Open access

Research of Immunology Markers of UC

Written By

David Díaz-Jiménez, Katya Carrillo, Rodrigo Quera and Marcela A. Hermoso

Submitted: 24 February 2011 Published: 18 January 2012

DOI: 10.5772/28097

From the Edited Volume

Ulcerative Colitis from Genetics to Complications

Edited by Mustafa M. Shennak

Chapter metrics overview

2,247 Chapter Downloads

View Full Metrics

1. Introduction

Several risk factors are recognized to increase an individual’s susceptibility to develop inflammatory bowel disease (IBD) that are related to molecules that play a role in intestinal homeostasis and mucosal immune response to luminal antigens. The hallmark of IBD is a chronic, recurrent inflammation of a particular segment of the gastrointestinal tract, which is presented as a loss and damage of the intestinal epithelial barrier, exposing immune cells to luminal antigens, that might finally trigger the recruitment and activation of other immune cells and unleashing an exaggerated immune response (Abraham and Cho, 2009; Kaser et al., 2010).

Mucosa immune response in general includes different mechanisms of induction, regulation and resolution (Medzhitov, 2010). Different factors participating in these mechanisms will act as inductors, initiating an inflammatory response that will be detected by specialized sensors or sentinel cells. This process will subsequently lead to the production of inflammatory mediators that will affect different tissues; eliciting changes in their functional state that will optimize an adaptation process to the harmful condition associated to the inflammatory response (Medzhitov, 2010).

Due to the chronicity of the inflammatory response, in IBD the mechanisms that regulate and resolve the induction of the inflammatory process are defective. Although the constant induction of the inflammatory process prevents an effective regulation and resolution, making difficult to estimate the key processes involved in the development of IBD.

Since that immune system and cytokines has been linked with the pathogenia of inflammatory disorders, including IBD, the clinical and pathological significance of IL-33/ST2L system in UC, is consistently supported by in vitro and in vivo studies. The involvement of the inflammatory mediator IL-33 in the activation the other immune cells might result in a chronic inflammatory response in the colonic mucosa that is reflected at the systemic level. To avoid an exaggerated immune response, the host has developed mechanisms to counteract the resulting inflammation through the release of soluble receptors, such as sST2. These decoy molecules are potential surrogate of immunological markers for UC.

Advertisement

2. Intestinal inflammatory process in ulcerative colitis

IBD is a chronic, relapsing-remitting condition that affects the gastrointestinal tract. The aetiology of IBD has not been fully elucidated, although, it has been described as a multifactorial disease, in which genetics, environmental factors and immune system have a leading role (Abraham and Cho, 2009; Xavier and Podolsky, 2007). IBD is a complex polygenic disease in which many genes, related or not, through their contribution and interaction with environmental factors are involved in the final disease manifestation (Bouma and Strober, 2003; McGovern et al., 2010; Risch and Merikangas, 1996; Thompson and Lees, 2011).

The two major types of IBD are Crohn’s disease (CD) and ulcerative colitis (UC), both with unique characteristics that make them different at the clinical, cellular and molecular level (Thompson and Lees, 2011). CD may affect a portion of the intestine in a segmental fashion and present a transmural inflammation that extends the entire intestinal wall; whereas in UC a diffuse and continuous inflammation is confined to the mucosa of the colon (Baumgart and Carding, 2007). Histopathologic features of UC confirm that the inflammatory process is limited to the mucosa and typically consists in an increase of inflammatory cells, such as polymorphonuclear granulocytes (Nishida et al., 2002), that extends through the crypt wall (cryptitis) or inside glands with subsequent formation of cryptic abscesses. Moreover, UC is also characterized by crypt architectural distortion due to epithelial injury, shortening or branching of the glands, goblet cells depletion; and presence of lymphoid aggregates associated to oedematous and congestive lamina propria (Silverberg et al., 2005). According to the features previously described, UC pathogenesis can be explained by a deregulation of the inflammatory response of the intestinal mucosa due to epithelial barrier defects to luminal antigens in genetically susceptible individuals. Thus, those diseases characterized by the presence of a defective epithelial barrier show a deregulation of inflammatory processes (Kaser et al., 2010).

Inflammation in UC is restricted to the most superficial layer of the colonic mucosa. Mechanisms that possible may lead to the epithelial injury are reflected by architectural crypt distortion, an increase in the distance between crypts or a decrease in the crypt number; however they are not fully understood. Nonetheless, these mechanisms are likely responsible for the induction of the distinctive inflammatory process of the disease.

Recently, considerable evidence (murine models, genetic studies, in vitro assays, etc) has demonstrated that UC and CD involve an uncontrolled primary response of the innate immune system against intestinal luminal compounds, mainly mediated by macrophages, mast cells and neutrophils (Kaser et al., 2010). This uncontrolled inflammation will redound in a scarcely resolutive T and B cells-mediated adaptive immune response. However, in spite of all this information the mechanism involved in the activation of the cellular response is still unknown. This enquiry has highlighted several line of research focused in the study role of the innate immune system in IBD pathogenesis.

2.1. Innate immune receptors in ulcerative colitis pathogenesis

Innate immune response is the first line of defence that protects the host from invasive pathogens and is responsible for their rapid recognition, detection, and elimination. This response initiates and defines the adaptive immunity that is executed by B and T cells. The strategies of recognition in the innate immune system are based on identification of pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs) located on the cell surface or in intracellular compartments, such as endosomes. PRRs are also responsible of the initial recognition of damage-associated molecular pattern (DAMPs) and participate in phagocytosis, activation of pro-inflammatory intracellular pathways, opsonization, complement activation and induction of apoptosis (Medzhitov, 2001). PRRs include Toll-like receptors (TLRs), Nod like receptors (NLRs), RIG-I-like receptor (RLRs), and C-type lectin receptors (CLR) (Kawai and Akira, 2010; Kumagai and Akira, 2010). TLRs are the best characterized family of PRRs that are involved in immune response mechanisms to protect epithelial barrier integrity and invasive microorganisms elimination, contributing to the tolerance and the homeostatic balance of the intestinal mucosa (Podolsky, 2002). TLRs belong to the IL-1 receptor/Toll-like receptor (TLR-IL-1R) superfamily and contain several leucine reach repeats in their extracellular domain and intracellular toll/IL-1 receptor (TIR) domain. TLRs are expressed in both innate immune cells, including macrophages and dendritic cells (DCs) and epithelial cells. The major TLR signalling pathways is the activation of the transcription factors, such as nuclear factor-κB (NFκB) and activating protein-1 (AP-1) that direct to the production of pro-inflammatory cytokines, chemokines, and adhesion molecules (Wang et al., 2001). In pathological conditions, such as in IBD, over-activation of TLRs may induce defective signalling, allowing the induction, amplification and perpetuation of harmful immune responses and the development of a chronic inflammation reflected by an impaired function of the epithelial barrier (Cario, 2010; Kamada et al., 2008).

2.1.1. Pathogenic role of TLR2 in ulcerative colitis

In murine models of colitis, Rakoff-Nahoum et al demonstrated that in TLR2 and adaptor MyD88- deficient mice, microflora-dependent TLR2 signalling is required for the homeostasis of the intestinal epithelium and protects gut epithelia (Rakoff-Nahoum et al., 2004). Clinical evidence supports the close relationship between TLR2 over-expression and UC (Cario et al., 2000). Genetic factor involved in UC, some TLR2 polymorphisms have been described, such as SNP Arg753Gly, which affects the recruitment of signalling pathway molecules and influences over inflammation and disease severity in UC patients with no impact on its susceptibility (Pierik et al., 2006). The nucleotide deletion of TLR2 gene at position –196 to −174 might be associated to a higher risk of severe corticoid-dependence in UC (Wang et al., 2007). Intestinal mucosa isolated lamina propria mononuclear cells show a higher expression of TLR2 in IBD patients than in healthy controls (Cario et al., 2000) and submitted data). In addition, peripheral blood monocytes obtained from IBD patients have a high content of TLR2 on the cell surface, which correlate to a high production of TNF-α in response to receptor agonists (Canto et al., 2006). On the other hand, we have recently shown high levels of TLR2 in colonic mucosa of UC patients and that these finding might be related to a higher expression of TLR2 in CD33+CX3CR1+ macrophage surface in comparison to controls (submitted results). UC patients also presented elevated levels of soluble TLR2 (sTLR2), which has been shown to sequester TLR2 ligands thus, reversing TLR2 pathway activation (LeBouder et al., 2003). At the moment, evidence indicates that sTLR2 generation might be related to post-transduction mechanisms, suggesting that high levels of transmembrane TLR2 in UC intestinal macrophages might be the main cellular source of this decoy receptor. The generation of sTLR2 might be explaining a compensatory mechanism to restrain the exaggerated inflammation triggered by over-activation of TLR2 in intestinal mucosa of UC patients. This inflammatory condition might explain the participation of sTLR2 to counteract the epithelial damage generated as a consequence of activation of pro-inflammatory signalling pathways, without restabilising the mucosa homeostasis.

2.2. Epithelium in innate immune response

The intestinal epithelial cells (ECs) are continually exposed to bacteria (microbiota and enteric pathogens); however, this interaction does not usually generate a pathological inflammatory response. To maintain integrity and normal function of the intestinal tract, ECs not only constitute a physical barrier that keeps a balance between local homeostatic response and host defence against microbiota and pathogens, but also, provides important functions in the regulation of the mucosal immunity. Recent studies indicate that in response to challenges, intestinal ECs through PRRs drives the expression of critical Th2-driving cytokines, such as IL-25, TSLP and IL-33, which mediate the initiation and interplay between innate and adaptive immunity (Bulek et al., 2010; Schleimer et al., 2007).

IL-33 expression is induced in the intestinal epithelium by exogenous stimuli, including allergens, microbiota, pathogens and pro-inflammatory cytokines (IL-1 and TNF-α), coordinating the immune regulation to maintain homeostasis and drive a protective Th2 phenotype (Schmitz et al., 2005). However, elevated production of these cytokines will be associated with inflammatory Th2 condition in lesions of the mucosa producing pathological changes in the tissue (Schmitz et al., 2005). The IL-33 signalling pathways might exert distinct impact on other inflammatory cells that amplified and perpetuated the immune responses permitting the development of a chronic inflammation (Figure 1).

During inflammatory episodes, different cells, such as lymphocytes, macrophages, neutrophils and mast cells infiltrate the intestinal mucosa, promoting increased production of pro-inflammatory cytokines associated with different immune profiles. In patients diagnosed with UC, Th2 cytokines such as IL-4, IL-5 and IL-13 have been associated (Beltran et al., 2009; Bernstein et al., 2005). In relation to Th2 response that characterized UC, IL-33 is also able to polarize naive T cells into Th2 cells and induce production of IL-4, IL-5 and IL-13 that resulted in pathological changes in the intestinal architecture that includes eosinophilic infiltrates, increased mucus production and epithelial cells hyperplasia and hypertrophy (Figure 1).

2.3. Pathogenic role of the IL-33/ST2 system in ulcerative colitis

Recently, we and others have reported that IL-33 expression is increased in colonic mucosa of UC patients, particularly in those with moderate to severe activity of the disease (Beltran et al., 2010; Kobori et al., 2010; Pastorelli et al., 2010; Seidelin et al., 2010). It has been proposed that in UC, IL-33 may be released by injured epithelial cells to induce pro-inflammatory cytokines production (i.e. IL-1, IL-6, TNF-α, IL-5 and IL-13) through activation of ST2L in mast cells, macrophages, eosinophils and neutrophils (Luthi et al., 2009). Moreover, IL-33 expression is restricted to the epithelial layer of the intestine (Beltran et al., 2010; Pastorelli et al., 2010). In addition, activation of ST2L in dendritic cells may contribute to the polarization to IL-5 and IL-13-producing Th2 cells (Rank et al., 2009) and in basophiles the induction of IL-13-dependent fibrosis (Pecaric-Petkovic et al., 2009). Those cytokines induced by IL-33, mostly IL-13, may have detrimental effects on epithelial barrier function (Heller et al., 2005). Together, the effects induced by IL-33 might amplify the local inflammatory response, and therefore, contributing to perpetuation of pathogenic inflammatory process that is characteristic of the disease (Palmer and Gabay, 2011).

Figure 1.

Role of IL-33/ST2 receptor system in UC. In ulcerative colitis (UC) epithelia is more exposed to pathogens, as mucus layer is deficient to prevent their access to the barrier, with persistent inflammation that also promote epithelial disruption. The tissue injury can be a consequence of infections or the access of the microbiota, which are linked to the development of IBD. The over-activation of TLR2 present in the macrophage cell surface (1) will produce inflammatory cytokines, as well as, tumor necrosis factor (TNF-α) and IL-1 (2), that promote the epithelial injury (3). Tissue damage leads to the release of interleukin-33 (IL-33) from epithelial cells (4), which acts as an early inducer of inflammation. IL-33 induce the expression of pro-inflammatory cytokines in cells that express ST2 receptor (mast cells (5), neutophils activated, eosinophils and basophils (6)). Moreover, IL-33 may drive antigen sensitization and polarization to T helper 2 (Th2) -mediated inflammation (7) during the development of UC owing to its ability to activate dendritic cells (DCs) and to recruit, polarize and activate Th2 cells that also express ST2 receptor. IL-33 can induce eosinophilia by mast cells through the induction of IL-13 secretion. IL-13 exerts detrimental effects on epithelial barrier function, favoring the effects of IL-33 (8). Mast cell-mediated inflammation may drive a robust proliferation of fibroblast toward fibrosis formation, however high levels of sST2 might counteract this cellular effect of IL-33. (M: M cells; G: Goblet cells and E: Epithelial cells).

2.3.1. Components of the IL-33/ST2 system

Clinical and experimental data have shown that activation of IL-33/ST2 pathway is primarily occurring in diseases that affect epithelial barriers, such as asthma, arthritis and UC (Palmer and Gabay, 2011). ST2 protein (IL1RL1) is encoded by a single gene located on chromosome 2q12 (Tominaga et al., 1991), that is part of the TLR-IL-1R superfamily. Three types of st2 gene products are generated by alternative splicing: ST2L, which is the complete form of protein, the receptor itself, that has a TIR intracellular domain (similar to Toll/Interleukin-1 receptors domain), a transmembrane and an extracellular domain formed by three immunoglobulin-like domains that binds IL-33; a soluble form of ST2 (sST2) that lacks the transmembrane and intracellular domains, but can also recognize IL-33; and a form bound to the plasma membrane (vST2) that,similar to sST2, lacks the intracellular domain. The production of these isoforms is under the control of two distinct promoters (proximal and distal) which have a differential activity depending on the cell type (Gachter et al., 1996; Iwahana et al., 1999). The differential function of the promoters allows a 3’ differential processing of st2 mRNA to generate the STL2 and sST2 isoforms (Bergers et al., 1994). To date, the cellular and molecular context that might activate a defined promoter it is still uncertain, and the signalling pathways required to produce one protein over the other, is even less clear. Many studies recognized pro-inflammatory properties to ST2L receptor activation and anti-inflammatory effects to the soluble form sST2. However, since there is no an experimental model available where characteristics of one of the protein isoforms are conserved, the attribution of these functional effects to ST2 remains under speculation.

Since IL-33 cytokine was described as the ligand of ST2L receptor, its effect has been associated to a Th2 immune profile (Schmitz et al., 2005). The signalling pathway activated upon ligand binding to ST2L is common to all members of the TLR-IL-1R superfamily, involving recruitment of MyD88, IRAKs and TRAF6 adaptor molecule which leads to phosphorylation of Mitogen-Activated Protein Kinases (MAPK) such as ERK1, ERK2 and p38 pathways and the consequent activation of NFκB and AP-1 to induce pro-inflammatory gene expression (Palmer et al., 2008; Schmitz et al., 2005).

2.3.2. Cellular sources of IL-33/ST2 system

ST2L receptor expression has been mainly associated to immune cells, such as mast cells, macrophages, dendritic cells, NK cells, eosinophils, basophils, Th2 lymphocytes and activated neutrophils (Allakhverdi et al., 2007; Ho et al., 2007; Komai-Koma et al., 2007; Rank et al., 2009; Suzukawa et al., 2008a) (Figure 1). Polymorphonuclear leukocytes have been also demonstrated to produce the soluble form sST2. However, sST2 has been primarily associated to fibroblasts, epithelial and endothelial cells (Hayakawa et al., 2007). The ligand of ST2, IL-33, was initially described as a nuclear protein, and is constitutively expressed by cells in contact with external surfaces, such as epithelial and endothelial cells (Baekkevold et al., 2003), and is potentially released in response to tissue damage, to rapidly activate the innate immune system (Palmer and Gabay, 2011). Induced expression of IL-33 has been reported in different cell types, in resident as well as in infiltrating inflammatory cells (Oboki et al., 2011). IL-33 is the most recently described member of the IL-1 cytokine family (Schmitz et al., 2005), has been attributed to have similar functions to IL-1α exerting dual effects as a nuclear factor as well as a pro-inflammatory cytokine (Carriere et al., 2007; Cayrol and Girard, 2009; Roussel et al., 2008; Talabot-Ayer et al., 2009). IL-33 gene does not encode a secretion signal peptide, such as other cytokines, so that its secretion is not produced by conventional mechanisms (Lamkanfi and Dixit, 2009; Zhao and Hu, 2010). It has been suggested that IL-33 is released during cell necrosis, similar to what was previously described for the alarmins, as an inflammatory response that will produce early activation of innate immune system cells (Haraldsen et al., 2009; Lamkanfi and Dixit, 2009).

One of the major cellular sources of the receptor ST2L in the intestinal mucosa is mast cells (Figure 1) which have been demonstrated to have important roles in the distinctive inflammatory process of UC (Allakhverdi et al., 2007; Iikura et al., 2007; Lee et al., 2002; Moritz et al., 1998). Mast cells are considered true sensors of cell injury in tissue exposed to the exterior (Enoksson et al., 2011). These cells might be responsible of orchestrating and enhancing the innate immune response induced by IL-33 in the intestinal mucosa in UC patients, since mast granulatory products have been detected in inflamed areas of the intestine (Bischoff et al., 1996).

2.3.3. Regulation of IL-33/ST2 inflammatory pathway: Role of sST2

One of the hallmarks of UC is chronicity, and periods of active inflammation (flare-ups) and remission. This special feature of UC opens different questions about inflammation regulation. The clinical practice can demonstrate classic endoscopic and histologic patterns of active inflammation in patients, where, mild mucosa inflammation is generally characterized by vascular congestion, erythema, oedema and granularity (Pineton de Chambrun et al., 2010). When inflammation becomes severe in UC, friability, spontaneous bleeding and macroscopic ulcers of different sizes are mainly observed (D'Haens et al., 2007; Fefferman and Farrell, 2005). Therefore, a patient in remission, after a period with lesions, the mucosa might have a reduced inflammatory process, reflected by mucosal healing (MH) and a decrease in cellular infiltrates (Lichtenstein and Rutgeerts, 2010; Rutgeerts et al., 2007). The MH is characterized by restoration of a normal vascular pattern, absence of friability, bleeding, erosions and ulcers in all intestinal segments of the mucosa visualized in the intestine from UC patients (Lichtenstein and Rutgeerts, 2010; Rutgeerts et al., 2007). However, at a cellular level, there is no consensus on the processes leading to MH. Only restoration, proliferation and differentiation of epithelial cells adjacent to the injured area will allow intestinal wound healing (Iizuka and Konno, 2011). Many reports support the evidence that activation of IL-10 signalling pathway may have a leading role in regulation of the inflammatory process (Li and He, 2004; Shih and Targan, 2008). IL-10-deficient mice (IL-10-/-) spontaneously reproduce a colitis phenotype similar to human colitis (Bristol et al., 2000; Kuhn et al., 1993; Rennick et al., 1997). In this mice model, the intestine damage is characterized by the presence of large and thick crypts, and low number of goblet cells, allowing the development of spontaneous colitis (Thompson and Lees, 2011). However, since IL-10 participation might be primarily associated to cellular processes that regulate and resolve the inflammatory response, in chronic inflammation condition, such as UC, its contribution might be relevant to achieve a homeostatic balance (Mosser and Zhang, 2008). Clinical and experimental data have shown sST2 counteractive effect over the activation of IL-33/ST2L pathway and resolution of inflammation (Takezako et al., 2006). Soluble ST2 inhibits IL-33 activity in in vitro assays of mast cells stimulated with the cytokine thus blocking the signalling pathway and the release of pro-inflammatory cytokines (Hayakawa et al., 2007; Ho et al., 2007; Palmer et al., 2008; Sanada et al., 2007; Weinberg, 2009). In murine asthma models, pre-treatment with recombinant ST2 reduced IL-13 content in bronchoalveolar lavage fluid induced by intranasal administration of IL-33 (Hayakawa et al., 2007). Similarly, intraperitoneal administration of sST2 reduced the severity, extent of inflammation and number of affected joints, as well as plasma concentration of pro-inflammatory cytokines in collagen-induced arthritis in mice (Leung et al., 2004). Also, in methylated-BSA induced-rheumatoid arthritis mice model, therapeutic effect of sST2 was also manifested in decrease of neutrophils recruitment to affected joints (Verri et al., 2010). In UC, recent reports have demonstrated that sST2 levels correlate with the severity of the disease (Beltran et al., 2010; Diaz-Jimenez et al., 2011); hence a reduction in protein levels could be used as a biomarker to determine clinical remission. However, although information points to an anti-inflammatory role of sST2, evidence also shows a direct relationship to the disease. Given that UC patients cursing with severe activity have evidently increased plasma levels of sST2, this condition was shown to directly correlate with increased intestinal levels (Diaz-Jimenez et al., 2011). A possible and appealing explanation to this issue is that intestinal increase of sST2 levels reflected in plasma evident a mechanism to prevent an exaggerated immune response; however it might be insufficient to resolve the pathological inflammation distinctive to severe UC (Akhabir and Sandford, 2010).

2.4. IBD genetics

As previously mentioned, IBDs have an important genetic background. Relationship between certain genes and susceptibility to a particular disease has been possible due to molecular characterization made in the past decades (Hardy and Singleton, 2009; Manolio, 2010). Genetic factors relevant in IBD have been demonstrated, through the identification of risk polymorphisms, their loci and genes involved (Barrett et al., 2008; Vermeire et al., 2010). Nevertheless, genetic contributing to disease risk is more profoundly documented in CD than in UC. Many of these risk factors might be related to molecules that participate in the immune response directed to preserve intestinal homeostasis (Carter et al., 2001; Henckaerts et al., 2007). For example, mutations in NOD2/CARD15 gene have acquired great importance as a susceptibility gene for CD. Association between NOD2/CARD15 gene variants and susceptibility and severity of the disease suggest that these mutant alleles may have a prognostic value of an unfavourable outcome and high requirement of surgery (Alvarez-Lobos et al., 2005; Annese et al., 2005; Seiderer et al., 2006). In UC, genes encoding glycoprotein e-cadherin and laminins, such as ECM1, CDH1 and Lamb1, involved in epithelial barrier function and in regulation of inflammatory process, have emerged as significant determinants of susceptibility (Thompson and Lees, 2011). High levels of ST2L protein expression have been described in polygenic and multifactorial diseases recognized to be caused by inflammatory response and with a compromise of the epithelial barrier, such as asthma, atopic dermatitis and systemic lupus erythematosus (Ali et al., 2009; Kuroiwa et al., 2001; Mok et al., 2010; Oshikawa et al., 2001a; Shimizu et al., 2005). These pathologies are also characterized by a high number of local inflammatory infiltrates (mast cells, basophils, neutrophils and eosinophils) and high plasma concentration of sST2, as was also described for UC. Since the physiopathologic role of this protein has been already described, genetic studies have been searching for single nucleotide polymorphism (SNP) in the st2 gene locus. To date, two case-control studies, one in atopic dermatitis (Shimizu et al., 2005) and another in asthma (Ali et al., 2009), have analyzed the presence of SNPs located in the distal promoter of the st2 gene. Among these, only the A allele of SNP -26999G/A (rs6543116), could be related to an increase in gene transcription, higher serum levels of sST2, and a higher risk to develop the disease (Shimizu et al., 2005). Whereas in asthma the presence of the AA genotype was only found in a small fraction of the studied population, it seems to be more often in severe corticoid-dependent asthma patients associated to a worsen course of the disease (Ali et al., 2009).

At the moment, there are no studies to demonstrate a possible association between SNPs present in the st2 gene and IBD susceptibility. However, our preliminary results seem to not support this association. Recently, two publications related to Genome-Wide Association (GWA) have made possible the detection of risk genes and loci associated with complex diseases, such as IBD (Barrett et al., 2008; Hampe et al., 2007; Rioux et al., 2007). In these studies, the loci that contains the st2 gene has not been directly implicated in UC susceptibility. However, the locus containing genes for IL1R, IL18R1 and IL18RAP, previously related to UC and with a high linkage disequilibrium (LD) represents an interesting candidate that might be involved in the development and course of the disease (Akhabir and Sandford, 2010). In line with this, a new SNP located in the non-codifying region of st2 gene (rs1420101), has been associated with eosinophilia in asthma and other inflammatory diseases, such as chronic obstructive pulmonary disease (COPD) and myocardial infarction. Gudbjartsoon et al. (Gudbjartsson et al., 2009) showed that there is no correlation between this genetic variant and the increased number in eosinophils, or with previously described genetic variants associated to CD (rs917997) (Zhernakova et al., 2008) and celiac disease (rs13015714) (Hunt et al., 2008). Since the IL-33/ST2 signalling pathway has a role in maturation, survival and activation of eosinophils, also in recruitment and regulation of Th2 cell function, the available information is consistent with a greater contribution of this system in eosinophil-mediated inflammation (Stolarski et al., 2010; Suzukawa et al., 2008b). The role of eosinophils in the aetiology and pathogenesis of UC is not completely clear. However, evidence shows a 10-fold higher content of eosinophils in patients with active UC compared to healthy individuals (Kristjansson et al., 2004). In these patients, tissue eosinophilia induced by IL-33 might have a detrimental effect on intestinal structural integrity. As previously mentioned, polymorphonuclear cells constitute an important source of ST2, making an interesting issue the analysis of the presence of these new SNPs in samples of UC patients.

2.5. Soluble ST2 as a biomarker of activity in ulcerative colitis

Many symptoms of UC and CD are similar, but there are some subtle differences. Clinical presentation are rather unspecific, thus further studies are necessary to achieve a differential diagnosis of the diseases. Colonoscopy through biopsy analysis remains the foundation for IBD diagnosis. However, several studies have shown a considerable variability in endoscopic and histological changes of intestinal mucosa (higher inter- and intra-observer variability in determination of the disease activity; low precision and reproducibility of the observations and scarce correlation between endoscopic appearance and clinical indices with treatment response) (D'Haens et al., 2007; Osada et al., 2010; Regueiro et al., 2011). This discrepancy has led physicians and immunologists to search for clinically relevant molecules as biomarkers, non-invasive indicators that objectively assess the damage that is taking place at the intestinal mucosa ( Gisbert et al., 2007 ; Tibble and Bjarnason, 2001 ; Vermeire et al., 2006). The ideal marker not only might allow for differential diagnosis but also establish prognosis, activity and severity of the disease, risks or complications, relapses, recurrence and clinical remission, besides evaluation of treatment response ( Gisbert and Gomollon, 2007 ; Gisbert et al., 2007 ; Tibble and Bjarnason, 2001 ; Vermeire et al., 2006).

Since intestinal inflammation in IBD patients is associated with acute-phase response and recruitment of immune cells to the site of inflammation, a high production of protein in the mucosa might be detected in serum and/or stool. Among the most commonly used molecules associated with IBD in clinical practice are serological biomarkers, including anti-microbial antibodies (ASCA, ANCA, anti-OmpC, anti-Cbir, anti-I2 and anti-glycans), other plasma proteins (albumin, C-reactive protein (CRP), cytokines, adhesion molecules, among others) and stool proteins (calprotectin, lactoferrin, and neutrophil-elastase) (Li et al., 2008). Most of former serological biomarkers, (mainly ASCA and ANCA) individually have the disadvantage of moderate sensitivity and specificity to predict intestinal inflammation and do not correlate with clinical and disease activity (Papp et al., 2007). C-reactive protein is an objective inflammatory marker and correlates with severe disease activity (Karoui et al., 2007; Rodgers and Cummins, 2007; Solem et al., 2005), however, it is a systemic inflammatory marker elevated in several other inflammatory diseases and with low specificity by diagnosing patients with mild disease activity (Karoui et al., 2007; Rodgers and Cummins, 2007; Solem et al., 2005; Vermeire et al., 2004).

Faecal calprotectin is another biomarker; it is a calcium-binding protein mainly present in neutrophil granules and has the theoretical benefit of having greater specificity for the diagnosis of gastrointestinal diseases. Calprotectin concentration is directly proportional to the migration of neutrophil to the gastrointestinal tract and is not altered by extra-intestinal diseases (Gisbert and McNicholl, 2009; Langhorst et al., 2008; Schoepfer et al., 2010; Sipponen et al., 2008). In addition, calprotectin levels most directly correlate to histological features rather than with endoscopic indexes, suggesting that this marker has a better sensitivity for the evaluation of disease activity than endoscopic procedures (Burns et al., 2003; Poullis et al., 2002). Calprotectin is generally detected in a small sample using simple and low cost methods and is relatively stable in the stool. Although calprotectin has many advantages as a biomarker and is highly sensitive to detect intestinal inflammation (Langhorst et al., 2008), it is not specific for IBD since other gastrointestinal diseases, such as colorectal cancer (Roseth et al., 1993; Tibble et al., 2001 ) or gastrointestinal infections, were shown to present high levels of this protein (Summerton et al., 2002; Tibble et al., 2002; Pezzilli et al., 2008).

Several clinical studies have reported an increase in plasma levels of sST2 in patients with inflammatory processes, including asthma (Oshikawa et al., 2001b), autoimmune diseases (rheumatoid arthritis, erythematosus systemic lupus and progressive systemic scleroderma) (Kuroiwa et al., 2001), cardiovascular diseases (Weinberg et al., 2003), trauma and sepsis (Brunner et al., 2004). Currently, plasma level of sST2 might work as a biomarker for diagnosis and /or prognosis of several cardiac function conditions directly related to myocardial injury (Diez, 2008; Rehman et al., 2008; Weinberg, 2009; Weinberg et al., 2003). In the context of IBD, we have reported high levels of sST2 that has been proposed as a reliable biomarker of UC activity (Diaz-Jimenez et al., 2011). In UC, serum levels of sST2 grouped by active and inactive, according to endoscopic disease activity index (inactive defined as a Mayo endoscopic score ≥ 1 point), were 33.19 pg/mL and 235.80 pg/mL, respectively. Differences observed in serum concentration were statistically significant (p<0.0001) to discriminate between active and inactive condition. Also, the correlation ROC curve shown a cut-off for sST2 level at 74.87 pg/mL that allow the discrimination between the grade of disease activity (AUC = 0.92) with a sensitivity and specificity of 83.33%. Additionally, serum sST2 levels directly correlated with the degree of endoscopic activity (r = 0.76) according to Mayo endoscopic sub-score classification (0 = normal or inactive state, 1 = mild, 2 = moderate, and 3 = severe disease). According to histopathologic findings (Gomes et al., 1986), levels of sST2 also correlated to the specific index (r = 0.67), suggesting that, similar to calprotectin, serum sST2 levels might have a better sensitivity than endoscopic procedure to estimate the activity of UC patients. Furthermore, we demonstrated that serum sST2 levels behave similarly to other serum inflammation marker, such as TNF-α, in relation to endoscopic and histopathologic activity indexes. However, sST2 is potentially a better biomarker as is more stable than the previously reported cytokine (Dieplinger et al., 2010).

Finally, we also showed that total levels of ST2 in colonic mucosa of UC patients positively correlated with endoscopic (r = 0.62) and histopathologic (r = 0.60) UC activity indexes similar to what was described for serum levels. The clear association between intestinal and serum ST2 levels illustrate a valid activity biomarker, unbiased and distinctive of inflammatory process taking place in the intestinal mucosa of UC patients. Thus, ST2 detection might help physicians in the decision making on whether to periodically send patients to colonoscopy, an invasive and expensive test, or use alternative assessment techniques.

Advertisement

3. Conclusion

Current methodology, especially those related to genomics and transcriptomes, will soon allow the improvement in differential diagnosis in patients who show great clinical heterogeneity, with less invasive procedures than colonoscopy and biopsies. Measurement of serum ST2 levels is a reliable, quick and low cost technique for differential diagnosis in IBD and other gastrointestinal diseases, and might allow the assessment of disease activity. Furthermore, in certain subgroups of patients with well-known diagnosis, sST2 has been assigned to have a predictive value to define the course of the disease, ever since genetic analysis of st2 gene might relate to higher surgery rates, as has been previously shown for NOD2/CARD15 in CD. Here lies the biggest advantage of new biomarkers based on intestinal specific inflammation mechanisms. The future development of molecular classification of IBD, according to molecular biomarkers, may allow a better accuracy in diagnosis, clinical course and response to a determined therapy to induce and sustain remission in IBD.

References

  1. 1. Abraham C. Cho J. H. 2009 Inflammatory bowel disease. N Engl J Med 361 2066 78 .
  2. 2. Akhabir L. Sandford A. 2010 Genetics of interleukin 1 receptor-like 1 in immune and inflammatory diseases. Curr Genomics 11 591 606 .
  3. 3. Ali M. Zhang G. Thomas W. R. McLean C. J. Bizzintino J. A. Laing I. A. Martin A. C. Goldblatt J. Le Souef P. N. Hayden C. M. 2009 Investigations into the role of ST2 in acute asthma in children. Tissue Antigens 73 206 12 .
  4. 4. Alvarez-Lobos M. Arostegui J. I. Sans M. Tassies D. Plaza S. Delgado S. Lacy A. M. Pique J. M. Yague J. Panes J. 2005 Crohn’s disease patients carrying Nod2/CARD15 gene variants have an increased and early need for first surgery due to stricturing disease and higher rate of surgical recurrence. Ann Surg 242 693 700.
  5. 5. Annese V. Lombardi G. Perri F. D’Inca R. Ardizzone S. Riegler G. Giaccari S. Vecchi M. Castiglione F. Gionchetti P. Cocchiara E. Vigneri S. Latiano A. Palmieri O. Andriulli A. 2005 Variants of CARD15 are associated with an aggressive clinical course of Crohn’s disease--an IG-IBD study. Am J Gastroenterol 100 84 92 .
  6. 6. Baekkevold E. S. Roussigne M. Yamanaka T. Johansen F. E. Jahnsen F. L. Amalric F. Brandtzaeg P. Erard M. Haraldsen G. Girard J. P. 2003 Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules. Am J Pathol 163 69 79 .
  7. 7. Barrett J. C. Hansoul S. Nicolae D. L. Cho J. H. Duerr R. H. Rioux J. D. Brant S. R. Silverberg M. S. Taylor K. D. Barmada M. M. Bitton A. Dassopoulos T. Datta L. W. Green T. Griffiths A. M. Kistner E. O. Murtha M. T. Regueiro M. D. Rotter J. I. Schumm L. P. Steinhart A. H. Targan S. R. Xavier R. J. Libioulle C. Sandor C. Lathrop M. Belaiche J. Dewit O. Gut I. Heath S. Laukens D. Mni M. Rutgeerts P. Van Gossum A. Zelenika D. Franchimont D. Hugot J. P. de Vos M. Vermeire S. Louis E. Cardon L. R. Anderson C. A. Drummond H. Nimmo E. Ahmad T. Prescott N. J. Onnie C. M. Fisher S. A. Marchini J. Ghori J. Bumpstead S. Gwilliam R. Tremelling M. Deloukas P. Mansfield J. Jewell D. Satsangi J. Mathew C. G. Parkes M. Georges M. Daly M. J. 2008 Genome-wide association defines more than 30 distinct susceptibility loci for Crohn’s disease. Nat Genet 40 955 62 .
  8. 8. Baumgart D. C. Carding S. R. 2007 Inflammatory bowel disease: cause and immunobiology. Lancet 369 1627 40 .
  9. 9. Beltran C. J. Candia E. Erranz B. Figueroa C. Gonzalez M. J. Quera R. Hermoso M. A. 2009 Peripheral cytokine profile in Chilean patients with Crohn’s disease and ulcerative colitis. Eur Cytokine Netw 20 33 8 .
  10. 10. 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. 2010 Characterization of the novel ST2/IL-33 system in patients with inflammatory bowel disease. Inflamm Bowel Dis 16 1097 107 .
  11. 11. Bergers G. Reikerstorfer A. Braselmann S. Graninger P. Busslinger M. 1994 Alternative promoter usage of the Fos-responsive gene Fit-1 generates mRNA isoforms coding for either secreted or membrane-bound proteins related to the IL-1 receptor. EMBO J 13 1176 88 .
  12. 12. Bernstein C. N. Wajda A. Blanchard J. F. 2005 The clustering of other chronic inflammatory diseases in inflammatory bowel disease: a population-based study. Gastroenterology 129 827 36 .
  13. 13. Bischoff S. C. Wedemeyer J. Herrmann A. Meier P. N. Trautwein C. Cetin Y. Maschek H. Stolte M. Gebel M. Manns M. P. 1996 Quantitative assessment of intestinal eosinophils and mast cells in inflammatory bowel disease. Histopathology 28 1 13 .
  14. 14. Bouma G. Strober W. 2003 The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol 3 521 33 .
  15. 15. Bristol I. J. Farmer M. A. Cong Y. Zheng X. X. Strom T. B. Elson C. O. Sundberg J. P. Leiter E. H. 2000 Heritable susceptibility for colitis in mice induced by IL-10 deficiency. Inflamm Bowel Dis 6 290 302 .
  16. 16. Brunner M. Krenn C. Roth G. Moser B. Dworschak M. Jensen-Jarolim E. Spittler A. Sautner T. Bonaros N. Wolner E. Boltz-Nitulescu G. Ankersmit H. J. 2004 Increased levels of soluble ST2 protein and IgG1 production in patients with sepsis and trauma. Intensive Care Med 30 1468 73 .
  17. 17. Bulek K. Swaidani S. Aronica M. Li X. 2010 Epithelium: the interplay between innate and Th2 immunity. Immunol Cell Biol 88 257 68 .
  18. 18. Burns K. Janssens S. Brissoni B. Olivos N. Beyaert R. Tschopp J. 2003 Inhibition of interleukin 1 receptor/Toll-like receptor signaling through the alternatively spliced, short form of MyD88 is due to its failure to recruit IRAK-4. J Exp Med 197 263 8 .
  19. 19. Canto E. Ricart E. Monfort D. Gonzalez-Juan D. Balanzo J. Rodriguez-Sanchez J. L. Vidal S. 2006 TNF alpha production to TLR2 ligands in active IBD patients. Clin Immunol 119 156 65 .
  20. 20. Cario E. 2010 Toll-like receptors in inflammatory bowel diseases: a decade later. Inflamm Bowel Dis 16 1583 97 .
  21. 21. Cario E. Rosenberg I. M. Brandwein S. L. Beck P. L. Reinecker H. C. Podolsky D. K. 2000 Lipopolysaccharide activates distinct signaling pathways in intestinal epithelial cell lines expressing Toll-like receptors. J Immunol 164 966 72 .
  22. 22. Carriere V. Roussel L. Ortega N. Lacorre D. A. Americh L. Aguilar L. Bouche G. Girard J. P. 2007 IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci U S A 104 282 7 .
  23. 23. Carter M. J. di Giovine F. S. Jones S. Mee J. Camp N. J. Lobo A. J. Duff G. W. 2001 Association of the interleukin 1 receptor antagonist gene with ulcerative colitis in Northern European Caucasians. Gut 48 461 7 .
  24. 24. Cayrol C. Girard J. P. 2009 The IL-1-like cytokine IL-33 is inactivated after maturation by caspase-1. Proc Natl Acad Sci U S A 106 9021 6 .
  25. 25. D’Haens G. Sandborn W. J. Feagan B. G. Geboes K. Hanauer S. B. Irvine E. J. Lemann M. Marteau P. Rutgeerts P. Scholmerich J. Sutherland L. R. 2007 A review of activity indices and efficacy end points for clinical trials of medical therapy in adults with ulcerative colitis. Gastroenterology 132 763 86 .
  26. 26. Diaz-Jimenez D. Nunez L. E. Beltran C. J. Candia E. Suazo C. Alvarez-Lobos M. Gonzalez M. J. Hermoso M. A. Quera R. 2011 Soluble ST2: A new and promising activity marker in ulcerative colitis. World J Gastroenterol 17 2181 90 .
  27. 27. Dieplinger B. Gegenhuber A. Kaar G. Poelz W. Haltmayer M. Mueller T. 2010 Prognostic value of established and novel biomarkers in patients with shortness of breath attending an emergency department. Clin Biochem 43 714 9 .
  28. 28. Diez J. 2008 Serum soluble ST2 as a biochemical marker of acute heart failure: future areas of research. J Am Coll Cardiol 52 1466 7 .
  29. 29. Enoksson M. Lyberg K. Moller-Westerberg C. Fallon P. G. Nilsson G. Lunderius-Andersson C. 2011 Mast cells as sensors of cell injury through IL-33 recognition. J Immunol 186 2523 8 .
  30. 30. Fefferman D. S. Farrell R. J. 2005 Endoscopy in inflammatory bowel disease: indications, surveillance, and use in clinical practice. Clin Gastroenterol Hepatol 3 11 24 .
  31. 31. Gachter T. Werenskiold A. K. Klemenz R. 1996 Transcription of the interleukin-1 receptor-related T1 gene is initiated at different promoters in mast cells and fibroblasts. J Biol Chem 271 124 9 .
  32. 32. Gisbert J. P. Gomollon F. 2007 [Common errors in the management of the seriously ill patient with inflammatory bowel disease]. Gastroenterol Hepatol 30 294 314 .
  33. 33. Gisbert J. P. McNicholl A. G. 2009 Questions and answers on the role of faecal calprotectin as a biological marker in inflammatory bowel disease. Dig Liver Dis 41 56 66 .
  34. 34. Gisbert J. P. Gonzalez-Lama Y. Mate J. 2007 [Role of biological markers in inflammatory bowel disease]. Gastroenterol Hepatol 30 117 29 .
  35. 35. Gomes P. du Boulay C. Smith C. L. Holdstock G. 1986 Relationship between disease activity indices and colonoscopic findings in patients with colonic inflammatory bowel disease. Gut 27 92 5 .
  36. 36. Gudbjartsson D. F. Bjornsdottir U. S. Halapi E. Helgadottir A. Sulem P. Jonsdottir G. M. Thorleifsson G. Helgadottir H. Steinthorsdottir V. Stefansson H. Williams C. Hui J. Beilby J. Warrington N. M. James A. Palmer L. J. Koppelman G. H. Heinzmann A. Krueger M. Boezen H. M. Wheatley A. Altmuller J. Shin H. D. Uh S. T. Cheong H. S. Jonsdottir B. Gislason D. Park C. S. Rasmussen L. M. Porsbjerg C. Hansen J. W. Backer V. Werge T. Janson C. Jonsson U. B. Ng M. C. Chan J. So W. Y. Ma R. Shah S. H. Granger C. B. Quyyumi A. A. Levey A. I. Vaccarino V. Reilly M. P. Rader D. J. Williams M. J. van Rij A. M. Jones G. T. Trabetti E. Malerba G. Pignatti P. F. Boner A. Pescollderungg L. Girelli D. Olivieri O. Martinelli N. Ludviksson B. R. Ludviksdottir D. Eyjolfsson G. I. Arnar D. Thorgeirsson G. Deichmann K. Thompson P. J. Wjst M. Hall I. P. Postma D. S. Gislason T. Gulcher J. Kong A. Jonsdottir I. Thorsteinsdottir U. Stefansson K. 2009 Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction. Nat Genet 41 342 7 .
  37. 37. Hampe J. Franke A. Rosenstiel P. Till A. Teuber M. Huse K. Albrecht M. Mayr G. De La Vega F. M. Briggs J. Gunther S. Prescott N. J. Onnie C. M. Hasler R. Sipos B. Folsch U. R. Lengauer T. Platzer M. Mathew C. G. Krawczak M. Schreiber S. 2007 A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet 39 207 11.
  38. 38. Haraldsen G. Balogh J. Pollheimer J. Sponheim J. Kuchler A. M. 2009 Interleukin-33- cytokine of dual function or novel alarmin? Trends Immunol 30 227 33 .
  39. 39. Hardy J. Singleton A. 2009 Genomewide association studies and human disease. N Engl J Med 360 1759 68 .
  40. 40. Hayakawa H. Hayakawa M. Kume A. Tominaga S. 2007 Soluble ST2 blocks interleukin-33 signaling in allergic airway inflammation. J Biol Chem 282 26369 80 .
  41. 41. Heller F. Florian P. Bojarski C. Richter J. Christ M. Hillenbrand B. Mankertz J. Gitter A. H. Burgel N. Fromm M. Zeitz M. Fuss I. Strober W. Schulzke J. D. 2005 Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology 129 550 64 .
  42. 42. Henckaerts L. Pierik M. Joossens M. Ferrante M. Rutgeerts P. Vermeire S. 2007 Mutations in pattern recognition receptor genes modulate seroreactivity to microbial antigens in patients with inflammatory bowel disease. Gut 56 1536 42 .
  43. 43. Ho L. H. Ohno T. Oboki K. Kajiwara N. Suto H. Iikura M. Okayama Y. Akira S. Saito H. Galli S. J. Nakae S. 2007 IL-33 induces IL-13 production by mouse mast cells independently of IgE-FcepsilonRI signals. J Leukoc Biol 82 1481 90 .
  44. 44. Hunt K. A. Zhernakova A. Turner G. Heap G. A. Franke L. Bruinenberg M. Romanos J. Dinesen L. C. Ryan A. W. Panesar D. Gwilliam R. Takeuchi F. Mc Laren W. M. Holmes G. K. Howdle P. D. Walters J. R. Sanders D. S. Playford R. J. Trynka G. Mulder C. J. Mearin M. L. Verbeek W. H. Trimble V. Stevens F. M. O’Morain C. Kennedy N. P. Kelleher D. Pennington D. J. Strachan D. P. McArdle W. L. Mein C. A. Wapenaar M. C. Deloukas P. Mc Ginnis R. Mc Manus R. Wijmenga C. van Heel D. A. 2008 Newly identified genetic risk variants for celiac disease related to the immune response. Nat Genet 40 395 402 .
  45. 45. Iikura M. Suto H. Kajiwara N. Oboki K. Ohno T. Okayama Y. Saito H. Galli S. J. Nakae S. 2007 IL-33 can promote survival, adhesion and cytokine production in human mast cells. Lab Invest 87 971 8 .
  46. 46. Iizuka M. Konno S. 2011 Wound healing of intestinal epithelial cells. World J Gastroenterol 17 2161 71 .
  47. 47. Iwahana H. Yanagisawa K. Ito-Kosaka A. Kuroiwa K. Tago K. Komatsu N. Katashima R. Itakura M. Tominaga S. 1999 Different promoter usage and multiple transcription initiation sites of the interleukin-1 receptor-related human ST2 gene in UT-7 and TM12 cells. Eur J Biochem 264 397 406 .
  48. 48. Kamada N. Hisamatsu T. Okamoto S. Chinen H. Kobayashi T. Sato T. Sakuraba A. Kitazume M. T. Sugita A. Koganei K. Akagawa K. S. Hibi T. 2008 Unique CD14 intestinal macrophages contribute to the pathogenesis of Crohn disease via IL-23/IFN-gamma axis. J Clin Invest 118 2269 80 .
  49. 49. Karoui S. Ouerdiane S. Serghini M. Jomni T. Kallel L. Fekih M. Boubaker J. Filali A. 2007 Correlation between levels of C-reactive protein and clinical activity in Crohn’s disease. Dig Liver Dis 39 1006 10 .
  50. 50. Kaser A. Zeissig S. Blumberg R. S. 2010 Inflammatory bowel disease. Annu Rev Immunol 28 573 621 .
  51. 51. Kawai T. Akira S. 2010 The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol 11 373 84 .
  52. 52. Kobori A. Yagi Y. Imaeda H. Ban H. Bamba S. Tsujikawa T. Saito Y. Fujiyama Y. Andoh A. 2010 Interleukin-33 expression is specifically enhanced in inflamed mucosa of ulcerative colitis. J Gastroenterol 45 999 1007 .
  53. 53. Komai-Koma M. Xu D. Li Y. McKenzie A. N. McInnes I. B. Liew F. Y. 2007 IL-33 is a chemoattractant for human Th2 cells. Eur J Immunol 37 2779 86 .
  54. 54. Kristjansson G. Venge P. Wanders A. Loof L. Hallgren R. 2004 Clinical and subclinical intestinal inflammation assessed by the mucosal patch technique: studies of mucosal neutrophil and eosinophil activation in inflammatory bowel diseases and irritable bowel syndrome. Gut 53 1806 12 .
  55. 55. Kuhn R. Lohler J. Rennick D. Rajewsky K. Muller W. 1993 Interleukin-10-deficient mice develop chronic enterocolitis. Cell 75 263 74 .
  56. 56. Kumagai Y. Akira S. 2010 Identification and functions of pattern-recognition receptors. J Allergy Clin Immunol 125 985 92 .
  57. 57. Kuroiwa K. Arai T. Okazaki H. Minota S. Tominaga S. 2001 Identification of human ST2 protein in the sera of patients with autoimmune diseases. Biochem Biophys Res Commun 284 1104 8 .
  58. 58. Lamkanfi M. Dixit V. M. 2009 IL-33 raises alarm. Immunity 31 5 7 .
  59. 59. Langhorst J. Elsenbruch S. Koelzer J. Rueffer A. Michalsen A. Dobos G. J. 2008 Noninvasive markers in the assessment of intestinal inflammation in inflammatory bowel diseases: performance of fecal lactoferrin, calprotectin, and PMN-elastase, CRP, and clinical indices. Am J Gastroenterol 103 162 9 .
  60. 60. LeBouder E. Rey-Nores J. E. Rushmere N. K. Grigorov M. Lawn S. D. Affolter M. Griffin G. E. Ferrara P. Schiffrin E. J. Morgan B. P. Labeta M. O. 2003 Soluble forms of Toll-like receptor (TLR)2 capable of modulating TLR2 signaling are present in human plasma and breast milk. J Immunol 171 6680 9 .
  61. 61. Lee D. M. Friend D. S. Gurish M. F. Benoist C. Mathis D. Brenner M. B. 2002 Mast cells: a cellular link between autoantibodies and inflammatory arthritis. Science 297 1689 92 .
  62. 62. Leung B. P. Xu D. Culshaw S. McInnes I. B. Liew F. Y. 2004 A novel therapy of murine collagen-induced arthritis with soluble T1/ST2. J Immunol 173 145 50 .
  63. 63. Li M. C. He S. H. 2004 IL-10 and its related cytokines for treatment of inflammatory bowel disease. World J Gastroenterol 10 620 5 .
  64. 64. Li X. Conklin L. Alex P. 2008 New serological biomarkers of inflammatory bowel disease. World J Gastroenterol 14 5115 24 .
  65. 65. Lichtenstein G. R. Rutgeerts P. 2010 Importance of mucosal healing in ulcerative colitis. Inflamm Bowel Dis 16 338 46 .
  66. 66. Luthi A. U. Cullen S. P. McNeela E. A. Duriez P. J. Afonina I. S. Sheridan C. Brumatti G. Taylor R. C. Kersse K. Vandenabeele P. Lavelle E. C. Martin S. J. 2009 Suppression of interleukin-33 bioactivity through proteolysis by apoptotic caspases. Immunity 31 84 98 .
  67. 67. Manolio T. A. 2010 Genomewide association studies and assessment of the risk of disease. N Engl J Med 363 166 76 .
  68. 68. McGovern D. P. Gardet A. Torkvist L. Goyette P. Essers J. Taylor K. D. Neale B. M. Ong R. T. Lagace C. Li C. Green T. Stevens C. R. Beauchamp C. Fleshner P. R. Carlson M. D’Amato M. Halfvarson J. Hibberd M. L. Lordal M. Padyukov L. Andriulli A. Colombo E. Latiano A. Palmieri O. Bernard E. J. Deslandres C. Hommes D. W. de Jong D. J. Stokkers P. C. Weersma R. K. Sharma Y. Silverberg M. S. Cho J. H. Wu J. Roeder K. Brant S. R. Schumm L. P. Duerr R. H. Dubinsky M. C. Glazer N. L. Haritunians T. Ippoliti A. Melmed G. Y. Siscovick D. S. Vasiliauskas E. A. Targan S. R. Annese V. Wijmenga C. Pettersson S. Rotter J. I. Xavier R. J. Daly M. J. Rioux J. D. Seielstad M. 2010 Genome-wide association identifies multiple ulcerative colitis susceptibility loci. Nat Genet 42 332 7 .
  69. 69. Medzhitov R. 2001 Toll-like receptors and innate immunity. Nat Rev Immunol 1 135 45 .
  70. 70. Medzhitov R. 2010 Inflammation 2010: new adventures of an old flame. Cell 140 771 6 .
  71. 71. Mok M. Y. Huang F. P. Ip W. K. Lo Y. Wong F. Y. Chan E. Y. Lam K. F. Xu D. 2010 Serum levels of IL-33 and soluble ST2 and their association with disease activity in systemic lupus erythematosus. Rheumatology (Oxford) 49 520 7 .
  72. 72. Moritz D. R. Rodewald H. R. Gheyselinck J. Klemenz R. 1998 The IL-1 receptor-related T1 antigen is expressed on immature and mature mast cells and on fetal blood mast cell progenitors. J Immunol 161 4866 74 .
  73. 73. Mosser D. M. Zhang X. 2008 Interleukin-10: new perspectives on an old cytokine. Immunol Rev 226 205 18 .
  74. 74. Nishida Y. Murase K. Isomoto H. Furusu H. Mizuta Y. Riddell R. H. Kohno S. 2002 Different distribution of mast cells and macrophages in colonic mucosa of patients with collagenous colitis and inflammatory bowel disease. Hepatogastroenterology 49 678 82 .
  75. 75. Oboki K. Nakae S. Matsumoto K. Saito H. 2011 IL-33 and Airway Inflammation. Allergy Asthma Immunol Res 3 81 8 .
  76. 76. Osada T. Ohkusa T. Yokoyama T. Shibuya T. Sakamoto N. Beppu K. Nagahara A. Otaka M. Ogihara T. Watanabe S. 2010 Comparison of several activity indices for the evaluation of endoscopic activity in UC: inter- and intraobserver consistency. Inflamm Bowel Dis 16 192 7.
  77. 77. Oshikawa K. Kuroiwa K. Tokunaga T. Kato T. Hagihara S. I. Tominaga S. I. Sugiyama Y. 2001a Acute eosinophilic pneumonia with increased soluble ST2 in serum and bronchoalveolar lavage fluid. Respir Med 95 532 3 .
  78. 78. Oshikawa K. Kuroiwa K. Tago K. Iwahana H. Yanagisawa K. Ohno S. Tominaga S. I. Sugiyama Y. 2001b Elevated soluble ST2 protein levels in sera of patients with asthma with an acute exacerbation. Am J Respir Crit Care Med 164 277 81 .
  79. 79. Palmer G. Gabay C. 2011 Interleukin-33 biology with potential insights into human diseases. Nat Rev Rheumatol 7 321 9 .
  80. 80. Palmer G. Lipsky B. P. Smithgall M. D. Meininger D. Siu S. Talabot-Ayer D. Gabay C. Smith D. E. 2008 The IL-1 receptor accessory protein (AcP) is required for IL-33 signaling and soluble AcP enhances the ability of soluble ST2 to inhibit IL-33. Cytokine 42 358 64 .
  81. 81. Papp M. Norman G. L. Altorjay I. Lakatos P. L. 2007 Utility of serological markers in inflammatory bowel diseases: gadget or magic? World J Gastroenterol 13 2028 36 .
  82. 82. Pastorelli L. Garg R. R. Hoang S. B. Spina L. Mattioli B. Scarpa M. Fiocchi C. Vecchi M. Pizarro T. T. 2010 Epithelial-derived IL-33 and its receptor ST2 are dysregulated in ulcerative colitis and in experimental Th1/Th2 driven enteritis. Proc Natl Acad Sci U S A 107 8017 22 .
  83. 83. Pecaric-Petkovic T. Didichenko S. A. Kaempfer S. Spiegl N. Dahinden C. A. 2009 Human basophils and eosinophils are the direct target leukocytes of the novel IL-1 family member IL-33. Blood 113 1526 34 .
  84. 84. Pezzilli R. Barassi A. Morselli Labate. A. M. Finazzi S. Fantini L. Gizzi G. Lotzniker M. Villani V. Melzi d’Eril G. Corinaldesi R. 2008 Fecal calprotectin levels in patients with colonic polyposis. Dig Dis Sci 53 47 51 .
  85. 85. Pierik M. Joossens S. Van Steen K. Van Schuerbeek N. Vlietinck R. Rutgeerts P. Vermeire S. 2006 Toll-like receptor-1,-2, and-6 polymorphisms influence disease extension in inflammatory bowel diseases. Inflamm Bowel Dis 12 1 8 .
  86. 86. Pineton de Chambrun G. Peyrin-Biroulet L. Lemann M. Colombel J. F. 2010 Clinical implications of mucosal healing for the management of IBD. Nat Rev Gastroenterol Hepatol 7 15 29 .
  87. 87. Podolsky D. K. 2002 Inflammatory bowel disease. N Engl J Med 347 417 29 .
  88. 88. Poullis A. Foster R. Northfield T. C. Mendall M. A. 2002 Review article: faecal markers in the assessment of activity in inflammatory bowel disease. Aliment Pharmacol Ther 16 675 81 .
  89. 89. Rakoff-Nahoum S. Paglino J. Eslami-Varzaneh F. Edberg S. Medzhitov R. 2004 Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 118 229 41 .
  90. 90. Rank M. A. Kobayashi T. Kozaki H. Bartemes K. R. Squillace D. L. Kita H. 2009 IL-33-activated dendritic cells induce an atypical TH2-type response. J Allergy Clin Immunol 123 1047 54 .
  91. 91. Regueiro M. Rodemann J. Kip K. E. Saul M. Swoger J. Baidoo L. Schwartz M. Barrie A. Binion D. 2011 Physician assessment of ulcerative colitis activity correlates poorly with endoscopic disease activity. Inflamm Bowel Dis 17 1008 14 .
  92. 92. Rehman S. U. Mueller T. Januzzi J. L. Jr. 2008 Characteristics of the novel interleukin family biomarker ST2 in patients with acute heart failure. J Am Coll Cardiol 52 1458 65 .
  93. 93. Rennick D. M. Fort M. M. Davidson N. J. 1997 Studies with IL-10-/- mice: an overview. J Leukoc Biol 61 389 96 .
  94. 94. Rioux J. D. Xavier R. J. Taylor K. D. Silverberg M. S. Goyette P. Huett A. Green T. Kuballa P. Barmada M. M. Datta L. W. Shugart Y. Y. Griffiths A. M. Targan S. R. Ippoliti A. F. Bernard E. J. Mei L. Nicolae D. L. Regueiro M. Schumm L. P. Steinhart A. H. Rotter J. I. Duerr R. H. Cho J. H. Daly M. J. Brant S. R. 2007 Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet 39 596 604 .
  95. 95. Risch N. Merikangas K. 1996 The future of genetic studies of complex human diseases. Science 273 1516 7 .
  96. 96. Rodgers A. D. Cummins A. G. 2007 CRP correlates with clinical score in ulcerative colitis but not in Crohn’s disease. Dig Dis Sci 52 2063 8 .
  97. 97. Roseth A. G. Kristinsson J. Fagerhol M. K. Schjonsby H. Aadland E. Nygaard K. Roald B. 1993 Faecal calprotectin: a novel test for the diagnosis of colorectal cancer? Scand J Gastroenterol 28 1073 6 .
  98. 98. Roussel L. Erard M. Cayrol C. Girard J. P. 2008 Molecular mimicry between IL-33 and KSHV for attachment to chromatin through the H2A-H2B acidic pocket. EMBO Rep 9 1006 12 .
  99. 99. Rutgeerts P. Vermeire S. Van Assche G. 2007 Mucosal healing in inflammatory bowel disease: impossible ideal or therapeutic target? Gut 56 453 5 .
  100. 100. Sanada S. Hakuno D. Higgins L. J. Schreiter E. R. McKenzie A. N. Lee R. T. 2007 IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system. J Clin Invest 117 1538 49 .
  101. 101. Schleimer R. P. Kato A. Kern R. Kuperman D. Avila P. C. 2007 Epithelium: at the interface of innate and adaptive immune responses. J Allergy Clin Immunol 120 1279 84 .
  102. 102. Schmitz J. Owyang A. Oldham E. Song Y. Murphy E. McClanahan T. K. Zurawski G. Moshrefi M. Qin J. Li X. Gorman D. M. Bazan J. F. Kastelein R. A. 2005 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 23 479 90 .
  103. 103. Schoepfer A. M. Beglinger C. Straumann A. Trummler M. Vavricka S. R. Bruegger L. E. Seibold F. 2010 Fecal calprotectin correlates more closely with the Simple Endoscopic Score for Crohn’s disease (SES-CD) than CRP, blood leukocytes, and the CDAI. Am J Gastroenterol 105 162 9 .
  104. 104. Seidelin J. B. Bjerrum J. T. Coskun M. Widjaya B. Vainer B. Nielsen O. H. 2010 IL-33 is upregulated in colonocytes of ulcerative colitis. Immunol Lett 128 80 5 .
  105. 105. Seiderer J. Brand S. Herrmann K. A. Schnitzler F. Hatz R. Crispin A. Pfennig S. Schoenberg S. O. Goke B. Lohse P. Ochsenkuhn T. 2006 Predictive value of the CARD15 variant 1007fs for the diagnosis of intestinal stenoses and the need for surgery in Crohn’s disease in clinical practice: results of a prospective study. Inflamm Bowel Dis 12 1114 21 .
  106. 106. Shih D. Q. Targan S. R. 2008 Immunopathogenesis of inflammatory bowel disease. World J Gastroenterol 14 390 400 .
  107. 107. Shimizu M. Matsuda A. Yanagisawa K. Hirota T. Akahoshi M. Inomata N. Ebe K. Tanaka K. Sugiura H. Nakashima K. Tamari M. Takahashi N. Obara K. Enomoto T. Okayama Y. Gao P. S. Huang S. K. Tominaga S. Ikezawa Z. Shirakawa T. 2005 Functional SNPs in the distal promoter of the ST2 gene are associated with atopic dermatitis. Hum Mol Genet 14 2919 27 .
  108. 108. Silverberg M. S. Satsangi J. Ahmad T. Arnott I. D. Bernstein C. N. Brant S. R. Caprilli R. Colombel J. F. Gasche C. Geboes K. Jewell D. P. Karban A. Loftus E. V. Jr. Pena A. S. Riddell R. H. Sachar D. B. Schreiber S. Steinhart A. H. Targan S. R. Vermeire S. Warren B. F. 2005 Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: Report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Can J Gastroenterol 19 Suppl A:5-36.
  109. 109. Sipponen T. Savilahti E. Kolho K. L. Nuutinen H. Turunen U. Farkkila M. 2008 Crohn’s disease activity assessed by fecal calprotectin and lactoferrin: correlation with Crohn’s disease activity index and endoscopic findings. Inflamm Bowel Dis 14 40 6 .
  110. 110. Solem C. A. Loftus E. V. Jr Tremaine W. J. Harmsen W. S. Zinsmeister A. R. Sandborn W. J. 2005 Correlation of C-reactive protein with clinical, endoscopic, histologic, and radiographic activity in inflammatory bowel disease. Inflamm Bowel Dis 11 707 12 .
  111. 111. Stolarski B. Kurowska-Stolarska M. Kewin P. Xu D. Liew F. Y. 2010 IL-33 exacerbates eosinophil-mediated airway inflammation. J Immunol 185 3472 80 .
  112. 112. Summerton C. B. Longlands M. G. Wiener K. Shreeve D. R. 2002 Faecal calprotectin: a marker of inflammation throughout the intestinal tract. Eur J Gastroenterol Hepatol 14 841 5 .
  113. 113. Suzukawa M. Iikura M. Koketsu R. Nagase H. Tamura C. Komiya A. Nakae S. Matsushima K. Ohta K. Yamamoto K. Yamaguchi M. 2008a An IL-1 cytokine member, IL-33, induces human basophil activation via its ST2 receptor. J Immunol 181 5981 9 .
  114. 114. Suzukawa M. Koketsu R. Iikura M. Nakae S. Matsumoto K. Nagase H. Saito H. Matsushima K. Ohta K. Yamamoto K. Yamaguchi M. 2008b Interleukin-33 enhances adhesion, CD11b expression and survival in human eosinophils. Lab Invest 88 1245 53 .
  115. 115. Takezako N. Hayakawa M. Hayakawa H. Aoki S. Yanagisawa K. Endo H. Tominaga S. 2006 ST2 suppresses IL-6 production via the inhibition of IkappaB degradation induced by the LPS signal in THP-1 cells. Biochem Biophys Res Commun 341 425 32 .
  116. 116. Talabot-Ayer D. Lamacchia C. Gabay C. Palmer G. 2009 Interleukin-33 is biologically active independently of caspase-1 cleavage. J Biol Chem 284 19420 6 .
  117. 117. Thompson A. I. Lees C. W. 2011 Genetics of ulcerative colitis. Inflamm Bowel Dis 17 831 48 . DOI: 10.1002/ibd.21375.
  118. 118. Tibble J. Sigthorsson G. Foster R. Sherwood R. Fagerhol M. Bjarnason I. 2001 Faecal calprotectin and faecal occult blood tests in the diagnosis of colorectal carcinoma and adenoma. Gut 49 402 8 .
  119. 119. Tibble J. A. Bjarnason I. 2001 Non-invasive investigation of inflammatory bowel disease. World J Gastroenterol 7 460 5 .
  120. 120. Tibble J. A. Sigthorsson G. Foster R. Forgacs I. Bjarnason I. 2002 Use of surrogate markers of inflammation and Rome criteria to distinguish organic from nonorganic intestinal disease. Gastroenterology 123 450 60 .
  121. 121. Tominaga S. Jenkins N. A. Gilbert D. J. Copeland N. G. Tetsuka T. 1991 Molecular cloning of the murine ST2 gene. Characterization and chromosomal mapping. Biochim Biophys Acta 1090 1 8 .
  122. 122. Vermeire S. Van Assche G. Rutgeerts P. 2004 C-reactive protein as a marker for inflammatory bowel disease. Inflamm Bowel Dis 10 661 5 .
  123. 123. Vermeire S. Van Assche G. Rutgeerts P. 2006 Laboratory markers in IBD: useful, magic, or unnecessary toys? Gut 55 426 31 .
  124. 124. Vermeire S. Van Assche G. Rutgeerts P. 2010 Role of genetics in prediction of disease course and response to therapy. World J Gastroenterol 16 2609 15 .
  125. 125. Verri W. A. Jr. Souto F. O. Vieira S. M. Almeida S. C. Fukada S. Y. Xu D. Alves-Filho J. C. Cunha T. M. Guerrero A. T. Mattos-Guimaraes R. B. Oliveira F. R. Teixeira M. M. Silva J. S. Mc Innes I. B. Ferreira S. H. Louzada-Junior P. Liew F. Y. Cunha F. Q. 2010 IL-33 induces neutrophil migration in rheumatoid arthritis and is a target of anti-TNF therapy. Ann Rheum Dis 69 1697 703 .
  126. 126. Wang F. Tahara T. Arisawa T. Shibata T. Nakamura M. Fujita H. Iwata M. Kamiya Y. Nagasaka M. Takahama K. Watanabe M. Hirata I. Nakano H. 2007 Genetic polymorphisms of CD14 and Toll-like receptor-2 (TLR2) in patients with ulcerative colitis. J Gastroenterol Hepatol 22 925 9 .
  127. 127. Wang T. Lafuse W. P. Zwilling B. S. 2001 NFkappaB and Sp1 elements are necessary for maximal transcription of toll-like receptor 2 induced by Mycobacterium avium. J Immunol 167 6924 32 .
  128. 128. Weinberg E. O. 2009 ST2 protein in heart disease: from discovery to mechanisms and prognostic value. Biomark Med 3 495 511 .
  129. 129. Weinberg E. O. Shimpo M. Hurwitz S. Tominaga S. Rouleau J. L. Lee R. T. 2003 Identification of serum soluble ST2 receptor as a novel heart failure biomarker. Circulation 107 721 6 .
  130. 130. Xavier R. J. Podolsky D. K. 2007 Unravelling the pathogenesis of inflammatory bowel disease. Nature 448 427 34 .
  131. 131. Zhao W. Hu Z. 2010 The enigmatic processing and secretion of interleukin-33. Cell Mol Immunol 7 260 2 .
  132. 132. Zhernakova A. Festen E. M. Franke L. Trynka G. van Diemen C. C. Monsuur A. J. Bevova M. Nijmeijer R. M. van’t Slot R. Heijmans R. Boezen H. M. van Heel D. A. van Bodegraven A. A. Stokkers P. C. Wijmenga C. Crusius J. B. Weersma R. K. 2008 Genetic analysis of innate immunity in Crohn’s disease and ulcerative colitis identifies two susceptibility loci harboring CARD9 and IL18RAP. Am J Hum Genet 82 1202 10 .
  133. 133. Allakhverdi Z. Smith D. E. Comeau M. R. Delespesse G. 2007 Cutting edge: The ST2 ligand IL-33 potently activates and drives maturation of human mast cells. J Immunol 179 2051 4 .

Written By

David Díaz-Jiménez, Katya Carrillo, Rodrigo Quera and Marcela A. Hermoso

Submitted: 24 February 2011 Published: 18 January 2012