CD4+ T helper (Th) cells exist in a variety of epigenetic states that determine their function, phenotype, and capacity for persistence. These polarization states include Th1, Th2, Th17, and Foxp3+ T regulatory cells, as well as the more recently described T follicular helper, Th9, and Th22 cells. Th17 cells express the master transcriptional regulator retinoic acid–related orphan receptor γ thymus and produce canonical interleukin (IL)-17A and IL-17F cytokines. Th17 cells display a great degree of context-dependent plasticity, as they are capable of acquiring functional characteristics of Th1 cells. This late plasticity may contribute to the protection against microbes, plays a role in the development of autoimmunity, and is necessary for antitumor activity of Th17 cells in adoptive cell transfer therapy models. Moreover, plasticity of this subset is associated with higher in vivo survival and self-renewal capacity and less senescence than Th1 polarized cells, which have less plasticity and more phenotypic stability. New findings indicate that subset polarization of CD4+ T cells not only induces characteristic patterns of surface markers and cytokine production but also has a maturational aspect that affects a cell’s ability to survive, respond to secondary stimulation, and form long-term immune memory.

CD4+ T helper (Th) cells are central to the normal functioning of the entire immune system,1  coordinating the expansion and regulation of CD8+ T cells, facilitating B-cell responses, and recruiting and modulating multiple components of innate immunity.2-5  The initial antigenic encounter of naive CD4+ T cells varies depending on the anatomical site, pathogen type, and presence of assorted cytokines and costimulatory molecules, and these variations cause Th cell differentiation into antigen-experienced effectors with distinct functional characteristics termed polarization states.

In their early descriptions of the Th1/Th2 paradigm, Mossman, Coffman and colleagues6 -8 attempted to explain responses observed in many experimental models of infection, autoimmunity, and allergy.9  Th1 cells were considered essential for antiviral immunity and for providing help to CD8+ cytotoxic T cells and were viewed as the main perpetrators of autoimmunity.10,11  Th1 cells are promoted by interleukin (IL)-12 (via signal transducer and activator of transcription [Stat]4) signaling and interferon (IFN)-γ (via Stat1), which induce the expression of master transcription factor T-box 21 (tbx21, T-bet) and secretion of the hallmark cytokine IFN-γ.12-14  Th2 cells, induced by IL-4 via Stat6 signaling that up-regulates the transcription factor GATA3,15,16  were linked to humoral responses to extracellular organisms or parasites and development of atopy and allergic reactions,17-20  producing cytokines that include IL-4, IL-5, and IL-13. It seemed that Th1 and Th2 cells represented mutually exclusive binary epigenetic states because IL-12, IFN-γ, and the expression of T-bet inhibited Th2 differentiation, whereas IL-4 and Gata3 expression antagonized Th1 polarization.21-25  The discovery of FoxP3-expressing regulatory CD4+ T cells (Treg) that restrained immune responses complement these early views of Th differentiation.26  However, not all CD4+ T-cell activity could easily be classified as either type 1 or type 2. Finally, Th17 cells producing the canonical cytokine IL-17A emerged as the most prominent addition to the old paradigm of two-mode Th polarization.27,28 

Harrington et al29  and Park et al30  established that Th17 cells were a true distinct lineage whose in vitro generation was enhanced when IL-4 and IFN-γ were blocked and was independent of Th1- or Th2-related transcription factors (T-bet, Stat1, Stat4, and Stat6). Induction of the Th17 polarization program was initially attributed to the effect of IL-23 (a heterodimer of p40 shared with IL-12 and p19 subunit) signaling via its distinct IL-23 receptor (IL-23R), triggering the Stat3 pathway31 ; however, this process produced cultures containing only small fractions of IL-17A–secreting Th cells.32  Subsequently, Th17 cells were efficiently generated in the presence of transforming growth factor (TGF)-β1 and IL-6, which signaled via Smad family proteins (Smads) and Stat3, respectively (Figure 1).33 

Figure 1

Schematic representation of signaling and transcriptional regulation of Th17 polarization. Th17 cells are induced upon T-cell receptor activation in the presence of cytokines that activate Stat3, including IL-6, IL-21, and IL-23. IL-12 and IFN-γ, which signal via Stat4 and Stat1, respectively, promote type 1 differentiation and inhibit Th17 polarization. However, IL-23 also activates Stat4 (not shown), and it remains perplexing that animals deficient in Stat4 have impaired functionality of Th17 cells. Similarly, IL-4 signaling via Stat6 inhibits Th17 polarization and promotes type 2 differentiation. Phosphorylated Stat3 (pStat3) binds to the promoter regions and activates transcription of genes encoding master regulators of Th17 polarization: rorc (encoding Rorγt) and rora (encoding Rora) transcription factors. Batf and Irf4 form a functional complex that plays a central role in Rorγt-mediated activation of the type 17 molecular signature.41  Together with pStat3, Rorγt and Rorα activate the expression of genes encoding canonical Th17-associated cytokines IL-17A and IL-17F, as well as IL-21, IL-22, and others. IL-6–induced activation of Stat3 also augments the expression of the IL-23 receptor (IL-23R), thus increasing the sensitivity of early Th17 cells to the polarizing effects of IL-23. pStat3 also induces expression of Hif1α, which inhibits FoxP3 and promotes Th17 differentiation. IL-21 secreted by early Th17 cells acts in a self-amplified autocrine loop via the IL-21 receptor. IL-1 promotes Th17 polarization via activities of p38 mitogen-activated protein kinase (MAPK) and the Akt/mTOR pathway. IL-1 also induces Irf4, which directly augments IL-21 secretion. Th17 polarization is also increased by activation of aryl hydrocarbon receptors (Ahr). TGF-β1 signals via Smads that most likely limit expression of genes encoding T-bet, Gata3, and other Th1- and Th2-associated factors, thus increasing Th17 differentiation. TGF-β1 signaling in conjunction with retinoic acid (RA) and IL-2–induced pStat5 promotes FoxP3 expression and Treg differentiation. RA has been shown to either limit or (in lower concentrations) augment Th17 polarization. pStat5 directly inhibits pStat3 binding to IL-17 promoter (not shown). Both FoxP3 and Rorγt form complexes with Runx1 and reciprocally regulate each other.

Figure 1

Schematic representation of signaling and transcriptional regulation of Th17 polarization. Th17 cells are induced upon T-cell receptor activation in the presence of cytokines that activate Stat3, including IL-6, IL-21, and IL-23. IL-12 and IFN-γ, which signal via Stat4 and Stat1, respectively, promote type 1 differentiation and inhibit Th17 polarization. However, IL-23 also activates Stat4 (not shown), and it remains perplexing that animals deficient in Stat4 have impaired functionality of Th17 cells. Similarly, IL-4 signaling via Stat6 inhibits Th17 polarization and promotes type 2 differentiation. Phosphorylated Stat3 (pStat3) binds to the promoter regions and activates transcription of genes encoding master regulators of Th17 polarization: rorc (encoding Rorγt) and rora (encoding Rora) transcription factors. Batf and Irf4 form a functional complex that plays a central role in Rorγt-mediated activation of the type 17 molecular signature.41  Together with pStat3, Rorγt and Rorα activate the expression of genes encoding canonical Th17-associated cytokines IL-17A and IL-17F, as well as IL-21, IL-22, and others. IL-6–induced activation of Stat3 also augments the expression of the IL-23 receptor (IL-23R), thus increasing the sensitivity of early Th17 cells to the polarizing effects of IL-23. pStat3 also induces expression of Hif1α, which inhibits FoxP3 and promotes Th17 differentiation. IL-21 secreted by early Th17 cells acts in a self-amplified autocrine loop via the IL-21 receptor. IL-1 promotes Th17 polarization via activities of p38 mitogen-activated protein kinase (MAPK) and the Akt/mTOR pathway. IL-1 also induces Irf4, which directly augments IL-21 secretion. Th17 polarization is also increased by activation of aryl hydrocarbon receptors (Ahr). TGF-β1 signals via Smads that most likely limit expression of genes encoding T-bet, Gata3, and other Th1- and Th2-associated factors, thus increasing Th17 differentiation. TGF-β1 signaling in conjunction with retinoic acid (RA) and IL-2–induced pStat5 promotes FoxP3 expression and Treg differentiation. RA has been shown to either limit or (in lower concentrations) augment Th17 polarization. pStat5 directly inhibits pStat3 binding to IL-17 promoter (not shown). Both FoxP3 and Rorγt form complexes with Runx1 and reciprocally regulate each other.

Close modal

Retinoic acid (RA)–related orphan receptor γ thymus (Rorγt), encoded by the gene Rorc, was identified as the master transcription factor defining Th17 cells as a distinct lineage.34  RA-related orphan receptor α (Rorα) was recognized as the additional transcription factor critical for Th17 differentiation.35  The genetic deletion of Stat3 abrogates Th17 differentiation, and genomewide chromatin-immunoprecipitation sequencing (Chip-seq) revealed a broad and pleiotropic function of Stat3 as a promoter of Rorc, Il17, Il17f, and multiple other genes associated with Th17 polarization and survival.36  The transcriptional network controlling type 17 differentiation also includes Batf/Irf4 complexes that mediate chromatin accessibility, cMaf, Runx1, and hypoxia inducible factor 1 alpha (Hif1α), among others.37-41  IL-2 signaling via Stat5 attenuates Th17 polarization, and Stat5 has a direct negative effect on IL-17A gene expression.42,43  IFN-γ and IL-27 (via Stat1) and IL-12 (via Stat4) clearly block Th17 differentiation, but the underlying mechanism remains unclear and possibly related to T-bet repressor activity.44,45  Perplexingly, Stat4 deficiency also impairs type 17 polarization, possibly because of lower IL-23R expression.46 

Naïve CD4+ T cells express very low levels of IL-23R, but its expression is induced on exposure to TGF-β1 and IL-6.47  Naïve CD4+ T cells cultured in only IL-6 and TGF-β and not in IL-23 secrete IL-17A but also produce high levels of IL-10 and fail to induce experimental autoimmune encephalitis (EAE) on adoptive cell transfer.48  Furthermore, in vivo IL-23R–deficient CD4+ Th17 cells fail to maintain their phenotype and demonstrate reduced persistence.49  Thus, IL-23 is not required for the initial Th17 polarization but instead stabilizes and expands pathogenic Th17-polarized cells.

Many additional cytokines and other molecules have been identified as factors regulating Th17 induction and expansion. Notably IL-21 (via Stat3 signaling) and TGF-β are sufficient to induce Rorγt and IL-23R expression and drive robust Th17 polarization in the absence of IL-6.50  In the presence of IL-6, stimulated CD4+ T cells produce IL-21 themselves, which further augments its secretion in an autocrine self-amplifying loop.51,52  IL-1β has emerged as another crucial factor driving Th17 polarization in a variety of inflammatory conditions.53  IL-1 serves as a prototypic inflammasome-related cytokine produced by cells of myeloid origin.54  IL-1 signaling promotes proliferation and survival of antigenically stimulated Th cells. This process involves activation of the serine-threonine protein kinase Akt-mammalian target of rapamycin (Akt-mTOR) pathway via induction of Ikbke inhibitor of kappaB kinase epsilon (IKKi) that inhibits glycogen synthtase kinase 3 (GSK3)α.55  IL-1 also induces Irf4, a regulator of the IL-21 autocrine signaling loop.56-58  Irf4-deficient animals have increased numbers of Foxp3+ Tregs and a diminished ability to form Th17 responses despite intact Stat3 signaling.39 

The contribution of TGF-β1 to Th17 polarization remains a matter of debate, perhaps in part because it can be found in biologically significant concentrations in culture media containing animal or human sera.59,60  Animals deficient in TGF-β signaling have Th17 cells in their gut mucosa,53  and Th17 cells generated using serum-free (thus, TGF-β–free) media in the presence of IL-1, IL-6, and IL-23 coexpress RORγt and T-bet and cause aggressive EAE.53  TGF-β1 inhibits IL-2–mediated STAT5 signaling and diminishes the expression of the Th1- and Th2-defining transcription factors T-bet and Gata3, respectively.61  Thus, at low concentrations, TGF-β1 contributes indirectly to the initial development of Th17 responses but is not critical to the inflammatory milieu that drives type 17 responses.62  At high concentrations, TGF-β signaling inhibits IL-23R expression and converts naïve Th cells into induced FoxP3+ regulatory cells (iTregs) by antagonizing Rorγt.63  This interaction involves the Runx1 transcription factor that binds either Rorγt or FoxP3.40  Thus, there is an active balance between the development of either iTregs or Th17 cells and even plasticity with FoxP3+ T cells capable of converting into FoxP3 Th17 progeny.64  All-trans retinoic acid (ATRA), a vitamin A metabolite produced in the gut mucosa, antagonizes the expression of rorc and promotes in vitro foxp3 expression via its nuclear receptors.65  Akin to the inhibitory function of TGF-β, ATRA also has been reported to negatively regulate Th1 and Th2 polarization. Both TGF-β and ATRA induce expression of Mir10a that might stabilize iTreg phenotype and limit Th17 differentiation.66  The same report indicated that physiological concentrations of ATRA might promote, rather than impair, Th17 polarization.66  Indeed, in vivo ATRA has a proinflammatory effect and is required for the efficient Th17 responses against mucosal pathogens.67 

Aryl hydrocarbon receptors (AHRs) are the cytosolic receptors with transcription factor activity that have been reported to promote Th17 polarization.68  AHRs sense a variety of small natural and man-made compounds including products of tryptophan metabolism and highly toxic dioxins, providing an intriguing link between the environment and autoimmunity.69  Early Th17 polarization induces high expression of AHRs, and AHR-deficient animals are partly protected from the induction of EAE.68 

Gut-residing commensal microbiota can profoundly affect Th17-mediated immunity and autoimmunity. Germ-free animals have fewer intraepithelial intestinal Th17 cells and an attenuated form of autoimmune arthritis but develop severe arthritis after exposure to segmented filamentous bacteria.70,71  Conversely, infected nonobese diabetic (NOD) mice were protected from the progression of diabetes.72 

Under physiological conditions, IL-17–producing CD4+ T cells reside mainly in the lamina propria of the small intestine but are readily induced at other mucosal sites during infection or vaccination.34,71,73,74  Consequently, Th17 cells and type 17–associated cytokines contribute to the protection against a variety of predominantly extracellular bacterial and fungal pathogens in the gastrointestinal tract, airway, lungs, and skin.75,76 

Th17 cells secrete IL-17 (IL-17A), IL-17F, and IL-17A/F heterodimers, as well as IL-21, IL-22, granulocyte macrophage–colony-stimulating factor (GM-CSF), and many other factors.77  The proinflammatory effects of IL-17 are mediated through the IL-17 receptor (IL-17R), composed of IL-17RA and IL-17RC subunits.78  IL-17R is widely expressed by immune system cells (T and B lymphocytes and neutrophils), and by many other tissues (epithelium, endothelium, fibroblasts, mesenchymal stromal cells, and keratinocytes).78  Thus, IL-17 is highly pleiotropic, causing a variety of inflammatory effects that link adaptive and innate immunity.75  Other rich sources of IL-17 include γσ T cells and innate lymphocytes.

IL-17A promotes granulopoiesis by triggering the secretion of granulocyte–colony-stimulating factor in bone marrow stroma,79  and IL-17R–deficient mice show a deeply impaired ability to reconstitute following irradiation.80  In the gut, IL-17 maintains mucosal homeostasis and integrity.81  IL-17 induces GM-CSF, tumor necrosis factor (TNF)α, IL-1β, IL-6, and multiple chemokines and chemokine receptors that act as powerful chemo-attractants for granulocytes. IL-17 also mediates formation of inducible secondary lymphoid tissues following local infection.82  IL-17 and Th17 cells play an important role in immunity against Klebsiella pneumoniae73  and many other bacteria including Staphylococcus aureus, Citrobacter rodentium, Salmonella and Shigella sp., and Bordetella pertusis.76,83  Th17 cells may augment Th1 responses against some intracellular pathogens (Mycobacteria and Francisella tularensis).84,85  Th17 cells are also involved in protection against mucocutanoeous candidiasis via the stimulation of pattern recognition elements (Dectin 1 and Dectin 2) in macrophages and dendritic cells.86  The role for Th17 cells and IL-17 in immunity against Asperillus fumigatus and some other fungi is more controversial and possibly detrimental.75,87 

IL-22 and IL-17 synergistically trigger production of many antimicrobial peptides and proteins (β-defensins, S100 proteins, regenerating islet-derived protein 3 γ, and lipocalin 2).88  The IL-22 receptor (IL-22R) is predominantly expressed by cells of nonhematopoietic origin (keratinocytes, hepatocytes, and enterocytes) and enables tissue-protective effects against invasion by Toxoplasma gondii and various gram-negative bacteria.89  IL-21 regulates CD8-mediated responses and facilitates B-cell responses and memory formation.90 

The importance of the Th17 subset in protecting humans is vividly demonstrated in patients with an autosomal-dominant form of hyper-IgE syndrome (Job’s syndrome) that have an inactivating STAT3 mutation; thus, they are incapable of generating Th17 cells.91,92  Patients with hyper-IgE syndrome suffer from recurrent staphylococcal soft tissue infections, pneumonias and bronchoalveolar infections, pneumatoceles, eczema, and mucocutaneous candidiasis. They also show impaired immunity against varicella zoster (VZV), Epstein Barr (EBV), and defective proliferative responses and reduced central memory formation caused by STAT3 dysfunction.93  Similar sequelae are seen in HIV infection, which is associated with early preferential loss of Th17-mediated mucosal immunity in the gut.92  Th17 depletion in the gastrointestinal tract leads to rapid disease progression in simian immunodeficiency virus–infected macaques, whereas sooty mangabeys that retain Th17 cells in the gastrointestinal mucosa are protected.94 

Early discoveries of IL-23 as an inducer of Th17 cells revealed their capacity to induce EAE, a disease initially attributed to Th1 cells and induced in the presence of IL-12 (a heterodimer of IL-12 p40 and p35 subunits). However, animals deficient in the IL-12 p35 subunit developed EAE despite diminished Th1 responses95  because they retained IL-23, which is a heterodimer of IL-12 p40 and a distinct p19 subunit.31  These experimental data were complemented by studies linking genetic polymorphisms of IL-23R and its downstream signaling pathways with susceptibility to human autoimmune diseases including inflammatory bowel disease, psoriasis, and aplastic anemia.96-98 

Tissue damage by Th17 cells might be caused by direct recognition of the antigen-specific target, or it can result from the recruitment of neutrophils and macrophages into the microenvironment.99  In EAE, IL-17 induces reactive oxygen species production in the endothelium, leading to a disruption of the blood–brain barrier.100  IL-17 promotes inflammatory cytokines (IL-1 and IL-6) and CCL20 (MIP3α), a potent chemotactic factor for myeloid cells, and additional Th17 cells expressing high levels of its receptor CCR6.101  IL-9 produced by Th17 cells exacerbates EAE via its effect on T cells and IL-6–secreting macrophages.102  Th17 cells also produce GM-CSF,103,104  which acts on myeloid cells by induction of IL-1, IL-6, and IL-23 and major histocompatibility complex class II expression.103  Blockade of GM-CSF signaling effectively protected mice from autoimmunity.104  Nevertheless, despite the latest emphasis on Th17 cells, both Th1 and Th17 cells can cause EAE and murine uveitis manifestations, albeit with distinct histopathological features.105-107  Furthermore, both type 1– and type 17–related factors can be produced by pathogenic Th17 cells, but both subset-defining cytokines, IL-17A and IFN-γ, are dispensable, whereas T-bet, thought of as the master transcription factor for Th1 cells, may be essential for pathogenicity of Th17 cells in EAE.108  Recently, TGF-β3 was identified as a critical IL-23– and T-bet–dependent factor produced by the highly pathogenic Th17 population.109  Moreover, the presence of TGF-β3 during initial Th17 polarization induced highly encephalitogenic T-bet–independent type 17 cells.

In patients with multiple sclerosis, elevated frequencies of Th17 cells were found in peripheral blood, and double producers of IL-17A and IFN-γ have been reported in central nervous system (CNS) lesions, mimicking the findings observed in mice with EAE. However, depletion of IL-23 in patients with multiple sclerosis caused only minimal improvement in disease control, possibly because of poor CNS penetration of anti–IL-23 antibody formulation used clinically or deficiencies in the mouse model.110 

Th17 responses are associated with psoriasis, where IL-23, IL-17, and IL-22 seem to contribute to the development of the human disease.111,112  In mice, IL-22 is crucial in inducing psoriatic-like lesions with acanthocytosis in kerationocytes.113  Targeting the p40 subunit of IL-12/IL-23 with the monoclonal antibody ustekinumab appears to be highly effective in patients with psoriasis.114,115  Recently, a phase 2 clinical trial of the anti–IL-17 receptor monoclonal antibody brodalumab in patients with psoriasis demonstrated significant improvement in a majority of patients.116 

IL-23 directly drives pathogenic Th cells in a mouse model of colitis.117  Whereas IFN-γ is required for tissue damage, the role of IL-17 remains disputed.118  IL-17 might protect the colon via inhibition of colitogenic Th1 differentiation.119  Likewise, IL-22 from Th17 and natural killer cells might attenuate the development of intestinal pathology via Stat3-mediated effects on epithelial cells.120,121  In contrast, IL-21 significantly potentiates pathogenic effects of Th17 cells in the gut.122,123  Increased frequency of IL-17–secreting CD4+ T cells can be found in the gut of patients with Crohn disease,124  and these cells express CD161 in addition to the previously described CCR6 and CCR4 chemokine receptors.101,125  In the clinical randomized proof of concept trial, the human anti–IL-17A monoclonal antibody failed to control the symptoms of Crohn disease and was associated with increased adverse events in comparison with the placebo,126  whereas depletion of IL-23 with ustekinumab was more effective.127  This might underscore the role of IL-23 not only in Th17 responses but also as a driver of inflammation mediated by the innate lymphoid cells that are abundant in the colonic mucosa.128 

Multiple other autoimmune diseases including rheumatoid arthritis, uveitis, atherosclerosis, emphysema, and allergies have been associated with Th17 responses in murine models and in patients.129,130  Currently, various agents modulating the type 17 axis of inflammation are under development. Recently, digoxin has been shown to inhibit Th17 development, and novel small molecules that modulate Rora and Rorγt activity have been described.131,132  Nevertheless, IL-23 rather than IL-17 plays a central role in Th17-related autoimmunity, and pathogenic Th17 cells usually evolve and display Th1-like characteristics.

Graft-versus-host disease (GVHD) is a complex immune syndrome that develops in organ-specific sites under influences of a variety of inflammatory conditions.133  The resemblance of GVHD to some autoimmune diseases has shed light on Th17 biology.134  IL-23R polymorphism might be a prognostic factor in patients undergoing allogeneic stem cell transplantation,135  and highly polarized allogeneic Th17 cells can induce severe forms of GVHD in mice.136  However, Th17 responses are frequently described as “sufficient but not necessary” for the development of GVHD, as evidenced by pathogenicity of IL-17A or Rorγt-deficient Th cells.137,138  Concurrent elimination of T-bet and Rorγt was reported as protective against GVHD development in fully mismatched settings (C57/BL6 into BALB/c), whereas the graft-versus-leukemia effect was spared.139  Perplexingly, a recent report demonstrated reduced numbers of Th17 cells within the lamina propria of animals with GVHD, but the development of GVHD colitis was attributed to the critical role of STAT3 signaling that promoted instability and plasticity of the nTreg compartment and blockade of iTreg generation.140  In a more clinically relevant haploidentical transplant model, rorc-deficient CD25-depleted CD4+ T cells induced only a mild form of GVHD.141  This protective effect correlated with diminished systemic levels of TNF-α and IL-17A, but colonic levels of IL-17A were preserved in mice treated with rorc-deficient CD4+ T cells. In the same model, tbx21−/− cells caused marked GVHD.

Undoubtedly, the functional plasticity of the Th17 subset that can produce Th1-like progeny is important for a better understanding of their complex role in mediating GVHD.134  Reminiscent of some autoimmune conditions, the role of IL-17 in GVHD is unclear, and IL-22 might be protective to colonic mucosa. IL-21, IL-6, IL-23, STAT3, and RA receptors have been reported as potentially useful targets for therapeutic intervention.142-145 

Carcinogenesis as a consequence of chronic inflammation was described in the 19th century by Rudolph Virchow. There is a strong correlation between cancer and autoimmunity (eg, in inflammatory bowel disease) or prolonged inflammation caused by cigarette smoke or chronic infection, such as Helicobacter pylori or hepatitis.146,147  Inflammation, especially involving Stat3 signaling pathways, plays an important role in the induction, progression, and metastasis of tumors either directly or by modulation of the various components of tumor stroma, recruitment of myeloid cells, and impairment of T-cell responses.148,149  IL-1, IL-6, IL-21, and IL-23 are frequently induced in the proinflammatory tumorigenic microenvironment, and Th17 cells are commonly found in precancerous and cancerous lesions.150,151 

The role of Th17 cells in protection or progression of tumors is controversial.152,153  IL-17 promoted tumor neovascularization in nude mice,154  supported neoplastic growth directly or via induction of IL-6,155  and induced myeloid suppressor cells,156  but could be also protective in immunocompetent animals.157,158  Th17 cells can suppress CD8-mediated immunosurvillance.159  Genetic Rorγt deficiency results in a slower growth of transplantable melanoma, and elimination of IL-23 or IL-21 inhibits carcinogenesis in some models.160-162  On the other hand, exogenous inflammatory cytokines can activate immune responses against tumors.163,164  In humans with cancer, the presence of Th17 cells is a poor prognostic indicator in some studies and a favorable indicator in other reports.153,165 

The unknown antigenic specificity of the Th populations in question undermines many animal and clinical studies. Furthermore, Th17 cells are often viewed as static and incapable of plasticity. We investigated the relative antitumor activity of Th1 and Th17 cells in the setting of adoptive immunotherapy of murine B16.10 melanoma using T-cell receptor transgenic CD4+ T cells recognizing melanocyte differentiation antigen tyrosinase related protein 1 (TRP-1).166  Th17-polarized TRP-1 cells swiftly eradicated large (>1 cm2) B16.10 melanomas on transfer into tumor-bearing C57/B6 animals, whereas Th1-polarized TRP-1 cells were significantly less effective. TRP-1 Th17 cells recruited endogenous cytotoxic CD8+ T cells into pulmonary metastases.167  The potency of tumor-specific type 17 responses has also been demonstrated using murine CD8+ T cells (Tc17) and genetically engineered human CD4+ T cells.168,169 

The Th1/Th2 paradigm implied that polarized lineages were mutually exclusive and stable, because of the self-enforcing nature of the signals involved in the acquisition of each lineage. This view of fixed phenotype and function was also initially applied to the newly defined Th17 subset. However, IL-17A and IFN-γ double-producing cells are found in vivo, suggesting that Th17 cells could at least in part function like Th1 cells.77  In the setting of immunotherapy of melanoma with Th17-polarized TRP-1 cells, in vivo elimination of IFN-γ or T-bet abrogated the antitumor effect of adoptively transferred Th17 cells, suggesting evolution of a transferred population into a Th1-like subset.166,170  This late developmental plasticity of the Th17 lineage was examined in detail using the IL-17F reporter system by Lee et al,171  who demonstrated that type 17 cells could be maintained as such in the presence of TGF-β and IL-23 but were readily converted into the IFN-γ–secreting population on restimulation in the presence of IL-12 or IL-23 and in the absence of TGF-β. An analogous switch occurs in vivo where transferred IL-17F–expressing CD4+ T cells give rise to IFN-γ–producing colitogenic effectors.171  In a model of diabetes mellitus, Th17 cells converted into pathogenic IFN-γ–producing daughter cells in lymphopenic conditions, but in the intact (lymphoreplete) recipients, the same Th17 cells caused only benign insulitis and maintained a stable type 17 phenotype.172 

The observed stability or plasticity of Th subsets is governed by the epigenetic regulation of the key transcription factors and cytokines determining the polarization status.173  Whether a particular gene is poised for expression or not is determined by the chromatin structure, as well as histone and DNA methylation states. Wei et al174  evaluated histone 3H methylation status over the entire genome in a variety of CD4+ Th subsets. Predictably, gene promoters for tbx21 in Th1 cells and rorc in Th17 cells displayed a permissive methylation state (H3K4me3) associated with full expression of these master regulators in each lineage. However, gene promoters for tbx21 in Th17 cells had a bivalent status characterized by H3K4me3/H3K27me3 dual positivity, thus substantiating the relative instability and propensity of these cells to acquire type 1 features in the presence of IL-12 (Figure 2).

Figure 2

Epigenetic mechanisms explain the lineage relationship between Th17, Th1, and iTreg subsets. (A) Whether a particular gene is poised for expression or not is determined by the chromatin structure, as well as histone and DNA methylation states. Trimethylation of histone 3H on lysine 4 (H3K4me3) is considered permissive for gene expression, whereas trimethylation of lysine 27 on histone 3H (H3K27me3) is a marker of gene silencing. In some cases, both states can be found in a gene locus, thus making it susceptible for either expression or negative regulation. (B) Plasticity of Th1, Th17, and iTreg cells is constrained by the epigenetic status of genes encoding for the master transcriptional regulators of polarization and canonical cytokines. Th17 cells display permissive H3K4me3 histone modification over the rorc and il17 genes and bivalent, poised status over loci encoding for tbx21 and foxp3, thus substantiating the relative instability of this subset and its propensity to evolve into Th1 progeny. In contrast, Th1 cells display only repressive H3K27me3 methylation status over gene loci encoding for rorc and Il17, rendering them much more stable. iTregs are another relatively unstable subset that can acquire Th1 or Th17 properties, based on the poised bivalent status of type 1– or type 17–associated genes.

Figure 2

Epigenetic mechanisms explain the lineage relationship between Th17, Th1, and iTreg subsets. (A) Whether a particular gene is poised for expression or not is determined by the chromatin structure, as well as histone and DNA methylation states. Trimethylation of histone 3H on lysine 4 (H3K4me3) is considered permissive for gene expression, whereas trimethylation of lysine 27 on histone 3H (H3K27me3) is a marker of gene silencing. In some cases, both states can be found in a gene locus, thus making it susceptible for either expression or negative regulation. (B) Plasticity of Th1, Th17, and iTreg cells is constrained by the epigenetic status of genes encoding for the master transcriptional regulators of polarization and canonical cytokines. Th17 cells display permissive H3K4me3 histone modification over the rorc and il17 genes and bivalent, poised status over loci encoding for tbx21 and foxp3, thus substantiating the relative instability of this subset and its propensity to evolve into Th1 progeny. In contrast, Th1 cells display only repressive H3K27me3 methylation status over gene loci encoding for rorc and Il17, rendering them much more stable. iTregs are another relatively unstable subset that can acquire Th1 or Th17 properties, based on the poised bivalent status of type 1– or type 17–associated genes.

Close modal

Deeper insight into the evolution of Th17 responses in vivo was gained by Hirota et al175  using a reporter system permanently labeling Th cells that express the il17a gene, thus allowing for fate mapping of all “ex-Th17” cells in vivo by yellow fluorescent protein. In vaccination-induced EAE almost all myelin-specific CD4+ T cells infiltrating CNS were yellow fluorescent proteinhi and thus of Th17 origin, although they ceased secretion of IL-17A and switched to production of IFN-γ or other cytokines. This in vivo conversion was critically dependent on IL-23. In contrast, in acute cutaneous candidiasis, responding Th17 cells remained firmly committed to IL-17 production, possibly because of low local levels of IL-23. Thus, in vivo–induced murine Th17 cells display context-dependent stability or flexibility, just like Th17 cells generated in vitro. If the same phenomenon is true in the case of human Th17 cells remains to be determined; however, T-cell receptor clonotypes present in the Th17 cell compartment and Th1 compartment found in joint fluid recovered from patients with juvenile rheumatoid arthritis were remarkably similar, suggesting in vivo conversion.176  In another report, Th17 cells specific for the cancer testis antigen MAGE-A3 isolated from a patient with lung cancer were readily converted into IFN-γ–secreting Th1-like effectors.177 

Overall, the Th17 cells generated in vitro or in vivo can maintain the pure type 17 phenotype or can acquire certain type 1 characteristics on secondary stimulation. This plasticity, while significant, is clearly restrained by the epigenetic status of the genes encoding for the master transcriptional regulators and cytokines. As a result, the observed plasticity is asymmetrical because committed Th1 cells cannot easily acquire Th17 features. Thus, the plasticity of Th cell subsets is constrained and in this regard is similar to other self-renewing tissues with stem cell functionality as postulated by Hal Waddington almost 60 years ago; differentiation is strictly controlled in its directionality, and once differentiated, cells rarely if ever revert to the plastic progenitor state, in a way analogous to biochemical or physical process of entropy.178 

Development of long-term memory is one of the key features of adaptive immunity and is crucial for the ability to mount effective protection on the subsequent antigenic encounter. The formation of CD4+ T-cell memory is complicated by the existence of multiple polarized Th subsets, which can evolve and experience plasticity. It is thought that CD4-mediated memory responses might be less robust in contrast to more stable persistence of CD8+ memory T cells.179 

T-cell memory has been extensively studied in the CD8+ compartment using phenotypic markers wherein naïve CD44loCD62Lhi CD8+ T cells differentiate on stimulation into CD62LhiCD44hi central memory (TCM) cells or CD62LloCD44hi effector memory (TEM) cells. Recently a novel, less-differentiated, and very early memory cell subset termed memory stem cells (TSCM) has been described in the setting of GVHD and adoptive immunotherapy. TSCM displays stem cell–like features such as longevity, a capacity for self-renewal, and the ability to give rise to more differentiated (TCM and TEM) memory subsets.180,181  These cells display a naïve-like phenotype (CD62Lhi, CCR7hi, and CD44lo) but can be identified by the presence of some activation markers (Sca1 in mice and CD95 in humans).181 

TSCM also shares certain molecular traits common to other self-renewing populations including the activity of the Wnt/β-catenin pathway that contributes to the maintenance of normal and transformed stem cells and the development of thymocytes.182  Inhibition of GSK3β during antigenic stimulation of naïve CD8+ T cells stabilizes β-catenin and enhances TSCM formation.181  Stabilized β-catenin induces Tcf7 gene expression that encodes T-cell factor 1 (Tcf1). Tcf1 participates in maintenance of memory T cells and Tregs in vivo. Its forced overexpression prevents secretion of type 1 and type 17 effector molecules (IFN-γ and IL-17A, respectively) and promotes Th2 differentiation.183,184  CD8+ TEM cells are highly cytotoxic in vitro but display very poor persistence and antitumor effect in vivo; thus, they behave in a senescent manner.179,185,186  TCM and even more dramatically TSCM cells that do not achieve full terminal differentiation in vitro are far more effective in eradicating the tumor in vivo, and this is associated with superior persistence.181,185  Therefore, therapeutic activity and self-renewal potential can be used as surrogate measurements of the maturational stage of the particular effector population.179,185 

When judged by some common phenotypic markers, Th17 cells appear to resemble terminally differentiated TEM cells, with low expression of CD62L, CD45RA, and CD27, a costimulatory molecule important for survival of T cells that is down-regulated with advanced maturational stage.170  In contrast, Th1 cells maintain a high expression of CD27, and on in vitro polarization, they retain a higher frequency of CD62Lhi cells. Because of the terminally differentiated phenotype (CD27lo), Th17 cells were described as short lived and unable to form long-term memory.74  However, type 17 responses can be clearly protective against rechallenge in immunization models.73  This assertion was also at odds with observations from adoptive cell transfer experiments, where even highly purified Th17 cells can survive, persist, and retain functionality, as evidenced by protracted tissue damage in some models of autoimmunity or GVHD.136,187  The persistence of Th17 cells is robustly observed in the TRP-1 model of cancer immunotherapy, where Th17 cells are highly effective in eradicating the tumor, whereas Th1 cells are less efficient.166,170  Furthermore, TRP-1 Th17 cells purified for the lowest expression of CD27 showed the highest in vivo activity and survival. This finding is consistent with a recent report that CD27 signaling reduced the incidence and severity of disease in an EAE model.188  Therefore, we hypothesized that despite certain phenotypic features of advanced maturation, considerable multipotency and plasticity demonstrated by Th17 cells indicated that these cells are not terminally differentiated, at least when induced in vitro. Despite their phenotype, Th1 cells might represent a more differentiated subset with less capacity to expand, persist, and eliminate tumors in vivo (Figure 3).

Figure 3

Relationship between Th1 and Th17 polarization and maturational stage of Th memory cells. The initial polarization of naïve CD4+ T cells into the Th1 or Th17 subset not only induces a canonical set of transcription factors and cytokines but also affects CD4+ T-cell plasticity and fate. Th17 cells are relatively more plastic and less terminally differentiated than their Th1 cell counterparts. This is reflected by the higher ability of Th17 cells to self-renew, generate a distinctive highly functional Th1-like Th17 progeny, and form long-term memory following the secondary antigen experience. Th1 cells rapidly acquire a senescent phenotype and molecular signature, are less functional, and display less ability for self-renewal and memory formation. The lower panel demonstrates a proposed relationship between polarized CD4+ Th cells and a model of linear CD8+ T-cell memory formation. Upon antigen stimulation CD8+, T cells progress from naïve via self-renewing early memory stem cells (TSCM), to central memory (TCM and TEM) cells, and to the senescent terminally differentiated cells with no self-renewal potential. In vivo antitumor efficacy of adoptively transferred cells used for immunotherapy of cancer inversely correlates with the maturational stage of T cells.

Figure 3

Relationship between Th1 and Th17 polarization and maturational stage of Th memory cells. The initial polarization of naïve CD4+ T cells into the Th1 or Th17 subset not only induces a canonical set of transcription factors and cytokines but also affects CD4+ T-cell plasticity and fate. Th17 cells are relatively more plastic and less terminally differentiated than their Th1 cell counterparts. This is reflected by the higher ability of Th17 cells to self-renew, generate a distinctive highly functional Th1-like Th17 progeny, and form long-term memory following the secondary antigen experience. Th1 cells rapidly acquire a senescent phenotype and molecular signature, are less functional, and display less ability for self-renewal and memory formation. The lower panel demonstrates a proposed relationship between polarized CD4+ Th cells and a model of linear CD8+ T-cell memory formation. Upon antigen stimulation CD8+, T cells progress from naïve via self-renewing early memory stem cells (TSCM), to central memory (TCM and TEM) cells, and to the senescent terminally differentiated cells with no self-renewal potential. In vivo antitumor efficacy of adoptively transferred cells used for immunotherapy of cancer inversely correlates with the maturational stage of T cells.

Close modal

The developmental program associated with Th17 plasticity has been elucidated using global gene expression profiles of transferred Th1 and Th17 cells. Indeed, molecular signatures of persisting Th1 and Th17 cells underwent rapid convergence, and Th17 cells readily down-regulated expression of type 17–defining cytokines (IL-17A, IL-17F, IL-21, and IL-22) and up-regulated transcripts associated with type 1 polarization (tbx21 and Ifng). Recent reports describe a role of T-bet expression gradient as a regulator of CD4+ T-cell memory formation, with highly Th1-polarized T-bethi cells displaying end-effector features and a short lifespan, whereas Th cells with a lower expression of T-bet formed a long-lived stable TCM population.189,190  This to some degree mimicked our observations, as long-lived ex-Th17 cells acquired T-bet expression, albeit at levels lower than their Th1 counterparts.

Multiple other transcripts remained differentially expressed. This difference between persisting Th1- and Th17-derived cells was analyzed by gene set enrichment analysis that revealed the core molecular program of long-lived Th17 cells resembled that of CD8+ T cells at an early stage of differentiation, even after they converted to a Th1-like subset in vivo.191  Reciprocally, the Th1 population acquired a global signature of terminal differentiation and senescence, with high expression of molecular and phenotypic markers of an advanced maturational stage, including cytotoxic effector molecules (granzymes, perforin), klrg1 and other killer-like lectin receptors, and prdm1 (a gene encoding a molecular marker of terminal Th1 differentiation, Blimp1).170  Furthermore, Th17 cells overexpressed tcf7 and some other downstream molecules of the Wnt/β-catenin signaling pathway. In CD8+ T cells, the level of tcf7 expression closely correlates with the maturational status of T cells: it is highly expressed in naïve T cells and is gradually lost following antigenic stimulation and acquisition of the TCM phenotype.180  TEM cells have the lowest tcf7 expression. Thus, tcf7 expression might be used as a possible surrogate marker of maturational state and self-renewal capacity. Strikingly, Th17 polarization in vitro induced tcf7 expression higher than in a naïve starting population. This was associated with a massive accumulation of stable β-catenin in Th17 cells, therefore mimicking a molecular signature found in naïve and TSCM cells induced by activation of Wnt signaling or pharmacological inhibition of GSK3β,179  but in type 17 cells, it most likely resulted from downstream effects of IL-6 or IL-1 mediated by phosphatidylinositol 3-kinase/Akt activity. As indicated by Luckey and Weaver,192  many questions regarding the CD4+ memory and self-renewal remain unanswered, but functionally, Th17 cells displayed traits similar to TSCM, as they were long lived, demonstrated plasticity, and gave rise to more differentiated Th1-like progeny, but also retained the ability to self-renew as IL-17A producers.

The findings from the murine model were substantiated by the description of stem cell–like behavior and a self-renewal molecular program in human Th17 cells that express high levels of HIF1α and antiapoptotic molecule BCL2.193  Intriguingly, the Stat3 signaling pathway that induces Th17 differentiation is critical for survival of long-lived memory T cells but also plays an important role in maintenance of normal and malignant self-renewing populations with attributes of stemness.

The discovery of Th17 cells has been followed by the realization that Th effectors can produce various other cytokines alone or in combination in patterns not fitting the preconceived definitions of Th1/Th2 or Th17 subsets. These findings have led to the description of additional Th cell lineages, including Th22, Th9, and, most notably, follicular T helper cells (Tfh). The plasticity, stability, and potential for memory formation by each of these subsets remain yet to be elucidated.

Tfh cells express CXCR5 and predominantly traffic to lymph nodes where they provide help to B cells and produce IL-21 (just like Th17 cells).194  Tfh cell induction is governed by Stat3 and the expression of Bcl6, a molecule important for the maintenance of CD8+ memory. Bcl6 is a prosurvival transcriptional repressor that antagonizes Blimp1, a molecule that drives terminal differentiation of T cells; thus, the balance between these two factors might have maturational implications.195  Likewise, Tfh cells have a high capacity to form long-term memory.196  The relationship of Tfh with other Th cell subsets such as Th1, Th2, and Th17 cells remains unclear, and cells have been investigated mostly in the context of infection in vivo and humoral immunity.196,197  When observing the in vivo responses to infection, it is difficult to ascertain the sequence of maturational events with long-lived cells emerging on contraction of the massively expanded effectors induced by the presence of pathogen. Thus, it is conceivable that Tfh cells might represent a self-renewing, plastic, and non–terminally differentiated early memory subset.

Th22 cells producing only IL-22 but neither IFN-γ nor IL-17A have been identified in humans.198  They are induced in the presence of TNF-α and IL-6 and require ligation of Ahr. Phenotypically, they can be identified by expression of CCR10 and the presence of some typical markers of human Th17 cells (CCR4, CCR6).200  Th22 cells via IL-22 influence the function of mesenchymal and epithelial cells and have been implicated in the dermatopathology of psoriasis and atopic dermatitis.199-201 

Th9 cells are induced in the presence of IL-4 and TGF-β1, but their master transcription factor has not been identified, and the functional roles of Th9 cells and IL-9 are ill defined. Recent findings demonstrated that IL-9 is expressed only transiently and is predominantly produced in vivo not by Th9 cells but by a novel subset of innate lymphoid cells termed ILC2.202  Willhelm et al203  theorized that IL-9 might have a regulatory and prosurvival function for many lymphoid and myeloid cells. A recent report described the development of endogenous antitumor Th9 responses in Rorγt-deficient animals and proposed a protective role for IL-9 in tumor immunity.162,204 

Recent years have brought a new level of appreciation of the complexities involved in the function of effector CD4+ Th cells. The discovery of Th17 cells has filled the gaps and deficiencies existing in the previous simplistic Th1/Th2 paradigm and has revolutionized our understanding of immune responses against certain pathogens and pathophysiology associated with development and progression of some autoimmune diseases or genetic defects of host defense, progression of HIV and development of AIDS, solid organ transplantation rejection, and GVHD. In addition, Th17 cells have been described as mediators of carcinogenesis, by facilitating early progression of solid and hematologic malignancy through the direct effects of inflammatory cytokines on some cancer cells, but more frequently by impairing immune surveillance or promoting neovascularization and supporting stroma.

The investigation of Th17 cells led to a new appreciation of the flexibility and plasticity of Th cell–mediated immunity. In contrast to the previously postulated dichotomous Th1/Th2 division of labor, it is now clear that naïve Th cells can achieve a multiplicity of polarization states and experience remarkably different cell fates that are highly dependent on the context of their encounter with antigen. Th17 cells can provide an initial inflammatory response via their canonical type 17 cytokines and recruit various myeloid populations to the site of the immune response before transitioning into type 1–like cells that can also target the intracellular pathogens. This developmental plasticity has significant implications for the rational design of new treatments of the type 17–linked autoimmune syndromes such as multiple sclerosis or GVHD. It can also be harnessed for the design of novel adoptive cell transfer therapies, as evidenced by the antitumor efficacy of Th17-polarized tumor-specific cells, where superior engraftment and persistence of highly active transferred effectors are desired.

The initial differentiation toward the Th17 subset prevents the effector cells from a premature acquisition of terminal differentiation that is associated with highly polarized Th1 cells. Instead, these cells acquire a molecular program reminiscent of the recently described TSCM subset that is functionally characterized by self-renewal, persistence, and the ability to efficiently form memory and at the same time generate more differentiated progeny critical for in vivo effector function. Thus, polarization of Th cells conveys not only characteristic phenotypic and cytokine profiles but also has a maturational aspect in the spectrum from early self-renewing subsets to those that are senescent.

This work was supported by the Center for Cancer Research, National Cancer Institute–National Institutes of Health Intramural Research Program, the National Heart, Lung, and Blood Institute Intramural Program, and the National Institutes of Health Center for Regenerative Medicine.

Contribution: P.M. and N.P.R. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Pawel Muranski, Hematology Branch, NHLBI/NIH, 10 Center Dr, 3E-5288, Bethesda, MD 20892; e-mail: muranskp@mail.nih.gov.

1
Zhu
 
J
Paul
 
WE
 
CD4 T cells: fates, functions, and faults. Blood. 2008;112(5):1557-1569.
2
Sun
 
JC
Bevan
 
MJ
 
Defective CD8 T cell memory following acute infection without CD4 T cell help. Science (New York, N.Y.) 2003;300(5617):339-342.
3
Bevan
 
MJ
Helping the CD8(+) T-cell response.
Nat Rev Immunol
2004
, vol. 
4
 
8
(pg. 
595
-
602
)
4
Swain
 
SL
McKinstry
 
KK
Strutt
 
TM
Expanding roles for CD4⁺ T cells in immunity to viruses.
Nat Rev Immunol
2012
, vol. 
12
 
2
(pg. 
136
-
148
)
5
Muranski
 
P
Restifo
 
NP
Adoptive immunotherapy of cancer using CD4(+) T cells.
Curr Opin Immunol
2009
, vol. 
21
 
2
(pg. 
200
-
208
)
6
Mosmann
 
TR
Cherwinski
 
H
Bond
 
MW
, et al. 
Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins.
J Immunol
1986
, vol. 
136
 
7
(pg. 
2348
-
2357
)
7
Mosmann
 
TR
Coffman
 
RL
TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties.
Annu Rev Immunol
1989
, vol. 
7
 (pg. 
145
-
173
)
8
Heinzel
 
FP
Sadick
 
MD
Holaday
 
BJ
, et al. 
Reciprocal expression of interferon gamma or interleukin 4 during the resolution or progression of murine leishmaniasis. Evidence for expansion of distinct helper T cell subsets.
J Exp Med
1989
, vol. 
169
 
1
(pg. 
59
-
72
)
9
Romagnani
 
S
The Th1/Th2 paradigm.
Immunol Today
1997
, vol. 
18
 
6
(pg. 
263
-
266
)
10
Parronchi
 
P
Romagnani
 
P
Annunziato
 
F
, et al. 
Type 1 T-helper cell predominance and interleukin-12 expression in the gut of patients with Crohn’s disease.
Am J Pathol
1997
, vol. 
150
 
3
(pg. 
823
-
832
)
11
Leonard
 
JP
Waldburger
 
KE
Goldman
 
SJ
Prevention of experimental autoimmune encephalomyelitis by antibodies against interleukin 12.
J Exp Med
1995
, vol. 
181
 
1
(pg. 
381
-
386
)
12
Afkarian
 
M
Sedy
 
JR
Yang
 
J
, et al. 
T-bet is a STAT1-induced regulator of IL-12R expression in naïve CD4+ T cells.
Nat Immunol
2002
, vol. 
3
 
6
(pg. 
549
-
557
)
13
Szabo
 
SJ
Kim
 
ST
Costa
 
GL
, et al. 
A novel transcription factor, T-bet, directs Th1 lineage commitment.
Cell
2000
, vol. 
100
 
6
(pg. 
655
-
669
)
14
Oestreich
 
KJ
Weinmann
 
AS
Transcriptional mechanisms that regulate T helper 1 cell differentiation.
Curr Opin Immunol
2012
, vol. 
24
 
2
(pg. 
191
-
195
)
15
Kaplan
 
MH
Schindler
 
U
Smiley
 
ST
, et al. 
Stat6 is required for mediating responses to IL-4 and for development of Th2 cells.
Immunity
1996
, vol. 
4
 
3
(pg. 
313
-
319
)
16
Zheng
 
WP
Flavell
 
RA
The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells.
Cell
1997
, vol. 
89
 
4
(pg. 
587
-
596
)
17
Shimoda
 
K
van Deursen
 
J
Sangster
 
MY
, et al. 
Lack of IL-4-induced Th2 response and IgE class switching in mice with disrupted Stat6 gene.
Nature
1996
, vol. 
380
 
6575
(pg. 
630
-
633
)
18
Robinson
 
DS
Hamid
 
Q
Ying
 
S
, et al. 
Predominant TH2-like bronchoalveolar T-lymphocyte population in atopic asthma.
N Engl J Med
1992
, vol. 
326
 
5
(pg. 
298
-
304
)
19
Lambrecht
 
BN
De Veerman
 
M
Coyle
 
AJ
, et al. 
Myeloid dendritic cells induce Th2 responses to inhaled antigen, leading to eosinophilic airway inflammation.
J Clin Invest
2000
, vol. 
106
 
4
(pg. 
551
-
559
)
20
Pearce
 
EJ
Caspar
 
P
Grzych
 
JM
, et al. 
Downregulation of Th1 cytokine production accompanies induction of Th2 responses by a parasitic helminth, Schistosoma mansoni.
J Exp Med
1991
, vol. 
173
 
1
(pg. 
159
-
166
)
21
Szabo
 
SJ
Jacobson
 
NG
Dighe
 
AS
, et al. 
Developmental commitment to the Th2 lineage by extinction of IL-12 signaling.
Immunity
1995
, vol. 
2
 
6
(pg. 
665
-
675
)
22
Ouyang
 
W
Ranganath
 
SH
Weindel
 
K
, et al. 
Inhibition of Th1 development mediated by GATA-3 through an IL-4-independent mechanism.
Immunity
1998
, vol. 
9
 
5
(pg. 
745
-
755
)
23
Usui
 
T
Preiss
 
JC
Kanno
 
Y
, et al. 
T-bet regulates Th1 responses through essential effects on GATA-3 function rather than on IFNG gene acetylation and transcription.
J Exp Med
2006
, vol. 
203
 
3
(pg. 
755
-
766
)
24
Hwang
 
ES
Szabo
 
SJ
Schwartzberg
 
PL
Glimcher
 
LH
 
T helper cell fate specified by kinase-mediated interaction of T-bet with GATA-3. Science (New York, N.Y.) 2005;307(5708):430-433.
25
Oestreich
 
KJ
Weinmann
 
AS
T-bet employs diverse regulatory mechanisms to repress transcription.
Trends Immunol
2012
, vol. 
33
 
2
(pg. 
78
-
83
)
26
Hori
 
S
Nomura
 
T
Sakaguchi
 
S
 
Control of regulatory T cell development by the transcription factor Foxp3. Science (New York, N.Y.) 2003; 299(5609):1057-1061.
27
Cua
 
DJ
Sherlock
 
J
Chen
 
Y
, et al. 
Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain.
Nature
2003
, vol. 
421
 
6924
(pg. 
744
-
748
)
28
Murphy
 
CA
Langrish
 
CL
Chen
 
Y
, et al. 
Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation.
J Exp Med
2003
, vol. 
198
 
12
(pg. 
1951
-
1957
)
29
Harrington
 
LE
Hatton
 
RD
Mangan
 
PR
, et al. 
Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages.
Nat Immunol
2005
, vol. 
6
 
11
(pg. 
1123
-
1132
)
30
Park
 
H
Li
 
ZX
Yang
 
XO
, et al. 
A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17.
Nat Immunol
2005
, vol. 
6
 
11
(pg. 
1133
-
1141
)
31
Oppmann
 
B
Lesley
 
R
Blom
 
B
, et al. 
Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12.
Immunity
2000
, vol. 
13
 
5
(pg. 
715
-
725
)
32
Langrish
 
CL
Chen
 
Y
Blumenschein
 
WM
, et al. 
IL-23 drives a pathogenic T cell population that induces autoimmune inflammation.
J Exp Med
2005
, vol. 
201
 
2
(pg. 
233
-
240
)
33
Hirahara
 
K
Ghoreschi
 
K
Laurence
 
A
, et al. 
Signal transduction pathways and transcriptional regulation in Th17 cell differentiation.
Cytokine Growth Factor Rev
2010
, vol. 
21
 
6
(pg. 
425
-
434
)
34
Ivanov
 
II
McKenzie
 
BS
Zhou
 
L
, et al. 
The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells.
Cell
2006
, vol. 
126
 
6
(pg. 
1121
-
1133
)
35
Yang
 
XO
Pappu
 
BP
Nurieva
 
R
, et al. 
T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma.
Immunity
2008
, vol. 
28
 
1
(pg. 
29
-
39
)
36
Durant
 
L
Watford
 
WT
Ramos
 
HL
, et al. 
Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis.
Immunity
2010
, vol. 
32
 
5
(pg. 
605
-
615
)
37
Schraml
 
BU
Hildner
 
K
Ise
 
W
, et al. 
The AP-1 transcription factor Batf controls T(H)17 differentiation.
Nature
2009
, vol. 
460
 
7253
(pg. 
405
-
409
)
38
Shi
 
LZ
Wang
 
R
Huang
 
G
, et al. 
 
HIF1α–dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med. 2011;208(7):1367-1376.
39
Brüstle
 
A
Heink
 
S
Huber
 
M
, et al. 
The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4.
Nat Immunol
2007
, vol. 
8
 
9
(pg. 
958
-
966
)
40
Zhang
 
F
Meng
 
G
Strober
 
W
Interactions among the transcription factors Runx1, RORgammat and Foxp3 regulate the differentiation of interleukin 17-producing T cells.
Nat Immunol
2008
, vol. 
9
 
11
(pg. 
1297
-
1306
)
41
Ciofani
 
M
Madar
 
A
Galan
 
C
, et al. 
A validated regulatory network for Th17 cell specification.
Cell
2012
, vol. 
151
 
2
(pg. 
289
-
303
)
42
Yang
 
XP
Ghoreschi
 
K
Steward-Tharp
 
SM
, et al. 
Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5.
Nat Immunol
2011
, vol. 
12
 
3
(pg. 
247
-
254
)
43
Laurence
 
A
Tato
 
CM
Davidson
 
TS
, et al. 
Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation.
Immunity
2007
, vol. 
26
 
3
(pg. 
371
-
381
)
44
Lazarevic
 
V
Chen
 
X
Shim
 
JH
, et al. 
T-bet represses T(H)17 differentiation by preventing Runx1-mediated activation of the gene encoding RORγt.
Nat Immunol
2011
, vol. 
12
 
1
(pg. 
96
-
104
)
45
Diveu
 
C
McGeachy
 
MJ
Boniface
 
K
, et al. 
 
IL-27 blocks RORc expression to inhibit lineage commitment of Th17 cells. J Immunol. 2009;182(9):5748-5756.
46
Mathur
 
AN
Chang
 
HC
Zisoulis
 
DG
, et al. 
Stat3 and Stat4 direct development of IL-17-secreting Th cells.
J Immunol
2007
, vol. 
178
 
8
(pg. 
4901
-
4907
)
47
Yang
 
XO
Panopoulos
 
AD
Nurieva
 
R
, et al. 
STAT3 regulates cytokine-mediated generation of inflammatory helper T cells.
J Biol Chem
2007
, vol. 
282
 
13
(pg. 
9358
-
9363
)
48
McGeachy
 
MJ
Bak-Jensen
 
KS
Chen
 
Y
, et al. 
TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology.
Nat Immunol
2007
, vol. 
8
 
12
(pg. 
1390
-
1397
)
49
McGeachy
 
MJ
Chen
 
Y
Tato
 
CM
, et al. 
The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo.
Nat Immunol
2009
, vol. 
10
 
3
(pg. 
314
-
324
)
50
Chen
 
Z
Laurence
 
A
O’Shea
 
JJ
Signal transduction pathways and transcriptional regulation in the control of Th17 differentiation.
Semin Immunol
2007
, vol. 
19
 
6
(pg. 
400
-
408
)
51
Zhou
 
L
Ivanov
 
II
Spolski
 
R
, et al. 
IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways.
Nat Immunol
2007
, vol. 
8
 
9
(pg. 
967
-
974
)
52
Korn
 
T
Bettelli
 
E
Gao
 
W
, et al. 
IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells.
Nature
2007
, vol. 
448
 
7152
(pg. 
484
-
487
)
53
Ghoreschi
 
K
Laurence
 
A
Yang
 
XP
, et al. 
Generation of pathogenic T(H)17 cells in the absence of TGF-β signalling.
Nature
2010
, vol. 
467
 
7318
(pg. 
967
-
971
)
54
Dinarello
 
CA
 
Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009:27:519-550.
55
Gulen
 
MF
Bulek
 
K
Xiao
 
H
, et al. 
Inactivation of the enzyme GSK3α by the kinase IKKi promotes AKT-mTOR signaling pathway that mediates interleukin-1-induced Th17 cell maintenance.
Immunity
2012
, vol. 
37
 
5
(pg. 
800
-
812
)
56
Gulen
 
MF
Kang
 
Z
Bulek
 
K
, et al. 
The receptor SIGIRR suppresses Th17 cell proliferation via inhibition of the interleukin-1 receptor pathway and mTOR kinase activation.
Immunity
2010
, vol. 
32
 
1
(pg. 
54
-
66
)
57
Chung
 
Y
Chang
 
SH
Martinez
 
GJ
, et al. 
Critical regulation of early Th17 cell differentiation by interleukin-1 signaling.
Immunity
2009
, vol. 
30
 
4
(pg. 
576
-
587
)
58
Chen
 
Q
Yang
 
W
Gupta
 
S
, et al. 
IRF-4-binding protein inhibits interleukin-17 and interleukin-21 production by controlling the activity of IRF-4 transcription factor.
Immunity
2008
, vol. 
29
 
6
(pg. 
899
-
911
)
59
Oida
 
T
Weiner
 
HL
Depletion of TGF-β from fetal bovine serum.
J Immunol Methods
2010
, vol. 
362
 
1-2
(pg. 
195
-
198
)
60
Gutcher
 
I
Donkor
 
MK
Ma
 
Q
, et al. 
Autocrine transforming growth factor-β1 promotes in vivo Th17 cell differentiation.
Immunity
2011
, vol. 
34
 
3
(pg. 
396
-
408
)
61
Qin
 
H
Wang
 
L
Feng
 
T
, et al. 
TGF-beta promotes Th17 cell development through inhibition of SOCS3.
J Immunol
2009
, vol. 
183
 
1
(pg. 
97
-
105
)
62
Veldhoen
 
M
Hocking
 
RJ
Atkins
 
CJ
, et al. 
TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells.
Immunity
2006
, vol. 
24
 
2
(pg. 
179
-
189
)
63
Zhou
 
L
Lopes
 
JE
Chong
 
MM
, et al. 
TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function.
Nature
2008
, vol. 
453
 
7192
(pg. 
236
-
240
)
64
Valmori
 
D
Raffin
 
C
Raimbaud
 
I
, et al. 
Human RORγt+ TH17 cells preferentially differentiate from naive FOXP3+Treg in the presence of lineage-specific polarizing factors.
Proc Natl Acad Sci USA
2010
, vol. 
107
 
45
(pg. 
19402
-
19407
)
65
Mucida
 
D
Park
 
Y
Kim
 
G
, et al. 
 
Reciprocal T(H)17 and regulatory T cell differentiation mediated by retinoic acid. Science (New York, N.Y.) 2007;317(5835):256-260.
66
Takahashi
 
H
Kanno
 
T
Nakayamada
 
S
, et al. 
TGF-β and retinoic acid induce the microRNA miR-10a, which targets Bcl-6 and constrains the plasticity of helper T cells.
Nat Immunol
2012
, vol. 
13
 
6
(pg. 
587
-
595
)
67
Hall
 
JA
Cannons
 
JL
Grainger
 
JR
, et al. 
Essential role for retinoic acid in the promotion of CD4(+) T cell effector responses via retinoic acid receptor alpha.
Immunity
2011
, vol. 
34
 
3
(pg. 
435
-
447
)
68
Veldhoen
 
M
Hirota
 
K
Westendorf
 
AM
, et al. 
The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins.
Nature
2008
, vol. 
453
 
7191
(pg. 
106
-
109
)
69
Stockinger
 
B
Hirota
 
K
Duarte
 
J
, et al. 
External influences on the immune system via activation of the aryl hydrocarbon receptor.
Semin Immunol
2011
, vol. 
23
 
2
(pg. 
99
-
105
)
70
Wu
 
H-J
Ivanov
 
II
Darce
 
J
, et al. 
Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells.
Immunity
2010
, vol. 
32
 
6
(pg. 
815
-
827
)
71
Ivanov
 
II
Atarashi
 
K
Manel
 
N
, et al. 
Induction of intestinal Th17 cells by segmented filamentous bacteria.
Cell
2009
, vol. 
139
 
3
(pg. 
485
-
498
)
72
Kriegel
 
MA
Sefik
 
E
Hill
 
JA
, et al. 
 
Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice. Proc Natl Acad Sci U.S.A. 2011;108(28):11548-11553.
73
Chen
 
K
McAleer
 
JP
Lin
 
Y
, et al. 
Th17 cells mediate clade-specific, serotype-independent mucosal immunity.
Immunity
2011
, vol. 
35
 
6
(pg. 
997
-
1009
)
74
Pepper
 
M
Linehan
 
JL
Pagán
 
AJ
, et al. 
Different routes of bacterial infection induce long-lived TH1 memory cells and short-lived TH17 cells.
Nat Immunol
2010
, vol. 
11
 
1
(pg. 
83
-
89
)
75
Ouyang
 
WJ
Kolls
 
JK
Zheng
 
Y
The biological functions of T helper 17 cell effector cytokines in inflammation.
Immunity
2008
, vol. 
28
 
4
(pg. 
454
-
467
)
76
Khader
 
SA
Gaffen
 
SL
Kolls
 
JK
Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa.
Mucosal Immunol
2009
, vol. 
2
 
5
(pg. 
403
-
411
)
77
Korn
 
T
Bettelli
 
E
Oukka
 
M
, et al. 
IL-17 and Th17 Cells.
Annu Rev Immunol
2009
, vol. 
27
 
1
(pg. 
485
-
517
)
78
Gaffen
 
SL
Structure and signalling in the IL-17 receptor family.
Nat Rev Immunol
2009
, vol. 
9
 
8
(pg. 
556
-
567
)
79
Fossiez
 
F
Djossou
 
O
Chomarat
 
P
, et al. 
 
T cell interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines. J Exp Med. 1996;183(6):2593-2603.
80
Tan
 
W
Huang
 
W
Zhong
 
Q
, et al. 
IL-17 receptor knockout mice have enhanced myelotoxicity and impaired hemopoietic recovery following gamma irradiation.
J Immunol
2006
, vol. 
176
 
10
(pg. 
6186
-
6193
)
81
Kinugasa
 
T
Sakaguchi
 
T
Gu
 
XB
, et al. 
Claudins regulate the intestinal barrier in response to immune mediators.
Gastroenterology
2000
, vol. 
118
 
6
(pg. 
1001
-
1011
)
82
Rangel-Moreno
 
J
Carragher
 
DM
de la Luz Garcia-Hernandez
 
M
, et al. 
The development of inducible bronchus-associated lymphoid tissue depends on IL-17.
Nat Immunol
2011
, vol. 
12
 
7
(pg. 
639
-
646
)
83
Mangan
 
PR
Harrington
 
LE
O’Quinn
 
DB
, et al. 
Transforming growth factor-beta induces development of the T(H)17 lineage.
Nature
2006
, vol. 
441
 
7090
(pg. 
231
-
234
)
84
Khader
 
SA
Bell
 
GK
Pearl
 
JE
, et al. 
IL-23 and IL-17 in the establishment of protective pulmonary CD4+ T cell responses after vaccination and during Mycobacterium tuberculosis challenge.
Nat Immunol
2007
, vol. 
8
 
4
(pg. 
369
-
377
)
85
Lin
 
YY
Ritchea
 
S
Logar
 
A
, et al. 
Interleukin-17 is required for T helper 1 cell immunity and host resistance to the intracellular pathogen Francisella tularensis.
Immunity
2009
, vol. 
31
 
5
(pg. 
799
-
810
)
86
LeibundGut-Landmann
 
S
Gross
 
O
Robinson
 
MJ
, et al. 
Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17.
Nat Immunol
2007
, vol. 
8
 
6
(pg. 
630
-
638
)
87
Wuthrich
 
M
Deepe
 
GS
Klein
 
B
Paul
 
WE
Adaptive immunity to fungi.
Annual Review of Immunology
, vol. 
Vol 30
 
Palo Alto, CA
Annual Reviews; 2012:
(pg. 
115
-
148
)
88
Liang
 
SC
Tan
 
X-Y
Luxenberg
 
DP
, et al. 
 
Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med. 2006;203(10):2271-2279.
89
Aujla
 
SJ
Chan
 
YR
Zheng
 
MQ
, et al. 
IL-22 mediates mucosal host defense against Gram-negative bacterial pneumonia.
Nat Med
2008
, vol. 
14
 
3
(pg. 
275
-
281
)
90
Spolski
 
R
Leonard
 
WJ
 
Interleukin-21: basic biology and implications for cancer and autoimmunity. Annu Rev Immunol. 2008;26:57-79.
91
Milner
 
JD
Brenchley
 
JM
Laurence
 
A
, et al. 
Impaired T(H)17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome.
Nature
2008
, vol. 
452
 
7188
(pg. 
773
-
776
)
92
Milner
 
JD
Sandler
 
NG
Douek
 
DC
Th17 cells, Job’s syndrome and HIV: opportunities for bacterial and fungal infections.
Curr Opin HIV AIDS
2010
, vol. 
5
 
2
(pg. 
179
-
183
)
93
Siegel
 
AM
Heimall
 
J
Freeman
 
AF
, et al. 
A critical role for STAT3 transcription factor signaling in the development and maintenance of human T cell memory.
Immunity
2011
, vol. 
35
 
5
(pg. 
806
-
818
)
94
Brenchley
 
JM
Paiardini
 
M
Knox
 
KS
, et al. 
Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections.
Blood
2008
, vol. 
112
 
7
(pg. 
2826
-
2835
)
95
Becher
 
B
Durell
 
BG
Noelle
 
RJ
Experimental autoimmune encephalitis and inflammation in the absence of interleukin-12.
J Clin Invest
2002
, vol. 
110
 
4
(pg. 
493
-
497
)
96
Cargill
 
M
Schrodi
 
SJ
Chang
 
M
, et al. 
A large-scale genetic association study confirms IL12B and leads to the identification of IL23R as psoriasis-risk genes.
Am J Hum Genet
2007
, vol. 
80
 
2
(pg. 
273
-
290
)
97
Takaku
 
T
Calado
 
RT
Kajigaya
 
S
, et al. 
Interleukin-23 receptor (IL-23R) gene polymorphisms in acquired aplastic anemia.
Ann Hematol
2009
, vol. 
88
 
7
(pg. 
653
-
657
)
98
Zwiers
 
A
Kraal
 
L
van de Pouw Kraan
 
TCTM
Wurdinger
 
T
Bouma
 
G
Kraal
 
G
 
Cutting edge: A variant of the IL-23R gene associated with inflammatory bowel disease induces loss of microRNA regulation and enhanced protein production. J Immunol. 2012;188(4):1573-1577.
99
Spolski
 
R
Wang
 
L
Wan
 
CK
, et al. 
IL-21 promotes the pathologic immune response to pneumovirus infection.
J Immunol
2012
, vol. 
188
 
4
(pg. 
1924
-
1932
)
100
Huppert
 
J
Closhen
 
D
Croxford
 
A
, et al. 
Cellular mechanisms of IL-17-induced blood-brain barrier disruption.
FASEB J
2010
, vol. 
24
 
4
(pg. 
1023
-
1034
)
101
Acosta-Rodriguez
 
EV
Rivino
 
L
Geginat
 
J
, et al. 
Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells.
Nat Immunol
2007
, vol. 
8
 
6
(pg. 
639
-
646
)
102
Nowak
 
EC
Weaver
 
CT
Turner
 
H
, et al. 
IL-9 as a mediator of Th17-driven inflammatory disease.
J Exp Med
2009
, vol. 
206
 
8
(pg. 
1653
-
1660
)
103
El-Behi
 
M
Ciric
 
B
Dai
 
H
, et al. 
The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF.
Nat Immunol
2011
, vol. 
12
 
6
(pg. 
568
-
575
)
104
Codarri
 
L
Gyülvészi
 
G
Tosevski
 
V
, et al. 
RORγt drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation.
Nat Immunol
2011
, vol. 
12
 
6
(pg. 
560
-
567
)
105
Luger
 
D
Silver
 
PB
Tang
 
J
, et al. 
Either a Th17 or a Th1 effector response can drive autoimmunity: conditions of disease induction affect dominant effector category.
J Exp Med
2008
, vol. 
205
 
4
(pg. 
799
-
810
)
106
Kroenke
 
MA
Carlson
 
TJ
Andjelkovic
 
AV
, et al. 
IL-12- and IL-23-modulated T cells induce distinct types of EAE based on histology, CNS chemokine profile, and response to cytokine inhibition.
J Exp Med
2008
, vol. 
205
 
7
(pg. 
1535
-
1541
)
107
Palmer
 
MT
Weaver
 
CT
Autoimmunity: increasing suspects in the CD4+ T cell lineup.
Nat Immunol
2010
, vol. 
11
 
1
(pg. 
36
-
40
)
108
Yang
 
Y
Weiner
 
J
Liu
 
Y
, et al. 
T-bet is essential for encephalitogenicity of both Th1 and Th17 cells.
J Exp Med
2009
, vol. 
206
 
7
(pg. 
1549
-
1564
)
109
Lee
 
Y
Awasthi
 
A
Yosef
 
N
, et al. 
Induction and molecular signature of pathogenic TH17 cells.
Nat Immunol
2012
, vol. 
13
 
10
(pg. 
991
-
999
)
110
Segal
 
BM
Constantinescu
 
CS
Raychaudhuri
 
A
, et al. 
Ustekinumab MS Investigators
Repeated subcutaneous injections of IL12/23 p40 neutralising antibody, ustekinumab, in patients with relapsing-remitting multiple sclerosis: a phase II, double-blind, placebo-controlled, randomised, dose-ranging study.
Lancet Neurol
2008
, vol. 
7
 
9
(pg. 
796
-
804
)
111
Nograles
 
KE
Zaba
 
LC
Guttman-Yassky
 
E
, et al. 
Th17 cytokines interleukin (IL)-17 and IL-22 modulate distinct inflammatory and keratinocyte-response pathways.
Br J Dermatol
2008
, vol. 
159
 
5
(pg. 
1092
-
1102
)
112
Di Cesare
 
A
Di Meglio
 
P
Nestle
 
FO
The IL-23/Th17 axis in the immunopathogenesis of psoriasis.
J Invest Dermatol
2009
, vol. 
129
 
6
(pg. 
1339
-
1350
)
113
Ma
 
HL
Liang
 
S
Li
 
J
, et al. 
IL-22 is required for Th17 cell-mediated pathology in a mouse model of psoriasis-like skin inflammation.
J Clin Invest
2008
, vol. 
118
 
2
(pg. 
597
-
607
)
114
Leonardi
 
CL
Kimball
 
AB
Papp
 
KA
, 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
, vol. 
371
 
9625
(pg. 
1665
-
1674
)
115
Benson
 
JM
Sachs
 
CW
Treacy
 
G
, et al. 
Therapeutic targeting of the IL-12/23 pathways: generation and characterization of ustekinumab.
Nat Biotechnol
2011
, vol. 
29
 
7
(pg. 
615
-
624
)
116
Papp
 
KA
Leonardi
 
C
Menter
 
A
, et al. 
Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis.
N Engl J Med
2012
, vol. 
366
 
13
(pg. 
1181
-
1189
)
117
Ahern
 
PP
Schiering
 
C
Buonocore
 
S
, et al. 
Interleukin-23 drives intestinal inflammation through direct activity on T cells.
Immunity
2010
, vol. 
33
 
2
(pg. 
279
-
288
)
118
Monteleone
 
I
Sarra
 
M
Pallone
 
F
, et al. 
Th17-related cytokines in inflammatory bowel diseases: friends or foes?
Curr Mol Med
2012
, vol. 
12
 
5
(pg. 
592
-
597
)
119
O’Connor
 
W
Kamanaka
 
M
Booth
 
CJ
, et al. 
A protective function for interleukin 17A in T cell-mediated intestinal inflammation.
Nat Immunol
2009
, vol. 
10
 
6
(pg. 
603
-
609
)
120
Sugimoto
 
K
Ogawa
 
A
Mizoguchi
 
E
, et al. 
IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis.
J Clin Invest
2008
, vol. 
118
 
2
(pg. 
534
-
544
)
121
Zenewicz
 
LA
Yancopoulos
 
GD
Valenzuela
 
DM
, et al. 
Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease.
Immunity
2008
, vol. 
29
 
6
(pg. 
947
-
957
)
122
Fantini
 
MC
Rizzo
 
A
Fina
 
D
, et al. 
IL-21 regulates experimental colitis by modulating the balance between Treg and Th17 cells.
Eur J Immunol
2007
, vol. 
37
 
11
(pg. 
3155
-
3163
)
123
Peluso
 
I
Fantini
 
MC
Fina
 
D
, et al. 
IL-21 counteracts the regulatory T cell-mediated suppression of human CD4+ T lymphocytes.
J Immunol
2007
, vol. 
178
 
2
(pg. 
732
-
739
)
124
Annunziato
 
F
Cosmi
 
L
Santarlasci
 
V
, et al. 
 
Phenotypic and functional features of human Th17 cells. J Exp Med. 2007;204(8)1849-1861.
125
Cosmi
 
L
De Palma
 
R
Santarlasci
 
V
, et al. 
Human interleukin 17-producing cells originate from a CD161+CD4+ T cell precursor.
J Exp Med
2008
, vol. 
205
 
8
(pg. 
1903
-
1916
)
126
Hueber
 
W
Sands
 
BE
Lewitzky
 
S
, 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
, vol. 
61
 
12
(pg. 
1693
-
1700
)
127
Sandborn
 
WJ
Feagan
 
BG
Fedorak
 
RN
, et al. 
Ustekinumab Crohn’s Disease Study Group
A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn’s disease.
Gastroenterology
2008
, vol. 
135
 
4
(pg. 
1130
-
1141
)
128
Buonocore
 
S
Ahern
 
PP
Uhlig
 
HH
, et al. 
Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology.
Nature
2010
, vol. 
464
 
7293
(pg. 
1371
-
1375
)
129
Tesmer
 
LA
Lundy
 
SK
Sarkar
 
S
, et al. 
Th17 cells in human disease.
Immunol Rev
2008
, vol. 
223
 (pg. 
87
-
113
)
130
Wilke
 
CM
Bishop
 
K
Fox
 
D
, et al. 
Deciphering the role of Th17 cells in human disease.
Trends Immunol
2011
, vol. 
32
 
12
(pg. 
603
-
611
)
131
Solt
 
LA
Kumar
 
N
Nuhant
 
P
, et al. 
Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand.
Nature
2011
, vol. 
472
 
7344
(pg. 
491
-
494
)
132
Huh
 
JR
Leung
 
MWL
Huang
 
P
, et al. 
Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORγt activity.
Nature
2011
, vol. 
472
 
7344
(pg. 
486
-
490
)
133
Blazar
 
BR
Murphy
 
WJ
Abedi
 
M
Advances in graft-versus-host disease biology and therapy.
Nat Rev Immunol
2012
, vol. 
12
 
6
(pg. 
443
-
458
)
134
Serody
 
JS
Hill
 
GR
The IL-17 differentiation pathway and its role in transplant outcome.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
1 Suppl
(pg. 
S56
-
S61
)
135
Carvalho
 
A
Cunha
 
C
Di Ianni
 
M
, et al. 
Prognostic significance of genetic variants in the IL-23/Th17 pathway for the outcome of T cell-depleted allogeneic stem cell transplantation.
Bone Marrow Transplant
2010
, vol. 
45
 
11
(pg. 
1645
-
1652
)
136
Carlson
 
MJ
West
 
ML
Coghill
 
JM
Panoskaltsis-Mortari
 
A
Blazar
 
BR
Serody
 
JS
 
In vitro-differentiated TH17 cells mediate lethal acute graft-versus-host disease with severe cutaneous and pulmonary pathologic manifestations. Blood. 2009;113(6):1365-1374.
137
Kappel
 
LW
Goldberg
 
GL
King
 
CG
, et al. 
IL-17 contributes to CD4-mediated graft-versus-host disease.
Blood
2009
, vol. 
113
 
4
(pg. 
945
-
952
)
138
Iclozan
 
C
Yu
 
Y
Liu
 
C
, et al. 
T helper17 cells are sufficient but not necessary to induce acute graft-versus-host disease.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
2
(pg. 
170
-
178
)
139
Yu
 
Y
Wang
 
D
Liu
 
C
, et al. 
 
Prevention of GVHD while sparing GVL effect by targeting Th1 and Th17 transcription factor T-bet and RORγt in mice. Blood. 2011;118(18):5011-5020.
140
Laurence
 
A
Amarnath
 
S
Mariotti
 
J
, et al. 
STAT3 transcription factor promotes instability of nTreg cells and limits generation of iTreg cells during acute murine graft-versus-host disease.
Immunity
2012
, vol. 
37
 
2
(pg. 
209
-
222
)
141
Fulton
 
LM
Carlson
 
MJ
Coghill
 
JM
, et al. 
 
Attenuation of acute graft-versus-host disease in the absence of the transcription factor RORγt. J Immunol. 2012;189(4):1765-1772.
142
Bucher
 
C
Koch
 
L
Vogtenhuber
 
C
, et al. 
IL-21 blockade reduces graft-versus-host disease mortality by supporting inducible T regulatory cell generation.
Blood
2009
, vol. 
114
 
26
(pg. 
5375
-
5384
)
143
Chen
 
X
Das
 
R
Komorowski
 
R
, et al. 
Blockade of interleukin-6 signaling augments regulatory T-cell reconstitution and attenuates the severity of graft-versus-host disease.
Blood
2009
, vol. 
114
 
4
(pg. 
891
-
900
)
144
Das
 
R
Komorowski
 
R
Hessner
 
MJ
, et al. 
Blockade of interleukin-23 signaling results in targeted protection of the colon and allows for separation of graft-versus-host and graft-versus-leukemia responses.
Blood
2010
, vol. 
115
 
25
(pg. 
5249
-
5258
)
145
Nishimori
 
H
Maeda
 
Y
Teshima
 
T
, et al. 
Synthetic retinoid Am80 ameliorates chronic graft-versus-host disease by down-regulating Th1 and Th17.
Blood
2012
, vol. 
119
 
1
(pg. 
285
-
295
)
146
Grivennikov
 
SI
Greten
 
FR
Karin
 
M
Immunity, inflammation, and cancer.
Cell
2010
, vol. 
140
 
6
(pg. 
883
-
899
)
147
Li
 
N
Grivennikov
 
SI
Karin
 
M
The unholy trinity: inflammation, cytokines, and STAT3 shape the cancer microenvironment.
Cancer Cell
2011
, vol. 
19
 
4
(pg. 
429
-
431
)
148
Kujawski
 
M
Kortylewski
 
M
Lee
 
H
, et al. 
Stat3 mediates myeloid cell-dependent tumor angiogenesis in mice.
J Clin Invest
2008
, vol. 
118
 
10
(pg. 
3367
-
3377
)
149
Kortylewski
 
M
Kujawski
 
M
Wang
 
T
, et al. 
Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity.
Nat Med
2005
, vol. 
11
 
12
(pg. 
1314
-
1321
)
150
Kryczek
 
I
Wei
 
S
Zou
 
L
, et al. 
Cutting edge: Th17 and regulatory T cell dynamics and the regulation by IL-2 in the tumor microenvironment.
J Immunol
2007
, vol. 
178
 
11
(pg. 
6730
-
6733
)
151
Kryczek
 
I
Banerjee
 
M
Cheng
 
P
, et al. 
 
Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood. 2009;114(6):1141-1149.
152
Muranski
 
P
Restifo
 
NP
Does IL-17 promote tumor growth?
Blood
2009
, vol. 
114
 
2
(pg. 
231
-
232
)
153
Zou
 
W
Restifo
 
NPT
T(H)17 cells in tumour immunity and immunotherapy.
Nat Rev Immunol
2010
, vol. 
10
 
4
(pg. 
248
-
256
)
154
Tartour
 
E
Fossiez
 
F
Joyeux
 
I
, et al. 
Interleukin 17, a T-cell-derived cytokine, promotes tumorigenicity of human cervical tumors in nude mice.
Cancer Res
1999
, vol. 
59
 
15
(pg. 
3698
-
3704
)
155
Wang
 
L
Yi
 
T
Kortylewski
 
M
, et al. 
IL-17 can promote tumor growth through an IL-6-Stat3 signaling pathway.
J Exp Med
2009
, vol. 
206
 
7
(pg. 
1457
-
1464
)
156
He
 
D
Li
 
H
Yusuf
 
N
, et al. 
IL-17 promotes tumor development through the induction of tumor promoting microenvironments at tumor sites and myeloid-derived suppressor cells.
J Immunol
2010
, vol. 
184
 
5
(pg. 
2281
-
2288
)
157
Benchetrit
 
F
Ciree
 
A
Vives
 
V
, et al. 
Interleukin-17 inhibits tumor cell growth by means of a T-cell-dependent mechanism.
Blood
2002
, vol. 
99
 
6
(pg. 
2114
-
2121
)
158
Kryczek
 
I
Wei
 
S
Szeliga
 
W
, et al. 
Endogenous IL-17 contributes to reduced tumor growth and metastasis.
Blood
2009
, vol. 
114
 
2
(pg. 
357
-
359
)
159
Chalmin
 
F
Mignot
 
G
Bruchard
 
M
, et al. 
Stat3 and Gfi-1 transcription factors control Th17 cell immunosuppressive activity via the regulation of ectonucleotidase expression.
Immunity
2012
, vol. 
36
 
3
(pg. 
362
-
373
)
160
Langowski
 
JL
Zhang
 
X
Wu
 
L
, et al. 
IL-23 promotes tumour incidence and growth.
Nature
2006
, vol. 
442
 
7101
(pg. 
461
-
465
)
161
Stolfi
 
C
Rizzo
 
A
Franzè
 
E
, et al. 
Involvement of interleukin-21 in the regulation of colitis-associated colon cancer.
J Exp Med
2011
, vol. 
208
 
11
(pg. 
2279
-
2290
)
162
Purwar
 
R
Schlapbach
 
C
Xiao
 
S
, et al. 
Robust tumor immunity to melanoma mediated by interleukin-9-producing T cells.
Nat Med
2012
, vol. 
18
 
8
(pg. 
1248
-
1253
)
163
Overwijk
 
WW
de Visser
 
KE
Tirion
 
FH
, et al. 
Immunological and antitumor effects of IL-23 as a cancer vaccine adjuvant.
J Immunol
2006
, vol. 
176
 
9
(pg. 
5213
-
5222
)
164
Tong
 
Z
Yang
 
XO
Yan
 
H
, et al. 
A protective role by interleukin-17F in colon tumorigenesis.
PLoS ONE
2012
, vol. 
7
 
4
pg. 
e34959
 
165
Cho
 
BS
Lim
 
JY
Yahng
 
SA
, et al. 
Circulating IL-17 levels during the peri-transplant period as a predictor for early leukemia relapse after myeloablative allogeneic stem cell transplantation.
Ann Hematol
2012
, vol. 
91
 
3
(pg. 
439
-
448
)
166
Muranski
 
P
Boni
 
A
Antony
 
PA
, et al. 
Tumor-specific Th17-polarized cells eradicate large established melanoma.
Blood
2008
, vol. 
112
 
2
(pg. 
362
-
373
)
167
Martin-Orozco
 
N
Muranski
 
P
Chung
 
Y
, et al. 
T helper 17 cells promote cytotoxic T cell activation in tumor immunity.
Immunity
2009
, vol. 
31
 
5
(pg. 
787
-
798
)
168
Paulos
 
CM
Carpenito
 
C
Plesa
 
G
, et al. 
The inducible costimulator (ICOS) is critical for the development of human T(H)17 cells.
Sci Transl Med
2010
, vol. 
2
 
55
pg. 
55ra78
 
169
Hinrichs
 
CS
Kaiser
 
A
Paulos
 
CM
, et al. 
Type 17 CD8+ T cells display enhanced anti-tumor immunity [published online ahead of print May 26, 2009].
Blood
2009
, vol. 
114
 
3
(pg. 
596
-
599
)
170
Muranski
 
P
Borman
 
ZA
Kerkar
 
SP
, et al. 
Th17 cells are long lived and retain a stem cell-like molecular signature.
Immunity
2011
, vol. 
35
 
6
(pg. 
972
-
985
)
171
Lee
 
YK
Turner
 
H
Maynard
 
CL
, et al. 
Late developmental plasticity in the T helper 17 lineage.
Immunity
2009
, vol. 
30
 
1
(pg. 
92
-
107
)
172
Martin-Orozco
 
N
Chung
 
Y
Chang
 
SH
, et al. 
Th17 cells promote pancreatic inflammation but only induce diabetes efficiently in lymphopenic hosts after conversion into Th1 cells.
Eur J Immunol
2009
, vol. 
39
 
1
(pg. 
216
-
224
)
173
O’Shea
 
JJ
Lahesmaa
 
R
Vahedi
 
G
, et al. 
Genomic views of STAT function in CD4+ T helper cell differentiation.
Nat Rev Immunol
2011
, vol. 
11
 
4
(pg. 
239
-
250
)
174
Wei
 
G
Wei
 
L
Zhu
 
J
, et al. 
Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells.
Immunity
2009
, vol. 
30
 
1
(pg. 
155
-
167
)
175
Hirota
 
K
Duarte
 
JH
Veldhoen
 
M
, et al. 
Fate mapping of IL-17-producing T cells in inflammatory responses.
Nat Immunol
2011
, vol. 
12
 
3
(pg. 
255
-
263
)
176
Cosmi
 
L
Cimaz
 
R
Maggi
 
L
, et al. 
Evidence of the transient nature of the Th17 phenotype of CD4+CD161+ T cells in the synovial fluid of patients with juvenile idiopathic arthritis.
Arthritis Rheum
2011
, vol. 
63
 
8
(pg. 
2504
-
2515
)
177
Hamaï
 
A
Pignon
 
P
Raimbaud
 
I
, et al. 
Human T(H)17 immune cells specific for the tumor antigen MAGE-A3 convert to IFN-γ-secreting cells as they differentiate into effector T cells in vivo.
Cancer Res
2012
, vol. 
72
 
5
(pg. 
1059
-
1063
)
178
Waddington
 
CH
The Strategy of the Genes
1957
London, United Kingdom
George Allen & Unwin
179
MacLeod
 
MKL
Clambey
 
ET
Kappler
 
JW
Marrack
 
P
CD4 memory T cells: What are they and what can they do?
Seminars in Immunology
2009
, vol. 
21
 (pg. 
53
-
61
)
180
Gattinoni
 
L
Lugli
 
E
Ji
 
Y
, et al. 
A human memory T cell subset with stem cell-like properties.
Nat Med
2011
, vol. 
17
 
10
(pg. 
1290
-
1297
)
181
Gattinoni
 
L
Zhong
 
XS
Palmer
 
DC
, et al. 
Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells.
Nat Med
2009
, vol. 
15
 
7
(pg. 
808
-
813
)
182
Gattinoni
 
L
Ji
 
Y
Restifo
 
NP
Wnt/beta-catenin signaling in T-cell immunity and cancer immunotherapy.
Clin Cancer Res
2010
, vol. 
16
 
19
(pg. 
4695
-
4701
)
183
Yu
 
Q
Sharma
 
A
Ghosh
 
A
, et al. 
T cell factor-1 negatively regulates expression of IL-17 family of cytokines and protects mice from experimental autoimmune encephalomyelitis.
J Immunol
2011
, vol. 
186
 
7
(pg. 
3946
-
3952
)
184
Yu
 
Q
Sharma
 
A
Oh
 
SY
, et al. 
T cell factor 1 initiates the T helper type 2 fate by inducing the transcription factor GATA-3 and repressing interferon-gamma.
Nat Immunol
2009
, vol. 
10
 
9
(pg. 
992
-
999
)
185
Gattinoni
 
L
Klebanoff
 
CA
Palmer
 
DC
, et al. 
Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells.
J Clin Invest
2005
, vol. 
115
 
6
(pg. 
1616
-
1626
)
186
Klebanoff
 
CA
Gattinoni
 
L
Restifo
 
NP
CD8+ T-cell memory in tumor immunology and immunotherapy.
Immunol Rev
2006
, vol. 
211
 (pg. 
214
-
224
)
187
Shi
 
G
Ramaswamy
 
M
Vistica
 
BP
, et al. 
Unlike Th1, Th17 cells mediate sustained autoimmune inflammation and are highly resistant to restimulation-induced cell death.
J Immunol
2009
, vol. 
183
 
11
(pg. 
7547
-
7556
)
188
Coquet
 
JM
Middendorp
 
S
van der Horst
 
G
, et al. 
 
The CD27 and CD70 costimulatory pathway inhibits effector function of T helper 17 cells and attenuates associated autoimmunity. Immunity. 2013;38(1):53-65.
189
Pepper
 
M
Pagán
 
AJ
Igyártó
 
BZ
, et al. 
Opposing signals from the Bcl6 transcription factor and the interleukin-2 receptor generate T helper 1 central and effector memory cells.
Immunity
2011
, vol. 
35
 
4
(pg. 
583
-
595
)
190
Marshall
 
HD
Chandele
 
A
Jung
 
YW
, et al. 
Differential expression of Ly6C and T-bet distinguish effector and memory Th1 CD4(+) cell properties during viral infection.
Immunity
2011
, vol. 
35
 
4
(pg. 
633
-
646
)
191
Wirth
 
TC
Xue
 
HH
Rai
 
D
, et al. 
Repetitive antigen stimulation induces stepwise transcriptome diversification but preserves a core signature of memory CD8(+) T cell differentiation.
Immunity
2010
, vol. 
33
 
1
(pg. 
128
-
140
)
192
Luckey
 
CJ
Weaver
 
CT
Stem-cell-like qualities of immune memory; CD4+ T cells join the party.
Cell Stem Cell
2012
, vol. 
10
 
2
(pg. 
107
-
108
)
193
Kryczek
 
I
Zhao
 
ED
Liu
 
Y
, et al. 
Human TH17 cells are long-lived effector memory cells.
Sci Transl Med
2011
, vol. 
3
 
104
pg. 
ra100
 
194
Ma
 
CS
Deenick
 
EK
Batten
 
M
, et al. 
The origins, function, and regulation of T follicular helper cells.
J Exp Med
2012
, vol. 
209
 
7
(pg. 
1241
-
1253
)
195
Crotty
 
S
Johnston
 
RJ
Schoenberger
 
SP
Effectors and memories: Bcl-6 and Blimp-1 in T and B lymphocyte differentiation.
Nat Immunol
2010
, vol. 
11
 
2
(pg. 
114
-
120
)
196
Liu
 
X
Yan
 
X
Zhong
 
B
, et al. 
 
Bcl6 expression specifies the T follicular helper cell program in vivo. J Exp Med. 2012;209(10):1841-1852.
197
Crotty
 
S
Paul
 
WE
Littman
 
DR
Yokoyama
 
WM
Follicular helper CD4 T cells (T-FH).
Annual Review of Immunology
, vol. 
Vol 29
 
Palo Alto, CA
Annual Reviews; 2011:
(pg. 
621
-
663
)
198
Eyerich
 
S
Eyerich
 
K
Pennino
 
D
, et al. 
Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling.
J Clin Invest
2009
, vol. 
119
 
12
(pg. 
3573
-
3585
)
199
Trifari
 
S
Spits
 
H
IL-22-producing CD4+ T cells: middle-men between the immune system and its environment.
Eur J Immunol
2010
, vol. 
40
 
9
(pg. 
2369
-
2371
)
200
Duhen
 
T
Geiger
 
R
Jarrossay
 
D
Lanzavecchia
 
A
Sallusto
 
F
Production of interleukin 22 but not interleukin 17 by a subset of human skin-homing memory T cells.
Nat Immunol
2009
, vol. 
10
 (pg. 
857
-
863
)
201
Kagami
 
S
Rizzo
 
HL
Lee
 
JJ
, et al. 
Circulating Th17, Th22, and Th1 cells are increased in psoriasis.
J Invest Dermatol
2010
, vol. 
130
 
5
(pg. 
1373
-
1383
)
202
Wilhelm
 
C
Hirota
 
K
Stieglitz
 
B
, et al. 
An IL-9 fate reporter demonstrates the induction of an innate IL-9 response in lung inflammation.
Nat Immunol
2011
, vol. 
12
 
11
(pg. 
1071
-
1077
)
203
Wilhelm
 
C
Turner
 
JE
Van Snick
 
J
, et al. 
The many lives of IL-9: a question of survival?
Nat Immunol
2012
, vol. 
13
 
7
(pg. 
637
-
641
)
204
Zou
 
W
Restifo
 
NP
Nine lives for TH9s?
Nat Med
2012
, vol. 
18
 
8
(pg. 
1177
-
1178
)
Sign in via your Institution