Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Jan 7, 2014; 20(1): 91-99
Published online Jan 7, 2014. doi: 10.3748/wjg.v20.i1.91
Inflammatory bowel disease: Pathogenesis
Yi-Zhen Zhang, Yong-Yu Li, Department of Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
Author contributions: Zhang YZ and Li YY have substantial contributions to conception and design, and acquisition of data; Zhang YZ drafted the article; Li YY revised the manuscript critically for important intellectual content; Zhang YZ and Li YY worked together for the final approval of the version to be published.
Supported by Grants from the National Natural Science Foundation of China, No. 81270477
Correspondence to: Yong-Yu Li, MD, Department of Pathophysiology, Tongji University School of Medicine, Siping Road 1239, Shanghai 200092, China. liyongyu@tongji.edu.cn
Telephone: +86-21-65981021 Fax: +86-21-65987071
Received: September 29, 2013
Revised: November 5, 2013
Accepted: November 28, 2013
Published online: January 7, 2014

Abstract

Inflammatory bowel disease (IBD), including Crohn’s disease and ulcerative colitis, is characterized by chronic relapsing intestinal inflammation. It has been a worldwide health-care problem with a continually increasing incidence. It is thought that IBD results from an aberrant and continuing immune response to the microbes in the gut, catalyzed by the genetic susceptibility of the individual. Although the etiology of IBD remains largely unknown, it involves a complex interaction between the genetic, environmental or microbial factors and the immune responses. Of the four components of IBD pathogenesis, most rapid progress has been made in the genetic study of gut inflammation. The latest internationally collaborative studies have ascertained 163 susceptibility gene loci for IBD. The genes implicated in childhood-onset and adult-onset IBD overlap, suggesting similar genetic predispositions. However, the fact that genetic factors account for only a portion of overall disease variance indicates that microbial and environmental factors may interact with genetic elements in the pathogenesis of IBD. Meanwhile, the adaptive immune response has been classically considered to play a major role in the pathogenesis of IBD, as new studies in immunology and genetics have clarified that the innate immune response maintains the same importance in inducing gut inflammation. Recent progress in understanding IBD pathogenesis sheds lights on relevant disease mechanisms, including the innate and adaptive immunity, and the interactions between genetic factors and microbial and environmental cues. In this review, we provide an update on the major advances that have occurred in above areas.

Key Words: Inflammatory bowel disease, Ulcerative colitis, Crohn’s disease, Pathogenesis, Genetics, Microbial factors, Immune responses

Core tip: Inflammatory bowel disease (IBD) includes Crohn’s disease and ulcerative colitis. Recent research indicated that the individual’s genetic susceptibility, external environment, intestinal microbial flora and immune responses are all involved and functionally integrated in the pathogenesis of IBD. The main purpose of this review is to offer an update that have occurred in each of the above four areas, and to highlight the future work to find a clear understanding of IBD pathogenesis.



INTRODUCTION

Inflammatory bowel disease (IBD) has been a global healthcare problem with a sustained increasing incidence[1]. It includes two major forms, Crohn’s disease (CD) and ulcerative colitis (UC), which are distinct chronic bowel-relapsing inflammatory disorders. CD can cause transmural inflammation and affect any part of the gastrointestinal tract (most commonly, the terminal ileum or the perianal region) in a non-continuous type. Unlike UC, CD is commonly associated with complications such as abscesses, fistulas and strictures. In contrast, UC is typified by mucosal inflammation and limited to the colon[2]. Although the etiology of IBD remains largely unknown, recent research indicated that the individual’s genetic susceptibility, external environment, intestinal microbial flora and immune responses are all involved and functionally integrated in the pathogenesis of IBD[3-5]. The main purpose of this review is to offer an update that have occurred in each of the above four areas, and to highlight the future work to find a clear understanding of IBD pathogenesis.

GENETICS

Over the past decades, there have been huge advances in our understanding of genetic contributions to IBD[6]. This is due to the technological advances in DNA analysis and sequencing and the use of huge multinational databases[7]. Advances in genetic testing and analyzing technologies have allowed for the completion of many genome-wide association studies (GWAS) which identify single nucleotide polymorphisms (SNPs). Recent studies have brought the number of IBD-associated gene loci to 163, of which 110 are associated with both diseases, 30 CD specific and 23 UC specific[8]. Studies of gene loci shared by UC and CD may provide new way to find their common pathogenesis.

The era of modern IBD genetic research began in 2001 with the discovery of NOD2 (nucleotide-binding oligomerization domaincontaining 2), the first susceptibility gene for CD[9]. The NOD2 gene codes for a protein that was originally described as an intracellular receptor recognizing the muramyl dipeptide (MDP), a conserved motif present in peptidoglycan from both Gram-positive and -negative bacteria[10]. MDP stimulation induces autophagy which controls bacterial replication and antigen presentation[11,12], and modulates both innate and adaptive immune responses[13]. NOD2 participates in distinct MDP-independent pathways such as the regulation of the T-cell response[14]. The association between CD and NOD2 has already been replicated at the genome-wide significance level[15].

Genetic analyses have shown an indispensable role for autophagy in immune responses in IBD, and reported two autophagy-related genes named ATG16L1 and IRGM[16-18]. Autophagy is involved in intracellular homeostasis, contributing to the degradation and recycling of cytosolic contents and organelles, as well as to the resistance against infection and removal of intracellular microbes[19]. ATG16L1 is essential for all forms of autophagy, and the coding mutation T300A is associated with an increased risk of CD. IRGM belongs to the p47 immunity-related GTPase family. CD-associated polymorphisms in IRGM lead to reduced protein expression. Epithelial cells and dendritic cells containing ATG16L1 and NOD2 variants show defects in antibacterial autophagy[12,20].

With the widespread use of GWAS and SNPs, a significant association between IBD and the IL23R gene has recently been described[21]. The IL23R gene encodes a subunit of the receptor for the pro-inflammatory cytokine interleukin (IL)-23, a peptide involved in the generation of Th17 cells. The Th17 and IL-23 pathway is well established in the pathogenesis of IBD, with susceptibility gene loci IL23R, IL12B, JAK2, and STAT3 having been identified in both UC and CD[22,23]. Variants in IL12B, which encodes the p40 subunit of IL-12 and IL-23, have been associated with IBD and other immune disorders. Defects in the function of IL-10 have also been associated with CD and UC[24]. Other susceptibility genes that regulate immune function include CARD9, IL1R2, REL, SMAD3 and PRDM1.

Recent progress in the genetics of IBD holds several key messages in regard to the underlying mechanism of the disease. On one hand, the expanding number of susceptibility gene loci described in IBD indicates that genetic influences are critical components of the disease pathogenesis; while on the other hand, explainable susceptibility loci discovered so far account for only 20%-25% of the heritability found in the above-mentioned studies. This is not only true for IBD, but also true for many other polygenetic diseases, and the phenomenon has been called “the mystery of missing heritability of common traits” or “genetic vacuum”[25]. This issue is further proved by GWAS that has failed to add new susceptibility gene loci to fill the “genetic vacuum”[26,27]. The possibility was then proposed that, instead of missing genes, the interactions among genes and their products could explain much of the apparent vacuum and account for a considerable number of IBD[25]. These new insights into genetics and heritability of IBD implicate that future explorations of gene-gene interactions, gene-pathway interactions and gene-environment interactions are likely to give us more insights into IBD pathogenesis than finding new rare variants.

ENVIRONMENT

There is no doubt that environmental factors play an important role in the pathogenesis of IBD. A large number of environmental factors are considered risk factors for IBD, including smoking, diet, drugs, geography, social stress, and psychological element[28]. Among them, smoking remains the most widely studied and replicated environmental prompter for IBD. Since the first described inverse association between UC and smoking in 1982, subsequent studies have confirmed the protective effect of heavy smoking on the development of UC with a lower rate of relapse[29-31]. Contrary to its effect on UC, smoking increases the risk of CD and is associated with a higher rate of postoperative disease[32].

Traditional conception for vitamin D’s role is concentrated in calcium metabolism and bone health. Nowadays, there has been increasing recognition of the immunologic role of vitamin D[33]. Recent literature suggests that the role of vitamin D is multifarious and associated with diverse diseases including IBD. Leslie et al[34] found that vitamin D deficiency had been common in diagnosed IBD patients and pointed out that low vitamin D had contributed to the increased risk of IBD. In mouse models, vitamin D deficiency is associated with an increased susceptibility to dextran sodium sulfate-induced colitis and 1,25(OH)2D3 supplementation ameliorates the severity of intestinal inflammation[35].

The effect of aspirin and nonsteroidal anti-inflammatory drugs (NSAIDs) in the gastrointestinal tract is well recognized. However, limited high quality evidence is available to support the notion that aspirin and NSAIDs have an effect in triggering onset or relapse of IBD. Ananthakrishnan et al[36] found no association between the dose, duration, or frequency of aspirin use and the risk for CD or UC; but the high dose, prolonged using duration, and frequent use of NSAIDs had been associated with an increased risk of CD and UC. A recent study has found that the use of antibiotics is an important environmental factor, influencing the risk of IBD through their effect on the microbiome. Antibiotic use within the first year of life is more common among pediatric IBD cases compared to controls[37].

Stress has long been proposed to play a role in the pathogenesis of CD and UC[38-40]. Bitton et al[41] suggested that individuals with lower levels of stress had a reduced risk of the disease onset. Mood components of perceived stress, including depression and anxiety, may play a strong role in mediating the deterioration of IBD[42]. A retrospective study by Goodhand et al[43] found a reduction in the number of symptomatic relapses in participants, and the antidepressants beneficially impact the course of IBD. However, a Cochrane review shows no benefit of psychological interventions in IBD[44].

Recent ecological and epidemiologic evidence suggests that air pollution may contribute to the risk of CD and UC. The rising incidence of CD and UC in developing countries parallels the development of industrialization[45]. Elevated air pollution is associated with an augment in circulating polymorphonuclear leukocytes and plasma cytokines[46,47]. Kaplan et al[48] using The Health Improvement Network Database in the United Kingdom, found that high levels of NO2 and SO2 correlate with the increased risk of CD and UC. In another study, total pollutant emission has been linked to increased rates of hospitalizations for both CD and UC, suggesting that ambient air pollution may also influence these established diseases[49].

MICROBIAL FACTORS

The whole human gut microbiome consists of approximately 1150 bacterial species, with each individual host having roughly 160 species[50]. Gut microbiome is established within the first 2 wk of life and then usually remains remarkably stable thereafter. Although it is only possible to culture 20%-30% of the gut microbiome, the association between the changes in the microbiome and IBD has been established[51]. Many studies have examined the gut flora in CD and UC in both inflamed and non-inflamed segments, and found that there is a significantly reduced biodiversity in faecal microbiome in IBD patients compared to that in healthy controls[52]. Other research has also found that the microbiota in IBD patients is unstable than that in healthy individual[53]. In healthy intestine, the Firmicutes and Bacteroidetes phyla predominate, and contribute to the production of epithelial metabolic substrates. In contrast, the microbiota is characterized by a relative lack of Firmicutes and Bacteroidetes, and an over-representation of enterobacteria in CD; meanwhile, a reduction in Clostridium spp. and an increase in Escherichia coli (E. coli) have been reported in UC[54].

In healthy colon, there is a continuous mucus coating consisting of two layers of sub-structures: the outer is a loosely adherent layer, good for bacterial growth; while the inner is a tightly adherent layer, normally sterile. In IBD, particularly CD, there is a marked increase in bacteria associated with the colonic adherent mucus layer[55,56]. In CD, a consistent increase in mucosa-associated E. coli and reduction in Firmicutes are reported[57,58]. There is strong evidence for an increase in mucosa-associated E. coli in both the ileum and colon, and their presence within the granulomas in CD implicates a primary pathogenic role[59]. An adherent and invasive E. coli (AIEC) phenotype has been found in CD, which is typified by bacterial invasion into epithelial cells and replication within macrophages[60]. AIEC has also been shown to induce granuloma in vitro and granulomatous colitis in Boxer dogs[61].

IMMUNOLOGICAL FACTORS

The investigation of IBD pathogenesis has been dominated for a long time by the studies of mucosal immunity, especially the T cell response. Available evidence suggests that the dysfunctions of innate and adaptive immune pathways contribute to the aberrant intestinal inflammatory response in patients with IBD. Most studies in the last two decades have focused on the role of abnormal adaptive immune responses in the pathogenesis of IBD. The focus on the adaptive immune response has ultimately led to the notion that the two main types of IBD represent clearly distinct forms of gut inflammation: CD has long been considered to be driven by a Th1 response and UC has been associated with a non-conventional Th2 response[62,63]. The newly described Th17 cells are also involved in the gut inflammatory response in IBD[64]. Immunological studies have recently focused on the mucosal innate immune responses, such as epithelial barrier integrity, innate microbial sensing, autophagy and unfolded protein response.

Innate immunity

The innate immune response represents our first line of defense against pathogens. It is non-specific, allowing the body to quickly respond to stimuli often within minutes or hours. The innate immune response is mediated by a large variety of different cell types including epithelial cells, neutrophils, dendritic cells, monocytes, macrophages and natural killer cells[65]. This form of immunity is initiated by the recognition of microbial antigens, which is mediated by pattern recognition receptors including toll-like receptors (TLRs) on the cell surface and NOD-like receptors in the cytoplasm[66]. Recent studies have found that the behavior of the cells mediating innate immunity and the expression and function of both TLRs and NOD proteins are altered significantly in individuals with IBD.

A British study shows that mucosal neutrophil accumulation and production of IL-1β and IL-8 in response to trauma are selectively reduced in CD patients but not in UC patients[67]. GWAS reveal that the NOD2 mutations most commonly associated with CD induce a defective ability of the gut to respond to LPS, and this defect may contribute to disease susceptibility[68]. Although the functional role of NOD2 mutations is still controversial, available evidence suggests that they represent loss-of-function mutations that lead to reduced activation of NF-κB[69]. This inadequate response might result in reduced antibacterial agent production and pathogenic microbial invasion[70]. Other studies suggest that the loss of function of NOD2 may result in the lack of inhibition of TLR2 stimulation, leading to activation of inflammatory pathways and excessive Th-1 responses[71]. Furthermore, NOD2 also contributes to immune tolerance. These effects are impaired in cells from patients with NOD2 mutation 3020insC[72].

IL-23 is a key cytokine both in innate and adaptive immunity and possesses a central role in driving early responses against microbes. IL23R polymorphisms have been associated with both CD and UC, suggesting that IL-23 may represent a shared inflammatory molecule in chronic intestinal inflammation. Recent studies have shown that, besides its activity on Th17 cells, IL-23 can also act on cells of the innate immune system. IL-23 has been shown to induce Th17 cytokine production by innate lymphoid cells (ILCs) that share the phenotype of lymphoid tissue-induced cells[73].

CD has also been associated with ATG16L1 and IRGM genes, which are involved in autophagy. ATG16L1 is essential for all forms of autophagy, and the coding mutation T300A is associated with an increased risk of CD. Autophagy is one of the mechanisms for maintaining cellular homeostasis and considered very important for host defense against intracellular microorganisms. Normally, autophagy is induced by bactericidal effects and presentation of endogenous antigens, and these processes are impaired in patients with mutations in NOD2 or Atg16L1. Closely relating to autophagy and innate immunity, dysregulation of the unfolded protein response may also contribute to IBD pathogenesis. This response is induced by endoplasmic reticulum stress and finally induces apoptotic cell death and causes IBD[74].

In addition, defective epithelial barrier and increased intestinal permeability have long been observed in IBD patients[75]. The first physical barrier that intestinal bacteria and food antigens encounter on the mucosal surface is represented by the mucous layer that covers the intestinal epithelium. The importance of mucus in the prevention of bacterial break-through and intestinal inflammation has been proved by many studies[76]. The second line of defense against bacterial invasion is formed by the intestinal epithelium which consists of enterocytes and specialized epithelial cells, such as goblet cells and Paneth cells. Besides forming a physical barrier against bacteria, epithelial cells can secrete a number of antimicrobial peptides. Defective expression of antimicrobial peptides has been observed in patients with CD[77].

Adaptive immunity

As opposed to the innate immune response, the adaptive immunity is highly specific, often takes several days to respond and depends on the type and number of T cells. Th1 cells, induced by IL-12, produce a high amount of IFN-γ, whereas Th2 cells release IL-4, IL-5 and IL-13[78]. An abnormal Th1 immune response is thought to cause intestinal inflammation in CD, and it has been observed that mucosal T cells from CD patients produce higher amounts of IL-2 and IFN-γ than T cells from UC patients or controls do[79]. It has also been shown that in UC, atypical NK T cells release higher amounts of the Th2 cytokine IL-13 than T cells from controls or CD patients do[80,81]. Therefore, CD has been thought to be characterized by a Th1 immune response, while UC has been considered as a Th2-mediated disease[82]. However, there have also been different observations about mucosal Th1 and Th2 cytokines in IBD. Both UC and CD biopsies cultured in vitro release high and comparable amounts of IFN-γ[83]. Lower levels of IL-13 are found in the colonic mucosa of UC patients compared to those in CD patients and subjects of the control group. Recent studies on experimental colitis have suggested an anti-inflammatory effect of IL-13 in the gut[84,85]. It has been also observed that IL-13 levels in the supernatants of intestinal biopsies cultured in vitro are lower than IFN-γ concentration and the concentrations are comparable among CD, UC and control groups[86]. Similar observations have been reported by Bernardo et al[87] who have described the presence of a mixed cytokine profile with predominance of IL-6 and the absence of IL-13 in supernatants of UC biopsies cultured in vitro. Collectively, these data should lead us to reconsider the Th1/Th2 paradigm in CD and UC[82], but the conception that UC is a Th2-mediated disease remains controversial.

Th17 cells are a T cell subset characterized by the production of large amounts of IL-17A, IL-17F, IL-21 and IL-22. They are induced by a combination of IL-6 and transforming growth factor (TGF)-β, and their expansion is promoted by IL-23[88]. The involvement of Th17 cells and, in particular, their signature cytokine IL-17A in intestinal inflammation has been extensively studied. High transcript levels of IL-17A have been detected both in CD and UC mucosa in comparison to normal gut[89,90]. Moreover, the inflamed IBD mucosa cultured in vitro produces higher levels of IL-17A than the control[79]. Furthermore, Th17 cells are an important source of IL-21, an IL-2-related cytokine which is up-regulated in inflamed IBD mucosa[91,92]. The true role of Th17 cells in IBD pathogenesis is currently undergoing intense scrutiny, and it is particularly fascinating that Th17 cells express the IL-23R on their surface[93].

CONCLUSION

There is no doubt that an unprecedented progress in our understanding of IBD pathogenesis has been achieved during the past few years. The key factors responsible for IBD include genetic components, environmental elements, microbial flora and immune responses. It is hard to dispute the popular belief that IBD arises from an extremely complex interaction among genetic and environmental elements, dysregulated immune responses and alterations of the microbiome, and that none of these factors alone is likely to cause the disease. More detailed information on their composition, function, and interaction is becoming increasingly accessible through high genomic approaches, investigation of environmental changes, molecular analysis of gut bacteria flora, and a more integrated understanding of the interaction between innate and adaptive immune responses. The growing number and diversity of genetic loci associated with IBD provide major challenges to the investigation of how they impact immunity and inflammation in susceptible individuals. Future research needs to further clarify and integrate the effects of the microbiome and environment on the immune response, and it shall be essential to gain further insights into the mechanisms and pathways of how bacteria, viruses or even fungi can modulate innate and adaptive immune responses.

Footnotes

P- Reviewers: Manguso F, Osawa S S- Editor: Ma YJ L- Editor: Wang TQ E- Editor: Zhang DN

References
1.  Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448:427-434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2894]  [Cited by in F6Publishing: 3106]  [Article Influence: 182.7]  [Reference Citation Analysis (8)]
2.  Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med. 2009;361:2066-2078.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1967]  [Cited by in F6Publishing: 2028]  [Article Influence: 135.2]  [Reference Citation Analysis (5)]
3.  Danese S, Fiocchi C. Etiopathogenesis of inflammatory bowel diseases. World J Gastroenterol. 2006;12:4807-4812.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Kugathasan S, Fiocchi C. Progress in basic inflammatory bowel disease research. Semin Pediatr Surg. 2007;16:146-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
5.  Podolsky DK. Inflammatory bowel disease. N Engl J Med. 2002;347:417-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2693]  [Cited by in F6Publishing: 2682]  [Article Influence: 121.9]  [Reference Citation Analysis (2)]
6.  Gaya DR, Russell RK, Nimmo ER, Satsangi J. New genes in inflammatory bowel disease: lessons for complex diseases? Lancet. 2006;367:1271-1284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 168]  [Article Influence: 9.3]  [Reference Citation Analysis (1)]
7.  Duerr RH. Genome-wide association studies herald a new era of rapid discoveries in inflammatory bowel disease research. Gastroenterology. 2007;132:2045-2049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
8.  Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, Lee JC, Schumm LP, Sharma Y, Anderson CA. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:119-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3465]  [Cited by in F6Publishing: 3309]  [Article Influence: 275.8]  [Reference Citation Analysis (0)]
9.  Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, Britton H, Moran T, Karaliuskas R, Duerr RH. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature. 2001;411:603-606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3555]  [Cited by in F6Publishing: 3393]  [Article Influence: 147.5]  [Reference Citation Analysis (0)]
10.  Inohara N, Ogura Y, Fontalba A, Gutierrez O, Pons F, Crespo J, Fukase K, Inamura S, Kusumoto S, Hashimoto M. Host recognition of bacterial muramyl dipeptide mediated through NOD2. Implications for Crohn’s disease. J Biol Chem. 2003;278:5509-5512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1255]  [Cited by in F6Publishing: 1219]  [Article Influence: 58.0]  [Reference Citation Analysis (0)]
11.  Cooney R, Baker J, Brain O, Danis B, Pichulik T, Allan P, Ferguson DJ, Campbell BJ, Jewell D, Simmons A. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med. 2010;16:90-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 760]  [Cited by in F6Publishing: 766]  [Article Influence: 51.1]  [Reference Citation Analysis (0)]
12.  Travassos LH, Carneiro LA, Ramjeet M, Hussey S, Kim YG, Magalhães JG, Yuan L, Soares F, Chea E, Le Bourhis L. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat Immunol. 2010;11:55-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 916]  [Cited by in F6Publishing: 967]  [Article Influence: 64.5]  [Reference Citation Analysis (0)]
13.  Shaw MH, Kamada N, Warner N, Kim YG, Nuñez G. The ever-expanding function of NOD2: autophagy, viral recognition, and T cell activation. Trends Immunol. 2011;32:73-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
14.  Sabbah A, Chang TH, Harnack R, Frohlich V, Tominaga K, Dube PH, Xiang Y, Bose S. Activation of innate immune antiviral responses by Nod2. Nat Immunol. 2009;10:1073-1080.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 515]  [Cited by in F6Publishing: 556]  [Article Influence: 37.1]  [Reference Citation Analysis (0)]
15.  Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, Ahmad T, Lees CW, Balschun T, Lee J, Roberts R. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet. 2010;42:1118-1125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1894]  [Cited by in F6Publishing: 1912]  [Article Influence: 136.6]  [Reference Citation Analysis (0)]
16.  Rioux JD, Xavier RJ, Taylor KD, Silverberg MS, Goyette P, Huett A, Green T, Kuballa P, Barmada MM, Datta LW. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet. 2007;39:596-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1392]  [Cited by in F6Publishing: 1347]  [Article Influence: 79.2]  [Reference Citation Analysis (0)]
17.  Hampe J, Franke A, Rosenstiel P, Till A, Teuber M, Huse K, Albrecht M, Mayr G, De La Vega FM, Briggs J. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet. 2007;39:207-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1387]  [Cited by in F6Publishing: 1402]  [Article Influence: 77.9]  [Reference Citation Analysis (0)]
18.  McCarroll SA, Huett A, Kuballa P, Chilewski SD, Landry A, Goyette P, Zody MC, Hall JL, Brant SR, Cho JH. Deletion polymorphism upstream of IRGM associated with altered IRGM expression and Crohn’s disease. Nat Genet. 2008;40:1107-1112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 504]  [Cited by in F6Publishing: 510]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
19.  Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of inflammatory bowel disease. Nature. 2011;474:307-317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1693]  [Cited by in F6Publishing: 1705]  [Article Influence: 131.2]  [Reference Citation Analysis (1)]
20.  Kuballa P, Huett A, Rioux JD, Daly MJ, Xavier RJ. Impaired autophagy of an intracellular pathogen induced by a Crohn’s disease associated ATG16L1 variant. PLoS One. 2008;3:e3391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 278]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
21.  Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, Daly MJ, Steinhart AH, Abraham C, Regueiro M, Griffiths A. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006;314:1461-1463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2184]  [Cited by in F6Publishing: 2199]  [Article Influence: 122.2]  [Reference Citation Analysis (0)]
22.  Anderson CA, Boucher G, Lees CW, Franke A, D’Amato M, Taylor KD, Lee JC, Goyette P, Imielinski M, Latiano A. Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47. Nat Genet. 2011;43:246-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 977]  [Cited by in F6Publishing: 998]  [Article Influence: 76.8]  [Reference Citation Analysis (0)]
23.  Brand S. Crohn’s disease: Th1, Th17 or both? The change of a paradigm: new immunological and genetic insights implicate Th17 cells in the pathogenesis of Crohn’s disease. Gut. 2009;58:1152-1167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 467]  [Cited by in F6Publishing: 474]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
24.  Tremelling M, Cummings F, Fisher SA, Mansfield J, Gwilliam R, Keniry A, Nimmo ER, Drummond H, Onnie CM, Prescott NJ. IL23R variation determines susceptibility but not disease phenotype in inflammatory bowel disease. Gastroenterology. 2007;132:1657-1664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 144]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
25.  Zuk O, Hechter E, Sunyaev SR, Lander ES. The mystery of missing heritability: Genetic interactions create phantom heritability. Proc Natl Acad Sci USA. 2012;109:1193-1198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1086]  [Cited by in F6Publishing: 1010]  [Article Influence: 84.2]  [Reference Citation Analysis (0)]
26.  Momozawa Y, Mni M, Nakamura K, Coppieters W, Almer S, Amininejad L, Cleynen I, Colombel JF, de Rijk P, Dewit O. Resequencing of positional candidates identifies low frequency IL23R coding variants protecting against inflammatory bowel disease. Nat Genet. 2011;43:43-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 154]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
27.  Rivas MA, Beaudoin M, Gardet A, Stevens C, Sharma Y, Zhang CK, Boucher G, Ripke S, Ellinghaus D, Burtt N. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nat Genet. 2011;43:1066-1073.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 619]  [Cited by in F6Publishing: 565]  [Article Influence: 43.5]  [Reference Citation Analysis (0)]
28.  Loftus EV. Clinical epidemiology of inflammatory bowel disease: Incidence, prevalence, and environmental influences. Gastroenterology. 2004;126:1504-1517.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Cosnes J. Tobacco and IBD: relevance in the understanding of disease mechanisms and clinical practice. Best Pract Res Clin Gastroenterol. 2004;18:481-496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 175]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
30.  Cosnes J. What is the link between the use of tobacco and IBD? Inflamm Bowel Dis. 2008;14 Suppl 2:S14-S15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
31.  Lakatos PL, Szamosi T, Lakatos L. Smoking in inflammatory bowel diseases: good, bad or ugly? World J Gastroenterol. 2007;13:6134-6139.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Birrenbach T, Böcker U. Inflammatory bowel disease and smoking: a review of epidemiology, pathophysiology, and therapeutic implications. Inflamm Bowel Dis. 2004;10:848-859.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Garg M, Lubel JS, Sparrow MP, Holt SG, Gibson PR. Review article: vitamin D and inflammatory bowel disease--established concepts and future directions. Aliment Pharmacol Ther. 2012;36:324-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 70]  [Article Influence: 5.8]  [Reference Citation Analysis (1)]
34.  Leslie WD, Miller N, Rogala L, Bernstein CN. Vitamin D status and bone density in recently diagnosed inflammatory bowel disease: the Manitoba IBD Cohort Study. Am J Gastroenterol. 2008;103:1451-1459.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 99]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
35.  Cantorna MT, Munsick C, Bemiss C, Mahon BD. 1,25-Dihydroxycholecalciferol prevents and ameliorates symptoms of experimental murine inflammatory bowel disease. J Nutr. 2000;130:2648-2652.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Ananthakrishnan AN, Higuchi LM, Huang ES, Khalili H, Richter JM, Fuchs CS, Chan AT. Aspirin, nonsteroidal anti-inflammatory drug use, and risk for Crohn disease and ulcerative colitis: a cohort study. Ann Intern Med. 2012;156:350-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 161]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
37.  Shaw SY, Blanchard JF, Bernstein CN. Association between the use of antibiotics in the first year of life and pediatric inflammatory bowel disease. Am J Gastroenterol. 2010;105:2687-2692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 275]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
38.  Maunder RG. Evidence that stress contributes to inflammatory bowel disease: evaluation, synthesis, and future directions. Inflamm Bowel Dis. 2005;11:600-608.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Mawdsley JE, Rampton DS. Psychological stress in IBD: new insights into pathogenic and therapeutic implications. Gut. 2005;54:1481-1491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 387]  [Cited by in F6Publishing: 411]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
40.  Mawdsley JE, Rampton DS. The role of psychological stress in inflammatory bowel disease. Neuroimmunomodulation. 2006;13:327-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 79]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
41.  Bitton A, Dobkin PL, Edwardes MD, Sewitch MJ, Meddings JB, Rawal S, Cohen A, Vermeire S, Dufresne L, Franchimont D. Predicting relapse in Crohn’s disease: a biopsychosocial model. Gut. 2008;57:1386-1392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 173]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
42.  Cámara RJ, Schoepfer AM, Pittet V, Begré S, von Känel R. Mood and nonmood components of perceived stress and exacerbation of Crohn’s disease. Inflamm Bowel Dis. 2011;17:2358-2365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 37]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
43.  Goodhand JR, Greig FI, Koodun Y, McDermott A, Wahed M, Langmead L, Rampton DS. Do antidepressants influence the disease course in inflammatory bowel disease? A retrospective case-matched observational study. Inflamm Bowel Dis. 2012;18:1232-1239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
44.  Timmer A, Preiss JC, Motschall E, Rücker G, Jantschek G, Moser G. Psychological interventions for treatment of inflammatory bowel disease. Cochrane Database Syst Rev. 2011;CD006913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 81]  [Article Influence: 6.2]  [Reference Citation Analysis (1)]
45.  Thia KT, Loftus EV, Sandborn WJ, Yang SK. An update on the epidemiology of inflammatory bowel disease in Asia. Am J Gastroenterol. 2008;103:3167-3182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 371]  [Cited by in F6Publishing: 393]  [Article Influence: 24.6]  [Reference Citation Analysis (0)]
46.  Tan WC, Qiu D, Liam BL, Ng TP, Lee SH, van Eeden SF, D’Yachkova Y, Hogg JC. The human bone marrow response to acute air pollution caused by forest fires. Am J Respir Crit Care Med. 2000;161:1213-1217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 184]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
47.  van Eeden SF, Tan WC, Suwa T, Mukae H, Terashima T, Fujii T, Qui D, Vincent R, Hogg JC. Cytokines involved in the systemic inflammatory response induced by exposure to particulate matter air pollutants (PM(10)). Am J Respir Crit Care Med. 2001;164:826-830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 481]  [Cited by in F6Publishing: 480]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
48.  Kaplan GG, Hubbard J, Korzenik J, Sands BE, Panaccione R, Ghosh S, Wheeler AJ, Villeneuve PJ. The inflammatory bowel diseases and ambient air pollution: a novel association. Am J Gastroenterol. 2010;105:2412-2419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 164]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
49.  Ananthakrishnan AN, McGinley EL, Binion DG, Saeian K. Ambient air pollution correlates with hospitalizations for inflammatory bowel disease: an ecologic analysis. Inflamm Bowel Dis. 2011;17:1138-1145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 107]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
50.  Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7052]  [Cited by in F6Publishing: 7084]  [Article Influence: 506.0]  [Reference Citation Analysis (3)]
51.  Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora. Science. 2005;308:1635-1638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5700]  [Cited by in F6Publishing: 5230]  [Article Influence: 275.3]  [Reference Citation Analysis (2)]
52.  Joossens M, Huys G, Cnockaert M, De Preter V, Verbeke K, Rutgeerts P, Vandamme P, Vermeire S. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut. 2011;60:631-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 692]  [Cited by in F6Publishing: 703]  [Article Influence: 54.1]  [Reference Citation Analysis (0)]
53.  Andoh A, Imaeda H, Aomatsu T, Inatomi O, Bamba S, Sasaki M, Saito Y, Tsujikawa T, Fujiyama Y. Comparison of the fecal microbiota profiles between ulcerative colitis and Crohn’s disease using terminal restriction fragment length polymorphism analysis. J Gastroenterol. 2011;46:479-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 151]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
54.  Martinez C, Antolin M, Santos J, Torrejon A, Casellas F, Borruel N, Guarner F, Malagelada JR. Unstable composition of the fecal microbiota in ulcerative colitis during clinical remission. Am J Gastroenterol. 2008;103:643-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 138]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
55.  Ott SJ, Musfeldt M, Wenderoth DF, Hampe J, Brant O, Fölsch UR, Timmis KN, Schreiber S. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53:685-693.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Johansson ME, Phillipson M, Petersson J, Velcich A, Holm L, Hansson GC. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc Natl Acad Sci USA. 2008;105:15064-15069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1326]  [Cited by in F6Publishing: 1381]  [Article Influence: 86.3]  [Reference Citation Analysis (1)]
57.  Martinez-Medina M, Aldeguer X, Gonzalez-Huix F, Acero D, Garcia-Gil LJ. Abnormal microbiota composition in the ileocolonic mucosa of Crohn’s disease patients as revealed by polymerase chain reaction-denaturing gradient gel electrophoresis. Inflamm Bowel Dis. 2006;12:1136-1145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 190]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
58.  Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci USA. 2007;104:13780-13785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3075]  [Cited by in F6Publishing: 3154]  [Article Influence: 185.5]  [Reference Citation Analysis (1)]
59.  Ryan P, Kelly RG, Lee G, Collins JK, O’Sullivan GC, O’Connell J, Shanahan F. Bacterial DNA within granulomas of patients with Crohn’s disease--detection by laser capture microdissection and PCR. Am J Gastroenterol. 2004;99:1539-1543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 132]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
60.  Meconi S, Vercellone A, Levillain F, Payré B, Al Saati T, Capilla F, Desreumaux P, Darfeuille-Michaud A, Altare F. Adherent-invasive Escherichia coli isolated from Crohn’s disease patients induce granulomas in vitro. Cell Microbiol. 2007;9:1252-1261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 97]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
61.  Simpson KW, Dogan B, Rishniw M, Goldstein RE, Klaessig S, McDonough PL, German AJ, Yates RM, Russell DG, Johnson SE. Adherent and invasive Escherichia coli is associated with granulomatous colitis in boxer dogs. Infect Immun. 2006;74:4778-4792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 185]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
62.  Cobrin GM, Abreu MT. Defects in mucosal immunity leading to Crohn’s disease. Immunol Rev. 2005;206:277-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 145]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
63.  Targan SR, Karp LC. Defects in mucosal immunity leading to ulcerative colitis. Immunol Rev. 2005;206:296-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 131]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
64.  Geremia A, Jewell DP. The IL-23/IL-17 pathway in inflammatory bowel disease. Expert Rev Gastroenterol Hepatol. 2012;6:223-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 69]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
65.  Medzhitov R, Janeway C. Innate immunity. N Engl J Med. 2000;343:338-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1517]  [Cited by in F6Publishing: 1393]  [Article Influence: 58.0]  [Reference Citation Analysis (0)]
66.  Abreu MT, Fukata M, Arditi M. TLR signaling in the gut in health and disease. J Immunol. 2005;174:4453-4460.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Marks DJ, Harbord MW, MacAllister R, Rahman FZ, Young J, Al-Lazikani B, Lees W, Novelli M, Bloom S, Segal AW. Defective acute inflammation in Crohn’s disease: a clinical investigation. Lancet. 2006;367:668-678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 280]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
68.  Bonen DK, Ogura Y, Nicolae DL, Inohara N, Saab L, Tanabe T, Chen FF, Foster SJ, Duerr RH, Brant SR. Crohn’s disease-associated NOD2 variants share a signaling defect in response to lipopolysaccharide and peptidoglycan. Gastroenterology. 2003;124:140-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 301]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
69.  Wehkamp J, Harder J, Weichenthal M, Schwab M, Schäffeler E, Schlee M, Herrlinger KR, Stallmach A, Noack F, Fritz P. NOD2 (CARD15) mutations in Crohn’s disease are associated with diminished mucosal alpha-defensin expression. Gut. 2004;53:1658-1664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 578]  [Cited by in F6Publishing: 578]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
70.  Abraham C, Cho JH. Functional consequences of NOD2 (CARD15) mutations. Inflamm Bowel Dis. 2006;12:641-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 82]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
71.  Watanabe T, Kitani A, Murray PJ, Strober W. NOD2 is a negative regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat Immunol. 2004;5:800-808.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 615]  [Cited by in F6Publishing: 589]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
72.  Noguchi E, Homma Y, Kang X, Netea MG, Ma X. A Crohn’s disease-associated NOD2 mutation suppresses transcription of human IL10 by inhibiting activity of the nuclear ribonucleoprotein hnRNP-A1. Nat Immunol. 2009;10:471-479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 147]  [Article Influence: 9.8]  [Reference Citation Analysis (1)]
73.  Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov II, Littman DR, O’Shea JJ. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med. 2009;206:35-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 577]  [Cited by in F6Publishing: 578]  [Article Influence: 38.5]  [Reference Citation Analysis (0)]
74.  Kaser A, Blumberg RS. Autophagy, microbial sensing, endoplasmic reticulum stress, and epithelial function in inflammatory bowel disease. Gastroenterology. 2011;140:1738-1747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 110]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
75.  Salim SY, Söderholm JD. Importance of disrupted intestinal barrier in inflammatory bowel diseases. Inflamm Bowel Dis. 2011;17:362-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 357]  [Cited by in F6Publishing: 397]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
76.  Van der Sluis M, De Koning BA, De Bruijn AC, Velcich A, Meijerink JP, Van Goudoever JB, Büller HA, Dekker J, Van Seuningen I, Renes IB. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology. 2006;131:117-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1057]  [Cited by in F6Publishing: 1101]  [Article Influence: 61.2]  [Reference Citation Analysis (0)]
77.  Wehkamp J, Harder J, Weichenthal M, Mueller O, Herrlinger KR, Fellermann K, Schroeder JM, Stange EF. Inducible and constitutive beta-defensins are differentially expressed in Crohn’s disease and ulcerative colitis. Inflamm Bowel Dis. 2003;9:215-223.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 Cells. Annu Rev Immunol. 2009;27:485-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3754]  [Cited by in F6Publishing: 3603]  [Article Influence: 240.2]  [Reference Citation Analysis (0)]
79.  Breese E, Braegger CP, Corrigan CJ, Walker-Smith JA, MacDonald TT. Interleukin-2- and interferon-gamma-secreting T cells in normal and diseased human intestinal mucosa. Immunology. 1993;78:127-131.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Heller F, Florian P, Bojarski C, Richter J, Christ M, Hillenbrand B, Mankertz J, Gitter AH, Bürgel 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 806]  [Cited by in F6Publishing: 834]  [Article Influence: 43.9]  [Reference Citation Analysis (0)]
81.  Fuss IJ, Heller F, Boirivant M, Leon F, Yoshida M, Fichtner-Feigl S, Yang Z, Exley M, Kitani A, Blumberg RS. Nonclassical CD1d-restricted NK T cells that produce IL-13 characterize an atypical Th2 response in ulcerative colitis. J Clin Invest. 2004;113:1490-1497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 516]  [Cited by in F6Publishing: 553]  [Article Influence: 27.7]  [Reference Citation Analysis (0)]
82.  Di Sabatino A, Biancheri P, Rovedatti L, MacDonald TT, Corazza GR. New pathogenic paradigms in inflammatory bowel disease. Inflamm Bowel Dis. 2012;18:368-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
83.  Rovedatti L, Kudo T, Biancheri P, Sarra M, Knowles CH, Rampton DS, Corazza GR, Monteleone G, Di Sabatino A, Macdonald TT. Differential regulation of interleukin 17 and interferon gamma production in inflammatory bowel disease. Gut. 2009;58:1629-1636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 270]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
84.  Vainer B, Nielsen OH, Hendel J, Horn T, Kirman I. Colonic expression and synthesis of interleukin 13 and interleukin 15 in inflammatory bowel disease. Cytokine. 2000;12:1531-1536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 54]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
85.  Kadivar K, Ruchelli ED, Markowitz JE, Defelice ML, Strogatz ML, Kanzaria MM, Reddy KP, Baldassano RN, von Allmen D, Brown KA. Intestinal interleukin-13 in pediatric inflammatory bowel disease patients. Inflamm Bowel Dis. 2004;10:593-598.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Wilson MS, Ramalingam TR, Rivollier A, Shenderov K, Mentink-Kane MM, Madala SK, Cheever AW, Artis D, Kelsall BL, Wynn TA. Colitis and intestinal inflammation in IL10-/- mice results from IL-13Rα2-mediated attenuation of IL-13 activity. Gastroenterology. 2011;140:254-264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 69]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
87.  Bernardo D, Vallejo-Díez S, Mann ER, Al-Hassi HO, Martínez-Abad B, Montalvillo E, Tee CT, Murugananthan AU, Núñez H, Peake ST. IL-6 promotes immune responses in human ulcerative colitis and induces a skin-homing phenotype in the dendritic cells and Tcells they stimulate. Eur J Immunol. 2012;42:1337-1353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 53]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
88.  Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T, Levy DE, Leonard WJ, Littman DR. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol. 2007;8:967-974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1566]  [Cited by in F6Publishing: 1643]  [Article Influence: 96.6]  [Reference Citation Analysis (0)]
89.  Kobayashi T, Okamoto S, Hisamatsu T, Kamada N, Chinen H, Saito R, Kitazume MT, Nakazawa A, Sugita A, Koganei K. IL23 differentially regulates the Th1/Th17 balance in ulcerative colitis and Crohn’s disease. Gut. 2008;57:1682-1689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 408]  [Cited by in F6Publishing: 417]  [Article Influence: 26.1]  [Reference Citation Analysis (0)]
90.  Sugihara T, Kobori A, Imaeda H, Tsujikawa T, Amagase K, Takeuchi K, Fujiyama Y, Andoh A. The increased mucosal mRNA expressions of complement C3 and interleukin-17 in inflammatory bowel disease. Clin Exp Immunol. 2010;160:386-393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 90]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
91.  Monteleone G, Monteleone I, Fina D, Vavassori P, Del Vecchio Blanco G, Caruso R, Tersigni R, Alessandroni L, Biancone L, Naccari GC. Interleukin-21 enhances T-helper cell type I signaling and interferon-gamma production in Crohn’s disease. Gastroenterology. 2005;128:687-694.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Sarra M, Monteleone I, Stolfi C, Fantini MC, Sileri P, Sica G, Tersigni R, Macdonald TT, Pallone F, Monteleone G. Interferon-gamma-expressing cells are a major source of interleukin-21 in inflammatory bowel diseases. Inflamm Bowel Dis. 2010;16:1332-1339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 75]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
93.  Fina D, Sarra M, Fantini MC, Rizzo A, Caruso R, Caprioli F, Stolfi C, Cardolini I, Dottori M, Boirivant M. Regulation of gut inflammation and th17 cell response by interleukin-21. Gastroenterology. 2008;134:1038-1048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 209]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]