Abstract
The term Tc17 cells refers to interleukin 17 (IL-17)-producing CD8+ T cells. While IL-17 is an important mediator of mucosal defense, it is also centrally involved in driving the inflammatory response in immune-mediated diseases, such as psoriasis, multiple sclerosis, and inflammatory bowel disease. In this review, we aim to gather the current knowledge on the phenotypic and transcriptional profile, the in vitro and in vivo generation of Tc17 cells, and the evidence pointing towards a relevant role of Tc17 cells in human diseases such as infectious diseases, cancer, and immune-mediated diseases.
Keywords: T cell, IL-17, Tc17, Th17, inflammatory bowel disease
Introduction
T cells are a central component of the adaptive immune system. They can recognize and respond to specific antigens, eliminate infected or cancerous cells, and generate memory responses that protect the host from reinfection. T cells can be divided into several subtypes, including CD4+ T cells and CD8+ T cells. CD4+ T cells, also called T helper cells, coordinate the immune response by releasing cytokines that activate and recruit other immune cells. Specific cytokines can shape different immune responses: Type 1 responses are mediated by interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α)-producing Th1 cells and protect against intracellular pathogens, but are also associated with immune-mediated diseases. Type 2 responses involve interleukin (IL)-4, IL-5, and IL-13-producing Th2 cells and the activation of eosinophils, basophils, and mast cells as well as the production of immunoglobulin E, thus protecting against helminth infections. On the flip side, overreaching type 2 immunity is observed in allergic diseases. The more recently described type 17 immunity comprising the Th17 cell-secreted cytokines IL-17 and IL-22 recruits neutrophils and mononuclear phagocytes and induces antimicrobial responses by epithelial cells, thereby providing defense against fungal infections. Overreaching type 17 responses have, however, been implicated in the pathogenesis of immune-mediated diseases such as multiple sclerosis (MS) and inflammatory bowel disease (IBD) [1].
A major role for CD8+ T cells is to recognize presented antigenic peptides via their T cell receptor (TCR) and subsequently kill virus-infected or cancerous cells. This is a function of cytotoxic Tc1 cells. However, CD8+ T cells can also produce different cytokines and perform functions other than killing. In the last decades, similar to CD4+ T cell nomenclature, additional distinct CD8+ T cell subtypes like Tc2, Tc9, CD8+ T regulatory cells, and Tc17 cells have been identified and characterized [2, 3]. Among these specialized CD8+ T cell subsets, Tc17 cells have mainly been identified based on their ability to produce type 17 cytokines such as IL-17A and IL-17F. Both in healthy and diseased humans, most Tc17 cells have been found to produce IL-17A, but only about 20% of Tc17 cells coproduce IL-17F [4]. Besides the production of their namesake cytokine IL-17, Tc17 cells are also capable of producing additional cytokines, such as IFN-γ, TNF-α, GM-CSF, IL-21, and IL-22 [5]. Quantitatively, about 90% of Tc17 cells can coproduce TNF-α and about half of Tc17 cells can coproduce IFN-γ in healthy donors. In inflammatory conditions like active IBD, this coproduction profile remains unchanged, suggesting it is an inherent feature of the Tc17 program [4].
The IL-17 receptor is universally expressed by epithelial cells and stromal cells such as fibroblasts [6–8], which upon IL-17 stimulation, produce cytokines and chemokines such as IL-6, CXCL2, and CXCL8 and the hematopoietic factors G-CSF and GM-CSF, thereby enhancing the local and systemic immune response, e.g. by recruitment of neutrophils [9–12]. Tc17 cells have been implicated in a variety of diseases such as viral, bacterial, and fungal infections, gastrointestinal malignancies, and immune-mediated diseases of the skin, the gastrointestinal tract, and the central nervous system, and the IL-17 pathway has recently become a prominent target of novel biological therapies [6, 13, 14]. In this review, we will summarize the current knowledge on the phenotypic and transcriptional profile of Tc17 cells, their in vitro and in vivo generation, and their involvement in human diseases.
The IL-17 cytokine family
The IL-17 cytokine family includes six members: IL-17 (also called IL-17A), IL-17B, IL-17C, IL-17D, IL-17E (also called IL-25), and IL-17F [15]. IL-17A and IL-17F share the greatest homology and are the best-characterized family members [16–18]. Both induce the production of proinflammatory cytokines and chemokines in target cells, thereby leading to inflammation and recruitment of neutrophils. Less is known about IL-17B and IL-17C, although both are thought to also mediate the inflammatory response and be involved in arthritis [19]. IL-17E is the most unique family member and can regulate type 2 immune responses [15].
Tc17 cell phenotype
Tc17 cells share key transcriptional, phenotypic, and functional features with their Th17 counterparts, including the expression of the type 17 master regulator retinoic acid receptor-related orphan receptor γt (RORγt) [20–23]. They are further characterized by the expression of a variety of surface markers, such as the C-type lectin-like receptor CD161, the dipeptidyl peptidase IV (CD26), and the CC motif chemokine receptors CCR5 and CCR6 [24–28] (Figure 1). Tc17 cells have been found to be mostly part of the CD27-/+ CD28+ CD45RA- cell subsets in humans [28, 29]. These markers can help identify Tc17 cells in diagnostic and biomarker settings that do not allow for intracellular analysis.
Figure 1:
Key features of Tc17 cells in protection and pathology
CD161 is expressed on human circulating lymphocytes able to produce IL-17, and its expression can be induced by expression of the type 17 master transcription factor RORC in naïve T cells [30], supporting the close correlation between CD161 expression and type 17 programming. While the binding of CD161 to its ligand lectin-like transcript 1 (LLT1) has costimulatory effects on natural killer (NK) cells, the role of CD161:LLT1 in modulating the function of T cells remains unclear, in particular, it remains unclear if LLT1 modulates IL-17 production [31–33].
CD26 was initially described as a T cell activation marker and regulates T cell proliferation [34, 35]. It acts as an ectoenzyme that cleaves amino-terminal dipeptides from different chemokines [36]. CD26 expressed on IL-17-producing human T cells is enzymatically active and negatively regulates the chemotactic T cell response, which can be restored by inhibition of CD26 [24].
The expression of CCR5 and CCR6 is shared among both Tc17 and Th17 cells [28, 37, 38]. Mechanistically, the interaction of CCR6 and its ligand CCL20 plays an important role for type 17 T cell migration to epithelial sites such as the intestinal epithelium [39, 40] or to the infected lung [41], as well as for T cell localization close to the intrahepatic bile ducts in inflammatory liver diseases [42].
While the expression of none of these markers is exclusive to Tc17 cells per se, a signature of seven surface markers uniquely expressed by human Tc17 cells has recently been identified by our group and consists of a high expression of CD161 and CD26, in addition to CD6, CD69, CD39, PD-1, and a low expression of CD27 [4].
CD6 is involved in the fine-tuning of T cell activation signals [43] and T cell proliferation [44]. Its ligands include activated leukocyte cell adhesion molecule (ALCAM/CD166) and CD318 [45–47]. The cytosolic part of CD6 forms a complex signalosome and can both positively and negatively regulate T cell activation [43, 48, 49]. Importantly, inhibition of CD6 on human CD8+ T cells results in reduced production of IL-17A, IFN-γ, and TNF-α [4]. It has previously been suggested that anti-CD6 antibodies modulate activation and differentiation of Th17 cells through modulation of signal transducer and activator of transcription 3 (STAT3) signaling [50], however, we were not able to reproduce this in Tc17 cells [4].
An increase in the expression of the type II transmembrane protein of the C-type lectin family CD69 can be rapidly detected after stimulation of different immune cell types including T cells [51] and CD69 is consequently commonly used as a marker for cell activation. Deletion of CD69 leads to alterations in the disease activity in arthritis and colitis mouse models [52–55], supporting a functional role of CD69-signaling in the regulation of Th17 differentiation and production of cytokines [56, 57], although the reports regarding the question whether CD69 deletion is clinically beneficial in these disease contexts are contradictory. Interaction of CD69 with its ligand galectin-1 impairs type 17 differentiation of both human and murine naïve CD4+ T cells, an effect which is reversed by anti-CD69 antibodies [58]. Mechanistically, the cytoplasmic fraction of CD69 interacts with the JAK3/STAT5 pathway, resulting in the suppression of STAT3-mediated type 17 gene expression [57, 59]. In addition, CD69 can also associate with the sphingosine-1-phosphate receptor 1 (S1PR1) and the heterodimeric amino acid transporter complex LAT1/CD98 [60, 61]. The association with CD69 prevents S1PR1 from mediating tissue egress of T cells [60, 62]. Consequently, CD69 is a common marker for tissue-resident memory cells, which share some characteristics with Tc17 cells and can produce IL-17 [63].
The ectoenzyme CD39 (ENTPD1/ectonucleoside triphosphate diphosphorylase 1) catalyzes the extracellular conversion of adenosine triphosphate (ATP) and adenosine diphosphate to adenosine monophosphate [64, 65], which is then converted to adenosine by CD73 [64, 66, 67]. The activity of these two enzymes thereby regulates the balance between extracellular proinflammatory ATP and its immunosuppressive derivates. CD39 expressed on CD8+ T cells is enzymatically active and negatively regulates their cytokine production [68]. While the frequency of CD39+ cells in the peripheral blood is typically low in both healthy donors and cancer patients, an increased expression has been described for tumor-infiltrating CD8+ T cells [69, 70] and hepatitis C virus (HCV)- and HIV-specific CD8+ T cells [71]. CD39+ CD8+ T cells recognize tumor antigens [72] and display features of T cell exhaustion such as an increased expression of inhibitory receptors and impaired production of TNF-α and IL-2 [70]. CD39 is further involved in the pathogenesis of IBD, as polymorphisms in the ENTPD1 gene are associated with increased susceptibility to Crohn’s disease in humans and CD39-deficiency exacerbates disease severity in murine IBD models [68, 73].
Programmed cell death protein 1 (PD-1) is an inhibitory receptor expressed by various immune cells including T cells, whose ligands include PD-L1 and PD-L2. PD-1 engagement results in the reduction of downstream TCR and co-stimulatory signaling [74–76] and curtails the production of cytokines and T cell proliferation [77–80]. An increased expression of PD-1 can be detected on activated T cells and is, besides its induction upon T cell activation, also linked to T cell differentiation status [81–83]. Activity of PD-1 is vital for the prevention of autoimmunity and the maintenance of self-tolerance [84, 85]. In cancer and chronic viral infection, PD-1 signaling is critical for the development of T cell exhaustion [86–90]. Conversely, the blockade of PD-1 and other inhibitory receptors via therapeutic antibodies (a strategy termed immune checkpoint blockade) has been a breakthrough in cancer therapy [91–93].
In the chronic inflammatory skin disease psoriasis, the PD-1+ subset of the CD103+ CD8+ skin-infiltrating cells was shown to preferentially produce IL-17A compared to its PD-1- counterpart and the frequency of these PD-1+ CD103+ CD8+ cells could be correlated with disease severity [94]. Another recent study confirmed a high expression of inhibitory receptors by skin-infiltrating Tc17 cells in psoriasis and while these cells exhibited some features of dysfunction, their transcriptional profile was distinct from melanoma-infiltrating, bona fide exhausted CD8+ T cells [95].
CD27 is a transmembrane homodimer and part of the TNF-receptor family with costimulatory effects on T cell responses [96–98]. CD27-mediated costimulation is relevant for the naïve, effector, and memory stages of T cell differentiation [98–102]. Its ligand CD70 is, within immune cells, expressed on APCs, DCs, B cells, and T cells [98]. CD27 exerts its costimulatory effects through the activation of canonical and alternative nuclear factor κB (NFκB) pathways [97, 103] and has been shown to increase the expression of the anti-apoptotic molecule BCL-xL on CD4+ T cells [104]. Clinically, CD27 stimulation has been shown to synergize with PD-1 blockade to increase CD8+ T cell expansion and effector function in cancer immunotherapy [105–107].
Despite Th17 cells sharing the expression of CD161, CD26, CD6, and CD69 with their CD8+ T cell counterparts, CD27, PD-1, and CD39 were not found to be similarly differentially expressed in Th17 cells, suggesting that albeit Th17 and Tc17 cells share common characteristics, they also exhibit cell-type specific phenotypic features [3, 4, 24].
Diversity of IL-17-producing CD8+ cells
In addition to Tc17 cells that express a conventional TCR consisting of an α- and a β-chain and that recognize peptide-major histocompatibility complex (MHC)-I complexes, other types of IL-17A+ CD8+ cells comprise mucosa-associated invariant T cells (MAIT cells), γδ T cells, and natural killer T cells (NKT cells) [4]. Innate-like MAIT cells are MHC-related protein 1-restricted and express a semi-invariant αβ-TCR composed of a Vα7.2 and a TCR-β chain. Interestingly, MAIT cells in their final stage of development within the thymus can be subdivided into MAIT1 and MAIT17 cells, the latter of which share a transcriptional program closely related to the type 17 program of Tc17 and Th17 cells [108].
Unconventional γδ T cells express a namesake TCR composed of one TCR γ-chain and one TCR δ-chain and combine features of innate and adaptive immunity that allow them to act as the first line of defense during early phases of inflammation. A subset of γδ T cells, γδ 17 T cells, is able to rapidly produce IL-17A after pathogen contact, preferentially at barrier sites such as the skin or mucosal tissues [109, 110].
In the peripheral blood of healthy donors, the frequency of IL-17A+ CD8+ T cells is below 2% of CD8+ T cells, with significant increases in inflammatory diseases [4, 28, 111]. Comparing the different sources of IL-17A production, both MAIT cells and Tc17 cells make up the majority of IL-17A+ CD8+ T cells in healthy humans, followed by NKT cells and γδ T cells [4]. In an inflammatory setting, such as Crohn’s disease, the ratio of MAIT cells to conventional Tc17 cells shifts towards significantly more conventional Tc17 cells, suggesting that conventional Tc17 cells in particular drive the inflammatory pathogenesis [4]. Importantly, despite the fact that these IL-17-producing cell types share many common features, their differentiation also requires very distinct transcriptional adaptations, such as different dependence on interferon-regulatory factor 4 (IRF4), T cell factor-1 (TCF-1), and promyelocytic leukemia zinc finger [3, 112]. Prior work has largely relied on identifying IL-17-producing cells purely based on their production of IL-17 but has not necessarily distinguished these cells into the aforementioned conventional and unconventional CD8+ cell subsets. This might explain why the understanding of the protective vs. pathogenic role of Tc17 cells is still incomplete. More in-depth phenotyping of the IL-17-producing cell subsets and distinction between them in a variety of health and disease settings will help elucidate the pathogenic roles of the individual cell types.
Generation of Tc17 cells and key transcription factors
In vitro generation of Tc17 cells
CD8+ T cell differentiation is tightly regulated via a complex network of cytokines and lineage-defining transcription factors [113, 114]. While the exact requirements for Tc17 cell differentiation are still unknown, several lessons have been learned from human and murine in vitro and in vivo systems.
Based on evidence that specific cytokines such as TGF-β, IL-1β, IL-6, IL-21, and IL-23 are important for the generation of CD4+ Th17 cells [115–126], similar cytokines have been used to establish in vitro type 17 polarizing conditions (Table 1). These in vitro differentiation protocols have been used for the generation of both human and murine Tc17 cells. However, the overall in vitro human Tc17 cell yield in the literature has been low compared to the Tc17 frequencies achieved with in vitro differentiation protocols using murine cells, much like for Th17 cells [4, 28, 127–139]. The frequencies of IL-17-producing CD8+ T cells achieved under in vitro conditions using human cells range from 0.1% to 6% of CD8+ T cells, whereas up to 80% could be reached using murine T cells (Table 1). The frequencies of IL-17-producing cells achieved using different in vitro culture protocols are variable, but nevertheless some important principles and differences between in vitro and in vivo generation of Tc17 cells can be concluded.
Table 1:
Published in vitro culture conditions for generation of Tc17 cells
| Cell type | In vitro culture conditions* | Tc17 yield | Publication |
|---|---|---|---|
| Human | |||
| naïve CD8+ T cells | Culture for 5 days with anti-CD3 (2 µg/ml)/CD28 (1 µg/ml) + IL-1β IL-6 IL-23 TGF-β, further 4 days of culture with IL-2 (200 U/ml) |
0.11% IL-17A+ of CD8+ T cells | Kondo/Takata, J Immunol 2009 [28] |
| CD8+ T cells | Culture for 3 days with anti-CD3 (2 µg/ml)/CD28 (0.5 µg/ml) + IL-6 (50 ng/ml) TGF-β1 (10 ng/ml) |
25 pg/ml IL-17A (detected by ELISA) | Gras, Molecular Pharmaceutics 2012 [127] |
| Naïve CD8+ T cells | Culture for 7 days with anti-CD3 (0.5 µg/ml)/CD28 (0.5 µg/ml) + IL-1β (10 ng/ml) IL-6 (50 ng/ml) TGF-β1 (10 ng/ml) anti-IL-4 (7 µg/ml) anti-IFN-γ (7 µg/ml) |
1.9% IL-17+ of CD8+ T cells | Renavikar, Front Immunol 2020 [128] |
| PBMCs | Culture for 3 days with anti-CD3/CD28 + IL-1β (12.5 ng/ml) IL-6 (25 ng/ml) IL-23 (25 ng/ml) TGF-β (5 ng/ml) |
6.06% IL-17A+ of CD8+ T cells | Globig/Hipp, Nat Commun 2022 [4] |
| Mouse | |||
| Hapten-primed CD8+ T cells | Culture for 2 days with hapten-labeled bone-marrow-derived dendritic cells + IL-23 (5 ng/ml) |
1.4 ng/ml IL-17A (detected by ELISA), 1.5% IL-17A+ of CD8+ T cells |
He, J Immunol 2006 [129] |
| Mixed splenocytes from BALB/c and C57BL/6 mice | Culture for 5 days + IL-6 (100 ng/ml) TGF-β (1 ng/ml) |
32.8% IL-17A+ of CD8+ T cells | Liu/Tsai, J Leukoc Biol 2007 [130] |
| WT OT-1 and IFN-γ-/- OT-1 CD8+ T cells | Culture for 4 days with peptide-pulsed B cell blasts + IL-2 (4.7 ng/ml) IL-1β (10 ng/ml) IL-6 (20 ng/ml) TGF-β (3 ng/ml) anti-IL-4 (10 µg/ml) anti-IFN-γ (10 µg/ml) IL-21 (80 ng/ml) IL-23 (50 ng/ml) |
WT OT-1: 45% IL-17A+ of CD8+ T cells IFN-γ-/- OT-1: 60% IL-17A+ of CD8+ T cells |
Hamada, J Immunol 2009 [131] |
| Naïve CD8+ T cells | Culture for 2 days with peptide-pulsed-irradiated splenocytes + IL-1β (20 ng/ml) IL-6 (20 ng/ml) TGF-β (5 ng/ml) IL-23 (20 ng/ml) anti-IFN-γ (10 µg/ml) anti-IL-4 (10 µg/ml), further 3 days of resting |
43% IL-17A+ of CD8+ T cells | Yen, J Immunol 2009 [132] |
| CD8+ T cells | Culture for 3 days with anti-CD3 (1 µg/ml)/CD28 (1 µg/ml) + TGF-β1 (2 ng/ml) IL-6 (20 ng/ml) anti-IFN-γ (5µg/ml) anti-IL-2 |
70% IL-17A+ of CD8+ T cells | Ciric, J Immunol 2009 [133] |
| Naïve P14 CD8+ T cells | Culture for 5 days with GP33-41 peptide (300 ng/ml) + IL-6 (20 ng/ml) TGF-β1 (5 ng/ml) IL-23 (10 ng/ml) anti-IFN-γ anti-IL-4 |
56% IL-17A+ of CD8+ T cells | Curtis, J Immunol 2009 [134] |
| CD8+ T cells | Culture for 3 days with anti-CD3 (5 µg/ml)/CD28 (1.5 µg/ml) + TGF-β (2 ng/ml) IL-6 (50 ng/ml) or IL-21 (50 ng/ml) |
30% IL-17A+ of CD8+ T cells | Huber, Eur J Immunol 2009 [135] |
| CD8+ T cells | Culture for 3 days with anti-CD3 (4 µg/ml)/CD28 (1 µg/ml) and CD8+-depleted-irradiated splenocytes + TGF-β1 (2 ng/ml) IL-6 (100 ng/ml) anti-IFN-γ (10 µg/ml), further culture for 2 days with IL-2 (20 U/ml) |
35% IL-17A+ of CD8+ T cells | Yeh/Glosson, J Immunol 2010 [136] |
| CD8+ T cells | Culture for 3 days with anti-CD3 (3 µg/ml)/CD28 (0.5 µg/ml) + IL-6 (20 ng/ml) TGF-β1 (0.5 ng/ml) IL-2 (50 U/ml) anti-IFN-γ (5 µg/ml) |
18.8% IL-17A+ of CD8+ T cells | Huber, J Clin Invest 2013 [137] |
| CD8+ T cells | Culture for 4 days with anti-CD3 (1 µg/ml) and antigen-presenting cells + IL-6 (20 ng/ml) TGF-β (2 ng/ml) anti-IFN-γ (5 µg/ml) |
9 ng/ml IL-17A (detected by ELISA) 82% IL-17A+ of CD8+ T cells |
Flores-Santibáñez, Immunology 2015 [138] |
| CD8+ T cells | Culture for 3 days with SIINFEKL (OVA257–264) peptide (0.5 µg/ml) and CD90-depleted WT splenocytes (for OT-1) or anti-CD3 (3 µg/ml)/CD28 (0.25-4 µg/ml) (for WT B6) + IL-6 (10 ng/ml) TGF-β (2 ng/ml) IL-23 (25 ng/ml) IL-1β (5 ng/ml) anti-IFN-γ (10 µg/ml) |
55% IL-17A+ of CD8+ T cells | Arra, Oncoimmunology 2017 [139] |
*The condition with the highest Tc17 yield was listed when multiple type 17 polarizing conditions were used.
TGF-β is a common component of cytokine cocktails for the in vitro generation of Tc17 cells, but it has been shown to be dispensable for Tc17 cell generation in vivo: Transgenic animals carrying a dominant negative form of the TGF-β receptor II are still able to generate IL-17+ CD8+ T cells (but fail to generate IL-17+ CD4+ T cells), and neutralization of TGF-β only has little effect on Tc17 frequencies, highlighting different requirements for CD8+ and CD4+ IL-17+ T cell differentiation [140, 141]. The cellular requirement for IL-6 signaling to induce a Tc17 program depends on the cell’s activation status: While IL-6 is frequently used in in vitro differentiation cocktails for the generation of Tc17 cells from naïve CD8+ T cells, it might not be relevant for the conversion of already differentiated murine Tc1 cells into Tc17 cells [142]. This is likely due to the expression pattern of the IL-6 receptor, which is expressed on naïve, but not on activated CD8+ T cells [142]. Intracellular effects of IL-6, analyzed in murine CD4+ T cell differentiation, include a RORγt-independent upregulation of the Il21 gene, and RORγt-dependent effects such as the expression of Il17 [124]. In vitro addition of IL-21 increases IL-17A production by murine CD8+ T cells and further supports Tc17 polarization in combination protocols with other cytokines [131, 133, 135]. In turn, murine and human Tc17 cells themselves also produce IL-21 [143] and for Th17 cells, autocrine stimulation by IL-21 has been suggested [122, 124, 141]. Addition of IL-23 alone during activation with peptide-presenting APCs has been shown to be sufficient for in vitro induction of IL-17A production and increased expression of RORγt and RORα by some murine CD8+ T cells. This could be further enhanced by the addition of IL-6 and TGF-β [134]. Furthermore, IL-23 has been shown to be the critical signal for IL-17 production in response to Klebsiella pneumoniae infection in mice [144]. While IL-1β is a common component of the cytokine cocktails used for in vitro generation of Th17 cells, its addition only led to a minor increase in murine IL-17A-producing CD8+ T cells in one study and showed no effects in a second study [131, 132]. Despite these modest effects, it has been included in the cytokine cocktails used for in vitro Tc17 cell generation by several groups (Table 1).
Results from in vitro CD4+ T cell differentiation experiments suggest a sequential role of the aforementioned cytokines, as IL-6 appears to initially program Th17 cell differentiation together with TGF-β; and IL-21 and IL-23 act in subsequent sequential pathways that amplify Th17 programming [17, 124]. However, the applicability of this concept to CD8+ T cells is currently still unclear, particularly since the expression of the IL-23R subunits differs on naïve CD8+ T cells and naïve CD4+ T cells [145] and the effects of in vitro addition of IL-23 alone during activation of naïve murine CD8+ T cells are inconsistent [133, 134, 145]. IL-23 has also been shown to promote IL-22 production by murine Tc17 cells and alters the pathogenic capacity of Tc17 cells [133]. The addition of both TGF-β and IL-6 during activation of naïve murine CD8+ T cells induces IL-17 production with a small proportion of IFN-γ co-producing cells [130, 133]. In contrast to these Tc17-promoting cytokines, IFN-γ and IL-2 have been shown to suppress murine and human Tc17 differentiation [128, 131–133, 146, 147].
Transcriptional landscape of Tc17 cells
Most evidence for the transcriptional prerequisites of Tc17 programming stems from mouse models and Table 2 compares the key phenotypic, transcriptional, and functional features of murine and human Tc17 cells, based on the publications analyzed for this review.
Table 2:
Published features of human and murine Tc17 cells
| Human | Mouse | |
|---|---|---|
| Tc17-promoting cytokines | TGF-β, IL-6, IL-1β, IL-23 | TGF-β, IL-6, IL-1β, IL-23, IL-21, TNF-α |
| Tc17-repressing cytokines | IFN-γ, IL-4 | IFN-γ, IL-4, IL-2 |
| Tc17-promoting transcription factors | RORγt, STAT3 | RORγt, STAT3, IRF4 |
| Tc 17-repressing transcription factors | IRF3, EOMES, T-BET, BLIMP1, TCF-1 | |
| Tc17 effector functions | IL-17A, IL-17F, IFN-γ, TNF-α, IL-22, XCL-1 |
IL-17A, IL-17F, IFN-γ, TNF-α, GM-CSF, IL-13, IL-21, IL-22, IL-5 |
| Tc17 surface markers | CCR5, CCR6, IL-23R CD26, CD161, CD6, CD69, CD39, PD-1 |
CCR6, IL-23R |
Both IL-6 and IL-21 are potent activators of STAT3. Indeed, similar to Th17 cells that rely on the activity of STAT3 [21, 22, 124], STAT3 activity has also been found to be essential for the generation of murine Tc17 cells [132, 135, 143] and is upregulated in human Tc17 cells (Table 2) [4]. Mechanistically, activation of STAT3 is crucial for the expression of the type 17 master regulators of the ROR family, RORα and RORγt, and STAT3 has been found to direct the molecular switch between type 17 and cytotoxic lymphocyte differentiation programs in murine cells [20–23, 135]. Furthermore, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) has been identified as a stabilizing factor of IL-17 production and Tc17 differentiation via control of STAT3 activity. Consequently, murine CTLA-4-deficient T cells express low levels of type 17 signature molecules [139]. In contrast to the Tc17-promoting role of STAT3, STAT5 has been found to repress IL-17 production in both murine CD4+ Th17 and CD8+ Tc17 cells [146].
The orphan nuclear receptor RORγt is required for the differentiation of murine Th17 cells and the expression of the Il17a and Il17f genes [23]. While RORγt is also highly expressed in both murine and human Tc17 cells and, by binding to the IL-17 conserved noncoding sequence-2-enhancer, increases IL-17 production in CD8+ T cells, its overexpression alone in murine CD8+ T cells under non-Tc17 polarizing conditions only results in a minor increase in Tc17 cells [3, 135, 148], indicating additional transcriptional changes are needed for induction of Tc17 cells.
The downregulation of T-box transcription factors T-BET and EOMES, or T-BET and BLIMP-1 is closely correlated with the capacity of murine CD8+ T cells to produce IL-17 [134, 135, 147, 149–151]. Interestingly, Tbx21 and Eomes murine double knockout (KO) cells, but not Tbx21 or Eomes single KO CD8+ T cells, upregulate mRNA levels of the type 17 markers Rorc, Il23r, Il17a, Il21, and Il22 [151].
The transcription factor aryl hydrocarbon receptor (AhR) is upregulated in murine and human Th17 cells [152]. Inhibition or deletion of AhR during in vitro polarization impairs the production of IL-17A and abrogates the production of IL-22 by murine Th17 cells [152, 153], effects which imply that AhR is especially important for more plastic Th17 cell populations [154]. Mechanistic studies using murine CD4+ T cells under Th17 polarizing conditions show that AhR inhibits STAT1 [155] and reduces IL-2 expression by induction of Ikzf3, encoding the transcription factor AIOLOS [156]. During in vitro culture of murine Th17 cells, sustained AhR expression was dependent on the presence of TGF-β1 [157]. Genetic deletion or inhibition of AhR also impaired murine Tc17 cell development, but, unlike in CD4+ Th17 cells, binding of AhR to its ligand 6-formylindolo[3,2-b]carbazole (FICZ) did not increase IL-17A production in AhR+/- mouse CD8+ Tc17 cells [153]. These data suggest that, unlike in Th17 cells, the baseline activity of AhR might suffice for the induction of murine Tc17 cells.
Other orchestrators of Tc17 cell differentiation include Tcf7, Irf3, and Irf4: A loss of Tcf7 in murine cells modulates chromatin accessibility with increased accessibility of the Rorc, Il17a, and Il17f genes, and reduced accessibility of the Tbx21, Eomes, and Irf4 loci [3]. Furthermore, Tcf7 suppresses Tc17 differentiation via suppression of the MAF (MAF BZIP transcription factor) – RORγt axis [3]. IRFs are key components of the activation of innate antimicrobial response pathways and also contribute to crossroad decisions within T cell differentiation. IRF3 restrains Tc17 differentiation by directly interacting with RORγt through its IRF interaction domain and thereby preventing the binding of RORγt to the Il17 promoter [148]. In contrast, IRF4 promotes the generation of Tc17 and Th17 cells. Irf4 deletion prevents the generation of Tc17 and Th17 cells due to lower expression of the Tc17-promoting transcription factors Rorc and Rora, as well as lower expression of Il23r and Il21, but higher expression of the Tc17-repressing transcription factor Eomes [137]. In sum, the development and maintenance of Tc17 cells not only depend on the presence of specific cytokines but also require specific transcriptional adaptations.
Tc17 plasticity
Tc17 cells are a highly plastic cell type, especially towards Tc1 or Tc2 fates. CTLA-4 deficiency and reactive oxygen species/IL-2 signaling have both been found to skew Tc17 cell differentiation towards a Tc1 fate with increased IFN-γ, TNF-α, perforin, and granzyme production as well as increased EOMES, T-BET, and BLIMP-1 expression [136, 139, 146, 158]. In contrast, in the context of tissue injury, murine Tc17 cells can acquire a Tc2 phenotype with secretion of IL-5 and IL-13 and expression of GATA3 upon exposure to IL-1, IL-18, and IL-33 [159]. Despite this plasticity, stable Tc17 phenotypes have been described in the setting of murine fungal infection, where Tc17 cells form a TCF1hi, TBETlow, and EOMESlow IL-17-producing memory cell population that persists over long periods of time [41, 160–162]. In summary, Tc17 cells are known to be highly plastic and can differentiate into Tc1 or Tc2 cells in specific cytokine milieus, but can also maintain a stable Tc17 phenotype. Since the transfer of Tc17 cells is currently being evaluated in adoptive cell therapy settings to improve anti-cancer immunity [139, 163, 164], this high degree of plasticity needs to be considered.
Tc17 cells in infectious diseases
Fungal infection
One hallmark task of type 17 immunity is defense against fungal infections. This is corroborated by the clinical observation that immunodeficient patients with defects in IL-17A and IL-17F signaling due to genetic deficiency of the IL-17 receptor subunit IL-17RA, deficiency in IL-17F or mutations in the downstream adaptor protein NFκB activator 1 (ACT1) often suffer from chronic mucocutaneous candidiasis (CMC) [165–167]. Moreover, patients with dominant-negative mutations in STAT3 show diminished frequencies of IL-17A- and IL-22-producing T cells, and also frequently develop CMC [168]. Furthermore, mucocutaneous candidiasis is a known complication of anti-IL-17A antibody therapy [169], highlighting the importance of IL-17 for defense against fungal infections.
In line with this, IL-17RA KO mice are highly susceptible to local and systemic fungal infection [11, 170]. Mechanistically, IL-17 receptor signaling is responsible for the timely and localized migration of neutrophils to infected organs and for the production of antimicrobial peptides and chemokines [11, 170, 171]. As an additional mechanism for the control of fungal infections, IL-17A has been shown to be necessary for the development of functional NK cells that are crucial for antifungal defense [172].
Historically, Th17 cells were identified as the key producers of IL-17A in anti-fungal protective immunity [11, 166, 173]. Notably however, in a mouse model of local fungal infection using CD4+ T cell-deficient mice, IL-17A-secreting CD8+ T cells were able to compensate for the lack of Th17 cells and provided protection against oropharyngeal candidiasis [174]. Furthermore, fungal vaccination in CD4+ T cell-deficient mice led to the development of vaccine immunity mediated by Tc17 cells. These vaccine-elicited Tc17 cells were able to protect the CD4+-deficient hosts from lethal fungal pneumonia, persisted and provided long-lasting memory [41, 160]. Thus, Tc17 cells are sufficient to compensate for the lack of Th17 cells in the setting of fungal infection.
Viral infection
Compared to fungal infections, the role of Tc17 cells in viral infections is less understood. In mouse models of acute viral infection (influenza virus, vaccinia virus), virus-specific Tc17 cells expand, can acquire a cytotoxic phenotype, and provide protection from lethal infection [131, 136]. In influenza infection, this was associated with enhanced migration of neutrophils into the infected lung [131]. However, mice lacking IL-17A due to genetic deficiency or antibody-mediated depletion had increased viral burden but were still able to clear the virus [136, 175, 176], indicating that IL-17A is enhancing anti-viral immunity but is not necessary for viral clearance. Additionally, in vitro polarized Tc17 cells can acquire an IFN-γ-secreting Tc1 phenotype after adoptive transfer and clear vaccinia virus infection [136].
Tbx21 and Eomes double KO CD8+ T cells acquire a type 17 transcriptional profile during acute LCMV infection with the Armstrong strain. This is associated with CD8+ T cell-dependent inflammation and wasting syndrome accompanied by multi-organ infiltration of neutrophils in the double KO mice [151]. These findings highlight that Tc17 cells can drive immunopathology in viral infections, especially manifested in tissues, in accordance with their importance in immune-mediated tissue diseases such as IBD and psoriasis.
In humans with chronic HCV infection, the frequency of polyfunctional Tc17 cells able to coproduce IL-10, IFN-γ, or IL-21 alongside with IL-17A in the peripheral blood was found to be negatively correlated with the degree of hepatic damage, while the frequency of mono-functional Tc17 cells positively correlated with hepatic damage [177]. In contrast, in the liver of patients with chronic HCV infection and advanced fibrosis, Tc17 cells as well as IFN-γ-coproducing Tc17 cells were found to be increased compared to the peripheral blood of these patients [27, 178]. The role of Tc17 cells for the progression of liver fibrosis is still unclear: IL-17A can contribute to the progression of liver fibrosis by activating hepatic stellate cells [179]. However, the stage of fibrosis in patients with chronic HCV infection was inversely correlated with the frequency of intrahepatic IL-17A/IFN-γ-producing CD8+ T cells [27]. These seemingly contradictory findings might be explained by the presence of other IL-17A-producing cell types [179–182]. The literature regarding Tc17 cells in HIV-infected individuals is still inconclusive. While HIV-specific Tc17 cells have been described, observations on overall frequencies of Tc17 cells in HIV-infected individuals compared to control donors are so far variable [183, 184].
In summary, virus-specific Tc17 cells have been identified in both murine and human viral infections, and there is evidence that Tc17 cells enhance the antiviral immune response in murine infections. Importantly, Tc17 responses in viral infections are associated with increased tissue immunopathology.
Bacterial infection
Consistent with the pathognomonic role of IL-17 in barrier tissue immunity, elevated levels of IL-17A have been described in the gastric epithelium of humans infected with Helicobacter pylori and were associated with secretion of IL-8 by gastric epithelial cells, which in turn promoted neutrophil chemotaxis [185]. Furthermore, in mouse models, an important role of IL-17A and IL-17 receptor signaling in the defense against different bacteria like Streptococcus pneumoniae, Bordetella pertussis, and Klebsiella pneumoniae has been identified [10, 186–188]. IL-17A enhances the migration of neutrophils to the site of infection as well as their phagocytic activity [186] and promotes the eradication of intracellular bacteria like Chlamydia muridarum by increasing the activity of the inducible nitric oxide synthase [189]. Most of these IL-17 effects have been attributed to the activity of Th17 cells [186–188], have been demonstrated in mouse models, and suggest that the field is only starting to understand the role of Tc17 cells in the defense against bacterial infections in barrier tissues.
Tc17 cells in cancer
Tc17 cells have been comparatively well characterized in gastroenterological malignancies like gastric cancer, bile duct cancer, and hepatocellular carcinoma (HCC). While there is conflicting data on the frequency of circulating Tc17 cells in patients with gastric cancer [190–192], it has repeatedly been shown that Tc17 cells accumulate in the tumors [190, 192, 193]. Increased intra-tumoral Tc17 cell frequencies are associated with tumor progression and predict a lower overall survival and disease-free survival [190–192]. Tc17 accumulation has been described to occur upon production of IL-6, IL-1β, and IL-23 by tumor-activated monocytes [192]. Intratumoral Tc17 cells themselves then induce the production of CXCL12 by the tumor cells, which causes the recruitment of myeloid-derived suppressor cells and consecutive suppression of cytotoxic CD8+ T cell responses [192]. Single-cell RNA sequencing additionally revealed increases in receptor expression for Tc17-derived cytokines like IL17RA/IL17RC and IL22RA1 in gastric cancer cells and it has been hypothesized that these cytokine-receptor interactions promote tumor growth [193].
In accordance with findings in gastric cancer, Tc17 cells have been found to be enriched within HCC, and localize mainly to the tumor margin compared to both intra-tumoral tissue and surrounding non-tumor liver tissue [194, 195]. Most of these Tc17 cells in the tumor margin are able to produce IFN-γ in addition to IL-17A, as opposed to the dominant subset of circulating Tc17 cells in HCC patients, which do not coproduce IFN-γ [194]. Furthermore, intra-tumoral Tc17 cells also express CCL20, CCR4, CCR6, and CXCR6 which have previously been associated with tumor progression in HCC patients [196–198]. Suggested mechanisms for a contribution of Tc17 cells to an immunosuppressive tumor microenvironment include the recruitment of regulatory T cells (TREG) via the CCR6-CCL20 axis indicated by increased levels of CCL20 in HCC samples, especially in those infiltrated with IFN-γ-negative Tc17 cells [195]. Overall, the high intra-tumoral density of IL-17A-producing cells and increased intra-tumoral frequency of IFN-γ-negative Tc17 cells were associated with poorer prognosis in HCC [195, 199]. In patients with bile duct cancer, an elevated frequency of RORγt and T-bet co-expressing CD8+ T cells in the peripheral blood was described and Tc17 cells were characterized by a terminal differentiation phenotype with increased TCR signaling despite increased expression of the inhibitory receptors LAG3 and TIM3 [200].
In sum, the current literature points towards an association of Tc17 cells with tumor progression in humans, contrary to data generated in mouse models: Transfer of in vitro polarized tumor-reactive Tc17 cells into tumor-bearing mice led to enhanced anti-tumor effects compared to control unpolarized CD8+ T cells. Notably, these in vitro polarized transferred Tc17 cells differentiated into IFN-γ-producing effector cells [158] similar to observations in murine viral infection. Therefore in vitro differentiated Tc17 cells might not recapitulate all features of in vivo differentiated, disease-associated Tc17 cells and murine and human Tc17 cells appear to differ in their anti-tumor function.
Tc17 cells in immune-mediated diseases
Psoriasis
Consistent with the established role of IL-17 in barrier tissue immunity, in healthy human and primate skin, as well as in wild-caught mice, the presence of Tc17 cells is physiological and considered part of the equilibrium forming barrier immunity [201, 202]. In contrast, specific pathogen-free (SPF) mice have reduced Tc17 frequencies in the skin at baseline [201], but colonization of SPF mice with defined skin commensals like Staphylococcus epidermidis induces accumulation of CD8+ and Tc17 cells in the skin [201, 202].
Psoriasis is a chronic disease of the skin affecting ~1–2% of the population and is considered to be the most frequent immune-mediated disorder in humans [203, 204]. Most studies refer to plaque psoriasis, which is the most common form of the disease [203, 204]. The frequency of Tc17 cells in the peripheral blood of psoriasis patients correlates with the clinical severity of the disease [205]. Within psoriatic lesions, Tc17 cells are increased compared to the skin of healthy donors [206–209] and the IL-23/IL-17 axis has been shown to sustain psoriatic plaques in humans and mice [210–212]. The antigens targeted by Tc17 cells are presented by the main psoriasis risk gene HLA-Cw6 [213] and other HLA-class I alleles and include the antimicrobial peptide LL-37 (expressed by keratinocytes) [214], the melanocyte-derived antigen ADAMTS-like protein 5 [215], and keratin 17 [216].
Interestingly, in human skin, CD49a expression of CD8+ T cells differentiates between IFN-γ and IL-17 production, with CD49a+ T cells preferentially producing IFN-γ and CD49a- T cells preferentially producing IL-17. CD49a- CD8+ T cells isolated from psoriatic lesions mainly generate IL-17 responses and promote local inflammation [63]. Depleting CD8+ T cells prevents psoriasis development in mouse models, indicating that CD8+ T cells are already necessary at the early stages of the disease [217].
Around 20% of psoriasis patients develop psoriasis arthritis [218], which in contrast to rheumatoid arthritis is associated with the presence of Tc17 cells in synovial fluid [219, 220], suggesting the specific disease pathogenesis rather than localization of the inflammation determines the presence of Tc17 cells.
Based on this evidence, monoclonal antibodies targeting the IL-17 program have made their way into clinical practice: Treatment of psoriasis vulgaris with secukinumab (targeting IL-17A) or ustekinumab (targeting IL-12/IL-23) led to a rapid reduction of psoriasis symptoms in clinical studies, corroborating the detrimental role of the IL-23/IL-17-axis in the pathogenesis of psoriasis [221, 222]. Furthermore, ixekizumab (anti-IL-17A), brodalumab (anti-IL-17RA), and risankizumab (targeting IL-23A) have proven to be effective [13, 223, 224]. More recently, bimekizumab, a bispecific antibody targeting both IL-17A and IL-17F has been approved [225–227]. Interestingly, dual blockade of IL-17A and IL-17F led to a better clinical response compared to blockade of IL-17A alone with secukinumab, but was associated with a higher rate of oropharyngeal candidiasis [227]. Another dual-inhibitor strategy was tested in clinical trials with COVA322, which inhibits both IL-17A and TNF-α [228], but had to be terminated because of its safety profile (NCT02243787). In Asia, an antibody targeting CD6 expressed by Tc17 cells, itolizumab, is in clinical use for the treatment of psoriasis with the rationale of modulation of CD6-dependent signaling pathways in Tc17 cells [4, 43, 50, 229].
Multiple sclerosis
MS is the most prevalent chronic inflammatory disease of the central nervous system [230]. Early studies described IL-17A-expressing immune cells in the cerebrospinal fluid (CSF) of MS patients [231]. IL-17A production was initially mainly attributed to Th17 cells and their pathogenic role was comprehensively characterized in both patients and murine models of MS [232–235]. Other work then found the frequency of Tc17 cells to be increased in the CSF of MS patients compared to their peripheral blood, and compared to the CSF of control patients [137, 236, 237] and demonstrated a positive correlation between the Tc17 frequency in CSF and the level of disability [238], suggesting a causative link between Tc17 cells and the progression of MS. Furthermore, Tc17 cells were found to be infiltrating the CNS tissue and to be present in both acute and chronic demyelinating lesions of MS patients [233]. In relapsing-remitting MS (RRMS), the most common form of MS, higher frequencies of Tc17 cells, also able to produce IFN-γ and TNF-α, have been described in the peripheral blood in the relapse phase of the disease compared to the remission phase [239].
Murine models of IL-17-deficiency such as Irf4-deficient mice that fail to generate Tc17 and Th17 cells and IL-17A-deficient mice are resistant to induction of experimental autoimmune encephalitis (EAE), a common murine model for MS [137, 240, 241]. Interestingly, an adoptive transfer of either wild-type (WT) CD4+ or WT CD8+ T cells alone into Irf4-deficient mice failed to evoke EAE symptoms, only the combined transfer of both WT CD4+ and WT CD8+ T cells induced the disease. Mechanistically, IL-17A production by CD8+ T cells was necessary to induce differentiation and CNS infiltration of Th17 cells and subsequent development of EAE, suggesting that CD8+ Tc17 cell help promotes Th17 cell pathogenicity [137].
Currently, more than a dozen disease-modifying drugs to slow down the progression of MS and prolong relapse-free periods are available [230]. No effects of IFN-β, glatiramer acetate, or vitamin D supplementation on the frequency of Tc17 cells in the peripheral blood of MS patients could be detected [237]. In contrast, responders to treatment with dimethyl fumarate (DMF) had a significantly increased frequency of peripheral blood Tc17 cells before treatment initiation compared to non-responders. Clinical and radiological response in this subgroup of MS patients was in turn then linked to a marked decrease in Tc17 cells. Notably, the frequencies of CD4+ Th17 cells remained unaffected in responders and non-responders [146]. This points towards a prognostic role of Tc17 cells in the therapeutic response of MS patients and could support personalized therapy approaches. Mechanistically, in the EAE mouse model, DMF has been demonstrated to reduce Tc17-dependent Th17 pathogenicity by reducing IL-17A production by Tc17 cells [146].
Murine studies suggested that the administration of anti-IL-17A antibodies was especially efficient to prevent relapse in a relapsing-remitting model of EAE, while their administration at the peak of the disease had only limited effect [242]. In patients with RRMS, treatment with the IL-17A antibody secukinumab also reduced MR-detectable lesion activity in a proof-of-concept study without notable serious adverse events [243]. A subsequent placebo-controlled, randomized, phase II trial of secukinumab (NCT01874340) was however terminated based on the availability of another human anti-IL-17 antibody, CJM112, which is currently in a phase II trial [13, 244, 245]. In sum, these data suggest that targeting the IL-17 pathway might be a promising therapeutic direction in MS.
Inflammatory bowel disease
The intestinal immune system is tasked with the challenge of maintaining barrier function and defense against infection while simultaneously tolerating a vast number and variety of intestinal microbiota [246]. In IBD, a disease group comprising Crohn’s disease (CD) and ulcerative colitis (UC), a complex interplay of genetic and environmental factors and impaired intestinal barrier function predisposes individuals to the development of an excessive immune response with acute and chronic inflammatory processes. This leads to further epithelial damage, impaired barrier function, and complications such as abscess formation, stenosis, and increased risk of malignancy [114, 247, 248]. In both CD and UC, IL-17A levels are increased in the serum and mucosa of patients, compared to ischemic or infectious colitis or healthy controls [249]. The involvement of Th17 cells has been previously described [250, 251]. More recent studies have focused on the role of Tc17 cells.
In both healthy controls and IBD patients, Tc17 frequencies are higher in the intestine than in the peripheral blood, consistent with a role in the maintenance of mucosal immune homeostasis [111]. Increased inflammation was found to be associated with Tc17 cell expansion in UC [252]. In accordance with this, our group has published an enrichment of IL-17-producing CD8+ T cells with a potentially targetable signature in the peripheral blood and intestinal tissue of patients with active CD [4]. We found this enrichment to be mainly due to conventional αβ-CD8+ Tc17 cells rather than unconventional T cell subsets. Disease-associated Tc17 cells produced mainly IL-17A and only small amounts of IL-17F, and expressed a distinct immune signature of CD6+, CD39+, CD69+, PD-1+, CD26+, CD161+, and CD27low which was also linked to flare-free survival of CD patients [4]. Interestingly, in contrast to the increase in IL-17+ CD8+ T cells, neither IFN-γ+ CD8+ T cells nor TNF-α+ CD8+ T cells were significantly increased in the peripheral blood of CD patients. While the total population of IL-17+ CD8+ T cells was increased in active disease, their polyfunctionality profile did not change in active disease with a preferential coproduction of TNF-α over IFN-γ possibly intrinsically linked to the Tc17 differentiation program. Targeting CD6 reduced the production of proinflammatory cytokines by peripheral blood and intestinal Tc17 cells in vitro [4]. In accordance with human data, IL-17+ CD8+ T cells have been linked to the development of severe colitis in mice [253]. A further mechanistic link between Tc17 cells and dysregulated microbiota in IBD patients could be short-chain fatty acids (SCFAs): Some SCFAs were able to increase expression of IFN-γ and granzyme B, thereby increasing the cytotoxic activity of murine Tc17 cells in vitro [254].
In analogy to the successful targeting of the IL-17 axis in psoriasis, the anti-IL-17A antibody secukinumab and the IL-17R targeting antibody brodalumab were evaluated in clinical trials with IBD patients, but these trials unexpectedly had to be terminated. While secukinumab treatment was inefficient and led to an increased rate of infections, treatment with brodalumab was not efficient and even caused disease exacerbation in a disproportionate number of CD patients [255, 256]. Aside from the previously discussed role of IL-17 in the defense against fungal infections, this might be explained by the importance of IL-17 for the maintenance of gastrointestinal barrier integrity: IL-17A-dependent signaling regulates the tight junction protein occludin, and subsequently, IL-17 neutralization increases gut permeability in a DSS (Dextran sulfate sodium)-induced colitis mouse model [257, 258]. Furthermore, for Th17 cells, an autoregulatory loop limiting pathogenicity through IL-17 effects has been described and might have been therapeutically impaired [259].
In contrast to selective targeting of the cytokine IL-17, combined targeting of the IL-23/IL-17 axis with ustekinumab, a monoclonal antibody targeting the shared p40 subunit of IL-12 and IL-23, is an effective treatment strategy in IBD [260]. While ustekinumab is considered to inhibit the differentiation of Th1 and Th17 cells, it has also been shown to affect the differentiation of T follicular helper cells [261]. Another novel drug targeting the IL-23/IL-17 axis via targeting of an IL-23 subunit is risankizumab, which binds to the p19 subunit of IL-23, and has been shown to be effective in CD [262, 263]. In addition to affecting the Tc17 program, IL-23 triggers other pro-inflammatory processes such as reduced TREG function and increased T cell production of TNF-α and GM-CSF [258, 264]. Interestingly, protective IL-17A secretion by γδ T cells was not impaired in settings of IL-23 inhibition [257].
In summary, these clinical studies suggest that targeting the IL-17 axis, but not necessarily selectively targeting the cytokine IL-17 itself can be a viable strategy in the treatment of IBD. Therefore, further expanding our knowledge of how Tc17 cells are generated and how their function is regulated in vivo might allow us to identify novel therapeutic targets.
Conclusion
The last two decades have identified IL-17-producing T cells as uniquely polarized immune cells at the center of many immune-mediated diseases such as IBD. Targeting the IL-17 pathway is an effective treatment strategy for a variety of diseases. While most studies have focused on CD4+ Th17 cells as the classical IL-17-producing T cell population, a growing body of evidence suggests that CD8+ Tc17 cells are similarly involved in the pathogenesis of many immune-mediated diseases and infections. Due to the diversity of conventional and unconventional CD8-expressing cell types that can produce IL-17, such as αβ Tc17 cells, γδ T cells, MAIT cells, and NKT cells, the pathogenic versus protective role of IL-17-producing CD8+ T cells is still incompletely understood. Particularly in humans, the environmental and transcriptional requirements for the generation of Tc17 cells are not yet well defined. Future mechanistic studies are needed to allow a deeper understanding of this novel T cell population and will help to expand on currently available targeted therapies.
Acknowledgements
We would like to thank all members of the Translational Experimental Immunology lab Freiburg for helpful discussions. Figure 1 was created with BioRender.com. The editor-in-chief, Simon Milling, and handling editor, Emily Gwyer Findlay, would like to thank the following reviewers, Kathryn Steel and an anonymous reviewer, for their contribution to the publication of this article.
Glossary
Abbreviations:
- ADAMTS
a disintegrin and metalloproteinase with thrombospondin motifs
- AhR
aryl hydrocarbon receptor
- ALCAM
activated leukocyte cell adhesion molecule/CD166
- ATP
adenosine triphosphate
- BCL-xL
B-cell lymphoma-extra large
- Blimp-1
B lymphocyte-induced maturation protein-1
- CCL
C-C motif chemokine ligand
- CCR
C-C motif chemokine receptor
- CD
Crohn’s disease
- CMC
chronic mucocutaneous candidiasis
- CNS
central nervous system
- CSF
cerebrospinal fluid
- CTLA-4
cytotoxic T-lymphocyte-associated protein 4
- CXCL
chemokine (C-X-C motif) ligand
- CXCR
chemokine (C-X-C motif) receptor
- DMF
dimethyl fumarate
- DSS
dextran sulfate sodium
- EAE
experimental autoimmune encephalomyelitis
- ENTPD1
ectonucleoside triphosphate diphosphorylase 1/CD39
- Eomes
eomesodermin
- FICZ
6-formylindolo[3,2-b]carbazole
- G-CSF
granulocyte colony-stimulating factor
- GM-CSF
granulocyte-macrophage colony-stimulating factor
- HCC
hepatocellular carcinoma
- HCV
hepatitis C Virus
- HIV
human immunodeficiency virus
- HLA
human leukocyte antigen
- IBD
inflammatory bowel disease
- IFN-γ
interferon-γ
- Ikzf
Ikaros family zinc finger protein
- IL
interleukin
- IRF
interferon regulatory factor
- JAK
Janus kinase
- KO
knockout
- LAG3
lymphocyte-activation gene 3
- LAT1
L-type amino acid transporter 1
- LLT1
lectin-like transcript 1
- MAIT cell
mucosa associated invariant T cell
- MHC
major histocompatibility complex
- mRNA
messenger RNA
- MS
multiple sclerosis
- NFκB
nuclear factor κB
- NKT cell
natural killer T cell
- PD-1
programmed cell death protein 1
- PLZF
promyelocytic leukemia zinc finger
- RORγt
retinoic acid receptor-related orphan receptor-γt
- RRMS
relapsing remitting MS
- S1PR1
sphingosine-1-phosphate receptor 1
- SCFAs
short-chain fatty acids
- SPF
specific pathogen free
- STAT
signal transducer and activator of transcription
- T-bet
T-box expressed in T cells
- Tc1
cytotoxic T lymphocyte
- Tc17
interleukin 17-producing CD8+ T cell
- Tc2
Type 2 CD8+ T cell
- Tc9
interleukin-9-producing CD8+ T cell
- TCF-1
T cell factor-1
- TCR
T cell receptor
- TGF-β
transforming growth factor-β
- Th1
T helper 1 cell
- Th17
T helper 17 cell
- Th2
T helper 2 cell
- Tim3
T cell immunoglobulin and mucin-domain containing 3
- TNF-α
tumor necrosis factor-α
- Treg
regulatory T cell
- TRM
tissue resident memory cells
- UC
ulcerative colitis
- WT
wild type
Contributor Information
Anna Veronika Hipp, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany.
Bertram Bengsch, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany.
Anna-Maria Globig, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany.
Conflict of interest:
The authors declare no conflict of interest.
Funding
AMG was supported by the German Research Foundation (Deutsche Forschungsgemeinschaft, GL 991/1-1).
Data availability
No new data were generated in this study.
Author contributions
Conceptualization – A.M.G., B.B. Writing (original draft) – A.V.H. Writing (review and editing) – A.M.G., B.B., and A.V.H. Visualization – A.M.G., A.V.H. All authors have read and agreed to the published version of the manuscript.
References
- 1.Annunziato F, Romagnani C, Romagnani S.. The 3 major types of innate and adaptive cell-mediated effector immunity. J Allergy Clin Immunol 2015, 135, 626–35. doi: 10.1016/j.jaci.2014.11.001. [DOI] [PubMed] [Google Scholar]
- 2.Mittrücker HW, Visekruna A, Huber M.. Heterogeneity in the differentiation and function of CD8+ T cells. Arch Immunol Ther Exp (Warsz) 2014, 62, 449–58. doi: 10.1007/s00005-014-0293-y. [DOI] [PubMed] [Google Scholar]
- 3.Mielke LA, Liao Y, Clemens EB, Firth MA, Duckworth B, Huang Q, et al. TCF-1 limits the formation of Tc17 cells via repression of the MAF–RORγt axis. J Exp Med 2019, 216, 1682–99. doi: 10.1084/jem.20181778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Globig AM, Hipp AV, Otto-Mora P, Heeg M, Mayer LS, Ehl S, et al. High-dimensional profiling reveals Tc17 cell enrichment in active Crohn’s disease and identifies a potentially targetable signature. Nat Commun 2022, 13, 3688. doi: 10.1038/s41467-022-31229-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Liang Y, Pan H-F, Ye D-Q.. IL-17A-producing CD8+T cells as therapeutic targets in autoimmunity. Expert Opin Ther Targets 2015, 19, 651–61. doi: 10.1517/14728222.2014.997710. [DOI] [PubMed] [Google Scholar]
- 6.Mills KHG. IL-17 and IL-17-producing cells in protection versus pathology. Nat Rev Immunol 2023, 23, 38–54. doi: 10.1038/s41577-022-00746-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Miossec P, Korn T, Kuchroo VK.. Interleukin-17 and type 17 helper T cells. N Engl J Med 2009, 361, 888–98. doi: 10.1056/NEJMra0707449. [DOI] [PubMed] [Google Scholar]
- 8.Toy D, Kugler D, Wolfson M, Vanden Bos T, Gurgel J, Derry J, et al. Cutting edge: interleukin 17 signals through a heteromeric receptor complex. J Immunol 2006, 177, 36–9. doi: 10.4049/jimmunol.177.1.36. [DOI] [PubMed] [Google Scholar]
- 9.Veldhoen M. Interleukin 17 is a chief orchestrator of immunity. Nat Immunol 2017, 18, 612–21. doi: 10.1038/ni.3742. [DOI] [PubMed] [Google Scholar]
- 10.Ye P, Rodriguez FH, Kanaly S, Stocking KL, Schurr J, Schwarzenberger P, et al. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. J Exp Med 2001, 194, 519–27. doi: 10.1084/jem.194.4.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.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: 10.1084/jem.20081463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley K.. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity 2005, 22, 285–94. doi: 10.1016/j.immuni.2005.01.011. [DOI] [PubMed] [Google Scholar]
- 13.Omidian Z, Ahmed R, Giwa A, Donner T, Hamad ARA.. IL-17 and limits of success. Cell Immunol 2019, 339, 33–40. doi: 10.1016/j.cellimm.2018.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lückel C, Picard FSR, Huber M.. Tc17 biology and function: novel concepts. Eur J Immunol 2020, 50, 1257–67. doi: 10.1002/eji.202048627. [DOI] [PubMed] [Google Scholar]
- 15.Reynolds JM, Angkasekwinai P, Dong C.. IL-17 family member cytokines: regulation and function in innate immunity. Cytokine Growth Factor Rev 2010, 21, 413–23. doi: 10.1016/j.cytogfr.2010.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chang SH, Dong C.. IL-17F: regulation, signaling and function in inflammation. Cytokine 2009, 46, 7–11. doi: 10.1016/j.cyto.2008.12.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dong C. Differentiation and function of pro-inflammatory Th17 cells. Microbes Infect 2009, 11, 584–8. doi: 10.1016/j.micinf.2009.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.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: 10.1093/emboj/20.19.5332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yamaguchi Y, Fujio K, Shoda H, Okamoto A, Tsuno NH, Takahashi K, et al. IL-17B and IL-17C are associated with TNF-alpha production and contribute to the exacerbation of inflammatory arthritis. J Immunol 2007, 179, 7128–36. doi: 10.4049/jimmunol.179.10.7128. [DOI] [PubMed] [Google Scholar]
- 20.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: 10.1016/j.immuni.2007.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.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: 10.1016/j.immuni.2007.02.009. [DOI] [PubMed] [Google Scholar]
- 22.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: 10.1074/jbc.C600321200. [DOI] [PubMed] [Google Scholar]
- 23.Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, et al. The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006, 126, 1121–33. doi: 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
- 24.Bengsch B, Seigel B, Flecken T, Wolanski J, Blum HE, Thimme R.. Human Th17 cells express high levels of enzymatically active dipeptidylpeptidase IV (CD26). J Immunol 2012, 188, 5438–47. doi: 10.4049/jimmunol.1103801. [DOI] [PubMed] [Google Scholar]
- 25.Fergusson JR, Smith KE, Fleming VM, Rajoriya N, Newell EW, Simmons R, et al. CD161 defines a transcriptional and functional phenotype across distinct human T cell lineages. Cell Rep 2014, 9, 1075–88. doi: 10.1016/j.celrep.2014.09.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Fergusson JR, Hühn MH, Swadling L, Walker LJ, Kurioka A, Llibre A, et al. CD161intCD8+ T cells: a novel population of highly functional, memory CD8+ T cells enriched within the gut. Mucosal Immunol 2016, 9, 401–13. doi: 10.1038/mi.2015.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Billerbeck E, Kang Y-H, Walker L, Lockstone H, Grafmueller S, Fleming V, et al. Analysis of CD161 expression on human CD8+ T cells defines a distinct functional subset with tissue-homing properties. Proc Natl Acad Sci U S A 2010, 107, 3006–11. doi: 10.1073/pnas.0914839107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kondo T, Takata H, Matsuki F, Takiguchi M.. Cutting edge: phenotypic characterization and differentiation of human CD8+ T cells producing IL-17. J Immunol 2009, 182, 1794–8. doi: 10.4049/jimmunol.0801347. [DOI] [PubMed] [Google Scholar]
- 29.Appay V, Van Lier RAW, Sallusto F, Roederer M.. Phenotype and function of human T lymphocyte subsets: consensus and issues. Cytom Part A 2008, 73, 975–83. doi: 10.1002/cyto.a.20643. [DOI] [PubMed] [Google Scholar]
- 30.Maggi L, Santarlasci V, Capone M, Peired A, Frosali F, Crome SQ, et al. CD161 is a marker of all human IL-17-producing T-cell subsets and is induced by RORC. Eur J Immunol 2010, 40, 2174–81. doi: 10.1002/eji.200940257. [DOI] [PubMed] [Google Scholar]
- 31.Braud VM, Meghraoui-Kheddar A, Elaldi R, Petti L, Germain C, Anjuère F.. LLT1-CD161 interaction in cancer: promises and challenges. Front Immunol 2022, 13, 847576. doi: 10.3389/fimmu.2022.847576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Aldemir H, Prod’homme V, Dumaurier M-J, Retiere C, Poupon G, Cazareth J, et al. Cutting edge: lectin-like transcript 1 is a ligand for the CD161 receptor. J Immunol 2005, 175, 7791–5. doi: 10.4049/jimmunol.175.12.7791. [DOI] [PubMed] [Google Scholar]
- 33.Rosen DB, Cao W, Avery DT, Tangye SG, Liu Y-J, Houchins JP, et al. Functional consequences of interactions between human NKR-P1A and its ligand LLT1 expressed on activated dendritic cells and B cells. J Immunol 2008, 180, 6508–17. doi: 10.4049/jimmunol.180.10.6508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fox DA, Hussey RE, Fitzgerald KA, Acuto O, Poole C, Palley L, et al. Ta1, a novel 105 KD human T cell activation antigen defined by a monoclonal antibody. J Immunol 1984, 133, 1250–6. [PubMed] [Google Scholar]
- 35.Kitagawa N, Hamaguchi M, Majima S, Fukuda T, Kimura T, Hashimoto Y, et al. Dipeptidyl peptidase-4 inhibitors have adverse effects for the proliferation of human T cells. J Clin Biochem Nutr 2018, 63, 106–12. doi: 10.3164/jcbn.17-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Metzemaekers M, Van Damme J, Mortier A, Proost P.. Regulation of chemokine activity – a focus on the role of dipeptidyl peptidase IV/CD26. Front Immunol 2016, 7, 483. doi: 10.3389/fimmu.2016.00483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Singh SP, Zhang HH, Foley JF, Hedrick MN, Farber JM.. Human T cells that are able to produce IL-17 express the chemokine receptor CCR6. J Immunol 2008, 180, 214–21. doi: 10.4049/jimmunol.180.1.214. [DOI] [PubMed] [Google Scholar]
- 38.Zaunders J, Mee Ling Munier C, McGuire HM, et al. Mapping the extent of heterogeneity of human CCR5+ CD4+ T cells in peripheral blood and lymph nodes. AIDS 2020, 34, 833–48. doi: 10.3389/fnhum.2015.00181. [DOI] [PubMed] [Google Scholar]
- 39.Williams IR. CCR6 and CCL20: partners in intestinal immunity and lymphorganogenesis. Ann N Y Acad Sci 2006, 1072, 52–61. doi: 10.1196/annals.1326.036. [DOI] [PubMed] [Google Scholar]
- 40.Wang C, Kang SG, Lee J, Sun Z, Kim CH.. The roles of CCR6 in migration of Th17 cells and regulation of effector T-cell balance in the gut. Mucosal Immunol 2009, 2, 173–83. doi: 10.1038/mi.2008.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Nanjappa SG, Heninger E, Wüthrich M, Gasper DJ, Klein BS.. Tc17 cells mediate vaccine immunity against lethal fungal pneumonia in immune deficient hosts lacking CD4+ T cells. PLoS Pathog 2012, 8, e1002771. doi: 10.1371/journal.ppat.1002771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Oo YH, Banz V, Kavanagh D, Liaskou E, Withers DR, Humphreys E, et al. CXCR3-dependent recruitment and CCR6-mediated positioning of Th-17 cells in the inflamed liver. J Hepatol 2012, 57, 1044–51. doi: 10.1016/j.jhep.2012.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gonçalves CM, Henriques SN, Santos RF, Carmo AM.. CD6, a rheostat-type signalosome that tunes T cell activation. Front Immunol 2018, 9, 2994. doi: 10.3389/fimmu.2018.02994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zimmerman AW, Joosten B, Torensma R, Parnes JR, van Leeuwen FN, Figdor CG.. Long-term engagement of CD6 and ALCAM is essential for T-cell proliferation induced by dendritic cells. Blood 2006, 107, 3212–20. doi: 10.1182/blood-2005-09-3881. [DOI] [PubMed] [Google Scholar]
- 45.Bowen MA, Aruffo AA, Bajorath J.. Cell surface receptors and their ligands: in vitro analysis of CD6-CD166 interactions. Proteins 2000, 40, 420–8. [PubMed] [Google Scholar]
- 46.Smith NR, Davies PS, Levin TG, Gallagher AC, Keene DR, Sengupta SK, et al. Cell adhesion molecule CD166/ALCAM functions within the crypt to orchestrate murine intestinal stem cell homeostasis. Cell Mol Gastroenterol Hepatol 2017, 3, 389–409. doi: 10.1016/j.jcmgh.2016.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Enyindah-Asonye G, Li Y, Ruth JH, Spassov DS, Hebron KE, Zijlstra A, et al. CD318 is a ligand for CD6. Proc Natl Acad Sci U S A 2017, 114, E6912–21. doi: 10.1073/pnas.1704008114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Breuning J, Brown MH.. T cell costimulation by CD6 is dependent on bivalent binding of a GADS/SLP-76 complex. Mol Cell Biol 2017, 37, e00071–17. doi: 10.1128/MCB.00071-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Henriques SN, Oliveira L, Santos RF, Carmo AM.. CD6-mediated inhibition of T cell activation via modulation of Ras. Cell Commun Signal 2022, 20, 184. doi: 10.1186/s12964-022-00998-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bughani U, Saha A, Kuriakose A, Nair R, Sadashivarao RB, Venkataraman R, et al. T cell activation and differentiation is modulated by a CD6 domain 1 antibody Itolizumab. PLoS One 2017, 12, e0180088. doi: 10.1371/journal.pone.0180088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Simms PE, Ellis TM.. Utility of flow cytometric detection of CD69 expression as a rapid method for determining poly- and oligoclonal lymphocyte activation. Clin Diagn Lab Immunol 1996, 3, 301–4. doi: 10.1128/cdli.3.3.301-304.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hasegawa A, Iwamura C, Kitajima M, Hashimoto K, Otsuyama K-I, Ogino H, et al. Crucial role for CD69 in the pathogenesis of dextran sulphate sodium-induced colitis. PLoS One 2013, 8, e65494. doi: 10.1371/journal.pone.0065494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Radulovic K, Rossini V, Manta C, Holzmann K, Kestler HA, Niess JH.. The early activation marker CD69 regulates the expression of chemokines and CD4 T cell accumulation in intestine. PLoS One 2013, 8, e65413. doi: 10.1371/journal.pone.0065413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Sancho D, Gómez M, Viedma F, Esplugues E, Gordón-Alonso M, García-López MA, et al. CD69 downregulates autoimmune reactivity through active transforming growth factor-β production in collagen-induced arthritis. J Clin Invest 2003, 112, 872–82. doi: 10.1172/JCI19112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Murata K, Inami M, Hasegawa A, Kubo S, Kimura M, Yamashita M, et al. CD69-null mice protected from arthritis induced with anti-type II collagen antibodies. Int Immunol 2003, 15, 987–92. doi: 10.1093/intimm/dxg102. [DOI] [PubMed] [Google Scholar]
- 56.Cibrián D, Sánchez-Madrid F.. CD69: from activation marker to metabolic gatekeeper. Eur J Immunol 2017, 47, 946–53. doi: 10.1002/eji.201646837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Martín P, Gómez M, Lamana A, Cruz-Adalia A, Ramírez-Huesca M, Ursa MA, et al. CD69 association with Jak3/Stat5 proteins regulates Th17 cell differentiation. Mol Cell Biol 2010, 30, 4877–89. doi: 10.1128/MCB.00456-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.de la Fuente H, Cruz-Adalia A, Martinez del Hoyo G, Cibrián-Vera D, Bonay P, Pérez-Hernández D, et al. The leukocyte activation receptor CD69 controls T cell differentiation through its interaction with galectin-1. Mol Cell Biol 2014, 34, 2479–87. doi: 10.1128/MCB.00348-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yang X-P, Ghoreschi K, Steward-Tharp SM, Rodriguez-Canales J, Zhu J, Grainger JR, et al. Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5. Nat Immunol 2011, 12, 247–54. doi: 10.1038/ni.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Shiow LR, Rosen DB, Brdičková N, Xu Y, An J, Lanier Lewis L, et al. CD69 acts downstream of interferon-α/β to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 2006, 440, 540–4. doi: 10.1038/nature04606. [DOI] [PubMed] [Google Scholar]
- 61.Cibrian D, Saiz ML, De La Fuente H, Sánchez-Díaz R, Moreno-Gonzalo O, Jorge I, et al. CD69 controls the uptake of L-tryptophan through LAT1-CD98 and AhR-dependent secretion of IL-22 in psoriasis. Nat Immunol 2016, 17, 985–96. doi: 10.1038/ni.3504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mackay LK, Braun A, Macleod BL, Collins N, Tebartz C, Bedoui S, et al. Cutting edge: CD69 interference with sphingosine-1-phosphate receptor function regulates peripheral T cell retention. J Immunol 2015, 194, 2059–63. doi: 10.4049/jimmunol.1402256. [DOI] [PubMed] [Google Scholar]
- 63.Cheuk S, Schlums H, Gallais Sérézal I, Martini E, Chiang SC, Marquardt N, et al. CD49a expression defines tissue-resident CD8+ T cells poised for cytotoxic function in human skin. Immunity 2017, 46, 287–300. doi: 10.1016/j.immuni.2017.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 2007, 204, 1257–65. doi: 10.1084/jem.20062512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Timperi E, Barnaba V.. CD39 regulation and functions in T cells. Int J Mol Sci 2021, 22, 8068. doi: 10.3390/ijms22158068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ohta A, Sitkovsky M.. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 2001, 414, 916–20. doi: 10.1038/414916a. [DOI] [PubMed] [Google Scholar]
- 67.Wolberg G, Zimmerman TP, Hiemstra K, Winston M, Chu LC.. Adenosine inhibition of lymphocyte-mediated cytolysis: possible role of cyclic adenosine monophosphate. Science 1975, 187, 957–9. doi: 10.1126/science.167434. [DOI] [PubMed] [Google Scholar]
- 68.Globig AM, Mayer LS, Heeg M, Andrieux G, Ku M, Otto-Mora P, et al. Exhaustion of CD39-expressing CD8+ T cells in Crohn’s disease is linked to clinical outcome. Gastroenterology 2022, 163, 965–81.e31. doi: 10.1053/j.gastro.2022.06.045. [DOI] [PubMed] [Google Scholar]
- 69.Gallerano D, Ciminati S, Grimaldi A, Piconese S, Cammarata I, Focaccetti C, et al. Genetically driven CD39 expression shapes human tumor-infiltrating CD8+ T-cell functions. Int J Cancer 2020, 147, 2597–610. doi: 10.1002/ijc.33131. [DOI] [PubMed] [Google Scholar]
- 70.Canale FP, Ramello MC, Núñez N, Araujo Furlan CL, Bossio SN, Gorosito Serrán M, et al. CD39 expression defines cell exhaustion in tumor-infiltrating CD8+ T cells. Cancer Res 2018, 78, 115–28. doi: 10.1158/0008-5472.CAN-16-2684. [DOI] [PubMed] [Google Scholar]
- 71.Gupta PK, Godec J, Wolski D, Adland E, Yates K, Pauken KE, et al. CD39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog 2015, 11, e1005177. doi: 10.1371/journal.ppat.1005177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Simoni Y, Becht E, Fehlings M, Loh CY, Koo S-L, Teng KWW, et al. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 2018, 557, 575–9. doi: 10.1038/s41586-018-0130-2. [DOI] [PubMed] [Google Scholar]
- 73.Friedman DJ, Künzli BM, A-Rahim YI, Sevigny J, Berberat PO, Enjyoji K, et al. CD39 deletion exacerbates experimental murine colitis and human polymorphisms increase susceptibility to inflammatory bowel disease. Proc Natl Acad Sci U S A 2009, 106, 16788–93. doi: 10.1073/pnas.0902869106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Sheppard KA, Fitz LJ, Lee JM, Benander C, George JA, Wooters J, et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3ζ signalosome and downstream signaling to PKCθ. FEBS Lett 2004, 574, 37–41. doi: 10.1016/j.febslet.2004.07.083. [DOI] [PubMed] [Google Scholar]
- 75.Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017, 355, 1428–33. doi: 10.1126/science.aaf1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, et al. Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 2017, 355, 1423–7. doi: 10.1126/science.aaf0683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Patsoukis N, Brown J, Petkova V, Liu F, Li L, Boussiotis VA.. Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci Signal 2012, 5, ra46. doi: 10.1126/scisignal.2002796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL.. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol 2004, 173, 945–54. doi: 10.4049/jimmunol.173.2.945. [DOI] [PubMed] [Google Scholar]
- 79.Carter LL, Fouser LA, Jussif J, Fitz L, Deng B, Wood C R, et al. PD-1:PD-L inhibitory pathway affects both CD4+ and CD8+ T cells and is overcome by IL-2. Eur J Immunol 2002, 32, 634–43. doi:. [DOI] [PubMed] [Google Scholar]
- 80.Blackburn SD, Crawford A, Shin H, Polley A, Freeman GJ, Wherry EJ.. Tissue-specific differences in PD-1 and PD-L1 expression during chronic viral infection: implications for CD8 T-cell exhaustion. J Virol 2010, 84, 2078–89. doi: 10.1128/JVI.01579-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol 1996, 8, 765–72. doi: 10.1093/intimm/8.5.765. [DOI] [PubMed] [Google Scholar]
- 82.Vibhakar R, Juan G, Traganos F, Darzynkiewicz Z, Finger LR.. Activation-induced expression of human programmed death-1 gene in T- lymphocytes. Exp Cell Res 1997, 232, 25–8. doi: 10.1006/excr.1997.3493. [DOI] [PubMed] [Google Scholar]
- 83.Sauce D, Almeida JR, Larsen M, Haro L, Autran B, Freeman GJ, et al. PD-1 expression on human CD8 T cells depends on both state of differentiation and activation status. AIDS 2007, 21, 2005–13. doi: 10.1097/QAD.0b013e3282eee548. [DOI] [PubMed] [Google Scholar]
- 84.Gianchecchi E, Delfino DV, Fierabracci A.. Recent insights into the role of the PD-1/PD-L1 pathway in immunological tolerance and autoimmunity. Autoimmun Rev 2013, 12, 1091–100. doi: 10.1016/j.autrev.2013.05.003. [DOI] [PubMed] [Google Scholar]
- 85.Nishimura H, Honjo T.. PD-1: an inhibitory immunoreceptor involved in peripheral tolerance. Trends Immunol 2001, 22, 265–8. doi: 10.1016/s1471-4906(01)01888-9. [DOI] [PubMed] [Google Scholar]
- 86.Odorizzi PM, Pauken KE, Paley MA, Sharpe A, Wherry EJ.. Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J Exp Med 2015, 212, 1125–37. doi: 10.1084/jem.20142237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Blackburn SD, Shin H, Freeman GJ, Wherry EJ.. Selective expansion of a subset of exhausted CD8 T cells by αPD-L1 blockade. Proc Natl Acad Sci U S A 2008, 105, 15016–21. doi: 10.1073/pnas.0801497105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Bengsch B, Johnson AL, Kurachi M, Odorizzi PM, Pauken KE, Attanasio J, et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8+ T cell exhaustion. Immunity 2016, 45, 358–73. doi: 10.1016/j.immuni.2016.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Wherry EJ, Kurachi M.. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 2015, 15, 486–99. doi: 10.1038/nri3862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Schietinger A, Greenberg PD.. Tolerance and exhaustion: defining mechanisms of T cell dysfunction. Trends Immunol 2014, 35, 51–60. doi: 10.1016/j.it.2013.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sharma P, Allison JP.. The future of immune checkpoint therapy. Science 2015, 348, 56–61. doi: 10.1126/science.aaa8172. [DOI] [PubMed] [Google Scholar]
- 92.Page DB, Postow MA, Callahan MK, Allison JP, Wolchok JD.. Immune modulation in cancer with antibodies. Annu Rev Med 2014, 65, 185–202. doi: 10.1146/annurev-med-092012-112807. [DOI] [PubMed] [Google Scholar]
- 93.Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006, 439, 682–7. doi: 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
- 94.Phadungsaksawasdi P, Fujiyama T, Kurihara K, Ito T, Honda T, Tokura Y.. PD-1 expression defines epidermal CD8+CD103+ T cells preferentially producing IL-17A and using skewed TCR repertoire in psoriasis. J Invest Dermatol 2021, 141, 2426–35.e5. doi: 10.1016/j.jid.2021.03.011. [DOI] [PubMed] [Google Scholar]
- 95.Liu J, Chang HW, Huang ZM, Nakamura M, Sekhon S, Ahn R, et al. Single-cell RNA sequencing of psoriatic skin identifies pathogenic Tc17 cell subsets and reveals distinctions between CD8+ T cells in autoimmunity and cancer. J Allergy Clin Immunol 2021, 147, 2370–80. doi: 10.1016/j.jaci.2020.11.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.van Lier RA, Borst J, Vroom TM, Klein H, Van Mourik P, Zeijlemaker WP, et al. Tissue distribution and biochemical and functional properties of Tp55 (CD27), a novel T cell differentiation antigen. J Immunol 1987, 139, 1589–96. [PubMed] [Google Scholar]
- 97.Croft M. The role of TNF superfamily members in T-cell function and diseases. Nat Rev Immunol 2009, 9, 271–85. doi: 10.1038/nri2526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Grant EJ, Nüssing S, Sant S, Clemens EB, Kedzierska K.. The role of CD27 in anti-viral T-cell immunity. Curr Opin Virol 2017, 22, 77–88. doi: 10.1016/j.coviro.2016.12.001. [DOI] [PubMed] [Google Scholar]
- 99.Rowley TF, Al-Shamkhani A.. Stimulation by soluble CD70 promotes strong primary and secondary CD8+ cytotoxic T cell responses in vivo. J Immunol 2004, 172, 6039–46. doi: 10.4049/jimmunol.172.10.6039. [DOI] [PubMed] [Google Scholar]
- 100.Willoughby JE, Kerr JP, Rogel A, Taraban VY, Buchan SL, Johnson PWM, et al. Differential impact of CD27 and 4-1BB costimulation on effector and memory CD8 T cell generation following peptide immunization. J Immunol 2014, 193, 244–51. doi: 10.4049/jimmunol.1301217. [DOI] [PubMed] [Google Scholar]
- 101.Hendriks J, Xiao Y, Borst J.. CD27 promotes survival of activated T cells and complements CD28 in generation and establishment of the effector T cell pool. J Exp Med 2003, 198, 1369–80. doi: 10.1084/jem.20030916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Taraban VY, Rowley TF, Kerr JP, Willoughby JE, Johnson PMW, Al-Shamkhani A, et al. CD27 costimulation contributes substantially to the expansion of functional memory CD8+ T cells after peptide immunization. Eur J Immunol 2013, 43, 3314–23. doi: 10.1002/eji.201343579. [DOI] [PubMed] [Google Scholar]
- 103.Akiba H, Nakano H, Nishinaka S, Shindo M, Kobata T, Atsuta M, et al. CD27, a member of the tumor necrosis factor receptor superfamily, activates NF-κB and stress-activated protein kinase/c-Jun N-terminal kinase via TRAF2, TRAF5, and NF-κB-inducing kinase. J Biol Chem 1998, 273, 13353–8. doi: 10.1074/jbc.273.21.13353. [DOI] [PubMed] [Google Scholar]
- 104.van Oosterwijk MF, Juwana H, Arens R, Tesselaar K, van Oers MHJ, Eldering E, et al. CD27-CD70 interactions sensitise naive CD4+ T cells for IL-12-induced Th1 cell development. Int Immunol 2007, 19, 713–8. doi: 10.1093/intimm/dxm033. [DOI] [PubMed] [Google Scholar]
- 105.Wasiuk A, Testa J, Weidlick J, Sisson C, Vitale L, Widger J, et al. CD27-mediated regulatory T cell depletion and effector T cell costimulation both contribute to antitumor efficacy. J Immunol 2017, 199, 4110–23. doi: 10.4049/jimmunol.1700606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Buchan SL, Fallatah M, Thirdborough SM, Taraban VY, Rogel A, Thomas LJ, et al. PD-1 blockade and CD27 stimulation activate distinct transcriptional programs that synergize for CD8+ T-cell–driven antitumor immunity. Clin Cancer Res 2018, 24, 2383–94. doi: 10.1158/1078-0432.CCR-17-3057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Vitale LA, He L-Z, Thomas LJ, Wasiuk A, O'Neill T, Widger J, et al. Development of CDX-527: a bispecific antibody combining PD-1 blockade and CD27 costimulation for cancer immunotherapy. Cancer Immunol Immunother 2020, 69, 2125–37. doi: 10.1007/s00262-020-02610-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Koay H-F, Su S, Amann-Zalcenstein D, Daley SR, Comerford I, Miosge L, et al. A divergent transcriptional landscape underpins the development and functional branching of MAIT cells. Sci Immunol 2019, 4, eaay6039. doi: 10.1126/sciimmunol.aay6039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Chien Y, Meyer C, Bonneville M.. γδ T cells: first line of defense and beyond. Annu Rev Immunol 2014, 32, 121–55. doi: 10.1146/annurev-immunol-032713-120216. [DOI] [PubMed] [Google Scholar]
- 110.Chen YS, Chen IB, Pham G, Shao T-Y, Bangar H, Way SS, et al. IL-17-producing γδ T cells protect against Clostridium difficile infection. J Clin Invest 2020, 130, 2377–90. doi: 10.1172/JCI127242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tom MR, Li J, Ueno A, Fort Gasia M, Chan R, Hung DY, et al. Novel CD8+ T-cell subsets demonstrating plasticity in patients with inflammatory bowel disease. Inflamm Bowel Dis 2016, 22, 1596–608. doi: 10.1097/MIB.0000000000000848. [DOI] [PubMed] [Google Scholar]
- 112.Chien Y, Zeng X, Prinz I.. The natural and the inducible: interleukin (IL)-17-producing γδ T cells. Trends Immunol 2013, 34, 151–4. doi: 10.1016/j.it.2012.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Chung HK, McDonald B, Kaech SM.. The architectural design of CD8+ T cell responses in acute and chronic infection: parallel structures with divergent fates. J Exp Med 2021, 218, e20201730. doi: 10.1084/jem.20201730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Strober W, Fuss IJ.. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology 2011, 140, 1756–67. doi: 10.1053/j.gastro.2011.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Volpe E, Servant N, Zollinger R, Bogiatzi SI, Hupé P, Barillot E, et al. A critical function for transforming growth factor-β, interleukin 23 and proinflammatory cytokines in driving and modulating human TH-17 responses. Nat Immunol 2008, 9, 650–7. doi: 10.1038/ni.1613. [DOI] [PubMed] [Google Scholar]
- 116.Manel N, Unutmaz D, Littman DR.. The differentiation of human TH-17 cells requires transforming growth factor-β and induction of the nuclear receptor RORγt. Nat Immunol 2008, 9, 641–9. doi: 10.1038/ni.1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.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: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
- 118.Bettelli E, Oukka M, Kuchroo VK.. T(H)-17 cells in the circle of immunity and autoimmunity. Nat Immunol 2007, 8, 345–50. doi: 10.1038/ni0407-345. [DOI] [PubMed] [Google Scholar]
- 119.Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F.. Interleukins 1β and 6 but not transforming growth factor-β are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol 2007, 8, 942–9. doi: 10.1038/ni1496. [DOI] [PubMed] [Google Scholar]
- 120.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: 10.1038/ni1497. [DOI] [PubMed] [Google Scholar]
- 121.Chen Z, Tato CM, Muul L, Laurence A, O'Shea JJ.. Distinct regulation of interleukin-17 in human T helper lymphocytes. Arthritis Rheum 2007, 56, 2936–46. doi: 10.1002/art.22866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Korn T, Bettelli E, Gao W, Awasthi A, Jäger A, Strom TB, et al. IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells. Nature 2007, 448, 484–7. doi: 10.1038/nature05970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.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: 10.1016/j.immuni.2006.01.001. [DOI] [PubMed] [Google Scholar]
- 124.Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T, et al. IL-6 programs TH-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol 2007, 8, 967–74. doi: 10.1038/ni1488. [DOI] [PubMed] [Google Scholar]
- 125.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: 10.1038/nature04754. [DOI] [PubMed] [Google Scholar]
- 126.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: 10.1038/nature05969. [DOI] [PubMed] [Google Scholar]
- 127.Gras R, García MI, Gómez R, de la Mata FJ, Muñoz-Fernández MA, López-Fernández LA.. Carbosilane dendrimer 2G-NN16 represses Tc17 differentiation in primary T CD8+ lymphocytes. Mol Pharm 2012, 9, 102–10. doi: 10.1021/mp200305u. [DOI] [PubMed] [Google Scholar]
- 128.Renavikar PS, Sinha S, Brate AA, Borcherding N, Crawford MP, Steward-Tharp SM, et al. IL-12-induced immune suppressive deficit during CD8+ T-cell differentiation. Front Immunol 2020, 11, 568630. doi: 10.3389/fimmu.2020.568630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.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 respones. J Immunol 2006, 177, 6852–8. doi: 10.4049/jimmunol.177.10.6852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Liu S-J, Tsai J-P, Shen C-R, Sher Y-P, Hsieh C-L, Yeh Y-C, et al. Induction of a distinct CD8 Tnc17 subset by transforming growth factor-β and interleukin-6. J Leukoc Biol 2007, 82, 354–60. doi: 10.1189/jlb.0207111. [DOI] [PubMed] [Google Scholar]
- 131.Hamada H, de la Luz Garcia-Hernandez M, Reome JB, Misra SK, Strutt TM, McKinstry KK, et al. Tc17, a unique subset of CD8 T cells that can protect against lethal influenza challenge. J Immunol 2009, 182, 3469–81. doi: 10.4049/jimmunol.0801814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Yen H-R, Harris TJ, Wada S, Grosso JF, Getnet D, Goldberg MV, et al. Tc17 CD8 T cells: functional plasticity and subset diversity. J Immunol 2009, 183, 7161–8. doi: 10.4049/jimmunol.0900368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Ciric B, El-behi M, Cabrera R, Zhang G-X, Rostami A.. IL-23 drives pathogenic IL-17-producing CD8+ T cells. J Immunol 2009, 182, 5296–305. doi: 10.4049/jimmunol.0900036. [DOI] [PubMed] [Google Scholar]
- 134.Curtis MM, Way SS, Wilson CB.. IL-23 promotes the production of IL-17 by antigen-specific CD8 T cells in the absence of IL-12 and type-I interferons. J Immunol 2009, 183, 381–7. doi: 10.4049/jimmunol.0900939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Huber M, Heink S, Grothe H, Guralnik A, Reinhard K, Elflein K, et al. A Th17-like developmental process leads to CD8+ Tc17 cells with reduced cytotoxic activity. Eur J Immunol 2009, 39, 1716–25. doi: 10.1002/eji.200939412. [DOI] [PubMed] [Google Scholar]
- 136.Yeh N, Glosson NL, Wang N, Guindon L, McKinley C, Hamada H, et al. Tc17 cells are capable of mediating immunity to vaccinia virus by acquisition of a cytotoxic phenotype. J Immunol 2010, 185, 2089–98. doi: 10.4049/jimmunol.1000818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.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: 10.1172/JCI63681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Flores-Santibáñez F, Fernández D, Meza D, Tejón G, Vargas L, Varela-Nallar L, et al. CD73-mediated adenosine production promotes stem cell-like properties in mouse Tc17 cells. Immunology 2015, 146, 582–94. doi: 10.1111/imm.12529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Arra A, Lingel H, Kuropka B, Pick J, Schnoeder T, Fischer T, et al. The differentiation and plasticity of Tc17 cells are regulated by CTLA-4-mediated effects on STATs. Oncoimmunology 2017, 6, e1273300. doi: 10.1080/2162402X.2016.1273300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Gartlan KH, Markey KA, Varelias A, Bunting MD, Koyama M, Kuns RD, et al. Tc17 cells are a proinflammatory, plastic lineage of pathogenic CD8+ T cells that induce GVHD without antileukemic effects. Blood 2015, 126, 1609–20. doi: 10.1182/blood-2015-01-622662. [DOI] [PubMed] [Google Scholar]
- 141.Dwivedi VP, Tousif S, Bhattacharya D, Prasad DVR, Van Kaer L, Das J, et al. Transforming growth factor-β protein inversely regulates in vivo differentiation of interleukin-17 (IL-17)-producing CD4+ and CD8+ T cells. J Biol Chem 2012, 287, 2943–7. doi: 10.1074/jbc.C111.327627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Chen HW, Tsai JP, Yao TY, Hsieh C-L, Chen I-H, Liu S-J.. TGF-β and IL-21 cooperatively stimulate activated CD8+ T cells to differentiate into Tc17 cells. Immunol Lett 2016, 174, 23–7. doi: 10.1016/j.imlet.2016.04.006. [DOI] [PubMed] [Google Scholar]
- 143.Srenathan U, Steel K, Taams LS.. IL-17+ CD8+ T cells: differentiation, phenotype and role in inflammatory disease. Immunol Lett 2016, 178, 20–6. doi: 10.1016/j.imlet.2016.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Happel KI, Zheng M, Young E, Quinton LJ, Lockhart E, Ramsay AJ, et al. Cutting edge: roles of toll-like receptor 4 and IL-23 in IL-17 expression in response to Klebsiella pneumoniae infection. J Immunol 2003, 170, 4432–6. doi: 10.4049/jimmunol.170.9.4432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Vanden Eijnden S, Goriely S, De Wit D, Willems F, Goldman M.. IL-23 up-regulates IL-10 and induces IL-17 synthesis by polyclonally activated naive T cells in human. Eur J Immunol 2005, 35, 469–75. doi: 10.1002/eji.200425677. [DOI] [PubMed] [Google Scholar]
- 146.Lückel C, Picard F, Raifer H, Campos Carrascosa L, Guralnik A, Zhang Y, et al. IL-17+ CD8+ T cell suppression by dimethyl fumarate associates with clinical response in multiple sclerosis. Nat Commun 2019, 10, 5722. doi: 10.1038/s41467-019-13731-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Xin A, Masson F, Liao Y, Preston S, Guan T, Gloury R, et al. A molecular threshold for effector CD8+ T cell differentiation controlled by transcription factors Blimp-1 and T-bet. Nat Immunol 2016, 17, 422–32. doi: 10.1038/ni.3410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Ysebrant de Lendonck L, Tonon S, Nguyen M, Vandevenne P, Welsby I, Martinet V, et al. Interferon regulatory factor 3 controls interleukin-17 expression in CD8 T lymphocytes. Proc Natl Acad Sci USA 2013, 110, E3189–97. doi: 10.1073/pnas.1219221110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Rangachari M, Mauermann N, Marty RR, Dirnhofer S, Kurrer MO, Komnenovic V, et al. T-bet negatively regulates autoimmune myocarditis by suppressing local production of interleukin 17. J Exp Med 2006, 203, 2009–19. doi: 10.1084/jem.20052222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Burrell BE, Csencsits K, Lu G, Grabauskiene S, Bishop DK.. CD8+ Th17 mediate costimulation blockade-resistant allograft rejection in T-bet-deficient mice. J Immunol 2008, 181, 3906–14. doi: 10.4049/jimmunol.181.6.3906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Intlekofer AM, Banerjee A, Takemoto N, Gordon SM, Dejong CS, Shin H, et al. Anomalous type 17 response to viral infection by CD8+ T cells lacking T-bet and eomesodermin. Science 2008, 321, 408–11. doi: 10.1126/science.1159806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Veldhoen M, Hirota K, Westendorf AM, Buer J, Dumoutier L, Renauld J-C, et al. The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 2008, 453, 106–9. doi: 10.1038/nature06881. [DOI] [PubMed] [Google Scholar]
- 153.Hayes MD, Ovcinnikovs V, Smith AG, Kimber I, Dearman RJ.. The aryl hydrocarbon receptor: differential contribution to T helper 17 and T cytotoxic 17 cell development. PLoS One 2014, 9, e106955. doi: 10.1371/journal.pone.0106955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Gutiérrez-Vázquez C, Quintana FJ.. Regulation of the immune response by the aryl hydrocarbon receptor. Immunity 2018, 48, 19–33. doi: 10.1016/j.immuni.2017.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kimura A, Naka T, Nohara K, Fujii-Kuriyama Y, Kishimoto T.. Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells. Proc Natl Acad Sci USA 2008, 105, 9721–6. doi: 10.1073/pnas.0804231105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Quintana FJ, Jin H, Burns EJ, Nadeau M, Yeste A, Kumar D, et al. Aiolos promotes TH17 differentiation by directly silencing Il2 expression. Nat Immunol 2012, 13, 770–7. doi: 10.1038/ni.2363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.de Lima KA, Donate PB, Talbot J, Davoli-Ferreira M, Peres RS, Cunha TM, et al. TGFβ1 signaling sustains aryl hydrocarbon receptor (AHR) expression and restrains the pathogenic potential of TH17 cells by an AHR-independent mechanism. Cell Death Dis 2018, 9, 1130. doi: 10.1038/s41419-018-1107-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hinrichs CS, Kaiser A, Paulos CM, Cassard L, Sanchez-Perez L, Heemskerk B, et al. Type 17 CD8+ T cells display enhanced antitumor immunity. Blood 2009, 114, 596–9. doi: 10.1182/blood-2009-02-203935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Harrison OJ, Linehan JL, Shih H-Y, Bouladoux N, Han S-J, Smelkinson M, et al. Commensal-specific T cell plasticity promotes rapid tissue adaptation to injury. Science 2019, 363, eaat6280. doi: 10.1126/science.aat6280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Nanjappa SG, McDermott AJ, Fites JS, Galles K, Wüthrich M, Deepe GS, et al. Antifungal Tc17 cells are durable and stable, persisting as long-lasting vaccine memory without plasticity towards IFNγ cells. PLoS Pathog 2017, 13, e1006356. doi: 10.1371/journal.ppat.1006356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Nanjappa SG, Mudalagiriyappa S, Fites JS, Suresh M, Klein BS.. CBLB Constrains inactivated vaccine-induced CD8+ T cell responses and immunity against lethal fungal pneumonia. J Immunol 2018, 201, 1717–26. doi: 10.4049/jimmunol.1701241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Nanjappa SG, Hernández-Santos N, Galles K, Wüthrich M, Suresh M, Klein BS.. Intrinsic MyD88-Akt1-mTOR signaling coordinates disparate Tc17 and Tc1 responses during vaccine immunity against fungal pneumonia. PLoS Pathog 2015, 11, e1005161. doi: 10.1371/journal.ppat.1005161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Yu Y, Cho H-I, Wang D, Kaosaard K, Anasetti C, Celis E, et al. Adoptive transfer of Tc1 or Tc17 cells elicits antitumor immunity against established melanoma through distinct mechanisms. J Immunol 2013, 190, 1873–81. doi: 10.4049/jimmunol.1201989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.de la Luz Garcia-Hernandez M, Hamada H, Reome JB, Misra SK, Tighe MP, Dutton RW.. Adoptive transfer of tumor-specific Tc17 effector T cells controls the growth of B16 melanoma in mice. J Immunol 2010, 184, 4215–27. doi: 10.4049/jimmunol.0902995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.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: 10.1016/j.immuni.2013.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.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: 10.1126/science.1200439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Lévy R, Okada S, Béziat V, Moriya K, Liu C, Chai LYA, 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, E8277–85. doi: 10.1073/pnas.1618300114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Chandesris M-O, 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 (Baltim) 2012, 91, e1–e19. doi: 10.1097/MD.0b013e31825f95b9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Davidson L, van den Reek JMPA, Bruno M, van Hunsel F, Herings Ron MC, Matzaraki V, et al. Risk of candidiasis associated with interleukin-17 inhibitors: a real-world observational study of multiple independent sources. Lancet Reg Health Eur 2021, 13, 100266. doi: 10.1016/j.lanepe.2021.100266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.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: 10.1086/422329. [DOI] [PubMed] [Google Scholar]
- 171.Whibley N, Tritto E, Traggiai E, Kolbinger F, Moulin P, Brees D, et al. Antibody blockade of IL-17 family cytokines in immunity to acute murine oral mucosal candidiasis. J Leukoc Biol 2016, 99, 1153–64. doi: 10.1189/jlb.4A0915-428R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Bär E, Whitney PG, Moor K, Reis e Sousa C, LeibundGut-Landmann S.. IL-17 regulates systemic fungal immunity by controlling the functional competence of NK cells. Immunity 2014, 40, 117–27. doi: 10.1016/j.immuni.2013.12.002. [DOI] [PubMed] [Google Scholar]
- 173.Milner JD, Holland SM.. The cup runneth over: lessons from the ever-expanding pool of primary immunodeficiency diseases. Nat Rev Immunol 2013, 13, 635–48. doi: 10.1038/nri3493. [DOI] [PubMed] [Google Scholar]
- 174.Hernández-Santos N, Huppler AR, Peterson AC, Khader SA, McKenna KC, Gaffen SL.. Th17 cells confer long-term adaptive immunity to oral mucosal Candida albicans infections. Mucosal Immunol 2013, 6, 900–10. doi: 10.1038/mi.2012.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Kohyama S, Ohno S, Isoda A, Moriya O, Belladonna ML, Hayashi H, et al. IL-23 enhances host defense against vaccinia virus infection via a mechanism partly involving IL-17. J Immunol 2007, 179, 3917–25. doi: 10.4049/jimmunol.179.6.3917. [DOI] [PubMed] [Google Scholar]
- 176.Patera AC, Pesnicak L, Bertin J, Cohen JI.. Interleukin 17 modulates the immune response to vaccinia virus infection. Virology 2002, 299, 56–63. doi: 10.1006/viro.2002.1400. [DOI] [PubMed] [Google Scholar]
- 177.Cachem FCOF, Dias AS, Monteiro C, Castro JR, Fernandes G, Delphim L, et al. The proportion of different interleukin-17-producing T-cell subsets is associated with liver fibrosis in chronic hepatitis C. Immunology 2017, 151, 167–76. doi: 10.1111/imm.12720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Foster RG, Golden-Mason L, Rutebemberwa A, Rosen HR.. Interleukin (IL)-17/IL-22-producing T cells enriched within the liver of patients with chronic hepatitis C viral (HCV) infection. Dig Dis Sci 2012, 57, 381–9. doi: 10.1007/s10620-011-1997-z. [DOI] [PubMed] [Google Scholar]
- 179.Sun HQ, Zhang JY, Zhang H, Zou ZS, Wang FS, Jia JH.. Increased Th17 cells contribute to disease progression in patients with HBV-associated liver cirrhosis. J Viral Hepat 2012, 19, 396–403. doi: 10.1111/j.1365-2893.2011.01561.x. [DOI] [PubMed] [Google Scholar]
- 180.Rowan AG, Fletcher JM, Ryan EJ, Moran B, Hegarty JE, O'Farrelly C, et al. Hepatitis C virus-specific Th17 cells are suppressed by virus-induced TGF-β. J Immunol 2008, 181, 4485–94. doi: 10.4049/jimmunol.181.7.4485. [DOI] [PubMed] [Google Scholar]
- 181.Paquissi FC. Immunity and fibrogenesis: the role of Th17/IL-17 axis in HBV and HCV-induced chronic hepatitis and progression to cirrhosis. Front Immunol 2017, 8, 1195. doi: 10.3389/fimmu.2017.01195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Zhao J, Zhang Z, Luan Y, Zou Z, Sun Y, Li Y, et al. Pathological functions of interleukin-22 in chronic liver inflammation and fibrosis with hepatitis B virus infection by promoting T helper 17 cell recruitment. Hepatology 2014, 59, 1331–42. doi: 10.1002/hep.26916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Gaardbo JC, Hartling HJ, Thorsteinsson K, Ullum H, Nielsen SD.. CD3+CD8+CD161high Tc17 cells are depleted in HIV-infection. AIDS 2013, 27, 659–62. doi: 10.1097/QAD.0b013e32835b8cb3. [DOI] [PubMed] [Google Scholar]
- 184.Perdomo-Celis F, Feria MG, Taborda NA, Rugeles MT.. A low frequency of IL-17-producing CD8+ T-cells is associated with persistent immune activation in people living with HIV despite HAART-induced viral suppression. Front Immunol 2018, 9, 2502. doi: 10.3389/fimmu.2018.02502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Luzza F, Parrello T, Monteleone G, Sebkova L, Romano M, Zarrilli R, et al. Up-regulation of IL-17 is associated with bioactive IL-8 expression in Helicobacter pylori-infected human gastric mucosa. J Immunol 2000, 165, 5332–7. doi: 10.4049/jimmunol.165.9.5332. [DOI] [PubMed] [Google Scholar]
- 186.Lu YJ, Gross J, Bogaert D, Finn A, Bagrade L, Zhang Q, et al. Interleukin-17A mediates acquired immunity to pneumococcal colonization. PLoS Pathog 2008, 4, e1000159. doi: 10.1371/journal.ppat.1000159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Borkner L, Curham LM, Wilk MM, Moran B, Mills KHG.. IL-17 mediates protective immunity against nasal infection with Bordetella pertussis by mobilizing neutrophils, especially Siglec-F+ neutrophils. Mucosal Immunol 2021, 14, 1183–202. doi: 10.1038/s41385-021-00407-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Aujla SJ, Chan YR, Zheng M, Fei M, Askew DJ, Pociask DA, et al. IL-22 mediates mucosal host defense against gram-negative bacterial pneumonia. Nat Med 2008, 14, 275–81. doi: 10.1038/nm1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Zhang Y, Wang H, Ren J, Tang X, Jing Y, Xing D, et al. IL-17A synergizes with IFN-γ to upregulate iNOS and NO production and inhibit chlamydial growth. PLoS One 2012, 7, e39214. doi: 10.1371/journal.pone.0039214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Saito H, Yamada Y, Takaya S, Osaki T, Ikeguchi M.. Clinical relevance of the number of interleukin-17-producing CD 8+ T cells in patients with gastric cancer. Surg Today 2015, 45, 1429–35. doi: 10.1007/s00595-015-1165-8. [DOI] [PubMed] [Google Scholar]
- 191.Zhong F, Cui D, Tao H, Du H, Xing C.. IL-17A-producing T cells and associated cytokines are involved in the progression of gastric cancer. Oncol Rep 2015, 34, 2365–74. doi: 10.3892/or.2015.4246. [DOI] [PubMed] [Google Scholar]
- 192.Zhuang Y, Peng L-S, Zhao Y-L, Shi Y, Mao X-H, Chen W, et al. CD8+ T cells that produce interleukin-17 regulate myeloid-derived suppressor cells and are associated with survival time of patients with gastric cancer. Gastroenterology 2012, 143, 951–62.e8. doi: 10.1053/j.gastro.2012.06.010. [DOI] [PubMed] [Google Scholar]
- 193.Sun K, Xu R, Ma F, Yang N, Li Y, Sun X, et al. scRNA-seq of gastric tumor shows complex intercellular interaction with an alternative T cell exhaustion trajectory. Nat Commun 2022, 13, 4943. doi: 10.1038/s41467-022-32627-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Kuang D-M, Peng C, Zhao Q, Wu Y, Zhu L-Y, Wang J, et al. Tumor-activated monocytes promote expansion of IL-17-producing CD8+ T cells in hepatocellular carcinoma patients. J Immunol 2010, 185, 1544–9. doi: 10.4049/jimmunol.0904094. [DOI] [PubMed] [Google Scholar]
- 195.Lee YH, Chuah S, Nguyen PHD, Lim CJ, Lai HLH, Wasser M, et al. IFNγ-IL-17+ CD8 T cells contribute to immunosuppression and tumor progression in human hepatocellular carcinoma. Cancer Lett 2023, 552, 215977. doi: 10.1016/j.canlet.2022.215977. [DOI] [PubMed] [Google Scholar]
- 196.Chen K-J, Lin S-Z, Zhou L, Xie H-Y, Zhou W-H, Taki-Eldin A, et al. Selective recruitment of regulatory T cell through CCR6-CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. PLoS One 2011, 6, e24671. doi: 10.1371/journal.pone.0024671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Gao Q, Zhao YJ, Wang XY, Qiu S-J, Shi Y-H, Sun J, et al. CXCR6 upregulation contributes to a proinflammatory tumor microenvironment that drives metastasis and poor patient outcomes in hepatocellular carcinoma. Cancer Res 2012, 72, 3546–56. doi: 10.1158/0008-5472.CAN-11-4032. [DOI] [PubMed] [Google Scholar]
- 198.Cheng X, Wu H, Jin ZJ, Ma D, Yuen S, Jing X-Q, et al. Up-regulation of chemokine receptor CCR4 is associated with human hepatocellular carcinoma malignant behavior. Sci Rep 2017, 7, 12362. doi: 10.1038/s41598-017-10267-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Zhang JP, Yan J, Xu J, Pang X-H, Chen M-S, Li L, et al. Increased intratumoral IL-17-producing cells correlate with poor survival in hepatocellular carcinoma patients. J Hepatol 2009, 50, 980–9. doi: 10.1016/j.jhep.2008.12.033. [DOI] [PubMed] [Google Scholar]
- 200.Chellappa S, Hugenschmidt H, Hagness M, Subramani S, Melum E, Line PD, et al. CD8+ T cells that coexpress RORγt and T-bet are functionally impaired and expand in patients with distal bile duct cancer. J Immunol 2017, 198, 1729–39. doi: 10.4049/jimmunol.1600061. [DOI] [PubMed] [Google Scholar]
- 201.Linehan JL, Harrison OJ, Han SJ, Byrd AL, Vujkovic-Cvijin I, Villarino AV, et al. Non-classical immunity controls microbiota impact on skin immunity and tissue repair. Cell 2018, 172, 784–96.e18. doi: 10.1016/j.cell.2017.12.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Naik S, Bouladoux N, Linehan JL, Han S-J, Harrison OJ, Wilhelm C, et al. Commensal–dendritic-cell interaction specifies a unique protective skin immune signature. Nature 2015, 520, 104–8. doi: 10.1038/nature14052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Nestle FO, Kaplan DH, Barker J.. Psoriasis. N Engl J Med 2009, 361, 496–509. doi: 10.1056/nejmra0804595. [DOI] [PubMed] [Google Scholar]
- 204.Griffiths CE, Barker JN.. Pathogenesis and clinical features of psoriasis. Lancet 2007, 370, 263–71. doi: 10.1016/S0140-6736(07)61128-3. [DOI] [PubMed] [Google Scholar]
- 205.Teunissen MBM, Yeremenko NG, Baeten DLP, Chielie S, Spuls PI, de Rie MA, et al. The IL-17A-producing CD8+ T-cell population in psoriatic lesional skin comprises mucosa-associated invariant T cells and conventional T cells. J Invest Dermatol 2014, 134, 2898–907. doi: 10.1038/jid.2014.261. [DOI] [PubMed] [Google Scholar]
- 206.Ortega C, Fernández-A S, Carrillo JM, Romero P, Molina IJ, Moreno JC, et al. IL-17-producing CD8+ T lymphocytes from psoriasis skin plaques are cytotoxic effector cells that secrete Th17-related cytokines. J Leukoc Biol 2009, 86, 435–43. doi: 10.1189/JLB.0109046. [DOI] [PubMed] [Google Scholar]
- 207.Res PCM, Piskin G, de Boer OJ, van der Loos CM, Teeling P, Bos JD, et al. Overrepresentation of IL-17A and IL-22 producing CD8 T cells in lesional skin suggests their involvement in the pathogenesis of psoriasis. PLoS One 2010, 5, e14108. doi: 10.1371/journal.pone.0014108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Kryczek I, Bruce AT, Gudjonsson JE, Johnston A, Aphale A, Vatan L, et al. Induction of IL-17+ T cell trafficking and development by IFN-γ: mechanism and pathological relevance in psoriasis. J Immunol 2008, 181, 4733–41. doi: 10.4049/jimmunol.181.7.4733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Hijnen D, Knol EF, Gent YY, Giovannone B, Beijn SJP, Kupper TS, et al. CD8+ T cells in the lesional skin of atopic dermatitis and psoriasis patients are an important source of IFN-γ, IL-13, IL-17, and IL-22. J Invest Dermatol 2013, 133, 973–9. doi: 10.1038/jid.2012.456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Hueber W, Patel DD, Dryja T, Wright AM, Koroleva I, Bruin G, et al.; Psoriasis Study Group. Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med 2010, 2, 52ra72. doi: 10.1126/scitranslmed.3001107. [DOI] [PubMed] [Google Scholar]
- 211.Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y, et al.; PHOENIX 1 study investigators. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet 2008, 371, 1665–74. doi: 10.1016/S0140-6736(08)60725-4. [DOI] [PubMed] [Google Scholar]
- 212.Martin DA, Towne JE, Kricorian G, Klekotka P, Gudjonsson JE, Krueger JG, et al. The emerging role of IL-17 in the pathogenesis of psoriasis: preclinical and clinical findings. J Invest Dermatol 2013, 133, 17–26. doi: 10.1038/jid.2012.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Harden JL, Krueger JG, Bowcock AM.. The immunogenetics of Psoriasis: a comprehensive review. J Autoimmun 2015, 64, 66–73. doi: 10.1016/j.jaut.2015.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Lande R, Botti E, Jandus C, Dojcinovic D, Fanelli G, Conrad C, et al. The antimicrobial peptide LL37 is a T-cell autoantigen in psoriasis. Nat Commun 2014, 5, 5621. doi: 10.1038/ncomms6621. [DOI] [PubMed] [Google Scholar]
- 215.Arakawa A, Siewert K, Stöhr J, Besgen P, Kim S-M, Rühl G, et al. Melanocyte antigen triggers autoimmunity in human psoriasis. J Exp Med 2015, 212, 2203–12. doi: 10.1084/jem.20151093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Valdimarsson H, Thorleifsdottir RH, Sigurdardottir SL, Gudjonsson JE, Johnston A.. Psoriasis – as an autoimmune disease caused by molecular mimicry. Trends Immunol 2009, 30, 494–501. doi: 10.1016/j.it.2009.07.008. [DOI] [PubMed] [Google Scholar]
- 217.Di Meglio P, Villanova F, Navarini AA, Mylonas A, Tosi I, Nestle FO, et al. Targeting CD8(+) T cells prevents psoriasis development. J Allergy Clin Immunol 2016, 138, 274–6.e6. doi: 10.1016/j.jaci.2015.10.046. [DOI] [PubMed] [Google Scholar]
- 218.Alinaghi F, Calov M, Kristensen LE, Gladman DD, Coates LC, Jullien D, et al. Prevalence of psoriatic arthritis in patients with psoriasis: a systematic review and meta-analysis of observational and clinical studies. J Am Acad Dermatol 2019, 80, 251–265.e19. doi: 10.1016/j.jaad.2018.06.027. [DOI] [PubMed] [Google Scholar]
- 219.Steel KJA, Srenathan U, Ridley M, Durham LE, Wu S-Y, Ryan SE, et al. Polyfunctional, proinflammatory, tissue-resident memory phenotype and function of synovial interleukin-17A+CD8+ T cells in psoriatic arthritis. Arthritis Rheumatol 2020, 72, 435–47. doi: 10.1002/art.41156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Menon B, Gullick NJ, Walter GJ, Rajasekhar M, Garrood T, Evans HG, et al. Interleukin-17+CD8+ T cells are enriched in the joints of patients with psoriatic arthritis and correlate with disease activity and joint damage progression. Arthritis Rheumatol 2014, 66, 1272–81. doi: 10.1002/art.38376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CEM, Papp K, et al.; ERASURE Study Group. Secukinumab in plaque psoriasis – results of two phase 3 trials. N Engl J Med 2014, 371, 326–38. doi: 10.1056/NEJMoa1314258. [DOI] [PubMed] [Google Scholar]
- 222.Griffiths CEM, Strober BE, van de Kerkhof P, Ho V, Fidelus-Gort R, Yeilding N, et al.; ACCEPT Study Group. Comparison of ustekinumab and etanercept for moderate-to-severe psoriasis. N Engl J Med 2010, 362, 118–28. doi: 10.1056/NEJMoa0810652. [DOI] [PubMed] [Google Scholar]
- 223.Mease PJ, Genovese MC, Greenwald MW, Ritchlin CT, Beaulieu AD, Deodhar A, et al. Brodalumab, an anti-IL17RA monoclonal antibody, in psoriatic arthritis. N Engl J Med 2014, 370, 2295–306. doi: 10.1056/NEJMoa1315231. [DOI] [PubMed] [Google Scholar]
- 224.Gordon KB, Strober B, Lebwohl M, Augustin M, Blauvelt A, Poulin Y, et al. Efficacy and safety of risankizumab in moderate-to-severe plaque psoriasis (UltIMMa-1 and UltIMMa-2): results from two double-blind, randomised, placebo-controlled and ustekinumab-controlled phase 3 trials. Lancet 2018, 392, 650–61. doi: 10.1016/S0140-6736(18)31713-6. [DOI] [PubMed] [Google Scholar]
- 225.Reich K, Papp KA, Blauvelt A, Langley RG, Armstrong A, Warren RB, et al. Bimekizumab versus ustekinumab for the treatment of moderate to severe plaque psoriasis (BE VIVID): efficacy and safety from a 52-week, multicentre, double-blind, active comparator and placebo controlled phase 3 trial. Lancet 2021, 397, 487–98. doi: 10.1016/S0140-6736(21)00125-2. [DOI] [PubMed] [Google Scholar]
- 226.McInnes IB, Asahina A, Coates LC, Landewé R, Merola JF, Ritchlin CT, et al. Bimekizumab in patients with psoriatic arthritis, naive to biologic treatment: a randomised, double-blind, placebo-controlled, phase 3 trial (BE OPTIMAL). Lancet 2023, 401, 25–37. doi: 10.1016/S0140-6736(22)02302-9. [DOI] [PubMed] [Google Scholar]
- 227.Reich K, Warren RB, Lebwohl M, Gooderham M, Strober B, Langley RG, et al. Bimekizumab versus secukinumab in plaque psoriasis. N Engl J Med 2021, 385, 142–52. doi: 10.1056/NEJMoa2102383. [DOI] [PubMed] [Google Scholar]
- 228.Silacci M, Lembke W, Woods R, Attinger-Toller I, Baenziger-Tobler N, Batey S, et al. Discovery and characterization of COVA322, a clinical-stage bispecific TNF/IL-17A inhibitor for the treatment of inflammatory diseases. MAbs 2016, 8, 141–9. doi: 10.1080/19420862.2015.1093266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Krupashankar DS, Dogra S, Kura M, Saraswat A, Budamakuntla L, Sumathy TK, et al. Efficacy and safety of itolizumab, a novel anti-CD6 monoclonal antibody, in patients with moderate to severe chronic plaque psoriasis: Results of a double-blind, randomized, placebo-controlled, phase-III study. J Am Acad Dermatol 2014, 71, 484–92. doi: 10.1016/j.jaad.2014.01.897. [DOI] [PubMed] [Google Scholar]
- 230.Reich DS, Lucchinetti CF, Calabresi PA.. Multiple sclerosis. N Engl J Med 2018, 378, 169–80. doi: 10.1056/NEJMra1401483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Matusevicius D, Kivisäkk P, He B, Kostulas N, Ozenci V, Fredrikson S, et al. Interleukin-17 mRNA expression in blood and CSF mononuclear cells is augmented in multiple sclerosis. Mult Scler 1999, 5, 101–4. doi: 10.1177/135245859900500206. [DOI] [PubMed] [Google Scholar]
- 232.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: 10.1084/jem.20041257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Tzartos JS, Friese MA, Craner MJ, Palace J, Newcombe J, Esiri MM, et al. Interleukin-17 production in central nervous system-infiltrating T cells and glial cells is associated with active disease in multiple sclerosis. Am J Pathol 2008, 172, 146–55. doi: 10.2353/ajpath.2008.070690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Rostami A, Ciric B.. Role of Th17 cells in the pathogenesis of CNS inflammatory demyelination. J Neurol Sci 2013, 333, 76–87. doi: 10.1016/j.jns.2013.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Kebir H, Kreymborg K, Ifergan I, Dodelet-Devillers A, Cayrol R, Bernard M, et al. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat Med 2007, 13, 1173–5. doi: 10.1038/nm1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Wang HH, Dai YQ, Qiu W, Lu ZQ, Peng FH, Wang YG, et al. Interleukin-17-secreting T cells in neuromyelitis optica and multiple sclerosis during relapse. J Clin Neurosci 2011, 18, 1313–7. doi: 10.1016/j.jocn.2011.01.031. [DOI] [PubMed] [Google Scholar]
- 237.Peelen E, Thewissen M, Knippenberg S, Smolders J, Muris A-H, Menheere P, et al. Fraction of IL-10+ and IL-17+ CD8 T cells is increased in MS patients in remission and during a relapse, but is not influenced by immune modulators. J Neuroimmunol 2013, 258, 77–84. doi: 10.1016/j.jneuroim.2013.02.014. [DOI] [PubMed] [Google Scholar]
- 238.Lolli F, Martini H, Citro A, Franceschini D, Portaccio E, Amato MP, et al. Increased CD8+ T cell responses to apoptotic T cell-associated antigens in multiple sclerosis. J Neuroinflammation 2013, 10, 94. doi: 10.1186/1742-2094-10-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Salehi Z, Doosti R, Beheshti M, Janzamin E, Sahraian MA, Izad M.. Differential frequency of CD8+ T cell subsets in multiple sclerosis patients with various clinical patterns. PLoS One 2016, 11, e0159565. doi: 10.1371/journal.pone.0159565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.McGinley AM, Sutton CE, Edwards SC, Leane CM, DeCourcey J, Teijeiro A, et al. Interleukin-17A serves a priming role in autoimmunity by recruiting IL-1β-producing myeloid cells that promote pathogenic T cells. Immunity 2020, 52, 342–356.e6. doi: 10.1016/j.immuni.2020.01.002. [DOI] [PubMed] [Google Scholar]
- 241.Constantinescu CS, Farooqi N, O’Brien K, Gran B.. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 2011, 164, 1079–106. doi: 10.1111/j.1476-5381.2011.01302.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Mardiguian S, Serres S, Ladds E, Campbell SJ, Wilainam P, McFadyen C, et al. Anti-IL-17A treatment reduces clinical score and VCAM-1 expression detected by in vivo magnetic resonance imaging in chronic relapsing EAE ABH mice. Am J Pathol 2013, 182, 2071–81. doi: 10.1016/j.ajpath.2013.02.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Havrdová E, Belova A, Goloborodko A, Tisserant A, Wright A, Wallstroem E, et al. Activity of secukinumab, an anti-IL-17A antibody, on brain lesions in RRMS: results from a randomized, proof-of-concept study. J Neurol 2016, 263, 1287–95. doi: 10.1007/s00415-016-8128-x. [DOI] [PubMed] [Google Scholar]
- 244.Bittner S, Wiendl H.. Neuroimmunotherapies targeting T cells: from pathophysiology to therapeutic applications. Neurotherapeutics 2016, 13, 4–19. doi: 10.1007/s13311-015-0405-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Wiendl H, Dahlke F, Bennett D, Rosenkranz G, Wolf C B-OA.. IL-17 neutralization by subcutaneous CJM112, a fully human anti IL-17A monoclonal antibody for the treatment of relapsing-remitting multiple sclerosis: study design of a phase 2 trial. In: European Committee for Treatment and Research in Multiple Sclerosis 2015, Barcelona, 2015. [Google Scholar]
- 246.Belkaid Y, Harrison OJ.. Homeostatic immunity and the microbiota. Immunity 2017, 46, 562–76. doi: 10.1016/j.immuni.2017.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Neurath MF. Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat Immunol 2019, 20, 970–9. doi: 10.1038/s41590-019-0415-0. [DOI] [PubMed] [Google Scholar]
- 248.Moschen AR, Tilg H, Raine T.. IL-12, IL-23 and IL-17 in IBD: immunobiology and therapeutic targeting. Nat Rev Gastroenterol Hepatol 2019, 16, 185–96. doi: 10.1038/s41575-018-0084-8. [DOI] [PubMed] [Google Scholar]
- 249.Fujino S, Andoh A, Bamba S, Ogawa A, Hata K, Araki Y, et al. Increased expression of interleukin 17 in inflammatory bowel disease. Gut 2003, 52, 65–70. doi: 10.1136/gut.52.1.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Globig AM, Hennecke N, Martin B, Seidl M, Ruf G, Hasselblatt P, et al. Comprehensive intestinal T helper cell profiling reveals specific accumulation of IFN-γ+IL-17+coproducing CD4+ T cells in active inflammatory bowel disease. Inflamm Bowel Dis 2014, 20, 2321–9. doi: 10.1097/MIB.0000000000000210. [DOI] [PubMed] [Google Scholar]
- 251.Bhaumik S, Basu R.. Cellular and molecular dynamics of Th17 differentiation and its developmental plasticity in the intestinal immune response. Front Immunol 2017, 8, 254. doi: 10.3389/fimmu.2017.00254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Smillie CS, Biton M, Ordovas-Montanes J, Sullivan KM, Burgin G, Graham DB, et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell 2019, 178, 714–30.e22. doi: 10.1016/j.cell.2019.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Tajima M, Wakita D, Noguchi D, Chamoto K, Yue Z, Fugo K, et al. IL-6–dependent spontaneous proliferation is required for the induction of colitogenic IL-17–producing CD8+ T cells. J Exp Med 2008, 205, 1019–27. doi: 10.1084/jem.20071133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Luu M, Weigand K, Wedi F, Breidenbend C, Leister H, Pautz S, et al. Regulation of the effector function of CD8+ T cells by gut microbiota-derived metabolite butyrate. Sci Rep 2018, 8, 14430. doi: 10.1038/s41598-018-32860-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, Reinisch W, Higgins PDR, et al.; Secukinumab in Crohn's Disease Study Group. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo- controlled trial. Gut 2012, 61, 1693–700. doi: 10.1136/gutjnl-2011-301668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Targan SR, Feagan B, Vermeire S, Panaccione R, Melmed GY, Landers C, et al. A randomized, double-blind, placebo-controlled phase 2 study of brodalumab in patients with moderate-to-severe Crohn’s disease. Am J Gastroenterol 2016, 111, 1599–607. doi: 10.1038/ajg.2016.298. [DOI] [PubMed] [Google Scholar]
- 257.Lee JS, Tato CM, Joyce-Shaikh B, Gulen MF, Cayatte C, Chen Y, et al. Interleukin-23-independent IL-17 production regulates intestinal epithelial permeability. Immunity 2015, 43, 727–38. doi: 10.1016/j.immuni.2015.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Maxwell JR, Zhang Y, Brown WA, Smith CL, Byrne FR, Fiorino M, et al. Differential roles for interleukin-23 and interleukin-17 in intestinal immunoregulation. Immunity 2015, 43, 739–50. doi: 10.1016/j.immuni.2015.08.019. [DOI] [PubMed] [Google Scholar]
- 259.Chong WP, Mattapallil MJ, Raychaudhuri K, Bing SJ, Wu S, Zhong Y, et al. The cytokine IL-17A limits Th17 pathogenicity via a negative feedback loop driven by autocrine induction of IL-24. Immunity 2020, 53, 384–97.e5. doi: 10.1016/j.immuni.2020.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Feagan BG, Sandborn WJ, Gasink C, Jacobstein D, Lang Y, Friedman JR, et al.; UNITI–IM-UNITI Study Group. Ustekinumab as induction and maintenance therapy for Crohn’s disease. N Engl J Med 2016, 375, 1946–60. doi: 10.1056/NEJMoa1602773. [DOI] [PubMed] [Google Scholar]
- 261.Globig AM, Sommer NP, Wild K, Schardey J, Zoldan K, Thomann AK, et al. Ustekinumab inhibits T follicular helper cell differentiation in patients with Crohn’s disease. Cell Mol Gastroenterol Hepatol 2021, 11, 1–12. doi: 10.1016/j.jcmgh.2020.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.D’Haens G, Panaccione R, Baert F, Bossuyt P, Colombel J-F, Danese S, et al. Risankizumab as induction therapy for Crohn’s disease: results from the phase 3 ADVANCE and MOTIVATE induction trials. Lancet 2022, 399, 2015–30. doi: 10.1016/S0140-6736(22)00467-6. [DOI] [PubMed] [Google Scholar]
- 263.Ferrante M, Panaccione R, Baert F, Bossuyt P, Colombel J-F, Danese S, et al. Risankizumab as maintenance therapy for moderately to severely active Crohn’s disease: results from the multicentre, randomised, double-blind, placebo-controlled, withdrawal phase 3 FORTIFY maintenance trial. Lancet 2022, 399, 2031–46. doi: 10.1016/S0140-6736(22)00466-4. [DOI] [PubMed] [Google Scholar]
- 264.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: 10.1038/nri3707. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No new data were generated in this study.

