Journal of Advances In Allergy & Immunologic Diseases

ISSN: 2575-6184

Impact Factor: 0.431

VOLUME: 3 ISSUE: 1

The Differentiation and Roles of inflammatory Cytokine in the initiation of Inflammatory Bowel Diseases (IBD)


Affiliation

ALHASSAN Hussaini Mohammed (PhD)1,2, ALI-AKBAR Saboor-Yaraghi (PhD)2

1. Department of Immunology, Faculty of Medical Laboratory Sciences. Usmanu Danfodiyo University, Sokoto - Nigeria.

2. Department of Immunology, School of Public Health, Tehran University of Medical Sciences, International Campus. TUMS-IC, Tehran- Iran.

Citation

Alhassan Hussaini Mohammed, The Differentiation and Roles of inflammatory Cytokine in the initiation of Inflammatory Bowel Diseases (IBD)(2021)Journal of Advances In Allergy & Immunologic Diseases 3(1)

Abstract

The activation, proliferation and differentiation of naive T-cells are essentially significant in the productions of inflammatory cytokines and maintenance of immune homeostasis and development of various forms of inflammatory disorders. This activation and ultimate differentiation of this group of cells is brought about by the presence of an antigen and other inflammatory mediators in the micro-environment. Inflammatory cytokines like, IL-12 and IL-4 are of great significance in the resultant differentiation of these cells into various subsets, such as, the Th1, Th2, Th17 and T-regs, each of these subsets has a pattern and a specific cytokine profile, an imbalance in this profile may lead to the development of inflammation or other undesirable inflammatory responses, such as observed in inflammatory bowel diseases (IBD).

Crohn’s disease (CD) is a T helper-1-derived autoimmune disease while Ulcerative colitis (UC) are viewed as a T-helper 2-mediated inflammatory condition. Based on the role of this cytokine, they are categorized into pro-inflammatory and antiinflammatory cytokines. The pro-inflammatory cytokines, such as IL-12, IL-2, IL-17, IL-6, TNF-αand INF-γ are responsible for various forms of inflammatory responses. The aim of this review is to look into current research information about Naive T-cells proliferation and the roles of inflammatory cytokine in the initiation and development of inflammatory bowel diseases. Concrete understanding of the functional proliferation and development of this cells may contribute to finding reliable therapeutic modalities for the treatment of inflammatory bowel diseases.

Keywords:  Naive-T cell, Ulcerative Colitis, Crohn’s Diseases, cytokines, Dendritic cell

Introduction

Inflammatory bowel disease (IBD) is an inflammation of the bowel which can be idiopathic and chronic.  The aetiologic origins of IBD are yet to be fully understood, but available research report has attributed its emergence to genetic susceptibility, immune dysregulations, environmental factors or bacterial infections [1]. IBD is categorized into two major forms: Ulcerative colitis (UC) and Crohn’s disease (CD), each of this type has its own distinguishing characteristic, but generally, the development of UC and CD are characterised by Neutrophil recruitment, activation and infiltration of intestinal innate immune responses [2].

Both in children and adults, IBD is recognized as an important cause of gastrointestinal disorders. The disease has a worldwide occurrence, but it is more common in the industrialized nations such as the United States, United Kingdom, and Scandinavian countries than in the developing countries. The prevalence rates of IBD are between 40 to 100/ 100,000 persons [3], about 20% of all patients with IBD develop symptoms during childhood, with approximately, 6% being diagnosed before 10 years of life, 25% of children affected with IBD have a positive family history of IBD.IBD is of major types, Ulcerative colitis and Crohn’s diseases [4].

Ulcerative colitis (UC) is an inflammatory disorder in which the morphological changes are restricted to the colon [5], while in Crohn’s disease (CD), the changes may extend from the oesophagus to the anus, it may transcend all the length of the gastrointestinal tract, but the ileocecal region and the terminal ileum are the most commonly affected location [6].

 

ACTIVATION AND DIFFERENTIATION OF NAÏVE T-CELLS

The most important event in the differentiation of naive T-cells, is antigenic stimulation, which as a result of the interaction of CD4+ T-cells and T-cells receptor (TCR) with the major histocompatibility antigen complex II (MHC II), by an antigen presenting cells (APCs). The coupling of CD3 with TCR induces signal transduction, that activates naive-T-cells differentiation into effector and regulatory T-cells [7]. The predominant classes of cytokines produce depend on, the types and presence of inducing cytokines, the concentration of antigens, and type of APCs, and co-stimulatory molecules [7].

Dendritic cells (DCs) are a type of APCs, that has a high degree of potency in naive T cell sensitization [7]. This is done through the pattern recognition receptors, such as the toll-like receptors (TLR) and nucleotide oligomerization domain-like receptors (NOD) present on the cell surface [8]. There are various subsets of DCs, which can interfere in the proliferations of T-cell, for instance, in mice, CD8α+ DC was linked with a Th1 subset in the presence of IL-12, while the CD8α− subsets were linked to Th2 differentiation, under the influence of IL-6[9]. When dendritic cells are activated, direct stimulatory signals are sent to naive T-cells, which causes its Differentiation in effector T-cells and regulatory T-cells [10]. Both the effector and regulatory T-cells undergo further differentiation to form Th1, Th2, Th17 and ITreg. The balance between Th1/Th17 and Th2/Tregs is significant in the initiation and development of IBD [10]

 

THE DIFFERENTIATION OF NAÏVE T-CELLS TO TH1 SUBSETS.

The differentiation of Naive T-cell to Th1 is highly depended on the presence of interferon gamma (IFN-γ) and interleukin 12(IL-12). An exaggerated production of the Th1-derived cytokines which are mainly pro-inflammatory in action could leads to an undesirable inflammatory response, which may bring about the development of various forms of autoimmune diseases, such as IBD [11]. Large amounts of IL-12 are secreted by APCs after their initial activation by an antigen, through the pattern recognition receptors present on the cell surface [9,10] and this is followed by natural killer cells (NK) induction by IL-12 to produce IFN-γ. Th1 differentiation is regulated by the master regulator, T-bet transcription factor (T-box). T-bet possesses the ability to suppress and down-regulates the development of other T-cell subsets, such as Th2 and Th17, while facilitating IFNγ production [12].

RORγt in mouse and RORc in human is the principal transcription factor of Th17, the Th17 inhibition is brought about by the constant interaction of T-bet with race promoter, which encodes RORγt [13]. The inhibition of Th2 by T-bet is initiated by constant and frequent suppression of IL-4 transcription gene, obstructing the activity of GATA3[14]. IFNγ production can be induce by STAT4, creating a positive feedback mechanism for T-bet and IL12Rβ2 expression. T-bet and STAT4 are directly involved in locus transcription of IFNγ, while GATA3 and STAT6, involvement in Th2 differentiation formed repressive histone marks at IFN-γ locus, this is an indication that Th1 differentiation is determined by the activation of IFNγ locus [15]. Research findings have reported the role of T-bet’s as a repressor of transcription, this is by Bcl-6, a transcriptional repressor induction, which suppresses the effects of IFNγ locus of Th1 proliferation in secondary responses, this may functions as a protective model aimed at inhibiting IBD Immunopathogenesis by downregulating aberrant production of IFNγ [16].

 

THE DIFFERATION OF NAIVE T-CELLS TO TH2 SUBSETS

The Naive T-cells differentiation to Th2 subsets depends majorly on the presence of IL-4 and IL-2. STAT6 is induced by IL-4, which is an essential transcription factor in the differentiation of Th2 cell. Gata3 expression is up-regulated by the IL-4-induced STAT6[17]. Three different postulations that explain these mechanisms of Th2 differentiation are:

  • Selective differentiation of Th2 subsets through Gfi-1 recruitment, and
  • Inhibition of Th1 differentiation by direct inhibition of T-bet functions.
  • Enhanced Th2 subsets of cytokine production,

These mechanisms suppress Th1 differentiation by downregulating STAT4[18] and inhibition of GATA3 leads to the blockage of Th2 differentiation [19]. Studies have shown that GATA3 needed the assistance of STAT6 to control all the Th2-specific genes [20}, besides GATA3, Th2 cell differentiation involves several other cytokines, such as IL-2, IL-6, and IL-21.

The activation of signal transducer and activator of transcription-5 (STAT-5) is purely independent on IL-4 but dependent on IL-2 signalling [21]. Since GATA3 cannot activate all Th2-specific genes, a synergistic action of STAT5 and GATA3 is required, alone GATA3 cannot cause the production of IL4, this is because GATA3 and STAT5 selectively bind to different epitopes on the IL4 locus. GATA-3 binds to DNaseI hypersensitive site VA and CNS-1 sites of the IL4/IL13 loci and STAT5 binds to the DNase I hypersensitive sites (HSII and HSIII) on the second intron of the IL4 locus and [18].

In Th2 differentiation, IL6 has a dual function. It enhances Th2 differentiation, while on the other hands, inhibiting the Th1 production [22]. IL-6 also induces IL-4 production by naive T-cells, through the upregulation of nuclear factor of the activated T cells (NFAT). The downregulation of the Th1 differentiation occurs by the IL-6-induced upregulation of suppressor of cytokine signaling-1 (SOCS1) expression, which inhibits STAT1 activation, reducing IFN-γ signalling [22].

 

THE DIFFERENTIATION OF NAÏVE T-CELLS TO Th-17

The definition of Th17 majorly relies on TGF-β, IL-6, IL-21, and IL-23, with the participation of the master regulator, Retinoid-related orphan receptor gamma-T (RORγt). The Naïve T-cells differentiation to Th17 has three main components, these are:

  • The differentiation of naïve –T-cells mediated by TGF-β and IL-6,
  • The self-amplification step mediated by IL-21, and
  • The stabilization step mediated by IL-23.

The transforming growth factor-β (TGF-β) is one of the most important and principal cytokines in Th17 differentiation [23]. It is signalling plays important roles in inducible T-regulatory cells(I-Treg) development. I-Treg and Th17 are an antagonist in their mode of action to one another. At higher concentration TGF-β can divert the differentiation of Naive T-cells towards ITreg development, through FOXP3 induction [24], but under the influence of IL-6, and at a lower concentration, TGF-β initiates Th17 definition, up-regulations of IL23R expression, and IL-21 production. Suppression of TGF-β signalling pathway in the presence of IL6 leads to the activation of RORγt [25].

 

THE DIFFERENTIATION OF NAÏVE T-CELLS TO REGULATORY T CELLS

The TGF-β is responsible for the initiation of Inducible T-regs (ITreg) [26]. I-Treg cells are FOXP3+CD4+CD25+cells, that develop in the peripheral lymphoid organs after priming with antigen, unlike natural Treg (nTreg), which are released from the thymus as a separate line from an already expressed FOXP3 [26]. TGF-β is a critical cytokine responsible for initiation of adherence to iTreg cell lines. The Forkhead transcription factor, FOXP3 is specifically expressed in CD4 + CD25 + Treg cells and is a major factor involved in iTreg differentiation [27].

FOXP3, and other transcription factors, such as Smad2 and Smad3, which are also activated via the TGF-signalling pathways, are involved in the iTreg differentiation [28]. Smad3 can differentiate the development of iTreg by enhancing the regulation of FOXP3 expression and inhibiting Th17 differentiation by blocking RORγT. NFAT through interaction with FOXP3 contributed to the differentiation of Th17 [28]. It was found that STAT5 enhances the expression of FOXP3, and then proceeds to signal transduction of FOXP3 and promotes the development of iTreg [29]. STAT5 and STAT3, which communicate with several common sites in the IL17 locus, closely antagonizd each other. Activation of STAT5 by the IL2 signal blocked the binding of STAT3 to the sites on the locus and, consequently, enhances the differentiation of iTreg [29]. The disruption of the interaction of the mutated FOXP3 gene and NFAT resulted in a decrease in the expression of Treg-CTLA4 and CD25 markers [30]

 

INFLAMMATORY CYTOKINES AND IBD PATHOGENESIS

ROLE OF IL-12

The IL-12 superfamily is made up of IL-12, IL-23, IL-27 and IL-35. IL-12 and IL-23 are produced by antigen-presenting cells, mainly, dendritic cells and phagocytes [31,32]. Up-regulation of IL-12 expression was seen in the mucosa of patients with UC, and it correlated with the activity of the disease [33]. Research has shown that, in macrophages and mucosal immunity, the basic protein of leucine zipper, NFIL3, is the regulator of IL-12p40 [34]. The interaction of macrophages with the microflora of the colon induced NFIL3 to down -regulate their inflammatory ability. IL-23 facilitates the proliferation and differentiation of naive CD4+T cells into Th17 cells [35], It is found and widely distributed in virtually all the immune cells as its expressed by CD4+T cells, CD8+T cells, NKT cells, and NK cells, [36]. IL-23 plays a pro-inflammatory role in colitis, which is mediated by T-cells, caused by 2,4,6-trinitrobenzenesulfonic acid (TNBS), IL-23 can also be termed to have an anti-inflammatory function because it suppresses production of IL-12 [37]. The signalling of IL-23 occurs mainly through the transition molecule STAT3. IL-23 also facilitates the proliferation of naive T-cells into regulatory T cells (Tregs) [38].

IL-12 expression is increased in the mucosa of patients with active Crohn (CD) [39], and due to the up-regulation of the levels of IL-12p40 and IL-12Rβ2 in the primary phase of CD initiation, it is said that IL-12 may participate in the early induction of Th1 -polarization of naive T-cells [40]. IL-12- dependent synthesis of IFN-γ of mucosal T cells can be facilitated by cytokines that feed the signal through a common γ-chain receptor [41]. IL-23 has an elevated level of mucosa in Crohn's disease [42]. The production of IL-17 and IL-6 appears to mediate IL-23-induced inflammation of the intestine [43].

IL-27 is described as a heterodimeric novel member of the IL-12 family. In in-vitro studies, it was shown that IL-27 is produced by the presence of activated dendritic cells and monocytes, which initiates the proliferation of naive CD4+T cells and co-ordinates with IL-12 for IFN-γ production [43].

 

ROLE OF TNF-α

The family of TNF comprises of 18-type 2-protein molecules which occur either as a membrane bound or soluble form, their receptors are transmembrane proteins type 1 [44]. The receptor binding of TNF-like ligands triggers intracellular responses that are involved in the proliferation and differentiation and ultimate survival of the cells. TNF-α is the main and master cytokine in the pathogenesis of IBD [45]. It manifests its effects by the secretion of adhesion molecules, the proliferation of fibroblasts, and pro-coagulant factors, more-so through the initiation of cytotoxic, apoptotic and acute phase reactions [46]. These basic cytokines have the ability to increase the production of IL-1β, IL-6 and IL-33, and also modulate the expression of ST2 in epithelial cells [47].

Serum TNF-α levels correlated with ulcerative colitis (UC) and CD clinical activity [48]. Their roles in inflammation of the colon were the basis for the anti-TNF-α antibodies therapy in IBD. The tumor necrosis factor (TL1A), is another TNF family member, it stimulates the secretion of IFN-γ by binding to the death receptor 3 (DR3) [49]. Binding of TNF-α to the soluble TNF receptor 1 and 2 (sTNFR1 and 2) results in the initiation of inflammatory responses [50].

In CD patients, the levels of sTNFR1 and 2 are elevated when compared to UC and healthy controls, so it can be used as a biomarker for disease activity and a discriminatory factor between Ulcerative colitis and Crohn’s diseases [51]. The pro-inflammatory role of TNF-α has confirmed the use of antibodies against TNF-α in the treatment of CD [52]. Another important TNF family member is called the Light, it mediated pro-inflammatory activity in the human intestinal mucosa, and It was reported that in CD the IFN producing CD4 + T cells expresses LIGHT as a mediating Th1 response [53]

 

The roles of IL-17

IL-17 is produced by Th17 cells, it acts as a key mediator in increasing the production of chemokines and in the recruitment of monocytes and neutrophils in the inflammatory responses. IL-17A is Th17 cells, but in chronic inflammation, this cytokine is also produced by CD8+ T cells [54]. It was shown that IL-25 has a strong capacity to inhibit CD14+cell-derived cytokines, specifically, IL-12 production and experimental colitis caused by Th1 cells, which indicates its therapeutic potential in CD and UC [55].

The mucosal level of IL-17 in human is significantly increased in active CD also studies has also reported that, the level of circulating memory cells of CD161+cells, IL-17 and IFN-γ increases in patients with CD, [56]. More-so, there is an increased level of circulating IL-23R expressing T cells in patients with CD that respond to IL-23 with increased production of IL-17, IL-22 and IFN-γ, this is also enhanced by the presence of IL-1β [57]. In line with research findings, it was concluded that Th17 cells, producing both IL-17 and IFN-γ are identified as important effector cells in the inflammatory response in CD.

The production of IL-21 is significantly increased in the mucosa of CD [71]. IL-21 is produced exclusively by T-cells that produce CD4 + IFN-γ, but only a little proportion of IL-21 produces CD4+ T cells expressing IL-17A, which indicates that IL-21 is mainly produced by Th1, rather than Th17 subsets. Stimulation of CD4 + T-lymphocytes from the control GIT with an anti-CD3 antibody and IL-12 increases the number of IL-21-secreting Th1 cells. While the blockade of endogenous IL-12 in cell cultures of CD mucosa significantly reduces the percentage of IL-21 production [58].

 

Roles of IL-1

The IL-1 family has two structurally different forms: IL-1α and IL-1β. The endogenous IL-1 receptor antagonist (IL-1Ra) is a natural antagonist of IL-1, which maintain a healthy balance of immunity in the GIT [59]. An increase in the ratio of IL-1 / IL-1Ra is related to increase in disease activities in ulcerative colitis. IL-33 also known as IL-F11, has been identified as another the new family member of the IL-1 [60]. It involved is in allergy, inflammation of the respiratory tract and rheumatic diseases [61]. Schmitz et al., reported that IL-33 apparently, restores the protective barriers of the mucosa by increasing mucus secretion and enhancing the immune response through cytokines associated with type 2 helper T-cells (Th2) [60]. It has been shown that the expression of the IL-33 receptor, ST2, increases both in the wall of the colon and in the serum of patients with IBD [62]. In the large intestine, the axis of IL-33 / ST2 may have multiple functions in inflammatory bowel disease (IBD) pathogenesis. Stimuli of cytokines, such as TNF-α and IL-1β, and Immune responses from the pathogen-associated molecular pattern (PAMP) could result to up-regulation in the level of IL-33 in epithelial cells. In the case of epithelial damage, IL-33 is released from cells and facilitates immune responses via ST2, which expresses immune cells, this leads to an increase in the severity of the inflammation [63].

The proportional up-regulation of IL-1 / IL-1Ra is related to increase in flare in CD activities [64]. IL-18 is a member of the IL-1 family [65], In the inflamed mucosa of patients with CD, the level of IL-18 is up-regulated [66]. The balance between the proinflammatory cytokine and the IL-18 binding protein (IL-18BP), which is its natural inhibitor, has a huge contribution to the pathogenesis of IBD [67]. It has been reported by Leach et al., that IL-18, produced in the colon of patients with CD especially that in children, contributes significantly to local inflammatory responses and changes [68].

 

Roles of IL-6

The IL-6, IL-11, IL-31 are members of the IL-6 family. Circulating levels of IL-6 and sIL-6R correlate with disease activity [69] The pathogenic role of the IL-6 and sIL-6R signalling in interfering with T-cell resistance to CD apoptosis was confirmed by blocking IL-6 trans-signalling [70]. These research findings have shown that IL-6 can influence not only the chronic inflammatory pathways, but also the relapses that arise in the pathology of CD. The effects of IL-6 in IBD were demonstrated in Caco2 cells, where it was shown that IL-6 induces the activation of NF-kappa β and enhances the expression of intercellular adhesion 1 molecule. This adhesion molecule is important in the pathogenesis of IBD and is most likely required for the extra-intestinal manifestation of IBD [71]. All the available research data have suggested that blockade of the IL-6 / STAT3 signalling pathways and the use of antibodies against IL-6R have been suggested as a new and promising therapeutic strategy in the treatment of IBD [71].

 

Role of IL-8

IL-8 has been described as a heparin-binding small base protein, which belongs to the cysteine amino acid cysteine chemokine [72]. The activation, movement and neutrophilic migration in peripheral blood are primarily mediated by IL-8. Studies have shown that, in active UC, the level of tissue IL-8 was higher compared to normal tissue, more-so, its concentration in the serum was associated with the severity of UC.

Roles of IL-13

IL-13 is a Th2-derived cytokine, important in the development and manifestation of UC [73]. Expression of IL-13 mRNA in the mucosa of UC is increased and also, the ex vivo culture of LPMC from UC shows appreciable amounts of IL-13 during stimulation than that obtained from healthy and CD patients [73]. UC-specific CD161+NKT cells have a cytotoxic response against colon epithelium, whose effect, at least in part, depends on functional IL-13. Subsequent responses, such as the NKT cell cytotoxic activity, induced apoptosis of epithelial cells by IL-13, and disruption of dense compounds all constitute into an epithelial damage [74]. It has also been recently shown that IL-13 signalling through IL-13Rα2 leads to an increase in the production of the transforming growth factor (TGF) -β1, which favours the progression of colonic wall fibrosis [75].

 

Roles of IL-5 AND IL-21

IL-5 is a selective eosinophil-activating hormone, it is also referred to as an eosinophil differentiation factor. IL-5 belongs to the family member of common β-chain-dependent cytokine. A higher amount of IL-5 is produced in mononuclear cells in active UC, but not in CD [76]. IL-5 appears to play a regulatory role in the regeneration of eosinophils in the mucosa of UC [77].

IL-21 is a T-cell derived member of common γ-chain-dependent cytokine family, it acts to maintain Th1-mediated inflammation in the colon epithelium, inducing the production of IFN-γ [78]. The number of IL-21 cells is much higher in CD than seen in UC [79]. Research has also indicated that IL-21 enhances the expansion of NK cells [80]. It is expressed by immune T-and B-cells and non-immune cells, such as fibroblasts, where it activates the production of metalloproteinase 1 and transmits signals through its IL-21R receptor, it activates STAT-3 in T cells [81]. IL-21, like IL- 6 and IL-23, also participates in the proliferation of Th17 cells and is overexpressed in both CD and UC, with higher levels found in CD [82].

Conclusion

IBD is the result of an aberrant immunological response of Naive T–cells to exaggerated antigenic stimulation, which leads to a dysregulated immune response. The result of this review has shown that inflammatory cytokine is not only related to the maintenance of the immune haemostasis but are also concerned with protections against foreign invasion. So concerted research effort should be directed at understanding the immunological and signalling pathways and the exact mechanisms that are involved in the initiation of exaggerated immune responses. Understanding the immunological pathways underlying the pathogenesis will not only assist in the treatment of IBD but will also help in alleviating the burden of very many other inflammatory disorders and autoimmune diseases.

Acknowledgement

I wish to express my sincere appreciation to the management of Tehran University of Medical Sciences, Management of TUMS-International Campus, for providing me with an enabling environment and making available their facilities for this review.

References

  1. H. Alhassan Mohammed*, A Mirshafiey, H Vahedi, G Hemmasi, A. Moussavi Nasl Khameneh, K. Parastouei & A.A. Saboor-Yaraghi. Immunoregulation of inflammatory and inhibitory cytokines by Vitamin D3 in patients with inflammatory bowel diseases (IBD). Scandinavian Journal of Immunology 2017, 85(6):386-394, ISSN: 0300-9475. (DOI: 10.1111/sji.12547) PMid:28332200

    View Article      PubMed/NCBI     
  2. Korzenik, J. R.; Dieckgraefe, B. K.; Valentine, J. F.; Hausman, D. F.; Gilbert, M. J. Sargramostim for active Crohn's disease. New England Journal of Medicine 2005, 352, 2193-2201. PMid:15917384

    View Article      PubMed/NCBI     
  3. Kirsner, J. B.; Shorter, R. G. Recent developments in nonspecific inflammatory bowel disease. New England Journal of Medicine 1982, 306, 775-785. PMid:7038488

    View Article      PubMed/NCBI     
  4. Mir-Madjlessi, S. H.; Michener, W. M.; Farmer, R. G. Course and prognosis of idiopathic ulcerative proctosigmoiditis in young patients. Journal of paediatric gastroenterology and nutrition 1986, 5, 570-575.

    View Article           
  5. Michener, W. M.; Whelan, G.; Greenstreet, R. L.; Farmer, R. G. Comparison of the clinical features of Crohn's disease and ulcerative colitis with onset in childhood or adolescence. Cleveland Clinic Quarterly 1982, 49, 13. PMid:7094330

    View Article      PubMed/NCBI     
  6. Casini-Raggi, V.; Kam, L.; Chong, Y.; Fiocchi, C.; Pizarro, T. T.; Cominelli, F. Mucosal imbalance of IL-1 and IL-1 receptor antagonist in inflammatory bowel disease. A novel mechanism of chronic intestinal inflammation. The Journal of Immunology 1995, 154, 2434-2440.

  7. 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. The Journal of Immunology 1996, 157, 1261-1270.

    View Article           
  8. Bamias, G.; Nyce, M. R.; Sarah, A.; Cominelli, F. New concepts in the pathophysiology of inflammatory bowel disease. Annals of internal medicine 2005, 143, 895-904. PMid:16365470

    View Article      PubMed/NCBI     
  9. Tao, X.; Constant, S.; Jorritsma, P.; Bottomly, K. Strength of TCR signal determines the costimulatory requirements for Th1 and Th2 CD4+ T cell differentiation. The Journal of Immunology 1997, 159, 5956-5963.

  10. Steinman, R. M.; Hawiger, D.; Nussenzweig, M. C. Tolerogenic dendritic cells. Annual review of immunology 2003, 21, 685-711. PMid:12615891

    View Article      PubMed/NCBI     
  11. Kohu, K.; Ohmori, H.; Wong, W. F.; Onda, D.; Wakoh, T.; Kon, S.; Yamashita, M.; Nakayama, T.; Kubo, M.; Satake, M. The Runx3 transcription factor augments Th1 and down-modulates Th2 phenotypes by interacting with and attenuating GATA3. The Journal of Immunology 2009, 183, 7817-7824. PMid:19933870

    View Article      PubMed/NCBI     
  12. Alhassan Mohammed, H.; Saboor-Yaraghi, A. A.; Mirshafiey, A.; Vahedi, H.; Shiri-Shahsavar, M. R.; Mousavi Nasl Khameneh, A. Immunomodulatory and Immunosuppressive roles of 1α, 25 (OH) 2D3 In Autoimmune Diseases. Scandinavian Journal of Immunology, 2017, 85, 95-103 PMid:27896829

    View Article      PubMed/NCBI     
  13. Trinchieri, G.; Pflanz, S.; Kastelein, R. A. The IL-12 family of heterodimeric cytokines: new players in the regulation of T cell responses. Immunity 2003, 19, 641-644. 00296-6

    View Article           
  14. Afkarian, M.; Sedy, J. R.; Yang, J.; Jacobson, N. G.; Cereb, N.; Yang, S. Y.; Murphy, T. L.; Murphy, K. M. T-bet is a STAT1-induced regulator of IL-12R expression in naive CD4+ T cells. Nature immunology 2002, 3, 549-557. PMid:12006974

    View Article      PubMed/NCBI     
  15. Lazarevic, V.; Chen, X.; Shim, J.-H.; Hwang, E.-S.; Jang, E.; Bolm, A. N.; Oukka, M.; Kuchroo, V. K.; Glimcher, L. H. T-bet represses TH17 differentiation by preventing Runx1-mediated activation of the gene encoding RORℽT. Nature immunology 2011, 12, 96-104. PMid:21151104

    View Article      PubMed/NCBI     
  16. Hwang, E. S.; Szabo, S. J.; Schwartzberg, P. L.; Glimcher, L. H. T helper cell fate specified by kinasemediated interaction of T-bet with GATA-3. Science 2005, 307, 430-433. PMid:15662016

    View Article      PubMed/NCBI     
  17. Aune, T. M.; Collins, P. L.; Chang, S. Epigenetics and T helper 1 differentiation. Immunology 2009, 126, 299-305. PMid:19178593

    View Article      PubMed/NCBI     
  18. Oestreich, K. J.; Huang, A. C.; Weinmann, A. S. The lineage-defining factors T-bet and Bcl-6 collaborate to regulate Th1 gene expression patterns. Journal of Experimental Medicine 2011, Jem. 20102144. PMid:21518797

    View Article      PubMed/NCBI     
  19. Kaplan, M. H.; Schindler, U.; Smiley, S. T.; Grusby, M. J. Stat6 is required for mediating responses to IL-4 and for the development of Th2 cells. Immunity 1996, 4, 313-319. 80439-2

    View Article           
  20. Zhu, J.; Yamane, H.; Cote-Sierra, J.; Guo, L.; Paul, W. E. GATA-3 promotes Th2 responses through three different mechanisms: induction of Th2 cytokine production, selective growth of Th2 cells and inhibition of Th1 cell-specific factors. Cell research 2006, 16, 3-10. PMid:16467870

    View Article      PubMed/NCBI     
  21. Zhu, J.; Min, B.; Hu-Li, J.; Watson, C. J.; Grinberg, A.; Wang, Q.; Killeen, N.; Urban, J. F.; Guo, L.; Paul, W. E. Conditional deletion of Gata3 shows its essential function in TH1-TH2 responses. Nature immunology 2004, 5, 1157-1165. PMid:15475959

    View Article      PubMed/NCBI     
  22. Horiuchi, S.; Onodera, A.; Hosokawa, H.; Watanabe, Y.; Tanaka, T.; Sugano, S.; Suzuki, Y.; Nakayama, T. Genome-wide analysis reveals unique regulation of transcription of Th2-specific genes by GATA3. The Journal of Immunology 2011, 186, 6378-6389. PMid:21536806

    View Article      PubMed/NCBI     
  23. Kim, J. I.; Ho, I. -C.; Grusby, M. J.; Glimcher, L. H. The transcription factor c-Maf controls the production of interleukin-4 but not other Th2 cytokines. Immunity 1999, 10, 745-751. 80073-4

    View Article           
  24. Diehl, S.; Anguita, J.; Hoffmeyer, A.; Zapton, T.; Ihle, J. N.; Fikrig, E.; Rincon, M. Inhibition of Th1 differentiation by IL-6 is mediated by SOCS1. Immunity 2000, 13, 805-815. 00078-9

    View Article           
  25. Veldhoen, M.; Hocking, R. J.; Atkins, C. J.; Locksley, R. M.; Stockinger, B. TGFβ in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 2006, 24, 179-189. PMid:16473830

    View Article      PubMed/NCBI     
  26. Zhou, L.; Lopes, J. E.; Chong, M. M.; Ivanov, I. I.; Min, R.; Victora, G. D.; Shen, Y.; Du, J.; Rubtsov, Y. P.; Rudensky, A. Y. TGF-β-induced Foxp3 inhibits TH17 cell differentiation by antagonizing RORγt function. Nature 2008, 453, 236-240. PMid:18368049

    View Article      PubMed/NCBI     
  27. Yang, X. O.; Pappu, B. P.; Nurieva, R.; Akimzhanov, A.; Kang, H. S.; Chung, Y.; Ma, L.; Shah, B.; Panopoulos, A. D.; Schluns, K. S. T helper 17 lineage differentiation is programmed by orphan nuclear receptors RORα and RORγ. Immunity 2008, 28, 29- PMid:18164222

    View Article      PubMed/NCBI     
  28. Fontenot, J. D.; Gavin, M. A.; Rudensky, A. Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature Immunology 2003, 4, 330-336. PMid:12612578

    View Article      PubMed/NCBI     
  29. Yagi, H.; Nomura, T.; Nakamura, K.; Yamazaki, S.; Kitawaki, T.; Hori, S.; Maeda, M.; Onodera, M.; Uchiyama, T.; Fujii, S. Crucial role of FOXP3 in the development and function of human CD25+ CD4+ regulatory T cells. International immunology 2004, 16, 1643-1656. PMid:15466453

    View Article      PubMed/NCBI     
  30. Tone, Y.; Furuuchi, K.; Kojima, Y.; Tykocinski, M. L.; Greene, M. I.; Tone, M. Smad3 and NFAT cooperate to induce Foxp3 expression through its enhancer. Nature Immunology 2008, 9, 194-202. PMid:18157133

    View Article      PubMed/NCBI     
  31. Laurence, A.; Tato, C. M.; Davidson, T. S.; Kanno, Y.; Chen, Z.; Yao, Z.; Blank, R. B.; Meylan, F.; Siegel, R.; Hennighausen, L. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity 2007, 26, 371-381. PMid:17363300

    View Article      PubMed/NCBI     
  32. Wu, Y.; Borde, M.; Heissmeyer, V.; Feuerer, M.; Lapan, A. D.; Stroud, J. C.; Bates, D. L.; Guo, L.; Han, A.; Ziegler, S. F. FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell 2006, 126, 375-387. PMid:16873067

    View Article      PubMed/NCBI     
  33. Awasthi, A.; Carrier, Y.; Peron, J. P.; Bettelli, E.; Kamanaka, M.; Flavell, R. A.; Kuchroo, V. K.; Oukka, M.; Weiner, H. L. A dominant function for interleukin 27 in generating interleukin 10-producing anti-inflammatory T cells. Nature Immunology 2007, 8, 1380-1389. PMid:17994022

    View Article      PubMed/NCBI     
  34. Apetoh, L.; Quintana, F. J.; Pot, C.; Joller, N.; Xiao, S.; Kumar, D.; Burns, E. J.; Sherr, D. H.; Weiner, H. L.; Kuchroo, V. K. The Aryl hydrocarbon Receptor (AhR) interacts with c-Maf to promote the differentiation of IL-27-induced regulatory type 1 (TR1) cells. Nature immunology 2010, 11, 854. PMid:20676095

    View Article      PubMed/NCBI     
  35. Watford, W. T.; Hissong, B. D.; Bream, J. H.; Kanno, Y.; Muul, L.; O'shea, J. J. Signaling by IL-12 and IL-23 and the immunoregulatory roles of STAT4. Immunological reviews 2004, 202, 139-156. PMid:15546391

    View Article      PubMed/NCBI     
  36. Trinchieri, G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nature Reviews Immunology 2003, 3, 133-146. PMid:12563297

    View Article      PubMed/NCBI     
  37. Nielsen, O.; Kirman, I.; Rudiger, N.; Hendel, J.; Vainer, B. Upregulation of interleukin-12 and-17 in active inflammatory bowel disease. Scandinavian journal of gastroenterology 2003, 38, 180-185. PMid:12678335

    View Article      PubMed/NCBI     
  38. Kobayashi, T.; Matsuoka, K.; Sheikh, S. Z.; Elloumi, H. Z.; Kamada, N.; Hisamatsu, T.; Hansen, J. J.; Doty, K. R.; Pope, S. D.; Smale, S. T. NFIL3 is a regulator of IL-12 p40 in macrophages and mucosal immunity. The Journal of Immunology 2011, 186, 4649-4655. PMid:21383239

    View Article      PubMed/NCBI     
  39. Iwakura, Y.; Ishigame, H. The IL-23/IL-17 axis in inflammation. The Journal of clinical investigation 2006, 116, 1218-1222. PMid:16670765

    View Article      PubMed/NCBI     
  40. Maul, J.; Zeitz, M. Ulcerative colitis: immune function, tissue fibrosis and current therapeutic considerations. Langenbeck's Archives of Surgery 2012, 397, 1-10. PMid:21479621

    View Article      PubMed/NCBI     
  41. Cho, J. H.; Brant, S. R. Recent insights into the genetics of inflammatory bowel disease. Gastroenterology 2011, 140, 1704-1712. e1702. PMid:21530736

    View Article      PubMed/NCBI     
  42. Ivanov, I. I.; McKenzie, B. S.; Zhou, L.; Tadokoro, C. E.; Lepelley, A.; Lafaille, J. J.; Cua, D. J.; Littman, D. R. The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006, 126, 1121-1133. PMid:16990136

    View Article      PubMed/NCBI     
  43. Műzes, G.; Molnar, B.; Sipos, F. Regulatory T cells in inflammatory bowel diseases and colorectal cancer. World J Gastroenterol 2012, 18, 5688-5694. PMid:23155308

    View Article      PubMed/NCBI     
  44. Peluso, I.; Pallone, F.; Monteleone, G. Interleukin-12 and Th1 immune response in Crohn's disease: Pathogenetic relevance and therapeutic implications. World journal of gastroenterology 2006, 12, 5606. PMid:17007011

    View Article      PubMed/NCBI     
  45. Schmidt, C.; Giese, T.; Ludwig, B.; Mueller-Molaian, I.; Marth, T.; Zeuzem, S.; Meuer, S. C.; Stallmach, A. Expression of interleukin-12-related cytokine transcripts in inflammatory bowel disease: elevated interleukin-23p19 and interleukin-27p28 in Crohn's disease but not in ulcerative colitis. Inflammatory bowel diseases 2005, 11, 16-23. PMid:15674109

    View Article      PubMed/NCBI     
  46. Yen, D.; Cheung, J.; Scheerens, H.; Poulet, F.; McClanahan, T.; Mckenzie, B.; Kleinschek, M. A.; Owyang, A.; Mattson, J.; Blumenschein, W. IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6. The Journal of clinical investigation 2006, 116, 1310-1316. PMid:16670770

    View Article      PubMed/NCBI     
  47. Locksley, R. M.; Killeen, N.; Lenardo, M. J. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 2001, 104, 487-501. 00237-9

    View Article           
  48. Smith, C. A.; Farrah, T.; Goodwin, R. G. The TNF receptor superfamily of cellular and viral proteins: activation, costimulation, and death. Cell 1994, 76, 959-962. 90372-7

    View Article           
  49. Baumann, H.; Gauldie, J. The acute phase response. Immunology today 1994, 15, 74-80. 90137-6

    View Article           
  50. Sanchez-Munoz, F.; Dominguez-Lopez, A.; Yamamoto-Furusho, J. K. Role of cytokines in inflammatory bowel disease. World J Gastroenterol 2008, 14, 4280-4288. PMid:18666314

    View Article      PubMed/NCBI     
  51. Reimund, J.-M.; Wittersheim, C.; Dumont, S.; Muller, C.; Baumann, R.; Poindron, P.; Duclos, B. Mucosal inflammatory cytokine production by intestinal biopsies in patients with ulcerative colitis and Crohn's disease. Journal of clinical immunology 1996, 16, 144-150. PMid:8734357

    View Article      PubMed/NCBI     
  52. Bamias, G.; Martin, C.; Marini, M.; Hoang, S.; Mishina, M.; Ross, W. G.; Sachedina, M. A.; Friel, C. M.; Mize, J.; Bickston, S. J. Expression, localization, and functional activity of TL1A, a novel Th1-polarizing cytokine in inflammatory bowel disease. The Journal of Immunology 2003, 171, 4868-4874. PMid:14568967

    View Article      PubMed/NCBI     
  53. Podolsky, D. K. The current future understanding of inflammatory bowel disease. Best practice & research Clinical Gastroenterology 2002, 16, 933-943. PMid:12473299

    View Article      PubMed/NCBI     
  54. Spoettl, T.; Hausmann, M.; Klebl, F.; Dirmeier, A.; Klump, B.; Hoffmann, J.; Herfarth, H.; Timmer, A.; Rogler, G. Serum soluble TNF receptor I and II levels correlate with disease activity in IBD patients. Inflammatory bowel diseases 2007, 13, 727-732. PMid:17260368

    View Article      PubMed/NCBI     
  55. Klotz, U. The pharmacological profile and clinical use of Mesalazine (5-aminosalicylic acid). Arzneimittelforschung 2012, 62, 53-58. PMid:22344548

    View Article      PubMed/NCBI     
  56. Cohavy, O.; Zhou, J.; Granger, S. W.; Ware, C. F.; Targan, S. R. LIGHT expression by mucosal T cells may regulate IFN-γ expression in the intestine. The Journal of Immunology 2004, 173, 251-258. PMid:15210782

    View Article      PubMed/NCBI     
  57. 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-β induces development of the TH17 lineage. Nature 2006, 441, 231-234. PMid:16648837

    View Article      PubMed/NCBI     
  58. Caruso, R.; Sarra, M.; Stolfi, C.; Rizzo, A.; Fina, D.; Fantini, M. C.; Pallone, F.; MacDonald, T. T.; Monteleone, G. Interleukin-25 inhibits interleukin-12 production and Th1 cell-driven inflammation in the gut. Gastroenterology 2009, 136, 2270-2279. PMid:19505427

    View Article      PubMed/NCBI     
  59. Kleinschek, M. A.; Boniface, K.; Sadekova, S.; Grein, J.; Murphy, E. E.; Turner, S. P.; Raskin, L.; Desai, B.; Faubion, W. A.; de Waal Malefyt, R. Circulating and gut-resident human Th17 cells express CD161 and promote intestinal inflammation. Journal of Experimental Medicine 2009, 206, 525-534. PMid:19273624

    View Article      PubMed/NCBI     
  60. Strober, W.; Zhang, F.; Kitani, A.; Fuss, I.; Fichtner-Feigl, S. Pro-inflammatory cytokines underlying the inflammation of Crohn's disease. Current opinion in gastroenterology 2010, 26, 310. PMid:20473158

    View Article      PubMed/NCBI     
  61. Rochman, Y.; Spolski, R.; Leonard, W. J. New insights into the regulation of T cells by γc family cytokines. Nature Reviews Immunology 2009, 9, 480-490. PMid:19543225

    View Article      PubMed/NCBI     
  62. Casini-Raggi, V.; Kam, L.; Chong, Y.; Fiocchi, C.; Pizarro, T. T.; Cominelli, F. Mucosal imbalance of IL-1 and IL-1 receptor antagonist in inflammatory bowel disease. A novel mechanism of chronic intestinal inflammation. The Journal of Immunology 1995, 154, 2434-2440.

  63. Schmitz, J.; Owyang, A.; Oldham, E.; Song, Y.; Murphy, E.; McClanahan, T. K.; Zurawski, G.; Moshrefi, M.; Qin, J.; Li, X. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 2005, 23, 479-490. PMid:16286016

    View Article      PubMed/NCBI     
  64. Matsuba-Kitamura, S.; Yoshimoto, T.; Yasuda, K.; Futatsugi-Yumikura, S.; Taki, Y.; Muto, T.; Ikeda, T.; Mimura, O.; Nakanishi, K. Contribution of IL-33 to induction and augmentation of experimental allergic conjunctivitis. International immunology 2010, 22, 479-489. PMid:20501612

    View Article      PubMed/NCBI     
  65. Beltran, C. J.; Nunez, L. E.; Diaz-Jimenez, D.; Farfan, N.; Candia, E.; Heine, C.; Lopez, F.; Gonzalez, M. J.; Quera, R.; Hermoso, M. A. Characterization of the novel ST2/IL-33 system in patients with inflammatory bowel disease. Inflammatory bowel diseases 2010, 16, 1097-1107. PMid:20014018

    View Article      PubMed/NCBI     
  66. Artis, D. Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nature Reviews Immunology 2008, 8, 411-420. PMid:18469830

    View Article      PubMed/NCBI     
  67. Dionne, S.; D'agata, I.; Hiscott, J.; Vanounou, T.; Seidman, E. Colonic explant production of IL-1 and its receptor antagonist is imbalanced in inflammatory bowel disease (IBD). Clinical and experimental immunology 1998, 112, 435-442. PMid:9649212

    View Article      PubMed/NCBI     
  68. Dinarello, C. A. IL-18: A TH1-inducing, proinflammatory cytokine and new member of the IL-1 family. Journal of Allergy and Clinical Immunology 1999, 103, 11-24. 70518-X

    View Article           
  69. Schmidt, C.; Giese, T.; Goebel, R.; Schilling, M.; Marth, T.; Ruether, A.; Schreiber, S.; Zeuzem, S.; Meuer, S.; Stallmach, A. Interleukin-18 is increased only in a minority of patients with active Crohn's disease. International journal of colorectal disease 2007, 22, 1013-1020. PMid:17318554

    View Article      PubMed/NCBI     
  70. Leach, S. T.; Messina, I.; Lemberg, D. A.; Novick, D.; Rubenstein, M.; Day, A. S. Local and systemic interleukin-18 and interleukin-18-binding protein in children with inflammatory bowel disease. Inflammatory bowel diseases 2008, 14, 68-74. PMid:17879274

    View Article      PubMed/NCBI     
  71. Van Kemseke, C.; Belaiche, J.; Louis, E. Frequently relapsing Crohn's disease is characterized by a persistent elevation in interleukin-6 and soluble interleukin-2 receptor serum levels during remission. International journal of colorectal disease 2000, 15, 206-210. PMid:11008719

    View Article      PubMed/NCBI     
  72. Salmi, M.; Jalkanen, S. Human leukocyte subpopulations from inflamed gut bind to joint vasculature using distinct sets of adhesion molecules. The Journal of Immunology 2001, 166, 4650-4657. PMid:11254724

    View Article      PubMed/NCBI     
  73. Hull, J.; Ackerman, H.; Isles, K.; Usen, S.; Pinder, M.; Thomson, A.; Kwiatkowski, D. Unusual haplotypic structure of IL8, a susceptibility locus for a common respiratory virus. The American Journal of Human Genetics 2001, 69, 413-419. PMid:11431705

    View Article      PubMed/NCBI     
  74. Fuss, I. J.; Heller, F.; Boirivant, M.; Leon, F.; Yoshida, M.; Fichtner-Feigl, S.; Yang, Z.; Exley, M.; Kitani, A.; Blumberg, R. S. Nonclassical CD1d-restricted NK T cells that produce IL-13 characterize an atypical Th2 response in ulcerative colitis. The Journal of clinical investigation 2004, 113, 1490-1497. PMid:15146247

    View Article      PubMed/NCBI     
  75. Bamias, G.; Kaltsa, G.; Ladas, S. D. Cytokines in the pathogenesis of ulcerative colitis. Discovery medicine 2011, 11, 459-467. PMid:22007233

    View Article      PubMed/NCBI     
  76. Fichtner-Feigl, S.; Fuss, I. J.; Young, C. A.; Watanabe, T.; Geissler, E. K.; Schlitt, H.-J.; Kitani, A.; Strober, W. Induction of IL-13 triggers TGF-β1-dependent tissue fibrosis in chronic 2, 4, 6-trinitrobenzene sulfonic acid colitis. The Journal of Immunology 2007, 178, 5859-5870. PMid:17442970

    View Article      PubMed/NCBI     
  77. Heller, F.; Florian, P.; Bojarski, C.; Richter, J.; Christ, M.; Hillenbrand, B.; Mankertz, J.; Gitter, A. H.; Burgel, N.; Fromm, M. 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. PMid:16083712

    View Article      PubMed/NCBI     
  78. Denis, M. -A.; Louis, R.; Malaise, M.; Belaiche, J.; Louis, E. Increased response of blood eosinophils to various chemotactic agents in quiescent Crohn disease. Scandinavian journal of gastroenterology 2001, 36, 190-195. PMid:11252412

    View Article      PubMed/NCBI     
  79. Fina, D.; Caruso, R.; Pallone, F.; Monteleone, G. Interleukin-21 (IL-21) controls inflammatory pathways in the gut. Endocrine, Metabolic & Immune Disorders-Drug Targets (Formerly Current Drug Targets-Immune, Endocrine & Metabolic Disorders) 2007, 7, 288-291. PMid:18220949

    View Article      PubMed/NCBI     
  80. De Nitto, D.; Sarra, M.; Pallone, F.; Monteleone, G. Interleukin-21 triggers effector cell responses in the gut. World J Gastroenterol 2010, 16, 3638-3641. PMid:20677335

    View Article      PubMed/NCBI     
  81. Fiorentino, D. F.; Bond, M. W.; Mosmann, T. Two types of mouse T helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. Journal of Experimental Medicine 1989, 170, 2081-2095. PMid:2531194 PMCid:PMC2189521

    View Article      PubMed/NCBI     
  82. Monteleone, G.; Monteleone, I.; Fina, D.; Vavassori, P.; Blanco, G. D. V.; Caruso, R.; Tersigni, R.; Alessandroni, L.; Biancone, L.; Naccari, G. C. Interleukin-21 enhances T-helper cell type I signaling and interferon-γ production in Crohn's disease. Gastroenterology 2005, 128, 687-694. PMid:15765404

    View Article      PubMed/NCBI     

Journal Recent Articles