Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Nov 30.
Published in final edited form as: J Allergy Clin Immunol Pract. 2023 Apr 26;11(6):1624–1634. doi: 10.1016/j.jaip.2023.04.015

The Th17/IL-17 axis and host defence against fungal infections

Stuart G Tangye 1,2, Anne Puel 3,4,5
PMCID: PMC12664666  NIHMSID: NIHMS2111043  PMID: 37116791

Abstract

Chronic mucocutaneous candidiasis (CMC) was recognized as a primary immunodeficiency in the early 1970’s. However, for almost 40 years, its genetic etiology remained unknown. The progressive molecular and cellular description of inborn errors of immunity (IEI) with syndromic CMC pointed toward a possible role of IL-17-mediated immunity in protecting against fungal infection and CMC. Since 2011, novel IEI affecting either the response to or production of IL-17A and IL-17F (IL-17A/F) in patients with isolated or syndromic CMC provided formal proof of the pivotal role of the IL-17 axis in mucocutaneous immunity to Candida spp. And, to a lesser extent, to Staphylococcus aureus in humans. In contrast, IL-17-mediated immunity seems largely redundant against other common microbes in humans. In this review, we outline the current knowledge of IEI associated with impaired IL-17A/F-mediated immunity, highlighting our current understanding of the role of IL-17A/F in human immunity.

Keywords: Th17 cells, IL-17 cytokines, chronic mucocutaneous candidiasis, inborn errors of immunity, anti-fungal immunity, anti-cytokine autoantibodies, STATs

INTRODUCTION

Cytokines are proteins produced by many cell types and have critical roles in immunity. Specifically, cytokines regulate the development, activation, and differentiation of leukocytes and non-haemopoietic cells. These processes underlie protection against infectious diseases following natural pathogen infection or immunization, thereby providing the host with long-lived immunological memory. In contrast, aberrant cytokine signaling can cause immune dysregulation, eg early-onset autoimmunity. Thus, balanced signals provided by distinct cytokines, and delivered to specific cell subsets, are critical for immune homeostasis14.

Approximately 60 different cytokines have been identified, including interleukins (IL-1 through IL-40), interferons, transforming growth factors and members of the TNF superfamily1. Most cytokines have pleiotropic effects on different immune cells; there is also substantial overlap in their function. The biological effects of many cytokines are mediated by JAK/STAT signalling pathways2, 57. Four JAKs (JAK1, JAK2, JAK3, Tyk2) and seven STATs (STAT1, 2, 3, 4, 5a, 5b, 6) have been identified in mammalian genomes2, 5, 6. JAKs associate with the cytoplasmic domains of cytokine receptors and, following engagement by specific ligands, phosphorylate tyrosine residues to provide docking sites for STATs. Receptor-associated STATs undergo JAK-mediated phosphorylation, resulting in formation of multimers that translocate to the nucleus and bind specific DNA sequences, thereby regulating expression of target genes2, 57.

The essential and non-redundant functions of specific cytokines in host defense and immune dysregulation in humans have been elegantly and repeatedly revealed by the discovery of individuals with pathogenic variants in genes encoding cytokines, cytokines receptors, or downstream transcription factors that manifest as immunodeficient and/or autoimmune states4, 8, 9. Currently, variants, either loss-of function (LOF) or gain-of function (GOF), have been identified in genes encoding cytokines (IL10, IL17F, IL12B, IL21), cytokine receptors (IL2RA, IL2RB, IL2RG, IL7RA, IL6R, IL6ST, IL10RA, IL10RB, IL11RA, IL12RB1, IL12RB2, IL17RA, IL7RC, IL21R, IL23R, IFNGR1, IFNGR2, IFNAR1, IFNAR2), JAKs (JAK1, JAK3, TYK2), transcription factors activated by specific cytokines (STAT1, STAT2, STAT3, STAT5B, STAT6), and transcription factors that regulate lymphocyte fate (FOXP3, RORC, ZNF341, IRF1, IRF4), resulting in aberrant development and/or effector function of different human immune cells4, 811. These discoveries have not only defined genetic causes of unique inborn errors of immunity (IEI) but also established key roles for cytokine signalling and cell types in host defense against specific pathogens. Remarkably, infectious complications that define some IEI can also result from production of neutralising anti-cytokine autoantibodies which impede the function of specific cytokines12. Here, we provide an overview of how the study of IEI has contributed to our understanding of the molecular and cellular requirements for the generation and function of human IL-17-producing cells in health and disease, and how these findings reveal mechanisms of disease pathogenesis that may enable improved management and treatment of individuals with these conditions.

IL-17A and IL-17F production, regulation, and signaling.

Interleukin (IL)-17A, also named IL-17, belongs to the IL-17 family of cytokines, which includes IL-17B, IL-17C, IL-17D, IL-17E (also called IL-25) and IL-17F13. The founding and best characterized member - IL-17A - was cloned in 1993 and initially named cytotoxic T lymphocyte-associated antigen 8 (CTLA-8)14, 15. IL-17F is the most closely related member, sharing 55% similarity with IL-17A. Both IL-17A and IL-17F are usually produced by the same cell types16, and function as homodimers (IL-17A/IL-17A, IL-17F/IL-17F) or heterodimers (IL-17A/IL-17F)13, 1719. However, IL-17A homodimers more potently induce production of pro-inflammatory cytokines and chemokines than IL-17F homodimers by target cells expressing IL-17 receptors, such as fibroblasts, macrophages, or epithelial cells. IL-17A/IL-17F heterodimers have intermediate signaling strength1719.

CD4+ T cell differentiation

CD4+ T cells play critical roles in mediating immunity against a broad array of pathogens that cause serious infectious diseases. This reflects the extraordinary ability of CD4+ T cells to differentiate into distinct effector subsets that co-operate with other immune cells to protect the host against infection with specific pathogens. Naïve CD4+ T cells can differentiate into distinct subsets of cells that have unique roles in protection against infectious diseases3, 20. For instance, distinct and specialised CD4+ T cell subsets have been implicated in immunity against viral or intramacrophagocytic (Th1), parasitic (Th2) and fungal (Th17) infections, as well as regulating B-cell differentiation/humoral immunity (Tfh) and immune regulation/homeostasis (Tregs)3, 20. CD4+ T cell differentiation is mediated by the microenvironment in which CD4+ T cells encounter various signals provided by antigen-presenting cells (APCs). These take the form of MHC class II/peptide complexes and co-stimulatory signals provided by cell-cell interactions and cytokines which activate relevant signalling pathways and induce lineage-specific transcription factors3, 20. These pathways not only induce commitment of naïve CD4+ T cells to a specific fate, but also restrain differentiation of naïve CD4+ T cells to alternate effector fates. Thus, the molecular wiring of CD4+ T cells allows these cells to acquire and retain specialised functions to establish and maintain effective protection against pathogens3, 20.

Th17 and IL-17-producing cells

In 2005, a CD4+ T helper (Th) subset distinct from Th1 and Th2 cells was described in mice as the main source of IL-17A and IL-17F and was therefore designated Th172124. Since then, IL-17A and IL-17F have been found to be produced by additional immune cells in humans and mice, including cytotoxic CD8+ T (Tc)17 cells2527, γδ T cells2830, type 3 innate lymphoid cells (ILC)33135, invariant natural killer T (iNKT)3638, mucosal-associated invariant T (MAIT) cells3941, and Th1* cells42. Here, we will predominantly focus on IL-17-producing CD4+ Th17 cells.

Th17 cells have been well-characterised based on expression of surface receptors (CCR4, CCR6, CD161, IL-23R) and Th17-lineage specific transcription factors RORγt and RORα, which regulate production of canonical cytokines including IL-17A, IL-17F, IL-21 and IL-22 (and IL-26 in humans) and CCL203, 20 (Figure 1). Due to the production of pro-inflammatory cytokines, Th17 cells have been implicated in numerous autoimmune conditions, such as inflammatory bowel disease, psoriasis, and ankylosing spondylitis43. However, before IL-17 was identified as a canonical Th17 cytokine, studies in mice demonstrated a key role for IL-17A/IL-17AR signaling in host immunity against fungal infections44. This was extended to reveal that Th17 cells were a predominant source of the IL-17 required for mediating anti-fungal immunity in mice45. Seminal studies in humans also found that memory CD4+ T cells co-expressing CCR6 and CCR4 were enriched for (1) production of IL-17 and expression of RORC (encoding the transcription factor RORγt), and (2) Candida-specific CD4+ T cells46. These studies established a fundamental role for IL-17-producing Th17 cells in host defense against mucosal fungal infections in mice and humans. Mechanistically, IL-17 induced anti-fungal immunity by binding IL-17 receptors expressed on epithelial cells and inducing production of anti-microbial peptides and β-defensins by these cells47 (Figure 1). Consistent with these observations, individuals with IEI that compromise IL17-mediated immunity develop chronic mucocutaneous candidiasis (CMC). Analysis of these conditions have provided invaluable insights into the pathways that regulate Th17 cell differentiation and function48.

Figure 1: Inborn errors of immunity and phenocopies that disrupt IL-17-mediated immunity against fungal infections.

Figure 1:

In response to cytokines produced by myeloid cells, naïve CD4+ T cells differentiate into specialised subsets of effectors cells. Production of canonical cytokines enables elicitation of specific effector functions important for host defence against pathogen infections. In the setting of Th17 cells, integration of STAT3/ZNF341-dependent signals downstream of cytokines including IL-23 and IL-21 in CD4+ T cells leads to expression of the transcription factor RORγt and induction of the Th17 program. IL-17 cytokines activate epithelial cells via IL-17R/ACT1/JNK1, resulting in production of pro-inflammatory and anti-microbial peptides. Molecules shown in red have been found to be mutated in specific IEI and impair either the generation or function of Th17 cells causing CMC (and often Staph infection). Autoantibodies against IL-17A/IL-17F phenocopy inborn errors of IL-17-mediated immunity.

Molecular requirements for generating human Th17 cells

Transcription Factors:

Naïve CD4+ T cells give rise to specialised subsets following receipt of instructive signals provided by the cellular microenvironment established during T cell activation. Numerous studies, performed predominantly in mice, identified various cytokines and transcription factors required for in vivo and in vitro Th17 cell differentiation and maintenance, including the cytokines TGF-β, IL-1, IL-6, IL-21, IL-2323, 42, 4957, and transcription factors STAT35860, RORγt61, and RORα62. However, several of these findings have not been confirmed by other investigators63, 64. IL-6, IL-21, and IL-23 activate STAT3, providing the basis for the initial discovery in mice that STAT3 plays a critical role in generating murine Th17 cells5860. Autosomal dominant hyper IgE syndrome (AD-HIES) is caused by heterozygous germline dominant negative STAT3 (STAT3DN) variants and is characterised by recurrent opportunistic bacterial and fungal infections6567. Indeed, >85% of affected individuals suffer from CMC, and >90% from pneumonia caused by Staphylococcus aureus or Streptococcus pneumoniae67 (Table 1). Analysis of STAT3-deficient patients revealed a lack of Th17 cells ex vivo6872, as determined by significant decreases in CCR6+ CD4+ T cells, and impaired production of IL-17A, IL-17F, and IL-22 by memory CD4+ T cells under neutral and Th17-inducing culture conditions71. This defect was cell intrinsic, because naïve STAT3DN CD4+ T cells were unable to upregulate expression of RORC and differentiate into IL17A/IL-17F-secreting cells in vitro following culture under Th17-polarising conditions68, 73 (Table 1, Figure 1). Interestingly, STAT3DN patients had numerical reductions in MAIT cells, and the residual MAIT cells present were impaired in production of Th17 cytokines41. As these innate-type T cells recognise fungal antigens, it is possible that a deficiency in these cells also contributes to compromised anti-fungal immunity in AD HIES.

Table 1:

Inborn errors of immunity and phenocopies resulting in compromised IL-17-mediated immunity and fungal infections

Genetic defect IL-17A/F signaling IL-17A/F production; CMC Staph disease Other infectious diseases Auto-immunity Refs
AD IL-17F deficiency Impaired IL-17A: normal; IL-17F: NT + − (+) 107, 108
AR IL-17RC deficiency Abolished Normal + − (+) 111
AR IL-17RA deficiency Abolished Normal to increased + + − (+)a 107, 109, 110
AR ACT1 deficiency Abolished Normal to increased + + − (+)b 112117
AD JNK1 deficiency Impaired Low + + +/− c 119
AR IL-12p40 deficiency NT (presumed normal) Low ex vivo +/−d mycobacteriosis and Salmonellosis 127
AR IL-12Rβ1 deficiency NT (presumed normal) Low ex vivo and after in vitro differentiation +/−e mycobacteriosis and Salmonellosis 7173, 86
AR IL-12RB2 deficiency NT (presumed normal) Normal mycobacteriosis, pulmonary tuberculosis 87
AR IL-23R deficiency NT (presumed normal) Normal ex vivo, low after in vitro differentiation mycobacteriosis 87, 88
AR IL-6R deficiency NT (presumed normal) subnormal ex vivo, low after in vitro differentiation + +
Bacterial diseases
128, 129
AR GP130 deficiency* NT (presumed normal) Low to normal ex vivo f + +
Bacterial diseases
130, 131
AD GP130 deficiency NT (presumed normal) Normal ex vivo, low after in vitro differentiation g + +
Bacterial diseases and secondary aspergillosis), h
94
AR IL-21R deficiency NT (presumed normal) Impaired − (+)i +
Bacterial, parasitic, fungal
9093
AR RORγ deficiency NT (presumed normal) + + BCGosis 74
AD STAT3 deficiency NT + + Bacterial diseases 66, 6870, 72
AR ZNF341 deficiency NT Impaired + + Bacterial disease, Pulmonary disease 76
AD JNK1 deficiency Impaired Low to normal + + Bacterial diseases 119
AR DOCK8 deficiency NT (presumed normal) Impaired ++ + Viral (Molluscum, HSV), bacterial 7779
AD STAT1 GOF NT Low ++ ++ Bacterial, viral, fungal + 71, 73, 80, 81
AR CARD9 deficiency NT Impaired to normal + Invasive fungal diseases 101, 102
Anti IL-17 AutoAb + + 12, 124, 125

NT: not tested

a

2 cases of suspected pulmonary tuberculosis or tuberculous meningitis, respectively;

b

discoid lupus erythematosus reported in one consanguineous multiplex family;

c

urinary tract infections (pyelonephritis and cystitis);

d

~6% of patients with AR IL-12p40 deficiency display CMC

e

about 25% of patients with AR IL-12Rβ1 deficiency display CMC;

f

one patient had aphthous tongue ulcer suggestive of an undefined fungal lesion;

g

one patient reported with onychomycosis;

h

one patient reported with five episodes of shingles

i

one patient reported with esophageal candidiasis

Individuals have also been identified with recessive variants in RORC74. Perhaps not surprisingly, most (>80%) patients with RORγt deficiency develop CMC (Table 1). LOF RORC mutations abolished the generation of Th17 and MAIT cells in vivo and in vitro74 (Table 1), consistent with findings from STAT3DN patients whose CD4+ T cells failed to upregulate RORC to generate Th17 cells68, 73, and lacked MAIT cells41. Furthermore, while Candida specific CD4+ T cells could be generated in the absence of RORγt, these cells were completely devoid of production of Th17 cytokines74 (Figure 1). Interestingly, proportions of CCR6+ CD4+ memory T cells were intact in RORγt-deficient patients74, indicating that STAT3 signalling regulates CCR6 expression independently of RORγt.

An autosomal recessive (AR) form of HIES that clinically phenocopies STAT3DN HIES, including high penetrance of CMC, is caused by bi-allelic mutations in the transcription factor ZNF34175, 76 (Table 1). The mechanistic link between ZNF341 and STAT3 function was provided by the finding that ZNF341 binds the STAT3 promoter and is essential for STAT3 transcription-dependent autoinduction and sustained STAT3 activity. Consequently, ZNF341-deficient patients have low levels of STAT3 mRNA and protein and poor responses following stimulation with STAT3-activating cytokines75, 76. ZNF341-deficient patients had a paucity of CD4+ CCR6+ memory T cells. The lack of Th17-phenotype cells was confirmed functionally by demonstrating reductions in proportions of memory CD4+ T cells expressing intracellular IL17A, IL17F, and IL2275, as well as abolished production of these canonical Th17 cytokines by purified memory CD4+ T cells in vitro under non-polarising conditions and by naïve and memory CD4+ T cells cultured under Th17-polarising conditions75, 76 (Table 1, Figure 1).

AR HIES can also be caused by biallelic mutations in DOCK8, encoding Dedicator of Cytokinesis 8, a guanine nucleotide exchange factor involved in regulating the activity of Rho family GTP enzymes77. DOCK8 deficiency causes a combined immunodeficiency, including severe susceptibility to bacterial, viral, and fungal infections.77 Whilst there are many cellular defects that underpin multiple infectious diseases, DOCK8-deficiency compromises the ability of naïve CD4+ T cells to differentiate into Th17-type cells78. Interestingly, DOCK8 has been reported to associate with STAT3, and STAT3 activation is impaired in DOCK8-deficient cells79. These findings suggest a causal link between STAT3-deficiency, DOCK8 deficiency and CMC.

Activating STAT1 GOF mutations cause immune dysregulation characterised predominantly by recurrent bacterial, viral and fungal infections (CMC), with the latter affecting almost all patients in the form of oropharyngeal, cutaneous, esophageal, genital, or onycho-mycoses80 (Table 1). CD4+ T cells from these patients exhibit reduced, but not abolished, Th17 cells71, 73, 80 (Table 1). Whilst the mechanism underlying defective Th17 generation due to STAT1 GOF remains incompletely determined, it is possible that stronger or sustained cellular response to the STAT1-dependent cytokines IFN-γ, IFN-α and IL-27 inhibits Th17 development80, 81 (Figure 1). However, as impaired Th17 differentiation is also observed for naïve STAT1 GOF CD4+ T cells cultured in vitro under Th17 conditions but in the absence IFN-γ, IFN-α and IL-2773, additional cell-intrinsic mechanisms that impair Th17 generation must exist (Figure 1). For instance, binding of STAT3 to specific target genes and induction of STAT3-target genes including RORC, IL17, IL22, and SOCS are impaired in immune cells from patients with STAT1 GOF mutations82. Similarly, studies in mice revealed that STAT1 and STAT3 can counter-regulate the function of each other83. For instance, deletion of Stat1 enhances STAT3 activation, while reduced IL-6-mediated activation of STAT3 augments STAT1 activation. These effects are regulated by differential induction of SOCS383.

Studies of individuals with IEI due to STAT3 DN, RORC LOF, ZNF341 LOF or STAT1 GOF variants have provided valuable insights into infectious susceptibility and a cellular and molecular explanation for their clinical features. Specifically, these findings revealed the essential intrinsic roles of STAT3, RORγt and ZNF341 in regulating differentiation of naïve CD4+ T cells into effector Th17 cells, and the opposing effect of hyper-active STAT1 on this pathway. The findings also provide a mechanism for extreme susceptibility to Candida albicans and the high incidence (~85%) of CMC in these patients (Table 1). Furthermore, determining the molecular etiology of disease due to STAT1 GOF variants enabled targeted therapies of these individuals with JAK inhibitors. Whilst STAT1 GOF causes a panoply of clinical presentations, CMC is a prominent feature. Treatment of affected individuals with JAK inhibitors including ruxolitinib or baracitinib resulted in significant and rapid (1–8 weeks) improvement in CMC in >90% of patients. Thus, JAK inhibition as a precision medicine is highly effective for treating severe CMC in the setting of STAT1 GOF84, 85.

Cytokine Receptors:

While the findings described above established critical roles for STAT3 signaling in generating and/or maintaining Th17 cells, they did not directly identify the upstream STAT3-activating cytokine(s) required for this process. However, this has now been achieved by the identification and analysis of patients with inactivating mutations in specific cytokine receptors that signal through STAT3, namely IL-12R, IL-23R, IL-6R and IL-21R.

Patients with recessive mutations in genes encoding receptors for IL12 (IL12RB1/IL12RB2) or IL-23 (IL12RB1/IL23R) predominantly suffer from mycobacterial disease8688. However, ~30% of individuals with pathogenic variants in IL12RB1 or IL23R, but not IL12RB2, also endure recurrent fungal infections8689 (Table 1). IL-12RB1- or IL23R-deficient memory CD4+ T cells exhibit significantly reduced production of IL-17A, IL-17F and IL-22 ex vivo and in vitro, yet production of these cytokines by IL12RB2-deficient cells was intact71, 87, 88 (Table 1, Figure 1). Similar findings were obtained when in vitro differentiation of naïve CD4+ T cells from these patients was examined73, 87, 88. These findings implicate IL-23 in generating Th17 cells in humans. However, the reduction in Th17 cells in IL-12RB1/IL23R-deficiency was not as severe as in STAT3-deficiency71, 73, 87, 88, and the penetrance of CMC was incomplete72, 87, 88, 90, supporting a role for additional cytokines in Th17 generation.

Interestingly, a similar proportion of patients with IL-21R-deficient patients also have problems controlling fungal infections, although not strictly CMC (~30%)9093 (Table 1). IL21R LOF mutations dramatically reduced, but did not abolish, the in vivo, ex vivo and in vitro generation of Th17 cells71, 73, 90 (Table 1). Heterozygous DN mutations in IL6ST, encoding the shared gp130 component of the IL-6 family of cytokines, result in a clinical condition similar to AD-HIES due to STAT3DN variants, with the exception of a very low incidence of CMC (1/9 patients, Table 1)94. Consistent with this, naïve CD4+ T cells bearing IL6ST variants produced similar levels of IL-17A and IL-17F as naïve CD4+ T cells from healthy donors following culture under in vitro Th17 polarizing conditions. IL6ST mutant memory CD4+ T cells also secreted intact amounts of IL-17A, even though the proportions of these cells expressing Th17 cytokines was modestly reduced94 (Table 1). Taken together, it appears that IL-23 (via IL-12Rβ1/IL23R) and IL-21 are the predominant STAT3-activating cytokines involved in generating Th17 cells in humans, thereby explaining the incidence of CMC in ~30% of individuals with these pathogenic gene mutations (Figure 1). These data also highlight functional redundancy and/or co-operativity between these cytokines in CD4+ T cell differentiation inasmuch that the complete absence of signalling via IL-21, IL-23 or IL-6 is insufficient to completely abolish induction of Th17 cells.

DECTIN1:

Dectin-1 (or C-type lectin domain family 7 member A, CLEC7A) is a C-type lectin receptor (CLR), mostly expressed by myeloid cells, which recognizes β-glucans present in the cell wall of certain pathogens including fungi and some bacteria95, 96. Upon β-glucan recognition, Dectin-1 recruits and activates the spleen tyrosine kinase SYK, engaging the Caspase recruitment domain-containing protein 9 (CARD9) protein to promote production of pro-inflammatory cytokines (IL-6, IL-23) by dendritic cells, that induce differentiation of CD4+ T cells into IL-17-producing T cells95, 97. In 2009, three adult siblings with onychomycosis caused by Trichophyton rubrum (dermatophytosis) and vulvovaginitis caused by C. albicans were reported to be homozygous for a deleterious allele of DECTIN1 (p.Y238*)98. The heterozygous parents had a milder, but similar clinical phenotype. The authors showed impaired induction of IL-17-producing T cells in some experimental conditions, including upon stimulation with C. albicans. However, the p.Y238X DECTIN1 allele is a common polymorphism, with a frequency of ~7% in European populations and of up to 40% in the San population of South Africa98 (http://hapmap.ncbi.nlm.nih.gov.proxy.insermbiblio.inist.fr/). Considering the frequency of the p.Y238* DECTIN1 allele and that of CMC (~10−5), it remains difficult to consider Dectin-1 deficiency as a genetic etiology of CMC99.

CARD9:

The adaptor protein CARD9 is predominantly expressed in myeloid cells and transduces signals downstream of several pattern recognition receptors, including CLRs, and is critical for antifungal immunity (Figure 1) 100. AR CARD9 deficiency was first described in 2009 in a large multiplex Iranian consanguineous family with CMC and possibly central nervous system (CNS) Candida disease101. Since then, >80 CARD9 deficient patients have been reported102. Approximately one third display superficial fungal diseases (CMC or dermatophytosis), whereas most are highly vulnerable to life-threatening invasive (e.g. affecting bones, CNS, soft tissues) fungal infections, frequently caused by Candida spp., dermatophytes, or black fungi, but are resistant to other microorganisms102 (Table 1). CARD9-deficient macrophages, dendritic cells, or peripheral blood mononuclear cells (PBMC) exhibit reduced production of cytokines (including IL-6, IL23) and chemokines when stimulated in vitro with various fungal ligands (Figure 1). CARD9-deficient neutrophils have a selective killing defect toward some but not all fungi tested103. Impaired CARD9-dependant chemokine production resulting in impaired neutrophil recruitment – and subsequent reduced neutrophil killing - to the sites of infection104106, combined with impaired CARD9-dependant cytokine production, contribute to the susceptibility to invasive fungal diseases seen in CARD9 deficient patients102, 103. Variable production of IL-17, measured ex vivo or in vitro after stimulation of CARD9-deficient PBMCs, suggests CARD9 is important, but not critical, for Th17-cell differentiation, possibly through production of IL-23 by myeloid cells, accounting for the incomplete penetrance of CMC in patients with AR CARD9 deficiency102, 103.

Inborn errors of the IL-17A/F response pathway

As described above, dissection of the molecular and cellular bases of IEI impairing production of IL-17A/F in patients with CMC paved the way for the identification of inborn errors of IL-17A/F-mediated immunity that impair the ability of target cells to respond to the effector cytokines produced by Th17 cells, thus conferring CMC in otherwise healthy individuals.

In 2011, a candidate gene approach identified AD IL-17F and AR IL-17RA deficiencies, each in a single family, as the first genetic causes of isolated CMC103, 107 (Table 1, Figure 1). A pathogenic variant in IL17F was identified in five relatives from an Argentinian family with early-onset CMC. The index patient also had recurrent upper respiratory tract infections, asthma, and episodes of furunculosis since infancy. A 9-month-old female relative heterozygous for the variant was also reported but was asymptomatic at the time of the study. All patients tested displayed normal proportions of IL-17A- and IL-22-producing T cells ex vivo. The pathogenic variant impaired binding of IL-17F to its receptor on fibroblasts. Consequently, fibroblasts or keratinocytes stimulated with mutant IL-17F homodimers or heterodimers (i.e. mutant IL-17F/wild type IL-17F, mutant IL-17F/IL-17A) showed impaired production of IL-6 and Gro-α, demonstrating the mutant IL-17F was both hypomorphic and DN on wild-type IL-17F- and IL-17A-mediated responses107 (Table 1). A second family of Tunisian-German descent has also been reported, with a woman and her son carrying a heterozygous IL17F variant and both presenting CMC from early childhood but otherwise healthy108.

In parallel, AR complete IL-17RA was reported in a child born to first cousin-parents of Moroccan descent. The patient suffered from early-onset CMC and cutaneous S. aureus infection, and carried a homozygous nonsense variant affecting the extracellular domain of IL-17RA. Additional homozygous mutations have since been reported in 27 patients with AR IL-17RA deficiency, from 14 unrelated kindreds originating from Morocco, Turkey, Japan, Saudi Arabia, Algeria, Argentina, and Sri Lanka103, 109, 110. All patients suffered from early-onset CMC. In addition, ~75% also presented staphylococcal skin diseases, and ~30% developed recurrent bacterial respiratory tract infections103, 107, 109, 110 (Table 1). AR complete IL-17RC deficiency was subsequently identified in three unrelated patients born to consanguineous families originating from Turkey and Argentina, with early-onset CMC in the absence of any other infectious phenotype, staphylococcal disease in particular111.

TRAF3IP2 encodes ACT1, a key signaling adapter functioning downstream of the IL17R complex. AR ACT1 deficiency was identified in 2013 in two siblings born to consanguineous Algerian parents, with early-onset CMC and recurrent severe skin and scalp disease caused by S. aureus112 (Table 1). Both patients carried a homozygous missense mutation of TRAF3IP2112. Nine additional patients born to non-consanguineous Spanish or Portuguese parents or consanguineous Indian, Lebanese or Turkish parents, carrying biallelic variants of TRAF3IP2 with early-onset CMC, severe skin, scalp, and pulmonary diseases were subsequently reported113117. Fibroblasts of all IL-17RA-, IL-17RC-, and ACT1-deficient patients tested failed to respond to IL-17A and IL-17F homodimers and heterodimers in terms of IL-6 or Gro-α production107, 109112 (Table 1). IL-17RA- and ACT1-deficient PBMCs, unlike IL-17RC-deficient PBMCs, also failed to respond to IL17E/IL-25110, 112, 114, which signals through IL-17RA/IL-17RB in an ACT1-dependent manner118.

JNK1 belongs to the MAPK signaling pathway and is involved in various signaling pathways, including the IL-17 pathway13. AD JNK1 deficiency was recently reported in a multiplex French family with syndromic CMC. Three patients from three generations suffered from early-onset CMC, mucocutaneous S. aureus infections, and a connective tissue disorder and carried a private heterozygous LOF variant of MAPK8 encoding JNK1119 (Table 1). Accordingly, patients’ fibroblasts displayed impaired cellular responses to IL-17A and IL-17F. JNK1 also acts downstream of TGFβ1, which contributes to human Th17 differentiation in vitro57, 120, 121 (Table 1). Consistent with this, proportions of ex vivo and in vitro-differentiated Th17 cells were reduced, but not abolished, in the patients compared to healthy donors. AD JNK1 deficiency caused CMC by haploinsufficiency, impairing production of and cellular responses to IL-17A/F119 (Table 1, Figure 1).

Thus, these five IEI due to pathogenic variants that prevent production of IL-17F homodimers or IL-17A/F heterodimers, abolish expression/function of IL17RA or IL17RC (receptors for IL-17A/F) or cripple signaling downstream of IL-17RA/C further highlight the critical role of IL-17A- and IL-17F-mediated immunity in mucocutaneous protection against Candida and S. aureus. They also suggest that IL-17A- and IL-17F-dependent immunity is otherwise redundant for protection against fungi other than Candida, bacteria other than S. aureus, viruses, or against invasive candidiasis or staphylococcal disease103, 122.

Autoimmune phenocopies: autoantibodies against IL-17A and IL-17F

Since the early 1980s, autoantibodies (auto-Abs) against cytokines have progressively emerged as important host factors in susceptibility to specific infectious diseases. By blocking the function of their target cytokines, these auto-Abs underlie infectious phenocopies of IEI of the corresponding cytokine or response pathway12. CMC is one of the three most common clinical manifestations of patients with autoimmune polyendocrine syndrome type 1 (APS-1, also called APECED syndrome), and is usually the earliest to appear123. In 2010, two independent studies reported that, irrespective of age, >90% of APS-1 patients tested had high serum titers of neutralizing IgG auto-Abs against at least one of the IL-17 cytokines (IL-17A, IL-17F, IL-22; Table 1, Figure 1). None of the healthy controls, healthy heterozygous relatives, or other patients with various autoimmune or endocrine syndromes tested in parallel had such auto-Abs124, 125. The only exception were two patients with thymoma who displayed auto-Abs against IL-17A and IL-22, and who were the only two patients with CMC of these 35 thymoma patients tested125. Levels of auto-Abs against IL-17 cytokines were already high before the onset of CMC125. Furthermore, since 2012, CMC has been described in ~2–20% of patients treated with therapeutic antibodies blocking IL-17A and/or IL-17F or IL-17RA126. This “relatively” low proportion likely reflects incomplete blocking of IL-17 cytokines in IL-17-competent individuals from birth. Nevertheless, this observation provides near-experimental proof that auto-Abs neutralizing IL-17 cytokines can underlie CMC, and provides strong support for a role of auto-Abs against IL-17 cytokines in the onset of CMC in APS-1 patients126.

Conclusion

The identification of various inborn errors of IL-17A/F immunity or auto-Abs against IL-17A/F, as well as the use of therapeutic antibodies blocking IL-17A/F function, have altogether demonstrated the essential role of human IL-17A and IL-17F in protective immunity against mucocutaneous infections with C. albicans. These studies also suggested that IL-17RA-dependent immunity is involved in mucocutaneous protection against S. aureus, but is largely redundant in host defense against other common microbes, including other bacteria and fungi, as well as viruses. These “experiments of Nature” further demonstrate the importance of deciphering the molecular and cellular mechanisms underlying human infectious diseases, thereby shedding light on their critical and redundant function in human immunity, and illuminating possible pathways to improved therapeutic intervention for people affected by these IEI and microbial infections.

Abbreviations:

DN

dominant negative

GOF

gain of function

IEI

inborn errors of immunity

LOF

loss of function

STAT

signal transducer and activator of transcription

CMC

chronic mucocutaneous candidiasis

AR

autosomal recessive

AD

autosomal dominant

JAK

Janus activating kinase

CARD9

Caspase recruitment domain-containing protein 9

Footnotes

Conflict of interest: the authors have no conflicts to declare

References

  • 1.Akdis M, Aab A, Altunbulakli C, Azkur K, Costa RA, Crameri R, et al. Interleukins (from IL-1 to IL-38), interferons, transforming growth factor beta, and TNF-alpha: Receptors, functions, and roles in diseases. J Allergy Clin Immunol 2016; 138:984–1010. [DOI] [PubMed] [Google Scholar]
  • 2.Leonard WJ. Cytokines and immunodeficiency diseases. Nat Rev Immunol 2001; 1:200–8. [DOI] [PubMed] [Google Scholar]
  • 3.O’Shea JJ, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science 2010; 327:1098–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Tangye SG, Pelham SJ, Deenick EK, Ma CS. Cytokine-Mediated Regulation of Human Lymphocyte Development and Function: Insights from Primary Immunodeficiencies. J Immunol 2017; 199:1949–58. [DOI] [PubMed] [Google Scholar]
  • 5.Levy DE, Darnell JE Jr. Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol 2002; 3:651–62. [DOI] [PubMed] [Google Scholar]
  • 6.Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system. Nat Rev Immunol 2003; 3:900–11. [DOI] [PubMed] [Google Scholar]
  • 7.O’Shea JJ, Holland SM, Staudt LM. JAKs and STATs in immunity, immunodeficiency, and cancer. N Engl J Med 2013; 368:161–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Casanova JL, Holland SM, Notarangelo LD. Inborn errors of human JAKs and STATs. Immunity 2012; 36:515–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tangye SG, Al-Herz W, Bousfiha A, Cunningham-Rundles C, Franco JL, Holland SM, et al. Human Inborn Errors of Immunity: 2022 Update on the Classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sharma M, Leung D, Momenilandi M, Jones LCW, Pacillo L, James AE, et al. Human germline heterozygous gain-of-function STAT6 variants cause severe allergic disease. J Exp Med 2023; 220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.International Consortium I, Fornes O, Jia A, Kuehn HS, Min Q, Pannicke U, et al. A multimorphic mutation in IRF4 causes human autosomal dominant combined immunodeficiency. Sci Immunol 2023; 8:eade7953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Puel A, Bastard P, Bustamante J, Casanova JL. Human autoantibodies underlying infectious diseases. J Exp Med 2022; 219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Gaffen SL, Jain R, Garg AV, Cua DJ. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol 2014; 14:585–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rouvier E, Luciani MF, Mattei MG, Denizot F, Golstein P. CTLA-8, cloned from an activated T cell, bearing AU-rich messenger RNA instability sequences, and homologous to a herpesvirus saimiri gene. J Immunol 1993; 150:5445–56. [PubMed] [Google Scholar]
  • 15.Yao Z, Painter SL, Fanslow WC, Ulrich D, Macduff BM, Spriggs MK, et al. Human IL-17: a novel cytokine derived from T cells. J Immunol 1995; 155:5483–6. [PubMed] [Google Scholar]
  • 16.Hymowitz SG, Filvaroff EH, Yin JP, Lee J, Cai L, Risser P, et al. IL-17s adopt a cystine knot fold: structure and activity of a novel cytokine, IL-17F, and implications for receptor binding. Embo J 2001; 20:5332–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chang SH, Dong C. A novel heterodimeric cytokine consisting of IL-17 and IL-17F regulates inflammatory responses. Cell Res 2007; 17:435–40. [DOI] [PubMed] [Google Scholar]
  • 18.Liang SC, Long AJ, Bennett F, Whitters MJ, Karim R, Collins M, et al. An IL-17F/A heterodimer protein is produced by mouse Th17 cells and induces airway neutrophil recruitment. J Immunol 2007; 179:7791–9. [DOI] [PubMed] [Google Scholar]
  • 19.Wright JF, Guo Y, Quazi A, Luxenberg DP, Bennett F, Ross JF, et al. Identification of an interleukin 17F/17A heterodimer in activated human CD4+ T cells. J Biol Chem 2007; 282:13447–55. [DOI] [PubMed] [Google Scholar]
  • 20.Sallusto F Heterogeneity of Human CD4(+) T Cells Against Microbes. Annu Rev Immunol 2016; 34:317–34. [DOI] [PubMed] [Google Scholar]
  • 21.Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 2005; 6:1123–32. [DOI] [PubMed] [Google Scholar]
  • 22.Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol 2005; 6:1133–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 2005; 201:233–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 Cells. Annu Rev Immunol 2009; 27:485–517. [DOI] [PubMed] [Google Scholar]
  • 25.He D, Wu L, Kim HK, Li H, Elmets CA, Xu H. CD8+ IL-17-producing T cells are important in effector functions for the elicitation of contact hypersensitivity responses. J Immunol 2006; 177:6852–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ciric B, El-behi M, Cabrera R, Zhang GX, Rostami A. IL-23 drives pathogenic IL-17-producing CD8+ T cells. J Immunol 2009; 182:5296–305. [DOI] [PubMed] [Google Scholar]
  • 27.Huber M, Heink S, Pagenstecher A, Reinhard K, Ritter J, Visekruna A, et al. IL-17A secretion by CD8+ T cells supports Th17-mediated autoimmune encephalomyelitis. J Clin Invest 2013; 123:247–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sutton CE, Lalor SJ, Sweeney CM, Brereton CF, Lavelle EC, Mills KH. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity 2009; 31:331–41. [DOI] [PubMed] [Google Scholar]
  • 29.Roark CL, French JD, Taylor MA, Bendele AM, Born WK, O’Brien RL. Exacerbation of collagen-induced arthritis by oligoclonal, IL-17-producing gamma delta T cells. J Immunol 2007; 179:5576–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Cai Y, Shen X, Ding C, Qi C, Li K, Li X, et al. Pivotal role of dermal IL-17-producing gammadelta T cells in skin inflammation. Immunity 2011; 35:596–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Cupedo T, Crellin NK, Papazian N, Rombouts EJ, Weijer K, Grogan JL, et al. Human fetal lymphoid tissue-inducer cells are interleukin 17-producing precursors to RORC+ CD127+ natural killer-like cells. Nat Immunol 2009; 10:66–74. [DOI] [PubMed] [Google Scholar]
  • 32.Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov, II, et al. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med 2009; 206:35–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Buonocore S, Ahern PP, Uhlig HH, Ivanov II, Littman DR, Maloy KJ, et al. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 2010; 464:1371–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Villanova F, Flutter B, Tosi I, Grys K, Sreeneebus H, Perera GK, et al. Characterization of innate lymphoid cells in human skin and blood demonstrates increase of NKp44+ ILC3 in psoriasis. J Invest Dermatol 2014; 134:984–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. Innate lymphoid cells--a proposal for uniform nomenclature. Nat Rev Immunol 2013; 13:145–9. [DOI] [PubMed] [Google Scholar]
  • 36.Kronenberg M Toward an understanding of NKT cell biology: progress and paradoxes. Annu Rev Immunol 2005; 23:877–900. [DOI] [PubMed] [Google Scholar]
  • 37.Michel ML, Keller AC, Paget C, Fujio M, Trottein F, Savage PB, et al. Identification of an IL-17-producing NK1.1(neg) iNKT cell population involved in airway neutrophilia. J Exp Med 2007; 204:995–1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Michel ML, Mendes-da-Cruz D, Keller AC, Lochner M, Schneider E, Dy M, et al. Critical role of RORgammat in a new thymic pathway leading to IL-17-producing invariant NKT cell differentiation. Proc Natl Acad Sci U S A 2008; 105:19845–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Carolan E, Tobin LM, Mangan BA, Corrigan M, Gaoatswe G, Byrne G, et al. Altered distribution and increased IL-17 production by mucosal-associated invariant T cells in adult and childhood obesity. J Immunol 2015; 194:5775–80. [DOI] [PubMed] [Google Scholar]
  • 40.Dusseaux M, Martin E, Serriari N, Peguillet I, Premel V, Louis D, et al. Human MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-17-secreting T cells. Blood 2011; 117:1250–9. [DOI] [PubMed] [Google Scholar]
  • 41.Wilson RP, Ives ML, Rao G, Lau A, Payne K, Kobayashi M, et al. STAT3 is a critical cell-intrinsic regulator of human unconventional T cell numbers and function. J Exp Med 2015; 212:855–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol 2007; 8:942–9. [DOI] [PubMed] [Google Scholar]
  • 43.Teng MW, Bowman EP, McElwee JJ, Smyth MJ, Casanova JL, Cooper AM, et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med 2015; 21:719–29. [DOI] [PubMed] [Google Scholar]
  • 44.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; 190:624–31. [DOI] [PubMed] [Google Scholar]
  • 45.Conti HR, Shen F, Nayyar N, Stocum E, Sun JN, Lindemann MJ, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M, Lanzavecchia A, et al. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat Immunol 2007; 8:639–46. [DOI] [PubMed] [Google Scholar]
  • 47.Conti HR, Bruno VM, Childs EE, Daugherty S, Hunter JP, Mengesha BG, et al. IL-17 Receptor Signaling in Oral Epithelial Cells Is Critical for Protection against Oropharyngeal Candidiasis. Cell Host Microbe 2016; 20:606–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Okada S, Puel A, Casanova JL, Kobayashi M. Chronic mucocutaneous candidiasis disease associated with inborn errors of IL-17 immunity. Clin Transl Immunology 2016; 5:e114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, et al. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature 2006; 441:231–4. [DOI] [PubMed] [Google Scholar]
  • 50.Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 2006; 24:179–89. [DOI] [PubMed] [Google Scholar]
  • 51.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006; 441:235–8. [DOI] [PubMed] [Google Scholar]
  • 52.Wilson NJ, Boniface K, Chan JR, McKenzie BS, Blumenschein WM, Mattson JD, et al. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat Immunol 2007; 8:950–7. [DOI] [PubMed] [Google Scholar]
  • 53.Korn T, Bettelli E, Gao W, Awasthi A, Jager A, Strom TB, et al. IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells. Nature 2007; 448:484–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T, et al. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol 2007; 8:967–74. [DOI] [PubMed] [Google Scholar]
  • 55.Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD, Ma L, et al. Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature 2007; 448:480–3. [DOI] [PubMed] [Google Scholar]
  • 56.Aggarwal S, Ghilardi N, Xie MH, de Sauvage FJ, Gurney AL. Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17. J Biol Chem 2003; 278:1910–4. [DOI] [PubMed] [Google Scholar]
  • 57.Yang L, Anderson DE, Baecher-Allan C, Hastings WD, Bettelli E, Oukka M, et al. IL-21 and TGF-beta are required for differentiation of human T(H)17 cells. Nature 2008; 454:350–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, et al. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity 2007; 26:371–81. [DOI] [PubMed] [Google Scholar]
  • 59.Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS, et al. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem 2007; 282:9358–63. [DOI] [PubMed] [Google Scholar]
  • 60.Harris TJ, Grosso JF, Yen HR, Xin H, Kortylewski M, Albesiano E, et al. Cutting edge: An in vivo requirement for STAT3 signaling in TH17 development and TH17-dependent autoimmunity. J Immunol 2007; 179:4313–7. [DOI] [PubMed] [Google Scholar]
  • 61.Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006; 126:1121–33. [DOI] [PubMed] [Google Scholar]
  • 62.Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity 2008; 28:29–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Coquet JM, Chakravarti S, Smyth MJ, Godfrey DI. Cutting edge: IL-21 is not essential for Th17 differentiation or experimental autoimmune encephalomyelitis. J Immunol 2008; 180:7097–101. [DOI] [PubMed] [Google Scholar]
  • 64.Sonderegger I, Kisielow J, Meier R, King C, Kopf M. IL-21 and IL-21R are not required for development of Th17 cells and autoimmunity in vivo. Eur J Immunol 2008; 38:1833–8. [DOI] [PubMed] [Google Scholar]
  • 65.Holland SM, DeLeo FR, Elloumi HZ, Hsu AP, Uzel G, Brodsky N, et al. STAT3 mutations in the hyper-IgE syndrome. N Engl J Med 2007; 357:1608–19. [DOI] [PubMed] [Google Scholar]
  • 66.Minegishi Y, Saito M, Tsuchiya S, Tsuge I, Takada H, Hara T, et al. Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyper-IgE syndrome. Nature 2007; 448:1058–62. [DOI] [PubMed] [Google Scholar]
  • 67.Chandesris MO, Melki I, Natividad A, Puel A, Fieschi C, Yun L, et al. Autosomal dominant STAT3 deficiency and hyper-IgE syndrome: molecular, cellular, and clinical features from a French national survey. Medicine (Baltimore) 2012; 91:e1–e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ma CS, Chew GY, Simpson N, Priyadarshi A, Wong M, Grimbacher B, et al. Deficiency of Th17 cells in hyper IgE syndrome due to mutations in STAT3. J Exp Med 2008; 205:1551–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Renner ED, Rylaarsdam S, Anover-Sombke S, Rack AL, Reichenbach J, Carey JC, et al. Novel signal transducer and activator of transcription 3 (STAT3) mutations, reduced T(H)17 cell numbers, and variably defective STAT3 phosphorylation in hyper-IgE syndrome. J Allergy Clin Immunol 2008; 122:181–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Milner JD, Brenchley JM, Laurence A, Freeman AF, Hill BJ, Elias KM, et al. Impaired T(H)17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature 2008; 452:773–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ma CS, Wong N, Rao G, Avery DT, Torpy J, Hambridge T, et al. Monogenic mutations differentially affect the quantity and quality of T follicular helper cells in patients with human primary immunodeficiencies. J Allergy Clin Immunol 2015; 136:993–1006 e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.de Beaucoudrey L, Puel A, Filipe-Santos O, Cobat A, Ghandil P, Chrabieh M, et al. Mutations in STAT3 and IL12RB1 impair the development of human IL-17-producing T cells. J Exp Med 2008; 205:1543–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ma CS, Wong N, Rao G, Nguyen A, Avery DT, Payne K, et al. Unique and shared signaling pathways cooperate to regulate the differentiation of human CD4+ T cells into distinct effector subsets. J Exp Med 2016; 213:1589–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Okada S, Markle JG, Deenick EK, Mele F, Averbuch D, Lagos M, et al. IMMUNODEFICIENCIES. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations. Science 2015; 349:606–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Beziat V, Li J, Lin JX, Ma CS, Li P, Bousfiha A, et al. A recessive form of hyper-IgE syndrome by disruption of ZNF341-dependent STAT3 transcription and activity. Sci Immunol 2018; 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Frey-Jakobs S, Hartberger JM, Fliegauf M, Bossen C, Wehmeyer ML, Neubauer JC, et al. ZNF341 controls STAT3 expression and thereby immunocompetence. Sci Immunol 2018; 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Su HC, Jing H, Angelus P, Freeman AF. Insights into immunity from clinical and basic science studies of DOCK8 immunodeficiency syndrome. Immunol Rev 2019; 287:9–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Tangye SG, Pillay B, Randall KL, Avery DT, Phan TG, Gray P, et al. Dedicator of cytokinesis 8-deficient CD4(+) T cells are biased to a T(H)2 effector fate at the expense of T(H)1 and T(H)17 cells. J Allergy Clin Immunol 2017; 139:933–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Keles S, Charbonnier LM, Kabaleeswaran V, Reisli I, Genel F, Gulez N, et al. Dedicator of cytokinesis 8 regulates signal transducer and activator of transcription 3 activation and promotes TH17 cell differentiation. J Allergy Clin Immunol 2016; 138:1384–94 e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Toubiana J, Okada S, Hiller J, Oleastro M, Lagos Gomez M, Aldave Becerra JC, et al. Heterozygous STAT1 gain-of-function mutations underlie an unexpectedly broad clinical phenotype. Blood 2016; 127:3154–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Liu L, Okada S, Kong XF, Kreins AY, Cypowyj S, Abhyankar A, et al. Gain-of-function human STAT1 mutations impair IL-17 immunity and underlie chronic mucocutaneous candidiasis. J Exp Med 2011; 208:1635–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zheng J, van de Veerdonk FL, Crossland KL, Smeekens SP, Chan CM, Al Shehri T, et al. Gain-of-function STAT1 mutations impair STAT3 activity in patients with chronic mucocutaneous candidiasis (CMC). Eur J Immunol 2015; 45:2834–46. [DOI] [PubMed] [Google Scholar]
  • 83.Hong F, Jaruga B, Kim WH, Radaeva S, El-Assal ON, Tian Z, et al. Opposing roles of STAT1 and STAT3 in T cell-mediated hepatitis: regulation by SOCS. J Clin Invest 2002; 110:1503–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Forbes LR, Vogel TP, Cooper MA, Castro-Wagner J, Schussler E, Weinacht KG, et al. Jakinibs for the treatment of immune dysregulation in patients with gain-of-function signal transducer and activator of transcription 1 (STAT1) or STAT3 mutations. J Allergy Clin Immunol 2018; 142:1665–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Deya-Martinez A, Riviere JG, Roxo-Junior P, Ramakers J, Bloomfield M, Guisado Hernandez P, et al. Impact of JAK Inhibitors in Pediatric Patients with STAT1 Gain of Function (GOF) Mutations-10 Children and Review of the Literature. J Clin Immunol 2022; 42:1071–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.de Beaucoudrey L, Samarina A, Bustamante J, Cobat A, Boisson-Dupuis S, Feinberg J, et al. Revisiting human IL-12Rbeta1 deficiency: a survey of 141 patients from 30 countries. Medicine (Baltimore) 2010; 89:381–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Martinez-Barricarte R, Markle JG, Ma CS, Deenick EK, Ramirez-Alejo N, Mele F, et al. Human IFN-gamma immunity to mycobacteria is governed by both IL-12 and IL-23. Sci Immunol 2018; 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Philippot Q, Ogishi M, Bohlen J, Puchan J, Arias AA, Nguyen T, et al. Human IL-23 is essential for IFN-gamma-dependent immunity to mycobacteria. Sci Immunol 2023; 8:eabq5204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ouederni M, Sanal O, Ikinciogullari A, Tezcan I, Dogu F, Sologuren I, et al. Clinical features of Candidiasis in patients with inherited interleukin 12 receptor beta1 deficiency. Clin Infect Dis 2014; 58:204–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Cagdas D, Mayr D, Baris S, Worley L, Langley DB, Metin A, et al. Genomic Spectrum and Phenotypic Heterogeneity of Human IL-21 Receptor Deficiency. J Clin Immunol 2021; 41:1272–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kotlarz D, Zietara N, Uzel G, Weidemann T, Braun CJ, Diestelhorst J, et al. Loss-of-function mutations in the IL-21 receptor gene cause a primary immunodeficiency syndrome. J Exp Med 2013; 210:433–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Stepensky P, Keller B, Abuzaitoun O, Shaag A, Yaacov B, Unger S, et al. Extending the clinical and immunological phenotype of human interleukin-21 receptor deficiency. Haematologica 2015; 100:e72–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Erman B, Bilic I, Hirschmugl T, Salzer E, Cagdas D, Esenboga S, et al. Combined immunodeficiency with CD4 lymphopenia and sclerosing cholangitis caused by a novel loss-of-function mutation affecting IL21R. Haematologica 2015; 100:e216–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Beziat V, Tavernier SJ, Chen YH, Ma CS, Materna M, Laurence A, et al. Dominant-negative mutations in human IL6ST underlie hyper-IgE syndrome. J Exp Med 2020; 217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Brown GD. Dectin-1: a signalling non-TLR pattern-recognition receptor. Nat Rev Immunol 2006; 6:33–43. [DOI] [PubMed] [Google Scholar]
  • 96.Kalia N, Singh J, Kaur M. The role of dectin-1 in health and disease. Immunobiology 2021; 226:152071. [DOI] [PubMed] [Google Scholar]
  • 97.LeibundGut-Landmann S, Gross O, Robinson MJ, Osorio F, Slack EC, Tsoni SV, et al. Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17. Nat Immunol 2007; 8:630–8. [DOI] [PubMed] [Google Scholar]
  • 98.Ferwerda B, Ferwerda G, Plantinga TS, Willment JA, van Spriel AB, Venselaar H, et al. Human dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med 2009; 361:1760–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.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; 22:467–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Winthrop KL, Mariette X, Silva JT, Benamu E, Calabrese LH, Dumusc A, et al. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Soluble immune effector molecules [II]: agents targeting interleukins, immunoglobulins and complement factors). Clinical microbiology and infection : the official publication of the European Society of Clinical Microbiology and Infectious Diseases 2018; 24 Suppl 2:S21–S40. [DOI] [PubMed] [Google Scholar]
  • 101.Glocker EO, Hennigs A, Nabavi M, Schaffer AA, Woellner C, Salzer U, et al. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N Engl J Med 2009; 361:1727–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Corvilain E, Casanova JL, Puel A. Inherited CARD9 Deficiency: Invasive Disease Caused by Ascomycete Fungi in Previously Healthy Children and Adults. J Clin Immunol 2018; 38:656–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li J, Vinh DC, Casanova JL, Puel A. Inborn errors of immunity underlying fungal diseases in otherwise healthy individuals. Current opinion in microbiology 2017; 40:46–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Rieber N, Gazendam RP, Freeman AF, Hsu AP, Collar AL, Sugui JA, et al. Extrapulmonary Aspergillus infection in patients with CARD9 deficiency. JCI Insight 2016; 1:e89890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Drummond RA, Swamydas M, Oikonomou V, Zhai B, Dambuza IM, Schaefer BC, et al. CARD9(+) microglia promote antifungal immunity via IL-1beta- and CXCL1-mediated neutrophil recruitment. Nat Immunol 2019; 20:559–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Drummond RA, Collar AL, Swamydas M, Rodriguez CA, Lim JK, Mendez LM, et al. CARD9-Dependent Neutrophil Recruitment Protects against Fungal Invasion of the Central Nervous System. PLoS Pathog 2015; 11:e1005293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Puel A, Cypowyj S, Bustamante J, Wright JF, Liu L, Lim HK, et al. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science 2011; 332:65–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Bader O, Weig MS, Gross U, Schon MP, Mempel M, Buhl T. Photo quiz. A 32-year-old man with ulcerative mucositis, skin lesions, and nail dystrophy. Chronic mucocutaneous candidiasis by multidrug-resistant Candida albicans. Clin Infect Dis 2012; 54:972, 1035–6. [DOI] [PubMed] [Google Scholar]
  • 109.Fellmann F, Angelini F, Wassenberg J, Perreau M, Arenas Ramirez N, Simon G, et al. IL-17 receptor A and adenosine deaminase 2 deficiency in siblings with recurrent infections and chronic inflammation. J Allergy Clin Immunol 2016; 137:1189–96 e2. [DOI] [PubMed] [Google Scholar]
  • 110.Levy R, Okada S, Beziat V, Moriya K, Liu C, Chai LY, et al. Genetic, immunological, and clinical features of patients with bacterial and fungal infections due to inherited IL-17RA deficiency. Proc Natl Acad Sci U S A 2016; 113:E8277–E85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ling Y, Cypowyj S, Aytekin C, Galicchio M, Camcioglu Y, Nepesov S, et al. Inherited IL-17RC deficiency in patients with chronic mucocutaneous candidiasis. J Exp Med 2015; 212:619–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Boisson B, Wang C, Pedergnana V, Wu L, Cypowyj S, Rybojad M, et al. An ACT1 mutation selectively abolishes interleukin-17 responses in humans with chronic mucocutaneous candidiasis. Immunity 2013; 39:676–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Bhattad S, Dinakar C, Pinnamaraju H, Ganapathy A, Mannan A. Chronic Mucocutaneous Candidiasis in an Adolescent Boy Due to a Novel Mutation in TRAF3IP2. J Clin Immunol 2019; 39:596–9. [DOI] [PubMed] [Google Scholar]
  • 114.Shafer S, Yao Y, Comrie W, Cook S, Zhang Y, Yesil G, et al. Two patients with chronic mucocutaneous candidiasis caused by TRAF3IP2 deficiency. J Allergy Clin Immunol 2021; 148:256–61 e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Blanco Lobo P, Lei WT, Pelham SJ, Guisado Hernandez P, Villaoslada I, de Felipe B, et al. Biallelic TRAF3IP2 variants causing chronic mucocutaneous candidiasis in a child harboring a STAT1 variant. Pediatr Allergy Immunol 2021; 32:1804–12. [DOI] [PubMed] [Google Scholar]
  • 116.Marujo F, Pelham SJ, Freixo J, Cordeiro AI, Martins C, Casanova JL, et al. A Novel TRAF3IP2 Mutation Causing Chronic Mucocutaneous Candidiasis. J Clin Immunol 2021; 41:1376–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Nemer G, El-Hachem N, Eid E, Hamie L, Bardawil T, Khalil S, et al. A novel TRAF3IP2 variant causing familial scarring alopecia with mixed features of discoid lupus erythematosus and folliculitis decalvans. Clin Genet 2020; 98:116–25. [DOI] [PubMed] [Google Scholar]
  • 118.Gaffen SL. Structure and signalling in the IL-17 receptor family. Nat Rev Immunol 2009; 9:556–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Li J, Ritelli M, Ma CS, Rao G, Habib T, Corvilain E, et al. Chronic mucocutaneous candidiasis and connective tissue disorder in humans with impaired JNK1-dependent responses to IL-17A/F and TGF-beta. Sci Immunol 2019; 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Manel N, Unutmaz D, Littman DR. The differentiation of human T(H)-17 cells requires transforming growth factor-beta and induction of the nuclear receptor RORgammat. Nat Immunol 2008; 9:641–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Volpe E, Servant N, Zollinger R, Bogiatzi SI, Hupe P, Barillot E, et al. A critical function for transforming growth factor-beta, interleukin 23 and proinflammatory cytokines in driving and modulating human T(H)-17 responses. Nat Immunol 2008; 9:650–7. [DOI] [PubMed] [Google Scholar]
  • 122.Puel A Human inborn errors of immunity underlying superficial or invasive candidiasis. Hum Genet 2020; 139:1011–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Cheng MH, Anderson MS. Monogenic autoimmunity. Annu Rev Immunol 2012; 30:393–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Puel A, Doffinger R, Natividad A, Chrabieh M, Barcenas-Morales G, Picard C, 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; 207:291–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kisand K, Boe Wolff AS, Podkrajsek KT, Tserel L, Link M, Kisand KV, et al. Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines. J Exp Med 2010; 207:299–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Philippot Q, Casanova JL, Puel A. Candidiasis in patients with APS-1: low IL-17, high IFN-gamma, or both? Curr Opin Immunol 2021; 72:318–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Prando C, Samarina A, Bustamante J, Boisson-Dupuis S, Cobat A, Picard C, et al. Inherited IL-12p40 deficiency: genetic, immunologic, and clinical features of 49 patients from 30 kindreds. Medicine (Baltimore) 2013; 92:109–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Spencer S, Kostel Bal S, Egner W, Lango Allen H, Raza Syed I, Ma CA, et al. Loss of the interleukin 6 receptor causes immunodeficiency, atopy, and abnormal inflammatory responses. J Exp Med 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Nahum A, Sharfe N, Broides A, Dadi H, Naghdi Z, Mandola AB, et al. Defining the biological responses of IL-6 by the study of a novel IL-6 receptor chain immunodeficiency. J Allergy Clin Immunol 2020; 145:1011–5 e6. [DOI] [PubMed] [Google Scholar]
  • 130.Schwerd T, Twigg SRF, Aschenbrenner D, Manrique S, Miller KA, Taylor IB, et al. A biallelic mutation in IL6ST encoding the GP130 co-receptor causes immunodeficiency and craniosynostosis. J Exp Med 2017; 214:2547–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Shahin T, Aschenbrenner D, Cagdas D, Bal SK, Conde CD, Garncarz W, et al. Selective loss of function variants in IL6ST cause Hyper-IgE syndrome with distinct impairments of T-cell phenotype and function. Haematologica 2019; 104:609–21. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES