Next Article in Journal
rCsHscB Derived from Clonorchis sinensis: A Carcinogenic Liver Fluke Ameliorates LPS-Induced Acute Hepatic Injury by Repression of Inflammation
Next Article in Special Issue
Current Knowledge of Th22 Cell and IL-22 Functions in Infectious Diseases
Previous Article in Journal
Syphilis as Re-Emerging Disease, Antibiotic Resistance, and Vulnerable Population: Global Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Double Game Played by Th17 Cells in Infection: Host Defense and Immunopathology

1
Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
2
Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
3
Post Graduate School of Public Health, University of Siena, 53100 Siena, Italy
4
Department of Anatomy, Histology, Legal Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
5
Eye Clinic, Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
*
Author to whom correspondence should be addressed.
Pathogens 2022, 11(12), 1547; https://doi.org/10.3390/pathogens11121547
Submission received: 6 November 2022 / Revised: 9 December 2022 / Accepted: 14 December 2022 / Published: 15 December 2022
(This article belongs to the Special Issue 10th Anniversary of Pathogens: T Cells in Pathogenic Infections)

Abstract

:
T-helper 17 (Th17) cells represent a subpopulation of CD4+ T lymphocytes that play an essential role in defense against pathogens. Th17 cells are distinguished from Th1 and Th2 cells by their ability to produce members of the interleukin-17 (IL-17) family, namely IL-17A and IL-17F. IL-17 in turn induces several target cells to synthesize and release cytokines, chemokines, and metalloproteinases, thereby amplifying the inflammatory cascade. Th17 cells reside predominantly in the lamina propria of the mucosa. Their main physiological function is to maintain the integrity of the mucosal barrier against the aggression of infectious agents. However, in an appropriate inflammatory microenvironment, Th17 cells can transform into immunopathogenic cells, giving rise to inflammatory and autoimmune diseases. This review aims to analyze the complex mechanisms through which the interaction between Th17 and pathogens can be on the one hand favorable to the host by protecting it from infectious agents, and on the other hand harmful, potentially generating autoimmune reactions and tissue damage.

1. Introduction

T helper 17 cells represent a subset of CD4+ T lymphocytes discovered about 20 years ago [1,2]. It was evident from the beginning that this population played a key role in defense against bacteria, viruses, and fungi. It was shown that the defensive action of Th17 cells occurred primarily through the production of members of the IL-17 family, in particular IL-17A and IL-17F [3,4]. It was also found that the specific cytokine signature of Th17 cells was closely dependent on IL-23 produced by cells of innate immunity in response to stimulation by microbial agents. The dependence of IL-17 production on IL-23 led to the formulation of the term “IL-23/IL-17 axis” [5].
Although Th17 cells were shown to be essential in preserving the integrity of the mucosal barrier from attack by infectious agents, subsequent studies revealed that these cells were also involved in immunopathological processes. Indeed, in an appropriate inflammatory microenvironment, they were found to be responsible for immunopathology, promoting the induction and maintenance of inflammatory and autoimmune diseases [6,7,8,9,10].
The purpose of this review is therefore to summarize the discoveries that led to the identification of Th17 cells and to analyze the complex mechanisms of their differentiation and function, with a focus on how their interaction with pathogens can lead on the one hand to defense against the infectious agents and on the other hand to the generation of immunopathology, as emerged from experimental animal models and human study.

2. Th17 Cell Discovery: A Novel CD4+ T-Helper Cell Paradigm

For several years since the second half of the 1980s, CD4+ T cells have been subdivided into two subpopulations, T-helper 1 (Th1) cells and Th2 cells [11]. Th1 cells were characterized by their ability to produce interferon (IFN)-γ, cooperate with B cells to produce immunoglobulin (Ig)G, and give rise to delayed-type T-cell responses [12]. Conversely, Th2 cells were characterized by the ability to produce interleukin (IL)-4, which is required to promote isotope switching by B cells for IgE production [13,14]. While Th1 cells predominantly provided defense against intracellular pathogens, Th2 cells provided defense against parasites. In 1989, the so-called Th1/Th2 paradigm was thus formulated [15].
Subsequently, however, several pieces of evidence showed that IFN-γ production by Th1 cells did not fully explain the presence of pro-inflammatory CD4+ T-cell mediated responses in mouse models [16,17,18]. The Th1/Th2 paradigm was therefore destined to be overcome.

3. IL-17-Member Family

A critical event that preceded the discovery of Th17 cells was the cloning of a new interleukin with pro-inflammatory properties in 1993 [19,20]. This interleukin was initially defined as CTLA8 and later as interleukin-17A. That interleukin showed sequence homology with an open reading frame of the Herpesvirus saimiri, a herpes virus capable of infecting T cells [21,22]. Interleukin 17A was characterized by not having sequence homology with other known cytokines and deserved the definition of cytokine because it could induce the production of immune-active soluble factors by target cells [23]. Other cytokines structurally similar to interleukin 17A were then identified in sequence homology studies [24,25,26]. All these molecules were then grouped into a family defined as IL-17 that presently includes six members from 17A to IL-17F [27,28]. Five specific receptors expressed on different cell types were then cloned [29,30,31,32]. These receptors have molecular peculiarities that differentiate them from other interleukin receptors. In particular, they contain an intracytoplasmic motif termed SEFIR (SEF/IL-17 receptor) that has some similarities with a domain present in the Toll/IL-1 receptor (TIR) [26]. IL-17R signaling initiates with the recruitment of Act1 adaptor molecule, with subsequent IL-17R/Act-1 association [33,34,35] thus amplifying the signal transduction. This interaction is critical in the response to pathogens as demonstrated in murine models of knockout mice for the gene encoding IL-17RA as well as in humans with ACT1 mutations or with congenital deficiency of IL-17A or IL-17C, where a high susceptibility to fungal infections is observed [36]. Act1 possesses the peculiar ability to bind to E3 Ubiquitin [37]. Through this interaction, TNF-receptor associated factor(TRAF)6 is then recruited [34,38], resulting in activation of the nuclear factor kappa-light-chain-enhancer of activated B (NF-κB) and subsequent gene transcription of several antimicrobial proteins [20,38,39,40,41].
In the year 2000, a discovery would prove essential for the subsequent identification of new pro-inflammatory CD4+ T cells in addition to the classic Th1 and Th2 cells [42]. Indeed, a new cytokine called p19 was discovered. This cytokine was able to form a heterodimer with the p40 chain of IL-12. The association of these two proteins originated a new interleukin called IL-23. Interleukin-23 was able to bind a receptor to IL-23R constituted by IL23R/IL-12β1 heterodimer [43]. Later studies showed that IL-23 was produced mainly by dendritic cells after activation by prostaglandin E and adenosine triphosphate [44,45]. It was shown that IL-23 was able to induce IL-17 production by a subpopulation of CD4+ T cells distinct from both Th1 and Th2 cells [46,47]. Therefore, this subpopulation was termed Th17 [1,2]. The peculiar differentiation and function characteristics of this subpopulation were further defined [7]. Th17 cells predominantly produce a member of the IL-17 family, namely IL-17A. However, subsequent study showed that other soluble factors involved in the inflammatory response, such as IL-17F, IL-21, IL-22, IL-26, and the chemokines CXCL8 and CCL20, were also produced [5]. It was found that a peculiar feature of Th17 cells was that their differentiation depended on specific transcription factors retinoic-acid orphan gamma receptor t (RORγt) in mice and its human isoform retinoic-acid orphan receptor C (RORC) [48,49]. These factors in turn induced il17a gene transcription [6,50]. Th17 cells were shown to characteristically express CCR6 chemokine receptor on their surface [4]. Further studies identified the existence of an intermediate cell subtype termed Th1Th17 able to produce both IL-17 and IFN-γ [3,51,52].

4. Differentiation of Th17 Cells

Differentiation of Th17 cells is a rather intricate molecular process that has been clarified at least in part only recently. This process requires TCR engagement of naïve CD4+ T cells together with the action of different cytokines present in the microenvironment. In more detail, IL-6 and tissue growth factor-β (TGF-β) provide to chromatin remodeling of the il17 gene locus [17,53,54]. IL-6 enhances retinoic-acid orphan receptors (RORγt) transcription through signal transducer and activator of transcription 3 (STAT3) phosphorylation [17], whereas TGF-β regulates Th17 differentiation via Staufen1 (STAU1)-mediated mRNA decay (SMD)-dependent or -independent pathways. It is noteworthy that TGF-β is also capable to promote regulatory T-cell (Treg) differentiation, which in turn suppresses Th17 through the function of forkhead box P3 (FOXP3). However, this activity is counteracted by IL-6-phosphorylated STAT3, which downregulates FOXP3, with consequent induction of RORγt and transformation of inducible Tregs into Th17 cells [50]. An important role in the early stages of Th17 cell differentiation is also played by IL-1β. This interleukin upregulates the expression of interferon regulatory factor (IRF) 4 [55] and RORγt [56,57,58]. Once differentiated, Th17 cells express IL23R on their surface. Interaction with IL-23 present in the inflammatory microenvironment is required to maintain the phenotype of Th17 cells, increasing RORγt and IL-17 expression by the intervention of STAT3 [59], but does not participate in the Th17 differentiation process [8,60]. Figure 1 shows the differentiation process of Th17 cells.

5. Plasticity of Th17

An important feature of Th17 cells is their plasticity, defined by their ability to acquire functional and phenotypic characteristics of other CD4+ T lymphocyte subpopulations. Indeed, Th-17 cells can acquire the characteristics of Th1 cells in the presence of interleukin 12, which downregulates the expression of RORγt/RORC and induces the expression of T-bet, a major transcription factor of IFN-γ [61,62,63]. In a microenvironment rich in IL-4, they can acquire a Th2 phenotype [64]. Th17 cells present in the Peyer’s patches can acquire the follicular T cells (Tfh) phenotype and induce immunoglobulin (Ig) A production by geminal center B lymphocytes [65]. Finally, differentiation of naïve CD4+ T cells into Treg or Th17 is bidirectional. Interconnection between these two subpopulations depends on FOXP3/ RORγt/RORC balance, as demonstrated in numerous studies. This is regulated by the relative activity of several cytokines, including TGF-β, IL-6, IL-21, IL1β, and IL-23 [66,67,68]. Th17 cells exert their function on several cellular targets that are not only part of the immune system, but also express receptors for cytokines they produce.

6. The Physiological Function of Th17

The final effect of Th17-produced cytokines, mainly IL-17A, induces the consequent production of chemokines, interleukins, and chemokines by IL17R+ cells. These factors participate to the recruitment of neutrophils to the inflammatory site and induce secretion of anti-bacterial substances by epithelial cells [7]. Importantly, Th17 is localized mainly at the mucosal level where it exerts protective activity against bacteria and fungi [69]. The primary function of Th17 cells is therefore to maintain immune control of infection at the mucosal level and in the skin [70,71]. As discussed above, the maintenance of their differentiative state over time is strictly dependent on the cytokine environment. To this end, an important role is played by low levels of IL-1β produced by macrophage cells stimulated by intestinal commensal bacteria [72]. In the skin, commensal bacteria including S. epidermidis contribute to Th17 cell stability [71]. The presence of several metabolites including tryptophan can also favor the differentiation state of Th17 cells [73]. At the intestinal level, Th17 cells produce IL-22 and IL-21 in addition to IL-17. IL-17 and IL-22 locally exert an antimicrobial action through the production of bactericidal proteins [74]. Either in experimental animal models or rare primary immunodeficiencies in humans, alteration in the production of interleukin 17 or its receptors as well as in the case of RORγt mutations, loss of immunological defense against C. albicans and S. aureus has been observed at the skin, nail, and genital mucosa levels [75,76]. The observation that commensal segmented filamentous bacteria (SFBs) can induce a vigorous Th17 response in the intestine appears to be of considerable importance [77]. Such bacteria have a special ability to penetrate through the mucus that protects mucous membranes and thus can resist their removal by epithelial cell turnover and digestive processes [78]. It has been proposed that this SFB property can indirectly facilitate the transformation of Th17 cells from defensive to pathogenic. Importantly, Th17 cells play their role in the mucosal defense against pathogens not only in the gut or at the skin level, but also in the lung [79,80]. Figure 2 shows the main pro-inflammatory activity of Th17 cells.

7. Th17 and Viral Infections

7.1. Animal Models

Th17 cells play a key role in defense against viruses. In this regard, IL-17 has been shown to play a key role in H5N1 influenza virus infection. In experimental studies, mice double-knockout for the il17 gene have shown great susceptibility to the infection and reduced survival when compared with wild-type mice [81]. Moreover, the transfer of specific Th17 cells significantly increased respiratory parameters in H5N1-infected IL-10 deficient mice [82]. Some experimental studies have shown that IL-17A is also important for herpes simplex virus (HSV) infection [83,84]. In viral myocarditis, Th17 cells were shown to mediate both immunopathology and protection. In this regard, it has been shown that, in experimental myocarditis due to Coxsackievirus B3 (CVB3) infection, there is an increase in the number of Th17 cell during the acute phase of the disease which paradoxically contributes to viral replication [85]. Downregulation of Th17 cell activity resulted in a decrease in the disease severity [86,87]. Similarly, in experimental Dengue virus (DENV) infection, high IL-17A levels are related to a bad prognosis of the disease [88].

7.2. Human Studies

In humans, recovery from recurrent herpes labialis is associated with an increased Th17/Treg ratio in peripheral blood [89]. On the other hand, Th17 cells have been shown to play a key role in amplifying liver inflammation during chronic hepatitis B [90,91] and C [92,93]. In congenital Zika syndrome (CZS), which can follow Zika Virus (ZIKV) infection, babies who died from brain inflammation showed a higher number of tissue-infiltrating Th17 cells as compared to controls died for unrelated causes [94]. Th17 cells and increased levels of IL-17 in peripheral blood have been reported in severe dengue infection in humans, suggesting a role for IL-17 in both the protection and pathogenesis of the disease [95,96]. In human papilloma virus (HPV) infection, specific Th17 cells infiltrate the cervical tissue in an attempt to clear the virus. However, if neoplastic transformation occurs following infection by oncogenic strains of HPV and uterine cervical cancer (UCC) develops, the pro-inflammatory activity of Th17 may lead to tumor progression by promoting angiogenesis and metastatic spread due to the destruction of the extracellular matrix [97,98]. In human immunodeficiency virus (HIV) infection, Th17 cells represent a susceptible target of the virus. Their permissiveness to infection is higher than that of Th1 and Th2 cells, as evidenced by the high level of viral DNA capable of integrating into the nucleus of Th17 cells [99]. In addition to easily entering Th17 cells due to the presence of the numerous membrane co-receptors sa4b7, CCr5, and CXCR4 [100], HIV increases in the late stages of infection the phosphorylation and expression of the target of rapamycin complex (mTOR), with a consequent increase in its replication in Th17 cells [101]. This eventually leads to an early depletion of TH 17 cells both in the peripheral blood and at the mucosal sites [102,103,104]. Interestingly, the portion of mucosal TH 17 sites is not rapidly restored following antiviral therapy, leading to severe impairment of intestinal barrier function [102,105]. The facilitated penetration of bacteria and fungi through the mucosa induces systemic immune activation and tissue inflammation, further promoting HIV replication [106,107]. Infected Th17 cells are also a critical cell compartment of the HIV reservoir. It has been reported that although Th17 cells represent only 6.2% of all CD4+ T cells, they account for approximately 18% of all HIV-infected T cells [104].
Recently, the role of Th17 cells has been investigated in Coronavirus Disease 2019 (COVID-19). Th17 cells have been identified in the lung of patients with COVID-19 even after viral clearance. These cells can interact with both alveolar macrophages and CD8+ cytotoxic T cells, which, following activation, participate in the “cytokine storm” responsible of the severe form of the disease [108]. Therefore, it has been proposed to block the activity of Th17 cells with anti-interleukin-17 drugs for the therapy of severe COVID-19 [109,110]. Subsequent studies have further confirmed the role of Th17 cells in the immunopathogenesis of organ damage associated with severe COVID-19 [111,112].

8. Th17 Cells and Bacterial Infections

8.1. Animal Models

An important role in the activation of both innate and adaptive immunity is played by commensal bacteria present in the gut [113]. In this regard, it is known that germ-free adult mice lack Th17 cells at the gut level [114]. This shows that bacteria play a key role in the generation of this T-cell subpopulation [115]. An important role in the maintenance of the Th17 response by bacteria is played by so-called superantigens. It has been shown, as mentioned earlier, that SFBs play a key role in shaping Th17 cells responses. Although the process by which these bacteria can promote the generation of Th17 cells in the intestine has not been fully elucidated, several studies have shown how SFBs can induce genes coding for serum amyloid A (SAA) and the dual oxidase 2 (Duox2) enzyme by epithelial cells [78]. SSA has been shown to be able to sensitize splenic dendritic cells to produce IL-1β. SFBs are also able to induce the generation of Th17 cells through the induction of TGF-β and IL-6. On the other hand, experiments regarding the depletion of IL-17, IL-17R, and RORγt were strictly related with SFB overgrowth [116,117]. This phenomenon was explained by the fact that these bacteria are sensitive to the action of α-defensin produced by epithelial cells in response to IL-17 [116]. Stabilization of the Th17 phenotype in the intestine is further promoted by IL-23 produced by dendritic cells of the lamina propria after stimulation by SFBs [78,118]. On the other hand, the presence of bacteria that can penetrate the mucosa and persist within the intestine can promote self-reactive Th17 cell responses [78,119]. As already pointed out, Th17 cells play an important role in mucosal defense against extracellular bacteria. IL-17R-deficient mice succumb 100% following an attack by klebsiella as they cannot activate a defensive response by neutrophils [120]. Th17 cells also play a role against intracellular bacteria. In the case of M. tuberculosis, a Th17 response was found to be necessary for a defense against the primary infection [121,122]. Indeed, in the early stages, Th17 cells facilitated tissue recruitment of neutrophils, macrophages, and Th1 cells to the areas of mycobacterium penetration. Moreover, Th17 cells were found to be responsible for the development of protective Th1 cells in the later stages of infection [123]. It has been reported, however, that Th17 response can be pathological rather than protective since a correlation with pulmonary tuberculosis progression and distant dissemination of infection has been observed in experimental models [121,124]. With regard to respiratory allergic diseases, studies in animal models have shown that airway inflammation is attenuated in allergic mice double-knockout for the il17a gene. TNF-α blockade has been shown to reduce the production of both IFN-γ and IL-17, suggesting that M. catarrhalis infection is dependent on both cytokines [125]. In addition, it has been shown how H. influenzae infection can promote a Th17 response with the consequent recruitment of neutrophils to the mucous membranes of the respiratory tract [126], possibly leading to airway remodeling and an increase in mucin secretion during OVA-induced allergy sensitization. Although there is evidence that M. pneumoniae infection may also be a trigger for asthma, the exact mechanism is still unclear. However, some studies have found that IL-17A in the lungs is markedly elevated in mice after infection with this pathogen. Experiments in vitro have found that the levels of IL-17A in the culture medium increased with increasing M. pneumoniae concentration [127]. Therefore, there is evidence that the IL-23/Th17 axis can be critical for asthma onset in the course of M. pneumoniae infection. In support of this hypothesis, mice infected with M. pneumoniae exhibited increased IL-23 secretion by alveolar macrophages, and increased levels of IL-17A and IL-17F as well. After the pharmacological neutralization of IL-23, the production of IL-17A and IL-17F was blocked resulting in decreased neutrophil recruitment in the lungs [128]. It was also hypothesized that M. pneumoniae infection could activate IL-6/STAT3 promoting Th17 cell differentiation and cytokine secretion with the consequent development of asthma [129]. Finally, it has been shown that mice lacking IL-17R are more sensitive to P. gingivalis-induced bone loss, demonstrating a protective role of IL-17 in bone homeostasis [130].

8.2. Human Studies

It has been observed that the superantigen Toxic Shock Syndrome Toxin-1 (TSST-1) produced by Staphylococcus aureus can stimulate autoreactive T cells in an antigen-independent manner [131]. Induction of interleukin 17 production by Th17 cells has been observed during S. aureus infection in response to enterotoxin B (SEB). In this regard, it is interesting to note that Toll-like receptors (TLRs) receptors expressed by cells of the innate immune system play an important role in defense against S. aureus after activation by pathogen-associated molecular patterns (PAMPs). The stimulation of TLR2 promotes a Th17-mediated reaction with possible onset of granulomatosis with polyangiitis [132]. Reduced Firmicutes:Bacteroidetes ratio in the microbiota has been associated with increased Th17 cell activity in patients with systemic lupus erythematosus (SLE) [133]. Gut dysbiosis has been also associated with other autoimmune diseases, including inflammatory bowel disease [134], multiple sclerosis [135], rheumatoid arthritis [136], and myasthenia gravis [137]. A particularly important role is played by Th17 cells in the genesis of chronic inflammatory bowel disease (IBD). In such patients, the percentage of Th17 cells is significantly increased in both blood and intestinal mucosa. However, in the case of abnormal activation of Th17 cells, increased levels of IL-17, IL-21 21, and IL-23 are detected with possible induction of IBDs [138]. Under such conditions, increased stimulation of fibroblasts and epithelial damage through the production of metalloproteinase were observed [139]. The close correlation between gut bacterial flora and the protective or immuno-pathogenic function of Th17 cells led to the hypothesis that manipulation of the gut microbiota could have a therapeutic effect on IBDs [140]. It has been shown in this regard that sterilization of the microbiota by antibiotic therapy can significantly reduce the differentiation of Th17 cells, and this can be restored after fecal transplantation [141]. Some studies have also shown that the use of probiotics that restore the homeostasis of intestinal bacterial flora can inhibit the proinflammatory effect of Th17 cells by improving the IBD progression [142,143]. In another study, the use of polysaccharide A derived from Bacteroides fragilis resulted in the amelioration of such inflammatory diseases by suppressing the pro-inflammatory response of intestinal Th17 cells through the stimulation of TLR2 [144]. Other studies have shown how gut barrier dysfunction contributes to worsening disease in patients with advanced cirrhosis [145,146]. Moreover, alterations in intestinal bacterial flora with increased Th17 activity were found to play an important role in the pathogenesis of inflammatory rheumatic diseases, including rheumatoid arthritis [147] and HLA-B27-associated spondylarthritis [148].
Th17 cells play also an important role in the genesis of many respiratory diseases. In particular, these T cells have been implicated in the occurrence of asthma. Several studies have shown how children who were infected perinatally with pathogens, such as H. influenzae, S. pneumoniae, and M. catarrhalis, have an increased risk of developing bronchial asthma [149].
S. pneumoniae is a pathogen capable of causing community-acquired pneumonia. Perinatal infection with this pathogen is closely related to the subsequent development of asthma. Several studies show that S. Pneumoniae infection is associated with increased numbers of Th17 cells in the airways with concomitant overproduction of interleukin 17A [150,151]. M. catarrhalis is another mucosal pathogen that causes respiratory illness in children. Infection by this pathogen has been found to increase the risk of asthma in both infants and adults. It has been also observed that IL-17A levels are significantly increased in the lungs of dust mite-allergic mice infected with M. catarrhalis.
M. pneumoniae is an airway pathogen that mainly adheres to the surface of the respiratory tract. M. pneumoniae infection can cause chronic inflammation of the lower airways by impairing ciliary clearance and increasing mucus secretion. Interestingly, it has been shown that a significant percentage of subjects with refractory asthma are infected with this pathogen [152,153]. Th17 cells were also found at the level of gingival tissues [130]. Although IL-17 may have protective functions in the oral cavity, several studies indicate that its excessive production is associated with periodontitis [154]. In this regard, it has been shown that IL-17 is responsible for pro-inflammatory activities by inducing cytokines via target cells and recruiting inflammatory cells such as neutrophils in the oral cavity. In addition, IL-17 facilitates the access of these cells to tissues through the regulation of chemokine ligand expression and granulocyte-macrophage colony-stimulating factors (GM-CSF). Upregulation of IL-17 can lead to the overactivation and mobilization of macrophages eventually leading to oral tissue damage [155]. In addition, IL-17 acts synergistically with other inflammatory cytokines to increase chemokine production by human gingival fibroblasts. This stimulates further recruitment of Th17 cells and, consequently, IL-17 production in inflamed periodontal tissues. A significant increase in IL-17 levels has been found not only in the bone and gingiva of periodontitis patients, but also in serum [156]. Expression levels of the RORC encoding gene have been found to be increased in patients with periodontitis [157].

9. Th17 Cells and Fungal Infections

9.1. Animal Models

Th17 cells have been proven to be critical in defense against fungi and in particular against Candida albicans, a mucocutaneous fungal pathobiont [158]. Animals made deficient for the expression of IL-17, IL-17R, or RORγt proved particularly susceptible to Candida albicans infection [75]. However, repeated fungal infections have been shown to activate Th17 cells and induce immunopathology. With regard to other fungal infections, it has been shown that A. alternata can induce a Th17 response in the lungs, activating neutrophils by β-glucan, which is able in turn to induce Th17 cells by binding Dectin-1. This promotes the secretion of several cytokines with consequent amplification of the inflammatory response [159,160,161].

9.2. Human Studies

Although fungus-specific Th17 cells are present in peripheral blood in almost all healthy subjects [162], it has been suggested that the role of fungi in inducing immunopathology is significantly underestimated probably due to a lack of adequate diagnostic tools [163,164]. C. albicans in particular is the most potent pathogen in humans capable of stimulating a Th17 response even more than S. aureus [51,165]. Such a response is induced by the peptide toxin Candidalysin. This protein not only protects against fungal invasion, but is also capable of damaging the intestinal epithelium [166,167]. Moreover, it has recently been demonstrated that A. fumigatus-specific T cells re-stimulated with C. albicans could acquire a Th17 phenotype, although these cells have been proven not to be essential for defense against this fungus [158,162]. It was also observed that patients with lung diseases are more easily colonized by A. fumigatus [168,169] and that such patients show more severe forms of pulmonary disease when the lung is infiltrated by Th17 cells [170,171,172]. This demonstrates the immunopathological activity of this T-cell subset in the lung. Therefore, it can be speculated that Th17 cells present at the mucosal level where they are protective against C. albicans can generate pathogenic A. fumigatus-specific through a heterologous cross-reactive immunologic response [173]. These findings also have therapeutic implications. Indeed, the overuse of antibiotic therapy results in increased intestinal colonization by C. albicans [174,175]. This has been correlated with pathology at sites far from the gut including the lung [176]. Therefore, preservation of the microbiota may be a key factor in counteracting the immunopathologic remote effects mediated by cross-reactive Th17 cells. Figure 3 represents the dual effect of the interaction of Th17 cells with pathogens.

10. Conclusions

Several factors can transform Th17 cells from an effective defensive arm against infectious pathogens into harmful self-reactive effectors. The pathogens themselves play a key role in this conversion. It is quite clear that gut commensal bacteria in the microbiota play a key role. However, as discussed in the previous sections, viruses and fungi can also induce Th17 cells to acquire a pathogenic phenotype. Th17 cells rendered pathogenic after interaction with various microbial agents are involved in the genesis of several autoimmune rheumatic diseases, such as rheumatoid arthritis, systemic lupus erythematosus, myasthenia gravis, and vasculitis, but this list is not certainly exhaustive. They also play a crucial role in psoriasis. An important role of Th17s in promoting the genesis of various neoplasms is also beginning to emerge [177]. In the case of HIV infection, moreover, Th17 cells perform a peculiar function by constituting a long-term HIV reservoir within the body. Quite recently Th17 cells are critical in the genesis of the “cytokine storm” characteristic of severe COVID-19. Luckily, after the identification of the pathogenetic role of Th17 cells, powerful therapeutic tools, such as monoclonal antibodies blocking the IL 23-IL 17 axis, have been developed. Future studies are needed to further elucidate the mechanisms by which Th17 cells can cause tissue damage following interaction with infectious agents and how to therapeutically modulate the activity of this cell subpopulation to be beneficial but not harmful to the host.

Author Contributions

Conceptualization, M.P., R.C. and M.P.P.; methodology, M.P., R.C., L.S. and M.P.P.; software, V.C.; validation, M.P., R.C., L.S. and M.P.P.; formal analysis, M.P.; investigation, M.P., R.C. M.T.F., V.C. and M.P.P.; resources, M.P. and R.C.; data curation, M.P., R.C., M.T.F., L.S., S.G., V.C. and M.P.P.; writing—original draft preparation, M.P.; writing—review and editing, M.P. and M.P.P.; visualization, M.P. and R.C.; supervision, M.P., R.C. and S.G.; project administration, M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harrington, L.E.; Hatton, R.D.; Mangan, P.R.; Turner, H.; Murphy, T.L.; Murphy, K.M.; Weaver, C.T. 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, 1123–1132. [Google Scholar] [CrossRef] [PubMed]
  2. Park, H.; Li, Z.; Yang, X.O.; Chang, S.H.; Nurieva, R.; Wang, Y.H.; Wang, Y.; Hood, L.; Zhu, Z.; Tian, Q.; et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat. Immunol. 2005, 6, 1133–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Acosta-Rodriguez, E.V.; 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. Nat. Immunol. 2007, 8, 639–646. [Google Scholar] [CrossRef]
  4. Annunziato, F.; Cosmi, L.; Santarlasci, V.; Maggi, L.; Liotta, F.; Mazzinghi, B.; Parente, E.; Fili, L.; Ferri, S.; Frosali, F.; et al. Phenotypic and functional features of human Th17 cells. J. Exp. Med. 2007, 204, 1849–1861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gaffen, S.L.; Jain, R.; Garg, A.V.; Cua, D.J. The IL-23-IL-17 immune axis: From mechanisms to therapeutic testing. Nat. Rev. Immunol. 2014, 14, 585–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Capone, A.; Volpe, E. Transcriptional Regulators of T Helper 17 Cell Differentiation in Health and Autoimmune Diseases. Front. Immunol. 2020, 11, 348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Korn, T.; Bettelli, E.; Oukka, M.; Kuchroo, V.K. IL-17 and Th17 Cells. Annu. Rev. Immunol. 2009, 27, 485–517. [Google Scholar] [CrossRef]
  8. Toussirot, E. The IL23/Th17 pathway as a therapeutic target in chronic inflammatory diseases. Inflamm. Allergy Drug Targets 2012, 11, 159–168. [Google Scholar] [CrossRef]
  9. Paroli, M.; Spadea, L.; Caccavale, R.; Spadea, L.; Paroli, M.P.; Nante, N. The Role of Interleukin-17 in Juvenile Idiopathic Arthritis: From Pathogenesis to Treatment. Medicina 2022, 58, 1552. [Google Scholar] [CrossRef]
  10. Chimenti, M.S.; Fonti, G.L.; Conigliaro, P.; Sunzini, F.; Scrivo, R.; Navarini, L.; Triggianese, P.; Peluso, G.; Scolieri, P.; Caccavale, R.; et al. One-year effectiveness, retention rate, and safety of secukinumab in ankylosing spondylitis and psoriatic arthritis: A real-life multicenter study. Expert Opin. Biol. Ther. 2020, 20, 813–821. [Google Scholar] [CrossRef]
  11. Mosmann, T.R.; Cherwinski, H.; Bond, M.W.; Giedlin, M.A.; Coffman, R.L. 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. [Google Scholar] [PubMed]
  12. Cher, D.J.; Mosmann, T.R. Two types of murine helper T cell clone. II. Delayed-type hypersensitivity is mediated by TH1 clones. J. Immunol. 1987, 138, 3688–3694. [Google Scholar] [PubMed]
  13. Coffman, R.L.; Carty, J. A T cell activity that enhances polyclonal IgE production and its inhibition by interferon-gamma. J. Immunol. 1986, 136, 949–954. [Google Scholar] [PubMed]
  14. Hu-Li, J.; Shevach, E.M.; Mizuguchi, J.; Ohara, J.; Mosmann, T.; Paul, W.E. B cell stimulatory factor 1 (interleukin 4) is a potent costimulant for normal resting T lymphocytes. J. Exp. Med. 1987, 165, 157–172. [Google Scholar] [CrossRef] [PubMed]
  15. Mosmann, T.R.; Coffman, R.L. TH1 and TH2 cells: Different patterns of lymphokine secretion lead to different functional properties. Annu. Rev. Immunol. 1989, 7, 145–173. [Google Scholar] [CrossRef]
  16. Krakowski, M.; Owens, T. Interferon-gamma confers resistance to experimental allergic encephalomyelitis. Eur. J. Immunol. 1996, 26, 1641–1646. [Google Scholar] [CrossRef]
  17. Bettelli, E.; Sullivan, B.; Szabo, S.J.; Sobel, R.A.; Glimcher, L.H.; Kuchroo, V.K. Loss of T-bet, but not STAT1, prevents the development of experimental autoimmune encephalomyelitis. J. Exp. Med. 2004, 200, 79–87. [Google Scholar] [CrossRef] [Green Version]
  18. Duong, T.T.; Finkelman, F.D.; Singh, B.; Strejan, G.H. Effect of anti-interferon-gamma monoclonal antibody treatment on the development of experimental allergic encephalomyelitis in resistant mouse strains. J. Neuroimmunol. 1994, 53, 101–107. [Google Scholar] [CrossRef]
  19. Rouvier, E.; Luciani, M.F.; Mattei, M.G.; 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, 5445–5456. [Google Scholar]
  20. Yao, Z.; Painter, S.L.; Fanslow, W.C.; Ulrich, D.; Macduff, B.M.; Spriggs, M.K.; Armitage, R.J. Human IL-17: A novel cytokine derived from T cells. J. Immunol. 1995, 155, 5483–5486. [Google Scholar]
  21. Gaffen, S.L. Structure and signalling in the IL-17 receptor family. Nat. Rev. Immunol. 2009, 9, 556–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Yao, Z.; Fanslow, W.C.; Seldin, M.F.; Rousseau, A.M.; Painter, S.L.; Comeau, M.R.; Cohen, J.I.; Spriggs, M.K. Herpesvirus saimiri encodes a new cytokine, IL-17, which binds to a novel cytokine receptor. Immunity 1995, 3, 811–821. [Google Scholar] [CrossRef] [PubMed]
  23. Hymowitz, S.G.; Filvaroff, E.H.; Yin, J.P.; Lee, J.; Cai, L.; Risser, P.; Maruoka, M.; Mao, W.; Foster, J.; Kelley, R.F.; 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, 5332–5341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Buckley, K.M.; Ho, E.C.H.; Hibino, T.; Schrankel, C.S.; Schuh, N.W.; Wang, G.; Rast, J.P. IL17 factors are early regulators in the gut epithelium during inflammatory response to Vibrio in the sea urchin larva. Elife 2017, 6, e23481. [Google Scholar]
  25. Han, Q.; Das, S.; Hirano, M.; Holland, S.J.; McCurley, N.; Guo, P.; Rosenberg, C.S.; Boehm, T.; Cooper, M.D. Characterization of Lamprey IL-17 Family Members and Their Receptors. J. Immunol. 2015, 195, 5440–5451. [Google Scholar] [CrossRef] [Green Version]
  26. Novatchkova, M.; Leibbrandt, A.; Werzowa, J.; Neubuser, A.; Eisenhaber, F. The STIR-domain superfamily in signal transduction, development and immunity. Trends Biochem. Sci. 2003, 28, 226–229. [Google Scholar] [CrossRef]
  27. Iwakura, Y.; Ishigame, H.; Saijo, S.; Nakae, S. Functional specialization of interleukin-17 family members. Immunity 2011, 34, 149–162. [Google Scholar] [CrossRef] [Green Version]
  28. McGeachy, M.J.; Cua, D.J.; Gaffen, S.L. The IL-17 Family of Cytokines in Health and Disease. Immunity 2019, 50, 892–906. [Google Scholar] [CrossRef]
  29. Goepfert, A.; Lehmann, S.; Blank, J.; Kolbinger, F.; Rondeau, J.M. Structural Analysis Reveals that the Cytokine IL-17F Forms a Homodimeric Complex with Receptor IL-17RC to Drive IL-17RA-Independent Signaling. Immunity 2020, 52, 499–512.e495. [Google Scholar] [CrossRef]
  30. Su, Y.; Huang, J.; Zhao, X.; Lu, H.; Wang, W.; Yang, X.O.; Shi, Y.; Wang, X.; Lai, Y.; Dong, C. Interleukin-17 receptor D constitutes an alternative receptor for interleukin-17A important in psoriasis-like skin inflammation. Sci. Immunol. 2019, 4, eaau9657. [Google Scholar] [CrossRef]
  31. Reynolds, J.M.; Lee, Y.H.; Shi, Y.; Wang, X.; Angkasekwinai, P.; Nallaparaju, K.C.; Flaherty, S.; Chang, S.H.; Watarai, H.; Dong, C. Interleukin-17B Antagonizes Interleukin-25-Mediated Mucosal Inflammation. Immunity 2015, 42, 692–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Huang, J.; Lee, H.Y.; Zhao, X.; Han, J.; Su, Y.; Sun, Q.; Shao, J.; Ge, J.; Zhao, Y.; Bai, X.; et al. Interleukin-17D regulates group 3 innate lymphoid cell function through its receptor CD93. Immunity 2021, 54, 673–686.e674. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, C.; Swaidani, S.; Qian, W.; Kang, Z.; Sun, P.; Han, Y.; Wang, C.; Gulen, M.F.; Yin, W.; Zhang, C.; et al. A CC’ loop decoy peptide blocks the interaction between Act1 and IL-17RA to attenuate IL-17- and IL-25-induced inflammation. Sci. Signal. 2011, 4, ra72. [Google Scholar] [CrossRef] [PubMed]
  34. Qian, Y.; Liu, C.; Hartupee, J.; Altuntas, C.Z.; Gulen, M.F.; Jane-Wit, D.; Xiao, J.; Lu, Y.; Giltiay, N.; Liu, J.; et al. The adaptor Act1 is required for interleukin 17-dependent signaling associated with autoimmune and inflammatory disease. Nat. Immunol. 2007, 8, 247–256. [Google Scholar] [CrossRef] [PubMed]
  35. Sonder, S.U.; Saret, S.; Tang, W.; Sturdevant, D.E.; Porcella, S.F.; Siebenlist, U. IL-17-induced NF-kappaB activation via CIKS/Act1: Physiologic significance and signaling mechanisms. J. Biol. Chem. 2011, 286, 12881–12890. [Google Scholar] [CrossRef] [Green Version]
  36. Conti, H.R.; Gaffen, S.L. IL-17-Mediated Immunity to the Opportunistic Fungal Pathogen Candida albicans. J. Immunol. 2015, 195, 780–788. [Google Scholar] [CrossRef] [Green Version]
  37. Liu, C.; Qian, W.; Qian, Y.; Giltiay, N.V.; Lu, Y.; Swaidani, S.; Misra, S.; Deng, L.; Chen, Z.J.; Li, X. Act1, a U-box E3 ubiquitin ligase for IL-17 signaling. Sci. Signal. 2009, 2, ra63. [Google Scholar] [CrossRef] [Green Version]
  38. Schwandner, R.; Yamaguchi, K.; Cao, Z. Requirement of tumor necrosis factor receptor-associated factor (TRAF)6 in interleukin 17 signal transduction. J. Exp. Med. 2000, 191, 1233–1240. [Google Scholar] [CrossRef] [Green Version]
  39. Karlsen, J.R.; Borregaard, N.; Cowland, J.B. Induction of neutrophil gelatinase-associated lipocalin expression by co-stimulation with interleukin-17 and tumor necrosis factor-alpha is controlled by IkappaB-zeta but neither by C/EBP-beta nor C/EBP-delta. J. Biol. Chem. 2010, 285, 14088–14100. [Google Scholar] [CrossRef] [Green Version]
  40. Ruddy, M.J.; Wong, G.C.; Liu, X.K.; Yamamoto, H.; Kasayama, S.; Kirkwood, K.L.; Gaffen, S.L. Functional cooperation between interleukin-17 and tumor necrosis factor-alpha is mediated by CCAAT/enhancer-binding protein family members. J. Biol. Chem. 2004, 279, 2559–2567. [Google Scholar] [CrossRef] [Green Version]
  41. Tohyama, M.; Shirakata, Y.; Hanakawa, Y.; Dai, X.; Shiraishi, K.; Murakami, M.; Miyawaki, S.; Mori, H.; Utsunomiya, R.; Masuda, K.; et al. Bcl-3 induced by IL-22 via STAT3 activation acts as a potentiator of psoriasis-related gene expression in epidermal keratinocytes. Eur. J. Immunol. 2018, 48, 168–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Oppmann, B.; Lesley, R.; Blom, B.; Timans, J.C.; Xu, Y.; Hunte, B.; Vega, F.; Yu, N.; Wang, J.; Singh, K.; et al. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity 2000, 13, 715–725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Chyuan, I.T.; Lai, J.H. New insights into the IL-12 and IL-23: From a molecular basis to clinical application in immune-mediated inflammation and cancers. Biochem. Pharmacol. 2020, 175, 113928. [Google Scholar] [CrossRef] [PubMed]
  44. Schnurr, M.; Toy, T.; Shin, A.; Wagner, M.; Cebon, J.; Maraskovsky, E. Extracellular nucleotide signaling by P2 receptors inhibits IL-12 and enhances IL-23 expression in human dendritic cells: A novel role for the cAMP pathway. Blood 2005, 105, 1582–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sheibanie, A.F.; Tadmori, I.; Jing, H.; Vassiliou, E.; Ganea, D. Prostaglandin E2 induces IL-23 production in bone marrow-derived dendritic cells. FASEB J. 2004, 18, 1318–1320. [Google Scholar] [CrossRef]
  46. Aggarwal, S.; Ghilardi, N.; Xie, M.H.; de Sauvage, F.J.; Gurney, A.L. Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17. J. Biol. Chem. 2003, 278, 1910–1914. [Google Scholar] [CrossRef] [Green Version]
  47. Cua, D.J.; Sherlock, J.; Chen, Y.; Murphy, C.A.; Joyce, B.; Seymour, B.; Lucian, L.; To, W.; Kwan, S.; Churakova, T.; et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature 2003, 421, 744–748. [Google Scholar] [CrossRef] [PubMed]
  48. Ivanov, I.I.; McKenzie, B.S.; Zhou, L.; Tadokoro, C.E.; Lepelley, A.; Lafaille, J.J.; Cua, D.J.; Littman, D.R. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006, 126, 1121–1133. [Google Scholar] [CrossRef] [Green Version]
  49. Unutmaz, D. RORC2: The master of human Th17 cell programming. Eur. J. Immunol. 2009, 39, 1452–1455. [Google Scholar] [CrossRef]
  50. Yang, X.O.; Pappu, B.P.; Nurieva, R.; Akimzhanov, A.; Kang, H.S.; Chung, Y.; Ma, L.; Shah, B.; Panopoulos, A.D.; Schluns, K.S.; et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity 2008, 28, 29–39. [Google Scholar] [CrossRef] [Green Version]
  51. Zielinski, C.E.; Mele, F.; Aschenbrenner, D.; Jarrossay, D.; Ronchi, F.; Gattorno, M.; Monticelli, S.; Lanzavecchia, A.; Sallusto, F. Pathogen-induced human TH17 cells produce IFN-gamma or IL-10 and are regulated by IL-1beta. Nature 2012, 484, 514–518. [Google Scholar] [CrossRef] [PubMed]
  52. Duhen, T.; Campbell, D.J. IL-1beta promotes the differentiation of polyfunctional human CCR6+CXCR3+ Th1/17 cells that are specific for pathogenic and commensal microbes. J. Immunol. 2014, 193, 120–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Veldhoen, M.; Hirota, K.; Westendorf, A.M.; Buer, J.; Dumoutier, L.; Renauld, J.C.; Stockinger, B. The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 2008, 453, 106–109. [Google Scholar] [CrossRef]
  54. Akimzhanov, A.M.; Yang, X.O.; Dong, C. Chromatin remodeling of interleukin-17 (IL-17)-IL-17F cytokine gene locus during inflammatory helper T cell differentiation. J. Biol. Chem. 2007, 282, 5969–5972. [Google Scholar] [CrossRef] [Green Version]
  55. Huber, M.; Brustle, A.; Reinhard, K.; Guralnik, A.; Walter, G.; Mahiny, A.; von Low, E.; Lohoff, M. IRF4 is essential for IL-21-mediated induction, amplification, and stabilization of the Th17 phenotype. Proc. Natl. Acad. Sci. USA 2008, 105, 20846–20851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Chung, Y.; Chang, S.H.; Martinez, G.J.; Yang, X.O.; Nurieva, R.; Kang, H.S.; Ma, L.; Watowich, S.S.; Jetten, A.M.; Tian, Q.; et al. Critical regulation of early Th17 cell differentiation by interleukin-1 signaling. Immunity 2009, 30, 576–587. [Google Scholar] [CrossRef] [Green Version]
  57. Ikeda, S.; Saijo, S.; Murayama, M.A.; Shimizu, K.; Akitsu, A.; Iwakura, Y. Excess IL-1 signaling enhances the development of Th17 cells by downregulating TGF-beta-induced Foxp3 expression. J. Immunol. 2014, 192, 1449–1458. [Google Scholar] [CrossRef] [Green Version]
  58. Mailer, R.K.; Joly, A.L.; Liu, S.; Elias, S.; Tegner, J.; Andersson, J. IL-1beta promotes Th17 differentiation by inducing alternative splicing of FOXP3. Sci. Rep. 2015, 5, 14674. [Google Scholar] [CrossRef] [Green Version]
  59. Gooderham, M.J.; Papp, K.A.; Lynde, C.W. Shifting the focus—The primary role of IL-23 in psoriasis and other inflammatory disorders. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 1111–1119. [Google Scholar] [CrossRef] [Green Version]
  60. Zhu, J.; Yamane, H.; Paul, W.E. Differentiation of effector CD4 T cell populations. Annu. Rev. Immunol. 2010, 28, 445–489. [Google Scholar] [CrossRef] [Green Version]
  61. Rivino, L.; Messi, M.; Jarrossay, D.; Lanzavecchia, A.; Sallusto, F.; Geginat, J. Chemokine receptor expression identifies Pre-T helper (Th)1, Pre-Th2, and nonpolarized cells among human CD4+ central memory T cells. J. Exp. Med. 2004, 200, 725–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Abromson-Leeman, S.; Bronson, R.T.; Dorf, M.E. Encephalitogenic T cells that stably express both T-bet and ROR gamma t consistently produce IFNgamma but have a spectrum of IL-17 profiles. J. Neuroimmunol. 2009, 215, 10–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Boniface, K.; Blumenschein, W.M.; Brovont-Porth, K.; McGeachy, M.J.; Basham, B.; Desai, B.; Pierce, R.; McClanahan, T.K.; Sadekova, S.; de Waal Malefyt, R. Human Th17 cells comprise heterogeneous subsets including IFN-gamma-producing cells with distinct properties from the Th1 lineage. J. Immunol. 2010, 185, 679–687. [Google Scholar] [CrossRef] [PubMed]
  64. Cosmi, L.; Maggi, L.; Santarlasci, V.; Capone, M.; Cardilicchia, E.; Frosali, F.; Querci, V.; Angeli, R.; Matucci, A.; Fambrini, M.; et al. Identification of a novel subset of human circulating memory CD4(+) T cells that produce both IL-17A and IL-4. J. Allergy. Clin. Immunol. 2010, 125, 222–230.e4. [Google Scholar] [CrossRef]
  65. Hirota, K.; Turner, J.E.; Villa, M.; Duarte, J.H.; Demengeot, J.; Steinmetz, O.M.; Stockinger, B. Plasticity of Th17 cells in Peyer’s patches is responsible for the induction of T cell-dependent IgA responses. Nat. Immunol. 2013, 14, 372–379. [Google Scholar] [CrossRef]
  66. Koenen, H.J.; Smeets, R.L.; Vink, P.M.; van Rijssen, E.; Boots, A.M.; Joosten, I. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17-producing cells. Blood 2008, 112, 2340–2352. [Google Scholar] [CrossRef] [Green Version]
  67. Valmori, D.; Raffin, C.; Raimbaud, I.; Ayyoub, M. Human RORgammat+ TH17 cells preferentially differentiate from naive FOXP3+Treg in the presence of lineage-specific polarizing factors. Proc. Natl. Acad. Sci. USA 2010, 107, 19402–19407. [Google Scholar] [CrossRef] [Green Version]
  68. Hoechst, B.; Gamrekelashvili, J.; Manns, M.P.; Greten, T.F.; Korangy, F. Plasticity of human Th17 cells and iTregs is orchestrated by different subsets of myeloid cells. Blood 2011, 117, 6532–6541. [Google Scholar] [CrossRef]
  69. Wacleche, V.S.; Landay, A.; Routy, J.P.; Ancuta, P. The Th17 Lineage: From Barrier Surfaces Homeostasis to Autoimmunity, Cancer, and HIV-1 Pathogenesis. Viruses 2017, 9, 303. [Google Scholar] [CrossRef]
  70. Wong, M.T.; Ong, D.E.; Lim, F.S.; Teng, K.W.; McGovern, N.; Narayanan, S.; Ho, W.Q.; Cerny, D.; Tan, H.K.; Anicete, R.; et al. A High-Dimensional Atlas of Human T Cell Diversity Reveals Tissue-Specific Trafficking and Cytokine Signatures. Immunity 2016, 45, 442–456. [Google Scholar] [CrossRef] [Green Version]
  71. Naik, S.; Bouladoux, N.; Wilhelm, C.; Molloy, M.J.; Salcedo, R.; Kastenmuller, W.; Deming, C.; Quinones, M.; Koo, L.; Conlan, S.; et al. Compartmentalized control of skin immunity by resident commensals. Science 2012, 337, 1115–1119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Shaw, M.H.; Kamada, N.; Kim, Y.G.; Nunez, G. Microbiota-induced IL-1beta, but not IL-6, is critical for the development of steady-state TH17 cells in the intestine. J. Exp. Med. 2012, 209, 251–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Stockinger, B.; Omenetti, S. The dichotomous nature of T helper 17 cells. Nat. Rev. Immunol. 2017, 17, 535–544. [Google Scholar] [CrossRef] [PubMed]
  74. Zheng, Y.; Valdez, P.A.; Danilenko, D.M.; Hu, Y.; Sa, S.M.; Gong, Q.; Abbas, A.R.; Modrusan, Z.; Ghilardi, N.; de Sauvage, F.J.; et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat. Med. 2008, 14, 282–289. [Google Scholar] [CrossRef] [PubMed]
  75. Puel, A.; Cypowyj, S.; Bustamante, J.; Wright, J.F.; Liu, L.; Lim, H.K.; Migaud, M.; Israel, L.; Chrabieh, M.; Audry, M.; et al. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science 2011, 332, 65–68. [Google Scholar] [CrossRef] [Green Version]
  76. Hernandez-Santos, N.; Huppler, A.R.; Peterson, A.C.; Khader, S.A.; McKenna, K.C.; Gaffen, S.L. Th17 cells confer long-term adaptive immunity to oral mucosal Candida albicans infections. Mucosal Immunol. 2013, 6, 900–910. [Google Scholar] [CrossRef] [Green Version]
  77. Wu, H.J.; Ivanov, I.I.; Darce, J.; Hattori, K.; Shima, T.; Umesaki, Y.; Littman, D.R.; Benoist, C.; Mathis, D. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 2010, 32, 815–827. [Google Scholar] [CrossRef] [Green Version]
  78. Atarashi, K.; Tanoue, T.; Ando, M.; Kamada, N.; Nagano, Y.; Narushima, S.; Suda, W.; Imaoka, A.; Setoyama, H.; Nagamori, T.; et al. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell 2015, 163, 367–380. [Google Scholar] [CrossRef] [Green Version]
  79. Bystrom, J.; Al-Adhoubi, N.; Al-Bogami, M.; Jawad, A.S.; Mageed, R.A. Th17 lymphocytes in respiratory syncytial virus infection. Viruses 2013, 5, 777–791. [Google Scholar] [CrossRef] [Green Version]
  80. Chen, K.; Kolls, J.K. T cell-mediated host immune defenses in the lung. Annu. Rev. Immunol. 2013, 31, 605–633. [Google Scholar] [CrossRef] [Green Version]
  81. Wang, X.; Chan, C.C.; Yang, M.; Deng, J.; Poon, V.K.; Leung, V.H.; Ko, K.H.; Zhou, J.; Yuen, K.Y.; Zheng, B.J.; et al. A critical role of IL-17 in modulating the B-cell response during H5N1 influenza virus infection. Cell Mol. Immunol. 2011, 8, 462–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. McKinstry, K.K.; Strutt, T.M.; Buck, A.; Curtis, J.D.; Dibble, J.P.; Huston, G.; Tighe, M.; Hamada, H.; Sell, S.; Dutton, R.W.; et al. IL-10 deficiency unleashes an influenza-specific Th17 response and enhances survival against high-dose challenge. J. Immunol. 2009, 182, 7353–7363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Anipindi, V.C.; Bagri, P.; Roth, K.; Dizzell, S.E.; Nguyen, P.V.; Shaler, C.R.; Chu, D.K.; Jimenez-Saiz, R.; Liang, H.; Swift, S.; et al. Estradiol Enhances CD4+ T-Cell Anti-Viral Immunity by Priming Vaginal DCs to Induce Th17 Responses via an IL-1-Dependent Pathway. PLoS Pathog. 2016, 12, e1005589. [Google Scholar]
  84. Bagri, P.; Anipindi, V.C.; Nguyen, P.V.; Vitali, D.; Stampfli, M.R.; Kaushic, C. Novel Role for Interleukin-17 in Enhancing Type 1 Helper T Cell Immunity in the Female Genital Tract following Mucosal Herpes Simplex Virus 2 Vaccination. J. Virol. 2017, 91, e01234-17. [Google Scholar] [CrossRef]
  85. Yuan, J.; Yu, M.; Lin, Q.W.; Cao, A.L.; Yu, X.; Dong, J.H.; Wang, J.P.; Zhang, J.H.; Wang, M.; Guo, H.P.; et al. Th17 cells contribute to viral replication in coxsackievirus B3-induced acute viral myocarditis. J. Immunol. 2010, 185, 4004–4010. [Google Scholar] [CrossRef] [Green Version]
  86. Chen, R.; Cao, Y.; Tian, Y.; Gu, Y.; Lu, H.; Zhang, S.; Xu, H.; Su, Z. PGE2 ameliorated viral myocarditis development and promoted IL-10-producing regulatory B cell expansion via MAPKs/AKT-AP1 axis or AhR signaling. Cell Immunol. 2020, 347, 104025. [Google Scholar] [CrossRef]
  87. Huang, Y.; Li, Y.; Wei, B.; Wu, W.; Gao, X. CD80 Regulates Th17 Cell Differentiation in Coxsackie Virus B3-Induced Acute Myocarditis. Inflammation 2018, 41, 232–239. [Google Scholar] [CrossRef]
  88. Guabiraba, R.; Besnard, A.G.; Marques, R.E.; Maillet, I.; Fagundes, C.T.; Conceicao, T.M.; Rust, N.M.; Charreau, S.; Paris, I.; Lecron, J.C.; et al. IL-22 modulates IL-17A production and controls inflammation and tissue damage in experimental dengue infection. Eur. J. Immunol. 2013, 43, 1529–1544. [Google Scholar] [CrossRef]
  89. Mei, X.X.; Lei, S.S.; Xu, L.; Wu, S.; Gu, H.P.; Du, Y.; Zhao, T.; Xie, G.Q.; Fan, Y.S.; Pan, X.P.; et al. Herpes simplex virus type I-infected disorders alter the balance between Treg and Th17 cells in recurrent herpes labialis patients. Int. J. Immunopathol. Pharmacol. 2020, 34, 2058738420933099. [Google Scholar] [CrossRef]
  90. Wang, Q.; Zhou, J.; Zhang, B.; Tian, Z.; Tang, J.; Zheng, Y.; Huang, Z.; Tian, Y.; Jia, Z.; Tang, Y.; et al. Hepatitis B virus induces IL-23 production in antigen presenting cells and causes liver damage via the IL-23/IL-17 axis. PLoS Pathog. 2013, 9, e1003410. [Google Scholar] [CrossRef] [Green Version]
  91. Huang, Z.; van Velkinburgh, J.C.; Ni, B.; Wu, Y. Pivotal roles of the interleukin-23/T helper 17 cell axis in hepatitis B. Liver Int. 2012, 32, 894–901. [Google Scholar] [CrossRef] [PubMed]
  92. Ashrafi Hafez, A.; Ahmadi Vasmehjani, A.; Baharlou, R.; Mousavi Nasab, S.D.; Davami, M.H.; Najafi, A.; Joharinia, N.; Rezanezhad, H.; Ahmadi, N.A.; Imanzad, M. Analytical assessment of interleukin -23 and -27 cytokines in healthy people and patients with hepatitis C virus infection (genotypes 1 and 3a). Hepat. Mon. 2014, 14, e21000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Wang, J.M.; Shi, L.; Ma, C.J.; Ji, X.J.; Ying, R.S.; Wu, X.Y.; Wang, K.S.; Li, G.; Moorman, J.P.; Yao, Z.Q. Differential regulation of interleukin-12 (IL-12)/IL-23 by Tim-3 drives T(H)17 cell development during hepatitis C virus infection. J. Virol. 2013, 87, 4372–4383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Azevedo, R.S.S.; de Sousa, J.R.; Araujo, M.T.F.; Martins Filho, A.J.; de Alcantara, B.N.; Araujo, F.M.C.; Queiroz, M.G.L.; Cruz, A.C.R.; Vasconcelos, B.H.B.; Chiang, J.O.; et al. In situ immune response and mechanisms of cell damage in central nervous system of fatal cases microcephaly by Zika virus. Sci. Rep. 2018, 8, 1–11. [Google Scholar] [CrossRef]
  95. Jain, A.; Pandey, N.; Garg, R.K.; Kumar, R. IL-17 level in patients with Dengue virus infection & its association with severity of illness. J. Clin. Immunol. 2013, 33, 613–618. [Google Scholar]
  96. Inizan, C.; O’Connor, O.; Worwor, G.; Cabemaiwai, T.; Grignon, J.C.; Girault, D.; Minier, M.; Prot, M.; Ballan, V.; Pakoa, G.J.; et al. Molecular Characterization of Dengue Type 2 Outbreak in Pacific Islands Countries and Territories, 2017–2020. Viruses 2020, 12, 1081. [Google Scholar] [CrossRef]
  97. Alves, J.J.P.; De Medeiros Fernandes, T.A.A.; De Araujo, J.M.G.; Cobucci, R.N.O.; Lanza, D.C.F.; Bezerra, F.L.; Andrade, V.S.; Fernandes, J.V. Th17 response in patients with cervical cancer. Oncol. Lett. 2018, 16, 6215–6227. [Google Scholar] [CrossRef] [Green Version]
  98. Walch-Ruckheim, B.; Mavrova, R.; Henning, M.; Vicinus, B.; Kim, Y.J.; Bohle, R.M.; Juhasz-Boss, I.; Solomayer, E.F.; Smola, S. Stromal Fibroblasts Induce CCL20 through IL6/C/EBPbeta to Support the Recruitment of Th17 Cells during Cervical Cancer Progression. Cancer Res. 2015, 75, 5248–5259. [Google Scholar] [CrossRef] [Green Version]
  99. Prendergast, A.; Prado, J.G.; Kang, Y.H.; Chen, F.; Riddell, L.A.; Luzzi, G.; Goulder, P.; Klenerman, P. HIV-1 infection is characterized by profound depletion of CD161+ Th17 cells and gradual decline in regulatory T cells. AIDS 2010, 24, 491–502. [Google Scholar] [CrossRef]
  100. Gosselin, A.; Monteiro, P.; Chomont, N.; Diaz-Griffero, F.; Said, E.A.; Fonseca, S.; Wacleche, V.; El-Far, M.; Boulassel, M.R.; Routy, J.P.; et al. Peripheral blood CCR4+CCR6+ and CXCR3+CCR6+CD4+ T cells are highly permissive to HIV-1 infection. J. Immunol. 2010, 184, 1604–1616. [Google Scholar] [CrossRef] [Green Version]
  101. Planas, D.; Zhang, Y.; Monteiro, P.; Goulet, J.P.; Gosselin, A.; Grandvaux, N.; Hope, T.J.; Fassati, A.; Routy, J.P.; Ancuta, P. HIV-1 selectively targets gut-homing CCR6+CD4+ T cells via mTOR-dependent mechanisms. JCI Insight 2017, 2, e93230. [Google Scholar] [CrossRef] [Green Version]
  102. Chege, D.; Sheth, P.M.; Kain, T.; Kim, C.J.; Kovacs, C.; Loutfy, M.; Halpenny, R.; Kandel, G.; Chun, T.W.; Ostrowski, M.; et al. Sigmoid Th17 populations, the HIV latent reservoir, and microbial translocation in men on long-term antiretroviral therapy. AIDS 2011, 25, 741–749. [Google Scholar] [CrossRef] [PubMed]
  103. McKinnon, L.R.; Nyanga, B.; Kim, C.J.; Izulla, P.; Kwatampora, J.; Kimani, M.; Shahabi, K.; Mugo, N.; Smith, J.S.; Anzala, A.O.; et al. Early HIV-1 infection is associated with reduced frequencies of cervical Th17 cells. J. Acquir. Immune Defic. Syndr. 2015, 68, 6–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Christensen-Quick, A.; Lafferty, M.; Sun, L.; Marchionni, L.; DeVico, A.; Garzino-Demo, A. Human Th17 Cells Lack HIV-Inhibitory RNases and Are Highly Permissive to Productive HIV Infection. J. Virol. 2016, 90, 7833–7847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. El Hed, A.; Khaitan, A.; Kozhaya, L.; Manel, N.; Daskalakis, D.; Borkowsky, W.; Valentine, F.; Littman, D.R.; Unutmaz, D. Susceptibility of human Th17 cells to human immunodeficiency virus and their perturbation during infection. J. Infect. Dis. 2010, 201, 843–854. [Google Scholar] [CrossRef]
  106. Isnard, S.; Lin, J.; Bu, S.; Fombuena, B.; Royston, L.; Routy, J.P. Gut Leakage of Fungal-Related Products: Turning Up the Heat for HIV Infection. Front. Immunol. 2021, 12, 656414. [Google Scholar] [CrossRef]
  107. Peng, X.; Isnard, S.; Lin, J.; Fombuena, B.; Bessissow, T.; Chomont, N.; Routy, J.P. Differences in HIV burden in the inflamed and non-inflamed colon from a person living with HIV and ulcerative colitis. J. Virus. Erad. 2021, 7, 100033. [Google Scholar] [CrossRef]
  108. Zhao, Y.; Kilian, C.; Turner, J.E.; Bosurgi, L.; Roedl, K.; Bartsch, P.; Gnirck, A.C.; Cortesi, F.; Schultheiss, C.; Hellmig, M.; et al. Clonal expansion and activation of tissue-resident memory-like Th17 cells expressing GM-CSF in the lungs of severe COVID-19 patients. Sci. Immunol. 2021, 6, eabf6692. [Google Scholar] [CrossRef]
  109. Toor, S.M.; Saleh, R.; Sasidharan Nair, V.; Taha, R.Z.; Elkord, E. T-cell responses and therapies against SARS-CoV-2 infection. Immunology 2021, 162, 30–43. [Google Scholar] [CrossRef]
  110. De Biasi, S.; Meschiari, M.; Gibellini, L.; Bellinazzi, C.; Borella, R.; Fidanza, L.; Gozzi, L.; Iannone, A.; Lo Tartaro, D.; Mattioli, M.; et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat. Commun. 2020, 11, 3434. [Google Scholar] [CrossRef]
  111. Orlov, M.; Wander, P.L.; Morrell, E.D.; Mikacenic, C.; Wurfel, M.M. A Case for Targeting Th17 Cells and IL-17A in SARS-CoV-2 Infections. J. Immunol. 2020, 205, 892–898. [Google Scholar] [CrossRef] [PubMed]
  112. Aghbash, P.S.; Hemmat, N.; Nahand, J.S.; Shamekh, A.; Memar, M.Y.; Babaei, A.; Baghi, H.B. The role of Th17 cells in viral infections. Int. Immunopharmacol. 2021, 91, 107331. [Google Scholar] [CrossRef] [PubMed]
  113. Hooper, L.V.; Littman, D.R.; Macpherson, A.J. Interactions between the microbiota and the immune system. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ivanov, I.I.; Frutos Rde, L.; Manel, N.; Yoshinaga, K.; Rifkin, D.B.; Sartor, R.B.; Finlay, B.B.; Littman, D.R. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 2008, 4, 337–349. [Google Scholar] [CrossRef] [Green Version]
  115. Macpherson, A.J.; Harris, N.L. Interactions between commensal intestinal bacteria and the immune system. Nat. Rev. Immunol. 2004, 4, 478–485. [Google Scholar] [CrossRef]
  116. Kumar, P.; Monin, L.; Castillo, P.; Elsegeiny, W.; Horne, W.; Eddens, T.; Vikram, A.; Good, M.; Schoenborn, A.A.; Bibby, K.; et al. Intestinal Interleukin-17 Receptor Signaling Mediates Reciprocal Control of the Gut Microbiota and Autoimmune Inflammation. Immunity 2016, 44, 659–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Ishigame, H.; Kakuta, S.; Nagai, T.; Kadoki, M.; Nambu, A.; Komiyama, Y.; Fujikado, N.; Tanahashi, Y.; Akitsu, A.; Kotaki, H.; et al. Differential roles of interleukin-17A and -17F in host defense against mucoepithelial bacterial infection and allergic responses. Immunity 2009, 30, 108–119. [Google Scholar] [CrossRef] [Green Version]
  118. Sano, T.; Huang, W.; Hall, J.A.; Yang, Y.; Chen, A.; Gavzy, S.J.; Lee, J.Y.; Ziel, J.W.; Miraldi, E.R.; Domingos, A.I.; et al. An IL-23R/IL-22 Circuit Regulates Epithelial Serum Amyloid A to Promote Local Effector Th17 Responses. Cell 2015, 163, 381–393. [Google Scholar] [CrossRef] [Green Version]
  119. Kleinewietfeld, M.; Manzel, A.; Titze, J.; Kvakan, H.; Yosef, N.; Linker, R.A.; Muller, D.N.; Hafler, D.A. Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells. Nature 2013, 496, 518–522. [Google Scholar] [CrossRef] [Green Version]
  120. Campisi, L.; Barbet, G.; Ding, Y.; Esplugues, E.; Flavell, R.A.; Blander, J.M. Apoptosis in response to microbial infection induces autoreactive TH17 cells. Nat. Immunol. 2016, 17, 1084–1092. [Google Scholar] [CrossRef]
  121. Khader, S.A.; Pearl, J.E.; Sakamoto, K.; Gilmartin, L.; Bell, G.K.; Jelley-Gibbs, D.M.; Ghilardi, N.; de Sauvage, F.; Cooper, A.M. IL-23 compensates for the absence of IL-12p70 and is essential for the IL-17 response during tuberculosis but is dispensable for protection and antigen-specific IFN-gamma responses if IL-12p70 is available. J. Immunol. 2005, 175, 788–795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Aujla, S.J.; Dubin, P.J.; Kolls, J.K. Th17 cells and mucosal host defense. Semin. Immunol. 2007, 19, 377–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Umemura, M.; Yahagi, A.; Hamada, S.; Begum, M.D.; Watanabe, H.; Kawakami, K.; Suda, T.; Sudo, K.; Nakae, S.; Iwakura, Y.; et al. IL-17-mediated regulation of innate and acquired immune response against pulmonary Mycobacterium bovis bacille Calmette-Guerin infection. J. Immunol. 2007, 178, 3786–3796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Cruz, A.; Fraga, A.G.; Fountain, J.J.; Rangel-Moreno, J.; Torrado, E.; Saraiva, M.; Pereira, D.R.; Randall, T.D.; Pedrosa, J.; Cooper, A.M.; et al. Pathological role of interleukin 17 in mice subjected to repeated BCG vaccination after infection with Mycobacterium tuberculosis. J. Exp. Med. 2010, 207, 1609–1616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Alnahas, S.; Hagner, S.; Raifer, H.; Kilic, A.; Gasteiger, G.; Mutters, R.; Hellhund, A.; Prinz, I.; Pinkenburg, O.; Visekruna, A.; et al. IL-17 and TNF-alpha Are Key Mediators of Moraxella catarrhalis Triggered Exacerbation of Allergic Airway Inflammation. Front. Immunol. 2017, 8, 1562. [Google Scholar] [CrossRef]
  126. Essilfie, A.T.; Simpson, J.L.; Horvat, J.C.; Preston, J.A.; Dunkley, M.L.; Foster, P.S.; Gibson, P.G.; Hansbro, P.M. Haemophilus influenzae infection drives IL-17-mediated neutrophilic allergic airways disease. PLoS Pathog. 2011, 7, e1002244. [Google Scholar] [CrossRef]
  127. Kurata, S.; Osaki, T.; Yonezawa, H.; Arae, K.; Taguchi, H.; Kamiya, S. Role of IL-17A and IL-10 in the antigen induced inflammation model by Mycoplasma pneumoniae. BMC Microbiol. 2014, 14, 156. [Google Scholar] [CrossRef] [Green Version]
  128. Wu, Q.; Martin, R.J.; Rino, J.G.; Breed, R.; Torres, R.M.; Chu, H.W. IL-23-dependent IL-17 production is essential in neutrophil recruitment and activity in mouse lung defense against respiratory Mycoplasma pneumoniae infection. Microbes Infect. 2007, 9, 78–86. [Google Scholar] [CrossRef] [Green Version]
  129. Gavino, A.C.; Nahmod, K.; Bharadwaj, U.; Makedonas, G.; Tweardy, D.J. STAT3 inhibition prevents lung inflammation, remodeling, and accumulation of Th2 and Th17 cells in a murine asthma model. Allergy 2016, 71, 1684–1692. [Google Scholar] [CrossRef]
  130. Dutzan, N.; Abusleme, L.; Bridgeman, H.; Greenwell-Wild, T.; Zangerle-Murray, T.; Fife, M.E.; Bouladoux, N.; Linley, H.; Brenchley, L.; Wemyss, K.; et al. On-going Mechanical Damage from Mastication Drives Homeostatic Th17 Cell Responses at the Oral Barrier. Immunity 2017, 46, 133–147. [Google Scholar] [CrossRef]
  131. Popa, E.R.; Stegeman, C.A.; Abdulahad, W.H.; van der Meer, B.; Arends, J.; Manson, W.M.; Bos, N.A.; Kallenberg, C.G.; Tervaert, J.W. Staphylococcal toxic-shock-syndrome-toxin-1 as a risk factor for disease relapse in Wegener’s granulomatosis. Rheumatology 2007, 46, 1029–1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Abdulahad, W.H.; Stegeman, C.A.; Kallenberg, C.G. Review article: The role of CD4(+) T cells in ANCA-associated systemic vasculitis. Nephrology 2009, 14, 26–32. [Google Scholar] [CrossRef] [PubMed]
  133. Lopez, P.; de Paz, B.; Rodriguez-Carrio, J.; Hevia, A.; Sanchez, B.; Margolles, A.; Suarez, A. Th17 responses and natural IgM antibodies are related to gut microbiota composition in systemic lupus erythematosus patients. Sci. Rep. 2016, 6, 24072. [Google Scholar] [CrossRef] [Green Version]
  134. Nishida, A.; Inoue, R.; Inatomi, O.; Bamba, S.; Naito, Y.; Andoh, A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin. J. Gastroenterol. 2018, 11, 1–10. [Google Scholar] [CrossRef] [Green Version]
  135. Shahi, S.K.; Freedman, S.N.; Mangalam, A.K. Gut microbiome in multiple sclerosis: The players involved and the roles they play. Gut Microbes 2017, 8, 607–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Horta-Baas, G.; Romero-Figueroa, M.D.S.; Montiel-Jarquin, A.J.; Pizano-Zarate, M.L.; Garcia-Mena, J.; Ramirez-Duran, N. Intestinal Dysbiosis and Rheumatoid Arthritis: A Link between Gut Microbiota and the Pathogenesis of Rheumatoid Arthritis. J. Immunol. Res. 2017, 2017, 4835189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Rinaldi, E.; Consonni, A.; Guidesi, E.; Elli, M.; Mantegazza, R.; Baggi, F. Gut microbiota and probiotics: Novel immune system modulators in myasthenia gravis? Ann. NY. Acad. Sci. 2018, 1413, 49–58. [Google Scholar] [CrossRef] [PubMed]
  138. Raza, A.; Shata, M.T. Letter: Pathogenicity of Th17 cells may differ in ulcerative colitis compared with Crohn’s disease. Aliment. Pharmacol. Ther. 2012, 36, 204. [Google Scholar] [CrossRef]
  139. Monteleone, I.; Sarra, M.; Pallone, F.; Monteleone, G. Th17-related cytokines in inflammatory bowel diseases: Friends or foes? Curr. Mol. Med. 2012, 12, 592–597. [Google Scholar] [CrossRef]
  140. Chu, H.; Khosravi, A.; Kusumawardhani, I.P.; Kwon, A.H.; Vasconcelos, A.C.; Cunha, L.D.; Mayer, A.E.; Shen, Y.; Wu, W.L.; Kambal, A.; et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science 2016, 352, 1116–1120. [Google Scholar] [CrossRef] [Green Version]
  141. Kamada, N.; Seo, S.U.; Chen, G.Y.; Nunez, G. Role of the gut microbiota in immunity and inflammatory disease. Nat. Rev. Immunol. 2013, 13, 321–335. [Google Scholar] [CrossRef] [PubMed]
  142. Cani, P.D.; de Vos, W.M. Next-Generation Beneficial Microbes: The Case of Akkermansia muciniphila. Front. Microbiol. 2017, 8, 1765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Park, J.S.; Choi, J.; Kwon, J.Y.; Jung, K.A.; Yang, C.W.; Park, S.H.; Cho, M.L. A probiotic complex, rosavin, zinc, and prebiotics ameliorate intestinal inflammation in an acute colitis mouse model. J. Transl. Med. 2018, 16, 37. [Google Scholar] [CrossRef]
  144. Round, J.L.; Lee, S.M.; Li, J.; Tran, G.; Jabri, B.; Chatila, T.A.; Mazmanian, S.K. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 2011, 332, 974–977. [Google Scholar] [CrossRef] [Green Version]
  145. Patel, V.C.; Lee, S.; McPhail, M.J.W.; Da Silva, K.; Guilly, S.; Zamalloa, A.; Witherden, E.; Stoy, S.; Manakkat Vijay, G.K.; Pons, N.; et al. Rifaximin-alpha reduces gut-derived inflammation and mucin degradation in cirrhosis and encephalopathy: RIFSYS randomised controlled trial. J. Hepatol. 2022, 76, 332–342. [Google Scholar] [CrossRef]
  146. Craven, L.; Rahman, A.; Nair Parvathy, S.; Beaton, M.; Silverman, J.; Qumosani, K.; Hramiak, I.; Hegele, R.; Joy, T.; Meddings, J.; et al. Allogenic Fecal Microbiota Transplantation in Patients with Nonalcoholic Fatty Liver Disease Improves Abnormal Small Intestinal Permeability: A Randomized Control Trial. Am. J. Gastroenterol. 2020, 115, 1055–1065. [Google Scholar] [CrossRef]
  147. Maeda, Y.; Kurakawa, T.; Umemoto, E.; Motooka, D.; Ito, Y.; Gotoh, K.; Hirota, K.; Matsushita, M.; Furuta, Y.; Narazaki, M.; et al. Dysbiosis Contributes to Arthritis Development via Activation of Autoreactive T Cells in the Intestine. Arthritis Rheumatol 2016, 68, 2646–2661. [Google Scholar] [CrossRef] [PubMed]
  148. Asquith, M.J.; Stauffer, P.; Davin, S.; Mitchell, C.; Lin, P.; Rosenbaum, J.T. Perturbed Mucosal Immunity and Dysbiosis Accompany Clinical Disease in a Rat Model of Spondyloarthritis. Arthritis Rheumatol 2016, 68, 2151–2162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Bisgaard, H.; Hermansen, M.N.; Buchvald, F.; Loland, L.; Halkjaer, L.B.; Bonnelykke, K.; Brasholt, M.; Heltberg, A.; Vissing, N.H.; Thorsen, S.V.; et al. Childhood asthma after bacterial colonization of the airway in neonates. N. Engl. J. Med. 2007, 357, 1487–1495. [Google Scholar] [CrossRef]
  150. Yang, B.; Liu, R.; Yang, T.; Jiang, X.; Zhang, L.; Wang, L.; Wang, Q.; Luo, Z.; Liu, E.; Fu, Z. Neonatal Streptococcus pneumoniae infection may aggravate adulthood allergic airways disease in association with IL-17A. PLoS ONE 2015, 10, e0123010. [Google Scholar] [CrossRef]
  151. Peng, X.; Wu, Y.; Kong, X.; Chen, Y.; Tian, Y.; Li, Q.; Tian, X.; Zhang, G.; Ren, L.; Luo, Z. Neonatal Streptococcus pneumoniae Pneumonia Induces an Aberrant Airway Smooth Muscle Phenotype and AHR in Mice Model. Biomed. Res. Int. 2019, 2019, 1948519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Wood, P.R.; Hill, V.L.; Burks, M.L.; Peters, J.I.; Singh, H.; Kannan, T.R.; Vale, S.; Cagle, M.P.; Principe, M.F.; Baseman, J.B.; et al. Mycoplasma pneumoniae in children with acute and refractory asthma. Ann. Allergy Asthma Immunol. 2013, 110, 328–334.e321. [Google Scholar] [CrossRef] [Green Version]
  153. Kassisse, E.; Garcia, H.; Prada, L.; Salazar, I.; Kassisse, J. Prevalence of Mycoplasma pneumoniae infection in pediatric patients with acute asthma exacerbation. Arch Argent Pediatr. 2018, 116, 179–185. [Google Scholar]
  154. Awang, R.A.; Lappin, D.F.; MacPherson, A.; Riggio, M.; Robertson, D.; Hodge, P.; Ramage, G.; Culshaw, S.; Preshaw, P.M.; Taylor, J.; et al. Clinical associations between IL-17 family cytokines and periodontitis and potential differential roles for IL-17A and IL-17E in periodontal immunity. Inflamm. Res. 2014, 63, 1001–1012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Cheng, W.C.; Hughes, F.J.; Taams, L.S. The presence, function and regulation of IL-17 and Th17 cells in periodontitis. J. Clin. Periodontol. 2014, 41, 541–549. [Google Scholar] [CrossRef] [PubMed]
  156. Schenkein, H.A.; Koertge, T.E.; Brooks, C.N.; Sabatini, R.; Purkall, D.E.; Tew, J.G. IL-17 in sera from patients with aggressive periodontitis. J. Dent. Res. 2010, 89, 943–947. [Google Scholar] [CrossRef]
  157. Behfarnia, P.; Birang, R.; Pishva, S.S.; Hakemi, M.G.; Khorasani, M.M. Expression levels of th-2 and th-17 characteristic genes in healthy tissue versus periodontitis. J. Dent. (Tehran) 2013, 10, 23–31. [Google Scholar]
  158. Li, J.; Casanova, J.L.; Puel, A. Mucocutaneous IL-17 immunity in mice and humans: Host defense vs. excessive inflammation. Mucosal Immunol. 2018, 11, 581–589. [Google Scholar] [CrossRef] [Green Version]
  159. Valladao, A.C.; Frevert, C.W.; Koch, L.K.; Campbell, D.J.; Ziegler, S.F. STAT6 Regulates the Development of Eosinophilic versus Neutrophilic Asthma in Response to Alternaria alternata. J. Immunol. 2016, 197, 4541–4551. [Google Scholar] [CrossRef] [Green Version]
  160. Zhang, Z.; Biagini Myers, J.M.; Brandt, E.B.; Ryan, P.H.; Lindsey, M.; Mintz-Cole, R.A.; Reponen, T.; Vesper, S.J.; Forde, F.; Ruff, B.; et al. beta-Glucan exacerbates allergic asthma independent of fungal sensitization and promotes steroid-resistant TH2/TH17 responses. J. Allergy Clin. Immunol. 2017, 139, 54–65.e58. [Google Scholar] [CrossRef] [Green Version]
  161. Patel, D.; Gaikwad, S.; Challagundla, N.; Nivsarkar, M.; Agrawal-Rajput, R. Spleen tyrosine kinase inhibition ameliorates airway inflammation through modulation of NLRP3 inflammosome and Th17/Treg axis. Int. Immunopharmacol. 2018, 54, 375–384. [Google Scholar] [CrossRef] [PubMed]
  162. Bacher, P.; Kniemeyer, O.; Schonbrunn, A.; Sawitzki, B.; Assenmacher, M.; Rietschel, E.; Steinbach, A.; Cornely, O.A.; Brakhage, A.A.; Thiel, A.; et al. Antigen-specific expansion of human regulatory T cells as a major tolerance mechanism against mucosal fungi. Mucosal Immunol. 2014, 7, 916–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Brown, G.D.; Denning, D.W.; Gow, N.A.; Levitz, S.M.; Netea, M.G.; White, T.C. Hidden killers: Human fungal infections. Sci. Transl. Med. 2012, 4, 165rv13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Scheffold, A.; Schwarz, C.; Bacher, P. Fungus-Specific CD4 T Cells as Specific Sensors for Identification of Pulmonary Fungal Infections. Mycopathologia 2018, 183, 213–226. [Google Scholar] [CrossRef]
  165. Bacher, P.; Heinrich, F.; Stervbo, U.; Nienen, M.; Vahldieck, M.; Iwert, C.; Vogt, K.; Kollet, J.; Babel, N.; Sawitzki, B.; et al. Regulatory T Cell Specificity Directs Tolerance versus Allergy against Aeroantigens in Humans. Cell 2016, 167, 1067–1078.e1016. [Google Scholar] [CrossRef] [Green Version]
  166. Moyes, D.L.; Wilson, D.; Richardson, J.P.; Mogavero, S.; Tang, S.X.; Wernecke, J.; Hofs, S.; Gratacap, R.L.; Robbins, J.; Runglall, M.; et al. Candidalysin is a fungal peptide toxin critical for mucosal infection. Nature 2016, 532, 64–68. [Google Scholar] [CrossRef] [Green Version]
  167. Verma, A.H.; Richardson, J.P.; Zhou, C.; Coleman, B.M.; Moyes, D.L.; Ho, J.; Huppler, A.R.; Ramani, K.; McGeachy, M.J.; Mufazalov, I.A.; et al. Oral epithelial cells orchestrate innate type 17 responses to Candida albicans through the virulence factor candidalysin. Sci Immunol 2017, 2, 17. [Google Scholar] [CrossRef]
  168. Knutsen, A.P.; Bush, R.K.; Demain, J.G.; Denning, D.W.; Dixit, A.; Fairs, A.; Greenberger, P.A.; Kariuki, B.; Kita, H.; Kurup, V.P.; et al. Fungi and allergic lower respiratory tract diseases. J. Allergy Clin. Immunol. 2012, 129, 280–291. [Google Scholar] [CrossRef]
  169. Yii, A.C.; Koh, M.S.; Lapperre, T.S.; Tan, G.L.; Chotirmall, S.H. The emergence of Aspergillus species in chronic respiratory disease. Front. Biosci. 2017, 9, 127–138. [Google Scholar]
  170. Iwanaga, N.; Kolls, J.K. Updates on T helper type 17 immunity in respiratory disease. Immunology 2019, 156, 3–8. [Google Scholar] [CrossRef] [Green Version]
  171. Chan, Y.R.; Chen, K.; Duncan, S.R.; Lathrop, K.L.; Latoche, J.D.; Logar, A.J.; Pociask, D.A.; Wahlberg, B.J.; Ray, P.; Ray, A.; et al. Patients with cystic fibrosis have inducible IL-17+IL-22+ memory cells in lung draining lymph nodes. J. Allergy Clin. Immunol. 2013, 131, 1117–1129.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Jolink, H.; de Boer, R.; Hombrink, P.; Jonkers, R.E.; van Dissel, J.T.; Falkenburg, J.H.; Heemskerk, M.H. Pulmonary immune responses against Aspergillus fumigatus are characterized by high frequencies of IL-17 producing T-cells. J. Infect. 2017, 74, 81–88. [Google Scholar] [CrossRef] [PubMed]
  173. Bacher, P.; Hohnstein, T.; Beerbaum, E.; Rocker, M.; Blango, M.G.; Kaufmann, S.; Rohmel, J.; Eschenhagen, P.; Grehn, C.; Seidel, K.; et al. Human Anti-fungal Th17 Immunity and Pathology Rely on Cross-Reactivity against Candida albicans. Cell 2019, 176, 1340–1355.e15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Kim, Y.G.; Udayanga, K.G.; Totsuka, N.; Weinberg, J.B.; Nunez, G.; Shibuya, A. Gut dysbiosis promotes M2 macrophage polarization and allergic airway inflammation via fungi-induced PGE(2). Cell Host Microbe 2014, 15, 95–102. [Google Scholar] [CrossRef] [Green Version]
  175. Noverr, M.C.; Noggle, R.M.; Toews, G.B.; Huffnagle, G.B. Role of antibiotics and fungal microbiota in driving pulmonary allergic responses. Infect. Immun. 2004, 72, 4996–5003. [Google Scholar] [CrossRef] [Green Version]
  176. McAleer, J.P.; Kolls, J.K. Contributions of the intestinal microbiome in lung immunity. Eur. J. Immunol. 2018, 48, 39–49. [Google Scholar] [CrossRef] [Green Version]
  177. Brevi, A.; Cogrossi, L.L.; Grazia, G.; Masciovecchio, D.; Impellizzieri, D.; Lacanfora, L.; Grioni, M.; Bellone, M. Much More Than IL-17A: Cytokines of the IL-17 Family Between Microbiota and Cancer. Front. Immunol. 2020, 11, 565470. [Google Scholar] [CrossRef]
Figure 1. The process of Th17 cell differentiation. Dendritic cells after activation by microbial components activate CD4+ naive T cells in an antigen-dependent manner. In the presence of the appropriate cytokine environment, such cells acquire a Th17 phenotype by upregulating RORγt/RORC and downregulating Foxp3. IL-23 is required to stabilize their phenotype. Th17 cells are characterized by the ability to synthesize and secrete IL-17A and IL-17F in addition to other soluble factors. IL-6, TGFβ, and IL-1β contribute to the differentiation of naïve T cells into Th17 cells. IL-23 is required to stabilize their phenotype. Chemokine receptor CCR6 directs Th17 cells to barrier tissues.
Figure 1. The process of Th17 cell differentiation. Dendritic cells after activation by microbial components activate CD4+ naive T cells in an antigen-dependent manner. In the presence of the appropriate cytokine environment, such cells acquire a Th17 phenotype by upregulating RORγt/RORC and downregulating Foxp3. IL-23 is required to stabilize their phenotype. Th17 cells are characterized by the ability to synthesize and secrete IL-17A and IL-17F in addition to other soluble factors. IL-6, TGFβ, and IL-1β contribute to the differentiation of naïve T cells into Th17 cells. IL-23 is required to stabilize their phenotype. Chemokine receptor CCR6 directs Th17 cells to barrier tissues.
Pathogens 11 01547 g001
Figure 2. The pro-inflammatory function of Th17 cells. IL-17A and IL-17F produced by Th17 cells recognize different cellular targets that express their specific receptors. After activation, the signal transduction is triggered with formation of the ACT1/TRAF6 complex. The final event of this process includes activation of transcription factor NF-κB. Different pro-inflammatory factors are synthesized by the stimulated cells including cytokines, chemokines, growth factors and matrix metalloproteinases. IL-17A/IL-17F target cells include keratinocytes, fibroblasts, osteoblasts, epithelial cells, endothelial cells and macrophages.
Figure 2. The pro-inflammatory function of Th17 cells. IL-17A and IL-17F produced by Th17 cells recognize different cellular targets that express their specific receptors. After activation, the signal transduction is triggered with formation of the ACT1/TRAF6 complex. The final event of this process includes activation of transcription factor NF-κB. Different pro-inflammatory factors are synthesized by the stimulated cells including cytokines, chemokines, growth factors and matrix metalloproteinases. IL-17A/IL-17F target cells include keratinocytes, fibroblasts, osteoblasts, epithelial cells, endothelial cells and macrophages.
Pathogens 11 01547 g002
Figure 3. The dual role of Th17s after encountering pathogens. Th17 cells provide mucosal defense against viral, bacterial, and fungal agents. However, the same pathogens can induce altered activation of Th17 cells, which may in turn become responsible for immunopathology causing disease onset. Evidence that the Th17 response to pathogens can mediate immunopathology comes from findings in humans, animal models, or both.
Figure 3. The dual role of Th17s after encountering pathogens. Th17 cells provide mucosal defense against viral, bacterial, and fungal agents. However, the same pathogens can induce altered activation of Th17 cells, which may in turn become responsible for immunopathology causing disease onset. Evidence that the Th17 response to pathogens can mediate immunopathology comes from findings in humans, animal models, or both.
Pathogens 11 01547 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Paroli, M.; Caccavale, R.; Fiorillo, M.T.; Spadea, L.; Gumina, S.; Candela, V.; Paroli, M.P. The Double Game Played by Th17 Cells in Infection: Host Defense and Immunopathology. Pathogens 2022, 11, 1547. https://doi.org/10.3390/pathogens11121547

AMA Style

Paroli M, Caccavale R, Fiorillo MT, Spadea L, Gumina S, Candela V, Paroli MP. The Double Game Played by Th17 Cells in Infection: Host Defense and Immunopathology. Pathogens. 2022; 11(12):1547. https://doi.org/10.3390/pathogens11121547

Chicago/Turabian Style

Paroli, Marino, Rosalba Caccavale, Maria Teresa Fiorillo, Luca Spadea, Stefano Gumina, Vittorio Candela, and Maria Pia Paroli. 2022. "The Double Game Played by Th17 Cells in Infection: Host Defense and Immunopathology" Pathogens 11, no. 12: 1547. https://doi.org/10.3390/pathogens11121547

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop