cytokines (IL-17A–IL-17F) and 5 receptors (IL-17RA-IL-17RE). IL-17A and IL-17F form homo- and heterodimers and signal through a dimer of IL-17RA and IL-17RC (77–79). Upon engagement of ligand, IL-17RA/RC recruits Act1 and TRAF6, E3 ubiquitin ligases that trigger activation of downstream NF-κB, C/EBP and MAPK pathways (43). IL-17 signaling occurs primarily in nonhematopoietic cells, due to restricted expression of the IL-17RC subunit (80). However, exceptions to this paradigm have been observed in studies of fungal infection. Specifically, IL-17RC is reported to be expressed in human neutrophils during Aspergillus fumigatus infections, and PMNs were reported to both produce and respond to IL-17 (81). Additionally, IL-17 was shown to act on bone marrow to drive NK-dependent functions such as GM-CSF production during systemic candidiasis (56).
Because most nonhematopoietic cells respond to IL-17, the majority of characteristic IL-17 signature genes are upregulated in cells of mesenchymal, epithelial and endothelial origin. The profile of IL-17 target genes is illuminating regarding its function. Genes regulated by IL-17 encode proinflammatory cytokines such as IL-6 and factors important for neutrophil function and trafficking, such as G-CSF, CXC chemokines (CXCL1, CXCL2, CXCL5) and calprotectin (S100A8/9) (82). IL-17 also induces CCL20, the ligand for CCR6, a chemokine receptor characteristic of Type 17 cells. Upregulation of CCL20 may function to recruit more IL-17 producing immune cells to the infected tissue during ongoing fungal overgrowth (Figure 1). Some target genes, however, have a more restricted expression pattern. Of particular relevance to fungal immunity, IL-17 stimulation of epithelial cells or keratinocytes induces β-defensins (BDs), AMPs with potent candidacidal activity. In mice, BD3 is a dominant IL-17-dependent gene induced during acute OPC (40). Intriguingly, BD3 and its human orthologue, BD2 are ligands for CCR6, though it is not clear whether BD2 and BD3 actually function as chemoattractants for CCR6+ cells in situ (83). BD1 also plays an important protective role in murine OPC (84). Another class of AMPs with candidacidal activity are histatins, which are highly expressed in human salivary gland (85). Patients with Hyper-IgE/Job’s syndrome exhibit reduced Th17 frequencies due to mutations in STAT3 (section 6), and exhibit reduced levels of salivary β-defensins and histatins. Moreover, IL-17 can directly induce histatin expression in human salivary gland cells in vitro (86). Thus, immunity to OPC is a function of cumulative IL-17-dependent gene regulation.
IL-17 is part of a family of related cytokines with overlapping activities (87). IL-17F is the most conserved, but does not appear to participate in anti-Candida immunity based on knockout mouse or antibody blocking studies (47, 88). IL-17C signals through a receptor consisting of IL-17RA paired with IL-17RE, and has considerable functional overlap with IL-17A (89). Epithelial cells, not hematopoietic cells, produce IL-17C, inducing a gene profile strikingly similar to IL-17A (89–93). Unlike IL-17A, however, IL-17C plays no detectable role in protection to oral, dermal or disseminated candidiasis in mouse models (94). Since IL-17C is pathogenic in psoriasis, this cytokine may prove to be another effective therapeutic target that could avoid potentially adverse side effects in fungal susceptibility.
Th17 cells produce IL-22 as another key signature cytokine. Although not part of the IL-17 family based on sequence homology, IL-22 promotes immunity to OPC as well as to gastric candidiasis (22, 40, 95). Like IL-17, IL-22 exerts its primary activities on non-hematopoietic cells, particularly epithelial cells. However, the signaling mechanisms induced by IL-22 are strikingly different from those activated by IL-17; whereas IL-17 activates TRAF/NF-κB signaling, IL-22 activates the JAK-STAT pathway, primarily STAT3 (43). The sensitivity of certain human populations to candidiasis implicates IL-22 (section 6). For example, patients with autoimmune polyendocrinopathy syndrome-1 (APS-1) exhibit neutralizing Abs against both IL-17 family members as well as IL-22, and Job’s syndrome patients have mutations in STAT3 (10).
The microbiome has generated much interest of late, and certainly contributes to antifungal immunity. Antibiotics are a risk factor for candidiasis, and administration of antibacterial agents concomitantly increases the abundance of fungi in the intestine (96). In a related subject, fungi constitute a significant but often overlooked part of the microbiome, sometimes termed the “mycobiome” (17). A seminal study in 2012 showed that there is a complex community of fungal species in the murine intestine that influences host immunity, in part through Dectin-1 (59). This is relevant to human autoimmune diseases, as SNPs in the genes encoding Dectin-1 (CLEC7A) and CARD9 are associated with risk for inflammatory bowel disease (59, 97). Although less well studied for other autoimmune conditions, fungal components such as zymosan can exacerbate pathology, at least in autoimmune models (98).
6. Defects in the IL-17 pathway in humans
‘Experiments of nature’ have been remarkably enlightening in validating the correlates of immunity to candidiasis in humans. Genetic defects underlying CMC have been defined through candidate gene or whole exome sequencing approaches, and nearly all link directly to the IL-17/Th17 pathway (73). Deficiencies include pattern recognition of Candida (CARD9, DECTIN1), factors involved in Th17 cell differentiation (IL12B, IL12RB1, STAT1, STAT3, TYK2), IL-17 signaling (IL17F,IL17RA, ACT1, IL17RC) and anti-IL-17 autoantibodies (AIRE) (73, 99).
6A. Pattern Recognition
Dectin-1 engagement on C. albicans is an important initiator of Th17 responses (28), and a DECTIN1 polymorphism is associated with increased susceptibility to CMC. APCs from patients with these SNPs showed defective production of IL-6 upon exposure to Candida. While disease is typically mild, these individuals present with VVC and onychomycosis, thought to be due to decreased IL-17 production (100). This polymorphism was also associated with increased Candida colonization in hematopoietic stem cell transplant recipients (101). Consistently, Dectin-1−/− mice show increased susceptibility to GI colonization with C. albicans and disseminated candidiasis, though this is somewhat dependent on the strain of C. albicans employed (102). Common lab strains of C. albicans adapt to the in vivo environment, leading to differences in cell wall composition and nature, which can in turn influence dectin-1 recognition. For exaple, differences in chitin deposition influence dectin-1 recognition of β-glucans and consequent susceptibility to candidiasis (103). Understanding how to manipulate the fungal cell wall is a ripe area for therapeutic intervention; as noted (section 2), caspofungin disrupts fungal cell walls to unmask β-glucan moieties (31).
CARD9 is an adaptor downstream of most CLRs (104). CARD9-deficient mice are susceptible to disseminated candidiasis and Th17 differentiation capacity is impaired (105). CARD9 is likewise critical for adaptive Th17 responses to OPC, but was unexpectedly dispensable for IL-17-mediated innate responses in the oral cavity (75). This intriguing finding raises the possibility that CARD9-independent pathways, perhaps through Dectin-1/Raf signaling, may dominate in the innate response (28). Such a model would be consistent with the concept of “trained immunity” to Candida described by Netea and colleagues, in which primary exposure to a pathogen improves the activity of monocytes to respond to re-challenge (106). In humans, a rare loss of function mutation in CARD9 has been described that is associated with Th17-deficiency. Disease was far more severe than in patients with a DECTIN1 mutations, suggesting that additional CARD9-dependent CLRs contribute to the response to C. albicans (107). Notably, CARD9 deficiency is the only setting where invasive candidiasis is described in humans.
6B. JAK-STAT pathway
Several mutations in the JAK-STAT pathway are associated with CMC and the Th17 pathway. Hyper-IgE syndrome (HIES, Job’s syndrome) is a primary immunodeficiency characterized by elevated IgE, dermatitis, recurrent infections of the skin and lungs and CMC (73). Autosomal dominant-HIES is caused by dominant negative mutations in the DNA-binding or SH2 domains of STAT3 (108, 109). STAT3 signals downstream of IL-6, IL-21 and IL-23, and is required for development of conventional Th17 cells. The promiscuous role of STAT3 signaling in the IL-23/IL-17 axis helps explain the Th17-deficiency and the concomitant increased susceptibility to CMC in HIES (Figure 1). Surprisingly, mice lacking STAT3 in CD4+ cells are not susceptible to acute OPC, perhaps indicating a reduced requirement for this transcription factor in innate Type 17 cells (65, 110). In addition to Th17 cells, salivary components are also defective in HIES. Increased oral colonization with C. albicans occurs in HIES patients, correlating with defects in IL-17-regulated salivary components such as BD2 and histatins. Consistently, saliva from individuals with HIES had decreased Candida-killing capacity compared to controls (86). This was also true in mice, as saliva from IL-23- and IL-17RA-deficient mice exhibited decreased levels of BD3 (orthologue of BD2) and Candida-killing properties in vitro (40). Thus, STAT3 exerts multifunctional antifungal activities in the context of candidiasis.
Multiple CMC patients with gain-of-function mutations in STAT1 have been identified (111–113). The link to Th17 pathways is somewhat indirect; STAT1 is downstream of Th17-pathway inhibitors, including IL-27, IFNγ and IFNα/β. Indeed, these mutations are associated with reduced Th17 frequencies. STAT1 mutations are comparatively common, with one report describing 12 missense mutations in 47 patients from 20 kindred groups. It is unclear exactly why these patients are susceptible to candidiasis, since Type I interferons are implicated in Candida immunity (114). Nonetheless, STAT1 mutations clearly lead to CMC susceptibility, likely via downstream signaling pathways that impact Th17 generation.
6C. IL-17 pathway
Individuals with mutations in IL-17A or IL-17RA offer compelling evidence for a direct role of IL-17 in antifungal immunity. A homozygous null mutation in IL-17RA was indentified in which fibroblasts were refractory to IL-17A and IL-17F signaling (115). Two individuals with IL17RC mutations were also recently identified (99). A family with autosomal dominant CMC due to a lack of IL-17A, IL-22 and Th17 cells has also been described (116). Additionally, a dominant-negative mutation in IL17F was discovered in a family with autosomal dominant CMC (115). IL-17F is a weaker agonist than IL-17A, but IL-17F and IL-17A form a heterodimer with intermediate signaling capacity (79). Indeed, the mutant IL-17F protein blocked signaling through the IL-17A:F heterodimer (115). In addition, ACT1 mutations in patients with CMC have been found that disrupt Act1 association with the IL-17R, underscoring the importance of IL-17 signaling (117).
Autoimmune polyendocrinopathy syndrome-1 (APS-1) is caused by mutations in the autoimmune regulator (AIRE) gene, causing aberrant thymic self-tolerance and multiorgan autoimmune disease. Intriguingly, most AIRE patients present with CMC. This singular susceptibility to candidiasis was at least partly explained by the discovery of neutralizing autoantibodies against Th17-related cytokines in these patients (118, 119). The most common are directed against type I interferons and Th17-related cytokines. Neutralization of IL-17 and related cytokines by these autoantibodies is thought to account for the increased susceptibility of APS-1 patients to CMC.
Cumulatively, these rare but informative genetic disorders provide compelling evidence for the centrality of the Th17/ IL-17 pathway in controlling Candida infections. That most of these mutations (except CARD9) are restricted to mucosal disease indicates that mucosal barriers are maintained even without functional Th17/IL-17 activity. The direct link between neutralizing autoantibody production and Candida infection seen in APS-1 also raises concerns regarding the clinical use of anti-IL-17 antibody therapies, as outlined below.
7. Anti-IL-17 Therapies and implications for antifungal immunity
Aberrant IL-17 production is linked to inflammation in autoimmunity. Psoriasis is emerging as a particularly strong IL-17-driven disorder (43, 89). Psoriasis is linked to IL-23-mediated activation of CD4+Th17 cells and innate Type 17 cells. IL-17 and IL-22 produced by these subsets interact with skin-resident keratinocytes, fibroblasts and endothelial cells to promote cell division and production of cytokines, chemokines and AMPs (120). This enhanced inflammatory state leads to exacerbated recruitment of neutrophils, mast cells and macrophages, and ultimately epidermal hyperplasia. The roles of IL-23 and IL-17 in psoriasis consequently make them attractive therapeutic targets (43). A number of biologic drugs targeting IL-17A/F, IL-17RA, and the IL-12p40 or IL-23p19 subunits of IL-23 are being used or evaluated in patients with psoriasis with impressive efficacy (121–123).
On the flip side, an obvious prediction of biologic anti-IL-17 therapies is an increased risk of Candida infections. Trials with anti-IL-17A antibodies indeed indicate that OPC is a side effect, though all cases so far are mild (121). A meta-analysis of opportunistic infections from anti-TNF therapies in IBD revealed that candidiasis occurs more frequently than often realized (124), which could conceivably relate to the characteristic signaling synergy between TNFα and IL17. In this regard, PBMCs from RA patients showed increased colonization and decreased oral anti-Candida responses compared to healthy controls (125). Since other human fungal pathogens also seem to involve the Th17 response, monitoring for fungal infections will be important facet of anti-IL-17 biologic agent usage.
8. Conclusions
The importance of IL-17 in candidiasis is now firmly established. In response to Candida at mucosal surfaces, IL-17 induces a protective neutrophil influx and AMPs that cooperate to control overgrowth and morphotype switching of Candida. The relative contribution of each component is an active area of research, and many questions remain (40, 88). Individuals with a wide-range of underlying conditions receive therapies that induce susceptibility to Candida infections, including glucocorticoids, radiotherapy or antibiotics. How the IL-17R signaling pathway is impacted in each modality of immunosuppression is poorly defined. With the advent of specific anti-Th17 therapies, the potential pool of patients at risk for Candida and other fungal opportunistic infections may expand considerably. The lack of effective vaccines for fungi and the increasing problem of antifungal resistance makes it imperative to understand the components involved in protection to this important pathogen (3, 59).
Acknowledgments
We thank N. Whibley for critical reading.
SLG was supported by NIH grants DE022550 and AI107825. HRC was supported by F32-DE023293.
Abbreviations
- CMC
chronic mucocutaneous candidiasis
- OPC
oropharyngeal candidiasis
- BD
β-defensin
- AMP
antimicrobial peptide
- VVC
vulvovaginal candidiasis
- CLR
C-type lectin receptor
- HIES
hyper-IgE syndrome
- APS-1
autoimmune polyendocrinopathy syndrome
- PRR
pattern recognition receptor
References
- 1.Romani L. Immunity to fungal infections. Nat Rev Immunol. 2011;11:275–288. doi: 10.1038/nri2939. [DOI] [PubMed] [Google Scholar]
- 2.Dongari-Bagtoglou A, Fidel P. The host cytokine responses and protective immunity in oropharyngeal candidiasis. J Dent Res. 2005;84:966–977. doi: 10.1177/154405910508401101. [DOI] [PubMed] [Google Scholar]
- 3.Brown GD, Denning DW, Gow NA, Levitz SM, Netea MG, White TC. Hidden killers: human fungal infections. Sci Transl Med. 2012;4:165rv113. doi: 10.1126/scitranslmed.3004404. [DOI] [PubMed] [Google Scholar]
- 4.Merseguel KB, Nishikaku AS, Rodrigues AM, Padovan AC, RC EF, Salles de Azevedo Melo A, da Silva Briones MR, Colombo AL. Genetic diversity of medically important and emerging Candida species causing invasive infection. BMC Infect Dis. 2015;15:57. doi: 10.1186/s12879-015-0793-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Jacobsen ID, Wilson D, Wachtler B, Brunke S, Naglik JR, Hube B. Candida albicans dimorphism as a therapeutic target. Exp Rev Anti-Infect Ther. 2012;10:85–93. doi: 10.1586/eri.11.152. [DOI] [PubMed] [Google Scholar]
- 6.Kashem SW, Igyarto BZ, Gerami-Nejad M, Kumamoto Y, Mohammed J, Jarrett E, Drummond RA, Zurawski SM, Zurawski G, Berman J, Iwasaki A, Brown GD, Kaplan DH. Candida albicans morphology and dendritic cell subsets determine T helper cell differentiation. Immunity. 2015;42:356–366. doi: 10.1016/j.immuni.2015.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Cheng SC, van de Veerdonk FL, Lenardon M, Stoffels M, Plantinga T, Smeekens S, Rizzetto L, Mukaremera L, Preechasuth K, Cavalieri D, Kanneganti TD, van der Meer JW, Kullberg BJ, Joosten LA, Gow NA, Netea MG. The dectin-1/inflammasome pathway is responsible for the induction of protective T-helper 17 responses that discriminate between yeasts and hyphae of Candida albicans. J Leukoc Biol. 2011;90:357–366. doi: 10.1189/jlb.1210702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.d’Ostiani CF, Del Sero G, Bacci A, Montagnoli C, Spreca A, Mencacci A, Ricciardi-Castagnoli P, Romani L. Dendritic cells discriminate between yeasts and hyphae of the fungus Candida albicans. Implications for initiation of T helper cell immunity in vitro and in vivo. J Exp Med. 2000;191:1661–1674. doi: 10.1084/jem.191.10.1661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.van der Graaf CA, Netea MG, Verschueren I, van der Meer JW, Kullberg BJ. Differential cytokine production and Toll-like receptor signaling pathways by Candida albicans blastoconidia and hyphae. Infect Immun. 2005;73:7458–7464. doi: 10.1128/IAI.73.11.7458-7464.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Huppler AR, Bishu S, Gaffen SL. Mucocutaneous candidiasis: the IL-17 pathway and implications for targeted immunotherapy. Arthritis Res Ther. 2012;14:217. doi: 10.1186/ar3893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Glocker E, Grimbacher B. Chronic mucocutaneous candidiasis and congenital susceptibility to Candida. Curr Opin Allergy Clin Immunol. 2010;10:542–550. doi: 10.1097/ACI.0b013e32833fd74f. [DOI] [PubMed] [Google Scholar]
- 12.McClelland RS, Richardson BA, Hassan WM, Graham SM, Kiarie J, Baeten JM, Mandaliya K, Jaoko W, Ndinya-Achola JO, Holmes KK. Prospective study of vaginal bacterial flora and other risk factors for vulvovaginal candidiasis. J Infect Dis. 2009;199:1883–1890. doi: 10.1086/599213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Schmidt CS, White CJ, Ibrahim AS, Filler SG, Fu Y, Yeaman MR, Edwards JE, Jr, Hennessey JP., Jr NDV-3, a recombinant alum-adjuvanted vaccine for Candida and Staphylococcus aureus, is safe and immunogenic in healthy adults. Vaccine. 2012;30:7594–7600. doi: 10.1016/j.vaccine.2012.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yano J, Noverr MC, Fidel PL., Jr Cytokines in the host response to Candida vaginitis: Identifying a role for non-classical immune mediators, S100 alarmins. Cytokine. 2012;58:118–128. doi: 10.1016/j.cyto.2011.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Fidel PL., Jr Candida-Host Interactions in HIV Disease: Implications for Oropharyngeal Candidiasis. Adv Dent Res. 2011;23:45–49. doi: 10.1177/0022034511399284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Conti HR, Gaffen SL. Host responses to Candida albicans: Th17 cells and mucosal candidiasis. Microbes Infect. 2010;12:518–527. doi: 10.1016/j.micinf.2010.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mukherjee PK, Sendid B, Hoarau G, Colombel JF, Poulain D, Ghannoum MA. Mycobiota in gastrointestinal diseases. Nat Rev Gastroenterol Hepatol. 2015;12:77–87. doi: 10.1038/nrgastro.2014.188. [DOI] [PubMed] [Google Scholar]
- 18.Kirkpatrick CH. Chronic mucocutaneous candidiasis. PIDS J. 2001;20:197–206. doi: 10.1097/00006454-200102000-00017. [DOI] [PubMed] [Google Scholar]
- 19.Hernández-Santos N, Gaffen SL. Th17 cells in immunity to Candida albicans. Cell Host Microbe. 2012;11:425–435. doi: 10.1016/j.chom.2012.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Huang W, Na L, Fidel PL, Schwarzenberger P. Requirement of interleukin-17A for systemic anti-Candida albicans host defense in mice. J Infect Dis. 2004;190:624–631. doi: 10.1086/422329. [DOI] [PubMed] [Google Scholar]
- 21.Mencacci A, Cenci E, Del Soro G, Fe D’Ostiani C, Mosci P, Bistoni F, Trinchieri G, Adorini L, Romani L. IL-10 is required for development of protective Th1 responses in IL-12-deficient mice upon Candida albicans infection. J Immunol. 1998;161:6228–6237. [PubMed] [Google Scholar]
- 22.Zelante T, De Luca A, Bonifazi P, Montagnoli C, Bozza S, Moretti S, Belladonna ML, Vacca C, Conte C, Mosci P, Bistoni F, Puccetti P, Kastelein RA, Kopf M, Romani L. IL-23 and the Th17 pathway promote inflammation and impair antifungal immune resistance. Eur J Immunol. 2007;37:2695–2706. doi: 10.1002/eji.200737409. [DOI] [PubMed] [Google Scholar]
- 23.Solis NV, Filler SG. Mouse model of oropharyngeal candidiasis. Nat Protoc. 2012;7:637–642. doi: 10.1038/nprot.2012.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Conti HR, Huppler AR, Whibley N, Gaffen SL. Animal models for candidiasis. Curr Protocol Immunol. 2014;105:19 16 11–19 16 17. doi: 10.1002/0471142735.im1906s105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Clancy C, Cheng S, Nguyen M. Animal Models of Candidiasis. In: Cihlar R, Calderone R, editors. Candida albicans: Methods and Protocols. Humana Press; 2009. pp. 65–76. [Google Scholar]
- 26.Cheng S, Clancy CJ, Xu W, Schneider F, Hao B, Mitchell AP, Nguyen MH. Profiling of Candida albicans gene expression during intra-abdominal candidiasis identifies biologic processes involved in pathogenesis. J Infect Dis. 2013;208:1529–1537. doi: 10.1093/infdis/jit335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Becker KL, Ifrim DC, Quintin J, Netea MG, van de Veerdonk FL. Antifungal innate immunity: recognition and inflammatory networks. Semin Immunopathol. 2015;37:107–116. doi: 10.1007/s00281-014-0467-z. [DOI] [PubMed] [Google Scholar]
- 28.Dambuza IM, Brown GD. C-type lectins in immunity: recent developments. Curr Opin Immunol. 2015;32:21–27. doi: 10.1016/j.coi.2014.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Cunha C, Carvalho A, Esposito A, Bistoni F, Romani L. DAMP signaling in fungal infections and diseases. Front Immunol. 2012;3:286. doi: 10.3389/fimmu.2012.00286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Netea MG, Brown GD, Kullberg BJ, Gow NA. An integrated model of the recognition of Candida albicans by the innate immune system. Nat Rev Microbiol. 2008;6:67–78. doi: 10.1038/nrmicro1815. [DOI] [PubMed] [Google Scholar]
- 31.Wheeler RT, Kombe D, Agarwala SD, Fink GR. Dynamic, morphotype-specific Candida albicans beta-glucan exposure during infection and drug treatment. PLoS Pathog. 2008;4:e1000227. doi: 10.1371/journal.ppat.1000227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Steinman L. A brief history of T(H)17, the first major revision in the T(H)1/T(H)2 hypothesis of T cell-mediated tissue damage. Nature Med. 2007;13:139–145. doi: 10.1038/nm1551. [DOI] [PubMed] [Google Scholar]
- 33.Centers for Disease, C. Pneumocystis pneumonia--Los Angeles. MMWR. 1981;30:250–252. [PubMed] [Google Scholar]
- 34.Greenspan D, Greenspan JS. Oral mucosal manifestations of AIDS? Dermatol Clin. 1987;5:733–737. [PubMed] [Google Scholar]
- 35.Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M, Lanzavecchia A, Sallusto F, Napolitani G. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nature Immunol. 2007;8:639–646. doi: 10.1038/ni1467. [DOI] [PubMed] [Google Scholar]
- 36.Zielinski CE, Mele F, Aschenbrenner D, Jarrossay D, Ronchi F, Gattorno M, Monticelli S, Lanzavecchia A, Sallusto F. Pathogen-induced human T(H)17 cells produce IFN-gamma or IL-10 and are regulated by IL-1beta. Nature. 2012;484:514–518. doi: 10.1038/nature10957. [DOI] [PubMed] [Google Scholar]
- 37.Cenci E, Mencacci A, Spaccapelo R, Tonnetti L, Mosci P, Enssle KH, Puccetti P, Romani L, Bistoni F. T helper cell type 1 (Th1)- and Th2-like responses are present in mice with gastric candidiasis but protective immunity is associated with Th1 development. J Infect Dis. 1995;171:1279–1288. doi: 10.1093/infdis/171.5.1279. [DOI] [PubMed] [Google Scholar]
- 38.Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol. 1986;136:2348–2357. [PubMed] [Google Scholar]
- 39.Farah C, Hu Y, Riminton S, Ashman R. Distinct roles for interleukin-12p40 and tumour necrosis factor in resistance to oral candidiasis defined by gene targeting. Oral Microbiol Immunol. 2006;21:252–255. doi: 10.1111/j.1399-302X.2006.00288.x. [DOI] [PubMed] [Google Scholar]
- 40.Conti H, Shen F, Nayyar N, Stocum E, JN S, Lindemann M, Ho A, Hai J, Yu J, Jung J, Filler S, Masso-Welch P, Edgerton M, Gaffen S. Th17 cells and IL-17 receptor signaling are essential for mucosal host defense against oral candidiasis. J Exp Med. 2009;206:299–311. doi: 10.1084/jem.20081463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ferreira MC, Whibley N, Mamo AJ, Siebenlist U, Chan YR, Gaffen SL. Interleukin-17-induced protein lipocalin 2 is dispensable for immunity to oral candidiasis. Infect Immun. 2014;82:1030–1035. doi: 10.1128/IAI.01389-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ho A, Shen F, Conti H, Patel N, Childs E, Peterson A, Hernandez-Santos N, Kolls J, Kane L, Ouyang W, Gaffen S. IL-17RC is required for immune signaling via an extended SEFIR domain in the cytoplasmic tail . J Immunol. 2010;185:1063–1070. doi: 10.4049/jimmunol.0903739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gaffen SL, Jain R, Garg A, Cua D. IL-23-IL-17 immune axis: Discovery, mechanistic understanding and clinical therapy. Nat Rev Immunol. 2014;14:585–600. doi: 10.1038/nri3707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Fidel PL., Jr History and update on host defense against vaginal candidiasis. Am J Reprod Immunol. 2007;57:2–12. doi: 10.1111/j.1600-0897.2006.00450.x. [DOI] [PubMed] [Google Scholar]
- 45.Pietrella D, Rachini A, Pines M, Pandey N, Mosci P, Bistoni F, d’Enfert C, Vecchiarelli A. Th17 cells and IL-17 in protective immunity to vaginal candidiasis. PLoS One. 2011;6:e22770. doi: 10.1371/journal.pone.0022770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rosentul DC, Delsing CE, Jaeger M, Plantinga TS, Oosting M, Costantini I, Venselaar H, Joosten LA, van der Meer JW, Dupont B, Kullberg BJ, Sobel JD, Netea MG. Gene polymorphisms in pattern recognition receptors and susceptibility to idiopathic recurrent vulvovaginal candidiasis. Front Microbiol. 2014;5:483. doi: 10.3389/fmicb.2014.00483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Saijo S, Ikeda S, Yamabe K, Kakuta S, Ishigame H, Akitsu A, Fujikado N, Kusaka T, Kubo S, Chung SH, Komatsu R, Miura N, Adachi Y, Ohno N, Shibuya K, Yamamoto N, Kawakami K, Yamasaki S, Saito T, Akira S, Iwakura Y. Dectin-2 recognition of alpha-mannans and induction of Th17 cell differentiation is essential for host defense against Candida albicans. Immunity. 2010;32:681–691. doi: 10.1016/j.immuni.2010.05.001. [DOI] [PubMed] [Google Scholar]
- 48.van de Veerdonk FL, Kullberg BJ, Verschueren IC, Hendriks T, van der Meer JW, Joosten LA, Netea MG. Differential effects of IL-17 pathway in disseminated candidiasis and zymosan-induced multiple organ failure. Shock. 2010;34:407–411. doi: 10.1097/SHK.0b013e3181d67041. [DOI] [PubMed] [Google Scholar]
- 49.Netea MG, H. LJ, van Tits LJH, Curfs JHAJ, Amiot F, Meis JFGM, van der Meer JWM, Kullberg BJ. Increased susceptibility of TNF-a Lymphotoxin-a double knockout mice to systemic candidiasis through impaired recruitment of neutrophils and phagocytosis of Candida albicans. J Immunol. 1999;163:1498–1505. [PubMed] [Google Scholar]
- 50.Ferrante A. Tumor necrosis factor alpha potentiates neutrophil antimicrobial activity: increased fungicidal activity against Torulopsis glabrata and Candida albicans and associated increases in oxygen radical production and lysosomal enzyme release. Infect Immun. 1989;57:2115–2122. doi: 10.1128/iai.57.7.2115-2122.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Marino MW, Dunn A, Grail D, Inglese M, Noguchi Y, Richards E, Jungbluth A, Wada H, Moore M, Williamson B, Basu S, Old LJ. Characterization of tumor necrosis factor-deficient mice. Proc Natl Acad Sci U S A. 1997;94:8093–8098. doi: 10.1073/pnas.94.15.8093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Filler SG, Yeaman MR, Sheppard DC. Tumor necrosis factor inhibition and invasive fungal infections. Clin Infect Dis. 2005;3(41 Suppl):S208–S212. doi: 10.1086/430000. [DOI] [PubMed] [Google Scholar]
- 53.Balish E, Wagner RD, Vazquez-Torres A, Pierson C, Warner T. Candidiasis in interferon-gamma knockout (IFN-gamma−/−) mice. J Infect Dis. 1998;178:478–487. doi: 10.1086/515645. [DOI] [PubMed] [Google Scholar]
- 54.Kaposzta R, Tree P, Marodi L, Gordon S. Characteristics of invasive candidiasis in gamma interferon- and interleukin-4-deficient mice: role of macrophages in host defense against Candida albicans. Infect Immun. 1998;66:1708–1717. doi: 10.1128/iai.66.4.1708-1717.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lin L, Ibrahim AS, Xu X, Farber JM, Avanesian V, Baquir B, Fu Y, French SW, Edwards JE, Jr, Spellberg B. Th1-Th17 cells mediate protective adaptive immunity against Staphylococcus aureus and Candida albicans infection in mice. PLoS Pathog. 2009;5:e1000703. doi: 10.1371/journal.ppat.1000703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Bar E, Whitney PG, Moor K, Reis e Sousa C, LeibundGut-Landmann S. IL-17 regulates systemic fungal immunity by controlling the functional competence of NK cells. Immunity. 2014;40:117–127. doi: 10.1016/j.immuni.2013.12.002. [DOI] [PubMed] [Google Scholar]
- 57.Bär E, Gladiator A, Bastidas S, Roschitzki B, Acha-Orbea H, Oxenius A, LeibundGut-Landmann S. A novel Th cell epitope of Candida albicans mediates protection from fungal infection. J Immunol. 2012;188:5636–5643. doi: 10.4049/jimmunol.1200594. [DOI] [PubMed] [Google Scholar]
- 58.Drewniak A, Gazendam RP, Tool AT, van Houdt M, Jansen MH, van Hamme JL, van Leeuwen EM, Roos D, Scalais E, de Beaufort C, Janssen H, van den Berg TK, Kuijpers TW. Invasive fungal infection and impaired neutrophil killing in human CARD9 deficiency. Blood. 2013;121:2385–2392. doi: 10.1182/blood-2012-08-450551. [DOI] [PubMed] [Google Scholar]
- 59.Iliev ID, Funari VA, Taylor KD, Nguyen Q, Reyes CN, Strom SP, Brown J, Becker CA, Fleshner PR, Dubinsky M, Rotter JI, Wang HL, McGovern DP, Brown GD, Underhill DM. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science. 2012;336:1314–1317. doi: 10.1126/science.1221789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Whibley N, Jaycox JR, Reid D, Garg AV, Taylor JA, Clancy CJ, Nguyen MH, Biswas1 PS, McGeachy MJ, Brown GD, Gaffen SL. Delinking CARD9 and IL-17: CARD9 protects against Candida tropicalis infection through a TNFα-dependent, IL-17-independent mechanism. in revision. 2015 doi: 10.4049/jimmunol.1500870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Cua DJ, Tato CM. Innate IL-17-producing cells: the sentinels of the immune system. Nat Rev Immunol. 2010;10:479–489. doi: 10.1038/nri2800. [DOI] [PubMed] [Google Scholar]
- 62.Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie AN, Mebius RE, Powrie F, Vivier E. Innate lymphoid cells--a proposal for uniform nomenclature. Nat Rev Immunol. 2013;13:145–149. doi: 10.1038/nri3365. [DOI] [PubMed] [Google Scholar]
- 63.Taylor PR, Leal SM, Jr, Sun Y, Pearlman E. Aspergillus and Fusarium corneal infections are regulated by Th17 cells and IL-17-producing neutrophils. J Immunol. 2014;192:3319–3327. doi: 10.4049/jimmunol.1302235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Werner JL, Gessner MA, Lilly LM, Nelson MP, Metz AE, Horn D, Dunaway CW, Deshane J, Chaplin DD, Weaver CT, Brown GD, Steele C. Neutrophils produce interleukin 17A (IL-17A) in a dectin-1- and IL-23-dependent manner during invasive fungal infection. Infect Immun. 2011;79:3966–3977. doi: 10.1128/IAI.05493-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Conti H, Peterson A, Huppler A, Brane L, Hernández-Santos N, Whibley N, Garg A, Simpson-Abelson M, Gibson G, Mamo A, Osborne L, Bishu S, Ghilardi N, Siebenlist U, Watkins S, Artis D, McGeachy M, Gaffen S. Oral-resident ‘natural’ Th17 cells and γδ-T cells control opportunistic Candida albicans infections. J Exp Med. 2014;211:2075–2084. doi: 10.1084/jem.20130877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hernández-Santos N, Huppler AR, Peterson AC, Khader SA, KC M, Gaffen SL. Th17 cells confer long term adaptive immunity to oral mucosal Candida albicans infections. Mucosal Immunol. 2013;6:900–910. doi: 10.1038/mi.2012.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Suegara N, Siegel JE, Savage DC. Ecological determinants in microbial colonization of the murine gastrointestinal tract: adherence of Torulopsis pintolopesii to epithelial surfaces. Infect Immun. 1979;25:139–145. doi: 10.1128/iai.25.1.139-145.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hirota K, Duarte JH, Veldhoen M, Hornsby E, Li Y, Cua DJ, Ahlfors H, Wilhelm C, Tolaini M, Menzel U, Garefalaki A, Potocnik AJ, Stockinger B. Fate mapping of IL-17-producing T cells in inflammatory responses. Nature Immunol. 2011;12:255–263. doi: 10.1038/ni.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Gladiator A, Wangler N, Trautwein-Weidner K, Leibundgut-Landmann S. Cutting Edge: IL-17-Secreting Innate Lymphoid Cells Are Essential for Host Defense against Fungal Infection. J Immunol. 2013;190:521–525. doi: 10.4049/jimmunol.1202924. [DOI] [PubMed] [Google Scholar]
- 70.Pandiyan P, Conti H, Zheng L, Peterson A, Mathern D, Hernandez-Santos N, Edgerton M, Gaffen S, Lenardo M. CD4+CD25+Foxp3+ regulatory T cells promote Th17 cells in vitro and enhance host resistance in mouse Candida albicans Th17 infection model. Immunity. 2011;34:422–434. doi: 10.1016/j.immuni.2011.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Igyarto BZ, Haley K, Ortner D, Bobr A, Gerami-Nejad M, Edelson BT, Zurawski SM, Malissen B, Zurawski G, Berman J, Kaplan DH. Skin-resident murine dendritic cell subsets promote distinct and opposing antigen-specific T helper cell responses. Immunity. 2011;35:260–272. doi: 10.1016/j.immuni.2011.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Kagami S, Rizzo HL, Kurtz SE, Miller LS, Blauvelt A. IL-23 and IL-17A, but not IL-12 and IL-22, are required for optimal skin host defense against Candida albicans. J Immunol. 2010;185:5453–5462. doi: 10.4049/jimmunol.1001153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Milner J, Holland S. The cup runneth over: lessons from the ever-expanding pool of primary immunodeficiency diseases. Nat Rev Immunol. 2013;13:635–648. doi: 10.1038/nri3493. [DOI] [PubMed] [Google Scholar]
- 74.Puel A, Picard C, Cypowyj S, Lilic D, Abel L, Casanova JL. Inborn errors of mucocutaneous immunity to Candida albicans in humans: a role for IL-17 cytokines? Curr Opin Immunol. 2010;22:467–474. doi: 10.1016/j.coi.2010.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Bishu S, Hernandez-Santos N, Simpson-Abelson M, Huppler AR, Conti HR, Ghilardi N, Mamo A, Gaffen SL. CARD9 is required for adaptive but not innate immunity to oral mucosal Candida albicans infections. Infect Immun. 2014;82:1173–1180. doi: 10.1128/IAI.01335-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Moyes DL, Runglall M, Murciano C, Shen C, Nayar D, Thavaraj S, Kohli A, Islam A, Mora-Montes H, Challacombe SJ, Naglik JR. A biphasic innate immune MAPK response discriminates between the yeast and hyphal forms of Candida albicans in epithelial cells. Cell Host Microbe. 2010;8:225–235. doi: 10.1016/j.chom.2010.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Toy D, Kugler D, Wolfson M, Vanden Bos T, Gurgel J, Derry J, Tocker J, Peschon JJ. Cutting Edge: Interleukin-17 signals through a heteromeric receptor complex. J Immunol. 2006;177:36–39. doi: 10.4049/jimmunol.177.1.36. [DOI] [PubMed] [Google Scholar]
- 78.Chang SH, Dong C. A novel heterodimeric cytokine consisting of IL-17 and IL-17F regulates inflammatory responses. Cell Res. 2007;17:435–440. doi: 10.1038/cr.2007.35. [DOI] [PubMed] [Google Scholar]
- 79.Wright JF, Guo Y, Quazi A, Luxenberg DP, Bennett F, Ross JF, Qiu Y, Whitters MJ, Tomkinson KN, Dunussi-Joannopoulos K, Carreno BM, Collins M, Wolfman NM. Identification of an Interleukin 17F/17A Heterodimer in Activated Human CD4+ T Cells. J Biol Chem. 2007;282:13447–13455. doi: 10.1074/jbc.M700499200. [DOI] [PubMed] [Google Scholar]
- 80.Ishigame H, Kakuta S, Nagai T, Kadoki M, Nambu A, Komiyama Y, Fujikado N, Tanahashi Y, Akitsu A, Kotaki H, Sudo K, Nakae S, Sasakawa C, Iwakura Y. Differential roles of interleukin-17A and −17F in host defense against mucoepithelial bacterial infection and allergic responses. Immunity. 2009;30:108–119. doi: 10.1016/j.immuni.2008.11.009. [DOI] [PubMed] [Google Scholar]
- 81.Taylor PR, Roy S, Leal SM, Jr, Sun Y, Howell SJ, Cobb BA, Li X, Pearlman E. Activation of neutrophils by autocrine IL-17A–IL-17RC interactions during fungal infection is regulated by IL-6, IL-23, RORgammat and dectin-2. Nature Immunol. 2014;15:143–151. doi: 10.1038/ni.2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Shen F, Hu Z, Goswami J, Gaffen SL. Identification of common transcriptional regulatory elements in interleukin-17 target genes. J Biol Chem. 2006;281:24138–24148. doi: 10.1074/jbc.M604597200. [DOI] [PubMed] [Google Scholar]
- 83.Yang D, Chertov O, Bykovskaia SN, Chen Q, Buffo MJ, Shogan J, Anderson M, Schröder JM, Wang JM, Howard OMZ, Oppenheim JJ. b-Defensins: Linking innate immunity and adaptive immunity through dendritic and T cell CCR6. Science. 1999;286:525–528. doi: 10.1126/science.286.5439.525. [DOI] [PubMed] [Google Scholar]
- 84.Tomalka J, Azodi E, Narra HP, Patel K, O’Neill S, Cardwell C, Hall BA, Wilson JM, Hise AG. beta-Defensin 1 plays a role in acute mucosal defense against Candida albicans. J Immunol. 2015;194:1788–1795. doi: 10.4049/jimmunol.1203239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Puri S, Edgerton M. How does it kill?: understanding the candidacidal mechanism of salivary histatin 5. Eukaryot Cell. 2014;13:958–964. doi: 10.1128/EC.00095-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Conti H, Baker O, Freeman A, Jang W, Li R, Holland S, Edgerton M, Gaffen S. New mechanism of oral immunity to mucosal candidiasis in hyper-IgE syndrome. Mucosal Immunol. 2011;4:448–455. doi: 10.1038/mi.2011.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Iwakura Y, Ishigame H, Saijo S, Nakae S. Functional specialization of interleukin-17 family members. Immunity. 2011;34:149–162. doi: 10.1016/j.immuni.2011.02.012. [DOI] [PubMed] [Google Scholar]
- 88.Trautwein-Weidner K, Gladiator A, Nur S, Diethelm P, LeibundGut-Landmann S. IL-17-mediated antifungal defense in the oral mucosa is independent of neutrophils. Mucosal Immunol. 2015;8:221–231. doi: 10.1038/mi.2014.57. [DOI] [PubMed] [Google Scholar]
- 89.Golden JB, McCormick TS, Ward NL. IL-17 in psoriasis: implications for therapy and cardiovascular co-morbidities. Cytokine. 2013;62:195–201. doi: 10.1016/j.cyto.2013.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ramirez-Carrozzi V, Sambandam A, Luis E, Lin Z, Jeet S, Lesch J, Hackney J, Kim J, Zhou M, Lai J, Modrusan Z, Sai T, Lee W, Xu M, Caplazi P, Diehl L, de Voss J, Balazs M, Gonzalez L, Jr, Singh H, Ouyang W, Pappu R. IL-17C regulates the innate immune function of epithelial cells in an autocrine manner. Nature Immunol. 2011;12:1159–1166. doi: 10.1038/ni.2156. [DOI] [PubMed] [Google Scholar]
- 91.Song X, Zhu S, Shi P, Liu Y, Shi Y, Levin SD, Qian Y. IL-17RE is the functional receptor for IL-17C and mediates mucosal immunity to infection with intestinal pathogens. Nature Immunol. 2011;12:1151–1158. doi: 10.1038/ni.2155. [DOI] [PubMed] [Google Scholar]
- 92.Yamaguchi Y, Fujio K, Shoda H, Okamoto A, Tsuno NH, Takahashi K, Yamamoto K. IL-17B and IL-17C are associated with TNF-alpha production and contribute to the exacerbation of inflammatory arthritis. J Immunol. 2007;179:7128–7136. doi: 10.4049/jimmunol.179.10.7128. [DOI] [PubMed] [Google Scholar]
- 93.Li H, Chen J, Huang A, Stinson J, Heldens S, Foster J, Dowd P, Gurney AL, Wood WI. Cloning and characterization of IL-17B and IL-17C, two new members of the IL-17 cytokine family. Proc Natl Acad Sci U S A. 2000;97:773–778. doi: 10.1073/pnas.97.2.773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Conti HR, Whibley N, Coleman B, Garg A, Jaycox J, Gaffen S. Signaling through IL-17C/IL-17RE is dispensable for immunity to systemic, oral and dermal candidiasis. PLoS One. 2015;10:e0122807. doi: 10.1371/journal.pone.0122807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.De Luca A, Zelante T, D’Angelo C, Zagarella S, Fallarino F, Spreca A, Iannitti RG, Bonifazi P, Renauld JC, Bistoni F, Puccetti P, Romani L. IL-22 defines a novel immune pathway of antifungal resistance. Mucosal Immunol. 2010;3:361–373. doi: 10.1038/mi.2010.22. [DOI] [PubMed] [Google Scholar]
- 96.Dollive S, Chen YY, Grunberg S, Bittinger K, Hoffmann C, Vandivier L, Cuff C, Lewis JD, Wu GD, Bushman FD. Fungi of the murine gut: episodic variation and proliferation during antibiotic treatment. PLoS One. 2013;8:e71806. doi: 10.1371/journal.pone.0071806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Zhernakova A, Festen EM, Franke L, Trynka G, van Diemen C, Monsuur AJ, Bevova M, Nijmeijer RM, van ‘t Slot R, Heijmans R, Boezen HM, van Heel DA, van Bodegraven AA, Stokkers PC, Wijmenga C, Crusius JB, Weersma RK. Genetic analysis of innate immunity in Crohn’s disease and ulcerative colitis identifies two susceptibility loci harboring CARD9 and IL18RAP. Am J Hum Genet. 2008;82:1202–1210. doi: 10.1016/j.ajhg.2008.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Marijnissen RJ, Koenders MI, van de Veerdonk FL, Dulos J, Netea MG, Boots AM, Joosten LA, van den Berg WB. Exposure to Candida albicans polarizes a T-cell driven arthritis model towards Th17 responses, resulting in a more destructive arthritis. PLoS One. 2012;7:e38889. doi: 10.1371/journal.pone.0038889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ling Y, Cypowyj S, Aytekin C, Galicchio M, Camcioglu Y, Nepesov S, Ikinciogullari A, Dogu F, Belkadi A, Levy R, Migaud M, Boisson B, Bolze A, Itan Y, Goudin N, Cottineau J, Picard C, Abel L, Bustamante J, Casanova JL, Puel A. Inherited IL-17RC deficiency in patients with chronic mucocutaneous candidiasis. J Exp Med. 2015;212:619–631. doi: 10.1084/jem.20141065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ferwerda B, Ferwerda G, Plantinga TS, Willment JA, van Spriel AB, Venselaar H, Elbers CC, Johnson MD, Cambi A, Huysamen C, Jacobs L, Jansen T, Verheijen K, Masthoff L, Morre SA, Vriend G, Williams DL, Perfect JR, Joosten LA, Wijmenga C, van der Meer JW, Adema GJ, Kullberg BJ, Brown GD, Netea MG. Human dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med. 2009;361:1760–1767. doi: 10.1056/NEJMoa0901053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Plantinga TS, van der Velden WJ, Ferwerda B, van Spriel AB, Adema G, Feuth T, Donnelly JP, Brown GD, Kullberg BJ, Blijlevens NM, Netea MG. Early stop polymorphism in human DECTIN-1 is associated with increased candida colonization in hematopoietic stem cell transplant recipients. Clin Infect Dis. 2009;49:724–732. doi: 10.1086/604714. [DOI] [PubMed] [Google Scholar]
- 102.Drummond RA, Brown GD. The role of Dectin-1 in the host defence against fungal infections. Curr Opin Microbiol. 2011;14:392–399. doi: 10.1016/j.mib.2011.07.001. [DOI] [PubMed] [Google Scholar]
- 103.Marakalala MJ, Vautier S, Potrykus J, Walker LA, Shepardson KM, Hopke A, Mora-Montes HM, Kerrigan A, Netea MG, Murray GI, Maccallum DM, Wheeler R, Munro CA, Gow NA, Cramer RA, Brown AJ, Brown GD. Differential adaptation of Candida albicans in vivo modulates immune recognition by dectin-1. PLoS Pathog. 2013;9:e1003315. doi: 10.1371/journal.ppat.1003315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Plato A, Willment JA, Brown GD. C-type lectin-like receptors of the dectin-1 cluster: ligands and signaling pathways. Intl Rev Immunol. 2013;32:134–156. doi: 10.3109/08830185.2013.777065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gross O, Gewies A, Finger K, Schafer M, Sparwasser T, Peschel C, Forster I, Ruland J. Card9 controls a non-TLR signalling pathway for innate anti-fungal immunity. Nature. 2006;442:651–656. doi: 10.1038/nature04926. [DOI] [PubMed] [Google Scholar]
- 106.Quintin J, Saeed S, Martens JH, Giamarellos-Bourboulis EJ, Ifrim DC, Logie C, Jacobs L, Jansen T, Kullberg BJ, Wijmenga C, Joosten LA, Xavier RJ, van der Meer JW, Stunnenberg HG, Netea MG. Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe. 2012;12:223–232. doi: 10.1016/j.chom.2012.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Glocker EO, Hennigs A, Nabavi M, Schaffer AA, Woellner C, Salzer U, Pfeifer D, Veelken H, Warnatz K, Tahami F, Jamal S, Manguiat A, Rezaei N, Amirzargar AA, Plebani A, Hannesschlager N, Gross O, Ruland J, Grimbacher B. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N Engl J Med. 2009;361:1727–1735. doi: 10.1056/NEJMoa0810719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Holland SM, DeLeo FR, Elloumi HZ, Hsu AP, Uzel G, Brodsky N, Freeman AF, Demidowich A, Davis J, Turner ML, Anderson VL, Darnell DN, Welch PA, Kuhns DB, Frucht DM, Malech HL, Gallin JI, Kobayashi SD, Whitney AR, Voyich JM, Musser JM, Woellner C, Schaffer AA, Puck JM, Grimbacher B. STAT3 mutations in the hyper-IgE syndrome. N Engl J Med. 2007;357:1608–1619. doi: 10.1056/NEJMoa073687. [DOI] [PubMed] [Google Scholar]
- 109.Minegishi Y, Saito M, Tsuchiya S, Tsuge I, Takada H, Hara T, Kawamura N, Ariga T, Pasic S, Stojkovic O, Metin A, Karasuyama H. Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyper-IgE syndrome. Nature. 2007;448:1058–1062. doi: 10.1038/nature06096. [DOI] [PubMed] [Google Scholar]
- 110.Marks BR, Nowyhed HN, Choi JY, Poholek AC, Odegard JM, Flavell RA, Craft J. Thymic self-reactivity selects natural interleukin 17-producing T cells that can regulate peripheral inflammation. Nature Immunol. 2009;10:1125–1132. doi: 10.1038/ni.1783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Liu L, Okada S, Kong XF, Kreins AY, Cypowyj S, Abhyankar A, Toubiana J, Itan Y, Audry M, Nitschke P, Masson C, Toth B, Flatot J, Migaud M, Chrabieh M, Kochetkov T, Bolze A, Borghesi A, Toulon A, Hiller J, Eyerich S, Eyerich K, Gulacsy V, Chernyshova L, Chernyshov V, Bondarenko R, Maria Cortes Grimaldo R, Blancas-Galicia L, Madrigal Beas IM, Roesler J, Magdorf K, Engelhard D, Thumerelle C, Burgel PR, Hoernes M, Drexel B, Seger R, Kusuma T, Jansson AF, Sawalle-Belohradsky J, Belohradsky B, Jouanguy E, Bustamante J, Bue M, Karin N, Wildbaum G, Bodemer C, Lortholary O, Fischer A, Blanche S, Al-Muhsen S, Reichenbach J, Kobayashi M, Rosales FE, Lozano CT, Kilic SS, Oleastro M, Etzioni A, Traidl-Hoffmann C, Renner ED, Abel L, Picard C, Marodi L, Boisson-Dupuis S, Puel A, Casanova JL. Gain-of-function human STAT1 mutations impair IL-17 immunity and underlie chronic mucocutaneous candidiasis. J Exp Med. 2011;208:1635–1648. doi: 10.1084/jem.20110958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.van de Veerdonk FL, Plantinga TS, Hoischen A, Smeekens SP, Joosten LA, Gilissen C, Arts P, Rosentul DC, Carmichael AJ, Smits-van der Graaf CA, Kullberg BJ, van der Meer JW, Lilic D, Veltman JA, Netea MG. STAT1 mutations in autosomal dominant chronic mucocutaneous candidiasis. N Engl J Med. 2011;365:54–61. doi: 10.1056/NEJMoa1100102. [DOI] [PubMed] [Google Scholar]
- 113.Hori T, Ohnishi H, Teramoto T, Tsubouchi K, Naiki T, Hirose Y, Ohara O, Seishima M, Kaneko H, Fukao T, Kondo N. Autosomal-Dominant Chronic Mucocutaneous Candidiasis with STAT1-Mutation can be Complicated with Chronic Active Hepatitis and Hypothyroidism. J Clin Immunol. 2012;32:1213–1220. doi: 10.1007/s10875-012-9744-6. [DOI] [PubMed] [Google Scholar]
- 114.Smeekens SP, Ng A, Kumar V, Johnson MD, Plantinga TS, van Diemen C, Arts P, Verwiel ET, Gresnigt MS, Fransen K, van Sommeren S, Oosting M, Cheng SC, Joosten LA, Hoischen A, Kullberg BJ, Scott WK, Perfect JR, van der Meer JW, Wijmenga C, Netea MG, Xavier RJ. Functional genomics identifies type I interferon pathway as central for host defense against Candida albicans. Nat Commun. 2013;4:1342. doi: 10.1038/ncomms2343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Puel A, Cypowji S, Bustamante J, Wright J, Liu L, Lim H, Migaud M, Israel L, Chrabieh M, Audry M, Gumbleton M, Toulon A, Bodemer C, El-Baghdadi J, Whitters M, Paradis T, Brooks J, Collins M, Wolfman N, Al-Muhsen S, Galicchio M, Abel L, Picard C, Casanova J-L. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science. 2011;332:65–68. doi: 10.1126/science.1200439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Eyerich K, Foerster S, Rombold S, Seidl HP, Behrendt H, Hofmann H, Ring J, Traidl-Hoffmann C. Patients with chronic mucocutaneous candidiasis exhibit reduced production of Th17-associated cytokines IL-17 and IL-22. J Invest Derm. 2008;128:2640–2645. doi: 10.1038/jid.2008.139. [DOI] [PubMed] [Google Scholar]
- 117.Boisson B, Wang C, Pedergnana V, Wu L, Cypowyj S, Rybojad M, Belkadi A, Picard C, Abel L, Fieschi C, Puel A, Li X, Casanova J-L. A biallelic ACT1 mutation selectively abolishes interleukin-17 responses in humans with chronic mucocutaneous candidiasis. Immunity. 2013;39:676–686. doi: 10.1016/j.immuni.2013.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Kisand K, Boe Wolff AS, Podkrajsek KT, Tserel L, Link M, Kisand KV, Ersvaer E, Perheentupa J, Erichsen MM, Bratanic N, Meloni A, Cetani F, Perniola R, Ergun-Longmire B, Maclaren N, Krohn KJ, Pura M, Schalke B, Strobel P, Leite MI, Battelino T, Husebye ES, Peterson P, Willcox N, Meager A. Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines. J Exp Med. 2010;207:299–308. doi: 10.1084/jem.20091669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Puel A, Doffinger R, Natividad A, Chrabieh M, Barcenas-Morales G, Picard C, Cobat A, Ouachee-Chardin M, Toulon A, Bustamante J, Al-Muhsen S, Al-Owain M, Arkwright PD, Costigan C, McConnell V, Cant AJ, Abinun M, Polak M, Bougneres PF, Kumararatne D, Marodi L, Nahum A, Roifman C, Blanche S, Fischer A, Bodemer C, Abel L, Lilic D, Casanova JL. 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. doi: 10.1084/jem.20091983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Ha HL, Wang H, Pisitkun P, Kim JC, Tassi I, Tang W, Morasso MI, Udey MC, Siebenlist U. IL-17 drives psoriatic inflammation via distinct, target cell-specific mechanisms. Proc Natl Acad Sci U S A. 2014;111:E3422–E3431. doi: 10.1073/pnas.1400513111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CE, Papp K, Puig L, Nakagawa H, Spelman L, Sigurgeirsson B, Rivas E, Tsai TF, Wasel N, Tyring S, Salko T, Hampele I, Notter M, Karpov A, Helou S, Papavassilis C, Group ES, Group FS. Secukinumab in plaque psoriasis--results of two phase 3 trials. N Engl J Med. 2014;371:326–338. doi: 10.1056/NEJMoa1314258. [DOI] [PubMed] [Google Scholar]
- 122.Miossec P, Kolls JK. Targeting IL-17 and TH17 cells in chronic inflammation. Nat Rev Drug Discov. 2012;11:763–776. doi: 10.1038/nrd3794. [DOI] [PubMed] [Google Scholar]
- 123.Kopp T, Riedl E, Bangert C, Bowman EP, Greisenegger E, Horowitz A, Kittler H, Blumenschein WM, McClanahan TK, Marbury T, Zachariae C, Xu D, Hou XS, Mehta A, Zandvliet AS, Montgomery D, van Aarle F, Khalilieh S. Clinical improvement in psoriasis with specific targeting of interleukin-23. Nature. 2015 doi: 10.1038/nature14175. [DOI] [PubMed] [Google Scholar]
- 124.Ford AC, Peyrin-Biroulet L. Opportunistic Infections With Anti-Tumor Necrosis Factor-alpha Therapy in Inflammatory Bowel Disease: Meta-Analysis of Randomized Controlled Trials. Am J Gastroenterol. 2013;108:1268–1276. doi: 10.1038/ajg.2013.138. [DOI] [PubMed] [Google Scholar]
- 125.Bishu S, Su E, Wilkerson E, Reckley K, Jones D, McGeachy MJ, Gaffen SL, M L. RA patients exhibit impaired Candida albicans-specific Th17 responses but preserved protective oral immunity. Arth Res Ther. 2014;16:R50. doi: 10.1186/ar4480. [DOI] [PMC free article] [PubMed] [Google Scholar]