Next Article in Journal
Bacteriophages and the Microbiome in Dermatology: The Role of the Phageome and a Potential Therapeutic Strategy
Next Article in Special Issue
Comparative Evaluation of Four Commercially Available Immunoassays for Therapeutic Drug Monitoring of Infliximab and Adalimumab
Previous Article in Journal
Application of Antisense Conjugates for the Treatment of Myotonic Dystrophy Type 1
Previous Article in Special Issue
Pathophysiology of Inflammatory Bowel Disease: Innate Immune System
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CD4 T-Cell Subsets and the Pathophysiology of Inflammatory Bowel Disease

by
Raquel Gomez-Bris
1,2,†,
Angela Saez
1,3,†,
Beatriz Herrero-Fernandez
1,2,
Cristina Rius
4,5,6,
Hector Sanchez-Martinez
1 and
Jose M. Gonzalez-Granado
1,6,7,8,*
1
LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
2
Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
3
Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), 28223 Pozuelo de Alarcón, Spain
4
Department of History of Science and Information Science, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
5
UISYS Research Unit, University of Valencia, 46010 Valencia, Spain
6
CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
7
Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
8
Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(3), 2696; https://doi.org/10.3390/ijms24032696
Submission received: 4 January 2023 / Revised: 24 January 2023 / Accepted: 28 January 2023 / Published: 31 January 2023

Abstract

:
Inflammatory bowel disease (IBD) is an umbrella term for the chronic immune-mediated idiopathic inflammation of the gastrointestinal tract, manifesting as Crohn’s disease (CD) or ulcerative colitis (UC). IBD is characterized by exacerbated innate and adaptive immunity in the gut in association with microbiota dysbiosis and the disruption of the intestinal barrier, resulting in increased bacterial exposure. In response to signals from microorganisms and damaged tissue, innate immune cells produce inflammatory cytokines and factors that stimulate T and B cells of the adaptive immune system, and a prominent characteristic of IBD patients is the accumulation of inflammatory T-cells and their proinflammatory-associated cytokines in intestinal tissue. Upon antigen recognition and activation, CD4 T-cells differentiate towards a range of distinct phenotypes: T helper(h)1, Th2, Th9, Th17, Th22, T follicular helper (Tfh), and several types of T-regulatory cells (Treg). T-cells are generated according to and adapt to microenvironmental conditions and participate in a complex network of interactions among other immune cells that modulate the further progression of IBD. This review examines the role of the CD4 T-cells most relevant to IBD, highlighting how these cells adapt to the environment and interact with other cell populations to promote or inhibit the development of IBD.

1. Introduction

The immune system is divided into two main branches, the innate and adaptive immune responses. Innate immune cells, which include neutrophils, monocytes, macrophages, and dendritic cells (DCs), respond rapidly and non-specifically to pathogens or other foreign entities as a first line of defense. Innate immune cells express pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs) and NOD-like receptors (NLR), that allow them to recognize pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), provoking their activation. Once activated, innate immune cells provoke inflammation by releasing cytokines and chemokines, activating the complement cascade and phagocytosing pathogens and cell debris. Some innate immune cells take up, process, and present antigens to activate the adaptive immune response, acting as antigen-presenting cells (APCs). Adaptive immunity depends on this antigen presentation by APCs, and the cytokine milieu generated by the innate response and thus takes longer to activate than innate immunity, but the corollary is that adaptive immunity is highly specific. The key cells of the adaptive immune system are CD4 and CD8 T-cells and B cells. Natural killer T-cells (NKT cells) and γδ T-cells are cytotoxic T lymphocytes that sit at the boundary between innate and adaptive immunity [1]. Profuse data show that the innate and adaptive immune systems both play significant roles in the origin and development of IBD [2,3,4,5,6,7,8,9,10].
IBD is a chronic immune-mediated idiopathic inflammation of the gastrointestinal tract with a prolonged period of relapse and remission [11,12,13,14]. Worldwide, 6.8 million people suffer from IBD [15]. IBD is considered a global disease, and its evolution can be stratified into four epidemiological stages, including emergence, acceleration in incidence, compounding prevalence, and prevalence equilibrium. Developing countries and newly industrialized countries are in the emergence and the acceleration in incidence stages, respectively, and Western regions are in the compounding prevalence stage and will eventually transition to the prevalence equilibrium stage [15]. Despite the success of IBD therapy, patients have a mortality risk 1.5 times higher than that of the healthy population, and mortality linked to IBD continues to increase progressively [16]. The stages of IBD range from mild to moderate to severe [17,18]. The main clinical manifestations of IBD are Crohn’s disease (CD) and ulcerative colitis (UC). These diseases differ in some symptoms, disease location, and histopathological characteristics but share gastrointestinal symptoms such as diarrhea, mucus, and bloody stools, and abdominal pain, as well as extraintestinal symptoms, such as arthritis, oral ulcers, skin lesions, and ophthalmological problems [19,20,21,22]. The numerous IBD complications include strictures, abscesses, fistulas, and colitis-associated cancer [23].
Even though the pathophysiology of IBD has not been fully defined, the etiology of the disease is known to involve a combination of genetic, environmental, microbiological, and immunological factors that promote intestinal barrier dysfunction and tissue damage, and dysregulated innate and adaptive immune responses [13,24,25].
A key feature in the appearance, progression, and prognosis of IBD is an aberrant intestinal mucosal immune system, and a common disease denominator in all IBD patients is the infiltration of intestinal tissue by inflammatory T-cells [26,27] and the accumulation of several proinflammatory cytokines associated with activated T-cells [28,29,30].
T-cells are classified broadly into proinflammatory and anti-inflammatory populations that form three main groups. The proinflammatory CD8 T-cells have cytotoxic capacity and are implicated in the response to tumors, metastatic cells, and viral infections [31]. CD4 T helper (Th) cells regulate the inflammatory milieu, promoting antibody production, controlling innate immunity, and stimulating immunologic memory. The third category is an anti-inflammatory CD4+ population called regulatory T-cells (Tregs), which suppress inflammatory responses, promote immunological tolerance, and control immune responses to prevent autoimmunity [32,33].
With the exception of natural Tregs, which are generated in the thymus during the positive selection of maturing T-cells [32], post-thymic naïve CD4 T-cells are nonactivated T-cells that have no contact with their cognate antigen and retain multiple differentiation capabilities [34]. Naïve T-cells undergo a functional and transcriptional programming called differentiation upon the recognition of an antigen presented by an APC in a secondary lymphoid organ. This recognition involves the binding of T-cell antigen receptors (TCRs) to host major histocompatibility complex (MHC) molecules complexed with foreign peptides [35]. After antigen recognition and the formation of an immune synapse with the APC [36], naïve CD4 T-cells are activated and undergo clonal expansion before differentiation. Changes in CD4 T-cells are mediated by a variety of factors, including the strength of the TCR signal, the cytokine microenvironment, and co-stimulation by the APC. These changes include chromatin remodeling and the modification of DNA methylation and promote the activation or suppression of specific transcription factors that direct the differentiation towards at least seven distinct T helper cell subsets: Th1, Th2, Th9, Th17, Th22, T follicular helper (Tfh), and several types of Tregs [37,38,39]. These CD4 T-cell subsets are classified according to their cytokine production and expression of master transcription factors [40]. T-cell subsets are further distinguished by the production of different components of the signaling transducer and activator (STAT) family (Figure 1).
Detailed knowledge of the functions of these lymphocyte subpopulations is essential for defining the complicated molecular and cellular pathways underlying IBD. In the following sections, we outline the main characteristics of these CD4 T-cell subsets and their positive or negative influence in IBD onset and progression.

2. T-Cells in IBD

Under steady state conditions, the gut contains scattered interepithelial lymphocytes and innate lymphocytes in the epithelial layer of the intestinal mucosa, with very few CD4 T-cells [41]. In contrast, IBD is associated with an abundance of CD4 T-cells in the epithelial layer of the inflamed intestinal mucosa [42] or with normal numbers of lamina propria and epithelial CD4 T-cells [43,44] but showing increased activation [45,46,47] and phenotypic alterations [48].
T-cells release interleukin (IL)-2, which signals in an autocrine manner via the IL-2 receptor, whose α chain, called CD25, is expressed on T-cells upon antigen recognition and activation. IBD is characterized by elevated numbers of hiCD25+ cells, specifically affecting T-cells in CD and macrophages in UC [49]. Some intestinal CD4 T-cells from CD patients, but not UC patients, also express high levels of the activating natural killer group 2D receptor (NKG2D) [50], whose stimulation in combination with that of the TCR promotes the cytotoxic capacity of CD4 T-cells, plus the release of the pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and IL-17A [50,51].
CD has usually been considered a type 1-driven disease, with the exacerbated production and activation of Th1 and Th17 cells and an elevated presence of their major cytokines IL-12, IL-23, IFN-γ, and IL-17. In contrast, UC has been designated as type 2-driven inflammation, linked to an elevated participation of Th2 and Th9 cells and their principal cytokines IL-13, IL-5, and IL-9 [52,53].

3. T Helper 1 (Th1) Cells

Th1 cells facilitate the eradication of intracellular pathogens, including parasites, protozoa, viruses, and intracellular bacteria, and intervene in cell-mediated immunity and delayed-type hypersensitivity reactions [54]. Th1 cells release IFN-γ and TNF-α, which stimulate innate immune cells, such as neutrophils and macrophages, and non-immune cells, such as epithelial cells and fibroblasts [55,56]. Th1 cells also release IFN-γ and IL-2 to recruit CD8 effector cytotoxic T-cells (CD8 CTL) [57].
Upon antigen recognition and the activation of a naïve CD4 T-cell, Th1 differentiation is mediated by the binding of IL-12 produced by the cognate APC. IL-12 induces T-cell expression of the master Th1 transcription factor T-box-containing protein (T-bet), encoded by the gene TBX21, and the cytokine IFN-γ, in both cases through a process dependent on STAT4 signaling stimulation [55,56,58]. T-bet increases the expression of IL-12 receptor subunit β2 (IL-12Rβ2), allowing synergistic IL-12 and STAT4 signaling to further increase IFN-γ generation [26,59,60].
In intestinal homeostasis, Th1 cells can prevent pathogen invasion and pathogen-derived antigens from mediating intestinal inflammation. Beside their direct antibacterial action, Th1 cells also ameliorate intestinal inflammation by secreting IL-2 and IL-10 to promote Treg stimulation. Moreover, Th1 cells can facilitate intestinal stem cell (ISC) proliferation and intraepithelial cell self-restoration by releasing low concentrations of TNF-α. Th1 cells thus constitute an immune barrier indispensable for intestinal homeostasis [61].
A pathogenic role for Th1 cells has been described in the course of IBD (Figure 2). An excessive Th1 response has been observed in the inflamed mucosa and serum of IBD patients [62]. Classically, an exacerbated Th1 response has been linked to CD, whereas UC has been considered a Th2 cell-driven disease [63]. However, both UC and CD feature activated effector Th1 cells, suggesting that Th1 cells are implicated in the origin and development of mucosal inflammation in IBD [64].
The elevated levels of IL-12 and IL-18 detected in IBD support an involvement of exacerbated Th1 immune responses and intestinal inflammation in CD [65,66]. These two cytokines, which are produced by macrophages, stimulate the production of IFN-γ by Th1 cells, and the blockade of IL-12 or IL-18 reduces IFN-γ production [65,66]. Early CD also features increased mucosal levels of the typical Th1 cytokines IFN-γ and IL-21 [67].
Th1 differentiation and function depend on the proteins T-bet, IFN-γ, and TNF-α. Supporting the importance of Th1 differentiation in IBD, a lack of IFN-γ in CD4 T-cells prevents the development of dextran sulfate sodium (DSS)-induced colitis in mice [68]. In humans, the IBD-associated single nucleotide polymorphisms, rs1551398 and rs1551399, alter T-bet binding sites and predispose their carriers to increased mucosal inflammation [69]. The elevated levels of TNF-α and IFN-γ in the intestinal epithelium in IBD disturb intraepithelial cell functions. TNF-α promotes intraepithelial cell apoptosis and inflammation [70], whereas IFN-γ stimulates macrophages and neutrophils and promotes immune-cell recruitment by inducing the expression of adhesion molecules on intraepithelial cells [71]. The direct destruction of intraepithelial cells has been attributed to an epithelial cell adhesion molecule (EpCAM)-specific action of IFN-γ+ Th1 cells upon antigen presentation by DCs [72,73,74].
The evidence that Th1 cells play different roles in CD and UC includes the observation that Th1 cells isolated from the lamina propria of CD patients produce more IFN-γ than cells from UC patients or control individuals [75,76,77,78]. However, other authors did not detect these differences [79,80].
The transcription factor interferon regulatory factor 5 (IRF5) aggravates experimental colitis by increasing the CD4 T-cell expression of Th1- and Th17-related cytokines and reducing the expression of Th2-related cytokines [81]. Another regulator of transcription, among other functions, is the nuclear envelope protein lamin A/C, which promotes T-cell activation [82,83] and Th1 differentiation [84], inhibits Treg differentiation [85], and aggravates IBD in a cell adoptive transfer mouse model of colitis [86].
The specific binding of the integrin αEβ7 to E-cadherin on epithelial cells promotes the retention of Th1 in the intestinal mucosa [87]. Moreover, αEβ7+ Th1 cells express higher levels of IFN-γ and TNF-α than αEβ7- cells in the intestine of UC patients [88]. Finally, the inhibition of T-cell activation with a selective Ca2+ release-activated Ca2+ channel blocker inhibits IFN-γ production by organ culture biopsies from IBD patients [89]. In summary, Th1 cells influence the severity of intestinal inflammation.

4. T Helper 2 (Th2) Cells

Th2 cells participate in the elimination of extracellular microbes and intestinal helminths and support IgE-mediated B-cell responses by secreting IL-4, IL-5, IL-13, and IL-10 [90]. Th2 polarization is mediated by IL-4-ligation–dependent STAT6 signaling and the production of the Th2 master transcription factor GATA binding protein 3 (GATA-3) [91,92]. In addition to IL-4, Th2 cells produce the cytokines IL-5, IL-13, IL-21, and IL-25. Th2 cytokines prevent Th1 differentiation and promote the activation of macrophages [54,55]. Impaired Th2 responses are linked to allergies and asthma [93,94,95,96,97,98].
Oxazolone-induced colitis in mice involves a Th2 response featuring IL-5 and IL-4 production [99]. Another important Th2 cytokine is IL-33, which is elevated in UC patients and in mouse models of colitis induced with trinitrobenzenesulfonic acid (TNBS) or DSS. Moreover, IL-33 and the IL-33 receptor ST2 (suppression of tumorigenicity 2) are associated with IBD risk loci [53,100,101,102,103,104,105,106,107]. A lack of ST2 in mice diminishes colitis, whereas the administration of exogenous IL-33 aggravates the condition. These effects are associated with increased amounts of the Th2 cytokines IL-4, IL-5, and IL-13; major reductions in IL-17 and IFN- γ; damage to the epithelial barrier; and delayed wound recovery in the damaged colonic epithelium [53,100,101,102,105,106,107]. In contrast, IL-33 protects against intestinal inflammation by promoting the differentiation of forkhead box P3 (Foxp3)+ Tregs and innate lymphoid cells (ILCs) and by inducing the expression of amphiregulin [108,109].
Nevertheless, the treatment of UC with the anti-IL-13 monoclonal antibodies, tralokinumab and anrukinzumab, has not produced clinical benefits [110,111]. The levels of IL-36β, a member of the IL-1 cytokine family, are elevated in IBD patients, and IL-36β exacerbates DSS-induced colitis in mice by promoting Th2 responses in the lamina propria while reducing Foxp3+ Treg responses [112].

5. T Helper 9 (Th9) Cells

Th9 cells, like Th2 cells, intervene in the response to intestinal helminths [113] and have been linked to allergy and autoimmunity [114]. The differentiation of Th9 cells is induced by the concurrent action of IL-4 and transforming growth factor-beta (TGF-β). IL-4 binding to the IL-4 receptor triggers GATA3 transcription and the phosphorylation and dimerization of STAT6, promoting Th2 differentiation, whereas TGF-β activates FOXP3, inducing Treg differentiation [115,116]. In combination, IL-4 and TGF-β induce the production of IL-9 and the polarization of CD4 T-cells towards the Th9 phenotype [117,118,119]. Th9 differentiation depends on multiple transcription factors, including PU.1 and IRF4 [120,121]. Th9 differentiation can also be induced by other molecular combinations [122], such as IL-4 plus IL-1β [123]. Th9 cells are the main source of IL9, but also release IL-10 [118,124]. IL-9 can act as a proinflammatory cytokine, activating Th17 cells [125], and shares the same γ-chain receptor as IL-4, IL-2, and IL-15. IL-9 binding to its receptor activates janus kinase (JAK)1 and JAK3, which form dimers with STAT3, STAT5, or STAT1 [126,127,128].
The contribution of Th9 cells and their role in gut immunity have been demonstrated in several studies. Altered tissue integrity and continuous inflammation during flare-up episodes in UC are associated with IL-9 release by Th9 cells in the colon [129,130]. The presence of Th9-derived IL-9 is associated with alterations in the expression of tight junctions [131].
Several studies performed with IBD patient samples and mouse models have shown increased levels of IL-9 and Th9-related transcription factors [129,132,133,134]. Th9 cell numbers and activity are increased in the inflamed mucosa of UC patients [135]. The intestinal overproduction of IL-9 is likely to affect epithelial-barrier integrity and compromise tolerance to commensal bacteria, potentially progressing to inflammation [135].
Th2 and Th9 responses are interrelated, and elevated IL-9 and Th9 cell numbers, such as increased Th2 responses, are especially important in UC [136]. In the Th2-dominant oxazolone-induced colitis mouse model [3], IL-9 expression is increased throughout the intestinal tract, and the number of intestinal and splenic IL-9+ CD4+ T-cells is higher than in control mice [137]. A lack of IL-9 in the oxazolone-induced colitis model was found to reduce histological and disease symptoms and to enhance intestinal-barrier function [129]. Interestingly, in the TNBS-induced colitis mouse model, which features a potent Th1 response that resembles CD [3], mice lacking IL-9 show less severe inflammation and weight loss than wild-type mice. Moreover, IL-9-deficient mice in the TNBS-induced colitis model showed much less prominent goblet cell impairment, wound stimulation, and mononuclear cell deposition [130]. This discrepancy between models suggests that the role of Th9 and IL-9 in the development of IBD depends on the local microenvironment. This microenvironment is dominated by Th2 responses in the oxazolone-induced colitis model, provoking a more inflammatory Th9 response, and by Th1/Th17 responses in the TNBS-induced colitis model, promoting a tolerogenic-biased Th9 response [3,138]. The Th1/Th2 cytokine milieu of a third mouse model of colitis, induced with DSS, is considered to resemble both UC and CD [3]. These mice show elevated numbers of Th9 cells expressing PU.1 and CD3 markers, and IL-9 antibody-blockade was found to reduce disease symptoms and the presence of inflammatory mediators by reducing lymphocyte activity in the mouse intestinal lamia propria [139].
TNF-like factor (TL)1A and its receptor death receptor (DR)3 belong to the TNF and TNFR protein superfamilies. The attachment of APC-derived TL1A to lymphocyte DR3 provides co-stimulation to activated lymphocytes. DR3-dependent signaling modulates proliferative activity of and cytokine production by effector lymphocytes while also significantly impacting the generation and inhibitory capacity of Tregs [140]. Intestinal inflammation in chronic DSS-induced colitis is aggravated by elevated TL1A expression. In this model, TL1A may promote Th9 differentiation and IL-9 release by upregulating the expression of TGF-β, IL-4, and PU.1, suggesting a new target for IBD treatment [141]. In another study, inflamed tissue from UC patient intestine was found to contain elevated amounts of IL-9, IL-6, and IL-17A mRNA, and IL-9 mRNA levels correlated with the inflammation score [132]. The expression of TGF-β and IL-4, which potentiate Th9 cell differentiation [142], is increased in IBD patients and correlates with inflammation and disease symptoms [132].
IL-9-producing T-cells from UC patients show augmented expression of α4β7 integrin, which mediates the homing of Th9 cells to the intestine [143,144], and αEβ7 and α4β7-expressing T lymphocytes accumulate in UC-patient intestine. αEβ7 integrin binds to E-cadherin and MadCam in the intestine, promoting T-cell retention [144]. Th9 accumulation in UC patients can be abrogated by the blockade of the β7 subunit of integrin α4β7 and αEβ7 with the monoclonal antibody Etrolizumab [145].
In summary, mouse and human studies suggest that Th9 cells and their main cytokine IL-9 play a prominent role in IBD pathogenesis, especially in UC. Knowledge of how the recruitment and action of Th9 cells can be manipulated is necessary for improving therapeutic strategies.

6. T Helper 17 (Th17) Cells

Th17 cells protect the host from bacterial and fungal infections on mucosal surfaces but are also implicated in inflammatory and autoimmune diseases [146]. Th17 cells have thus been identified as pathogenic cells in relation to tissue inflammation and autoimmune disease [147,148,149]. However, it is becoming clear that Th17 cells also have a non-pathogenic phenotype with immune-modulatory functions [61,150,151,152,153].
Pathogenic and non-pathogenic Th17 cells can be polarized in vitro [154]. A combination of IL-6, IL-23, and IL-1β promotes pathogenic Th17 differentiation [155,156], whereas TGF-β1, in addition to IL-6, favors non-pathogenic Th17 cells [150,157,158,159]. IL-23 appears not to promote Th17  differentiation directly since naïve T-cells do not express the IL-23 receptor (IL-23R) in vitro, thus suggesting that IL-23 stabilizes the Th17 phenotype and promotes Th17 cell survival [156].
Pathogenic and non-pathogenic Th17 cells both express the transcription factor retinoic acid receptor-related orphan nuclear receptor gamma (RORγt) [160] in a STAT3-dependent manner [161] and produce IL-17 [61]; however, they have distinct genetic signatures, one contributing to immune injury and the other to immune homeostasis [154,162]. Pathogenic Th17 cells are characterized by the production of pro-inflammatory molecules, such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-23R, and by a low expression of immune-regulatory molecules, such as IL-10 and CD5 molecule like (CD5L). In contrast, non-pathogenic Th17 cells produce low amounts of GM-CSF and IL-23R and high amounts of IL-10 and CD5L, facilitating tissue homeostasis [150,151,156,163,164].
In settings of inflammation, Th17 cells produce IL-17A and IL-17F [160], members of the IL-17 family of proinflammatory cytokines, which run from IL-17A through F, with IL-17A frequently denoted as IL-17 [165]. IL-17 binds to a heterodimeric receptor (composed of IL-17RA and IL-17RC), which is expressed on many non-hematopoietic cells, including intestinal epithelial cells and on some activated T-cells [166]. The binding of IL-17 to its receptor regulates intestinal barrier function and the release of inflammatory chemokines and cytokines by target cells [167].
Other cytokines produced by Th17 cells include IL-22 and IL-21. At sites of inflammation, Th17 cells secrete several chemokines that promote the recruitment of neutrophils (chemokine (C-X-C motif) ligand (CXCL)1, CXCL2, CXCL5, and CXCL8) [168], as well as Tregs and more Th17 cells (chemokine receptor 6 (CCR6) and their ligands (C-C motif ligand (CCL20)) [165]. These Th17 cells also secrete granulopoiesis factors (granulocyte colony-stimulating factor (G-CSF)) and mediators of the acute phase response, including IL-6. IL-17 also stimulates the activity of matrix metalloproteinases (MMPs). Commensal bacteria promote IL-17 and IL-22 production, which in turn promote the production of barrier-protective cytokines and antimicrobial peptides [169,170].
Unlike Th1 and Th2 cells, Th17 cells show great plasticity and are able to differentiate into Th1, Tregs, and Tfh cells [61,171].
The binding of Th17-cell expressed CCR6 to the chemokine CCL20 recruits Th17 cells to the intestine, where they secrete IL-17, IL-21, IL-22, and TNF-α [172]. In homeostasis, Th17 cells control the proliferation and differentiation of lymphocytes, macrophages, and neutrophils; combat infection; and protect the integrity of the intestinal barrier [173,174]. However, alterations in Th17 cell number and functions can promote uncontrolled inflammation and mediate the development of IBD [30,61,171,172,175,176,177].
Th17 cells are more numerous in the peripheral blood of IBD patients, and several major Th17 cytokines, such as IL-17, IL-21, and IL-23, are abundant in the inflamed mucosa of these patients [178].
Compared with UC and healthy individuals, CD patients have elevated serum IL-17A and Th17 cell numbers in gut-draining lymph nodes [179]. Moreover, the numbers of IL-17-expressing cells are increased in the gut of patients with active CD and UC relative to the numbers in healthy individuals and patients with inactive CD or UC [180].
High levels of IL-17 and IL-21 promote the production of matrix MMPs by myofibroblasts, which results in the lysis of the extracellular matrix and epithelial cell damage. These cytokines also promote epithelial cells to release chemokines that stimulate inflammatory cell recruitment [181]. For example, IL-17 promotes IL-8 release by epithelial cells, stimulating the recruitment of neutrophils and Th17 cells to the inflamed tissue [175]. In line with these observations, a lack of the IL-17 receptor (IL-17R) in mice protects against TNBS-induced colitis [182], and a lack of IL-17F confers resistance to DSS-induced colitis [183]. However, the absence of IL-17 aggravates DSS-induced colitis, indicating that IL-17 also has beneficial effects [184]. IL-21 released by Th17 cells acts in an autocrine manner to promote their differentiation and the production of IL-17. Th17 responses are also supported by autocrine production of IL-23 [185]. Increased levels of IL-21 have been found in mice with chronic DSS-induced and TNBS-induced colitis, and IL-21 blockade by the addition of neutralizing IL-21R fusion proteins to DSS-treated mice mitigates colitis and inhibits the release of the main Th17 cytokines [186]. IL-21 enhances Th1 responses and IFNγ production by both Th1 and NK cells [187].
Th17 cells release TNF-α, which binds to the receptors TNFR-1 and TNFR-2 [188] and enhances IBD [189]. Th17 cells in the gut of CD patients can secrete IL-17 and IFNγ together, a finding confirmed by treating Th17 cells in vitro with the pro-Th1 cytokine IL-12 [190]. The transition of Th17 cells to the Th1 phenotype has also been reported in experimental mouse models, including of IBD [191]. Surprisingly, the blockade of IL-17A or IL-17R with the antibodies Secukinumab or Brodalumab in patients with moderate to severe CD generated more serious adverse events in the treatment group than in patients receiving the placebo [192,193]. Increased disease scores and symptoms were also observed in DSS-induced colitis upon the blockade of IL-17A [194] or in IL-17 KO mice [169]. Colitis-like disease is also promoted by the transfer of Th cells from mice lacking IL-17A or IL-17RA into mice that are deficient for recombination activating gene (RAG)1 [167].
Similarly to the situation described above for Th9 cells, mice genetically deficient for IL-17R or treated with IL-17R-Ig fusion protein develop less severe IBD upon treatment with TNBS [182], suggesting that the protective role of IL-17 may depend on the specific colitis model and the local microenvironmental inflammatory conditions.
A possible explanation for these conflicting results is that the inhibition of the Th17 response can potentiate the more proinflammatory Th1 phenotype, as reported in a study that found increased intestinal Ifng mRNA and Th1 polarization in the absence of IL-17A, reflecting the ability of IL-17 to diminish the expression of IFN-γ and thus potentially enhance the stability of the Th17 phenotype by limiting Th1 differentiation [167]. Alternatively, the protective function of IL-17A may be related to its ability to regulate the epithelial barrier function and gut homeostasis [195]. Supporting this, IL-17 antibody blockade enhances the permeability of the intestinal epithelial barrier in DSS-induced colitis in mice [196]. In both cases, the increase in permeability correlated with changes in epithelial tight junction gene expression and in occludin positioning within the damaged epithelial layer [195,196].
IL-17 also modulates anti-microbial peptide release, potentially modulating microbial populations within the gut in IBD [195]. IL-17, in concert with fibroblast growth factor 2, also controls both epithelial barrier maintenance and bacterial homeostasis in the intestine [169]. Together, these data indicate a proinflammatory effect of Th17 in concert with a role in maintaining a healthy epithelial barrier and an optimal bacterial balance.
IL-23 promotes the expansion of pathogenic Th17 cells by maintaining Th17 signature genes, upregulating effector genes, such as IL17A, IL17F, or IL22, or repressing suppressive factors. Moreover, IL17 and IL23 signaling promote pro-inflammatory molecules such as TNF, IFNγ, IL22, lymphotoxin, and IL1β [176]. Several mouse models of colitis have shown an augmented production of IL23 [197,198,199,200]. In patients, treatment with selective IL23 inhibitors promotes better response rates in the cohort of CD patients that failed prior anti-TNF therapy (reviewed in [176]), and IL23 targeting in UC patient, is safe and effective and promote and sustain clinical remission, low inflammation, mucosal healing, and an improved quality of life (reviewed in [201]). These experiments indicate the importance of the IL23/IL17 axis in mucosal inflammation.

7. T Helper 22 (Th22) Cells

Th22 cells protect against tissue damage and bacterial infection by producing the IL-10 family member IL-22 [202,203,204]. Th22 cells also produce IL-13, fibroblast growth factor, chemokines, and TNFα. IL-22 is also secreted by Th1 and Th17 cells, but Th22 cells are able to secrete IL-22 without producing IFN-γ or IL-17 [54,205]. IL-22 is also secreted by NKs, γδT cells, ILC3s, and some nonlymphoid cells [206]. Th22 cells express the chemokine receptors CCR10, CCR6, and CCR4, and their differentiation is promoted by the activation of STAT3 and the aryl hydrocarbon receptor (AHR) by IL-6, TNF-α, and IL-1β and is diminished by TGF-β [207,208].
IL-22 enhances innate immunity by modulating cell differentiation, chemokine secretion, and antimicrobial peptide (AMP) secretion [209,210,211]. In the intestinal epithelium, IL-22 promotes the secretion of AMPs, such as β defensins and lipocalin 2 and the mucin proteins MUC1 and MUC3 [212]. IL-22 can also promote the secretion by human colonic myofibroblasts of the anti-inflammatory factor IL-11 and inflammatory molecules, such as IL-6 and CXCL chemokines [213].
In healthy individuals, IL-22 is released mainly in the gastrointestinal tract, where it favors mucosal recovery [214,215]. This beneficial effect is mediated by the binding of IL-22 to the receptor IL-22R, whose expression is mostly limited to epithelial cells [214].
IL-22 maintains intestinal epithelial barrier function by promoting the release of antimicrobial peptides [202] and mucins [215], as well as by facilitating intestinal epithelial cell survival and proliferation [214]. IL-22 can increase the production of anti-inflammatory factors, such as IL-11, that also protect epithelial barrier function [216].
However, elevated levels of IL-22 can be detrimental [206], enhancing the production of inflammatory mediators, such as IL-6 and CXCL chemokines by human colonic myofibroblasts [217]. IL-22 modulates neutrophil recruitment to the colon by controlling the expression of neutrophil-active CXC-family chemokines in ulcerative colitis; by this mechanism, the augmented expression of IL-22 is associated with treatment resistance to an anti-IL-12/23 p40 subunit monoclonal antibody [218].
IL-22 is secreted at low levels, and is mostly maintained in a biologically inactive state through the action of IL-22 binding protein (IL-22BP, also known as IL-22RA2), produced by intestinal DCs and macrophages in the gut lamina propria and secondary lymphoid structures [219,220,221,222,223]. In inflamed intestinal tissue, the main producers of IL-22BP are CD4 T-cells [217,220]. IL-22BP is a soluble receptor homolog that attaches to IL-22 with greater affinity than IL-22R, preventing IL-22 from binding to its receptor and thereby blocking IL-22 signaling [224,225]. Elevated levels of IL-22 and IL-22BP mRNA and protein have been detected in inflamed tissue from CD and UC patients [220,226,227]. Consistent with these findings, the IL-22-associated protection against DSS-induced colitis is increased in IL-22BP deficient rats [228], and IL-22BP aggravates T-cell-mediated colitis in mice [220]. IL-22BP expression is reduced in infectious colitis but not in inflamed tissues in IBD, indicating potential pathophysiological significance for IL-22BP-dependent alterations in IL-22 bioactivity [220,228].These responses may vary between patients and differ according to the extent of histological damage. For example, CD patients with granuloma are reported to have increased frequencies of IL-22+ and IL-22+ IFN-γ+ cells in colonic tissue [229].
Results from experiments in mice thus seem to indicate that high levels of IL-22BP in IBD can provoke inflammation by interrupting IL-22-mediated mucosal healing [220]. Supporting this, gut CD4 T-cells from anti-TNF-α-treated IBD patients show lower amounts of IL-22BP but still express IL-22 [220] and may even up-regulate IL-22 generation [230].

8. Regulatory T-Cells (Treg)

Treg cells suppress immune responses and maintain peripheral tolerance and immune homeostasis [231]. Tregs are divided into thymic-derived Tregs, also called natural Treg cells (nTregs) [232], and post-thymic maturation peripheral Tregs (pTregs) [233,234,235,236,237]. Tregs induced in vitro by the addition of TGF-β and IL-2 to naïve CD4 T-cells are called inducible Tregs (iTregs) [237,238].
Tregs are characterized by the secretion of the inhibitory cytokines IL-10, IL-35, and TGF-β, and the expression of the transcription factor Foxp3, which mediates Treg development, lineage commitment, and regulatory functions [55]. Another marker of nTregs and pTregs is the IL-2 receptor α chain CD25 [55].
nTregs are positively selected in the thymus by the intermediate affinity of the TCR for self-peptides/MHC [232], whereas T-cells with a high-affinity TCR antigen are eliminated and those with low-affinity differentiate into naïve T-cells [239]. In humans, nTreg development seems to also depend on IL-2 and/or IL-15 [240,241,242].
In the thymus, a restricted number of autoreactive CD4 T-cells differentiate into nTregs, in a process called agonist selection that guarantees central tolerance to self-antigens, thus avoiding autoimmunity [236,243,244]. nTregs are already in an antigen-primed or antigen-activated state in the thymus [238].
pTregs differentiate from conventional CD4 T-cells in the periphery under tolerogenic conditions in secondary lymphoid tissues, in particular intestinal draining lymph nodes, upon the recognition of an antigen presented by an APC [245,246,247]. pTreg differentiation requires the sustained expression of FOXP3 and is dependent on high levels of TGF-β, an absence of proinflammatory cytokines [54], and the activation of naïve CD4 T-cells upon recognition of mainly exogenous antigens [248,249,250]. pTreg differentiation is also facilitated by vitamin-A derived retinoic acid [251,252,253].
pTregs are classified as central, effector, and tissue-resident pTregs [254]. Central pTregs are considered naïve and in mice are characterized by the expression of the markers CD62Lhigh CCR7+ or CD45RAhigh CD25low. Central pTregs are the main Treg type in the circulation and in secondary lymphoid organs. The marker profile of effector Tregs, also called effector memory or activated Tregs, is CD62Llow, CCR7low, CD44hi killer cell lectin like receptor G1 (KLRG1)+, CD103+, or CD25RAlow CD25hi. Effector memory pTregs are less frequent than central pTregs and are similar to conventional activated CD4 T-cells that have had recent contact with an antigen. Tissue-resident pTregs found In non-lymphoid tissues, such as the colon, and under steady state conditions account for most pTregs in the intestine [254].
Tregs are activated at much lower antigen/MHC concentrations than naïve T-cells, ensuring Treg-dependent self-tolerance [255]. Tregs are a frequent immune cell population in the intestine, where they limit inflammatory CD4 T-cells [256,257] and maintain immune homeostasis through several mechanisms [258]. FoxP3+ Tregs, especially effector Tregs, are constantly proliferating under steady state conditions, likely as a consequence of identifying self-antigens and antigens derived from commensal microbes [259,260].
The suppressor activity of Tregs is mostly mediated by cell-contact–dependent and humoral-factor–mediated mechanisms. These mechanisms include IL-2 scavenging; the secretion of regulatory cytokines, such as IL-10, [261], IL-35 [262], and TGF-β [263,264]; the surface expression of inhibitory molecules, such as CTLA-4 (cytotoxic lymphocyte antigen 4) and PD-1 (programmed cell death 1), TIGIT (T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibition motif domains), CD39, and CD73 [265,266]; cytolysis; and metabolic control [238]. Tregs also promote tissue through the release of growth factor amphiregulin [267].
Treg numbers are increased in the inflamed tissue of IBD patients [268]. This increase may be the result of a compensatory mechanism to control the exacerbated proinflammatory immune response, but would seem to imply that Tregs are inefficient at suppression, since transient FOXP3 expression has been observed in human activated non-regulatory CD4 T-cells [253]. Recent single-cell RNA sequencing (scRNAseq) studies have provided a more detailed picture of the cell populations, including Tregs, in the healthy [269] and inflamed tissue of IBD patients and mice [48,270,271,272,273,274,275,276,277]. The inflamed epithelium of CD patients contains depleted numbers of Tregs, CD8 T-cells, γδT cells, and Tfh cells and elevated numbers of activated Th17 cells, as revealed by scRNAseq and multi-parameter flow cytometry or mass cytometry experiments [271]. scRNAseq has also revealed the persistence and expansion of CTLA-4+ Tregs in patients with checkpoint inhibitor-induced colitis [278]. In another study, inflamed tissue from UC patients was found to contain increased numbers of Tregs expressing FOXP3 and basic leucine zipper ATF-like transcription factor (BATF) and IL1B/LYZ+ myeloid cells [279]. These contradictory results illustrate that Treg changes remain unclear, and hint at a heterogeneous response among different Treg cell subsets in IBD. For example, inflamed tissue from CD patients contains increased numbers of RORγt+FOXP3+ Tregs, which secrete IL-17 and IFNγ while maintaining their suppressive function [280]. A similar population has been detected in UC patients [277]. It would thus seem that, although the lineage stability of Tregs allows them to maintain suppressive capacity and FoxP3 production despite exposure to inflammatory stimuli [281], Tregs can alter their phenotype by expressing transcription factors and chemokine receptors without producing inflammatory cytokines, helping them to arrive at the inflammation site where they can exert their suppressive effect on target T effector cells [282]. Through this mechanism, Tregs can acquire phenotypes similar to Th1 [283,284], Th2 [285,286,287], Th17 [288,289,290], or Tfh cells [291,292,293].
In the intestine, Tregs can acquire several phenotypes expressing varying levels of GATA3, Helios, and RORγt. GATA3+Helios+ Tregs seem to have a thymic origin and react to the alarmin IL-33 produced in response to tissue damage, reducing tissue injury in colitis [294]. RORγt+Helios− Tregs, produced in response to intestinal microbiota, are considered pTregs and play a protective role in severe gut inflammation [289,290]. RORγt−Helios− Tregs are more abundant in the small intestine and participate in the amelioration of allergic responses to food antigens [295]. These observations indicate that Tregs are highly versatile cells that adapt to their environment in order to better contribute to tissue homeostasis. There is some interest in developing therapies to boost Treg cell number and function and thereby reduce intestinal inflammation in IBD [254,296,297].
Other CD4 T-cell subsets include Foxp3- type 1 regulatory T (Tr1) cells, which secrete the suppressive cytokines IL-10 and TGF β [26,298], and Tfh cells, which are a specialized CD4 T-cell subset involved in the induction and differentiation of B cells into plasma cells and memory cells [299,300,301], cell subsets whose role in IBD has recently been reviewed [26,254,302,303].

9. Conclusions

Current IBD therapy is making great strides in improving patient quality of life. However, the high phenotypic variability of these patients means that, in many cases, treatments fall short of their objectives or simply do not work. Recent technological advances have contributed to a more detailed knowledge of the cell types involved in the pathophysiology of IBD, revealing the wide range of different phenotypic T-cell subsets and plasticity between them. Understanding the mechanisms that control changes between these phenotypes and determine the disease-promoting or disease-alleviating behaviors of these different cell subsets could lead to more specific therapies for each patient and each stage of the disease.

Author Contributions

Conceptualization, J.M.G.-G.; writing, figure design, and editing, R.G.-B., A.S., B.H.-F., C.R., H.S.-M. and J.M.G.-G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Instituto de Salud Carlos III (ISCIII) (PI20/00306) with co-funding from the European Regional Development Fund (ERDF) “A way to build Europe”. The CNIC is supported by the ISCIII, the Ministerio de Ciencia, Innovación y Universidades (MCNU), and the Pro CNIC Foundation. B.H.-F. and R.G.-B. by the UAM and the MCNU FPU program (FPU18/00895, FPU19/01774); A.S. by Universidad Francisco de Vitoria; and H.S-M. by the Comunidad de Madrid YEI program (PEJ-2020-TL/BMD-17604).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created in this study. All the data reported in this review were found in original articles cited in the text.

Acknowledgments

The authors thank S. Bartlett for English editing. A.S. and R.G.-B. contributed equally to this manuscript. Parts of the figures were drawn by using pictures from Servier Medical Art. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/ accessed on 25 November 2022). Authors thank Servier Medical Art for the creation of a great repertoire of medical images, under a CC BY 3.0 Unported License.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AHRAryl hydrocarbon receptor
AMPAntimicrobial peptide
APCAntigen presenting cell
BATFBasic leucine zipper ATF-like transcription factor
CCLChemokine (C-C motif) ligand
CCRC-C motif chemokine receptor
CDCrohn’s disease
CD5LCD5 molecule like
CTLCytotoxic T cells
CTLA-4Cytotoxic lymphocyte antigen 4
CXCLChemokine (C-X-C motif) ligand
DAMPsDamage-associated molecular patterns
DCsDendritic cells
DRDeath receptor
DSSDextran sodium sulfate
EpCAMEpithelial cell adhesion molecule
Foxp3Forkhead box P3
GATA-3GATA binding protein 3
G-CSFGranulocyte colony-stimulating factor
GM-CSFGranulocyte-macrophage colony-stimulating factor
IBDInflammatory bowel disease
IECIntestinal epithelial cells
IFNInterferon
IgImmunoglobulin
ILCsInnate lymphoid cells
ILInterleukin
IL-12Rβ2IL-12 receptor subunit β2
IL-22BPIL-22 binding protein
IRFInterferon regulatory factor
ISCIntestinal stem cell
JAKActivates janus kinase
KLRG1Killer cell lectin like receptor G1
KOKnockout
MHCMajor histocompatibility complex
MMPsMatrix metalloproteinases
NKNatural killer
NKG2DNatural killer group 2D receptor
NLRNod-like receptors
PAMPsPathogen-associated molecular patterns
PD-1Programmed cell death 1
PRRsPattern recognition receptors
RAGRecombination activating gene
RORγtRetinoic acid receptor-related orphan receptor gamma t
scRNA-seqSingle cell RNA sequencing
STATSignal transducer and activator of transcription
T-bet T-box-containing protein
TCRT cell antigen receptor
TfhT follicular helper
TGFTransforming growth factor
ThT helper
TIGITT cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibition motif domains
TLTNF-like factor
TL1ATNF-like ligand 1 A
TLRsToll-like receptors
TNBS2,4,6-trinitrobenzene sulfonic acid
TNFTumor necrosis factor
TregT regulatory
Tr1T regulatory type 1
UCUlcerative colitis

References

  1. Herrero-Fernandez, B.; Gomez-Bris, R.; Somovilla-Crespo, B.; Gonzalez-Granado, J.M. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int. J. Mol. Sci. 2019, 20, 5293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Yao, H.; Tang, G. Macrophages in intestinal fibrosis and regression. Cell. Immunol. 2022, 381, 104614. [Google Scholar] [CrossRef] [PubMed]
  3. Katsandegwaza, B.; Horsnell, W.; Smith, K. Inflammatory Bowel Disease: A Review of Pre-Clinical Murine Models of Human Disease. Int. J. Mol. Sci. 2022, 23, 9344. [Google Scholar] [CrossRef] [PubMed]
  4. Kang, L.; Fang, X.; Song, Y.H.; He, Z.X.; Wang, Z.J.; Wang, S.L.; Li, Z.S.; Bai, Y. Neutrophil-Epithelial Crosstalk During Intestinal Inflammation. Cell. Mol. Gastroenterol. Hepatol. 2022, 14, 1257–1267. [Google Scholar] [CrossRef]
  5. Kaluzna, A.; Olczyk, P.; Komosinska-Vassev, K. The Role of Innate and Adaptive Immune Cells in the Pathogenesis and Development of the Inflammatory Response in Ulcerative Colitis. J. Clin. Med. 2022, 11, 400. [Google Scholar] [CrossRef]
  6. He, X.; Tan, S.; Shao, Z.; Wang, X. Latitudinal and longitudinal regulation of tissue macrophages in inflammatory diseases. Genes Dis. 2022, 9, 1194–1207. [Google Scholar] [CrossRef]
  7. Ghilas, S.; O’Keefe, R.; Mielke, L.A.; Raghu, D.; Buchert, M.; Ernst, M. Crosstalk between epithelium, myeloid and innate lymphoid cells during gut homeostasis and disease. Front. Immunol. 2022, 13, 944982. [Google Scholar] [CrossRef]
  8. Wardell, C.M.; MacDonald, K.N.; Levsings, M.K.; Cook, L. Cross talk between human regulatory T cells and antigen-presenting cells: Lessons for clinical applications. Eur. J. Immunol. 2021, 51, 27–38. [Google Scholar] [CrossRef]
  9. Schulz-Kuhnt, A.; Neurath, M.F.; Wirtz, S.; Atreya, I. Innate Lymphoid Cells as Regulators of Epithelial Integrity: Therapeutic Implications for Inflammatory Bowel Diseases. Front. Med. (Lausanne) 2021, 8, 656745. [Google Scholar] [CrossRef]
  10. Saez, A.; Gomez-Bris, R.; Herrero-Fernandez, B.; Mingorance, C.; Rius, C.; Gonzalez-Granado, J.M. Innate Lymphoid Cells in Intestinal Homeostasis and Inflammatory Bowel Disease. Int. J. Mol. Sci. 2021, 22, 7618. [Google Scholar] [CrossRef]
  11. Peloquin, J.M.; Goel, G.; Villablanca, E.J.; Xavier, R.J. Mechanisms of Pediatric Inflammatory Bowel Disease. Annu. Rev. Immunol. 2016, 34, 31–64. [Google Scholar] [CrossRef]
  12. Liu, T.C.; Stappenbeck, T.S. Genetics and Pathogenesis of Inflammatory Bowel Disease. Annu. Rev. Pathol. 2016, 11, 127–148. [Google Scholar] [CrossRef] [Green Version]
  13. Torres, J.; Mehandru, S.; Colombel, J.F.; Peyrin-Biroulet, L. Crohn’s disease. Lancet 2017, 389, 1741–1755. [Google Scholar] [CrossRef]
  14. Ungaro, R.; Mehandru, S.; Allen, P.B.; Peyrin-Biroulet, L.; Colombel, J.F. Ulcerative colitis. Lancet 2017, 389, 1756–1770. [Google Scholar] [CrossRef]
  15. Kaplan, G.G.; Windsor, J.W. The four epidemiological stages in the global evolution of inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 56–66. [Google Scholar] [CrossRef]
  16. Olen, O.; Askling, J.; Sachs, M.C.; Neovius, M.; Smedby, K.E.; Ekbom, A.; Ludvigsson, J.F. Mortality in adult-onset and elderly-onset IBD: A nationwide register-based cohort study 1964–2014. Gut 2020, 69, 453–461. [Google Scholar] [CrossRef]
  17. Peyrin-Biroulet, L.; Panes, J.; Sandborn, W.J.; Vermeire, S.; Danese, S.; Feagan, B.G.; Colombel, J.F.; Hanauer, S.B.; Rycroft, B. Defining Disease Severity in Inflammatory Bowel Diseases: Current and Future Directions. Clin. Gastroenterol. Hepatol. 2016, 14, 348–354.e17. [Google Scholar] [CrossRef] [Green Version]
  18. Pabla, B.S.; Schwartz, D.A. Assessing Severity of Disease in Patients with Ulcerative Colitis. Gastroenterol. Clin. N. Am. 2020, 49, 671–688. [Google Scholar] [CrossRef]
  19. Cleynen, I.; Gonzalez, J.R.; Figueroa, C.; Franke, A.; McGovern, D.; Bortlik, M.; Crusius, B.J.; Vecchi, M.; Artieda, M.; Szczypiorska, M.; et al. Genetic factors conferring an increased susceptibility to develop Crohn’s disease also influence disease phenotype: Results from the IBDchip European Project. Gut 2013, 62, 1556–1565. [Google Scholar] [CrossRef] [Green Version]
  20. Satsangi, J.; Silverberg, M.S.; Vermeire, S.; Colombel, J.F. The Montreal classification of inflammatory bowel disease: Controversies, consensus, and implications. Gut 2006, 55, 749–753. [Google Scholar] [CrossRef]
  21. Louis, E.; Van Kemseke, C.; Reenaers, C. Necessity of phenotypic classification of inflammatory bowel disease. Best Pract. Res. Clin. Gastroenterol. 2011, 25 (Suppl. 1), S2–S7. [Google Scholar] [CrossRef] [PubMed]
  22. Levine, A.; Griffiths, A.; Markowitz, J.; Wilson, D.C.; Turner, D.; Russell, R.K.; Fell, J.; Ruemmele, F.M.; Walters, T.; Sherlock, M.; et al. Pediatric modification of the Montreal classification for inflammatory bowel disease: The Paris classification. Inflamm. Bowel Dis. 2011, 17, 1314–1321. [Google Scholar] [CrossRef] [PubMed]
  23. Gecse, K.B.; Vermeire, S. Differential diagnosis of inflammatory bowel disease: Imitations and complications. Lancet Gastroenterol. Hepatol. 2018, 3, 644–653. [Google Scholar] [CrossRef]
  24. de Souza, H.S.P.; Fiocchi, C.; Iliopoulos, D. The IBD interactome: An integrated view of aetiology, pathogenesis and therapy. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 739–749. [Google Scholar] [CrossRef] [PubMed]
  25. Choy, M.C.; Visvanathan, K.; De Cruz, P. An Overview of the Innate and Adaptive Immune System in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2017, 23, 2–13. [Google Scholar] [CrossRef]
  26. Tindemans, I.; Joosse, M.E.; Samsom, J.N. Dissecting the Heterogeneity in T-Cell Mediated Inflammation in IBD. Cells 2020, 9, 110. [Google Scholar] [CrossRef] [Green Version]
  27. Giuffrida, P.; Di Sabatino, A. Targeting T cells in inflammatory bowel disease. Pharmacol. Res. 2020, 159, 105040. [Google Scholar] [CrossRef]
  28. Zenewicz, L.A.; Antov, A.; Flavell, R.A. CD4 T-cell differentiation and inflammatory bowel disease. Trends Mol. Med. 2009, 15, 199–207. [Google Scholar] [CrossRef]
  29. Friedrich, M.; Pohin, M.; Powrie, F. Cytokine Networks in the Pathophysiology of Inflammatory Bowel Disease. Immunity 2019, 50, 992–1006. [Google Scholar] [CrossRef] [Green Version]
  30. Cao, H.; Diao, J.; Liu, H.; Liu, S.; Liu, J.; Yuan, J.; Lin, J. The Pathogenicity and Synergistic Action of Th1 and Th17 Cells in Inflammatory Bowel Diseases. Inflamm. Bowel Dis. 2022, izac199. [Google Scholar] [CrossRef]
  31. Zhang, N.; Bevan, M.J. CD8(+) T cells: Foot soldiers of the immune system. Immunity 2011, 35, 161–168. [Google Scholar] [CrossRef] [Green Version]
  32. Sakaguchi, S.; Miyara, M.; Costantino, C.M.; Hafler, D.A. FOXP3+ regulatory T cells in the human immune system. Nat. Rev. Immunol. 2010, 10, 490–500. [Google Scholar] [CrossRef]
  33. Elahi, S.; Horton, H. Association of HLA-alleles with the immune regulation of chronic viral infections. Int. J. Biochem. Cell Biol. 2012, 44, 1361–1365. [Google Scholar] [CrossRef] [Green Version]
  34. Morel, P.A. Differential T-cell receptor signals for T helper cell programming. Immunology 2018, 155, 63–71. [Google Scholar] [CrossRef] [Green Version]
  35. Lee, G.R.; Kim, S.T.; Spilianakis, C.G.; Fields, P.E.; Flavell, R.A. T helper cell differentiation: Regulation by cis elements and epigenetics. Immunity 2006, 24, 369–379. [Google Scholar] [CrossRef] [Green Version]
  36. Bustos-Moran, E.; Blas-Rus, N.; Martin-Cofreces, N.B.; Sanchez-Madrid, F. Orchestrating Lymphocyte Polarity in Cognate Immune Cell-Cell Interactions. Int. Rev. Cell Mol. Biol. 2016, 327, 195–261. [Google Scholar]
  37. Hirahara, K.; Nakayama, T. CD4+ T-cell subsets in inflammatory diseases: Beyond the Th1/Th2 paradigm. Int. Immunol. 2016, 28, 163–171. [Google Scholar] [CrossRef] [Green Version]
  38. DuPage, M.; Bluestone, J.A. Harnessing the plasticity of CD4(+) T cells to treat immune-mediated disease. Nat. Rev. Immunol. 2016, 16, 149–163. [Google Scholar] [CrossRef] [Green Version]
  39. Li, P.; Spolski, R.; Liao, W.; Leonard, W.J. Complex interactions of transcription factors in mediating cytokine biology in T cells. Immunol. Rev. 2014, 261, 141–156. [Google Scholar] [CrossRef] [Green Version]
  40. Mosmann, T.R.; Cherwinski, H.; Bond, M.W.; Giedlin, M.A.; Coffman, R.L. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J. Immunol. 1986, 136, 2348–2357. [Google Scholar] [CrossRef]
  41. Allaire, J.M.; Crowley, S.M.; Law, H.T.; Chang, S.Y.; Ko, H.J.; Vallance, B.A. The Intestinal Epithelium: Central Coordinator of Mucosal Immunity. Trends Immunol. 2018, 39, 677–696. [Google Scholar] [CrossRef] [PubMed]
  42. Basso, L.; Boue, J.; Auge, C.; Deraison, C.; Blanpied, C.; Cenac, N.; Lluel, P.; Vergnolle, N.; Dietrich, G. Mobilization of CD4+ T lymphocytes in inflamed mucosa reduces pain in colitis mice: Toward a vaccinal strategy to alleviate inflammatory visceral pain. Pain 2018, 159, 331–341. [Google Scholar] [CrossRef] [PubMed]
  43. Hirata, I.; Berrebi, G.; Austin, L.L.; Keren, D.F.; Dobbins, W.O., 3rd. Immunohistological characterization of intraepithelial and lamina propria lymphocytes in control ileum and colon and in inflammatory bowel disease. Dig. Dis. Sci. 1986, 31, 593–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Selby, W.S.; Janossy, G.; Bofill, M.; Jewell, D.P. Intestinal lymphocyte subpopulations in inflammatory bowel disease: An analysis by immunohistological and cell isolation techniques. Gut 1984, 25, 32–40. [Google Scholar] [CrossRef] [Green Version]
  45. Schreiber, S.; MacDermott, R.P.; Raedler, A.; Pinnau, R.; Bertovich, M.J.; Nash, G.S. Increased activation of isolated intestinal lamina propria mononuclear cells in inflammatory bowel disease. Gastroenterology 1991, 101, 1020–1030. [Google Scholar] [CrossRef]
  46. Rabe, H.; Malmquist, M.; Barkman, C.; Ostman, S.; Gjertsson, I.; Saalman, R.; Wold, A.E. Distinct patterns of naive, activated and memory T and B cells in blood of patients with ulcerative colitis or Crohn’s disease. Clin. Exp. Immunol. 2019, 197, 111–129. [Google Scholar] [CrossRef] [Green Version]
  47. Muller, S.; Lory, J.; Corazza, N.; Griffiths, G.M.; Z’Graggen, K.; Mazzucchelli, L.; Kappeler, A.; Mueller, C. Activated CD4+ and CD8+ cytotoxic cells are present in increased numbers in the intestinal mucosa from patients with active inflammatory bowel disease. Am. J. Pathol. 1998, 152, 261–268. [Google Scholar]
  48. Corridoni, D.; Antanaviciute, A.; Gupta, T.; Fawkner-Corbett, D.; Aulicino, A.; Jagielowicz, M.; Parikh, K.; Repapi, E.; Taylor, S.; Ishikawa, D.; et al. Single-cell atlas of colonic CD8(+) T cells in ulcerative colitis. Nat. Med. 2020, 26, 1480–1490. [Google Scholar] [CrossRef]
  49. Page, M.J.; Poritz, L.S.; Tilberg, A.F.; Zhang, W.J.; Chorney, M.J.; Koltun, W.A. Cd1d-restricted cellular lysis by peripheral blood lymphocytes: Relevance to the inflammatory bowel diseases. J. Surg. Res. 2000, 92, 214–221. [Google Scholar] [CrossRef]
  50. Allez, M.; Tieng, V.; Nakazawa, A.; Treton, X.; Pacault, V.; Dulphy, N.; Caillat-Zucman, S.; Paul, P.; Gornet, J.M.; Douay, C.; et al. CD4+NKG2D+ T cells in Crohn’s disease mediate inflammatory and cytotoxic responses through MICA interactions. Gastroenterology 2007, 132, 2346–2358. [Google Scholar] [CrossRef]
  51. Pariente, B.; Mocan, I.; Camus, M.; Dutertre, C.A.; Ettersperger, J.; Cattan, P.; Gornet, J.M.; Dulphy, N.; Charron, D.; Lemann, M.; et al. Activation of the receptor NKG2D leads to production of Th17 cytokines in CD4+ T cells of patients with Crohn’s disease. Gastroenterology 2011, 141, 217–226.e2. [Google Scholar] [CrossRef]
  52. de Souza, H.S.; Fiocchi, C. Immunopathogenesis of IBD: Current state of the art. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 13–27. [Google Scholar] [CrossRef]
  53. Mahapatro, M.; Erkert, L.; Becker, C. Cytokine-Mediated Crosstalk between Immune Cells and Epithelial Cells in the Gut. Cells 2021, 10, 111. [Google Scholar] [CrossRef]
  54. Raphael, I.; Nalawade, S.; Eagar, T.N.; Forsthuber, T.G. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine 2015, 74, 5–17. [Google Scholar] [CrossRef] [Green Version]
  55. Caza, T.; Landas, S. Functional and Phenotypic Plasticity of CD4(+) T Cell Subsets. Biomed. Res. Int. 2015, 2015, 521957. [Google Scholar] [CrossRef] [Green Version]
  56. Lazarevic, V.; Glimcher, L.H.; Lord, G.M. T-bet: A bridge between innate and adaptive immunity. Nat. Rev. Immunol. 2013, 13, 777–789. [Google Scholar] [CrossRef]
  57. Wang, P.; Zhang, Q.; Tan, L.; Xu, Y.; Xie, X.; Zhao, Y. The Regulatory Effects of mTOR Complexes in the Differentiation and Function of CD4(+) T Cell Subsets. J. Immunol. Res. 2020, 2020, 3406032. [Google Scholar] [CrossRef] [Green Version]
  58. Baumann, C.; Bonilla, W.V.; Frohlich, A.; Helmstetter, C.; Peine, M.; Hegazy, A.N.; Pinschewer, D.D.; Lohning, M. T-bet- and STAT4-dependent IL-33 receptor expression directly promotes antiviral Th1 cell responses. Proc. Natl. Acad. Sci. USA 2015, 112, 4056–4061. [Google Scholar] [CrossRef] [Green Version]
  59. Szabo, S.J.; Kim, S.T.; Costa, G.L.; Zhang, X.; Fathman, C.G.; Glimcher, L.H. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 2000, 100, 655–669. [Google Scholar] [CrossRef] [Green Version]
  60. Yang, R.; Mele, F.; Worley, L.; Langlais, D.; Rosain, J.; Benhsaien, I.; Elarabi, H.; Croft, C.A.; Doisne, J.M.; Zhang, P.; et al. Human T-bet Governs Innate and Innate-like Adaptive IFN-gamma Immunity against Mycobacteria. Cell 2020, 183, 1826–1847.e31. [Google Scholar] [CrossRef]
  61. Wu, X.; Tian, J.; Wang, S. Insight Into Non-Pathogenic Th17 Cells in Autoimmune Diseases. Front. Immunol. 2018, 9, 1112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Leppkes, M.; Neurath, M.F. Cytokines in inflammatory bowel diseases—Update 2020. Pharmacol. Res. 2020, 158, 104835. [Google Scholar] [CrossRef] [PubMed]
  63. Li, J.; Ueno, A.; Fort Gasia, M.; Luider, J.; Wang, T.; Hirota, C.; Jijon, H.B.; Deane, M.; Tom, M.; Chan, R.; et al. Profiles of Lamina Propria T Helper Cell Subsets Discriminate Between Ulcerative Colitis and Crohn’s Disease. Inflamm. Bowel Dis. 2016, 22, 1779–1792. [Google Scholar] [CrossRef] [PubMed]
  64. Ma, C.; Wu, W.; Lin, R.; Ge, Y.; Zhang, C.; Sun, S.; Cong, Y.; Li, X.; Liu, Z. Critical Role of CD6highCD4+ T Cells in Driving Th1/Th17 Cell Immune Responses and Mucosal Inflammation in IBD. J. Crohns Colitis 2019, 13, 510–524. [Google Scholar] [CrossRef]
  65. Monteleone, G.; Trapasso, F.; Parrello, T.; Biancone, L.; Stella, A.; Iuliano, R.; Luzza, F.; Fusco, A.; Pallone, F. Bioactive IL-18 expression is up-regulated in Crohn’s disease. J. Immunol. 1999, 163, 143–147. [Google Scholar] [CrossRef]
  66. Monteleone, G.; Biancone, L.; Marasco, R.; Morrone, G.; Marasco, O.; Luzza, F.; Pallone, F. Interleukin 12 is expressed and actively released by Crohn’s disease intestinal lamina propria mononuclear cells. Gastroenterology 1997, 112, 1169–1178. [Google Scholar] [CrossRef]
  67. Zorzi, F.; Monteleone, I.; Sarra, M.; Calabrese, E.; Marafini, I.; Cretella, M.; Sedda, S.; Biancone, L.; Pallone, F.; Monteleone, G. Distinct profiles of effector cytokines mark the different phases of Crohn’s disease. PLoS ONE 2013, 8, e54562. [Google Scholar] [CrossRef]
  68. Zimmermann, J.; Kuhl, A.A.; Weber, M.; Grun, J.R.; Loffler, J.; Haftmann, C.; Riedel, R.; Maschmeyer, P.; Lehmann, K.; Westendorf, K.; et al. T-bet expression by Th cells promotes type 1 inflammation but is dispensable for colitis. Mucosal Immunol. 2016, 9, 1487–1499. [Google Scholar] [CrossRef] [Green Version]
  69. Soderquest, K.; Hertweck, A.; Giambartolomei, C.; Henderson, S.; Mohamed, R.; Goldberg, R.; Perucha, E.; Franke, L.; Herrero, J.; Plagnol, V.; et al. Genetic variants alter T-bet binding and gene expression in mucosal inflammatory disease. PLoS Genet. 2017, 13, e1006587. [Google Scholar] [CrossRef] [Green Version]
  70. Alfen, J.S.; Larghi, P.; Facciotti, F.; Gagliani, N.; Bosotti, R.; Paroni, M.; Maglie, S.; Gruarin, P.; Vasco, C.M.; Ranzani, V.; et al. Intestinal IFN-gamma-producing type 1 regulatory T cells coexpress CCR5 and programmed cell death protein 1 and downregulate IL-10 in the inflamed guts of patients with inflammatory bowel disease. J. Allergy Clin. Immunol. 2018, 142, 1537–1547.e8. [Google Scholar] [CrossRef] [Green Version]
  71. Li, X.; Wang, Q.; Ding, L.; Wang, Y.X.; Zhao, Z.D.; Mao, N.; Wu, C.T.; Wang, H.; Zhu, H.; Ning, S.B. Intercellular adhesion molecule-1 enhances the therapeutic effects of MSCs in a dextran sulfate sodium-induced colitis models by promoting MSCs homing to murine colons and spleens. Stem Cell Res. Ther. 2019, 10, 267. [Google Scholar] [CrossRef]
  72. Ziegler, A.; Heidenreich, R.; Braumuller, H.; Wolburg, H.; Weidemann, S.; Mocikat, R.; Rocken, M. EpCAM, a human tumor-associated antigen promotes Th2 development and tumor immune evasion. Blood 2009, 113, 3494–3502. [Google Scholar] [CrossRef]
  73. Atanackovic, D.; Reinhard, H.; Meyer, S.; Spock, S.; Grob, T.; Luetkens, T.; Yousef, S.; Cao, Y.; Hildebrandt, Y.; Templin, J.; et al. The trifunctional antibody catumaxomab amplifies and shapes tumor-specific immunity when applied to gastric cancer patients in the adjuvant setting. Hum. Vaccin. Immunother. 2013, 9, 2533–2542. [Google Scholar] [CrossRef]
  74. Mashimo, M.; Fujii, T.; Ono, S.; Moriwaki, Y.; Misawa, H.; Kawashima, K. Minireview: Divergent roles of alpha7 nicotinic acetylcholine receptors expressed on antigen-presenting cells and CD4(+) T cells in the regulation of T cell differentiation. Int. Immunopharmacol. 2020, 82, 106306. [Google Scholar] [CrossRef]
  75. Fuss, I.J.; Heller, F.; Boirivant, M.; Leon, F.; Yoshida, M.; Fichtner-Feigl, S.; Yang, Z.; Exley, M.; Kitani, A.; Blumberg, R.S.; et al. Nonclassical CD1d-restricted NK T cells that produce IL-13 characterize an atypical Th2 response in ulcerative colitis. J. Clin. Investig. 2004, 113, 1490–1497. [Google Scholar] [CrossRef] [Green Version]
  76. Heller, F.; Florian, P.; Bojarski, C.; Richter, J.; Christ, M.; Hillenbrand, B.; Mankertz, J.; Gitter, A.H.; Burgel, N.; Fromm, M.; et al. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology 2005, 129, 550–564. [Google Scholar] [CrossRef]
  77. Camoglio, L.; Te Velde, A.A.; Tigges, A.J.; Das, P.K.; Van Deventer, S.J. Altered expression of interferon-gamma and interleukin-4 in inflammatory bowel disease. Inflamm. Bowel Dis. 1998, 4, 285–290. [Google Scholar] [CrossRef]
  78. Fuss, I.J.; Neurath, M.; Boirivant, M.; Klein, J.S.; de la Motte, C.; Strong, S.A.; Fiocchi, C.; Strober, W. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn’s disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J. Immunol. 1996, 157, 1261–1270. [Google Scholar] [CrossRef]
  79. Biancheri, P.; Di Sabatino, A.; Ammoscato, F.; Facciotti, F.; Caprioli, F.; Curciarello, R.; Hoque, S.S.; Ghanbari, A.; Joe-Njoku, I.; Giuffrida, P.; et al. Absence of a role for interleukin-13 in inflammatory bowel disease. Eur. J. Immunol. 2014, 44, 370–385. [Google Scholar] [CrossRef] [Green Version]
  80. Rovedatti, L.; Kudo, T.; Biancheri, P.; Sarra, M.; Knowles, C.H.; Rampton, D.S.; Corazza, G.R.; Monteleone, G.; Di Sabatino, A.; Macdonald, T.T. Differential regulation of interleukin 17 and interferon gamma production in inflammatory bowel disease. Gut 2009, 58, 1629–1636. [Google Scholar] [CrossRef]
  81. Yan, J.; Pandey, S.P.; Barnes, B.J.; Turner, J.R.; Abraham, C. T Cell-Intrinsic IRF5 Regulates T Cell Signaling, Migration, and Differentiation and Promotes Intestinal Inflammation. Cell Rep. 2020, 31, 107820. [Google Scholar] [CrossRef] [PubMed]
  82. Rocha-Perugini, V.; Gonzalez-Granado, J.M. Nuclear envelope lamin-A as a coordinator of T cell activation. Nucleus 2014, 5, 396–401. [Google Scholar] [CrossRef] [PubMed]
  83. Gonzalez-Granado, J.M.; Silvestre-Roig, C.; Rocha-Perugini, V.; Trigueros-Motos, L.; Cibrian, D.; Morlino, G.; Blanco-Berrocal, M.; Osorio, F.G.; Freije, J.M.P.; Lopez-Otin, C.; et al. Nuclear envelope lamin-A couples actin dynamics with immunological synapse architecture and T cell activation. Sci. Signal. 2014, 7, ra37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Toribio-Fernandez, R.; Zorita, V.; Rocha-Perugini, V.; Iborra, S.; Martinez Del Hoyo, G.; Chevre, R.; Dorado, B.; Sancho, D.; Sanchez-Madrid, F.; Andres, V.; et al. Lamin A/C augments Th1 differentiation and response against vaccinia virus and Leishmania major. Cell Death Dis. 2018, 9, 9. [Google Scholar] [CrossRef] [Green Version]
  85. Saez, A.; Herrero-Fernandez, B.; Gomez-Bris, R.; Somovilla-Crespo, B.; Rius, C.; Gonzalez-Granado, J.M. Lamin A/C and the Immune System: One Intermediate Filament, Many Faces. Int. J. Mol. Sci. 2020, 21, 6109. [Google Scholar] [CrossRef]
  86. Toribio-Fernandez, R.; Herrero-Fernandez, B.; Zorita, V.; Lopez, J.A.; Vazquez, J.; Criado, G.; Pablos, J.L.; Collas, P.; Sanchez-Madrid, F.; Andres, V.; et al. Lamin A/C deficiency in CD4(+) T-cells enhances regulatory T-cells and prevents inflammatory bowel disease. J. Pathol. 2019, 249, 509–522. [Google Scholar] [CrossRef]
  87. Cepek, K.L.; Parker, C.M.; Madara, J.L.; Brenner, M.B. Integrin alpha E beta 7 mediates adhesion of T lymphocytes to epithelial cells. J. Immunol. 1993, 150 Pt 1, 3459–3470. [Google Scholar] [CrossRef]
  88. Lamb, C.A.; Mansfield, J.C.; Tew, G.W.; Gibbons, D.; Long, A.K.; Irving, P.; Diehl, L.; Eastham-Anderson, J.; Price, M.B.; O’Boyle, G.; et al. alphaEbeta7 Integrin Identifies Subsets of Pro-Inflammatory Colonic CD4+ T Lymphocytes in Ulcerative Colitis. J. Crohns Colitis 2017, 11, 610–620. [Google Scholar]
  89. Di Sabatino, A.; Rovedatti, L.; Kaur, R.; Spencer, J.P.; Brown, J.T.; Morisset, V.D.; Biancheri, P.; Leakey, N.A.; Wilde, J.I.; Scott, L.; et al. Targeting gut T cell Ca2+ release-activated Ca2+ channels inhibits T cell cytokine production and T-box transcription factor T-bet in inflammatory bowel disease. J. Immunol. 2009, 183, 3454–3462. [Google Scholar] [CrossRef] [Green Version]
  90. Zeng, W.P. ‘All things considered’: Transcriptional regulation of T helper type 2 cell differentiation from precursor to effector activation. Immunology 2013, 140, 31–38. [Google Scholar] [CrossRef]
  91. Zheng, W.; Flavell, R.A. The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells. Cell 1997, 89, 587–596. [Google Scholar] [CrossRef] [Green Version]
  92. Zhang, D.H.; Cohn, L.; Ray, P.; Bottomly, K.; Ray, A. Transcription factor GATA-3 is differentially expressed in murine Th1 and Th2 cells and controls Th2-specific expression of the interleukin-5 gene. J. Biol. Chem. 1997, 272, 21597–21603. [Google Scholar] [CrossRef] [Green Version]
  93. Bryant, N.; Muehling, L.M. T-cell responses in asthma exacerbations. Ann. Allergy Asthma Immunol. 2022, 129, 709–718. [Google Scholar] [CrossRef]
  94. Shankar, A.; McAlees, J.W.; Lewkowich, I.P. Modulation of IL-4/IL-13 cytokine signaling in the context of allergic disease. J. Allergy Clin. Immunol. 2022, 150, 266–276. [Google Scholar] [CrossRef]
  95. Luo, W.; Hu, J.; Xu, W.; Dong, J. Distinct spatial and temporal roles for Th1, Th2, and Th17 cells in asthma. Front. Immunol. 2022, 13, 974066. [Google Scholar] [CrossRef]
  96. Habib, N.; Pasha, M.A.; Tang, D.D. Current Understanding of Asthma Pathogenesis and Biomarkers. Cells 2022, 11, 2764. [Google Scholar] [CrossRef]
  97. Salvati, L.; Liotta, F.; Annunziato, F.; Cosmi, L. Therapeutical Targets in Allergic Inflammation. Biomedicines 2022, 10, 2874. [Google Scholar] [CrossRef]
  98. Durham, S.R.; Shamji, M.H. Allergen immunotherapy: Past, present and future. Nat. Rev. Immunol. 2022, 1–12. [Google Scholar] [CrossRef]
  99. Boirivant, M.; Fuss, I.J.; Chu, A.; Strober, W. Oxazolone colitis: A murine model of T helper cell type 2 colitis treatable with antibodies to interleukin 4. J. Exp. Med. 1998, 188, 1929–1939. [Google Scholar] [CrossRef] [Green Version]
  100. Pushparaj, P.N.; Li, D.; Komai-Koma, M.; Guabiraba, R.; Alexander, J.; McSharry, C.; Xu, D. Interleukin-33 exacerbates acute colitis via interleukin-4 in mice. Immunology 2013, 140, 70–77. [Google Scholar] [CrossRef]
  101. Sedhom, M.A.K.; Pichery, M.; Murdoch, J.R.; Foligné, B.; Ortega, N.; Normand, S.; Mertz, K.; Sanmugalingam, D.; Brault, L.; Grandjean, T.; et al. Neutralisation of the interleukin-33/ST2 pathway ameliorates experimental colitis through enhancement of mucosal healing in mice. Gut 2013, 62, 1714–1723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Guan, Q.; Zhang, J. Recent Advances: The Imbalance of Cytokines in the Pathogenesis of Inflammatory Bowel Disease. Mediat. Inflamm. 2017, 2017, 4810258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Pastorelli, L.; De Salvo, C.; Vecchi, M.; Pizarro, T.T. The role of IL-33 in gut mucosal inflammation. Mediat. Inflamm. 2013, 2013, 608187. [Google Scholar] [CrossRef] [PubMed]
  104. Latiano, A.; Palmieri, O.; Pastorelli, L.; Vecchi, M.; Pizarro, T.T.; Bossa, F.; Merla, G.; Augello, B.; Latiano, T.; Corritore, G.; et al. Associations between genetic polymorphisms in IL-33, IL1R1 and risk for inflammatory bowel disease. PLoS ONE 2013, 8, e62144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Zhu, J.; Wang, Y.; Yang, F.; Sang, L.; Zhai, J.; Li, S.; Li, Y.; Wang, D.; Lu, C.; Sun, X. IL-33 alleviates DSS-induced chronic colitis in C57BL/6 mice colon lamina propria by suppressing Th17 cell response as well as Th1 cell response. Int. Immunopharmacol. 2015, 29, 846–853. [Google Scholar] [CrossRef]
  106. Zhu, J.; Xu, Y.; Zhu, C.; Zhao, J.; Meng, X.; Chen, S.; Wang, T.; Li, X.; Zhang, L.; Lu, C.; et al. IL-33 induces both regulatory B cells and regulatory T cells in dextran sulfate sodium-induced colitis. Int. Immunopharmacol. 2017, 46, 38–47. [Google Scholar] [CrossRef]
  107. Zhu, J.; Yang, F.; Sang, L.; Zhai, J.; Zhang, X.; Yue, D.; Li, S.; Li, Y.; Lu, C.; Sun, X. IL-33 Aggravates DSS-Induced Acute Colitis in Mouse Colon Lamina Propria by Enhancing Th2 Cell Responses. Mediat. Inflamm. 2015, 2015, 913041. [Google Scholar] [CrossRef] [Green Version]
  108. Monticelli, L.A.; Osborne, L.C.; Noti, M.; Tran, S.V.; Zaiss, D.M.; Artis, D. IL-33 promotes an innate immune pathway of intestinal tissue protection dependent on amphiregulin-EGFR interactions. Proc. Natl. Acad. Sci. USA 2015, 112, 10762–10767. [Google Scholar] [CrossRef] [Green Version]
  109. Duan, L.; Chen, J.; Zhang, H.; Yang, H.; Zhu, P.; Xiong, A.; Xia, Q.; Zheng, F.; Tan, Z.; Gong, F.; et al. Interleukin-33 ameliorates experimental colitis through promoting Th2/Foxp3(+) regulatory T-cell responses in mice. Mol. Med. 2012, 18, 753–761. [Google Scholar] [CrossRef]
  110. Reinisch, W.; Panes, J.; Khurana, S.; Toth, G.; Hua, F.; Comer, G.M.; Hinz, M.; Page, K.; O’Toole, M.; Moorehead, T.M.; et al. Anrukinzumab, an anti-interleukin 13 monoclonal antibody, in active UC: Efficacy and safety from a phase IIa randomised multicentre study. Gut 2015, 64, 894–900. [Google Scholar] [CrossRef]
  111. Danese, S.; Rudzinski, J.; Brandt, W.; Dupas, J.L.; Peyrin-Biroulet, L.; Bouhnik, Y.; Kleczkowski, D.; Uebel, P.; Lukas, M.; Knutsson, M.; et al. Tralokinumab for moderate-to-severe UC: A randomised, double-blind, placebo-controlled, phase IIa study. Gut 2015, 64, 243–249. [Google Scholar] [CrossRef]
  112. Zhu, J.; Xu, Y.; Li, Z.; Liu, S.; Fu, W.; Wei, Y. Interleukin-36beta exacerbates DSS-induce acute colitis via inhibiting Foxp3(+) regulatory T cell response and increasing Th2 cell response. Int. Immunopharmacol. 2022, 108, 108762. [Google Scholar] [CrossRef]
  113. Schmitt, E.; Bopp, T. Discovery and initial characterization of Th9 cells: The early years. Semin. Immunopathol. 2017, 39, 5–10. [Google Scholar] [CrossRef]
  114. Neurath, M.F.; Finotto, S. IL-9 signaling as key driver of chronic inflammation in mucosal immunity. Cytokine Growth Factor Rev. 2016, 29, 93–99. [Google Scholar] [CrossRef]
  115. Kaplan, M.H.; Schindler, U.; Smiley, S.T.; Grusby, M.J. Stat6 is required for mediating responses to IL-4 and for development of Th2 cells. Immunity 1996, 4, 313–319. [Google Scholar] [CrossRef] [Green Version]
  116. Lu, L.; Ma, J.; Wang, X.; Wang, J.; Zhang, F.; Yu, J.; He, G.; Xu, B.; Brand, D.D.; Horwitz, D.A.; et al. Synergistic effect of TGF-beta superfamily members on the induction of Foxp3+ Treg. Eur. J. Immunol. 2010, 40, 142–152. [Google Scholar] [CrossRef] [Green Version]
  117. Veldhoen, M.; Uyttenhove, C.; van Snick, J.; Helmby, H.; Westendorf, A.; Buer, J.; Martin, B.; Wilhelm, C.; Stockinger, B. Transforming growth factor-beta ‘reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nat. Immunol. 2008, 9, 1341–1346. [Google Scholar] [CrossRef]
  118. Dardalhon, V.; Awasthi, A.; Kwon, H.; Galileos, G.; Gao, W.; Sobel, R.A.; Mitsdoerffer, M.; Strom, T.B.; Elyaman, W.; Ho, I.C.; et al. IL-4 inhibits TGF-beta-induced Foxp3+ T cells and, together with TGF-beta, generates IL-9+ IL-10+ Foxp3(−) effector T cells. Nat. Immunol. 2008, 9, 1347–1355. [Google Scholar] [CrossRef] [Green Version]
  119. Cosmi, L.; Maggi, L.; Santarlasci, V.; Liotta, F.; Annunziato, F. T helper cells plasticity in inflammation. Cytom. Part A J. Int. Soc. Anal. Cytol. 2014, 85, 36–42. [Google Scholar] [CrossRef]
  120. Staudt, V.; Bothur, E.; Klein, M.; Lingnau, K.; Reuter, S.; Grebe, N.; Gerlitzki, B.; Hoffmann, M.; Ulges, A.; Taube, C.; et al. Interferon-regulatory factor 4 is essential for the developmental program of T helper 9 cells. Immunity 2010, 33, 192–202. [Google Scholar] [CrossRef] [Green Version]
  121. Chang, H.C.; Sehra, S.; Goswami, R.; Yao, W.; Yu, Q.; Stritesky, G.L.; Jabeen, R.; McKinley, C.; Ahyi, A.N.; Han, L.; et al. The transcription factor PU.1 is required for the development of IL-9-producing T cells and allergic inflammation. Nat. Immunol. 2010, 11, 527–534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Shohan, M.; Elahi, S.; Shirzad, H.; Rafieian-Kopaei, M.; Bagheri, N.; Soltani, E. Th9 Cells: Probable players in ulcerative colitis pathogenesis. Int. Rev. Immunol. 2018, 37, 192–205. [Google Scholar] [CrossRef] [PubMed]
  123. Xue, G.; Jin, G.; Fang, J.; Lu, Y. IL-4 together with IL-1beta induces antitumor Th9 cell differentiation in the absence of TGF-beta signaling. Nat. Commun. 2019, 10, 1376. [Google Scholar] [CrossRef] [PubMed]
  124. Wilhelm, C.; Turner, J.E.; Van Snick, J.; Stockinger, B. The many lives of IL-9: A question of survival? Nat. Immunol. 2012, 13, 637–641. [Google Scholar] [CrossRef]
  125. Schmitt, N.; Ueno, H. Regulation of human helper T cell subset differentiation by cytokines. Curr. Opin. Immunol. 2015, 34, 130–136. [Google Scholar] [CrossRef] [Green Version]
  126. Bauer, J.H.; Liu, K.D.; You, Y.; Lai, S.Y.; Goldsmith, M.A. Heteromerization of the gammac chain with the interleukin-9 receptor alpha subunit leads to STAT activation and prevention of apoptosis. J. Biol. Chem. 1998, 273, 9255–9260. [Google Scholar] [CrossRef] [Green Version]
  127. Demoulin, J.B.; Uyttenhove, C.; Van Roost, E.; DeLestre, B.; Donckers, D.; Van Snick, J.; Renauld, J.C. A single tyrosine of the interleukin-9 (IL-9) receptor is required for STAT activation, antiapoptotic activity, and growth regulation by IL-9. Mol. Cell. Biol. 1996, 16, 4710–4716. [Google Scholar] [CrossRef] [Green Version]
  128. Demoulin, J.B.; Van Roost, E.; Stevens, M.; Groner, B.; Renauld, J.C. Distinct roles for STAT1, STAT3, and STAT5 in differentiation gene induction and apoptosis inhibition by interleukin-9. J. Biol. Chem. 1999, 274, 25855–25861. [Google Scholar] [CrossRef] [Green Version]
  129. Gerlach, K.; Hwang, Y.; Nikolaev, A.; Atreya, R.; Dornhoff, H.; Steiner, S.; Lehr, H.A.; Wirtz, S.; Vieth, M.; Waisman, A.; et al. TH9 cells that express the transcription factor PU.1 drive T cell-mediated colitis via IL-9 receptor signaling in intestinal epithelial cells. Nat. Immunol. 2014, 15, 676–686. [Google Scholar] [CrossRef]
  130. Gerlach, K.; McKenzie, A.N.; Neurath, M.F.; Weigmann, B. IL-9 regulates intestinal barrier function in experimental T cell-mediated colitis. Tissue Barriers 2015, 3, e983777. [Google Scholar] [CrossRef] [Green Version]
  131. Turner, J.R. Intestinal mucosal barrier function in health and disease. Nat. Rev. Immunol. 2009, 9, 799–809. [Google Scholar] [CrossRef]
  132. Nalleweg, N.; Chiriac, M.T.; Podstawa, E.; Lehmann, C.; Rau, T.T.; Atreya, R.; Krauss, E.; Hundorfean, G.; Fichtner-Feigl, S.; Hartmann, A.; et al. IL-9 and its receptor are predominantly involved in the pathogenesis of UC. Gut 2015, 64, 743–755. [Google Scholar] [CrossRef] [Green Version]
  133. Atreya, R.; Neurath, M.F. IBD pathogenesis in 2014: Molecular pathways controlling barrier function in IBD. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 67–68. [Google Scholar] [CrossRef]
  134. Kim, H.S.; Chung, D.H. IL-9-producing invariant NKT cells protect against DSS-induced colitis in an IL-4-dependent manner. Mucosal Immunol. 2013, 6, 347–357. [Google Scholar] [CrossRef] [Green Version]
  135. Shohan, M.; Sabzevary-Ghahfarokhi, M.; Bagheri, N.; Shirzad, H.; Rahimian, G.; Soltani, A.; Ghatreh-Samani, M.; Deris, F.; Tahmasbi, K.; Shahverdi, E.; et al. Intensified Th9 Response is Associated with the Immunopathogenesis of Active Ulcerative Colitis. Immunol. Investig. 2018, 47, 700–711. [Google Scholar] [CrossRef]
  136. Singh, U.P.; Singh, N.P.; Murphy, E.A.; Price, R.L.; Fayad, R.; Nagarkatti, M.; Nagarkatti, P.S. Chemokine and cytokine levels in inflammatory bowel disease patients. Cytokine 2016, 77, 44–49. [Google Scholar] [CrossRef] [Green Version]
  137. Neurath, M.F.; Kaplan, M.H. Th9 cells in immunity and immunopathological diseases. Semin. Immunopathol. 2017, 39, 1–4. [Google Scholar] [CrossRef] [Green Version]
  138. Bilsborough, J.; Fiorino, M.F.; Henkle, B.W. Select animal models of colitis and their value in predicting clinical efficacy of biological therapies in ulcerative colitis. Expert Opin. Drug Discov. 2021, 16, 567–577. [Google Scholar] [CrossRef]
  139. Yuan, A.; Yang, H.; Qi, H.; Cui, J.; Hua, W.; Li, C.; Pang, Z.; Zheng, W.; Cui, G. IL-9 antibody injection suppresses the inflammation in colitis mice. Biochem. Biophys. Res. Commun. 2015, 468, 921–926. [Google Scholar] [CrossRef]
  140. Valatas, V.; Kolios, G.; Bamias, G. TL1A (TNFSF15) and DR3 (TNFRSF25): A Co-stimulatory System of Cytokines With Diverse Functions in Gut Mucosal Immunity. Front. Immunol. 2019, 10, 583. [Google Scholar] [CrossRef] [Green Version]
  141. Wang, D.; Li, H.; Duan, Y.Y.; Han, F.; Luo, Y.X.; Wu, M.Y.; Yang, M.Y.; Zhan, R.R.; Song, J.; Zhang, H.; et al. TL1A modulates the severity of colitis by promoting Th9 differentiation and IL-9 secretion. Life Sci. 2019, 231, 116536. [Google Scholar] [CrossRef] [PubMed]
  142. Perumal, N.B.; Kaplan, M.H. Regulating Il9 transcription in T helper cells. Trends Immunol. 2011, 32, 146–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Erle, D.J.; Briskin, M.J.; Butcher, E.C.; Garcia-Pardo, A.; Lazarovits, A.I.; Tidswell, M. Expression and function of the MAdCAM-1 receptor, integrin alpha 4 beta 7, on human leukocytes. J. Immunol. 1994, 153, 517–528. [Google Scholar] [CrossRef] [PubMed]
  144. Dotan, I.; Allez, M.; Danese, S.; Keir, M.; Tole, S.; McBride, J. The role of integrins in the pathogenesis of inflammatory bowel disease: Approved and investigational anti-integrin therapies. Med. Res. Rev. 2020, 40, 245–262. [Google Scholar] [CrossRef] [Green Version]
  145. Zundler, S.; Schillinger, D.; Fischer, A.; Atreya, R.; Lopez-Posadas, R.; Watson, A.; Neufert, C.; Atreya, I.; Neurath, M.F. Blockade of alphaEbeta7 integrin suppresses accumulation of CD8(+) and Th9 lymphocytes from patients with IBD in the inflamed gut in vivo. Gut 2017, 66, 1936–1948. [Google Scholar] [CrossRef] [Green Version]
  146. Acharya, S.; Timilshina, M.; Jiang, L.; Neupane, S.; Choi, D.Y.; Park, S.W.; Lee, S.Y.; Jeong, B.S.; Kim, J.A.; Nam, T.G.; et al. Amelioration of Experimental autoimmune encephalomyelitis and DSS induced colitis by NTG-A-009 through the inhibition of Th1 and Th17 cells differentiation. Sci. Rep. 2018, 8, 7799. [Google Scholar] [CrossRef] [Green Version]
  147. Korn, T.; Bettelli, E.; Oukka, M.; Kuchroo, V.K. IL-17 and Th17 Cells. Annu. Rev. Immunol. 2009, 27, 485–517. [Google Scholar] [CrossRef]
  148. Dong, C. Diversification of T-helper-cell lineages: Finding the family root of IL-17-producing cells. Nat. Rev. Immunol. 2006, 6, 329–333. [Google Scholar] [CrossRef]
  149. Weaver, C.T.; Harrington, L.E.; Mangan, P.R.; Gavrieli, M.; Murphy, K.M. Th17: An effector CD4 T cell lineage with regulatory T cell ties. Immunity 2006, 24, 677–688. [Google Scholar] [CrossRef] [Green Version]
  150. McGeachy, M.J.; Bak-Jensen, K.S.; Chen, Y.; Tato, C.M.; Blumenschein, W.; McClanahan, T.; Cua, D.J. 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, 8, 1390–1397. [Google Scholar] [CrossRef]
  151. Omenetti, S.; Bussi, C.; Metidji, A.; Iseppon, A.; Lee, S.; Tolaini, M.; Li, Y.; Kelly, G.; Chakravarty, P.; Shoaie, S.; et al. The Intestine Harbors Functionally Distinct Homeostatic Tissue-Resident and Inflammatory Th17 Cells. Immunity 2019, 51, 77–89.e6. [Google Scholar] [CrossRef] [Green Version]
  152. Esplugues, E.; Huber, S.; Gagliani, N.; Hauser, A.E.; Town, T.; Wan, Y.Y.; O’Connor, W., Jr.; Rongvaux, A.; Van Rooijen, N.; Haberman, A.M.; et al. Control of TH17 cells occurs in the small intestine. Nature 2011, 475, 514–518. [Google Scholar] [CrossRef] [Green Version]
  153. Noster, R.; de Koning, H.D.; Maier, E.; Prelog, M.; Lainka, E.; Zielinski, C.E. Dysregulation of proinflammatory versus anti-inflammatory human T(H)17 cell functionalities in the autoinflammatory Schnitzler syndrome. J. Allergy Clin. Immunol. 2016, 138, 1161–1169.e6. [Google Scholar] [CrossRef]
  154. Stockinger, B.; Omenetti, S. The dichotomous nature of T helper 17 cells. Nat. Rev. Immunol. 2017, 17, 535–544. [Google Scholar] [CrossRef]
  155. Ghoreschi, K.; Laurence, A.; Yang, X.P.; Tato, C.M.; McGeachy, M.J.; Konkel, J.E.; Ramos, H.L.; Wei, L.; Davidson, T.S.; Bouladoux, N.; et al. Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling. Nature 2010, 467, 967–971. [Google Scholar] [CrossRef] [Green Version]
  156. Lee, Y.; Awasthi, A.; Yosef, N.; Quintana, F.J.; Xiao, S.; Peters, A.; Wu, C.; Kleinewietfeld, M.; Kunder, S.; Hafler, D.A.; et al. Induction and molecular signature of pathogenic TH17 cells. Nat. Immunol. 2012, 13, 991–999. [Google Scholar] [CrossRef] [Green Version]
  157. Mangan, P.R.; Harrington, L.E.; O’Quinn, D.B.; Helms, W.S.; Bullard, D.C.; Elson, C.O.; Hatton, R.D.; Wahl, S.M.; Schoeb, T.R.; Weaver, C.T. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature 2006, 441, 231–234. [Google Scholar] [CrossRef] [PubMed]
  158. Bettelli, E.; Carrier, Y.; Gao, W.; Korn, T.; Strom, T.B.; Oukka, M.; Weiner, H.L.; Kuchroo, V.K. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006, 441, 235–238. [Google Scholar] [CrossRef] [PubMed]
  159. Veldhoen, M.; Hocking, R.J.; Atkins, C.J.; Locksley, R.M.; Stockinger, B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 2006, 24, 179–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Tesmer, L.A.; Lundy, S.K.; Sarkar, S.; Fox, D.A. Th17 cells in human disease. Immunol. Rev. 2008, 223, 87–113. [Google Scholar] [CrossRef]
  161. Yang, X.O.; Panopoulos, A.D.; Nurieva, R.; Chang, S.H.; Wang, D.; Watowich, S.S.; Dong, C. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J. Biol. Chem. 2007, 282, 9358–9363. [Google Scholar] [CrossRef] [Green Version]
  162. Ghoreschi, K.; Laurence, A.; Yang, X.P.; Hirahara, K.; O’Shea, J.J. T helper 17 cell heterogeneity and pathogenicity in autoimmune disease. Trends Immunol. 2011, 32, 395–401. [Google Scholar] [CrossRef] [Green Version]
  163. Wang, C.; Yosef, N.; Gaublomme, J.; Wu, C.; Lee, Y.; Clish, C.B.; Kaminski, J.; Xiao, S.; Meyer Zu Horste, G.; Pawlak, M.; et al. CD5L/AIM Regulates Lipid Biosynthesis and Restrains Th17 Cell Pathogenicity. Cell 2015, 163, 1413–1427. [Google Scholar] [CrossRef]
  164. Gaublomme, J.T.; Yosef, N.; Lee, Y.; Gertner, R.S.; Yang, L.V.; Wu, C.; Pandolfi, P.P.; Mak, T.; Satija, R.; Shalek, A.K.; et al. Single-Cell Genomics Unveils Critical Regulators of Th17 Cell Pathogenicity. Cell 2015, 163, 1400–1412. [Google Scholar] [CrossRef] [Green Version]
  165. Singh, R.P.; Hasan, S.; Sharma, S.; Nagra, S.; Yamaguchi, D.T.; Wong, D.T.; Hahn, B.H.; Hossain, A. Th17 cells in inflammation and autoimmunity. Autoimmun. Rev. 2014, 13, 1174–1181. [Google Scholar] [CrossRef]
  166. Gaffen, S. IL-17 receptor composition. Nat. Rev. Immunol. 2016, 16, 4. [Google Scholar] [CrossRef]
  167. O’Connor, W., Jr.; Kamanaka, M.; Booth, C.J.; Town, T.; Nakae, S.; Iwakura, Y.; Kolls, J.K.; Flavell, R.A. A protective function for interleukin 17A in T cell-mediated intestinal inflammation. Nat. Immunol. 2009, 10, 603–609. [Google Scholar] [CrossRef]
  168. Franke, A.; Balschun, T.; Karlsen, T.H.; Hedderich, J.; May, S.; Lu, T.; Schuldt, D.; Nikolaus, S.; Rosenstiel, P.; Krawczak, M.; et al. Replication of signals from recent studies of Crohn’s disease identifies previously unknown disease loci for ulcerative colitis. Nat. Genet. 2008, 40, 713–715. [Google Scholar] [CrossRef]
  169. Song, X.; Dai, D.; He, X.; Zhu, S.; Yao, Y.; Gao, H.; Wang, J.; Qu, F.; Qiu, J.; Wang, H.; et al. Growth Factor FGF2 Cooperates with Interleukin-17 to Repair Intestinal Epithelial Damage. Immunity 2015, 43, 488–501. [Google Scholar] [CrossRef] [Green Version]
  170. Hegazy, A.N.; West, N.R.; Stubbington, M.J.T.; Wendt, E.; Suijker, K.I.M.; Datsi, A.; This, S.; Danne, C.; Campion, S.; Duncan, S.H.; et al. Circulating and Tissue-Resident CD4(+) T Cells With Reactivity to Intestinal Microbiota Are Abundant in Healthy Individuals and Function Is Altered During Inflammation. Gastroenterology 2017, 153, 1320–1337.e16. [Google Scholar] [CrossRef] [Green Version]
  171. Ueno, A.; Jeffery, L.; Kobayashi, T.; Hibi, T.; Ghosh, S.; Jijon, H. Th17 plasticity and its relevance to inflammatory bowel disease. J. Autoimmun. 2018, 87, 38–49. [Google Scholar] [CrossRef]
  172. Zhao, J.; Lu, Q.; Liu, Y.; Shi, Z.; Hu, L.; Zeng, Z.; Tu, Y.; Xiao, Z.; Xu, Q. Th17 Cells in Inflammatory Bowel Disease: Cytokines, Plasticity, and Therapies. J. Immunol. Res. 2021, 2021, 8816041. [Google Scholar] [CrossRef] [PubMed]
  173. Liang, S.C.; Tan, X.Y.; Luxenberg, D.P.; Karim, R.; Dunussi-Joannopoulos, K.; Collins, M.; Fouser, L.A. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J. Exp. Med. 2006, 203, 2271–2279. [Google Scholar] [CrossRef] [PubMed]
  174. Ye, P.; Rodriguez, F.H.; Kanaly, S.; Stocking, K.L.; Schurr, J.; Schwarzenberger, P.; Oliver, P.; Huang, W.; Zhang, P.; Zhang, J.; 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, 519–527. [Google Scholar] [CrossRef] [PubMed]
  175. Jiang, P.; Zheng, C.; Xiang, Y.; Malik, S.; Su, D.; Xu, G.; Zhang, M. The involvement of TH17 cells in the pathogenesis of IBD. Cytokine Growth Factor Rev. 2022. [Google Scholar] [CrossRef] [PubMed]
  176. Schmitt, H.; Neurath, M.F.; Atreya, R. Role of the IL23/IL17 Pathway in Crohn’s Disease. Front. Immunol. 2021, 12, 622934. [Google Scholar] [CrossRef]
  177. Yan, J.B.; Luo, M.M.; Chen, Z.Y.; He, B.H. The Function and Role of the Th17/Treg Cell Balance in Inflammatory Bowel Disease. J. Immunol. Res. 2020, 2020, 8813558. [Google Scholar] [CrossRef]
  178. Raza, A.; Shata, M.T. Letter: Pathogenicity of Th17 cells may differ in ulcerative colitis compared with Crohn’s disease. Aliment. Pharmacol. Ther. 2012, 36, 204. [Google Scholar] [CrossRef]
  179. Sakuraba, A.; Sato, T.; Kamada, N.; Kitazume, M.; Sugita, A.; Hibi, T. Th1/Th17 immune response is induced by mesenteric lymph node dendritic cells in Crohn’s disease. Gastroenterology 2009, 137, 1736–1745. [Google Scholar] [CrossRef]
  180. Fujino, S.; Andoh, A.; Bamba, S.; Ogawa, A.; Hata, K.; Araki, Y.; Bamba, T.; Fujiyama, Y. Increased expression of interleukin 17 in inflammatory bowel disease. Gut 2003, 52, 65–70. [Google Scholar] [CrossRef]
  181. Monteleone, I.; Sarra, M.; Pallone, F.; Monteleone, G. Th17-related cytokines in inflammatory bowel diseases: Friends or foes? Curr. Mol. Med. 2012, 12, 592–597. [Google Scholar] [CrossRef]
  182. Zhang, Z.; Zheng, M.; Bindas, J.; Schwarzenberger, P.; Kolls, J.K. Critical role of IL-17 receptor signaling in acute TNBS-induced colitis. Inflamm. Bowel Dis. 2006, 12, 382–388. [Google Scholar] [CrossRef]
  183. Yang, X.O.; Chang, S.H.; Park, H.; Nurieva, R.; Shah, B.; Acero, L.; Wang, Y.H.; Schluns, K.S.; Broaddus, R.R.; Zhu, Z.; et al. Regulation of inflammatory responses by IL-17F. J. Exp. Med. 2008, 205, 1063–1075. [Google Scholar] [CrossRef]
  184. Strober, W.; Fuss, I.J.; Blumberg, R.S. The immunology of mucosal models of inflammation. Annu. Rev. Immunol. 2002, 20, 495–549. [Google Scholar] [CrossRef]
  185. Sandborn, W.J.; Feagan, B.G.; Fedorak, R.N.; Scherl, E.; Fleisher, M.R.; Katz, S.; Johanns, J.; Blank, M.; Rutgeerts, P.; 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, 135, 1130–1141. [Google Scholar] [CrossRef] [Green Version]
  186. Fina, D.; Sarra, M.; Fantini, M.C.; Rizzo, A.; Caruso, R.; Caprioli, F.; Stolfi, C.; Cardolini, I.; Dottori, M.; Boirivant, M.; et al. Regulation of gut inflammation and th17 cell response by interleukin-21. Gastroenterology 2008, 134, 1038–1048. [Google Scholar] [CrossRef]
  187. Strengell, M.; Sareneva, T.; Foster, D.; Julkunen, I.; Matikainen, S. IL-21 up-regulates the expression of genes associated with innate immunity and Th1 response. J. Immunol. 2002, 169, 3600–3605. [Google Scholar] [CrossRef] [Green Version]
  188. Gahring, L.C.; Carlson, N.G.; Kulmar, R.A.; Rogers, S.W. Neuronal expression of tumor necrosis factor alpha in the murine brain. Neuroimmunomodulation 1996, 3, 289–303. [Google Scholar] [CrossRef]
  189. Perrier, C.; de Hertogh, G.; Cremer, J.; Vermeire, S.; Rutgeerts, P.; Van Assche, G.; Szymkowski, D.E.; Ceuppens, J.L. Neutralization of membrane TNF, but not soluble TNF, is crucial for the treatment of experimental colitis. Inflamm. Bowel Dis. 2013, 19, 246–253. [Google Scholar] [CrossRef]
  190. Annunziato, F.; Cosmi, L.; Santarlasci, V.; Maggi, L.; Liotta, F.; Mazzinghi, B.; Parente, E.; Fili, L.; Ferri, S.; Frosali, F.; et al. Phenotypic and functional features of human Th17 cells. J. Exp. Med. 2007, 204, 1849–1861. [Google Scholar] [CrossRef] [Green Version]
  191. Lee, Y.K.; Turner, H.; Maynard, C.L.; Oliver, J.R.; Chen, D.; Elson, C.O.; Weaver, C.T. Late developmental plasticity in the T helper 17 lineage. Immunity 2009, 30, 92–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Hueber, W.; Sands, B.E.; Lewitzky, S.; Vandemeulebroecke, M.; Reinisch, W.; Higgins, P.D.; Wehkamp, J.; Feagan, B.G.; Yao, M.D.; Karczewski, M.; et al. 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, 1693–1700. [Google Scholar] [CrossRef] [Green Version]
  193. Targan, S.R.; Feagan, B.; Vermeire, S.; Panaccione, R.; Melmed, G.Y.; Landers, C.; Li, D.; Russell, C.; Newmark, R.; Zhang, N.; 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, 1599–1607. [Google Scholar] [CrossRef] [PubMed]
  194. Ogawa, A.; Andoh, A.; Araki, Y.; Bamba, T.; Fujiyama, Y. Neutralization of interleukin-17 aggravates dextran sulfate sodium-induced colitis in mice. Clin. Immunol. 2004, 110, 55–62. [Google Scholar] [CrossRef] [PubMed]
  195. Maxwell, J.R.; Zhang, Y.; Brown, W.A.; Smith, C.L.; Byrne, F.R.; Fiorino, M.; Stevens, E.; Bigler, J.; Davis, J.A.; Rottman, J.B.; et al. Differential Roles for Interleukin-23 and Interleukin-17 in Intestinal Immunoregulation. Immunity 2015, 43, 739–750. [Google Scholar] [CrossRef] [Green Version]
  196. Lee, J.S.; Tato, C.M.; Joyce-Shaikh, B.; Gulen, M.F.; Cayatte, C.; Chen, Y.; Blumenschein, W.M.; Judo, M.; Ayanoglu, G.; McClanahan, T.K.; et al. Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability. Immunity 2015, 43, 727–738. [Google Scholar] [CrossRef] [Green Version]
  197. Arnold, I.C.; Mathisen, S.; Schulthess, J.; Danne, C.; Hegazy, A.N.; Powrie, F. CD11c(+) monocyte/macrophages promote chronic Helicobacter hepaticus-induced intestinal inflammation through the production of IL-23. Mucosal Immunol. 2016, 9, 352–363. [Google Scholar] [CrossRef] [Green Version]
  198. Becker, C.; Dornhoff, H.; Neufert, C.; Fantini, M.C.; Wirtz, S.; Huebner, S.; Nikolaev, A.; Lehr, H.A.; Murphy, A.J.; Valenzuela, D.M.; et al. Cutting edge: IL-23 cross-regulates IL-12 production in T cell-dependent experimental colitis. J. Immunol. 2006, 177, 2760–2764. [Google Scholar] [CrossRef] [Green Version]
  199. Yen, D.; Cheung, J.; Scheerens, H.; Poulet, F.; McClanahan, T.; McKenzie, B.; Kleinschek, M.A.; Owyang, A.; Mattson, J.; Blumenschein, W.; et al. IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6. J. Clin. Investig. 2006, 116, 1310–1316. [Google Scholar] [CrossRef] [Green Version]
  200. Karaboga, I.; Demirtas, S.; Karaca, T. Investigation of the relationship between the Th17/IL-23 pathway and innate-adaptive immune system in TNBS-induced colitis in rats. Iran. J. Basic Med. Sci. 2017, 20, 870–879. [Google Scholar]
  201. Noviello, D.; Mager, R.; Roda, G.; Borroni, R.G.; Fiorino, G.; Vetrano, S. The IL23-IL17 Immune Axis in the Treatment of Ulcerative Colitis: Successes, Defeats, and Ongoing Challenges. Front. Immunol. 2021, 12, 611256. [Google Scholar] [CrossRef]
  202. Zheng, Y.; Valdez, P.A.; Danilenko, D.M.; Hu, Y.; Sa, S.M.; Gong, Q.; Abbas, A.R.; Modrusan, Z.; Ghilardi, N.; de Sauvage, F.J.; et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat. Med. 2008, 14, 282–289. [Google Scholar] [CrossRef]
  203. Pickert, G.; Neufert, C.; Leppkes, M.; Zheng, Y.; Wittkopf, N.; Warntjen, M.; Lehr, H.A.; Hirth, S.; Weigmann, B.; Wirtz, S.; et al. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. J. Exp. Med. 2009, 206, 1465–1472. [Google Scholar] [CrossRef] [Green Version]
  204. Basu, R.; O’Quinn, D.B.; Silberger, D.J.; Schoeb, T.R.; Fouser, L.; Ouyang, W.; Hatton, R.D.; Weaver, C.T. Th22 cells are an important source of IL-22 for host protection against enteropathogenic bacteria. Immunity 2012, 37, 1061–1075. [Google Scholar] [CrossRef]
  205. Dudakov, J.A.; Mertelsmann, A.M.; O’Connor, M.H.; Jenq, R.R.; Velardi, E.; Young, L.F.; Smith, O.M.; Boyd, R.L.; van den Brink, M.R.M.; Hanash, A.M. Loss of thymic innate lymphoid cells leads to impaired thymopoiesis in experimental graft-versus-host disease. Blood 2017, 130, 933–942. [Google Scholar] [CrossRef] [Green Version]
  206. Shohan, M.; Dehghani, R.; Khodadadi, A.; Dehnavi, S.; Ahmadi, R.; Joudaki, N.; Houshmandfar, S.; Shamshiri, M.; Shojapourian, S.; Bagheri, N. Interleukin-22 and intestinal homeostasis: Protective or destructive? IUBMB Life 2020, 72, 1585–1602. [Google Scholar] [CrossRef]
  207. Plank, M.W.; Kaiko, G.E.; Maltby, S.; Weaver, J.; Tay, H.L.; Shen, W.; Wilson, M.S.; Durum, S.K.; Foster, P.S. Th22 Cells Form a Distinct Th Lineage from Th17 Cells In Vitro with Unique Transcriptional Properties and Tbet-Dependent Th1 Plasticity. J. Immunol. 2017, 198, 2182–2190. [Google Scholar] [CrossRef] [Green Version]
  208. 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, 10, 857–863. [Google Scholar] [CrossRef]
  209. Li, J.; Chen, S.; Xiao, X.; Zhao, Y.; Ding, W.; Li, X.C. IL-9 and Th9 cells in health and diseases-From tolerance to immunopathology. Cytokine Growth Factor Rev. 2017, 37, 47–55. [Google Scholar] [CrossRef]
  210. Mock, B.A.; Krall, M.; Kozak, C.A.; Nesbitt, M.N.; McBride, O.W.; Renauld, J.C.; Van Snick, J. IL9 maps to mouse chromosome 13 and human chromosome 5. Immunogenetics 1990, 31, 265–270. [Google Scholar] [CrossRef]
  211. Stassen, M.; Schmitt, E.; Bopp, T. From interleukin-9 to T helper 9 cells. Ann. N. Y. Acad. Sci. 2012, 1247, 56–68. [Google Scholar] [CrossRef] [PubMed]
  212. Steenwinckel, V.; Louahed, J.; Orabona, C.; Huaux, F.; Warnier, G.; McKenzie, A.; Lison, D.; Levitt, R.; Renauld, J.C. IL-13 mediates in vivo IL-9 activities on lung epithelial cells but not on hematopoietic cells. J. Immunol. 2007, 178, 3244–3251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Arshad, T.; Mansur, F.; Palek, R.; Manzoor, S.; Liska, V. A Double Edged Sword Role of Interleukin-22 in Wound Healing and Tissue Regeneration. Front. Immunol. 2020, 11, 2148. [Google Scholar] [CrossRef] [PubMed]
  214. Lindemans, C.A.; Calafiore, M.; Mertelsmann, A.M.; O’Connor, M.H.; Dudakov, J.A.; Jenq, R.R.; Velardi, E.; Young, L.F.; Smith, O.M.; Lawrence, G.; et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature 2015, 528, 560–564. [Google Scholar] [CrossRef]
  215. Sugimoto, K.; Ogawa, A.; Mizoguchi, E.; Shimomura, Y.; Andoh, A.; Bhan, A.K.; Blumberg, R.S.; Xavier, R.J.; Mizoguchi, A. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J. Clin. Investig. 2008, 118, 534–544. [Google Scholar] [CrossRef] [Green Version]
  216. Gibson, D.L.; Montero, M.; Ropeleski, M.J.; Bergstrom, K.S.; Ma, C.; Ghosh, S.; Merkens, H.; Huang, J.; Mansson, L.E.; Sham, H.P.; et al. Interleukin-11 reduces TLR4-induced colitis in TLR2-deficient mice and restores intestinal STAT3 signaling. Gastroenterology 2010, 139, 1277–1288. [Google Scholar] [CrossRef]
  217. Andoh, A.; Zhang, Z.; Inatomi, O.; Fujino, S.; Deguchi, Y.; Araki, Y.; Tsujikawa, T.; Kitoh, K.; Kim-Mitsuyama, S.; Takayanagi, A.; et al. Interleukin-22, a member of the IL-10 subfamily, induces inflammatory responses in colonic subepithelial myofibroblasts. Gastroenterology 2005, 129, 969–984. [Google Scholar] [CrossRef]
  218. Pavlidis, P.; Tsakmaki, A.; Pantazi, E.; Li, K.; Cozzetto, D.; Digby-Bell, J.; Yang, F.; Lo, J.W.; Alberts, E.; Sa, A.C.C.; et al. Interleukin-22 regulates neutrophil recruitment in ulcerative colitis and is associated with resistance to ustekinumab therapy. Nat. Commun. 2022, 13, 5820. [Google Scholar] [CrossRef]
  219. Huber, S.; Gagliani, N.; Zenewicz, L.A.; Huber, F.J.; Bosurgi, L.; Hu, B.; Hedl, M.; Zhang, W.; O’Connor, W., Jr.; Murphy, A.J.; et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature 2012, 491, 259–263. [Google Scholar] [CrossRef] [Green Version]
  220. Pelczar, P.; Witkowski, M.; Perez, L.G.; Kempski, J.; Hammel, A.G.; Brockmann, L.; Kleinschmidt, D.; Wende, S.; Haueis, C.; Bedke, T.; et al. A pathogenic role for T cell-derived IL-22BP in inflammatory bowel disease. Science 2016, 354, 358–362. [Google Scholar] [CrossRef]
  221. Jinnohara, T.; Kanaya, T.; Hase, K.; Sakakibara, S.; Kato, T.; Tachibana, N.; Sasaki, T.; Hashimoto, Y.; Sato, T.; Watarai, H.; et al. IL-22BP dictates characteristics of Peyer’s patch follicle-associated epithelium for antigen uptake. J. Exp. Med. 2017, 214, 1607–1618. [Google Scholar] [CrossRef]
  222. Martin, J.C.; Beriou, G.; Heslan, M.; Chauvin, C.; Utriainen, L.; Aumeunier, A.; Scott, C.L.; Mowat, A.; Cerovic, V.; Houston, S.A.; et al. Interleukin-22 binding protein (IL-22BP) is constitutively expressed by a subset of conventional dendritic cells and is strongly induced by retinoic acid. Mucosal Immunol. 2014, 7, 101–113. [Google Scholar] [CrossRef] [Green Version]
  223. Savage, A.K.; Liang, H.E.; Locksley, R.M. The Development of Steady-State Activation Hubs between Adult LTi ILC3s and Primed Macrophages in Small Intestine. J. Immunol. 2017, 199, 1912–1922. [Google Scholar] [CrossRef] [Green Version]
  224. Zenewicz, L.A. IL-22 Binding Protein (IL-22BP) in the Regulation of IL-22 Biology. Front. Immunol. 2021, 12, 766586. [Google Scholar] [CrossRef]
  225. Kotenko, S.V.; Izotova, L.S.; Mirochnitchenko, O.V.; Esterova, E.; Dickensheets, H.; Donnelly, R.P.; Pestka, S. Identification, cloning, and characterization of a novel soluble receptor that binds IL-22 and neutralizes its activity. J. Immunol. 2001, 166, 7096–7103. [Google Scholar] [CrossRef] [Green Version]
  226. Schmechel, S.; Konrad, A.; Diegelmann, J.; Glas, J.; Wetzke, M.; Paschos, E.; Lohse, P.; Goke, B.; Brand, S. Linking genetic susceptibility to Crohn’s disease with Th17 cell function: IL-22 serum levels are increased in Crohn’s disease and correlate with disease activity and IL23R genotype status. Inflamm. Bowel Dis. 2008, 14, 204–212. [Google Scholar] [CrossRef]
  227. Wolk, K.; Witte, E.; Hoffmann, U.; Doecke, W.D.; Endesfelder, S.; Asadullah, K.; Sterry, W.; Volk, H.D.; Wittig, B.M.; Sabat, R. IL-22 induces lipopolysaccharide-binding protein in hepatocytes: A potential systemic role of IL-22 in Crohn’s disease. J. Immunol. 2007, 178, 5973–5981. [Google Scholar] [CrossRef] [Green Version]
  228. Martin, J.C.; Beriou, G.; Heslan, M.; Bossard, C.; Jarry, A.; Abidi, A.; Hulin, P.; Menoret, S.; Thinard, R.; Anegon, I.; et al. IL-22BP is produced by eosinophils in human gut and blocks IL-22 protective actions during colitis. Mucosal Immunol. 2016, 9, 539–549. [Google Scholar] [CrossRef] [Green Version]
  229. Gui, X.; Li, J.; Ueno, A.; Iacucci, M.; Qian, J.; Ghosh, S. Histopathological Features of Inflammatory Bowel Disease are Associated With Different CD4+ T Cell Subsets in Colonic Mucosal Lamina Propria. J. Crohns Colitis 2018, 12, 1448–1458. [Google Scholar] [CrossRef]
  230. Fang, L.; Pang, Z.; Shu, W.; Wu, W.; Sun, M.; Cong, Y.; Liu, Z. Anti-TNF Therapy Induces CD4+ T-Cell Production of IL-22 and Promotes Epithelial Repairs in Patients With Crohn’s Disease. Inflamm. Bowel Dis. 2018, 24, 1733–1744. [Google Scholar] [CrossRef]
  231. Josefowicz, S.Z.; Lu, L.F.; Rudensky, A.Y. Regulatory T cells: Mechanisms of differentiation and function. Annu. Rev. Immunol. 2012, 30, 531–564. [Google Scholar] [CrossRef] [PubMed]
  232. Itoh, M.; Takahashi, T.; Sakaguchi, N.; Kuniyasu, Y.; Shimizu, J.; Otsuka, F.; Sakaguchi, S. Thymus and autoimmunity: Production of CD25+CD4+ naturally anergic and suppressive T cells as a key function of the thymus in maintaining immunologic self-tolerance. J. Immunol. 1999, 162, 5317–5326. [Google Scholar] [CrossRef] [PubMed]
  233. Fontenot, J.D.; Gavin, M.A.; Rudensky, A.Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat. Immunol. 2003, 4, 330–336. [Google Scholar] [CrossRef] [PubMed]
  234. Hori, S.; Nomura, T.; Sakaguchi, S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003, 299, 1057–1061. [Google Scholar] [CrossRef]
  235. Sakaguchi, S.; Sakaguchi, N.; Asano, M.; Itoh, M.; Toda, M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J. Immunol. 1995, 155, 1151–1164. [Google Scholar] [CrossRef]
  236. Josefowicz, S.Z.; Rudensky, A. Control of regulatory T cell lineage commitment and maintenance. Immunity 2009, 30, 616–625. [Google Scholar] [CrossRef] [Green Version]
  237. Abbas, A.K.; Benoist, C.; Bluestone, J.A.; Campbell, D.J.; Ghosh, S.; Hori, S.; Jiang, S.; Kuchroo, V.K.; Mathis, D.; Roncarolo, M.G.; et al. Regulatory T cells: Recommendations to simplify the nomenclature. Nat. Immunol. 2013, 14, 307–308. [Google Scholar] [CrossRef] [Green Version]
  238. Sakaguchi, S.; Mikami, N.; Wing, J.B.; Tanaka, A.; Ichiyama, K.; Ohkura, N. Regulatory T Cells and Human Disease. Annu. Rev. Immunol. 2020, 38, 541–566. [Google Scholar] [CrossRef] [Green Version]
  239. Hsieh, C.S.; Lee, H.M.; Lio, C.W. Selection of regulatory T cells in the thymus. Nat. Rev. Immunol. 2012, 12, 157–167. [Google Scholar] [CrossRef]
  240. Lancaster, J.N.; Li, Y.; Ehrlich, L.I.R. Chemokine-Mediated Choreography of Thymocyte Development and Selection. Trends Immunol. 2018, 39, 86–98. [Google Scholar] [CrossRef]
  241. Lancaster, J.N.; Thyagarajan, H.M.; Srinivasan, J.; Li, Y.; Hu, Z.; Ehrlich, L.I.R. Live-cell imaging reveals the relative contributions of antigen-presenting cell subsets to thymic central tolerance. Nat. Commun. 2019, 10, 2220. [Google Scholar] [CrossRef] [Green Version]
  242. Caramalho, I.; Nunes-Cabaco, H.; Foxall, R.B.; Sousa, A.E. Regulatory T-Cell Development in the Human Thymus. Front. Immunol. 2015, 6, 395. [Google Scholar] [CrossRef] [Green Version]
  243. Sakaguchi, S.; Yamaguchi, T.; Nomura, T.; Ono, M. Regulatory T cells and immune tolerance. Cell 2008, 133, 775–787. [Google Scholar] [CrossRef] [Green Version]
  244. Takaba, H.; Takayanagi, H. The Mechanisms of T Cell Selection in the Thymus. Trends Immunol. 2017, 38, 805–816. [Google Scholar] [CrossRef]
  245. Veenbergen, S.; Samsom, J.N. Maintenance of small intestinal and colonic tolerance by IL-10-producing regulatory T cell subsets. Curr. Opin. Immunol. 2012, 24, 269–276. [Google Scholar] [CrossRef]
  246. Worbs, T.; Bode, U.; Yan, S.; Hoffmann, M.W.; Hintzen, G.; Bernhardt, G.; Forster, R.; Pabst, O. Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J. Exp. Med. 2006, 203, 519–527. [Google Scholar] [CrossRef] [Green Version]
  247. Hauet-Broere, F.; Unger, W.W.; Garssen, J.; Hoijer, M.A.; Kraal, G.; Samsom, J.N. Functional CD25− and CD25+ mucosal regulatory T cells are induced in gut-draining lymphoid tissue within 48 h after oral antigen application. Eur. J. Immunol. 2003, 33, 2801–2810. [Google Scholar] [CrossRef]
  248. Davidson, T.S.; DiPaolo, R.J.; Andersson, J.; Shevach, E.M. Cutting Edge: IL-2 is essential for TGF-beta-mediated induction of Foxp3+ T regulatory cells. J. Immunol. 2007, 178, 4022–4026. [Google Scholar] [CrossRef] [Green Version]
  249. Kretschmer, K.; Apostolou, I.; Hawiger, D.; Khazaie, K.; Nussenzweig, M.C.; von Boehmer, H. Inducing and expanding regulatory T cell populations by foreign antigen. Nat. Immunol. 2005, 6, 1219–1227. [Google Scholar] [CrossRef]
  250. Chen, W.; Jin, W.; Hardegen, N.; Lei, K.J.; Li, L.; Marinos, N.; McGrady, G.; Wahl, S.M. Conversion of peripheral CD4+CD25− naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J. Exp. Med. 2003, 198, 1875–1886. [Google Scholar] [CrossRef]
  251. Coombes, J.L.; Siddiqui, K.R.; Arancibia-Carcamo, C.V.; Hall, J.; Sun, C.M.; Belkaid, Y.; Powrie, F. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J. Exp. Med. 2007, 204, 1757–1764. [Google Scholar] [CrossRef] [PubMed]
  252. Mucida, D.; Park, Y.; Kim, G.; Turovskaya, O.; Scott, I.; Kronenberg, M.; Cheroutre, H. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science 2007, 317, 256–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Wang, J.; Ioan-Facsinay, A.; van der Voort, E.I.; Huizinga, T.W.; Toes, R.E. Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur. J. Immunol. 2007, 37, 129–138. [Google Scholar] [CrossRef] [PubMed]
  254. Jacobse, J.; Li, J.; Rings, E.; Samsom, J.N.; Goettel, J.A. Intestinal Regulatory T Cells as Specialized Tissue-Restricted Immune Cells in Intestinal Immune Homeostasis and Disease. Front. Immunol. 2021, 12, 716499. [Google Scholar] [CrossRef]
  255. Nelson, R.W.; Beisang, D.; Tubo, N.J.; Dileepan, T.; Wiesner, D.L.; Nielsen, K.; Wuthrich, M.; Klein, B.S.; Kotov, D.I.; Spanier, J.A.; et al. T cell receptor cross-reactivity between similar foreign and self peptides influences naive cell population size and autoimmunity. Immunity 2015, 42, 95–107. [Google Scholar] [CrossRef] [Green Version]
  256. Tanoue, T.; Atarashi, K.; Honda, K. Development and maintenance of intestinal regulatory T cells. Nat. Rev. Immunol. 2016, 16, 295–309. [Google Scholar] [CrossRef]
  257. Atarashi, K.; Tanoue, T.; Shima, T.; Imaoka, A.; Kuwahara, T.; Momose, Y.; Cheng, G.; Yamasaki, S.; Saito, T.; Ohba, Y.; et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011, 331, 337–341. [Google Scholar] [CrossRef] [Green Version]
  258. Wing, J.B.; Tanaka, A.; Sakaguchi, S. Human FOXP3(+) Regulatory T Cell Heterogeneity and Function in Autoimmunity and Cancer. Immunity 2019, 50, 302–316. [Google Scholar] [CrossRef] [Green Version]
  259. Miyara, M.; Yoshioka, Y.; Kitoh, A.; Shima, T.; Wing, K.; Niwa, A.; Parizot, C.; Taflin, C.; Heike, T.; Valeyre, D.; et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 2009, 30, 899–911. [Google Scholar] [CrossRef] [Green Version]
  260. Fisson, S.; Darrasse-Jeze, G.; Litvinova, E.; Septier, F.; Klatzmann, D.; Liblau, R.; Salomon, B.L. Continuous activation of autoreactive CD4+ CD25+ regulatory T cells in the steady state. J. Exp. Med. 2003, 198, 737–746. [Google Scholar] [CrossRef]
  261. Asseman, C.; Mauze, S.; Leach, M.W.; Coffman, R.L.; Powrie, F. An essential role for interleukin 10 in the function of regulatory T cells that inhibit intestinal inflammation. J. Exp. Med. 1999, 190, 995–1004. [Google Scholar] [CrossRef]
  262. Wirtz, S.; Billmeier, U.; McHedlidze, T.; Blumberg, R.S.; Neurath, M.F. Interleukin-35 mediates mucosal immune responses that protect against T-cell-dependent colitis. Gastroenterology 2011, 141, 1875–1886. [Google Scholar] [CrossRef] [Green Version]
  263. Nakamura, K.; Kitani, A.; Fuss, I.; Pedersen, A.; Harada, N.; Nawata, H.; Strober, W. TGF-beta 1 plays an important role in the mechanism of CD4+CD25+ regulatory T cell activity in both humans and mice. J. Immunol. 2004, 172, 834–842. [Google Scholar] [CrossRef] [Green Version]
  264. Powrie, F.; Carlino, J.; Leach, M.W.; Mauze, S.; Coffman, R.L. A critical role for transforming growth factor-beta but not interleukin 4 in the suppression of T helper type 1-mediated colitis by CD45RB(low) CD4+ T cells. J. Exp. Med. 1996, 183, 2669–2674. [Google Scholar] [CrossRef]
  265. Read, S.; Greenwald, R.; Izcue, A.; Robinson, N.; Mandelbrot, D.; Francisco, L.; Sharpe, A.H.; Powrie, F. Blockade of CTLA-4 on CD4+CD25+ regulatory T cells abrogates their function in vivo. J. Immunol. 2006, 177, 4376–4383. [Google Scholar] [CrossRef] [Green Version]
  266. Read, S.; Malmstrom, V.; Powrie, F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J. Exp. Med. 2000, 192, 295–302. [Google Scholar] [CrossRef] [Green Version]
  267. Burzyn, D.; Kuswanto, W.; Kolodin, D.; Shadrach, J.L.; Cerletti, M.; Jang, Y.; Sefik, E.; Tan, T.G.; Wagers, A.J.; Benoist, C.; et al. A special population of regulatory T cells potentiates muscle repair. Cell 2013, 155, 1282–1295. [Google Scholar] [CrossRef] [Green Version]
  268. Maul, J.; Loddenkemper, C.; Mundt, P.; Berg, E.; Giese, T.; Stallmach, A.; Zeitz, M.; Duchmann, R. Peripheral and intestinal regulatory CD4+ CD25(high) T cells in inflammatory bowel disease. Gastroenterology 2005, 128, 1868–1878. [Google Scholar] [CrossRef]
  269. Wang, Y.; Song, W.; Yu, S.; Liu, Y.; Chen, Y.G. Intestinal cellular heterogeneity and disease development revealed by single-cell technology. Cell Regen. 2022, 11, 26. [Google Scholar] [CrossRef]
  270. Huang, B.; Chen, Z.; Geng, L.; Wang, J.; Liang, H.; Cao, Y.; Chen, H.; Huang, W.; Su, M.; Wang, H.; et al. Mucosal Profiling of Pediatric-Onset Colitis and IBD Reveals Common Pathogenics and Therapeutic Pathways. Cell 2019, 179, 1160–1176.e24. [Google Scholar] [CrossRef]
  271. Jaeger, N.; Gamini, R.; Cella, M.; Schettini, J.L.; Bugatti, M.; Zhao, S.; Rosadini, C.V.; Esaulova, E.; Di Luccia, B.; Kinnett, B.; et al. Single-cell analyses of Crohn’s disease tissues reveal intestinal intraepithelial T cells heterogeneity and altered subset distributions. Nat. Commun. 2021, 12, 1921. [Google Scholar] [CrossRef] [PubMed]
  272. Kinchen, J.; Chen, H.H.; Parikh, K.; Antanaviciute, A.; Jagielowicz, M.; Fawkner-Corbett, D.; Ashley, N.; Cubitt, L.; Mellado-Gomez, E.; Attar, M.; et al. Structural Remodeling of the Human Colonic Mesenchyme in Inflammatory Bowel Disease. Cell 2018, 175, 372–386.e17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Martin, J.C.; Chang, C.; Boschetti, G.; Ungaro, R.; Giri, M.; Grout, J.A.; Gettler, K.; Chuang, L.S.; Nayar, S.; Greenstein, A.J.; et al. Single-Cell Analysis of Crohn’s Disease Lesions Identifies a Pathogenic Cellular Module Associated with Resistance to Anti-TNF Therapy. Cell 2019, 178, 1493–1508.e20. [Google Scholar] [CrossRef] [PubMed]
  274. Parikh, K.; Antanaviciute, A.; Fawkner-Corbett, D.; Jagielowicz, M.; Aulicino, A.; Lagerholm, C.; Davis, S.; Kinchen, J.; Chen, H.H.; Alham, N.K.; et al. Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature 2019, 567, 49–55. [Google Scholar] [CrossRef] [PubMed]
  275. Smillie, C.S.; Biton, M.; Ordovas-Montanes, J.; Sullivan, K.M.; Burgin, G.; Graham, D.B.; Herbst, R.H.; Rogel, N.; Slyper, M.; Waldman, J.; et al. Intra- and Inter-cellular Rewiring of the Human Colon during Ulcerative Colitis. Cell 2019, 178, 714–730.e22. [Google Scholar] [CrossRef]
  276. West, N.R.; Hegazy, A.N.; Owens, B.M.J.; Bullers, S.J.; Linggi, B.; Buonocore, S.; Coccia, M.; Gortz, D.; This, S.; Stockenhuber, K.; et al. Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease. Nat. Med. 2017, 23, 579–589. [Google Scholar] [CrossRef] [Green Version]
  277. Mitsialis, V.; Wall, S.; Liu, P.; Ordovas-Montanes, J.; Parmet, T.; Vukovic, M.; Spencer, D.; Field, M.; McCourt, C.; Toothaker, J.; et al. Single-Cell Analyses of Colon and Blood Reveal Distinct Immune Cell Signatures of Ulcerative Colitis and Crohn’s Disease. Gastroenterology 2020, 159, 591–608.e10. [Google Scholar] [CrossRef]
  278. Luoma, A.M.; Suo, S.; Williams, H.L.; Sharova, T.; Sullivan, K.; Manos, M.; Bowling, P.; Hodi, F.S.; Rahma, O.; Sullivan, R.J.; et al. Molecular Pathways of Colon Inflammation Induced by Cancer Immunotherapy. Cell 2020, 182, 655–671.e22. [Google Scholar] [CrossRef]
  279. Devlin, J.C.; Axelrad, J.; Hine, A.M.; Chang, S.; Sarkar, S.; Lin, J.D.; Ruggles, K.V.; Hudesman, D.; Cadwell, K.; Loke, P. Single-Cell Transcriptional Survey of Ileal-Anal Pouch Immune Cells From Ulcerative Colitis Patients. Gastroenterology 2021, 160, 1679–1693. [Google Scholar] [CrossRef]
  280. Hovhannisyan, Z.; Treatman, J.; Littman, D.R.; Mayer, L. Characterization of interleukin-17-producing regulatory T cells in inflamed intestinal mucosa from patients with inflammatory bowel diseases. Gastroenterology 2011, 140, 957–965. [Google Scholar] [CrossRef] [Green Version]
  281. Rubtsov, Y.P.; Niec, R.E.; Josefowicz, S.; Li, L.; Darce, J.; Mathis, D.; Benoist, C.; Rudensky, A.Y. Stability of the regulatory T cell lineage in vivo. Science 2010, 329, 1667–1671. [Google Scholar] [CrossRef] [Green Version]
  282. Wing, J.B.; Sakaguchi, S. Multiple treg suppressive modules and their adaptability. Front. Immunol. 2012, 3, 178. [Google Scholar] [CrossRef] [Green Version]
  283. Levine, A.G.; Mendoza, A.; Hemmers, S.; Moltedo, B.; Niec, R.E.; Schizas, M.; Hoyos, B.E.; Putintseva, E.V.; Chaudhry, A.; Dikiy, S.; et al. Stability and function of regulatory T cells expressing the transcription factor T-bet. Nature 2017, 546, 421–425. [Google Scholar] [CrossRef] [Green Version]
  284. Koch, M.A.; Tucker-Heard, G.; Perdue, N.R.; Killebrew, J.R.; Urdahl, K.B.; Campbell, D.J. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat. Immunol. 2009, 10, 595–602. [Google Scholar] [CrossRef]
  285. Zheng, Y.; Chaudhry, A.; Kas, A.; deRoos, P.; Kim, J.M.; Chu, T.T.; Corcoran, L.; Treuting, P.; Klein, U.; Rudensky, A.Y. Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature 2009, 458, 351–356. [Google Scholar] [CrossRef] [Green Version]
  286. Cretney, E.; Xin, A.; Shi, W.; Minnich, M.; Masson, F.; Miasari, M.; Belz, G.T.; Smyth, G.K.; Busslinger, M.; Nutt, S.L.; et al. The transcription factors Blimp-1 and IRF4 jointly control the differentiation and function of effector regulatory T cells. Nat. Immunol. 2011, 12, 304–311. [Google Scholar] [CrossRef]
  287. Wohlfert, E.A.; Grainger, J.R.; Bouladoux, N.; Konkel, J.E.; Oldenhove, G.; Ribeiro, C.H.; Hall, J.A.; Yagi, R.; Naik, S.; Bhairavabhotla, R.; et al. GATA3 controls Foxp3(+) regulatory T cell fate during inflammation in mice. J. Clin. Investig. 2011, 121, 4503–4515. [Google Scholar] [CrossRef] [Green Version]
  288. Chaudhry, A.; Rudra, D.; Treuting, P.; Samstein, R.M.; Liang, Y.; Kas, A.; Rudensky, A.Y. CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science 2009, 326, 986–991. [Google Scholar] [CrossRef] [Green Version]
  289. Ohnmacht, C.; Park, J.H.; Cording, S.; Wing, J.B.; Atarashi, K.; Obata, Y.; Gaboriau-Routhiau, V.; Marques, R.; Dulauroy, S.; Fedoseeva, M.; et al. MUCOSAL IMMUNOLOGY. The microbiota regulates type 2 immunity through RORgammat(+) T cells. Science 2015, 349, 989–993. [Google Scholar] [CrossRef]
  290. Sefik, E.; Geva-Zatorsky, N.; Oh, S.; Konnikova, L.; Zemmour, D.; McGuire, A.M.; Burzyn, D.; Ortiz-Lopez, A.; Lobera, M.; Yang, J.; et al. MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce a distinct population of RORgamma(+) regulatory T cells. Science 2015, 349, 993–997. [Google Scholar] [CrossRef] [Green Version]
  291. Linterman, M.A.; Pierson, W.; Lee, S.K.; Kallies, A.; Kawamoto, S.; Rayner, T.F.; Srivastava, M.; Divekar, D.P.; Beaton, L.; Hogan, J.J.; et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat. Med. 2011, 17, 975–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Chung, Y.; Tanaka, S.; Chu, F.; Nurieva, R.I.; Martinez, G.J.; Rawal, S.; Wang, Y.H.; Lim, H.; Reynolds, J.M.; Zhou, X.H.; et al. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat. Med. 2011, 17, 983–988. [Google Scholar] [CrossRef] [PubMed]
  293. Wollenberg, I.; Agua-Doce, A.; Hernandez, A.; Almeida, C.; Oliveira, V.G.; Faro, J.; Graca, L. Regulation of the germinal center reaction by Foxp3+ follicular regulatory T cells. J. Immunol. 2011, 187, 4553–4560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Schiering, C.; Krausgruber, T.; Chomka, A.; Frohlich, A.; Adelmann, K.; Wohlfert, E.A.; Pott, J.; Griseri, T.; Bollrath, J.; Hegazy, A.N.; et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 2014, 513, 564–568. [Google Scholar] [CrossRef]
  295. Kim, K.S.; Hong, S.W.; Han, D.; Yi, J.; Jung, J.; Yang, B.G.; Lee, J.Y.; Lee, M.; Surh, C.D. Dietary antigens limit mucosal immunity by inducing regulatory T cells in the small intestine. Science 2016, 351, 858–863. [Google Scholar] [CrossRef]
  296. Negi, S.; Saini, S.; Tandel, N.; Sahu, K.; Mishra, R.P.N.; Tyagi, R.K. Translating Treg Therapy for Inflammatory Bowel Disease in Humanized Mice. Cells 2021, 10, 1847. [Google Scholar] [CrossRef]
  297. Cassinotti, A.; Passamonti, F.; Segato, S. Cell Therapy in Inflammatory Bowel Disease. Pharmacol. Res. 2021, 163, 105247. [Google Scholar] [CrossRef]
  298. Bacchetta, R.; Bigler, M.; Touraine, J.L.; Parkman, R.; Tovo, P.A.; Abrams, J.; de Waal Malefyt, R.; de Vries, J.E.; Roncarolo, M.G. High levels of interleukin 10 production in vivo are associated with tolerance in SCID patients transplanted with HLA mismatched hematopoietic stem cells. J. Exp. Med. 1994, 179, 493–502. [Google Scholar] [CrossRef]
  299. He, J.; Tsai, L.M.; Leong, Y.A.; Hu, X.; Ma, C.S.; Chevalier, N.; Sun, X.; Vandenberg, K.; Rockman, S.; Ding, Y.; et al. Circulating precursor CCR7(lo)PD-1(hi) CXCR5(+) CD4(+) T cells indicate Tfh cell activity and promote antibody responses upon antigen reexposure. Immunity 2013, 39, 770–781. [Google Scholar] [CrossRef] [Green Version]
  300. Ueno, H.; Banchereau, J.; Vinuesa, C.G. Pathophysiology of T follicular helper cells in humans and mice. Nat. Immunol. 2015, 16, 142–152. [Google Scholar] [CrossRef] [Green Version]
  301. Moore, K.W.; de Waal Malefyt, R.; Coffman, R.L.; O’Garra, A. Interleukin-10 and the interleukin-10 receptor. Annu. Rev. Immunol. 2001, 19, 683–765. [Google Scholar] [CrossRef]
  302. Jia, X.; Zhai, T.; Wang, B.; Yao, Q.; Li, Q.; Mu, K.; Zhang, J.A. Decreased number and impaired function of type 1 regulatory T cells in autoimmune diseases. J. Cell. Physiol. 2019, 234, 12442–12450. [Google Scholar] [CrossRef]
  303. Sun, L.; Kong, R.; Li, H.; Wang, D. The Role of T Follicular Helper Cells and Interleukin-21 in the Pathogenesis of Inflammatory Bowel Disease. Gastroenterol. Res. Pract. 2021, 2021, 9621738. [Google Scholar] [CrossRef]
Figure 1. T-cell differentiation, subsets, and main functions. Naive CD4 T-cells can undergo differentiation into distinct effector subsets (e.g., Th1, Th2, Th9, Th17, and Th22 cells), follicular helper T (Tfh) cells, and regulatory phenotypes (Treg), each producing a characteristic set of cytokines (unfilled boxes next to cells). This differentiation process is mediated in part by the local cytokine microenvironment (arrows), which activates specific transcription factors and signaling molecules (text inside cells). Color-filled boxes next to cells list functions in homeostasis and in IBD (bold text). IBD is associated with changes in T-cell populations.
Figure 1. T-cell differentiation, subsets, and main functions. Naive CD4 T-cells can undergo differentiation into distinct effector subsets (e.g., Th1, Th2, Th9, Th17, and Th22 cells), follicular helper T (Tfh) cells, and regulatory phenotypes (Treg), each producing a characteristic set of cytokines (unfilled boxes next to cells). This differentiation process is mediated in part by the local cytokine microenvironment (arrows), which activates specific transcription factors and signaling molecules (text inside cells). Color-filled boxes next to cells list functions in homeostasis and in IBD (bold text). IBD is associated with changes in T-cell populations.
Ijms 24 02696 g001
Figure 2. T-cell subsets and functions in the intestinal mucosa in inflammatory bowel disease. The development of IBD is induced by multiple phenomena occurring in the gastrointestinal tract: microbial dysbiosis, disruption of the mucus layer, dysregulation of epithelial tight junctions, defects in the number and function of Paneth cells, and increased intestinal permeability. These events massively increase bacterial exposure. In this context, antigen-bearing DCs capture antigens and migrate to secondary lymphoid organs, where they present antigens to naive T-cells. Once activated, CD4 T-cells undergo proliferation and differentiation into different effector T-cell subsets (Th1, Th9, Th17, and Th2 cells). Differentiated Th cells migrate back to the gut, where they carry out inflammatory functions, such as production of IFN-γ in the case of Th1 cells or IL-17A (which plays an important role in recruiting neutrophils to sites of active inflammation) and IL-21 (which induces MMP production by stromal cells) in the case of Th17 cells. Cytokines released by Th1 cells favor activation of macrophages, which release TNF-α and trigger epithelial-cell apoptosis. Th9 cells produce IL-9, which can act as a proinflammatory cytokine, activating Th17 cells. The presence of IL-9 is associated with alterations in the expression of tight junctions, and intestinal overproduction of IL-9 is likely to impair epithelial-barrier integrity and compromise tolerance to commensal bacteria, eventually progressing to inflammation. IL-33 is upregulated in UC patients and drives a Th2-like cytokine response. Elevated IL-33 production Th2 cells have also been reported in UC patients. Proinflammatory signals in IBD are counterbalanced by IL-10 produced by Tregs. IL-22 released by Th22 cells maintains intestinal epithelial barrier function. In inflamed intestinal tissue, CD4 T-cells are a major source of IL-22BP, which blocks IL-22 signaling.
Figure 2. T-cell subsets and functions in the intestinal mucosa in inflammatory bowel disease. The development of IBD is induced by multiple phenomena occurring in the gastrointestinal tract: microbial dysbiosis, disruption of the mucus layer, dysregulation of epithelial tight junctions, defects in the number and function of Paneth cells, and increased intestinal permeability. These events massively increase bacterial exposure. In this context, antigen-bearing DCs capture antigens and migrate to secondary lymphoid organs, where they present antigens to naive T-cells. Once activated, CD4 T-cells undergo proliferation and differentiation into different effector T-cell subsets (Th1, Th9, Th17, and Th2 cells). Differentiated Th cells migrate back to the gut, where they carry out inflammatory functions, such as production of IFN-γ in the case of Th1 cells or IL-17A (which plays an important role in recruiting neutrophils to sites of active inflammation) and IL-21 (which induces MMP production by stromal cells) in the case of Th17 cells. Cytokines released by Th1 cells favor activation of macrophages, which release TNF-α and trigger epithelial-cell apoptosis. Th9 cells produce IL-9, which can act as a proinflammatory cytokine, activating Th17 cells. The presence of IL-9 is associated with alterations in the expression of tight junctions, and intestinal overproduction of IL-9 is likely to impair epithelial-barrier integrity and compromise tolerance to commensal bacteria, eventually progressing to inflammation. IL-33 is upregulated in UC patients and drives a Th2-like cytokine response. Elevated IL-33 production Th2 cells have also been reported in UC patients. Proinflammatory signals in IBD are counterbalanced by IL-10 produced by Tregs. IL-22 released by Th22 cells maintains intestinal epithelial barrier function. In inflamed intestinal tissue, CD4 T-cells are a major source of IL-22BP, which blocks IL-22 signaling.
Ijms 24 02696 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gomez-Bris, R.; Saez, A.; Herrero-Fernandez, B.; Rius, C.; Sanchez-Martinez, H.; Gonzalez-Granado, J.M. CD4 T-Cell Subsets and the Pathophysiology of Inflammatory Bowel Disease. Int. J. Mol. Sci. 2023, 24, 2696. https://doi.org/10.3390/ijms24032696

AMA Style

Gomez-Bris R, Saez A, Herrero-Fernandez B, Rius C, Sanchez-Martinez H, Gonzalez-Granado JM. CD4 T-Cell Subsets and the Pathophysiology of Inflammatory Bowel Disease. International Journal of Molecular Sciences. 2023; 24(3):2696. https://doi.org/10.3390/ijms24032696

Chicago/Turabian Style

Gomez-Bris, Raquel, Angela Saez, Beatriz Herrero-Fernandez, Cristina Rius, Hector Sanchez-Martinez, and Jose M. Gonzalez-Granado. 2023. "CD4 T-Cell Subsets and the Pathophysiology of Inflammatory Bowel Disease" International Journal of Molecular Sciences 24, no. 3: 2696. https://doi.org/10.3390/ijms24032696

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

Article Metrics

Back to TopTop