ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Immune and microbial mechanisms in the pathogenesis of inflammatory bowel disease

Immune and microbial mechanisms in the pathogenesis of inflammatory bowel disease
Literature review current through: Jan 2024.
This topic last updated: Jan 25, 2024.

INTRODUCTION — The immune response has long been implicated in the pathogenesis of inflammatory bowel disease (IBD), including both ulcerative colitis and Crohn disease. A vast body of literature has identified roles for both host and microbial factors in the pathogenesis of IBD, ultimately leading to inappropriate immune responses to microbes residing in the intestinal lumen [1].

This topic review will focus on our expanding understanding of the immune, microbial, and genetic factors involved in both the initiation and maintenance of IBD. The epidemiology, risk factors, clinical manifestations, diagnosis of IBD are discussed separately:

(See "Definitions, epidemiology, and risk factors for inflammatory bowel disease".)

(See "Clinical manifestations, diagnosis, and prognosis of Crohn disease in adults".)

(See "Clinical manifestations, diagnosis, and prognosis of ulcerative colitis in adults".)

The management of IBD (Crohn disease, ulcerative colitis) is discussed separately:

(See "Overview of the medical management of mild (low risk) Crohn disease in adults".)

(See "Medical management of moderate to severe Crohn disease in adults".)

(See "Medical management of low-risk adult patients with mild to moderate ulcerative colitis".)

(See "Management of the hospitalized adult patient with severe ulcerative colitis".)

(See "Management of moderate to severe ulcerative colitis in adults".)

THE MUCOSAL IMMUNE SYSTEM — In the process of carrying out the absorption of essential nutrients, the human intestine must discriminate between innocuous food antigens and infectious or toxic agents. To protect the host from the latter, the intestine relies upon an effective barrier and an innate and an acquired immune system (figure 1):

The effective barrier depends upon an intact intestinal epithelium, with its overlying surface mucus secreted by goblet cells, normal peristalsis, and the secretion of numerous protective factors (eg, antimicrobial proteins, growth factors). Studies have shown that, in addition to their role in producing mucus, goblet cells actively participate in the delivery of antigens from the intestinal lumen to cells in the intestinal lamina propria [2].

The innate immune system is comprised of myeloid-derived cells (neutrophils, monocytes, dendritic cells, and macrophages), natural killer cells, and innate lymphoid cells. These cells, in addition to epithelial cells, endothelial cells, and stromal cells, express pattern recognition receptors, which bind stereotypic microbial products. In combination, these cells provide the initial response to either pathogenic or commensal microorganisms.

The adaptive immune system is comprised primarily of B and T lymphocytes, which confer specific immunity. The acquired immune system is designed to respond to foreign antigens displayed by "professional" antigen-presenting cells (APCs) (eg, dendritic cells and macrophages) in association with molecules of the major histocompatibility complex (MHC).

Both humoral and cell-mediated mechanisms are involved in the acquired immune system. Humoral immunity is mediated by B cells within the gut, which secrete antibodies largely of the immunoglobulin (Ig) A class. (See "Selective IgA deficiency: Clinical manifestations, pathophysiology, and diagnosis".)

Cellular immunity is mediated by T lymphocytes, which can be functionally divided into CD4+ helper T cells (Th), CD8+ T cells (cytotoxic), and regulatory T cells. CD4+ T cells respond to processed antigen on professional APCs in association with MHC class II molecules. CD8+ T cells respond to processed antigen on all cell types in association with MHC class I molecules.

CD4+ Th cells can be further subdivided functionally into various T-cell subsets that are, in part, defined by the cytokines they produce. Examples of these include Th1 cells, Th2 cells, Th9 cells, Th17 cells, T follicular helper cells, tissue resident memory T cells, and regulatory T cells.

Th1 cells secrete predominantly interferon gamma (IFN-gamma), tumor necrosis factor (TNF)-alpha, and interleukin (IL)-2. IL-12 (consisting of the heterodimer p35/p40) is secreted by APCs and is critical for the generation of Th1 cells.

Th2 cells are induced by IL-4 and inhibited by IL-12. Th2 cells regulate B-cell differentiation by secreting predominantly IL-4, IL-5, and IL-13.

Th17 cells are a lineage of helper T cells that have a critical role in regulating the inflammatory process and responses at mucosal surfaces and may have a central role in immune-mediated diseases. Th17 cells secrete predominantly IL-17, IL-21, IL-22, and granulocyte-macrophage colony-stimulating factor (GM-CSF) [3,4]. These cells are somewhat heterogeneous, such that subsets of Th17 cells also can secrete IFN-gamma, TNF-alpha, or IL-10; the repertoire of combined cytokines help determine the behavior of Th17 cells at mucosal surfaces. The IL-12-related molecule, IL-23 (consisting of the heterodimer p19/p40), is important for maintenance of Th17 cells [3]. Mutations in the IL-23 receptor have been associated with susceptibility to inflammatory bowel disease (IBD) [5-7]. Furthermore, targeting of both the p19 subunit (unique to IL-23) and the p40 subunit (common to both IL-12 and IL-23) has demonstrated efficacy in clinical trials for treatment of IBD [8-10], thereby highlighting the importance of these cytokines in IBD pathogenesis; anti-IL-12p40 therapy has been approved for both Crohn disease and ulcerative colitis. Selective anti-IL23p19 therapy is being studied in ongoing clinical trials. (See "Medical management of moderate to severe Crohn disease in adults".)

In contrast, clinical trials blocking IL-17 in Crohn disease patients showed no efficacy, and, in fact, some patients developed worse disease [11]. Data have suggested that, in addition to pro-inflammatory activities, IL-17 has important roles in intestinal epithelial cell functions, which may have hindered clinical efficacy with anti-IL-17 targeted therapies [12,13].

Tissue resident CD4+ memory (TRM) T cells (defined as non-circulating, memory CD4 T cells) are retained locally in specific tissue sites and have the potential to rapidly express cytokines in response to antigen. In human intestine, these cells express CD69 and CD154, produce IFN-gamma and IL-17, and are broadly reactive to commensal organisms [14].

Regulatory T cells consist of various subsets including Th3, type 1 regulatory (Tr1), and CD4+CD25+ (mediated at least in part by the expression of the forkhead box P3 transcription factor [FOXP3]). They block or downregulate Th1, Th2, and Th17 cells either by production of specific cytokines (IL-10 and transforming growth factor (TGF)-beta) or via cell-cell contact mechanisms [15].

Immune cells, when located in the intestine, are collectively known as the gut-associated lymphoid tissue (GALT) and are distributed throughout the lamina propria, between epithelial cells, and in discrete lymphoid structures. Epithelial cells, "professional" APCs, and other leukocytes within the GALT also secrete a variety of soluble protein mediators (cytokines), which serve to regulate responses to foreign antigens. In addition, alterations in the intestinal microcirculation, trafficking molecules, and neuronal afferents can modify the composition and effector function of both immune and non-immune cells within the GALT.

KEY CONCEPTS EMERGING IN THE MUCOSAL IMMUNE SYSTEM

The immune response components must be properly balanced. As such, either too strong a response or an inadequate immune response to microbes in the intestinal lumen can ultimately result in intestinal inflammation.

The various arms of the intestinal immune system cooperate to defend against microbiota that gain access to intestinal tissues.

Given the high density of resident microbes in the intestinal lumen, the intestinal immune system has developed mechanisms to control excessive responses to these microbes. Cells in the intestinal immune system exhibit unique characteristics that are, in part, conditioned through the local intestinal environment. Examples of unique phenotypes of macrophages, dendritic cells, and T cells within intestinal tissues include:

Relative to the peripheral immune system, macrophages in the intestinal lamina propria do not produce high levels of proinflammatory cytokines upon exposure to microbial components but are more effective in clearing bacteria [16]. These combined characteristics likely contribute to eliminating bacteria in a manner that minimizes tissue injury. Intestinal macrophages also demonstrate anti-inflammatory characteristics [17,18].

Intestinal dendritic cells express high levels of retinoic acid-metabolizing enzymes. Retinoic acid, in turn, leads to expression of molecules that lead to enriched T-cell trafficking to intestinal tissues and regulatory T-cell function [19-24].

Mechanisms exist in intestinal tissues to eliminate T cells reactive to luminal bacteria [25] and to differentiate T cells into regulatory T-cell subsets.

While many distinct, differentiated intestinal immune cell subsets have been identified, a number of these same cells demonstrate plasticity. For example, Th17 cells can undergo transition to IL-10-producing regulatory T cells and Th2 cells, Th1 cells to Th2 cells, and Treg cells can transition to pathogenic IL-17-producing and IFN-gamma-producing T cells [26-29]. This has implications for therapy in IBD. On the one hand, it indicates that, when considering cell therapy with transfer of regulatory T-cell subsets for inhibiting inflammation, these cells may transition to pathogenic T cells rather than function in the protective manner intended. On the other hand, it provides possibilities for therapeutically inducing transition of pathogenic T cells to T cells with regulatory function. In addition, single-cell analysis technologies are allowing for more refined insight into the broad spectrum of cell subsets and how these cell subsets might transition during health and disease and with therapeutic interventions [30-35].

Conditions in the intestinal environment can influence the manner in which immune cells differentiate, with inflammation, secreted mediators, microbiota, metabolic products, dietary factors, and prior infectious exposures all able to influence these differentiation outcomes [26,27,36-44].

The enteric nervous system (ENS) can modulate the mucosal immune system and is comprised of both sympathetic and parasympathetic nerve fibers originating from the central nervous system. In addition, the intrinsic ENS is the network of neurons and glia that reside along the intestine and interact with the gastrointestinal tract by receiving and delivering modulating signals [30,45-47].

IMMUNE DYSREGULATION AND IBD — Many studies support the concept that inflammatory bowel disease (IBD) results from a dysregulated response by the mucosal immune system to the microbiota that reside within the intestinal lumen. This dysregulation can be both due to excessive immune reactivity and to inadequate immune responses to intestinal microbiota, highlighting the importance of a balanced immune response. The following describe alterations in the immune responses observed in IBD.

Dysregulation at the epithelial barrier — Alterations in intestinal mucus, high numbers of bacteria within mucus, and increased intestinal permeability have been associated with IBD [48-55]. Studies have revealed that mice with defects in epithelial barrier function can develop spontaneous colitis and/or demonstrate increased susceptibility to experimental models of colitis [56-59]. Epithelial cells can express some receptors also expressed on immune cells and can present antigens, similar to classical antigen-presenting cells [60-65]. Some studies have shown that abnormal antigen presentation by epithelial cells and/or interaction with intraepithelial lymphocytes is associated with IBD [66-69]. Moreover, abnormalities in the manner in which epithelial cells (in particular, highly secretory cells such as Paneth cells) handle unfolded intracellular proteins, a process that is associated with "stress" on the endoplasmic reticulum, has been associated with IBD [70-72]. Other abnormalities in Paneth cells have been identified in association with various immune pathway perturbations observed in IBD, which in some cases result in dysregulation in production of antimicrobial proteins [73,74].

Dysregulation in immune cells — Excessive immune cell recruitment and activation has been detected in multiple immune cell subsets. Myeloid cells with an "inflammatory" phenotype that produce increased levels of cytokines are present in the lamina propria of IBD patients [36,75-77]. Natural killer (NK) cells subsets are altered [78]. Mononuclear cells isolated from lesions of patients with IBD display numerous activation markers and cytokines [34,79]. T cells isolated from IBD mucosa may show increased proliferation and cytokine production to antigens in vitro, suggesting that they may respond abnormally to resident antigens [80]. Whether IBD results from an altered response by T cells to single or few antigens remains an unanswered question since some, but not all, studies have found a restricted T-cell receptor repertoire [52,81-86]. Innate lymphoid cells have also been found to be dysregulated in IBD [87].

The findings of increased numbers of mucosal and circulating B cells and plasma cells, autoantibodies (atypical perinuclear antineutrophil cytoplasmic antibodies [P-ANCA]), and increased antibodies to microbial components and cytokines (eg, anti-granulocyte-macrophage colony-stimulating factor) in patients with IBD suggest that abnormal B-cell regulation may also be involved in the pathogenesis of this disorder [53,54,88-94]. However, studies have not shown that antibody production is directly involved in the pathogenesis of IBD. As an example, the B-cell-depleting medication rituximab was not effective for treating ulcerative colitis [94]. Colonic-resident plasma cells are retained in the colonic mucosa of treated patients, which may contribute to the lack of efficacy [95].

Some inflammatory cells, such as neutrophils, found within diseased mucosa in IBD are not usually present in the lamina propria and therefore must be recruited from the blood vessels. This process, called "homing," requires several coordinated steps. Leukocytes "roll" along the endothelium, and chemokines secreted from the tissues activate adhesion molecules, thereby resulting in firm adhesion. Once adherent, leukocytes traverse the endothelium (a process called "diapedesis") [22,96]. Enhanced expression of adhesion molecules on leukocytes and endothelial cells, increased chemokines, and increased leukocyte binding to vascular endothelial cells in patients with IBD have been described [55,97-99]. There is an increasing focus on stromal cell contributions to immune cell recruitment into intestinal tissues and intestinal inflammation, in addition to their known role in fibrosis [100,101]. Therapeutic strategies to block mucosal homing of leukocytes are in use as described below, with additional strategies under investigation.

One approach to reduce lymphocyte trafficking to the intestine is with vedolizumab, an antibody directed specifically against the alpha-4 beta-7 integrin heterodimer mediating lymphocyte trafficking to the lamina propria [63,64,102,103]. Other approaches have included modulators of the S1P/S1PR1 axis such as ozanimod [104], which blocks the egress of lymphocytes from lymph nodes, thereby reducing the number of circulating lymphocytes. Defining the integrins, chemokines, and other pathways regulating the homing of various distinct cell subsets to intestinal tissues remains an area of active investigation.

Dysregulation in secreted mediators — In addition to the dysregulated cell populations, abnormal levels of immunoregulatory and inflammatory cytokines correlate with active IBD [65,68,105].

CD4+ T lymphocytes isolated from patients with Crohn disease secrete large amounts of interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha, cytokines with a marked proinflammatory effect, thereby implicating Th1 cells in the pathogenesis of IBD. These findings are consistent with observations in several mouse models of chronic colitis [106-111]. These studies have led to therapeutic strategies using anti-TNF-alpha antibodies for Crohn disease [112]. The secretion of interleukin (IL)-4, IL-5, and IL-13 by Th2 cells suppresses Th1-mediated responses, but also leads to the infiltration of eosinophils and can contribute to intestinal inflammation, shown in both human IBD and mouse colitis studies [69,72,113-116]. Cytokines associated with Th17 cells are also observed in the intestinal mucosa of IBD patients [117-124]. IL-23-dependent signaling and secretion of cytokines by Th17 cells may account, at least in part, for some of the aberrant cytokine secretion previously attributed to Th1 cells [4,125-131]. Importantly, Th17 cells are heterogenous in nature and the conditioning of Th17 cells by IL-23 leads to Th17 cells that are more pathogenic in nature [132-134]. Ongoing studies are investigating optimal approaches for targeting the IL-12/IL-23 axis given its critical role in the generation of Th1/Th17 cells [8-10].

As noted above, mice and humans also exhibit subsets of regulatory CD4+ T cells, including CD4+ CD25+ Tregs (forkhead box P3 transcription factor [FOXP3] expressing), Th3 and Tr1 cells, which can act as "suppressor" cells, in part through the secretion of inhibitory IL-10 and TGF-beta cytokines. While some regulatory cells are generated in the thymus, other "induced" or "adaptive" regulatory cells can be generated in situ in the gut-associated lymphoid tissue (GALT). CD8+ T cells also secrete IL-10 and TGF-beta, which suppress inflammatory responses. Although aberrant regulatory cell function has not yet been implicated in IBD, therapeutic strategies to induce or enhance regulatory T cells are under active investigation [15,135].

The TL1A (TNFSF15)/DR3 axis has also been implicated in Crohn disease [136-138]. TL1A is a TNF-like cytokine that is increased in Crohn disease and interacts with the death domain receptor (DR3). Several studies have implicated the TL1A/DR3 axis in animal models of colitis [139-141], with studies suggesting that TL1A can lead to induction of cytokines from Th1, Th2, and Th17 cells; NK cells; myeloid cells; and innate lymphoid cells [138,139,141-145]. As noted below, TNFSF15 is a well-established susceptibility locus in Crohn disease [146-151].

In addition to protein mediators, several other biological products have been implicated in IBD pathogenesis when found in excess. Examples include arachidonic acid and its byproducts and reactive oxygen and nitrogen products (table 1).

ROLE OF MICROBES — The distal ileum and colon contain high concentrations of microbes. Bidirectional host-microbe interactions in the intestine can be mutually beneficial or have adverse effects that contribute to intestinal inflammation. On the one hand, intestinal microbial colonization is essential to nutrition, energy metabolism, and proper "conditioning" of the intestinal and peripheral immune systems. On the other hand, the intestinal lumen can contain microbiota and microbial-derived factors that may promote inflammatory bowel disease (IBD) in the context of an underlying genetic immune defect.

The intestinal microbiota is acquired at birth but changes rapidly during the first year of life. In adults, each person's unique population of fecal microbiota is fairly stable over time, but fluctuations occur in response to environmental and developmental factors and in disease [152]. Yet another important measure beyond the composition of intestinal microbiota is the manner in which intestinal microbiota modulate microbial functions such as energy metabolism [153,154]. Studies in mice have shown that intestinal microbiota and the ability of the host to recognize and respond to these microbiota are important in the generation and optimal function of intestinal antimicrobial proteins, epithelial cells, innate lymphoid cells, natural killer T cells, macrophages, interleukin(IL)-17-producing T cells, intestinal and peripheral regulatory T cells (Tregs), and immunoglobulin A (IgA) [42,155-171].

At the same time, the host immune system and various host conditions (eg, obesity) can influence intestinal microbial communities [57,172-177]. The microbial community alterations can, in turn, modulate intestinal inflammatory outcomes [71,173,174]. Environmental factors can also influence intestinal microbiota, including dietary factors [177-180], helminth exposure [181], and antibiotic usage. Changes in these environmental factors have likely contributed to the increased prevalence of IBD during the past century. For example, the increased use of antibiotics in early childhood has been associated with an increased likelihood of developing IBD [182-185].

In patients with IBD, alterations in both the diversity and density of bacteria, in specific bacteria directly associated with the mucosa, and in the functions of the bacteria present (eg, oxidative stress, nutritional regulation) have been described [186-191]. These alterations in microbial communities have been identified even at the time of initial diagnosis in children [190]. Whether these microbial alterations are primary drivers of IBD or secondary to the underlying intestinal inflammation observed with IBD is not yet clear. Interestingly, select communities of bacteria from ulcerative colitis patients can induce Th17 responses when transferred into mice [192,193], thereby demonstrating that these bacteria can contribute to the dysregulated T cell responses observed in patients. No single agent has been shown to have a consistent relationship to IBD. The prevailing hypothesis at this time is that changes in communities of intestinal bacteria can contribute to the initiation and/or perpetuation of inflammation associated with IBD.

Consistent with this hypothesis is the observation of immune responses directed against particular microbial components. For example, antibodies to the DNA segment (I2) in affected mucosa is observed in 54 percent of patients with Crohn disease compared with 4 to 10 percent of controls [194], and antibodies to bacterial flagellin are observed in complicated Crohn disease patients [195,196]. In general, the immunoreactivity to microbial antigens correlates with more aggressive IBD [197,198]. It has been unclear if these increased antibodies are in response to the enhanced exposure to intestinal bacteria due to epithelial barrier perturbations and/or to increased immune reactivity of intestinal immune cells in the context of inflammation, or if these antibodies also contribute to the pathogenesis of IBD.

The role of microbiota is also being examined in patients who have undergone an ileal pouch-anal anastomosis surgery for ulcerative colitis. Correlations have been identified between microbiota, mucosal gene expression, and clinical outcomes (eg, pouchitis) [199-201]. (See "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Etiopathogenesis'.)

In contrast to our limited understanding in human IBD, the importance of microbes in the protection, induction, and/or maintenance of disease has been demonstrated more clearly in murine models of IBD. As an example, various rodent models of colitis develop intestinal inflammation in the presence of a normal microflora, but not in germ-free conditions [197,202,203]. In some cases, bacteria from mice with colitis have been shown to promote intestinal inflammation when transferred from one animal to another [173,174]. In other cases, select bacteria result in inflammation specifically when combined with an underlying genetic defect [204]. There are numerous efforts underway to better identify these inflammation-promoting bacteria. As one such example, IgA-coated intestinal bacteria preferentially drive intestinal inflammation [205]. Moreover, certain combinations of bacteria (eg, probiotics, Clostridia) can mediate protection from inflammation through such mechanisms as inducing Treg cells [206] or modulating growth factors that promote epithelial restitution [207]. Beyond their ability to regulate colitis, studies in mice have shown that intestinal microbiota can modulate the development of other diseases such as metabolic syndrome, diabetes, and autism [208-210].

Additional analyses have revealed that specific microbial components, such as the polysaccharide A component of the resident intestinal microbe Bacteroides fragilis, and microbial-derived products, such as short-chain fatty acids, are mechanisms through which intestinal microbiota can modulate both intestinal immune system development and intestinal inflammation [211-216].

The microbial alterations in IBD have extended to those beyond bacteria. For example, changes in intestinal viruses have also been described in patients with IBD [217]. Further supporting contributions from intestinal viral communities are studies that have shown that in mice with alterations in genes associated with IBD, colonization with norovirus leads to intestinal abnormalities [218]. Fungi have also been shown to modulate intestinal inflammation in mouse studies [219], and studies are ongoing to determine how fungi may be altered in human IBD [220].

Genetic studies have identified susceptibility loci that regulate innate responses to the microbial flora. They provide further evidence for the role of microbes in the pathogenesis of IBD. (See 'Genetic susceptibility' below.)

GENETIC SUSCEPTIBILITY — Over 240 distinct susceptibility loci for inflammatory bowel disease (IBD) have been identified, with some contributing to unique risk and others contributing to combined risk for Crohn disease and ulcerative colitis [146,151,221,222]. In fact, approximately 70 percent of the genes are shared between Crohn disease and ulcerative colitis, thereby highlighting significant genetic overlap in these disease entities [146]. Many of the variants are located in intronic regions of the gene, such that they do not alter the amino acids within the proteins encoded by these genes, but rather likely modulate the expression of the proteins. This expression modulation is generally mild in degree, consistent with the fact that each of the susceptibility loci confers a slight increase in disease risk. A subset of the pathways associated with IBD susceptibility also contribute to very early-onset IBD when associated with genetic variants that result in more dramatic disruption of the same pathway [223]. A high percentage of the IBD-associated pathways are associated with other immune-mediated diseases and/or confer risk for immunodeficiencies and for susceptibility to mycobacterial diseases [146].

The IBD genetic discoveries have provided insight into pathways that contribute to IBD pathogenesis and that might ultimately be targeted in IBD. Studies are ongoing to define the specific genes within the identified susceptibility loci, the functional consequences of the variants within these genes, and the specific manner in which the associations are contributing to IBD pathogenesis. In many cases, the proteins encoded by the genes are expressed in multiple different cell subsets and thereby contribute to disease through distinct and cooperative mechanisms in these various cells. The following are illustrative of the range of findings:

Innate immune pathways – The first susceptibility gene identified in association with Crohn disease encodes for the protein NOD2 [224,225]. Of the genes identified to date, NOD2 confers the greatest risk of developing Crohn disease. The wild-type NOD2 protein responds to bacterial peptidoglycan, which then activates signaling pathways that lead to cytokine production and clearance of bacteria [226]. The variants associated with Crohn disease result in a loss of these various functional outcomes. Mice deficient in NOD2 are susceptible to enteric pathogens and have changes in their intestinal luminal bacteria [227,228]. The loss-of-function in NOD2 also leads to an impaired ability to instruct cells to ultimately downregulate inflammatory pathways in conditions that simulate the intestinal environment [229,230]. NOD2 is expressed in a variety of cell types, including intestinal epithelial cells, macrophages, dendritic cells, endothelial cells, and stromal cells [226], thereby highlighting the many different levels at which NOD2 contributes to intestinal immune function. Further, experimental model systems have demonstrated that the adverse consequences of impaired NOD2 function can be more severe when combined with additional genetic or environmental triggers [54,231].

An individual inheriting one mutant NOD2 allele has a 1.5- to 3.7-fold increase in risk of developing Crohn disease, while an individual inheriting two mutant NOD2 alleles has a 17- to 40-fold increase in risk [232-235]. Penetrance of Crohn disease in carriers of one mutant NOD2 allele is 0.54 percent and two mutant NOD2 alleles is 4.9 percent, compared with 0.18 percent for wild-type NOD2 carriers [235]. In contrast to the risk conferred to European populations, studies including populations from East Asia have not found an association between the common NOD2 variants and Crohn disease [151,236,237].

The variants in NOD2 confer susceptibility to developing Crohn disease at an earlier age, to ileal and fibrostenosing Crohn disease, and to worse outcomes following ileal pouch-anal anastomosis for ulcerative colitis [232,233,238-242]. A meta-analysis that included 49 studies with 8893 Crohn disease patients found that patients with NOD2 mutations were at increased risk for complicated disease (relative risk [RR] 1.17, 95% CI 1.10-1.24) and surgery (RR 1.58, 95% CI 1.38-1.80) compared with those without mutations [243].

In addition to NOD2, a number of other IBD-associated gene variants have been shown to modulate innate pathways. Interestingly, innate cells from healthy individuals demonstrate dramatic variation in responses upon exposure to microbial products. This variation between individuals is in part genetically determined and contributes to the balance in susceptibility to infectious diseases and to immune-mediated diseases. Various studies have examined innate cells from individuals carrying IBD risk variants. In some cases, the common risk variants that confer IBD risk have been shown to increase innate pathway responses (eg, IRF5, TNFSF15, TPL2, JAK2, STAT1/STAT4, STAT3/STAT5), thereby increasing cytokine production in the presence of microbial products [144,244-252]. In other cases, the variants decrease innate pathway responses (eg, ICOSLG, INAVA, LACC1, RNF186) [253-260], which may ultimately lead to less effective bacterial clearance mechanisms. These genetic findings once again highlight the need to balance bacterial clearance with tightly controlled immune responses in the intestinal environment.

Microbial clearance pathways – Defects in autophagy Related 16-Like 1 (ATG16L1) and immunity-related GTPase M (IRGM) proteins have been associated with Crohn disease [147,148,261-267]. These proteins are involved in promoting autophagy, a process that contributes to elimination of intracellular pathogens. Consequences of the mutation in ATG16L1 associated with Crohn disease include changes in Paneth cells and goblet cells, a decreased ability to clear bacteria, and an increased secretion of cytokines in studies in both mice and IBD patients [73,74,268,269]. In addition to loss-of-function variants in proteins promoting autophagy, gain-of-function variants in MTMR3, an inhibitor of autophagy, are associated with Crohn disease [221]. This, in turn, also leads to impaired autophagy [270].

Variants in the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase genes that regulate the generation of reactive oxygen species, which contribute to microbial clearance, are observed in both very early-onset IBD [271,272] and adult-onset IBD [146]. As above, there are also IBD-associated genetic variants leading an impaired induction of the microbial pathways observed with the activation of pattern recognition receptors. Given the numerous examples in mouse studies wherein host immune function can modulate microbiota within the intestinal lumen, studies are ongoing to determine how various IBD risk variants modulate human intestinal microbiota [273]. One such example is the alterations in colonic microbiota at both the compositional and functional levels observed in FUT2 IBD risk carriers [274].

Cytokine pathways – Consistent with the very important role of cytokines in mediating the inflammation observed in IBD, a variety of IBD genetic associations have been identified in pathways both leading to the production of and the response to cytokines. Below are examples of these associations:

Th17/IL-23 pathway – Certain noncoding variants of the IL-23 receptor have been associated with both Crohn disease and ulcerative colitis [5-7,146,148]. By contrast, an uncommon coding variant confers an approximately two- to threefold protection against the risk of IBD. As noted above, IL-23 is a proinflammatory cytokine that is important for the stabilization of Th17 cells, and in particular for conditioning more pathogenic Th17 cells. Individuals carrying the protective IL23R risk variant have less circulating Th17 cells, and their T cells signal less in response to IL-23 [275-277]. Interestingly, loci containing STAT3, JAK2, and TYK2 (signaling molecules downstream of IL-23-dependent signaling); IL12B (the p40 subunit that is contained within both IL-12 and IL-23); and RORC (a transcription factor associated with Th17 cells) are associated with IBD risk [146-148,263], thereby highlighting the convergence of multiple genes in the same pathway. Colonic tissues from STAT3 risk-variant IBD patients have increased expression of cytokines and chemokines [278], and myeloid cells from STAT3 IBD risk-variant individuals also demonstrate increased cytokines in response to microbial components [252].

IL-10 pathway – IL-10 functions to inhibit a broad array of inflammatory outcomes [279-283]. Interestingly, common IL-10 variants are associated with the common form of IBD [146]; rare loss-of-function IL-10 and IL-10 receptor mutations are associated with very early-onset IBD [284-286]; and IL-10 and IL-10 receptor deficient mice develop spontaneous colitis [106,111]. These provide examples of both common and rare variants in human disease, as well as convergence of human and mouse models in a single critical regulatory pathway. Interestingly, one of the inflammatory outcomes which IL-10 inhibits is the IL-1 beta pathway. As such, very early-onset IBD patients with mutations leading to IL-10 deficiency have demonstrated improvement upon therapy with IL-1 beta pathway blockade [287].

TNFSF15 pathway – Variants associated with IBD [146,148,288] that result in increased TNFSF15 (TL1A) expression enhance innate cell responses to microbial products [144,250]. In addition to European-ancestry IBD, TNFSF15 variants are strongly associated with IBD in Asian-ancestry individuals [149-151], providing an important example of an IBD risk gene that is shared across populations. As above, TNFSF15 is increased in intestinal tissues from IBD patients, and several studies have implicated the TNFSF15/DR3 axis in animal models of colitis and in the fibrosis observed in these models [139-141,289].

IL18R/IL1R pathways – Genetic variants in a region that includes a cluster of cytokine receptors from the IL1 receptor and IL18 receptor family are associated with both Crohn disease and ulcerative colitis [146]. The IBD risk variants lead to decreased expression of IL18RAP, IL18R1, and IL1R1 on innate cells. This decreased expression leads to decreased responses to IL-18 and IL-1beta and to the IL-18 and IL-1beta amplification of signaling and cytokines observed upon exposure to microbial products [290].

Adaptive immune pathways – The innate immune system is continuously interacting with the adaptive immune system. As a result, a number of the IBD gene variants that regulate innate immune responses will likely also modulate adaptive immune responses through both direct and indirect mechanisms. Examples of IBD risk variants that directly regulate adaptive immune pathways include those in IRF5 [291], IL23R, PTPN2 and PTPN22. Crohn disease patients who carry the IBD risk PTPN2 variants show increased Th1 and Th17 cell but decreased Treg markers in serum and intestinal tissues [292].

Epithelial pathways – Several susceptibility loci include genes that regulate epithelial cell functions. For example, X-box binding protein 1 (XBP1) and ORMDL3 are proteins that regulate the unfolded protein response, which is a critical cellular process by which highly secretory cells like epithelial cells handle endoplasmic reticulum stress. This process also regulates the process of autophagy described above [70]. Additional IBD susceptibility genes that can regulate outcomes in epithelial cells include ATG16L1, PTPN2, INAVA, RNF186, and A20; in some cases, these outcomes are particularly observed when alterations in these IBD-associated genes are combined with environmental triggers (eg, smoking, viruses) or other IBD-associated gene alterations [74,218,293-297].  

LESSONS FROM ANIMAL MODELS OF IBD — The availability of various animal models of inflammatory bowel disease (IBD) has facilitated investigation into the mechanisms essential for maintaining a well-balanced intestinal immune system, the underlying defects in the gut-associated lymphoid tissue (GALT) that can induce inflammation, and the essential factors required for the maintenance of disease. Numerous murine strains develop intestinal inflammation as a result of genetic manipulation or "knock out" of genes that affect immune function within the mucosal immune system [56,106,107,261,298-308]. The elimination of these genes from selection cell subsets has allowed for further refining of our understanding of the differential regulation of pathways in distinct cells subsets. This more refined understanding might ultimately allow for selectively targeting implicated IBD-associated pathways in pertinent cell subsets, while leaving the pathway(s) intact in cell subsets where maintained function might be important.  

Major themes have emerged from these animal models, including:

The absence or impaired function of diverse proteins (or cell types) involved in regulating the innate or adaptive mucosal immune system and epithelial cell function can result in mucosal inflammation or can enhance susceptibility to experimental models of colitis.

The presence of luminal bacteria is generally required for development of colitis.

There is continuous cross-talk between the host and intestinal microbes, which ultimately influences both protection and susceptibility to intestinal inflammation.

Importantly, these animal models provide a foundation for future investigation into the mechanisms responsible for IBD. They further provide a means for developing and testing therapeutic interventions for IBD, which will hopefully result in the future availability of novel therapeutic regimens.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Ulcerative colitis in adults" and "Society guideline links: Crohn disease in adults".)

SUMMARY AND RECOMMENDATIONS

A vast body of evidence indicates that the pathogenesis of inflammatory bowel disease (IBD) arises from dysregulated immune responses to luminal bacteria and/or their products. (See 'Immune dysregulation and IBD' above.)

Multiple lines of evidence suggest that patients with IBD have alterations in both the composition and function of intestinal microbiota. (See 'Role of microbes' above.)

Numerous genetic variations have been implicated in the pathogenesis of IBD, which provide insight into pathways important in regulating intestinal inflammation. (See 'Genetic susceptibility' above.)

Therapies targeting immune dysregulation, alterations in intestinal microbiota, and pathways identified through genetic studies are in active development. (See 'Immune dysregulation and IBD' above and 'Role of microbes' above and 'Genetic susceptibility' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Daniel Podolsky, MD, who contributed to an earlier version of this topic review, and E Richard Stiehm, MD, who contributed as a Section Editor to an earlier version of this topic review.

The UpToDate editorial staff also acknowledges Paul Rutgeerts, MD (deceased), who contributed as a section editor for UpToDate in Gastroenterology.

  1. Cho JH. The genetics and immunopathogenesis of inflammatory bowel disease. Nat Rev Immunol 2008; 8:458.
  2. McDole JR, Wheeler LW, McDonald KG, et al. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature 2012; 483:345.
  3. Gaffen SL, Jain R, Garg AV, Cua DJ. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol 2014; 14:585.
  4. Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol 2007; 25:821.
  5. Duerr RH, Taylor KD, Brant SR, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 2006; 314:1461.
  6. Tremelling M, Cummings F, Fisher SA, et al. IL23R variation determines susceptibility but not disease phenotype in inflammatory bowel disease. Gastroenterology 2007; 132:1657.
  7. Baldassano RN, Bradfield JP, Monos DS, et al. Association of variants of the interleukin-23 receptor gene with susceptibility to pediatric Crohn's disease. Clin Gastroenterol Hepatol 2007; 5:972.
  8. Feagan BG, Sandborn WJ, Gasink C, et al. Ustekinumab as Induction and Maintenance Therapy for Crohn's Disease. N Engl J Med 2016; 375:1946.
  9. Sands BE, Chen J, Feagan BG, et al. Efficacy and Safety of MEDI2070, an Antibody Against Interleukin 23, in Patients With Moderate to Severe Crohn's Disease: A Phase 2a Study. Gastroenterology 2017; 153:77.
  10. Feagan BG, Sandborn WJ, D'Haens G, et al. Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn's disease: a randomised, double-blind, placebo-controlled phase 2 study. Lancet 2017; 389:1699.
  11. Hueber W, Sands BE, Lewitzky S, et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut 2012; 61:1693.
  12. Lee JS, Tato CM, Joyce-Shaikh B, et al. Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability. Immunity 2015; 43:727.
  13. Maxwell JR, Zhang Y, Brown WA, et al. Differential Roles for Interleukin-23 and Interleukin-17 in Intestinal Immunoregulation. Immunity 2015; 43:739.
  14. Hegazy AN, West NR, Stubbington MJT, et al. Circulating and Tissue-Resident CD4+ T Cells With Reactivity to Intestinal Microbiota Are Abundant in Healthy Individuals and Function Is Altered During Inflammation. Gastroenterology 2017; 153:1320.
  15. Boden EK, Snapper SB. Regulatory T cells in inflammatory bowel disease. Curr Opin Gastroenterol 2008; 24:733.
  16. Smythies LE, Sellers M, Clements RH, et al. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Invest 2005; 115:66.
  17. Denning TL, Wang YC, Patel SR, et al. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol 2007; 8:1086.
  18. Manicassamy S, Reizis B, Ravindran R, et al. Activation of beta-catenin in dendritic cells regulates immunity versus tolerance in the intestine. Science 2010; 329:849.
  19. Mora JR, Bono MR, Manjunath N, et al. Selective imprinting of gut-homing T cells by Peyer's patch dendritic cells. Nature 2003; 424:88.
  20. Iwata M, Hirakiyama A, Eshima Y, et al. Retinoic acid imprints gut-homing specificity on T cells. Immunity 2004; 21:527.
  21. Sigmundsdottir H, Butcher EC. Environmental cues, dendritic cells and the programming of tissue-selective lymphocyte trafficking. Nat Immunol 2008; 9:981.
  22. Villablanca EJ, Cassani B, von Andrian UH, Mora JR. Blocking lymphocyte localization to the gastrointestinal mucosa as a therapeutic strategy for inflammatory bowel diseases. Gastroenterology 2011; 140:1776.
  23. Mucida D, Park Y, Kim G, et al. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science 2007; 317:256.
  24. Sun CM, Hall JA, Blank RB, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med 2007; 204:1775.
  25. Hepworth MR, Fung TC, Masur SH, et al. Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4⁺ T cells. Science 2015; 348:1031.
  26. Oldenhove G, Bouladoux N, Wohlfert EA, et al. Decrease of Foxp3+ Treg cell number and acquisition of effector cell phenotype during lethal infection. Immunity 2009; 31:772.
  27. Esplugues E, Huber S, Gagliani N, et al. Control of TH17 cells occurs in the small intestine. Nature 2011; 475:514.
  28. Panzer M, Sitte S, Wirth S, et al. Rapid in vivo conversion of effector T cells into Th2 cells during helminth infection. J Immunol 2012; 188:615.
  29. Gagliani N, Amezcua Vesely MC, Iseppon A, et al. Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation. Nature 2015; 523:221.
  30. Elmentaite R, Kumasaka N, Roberts K, et al. Cells of the human intestinal tract mapped across space and time. Nature 2021; 597:250.
  31. Fawkner-Corbett D, Antanaviciute A, Parikh K, et al. Spatiotemporal analysis of human intestinal development at single-cell resolution. Cell 2021; 184:810.
  32. Parikh K, Antanaviciute A, Fawkner-Corbett D, et al. Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature 2019; 567:49.
  33. Corridoni D, Antanaviciute A, Gupta T, et al. Single-cell atlas of colonic CD8+ T cells in ulcerative colitis. Nat Med 2020; 26:1480.
  34. Mitsialis V, Wall S, Liu P, et al. Single-Cell Analyses of Colon and Blood Reveal Distinct Immune Cell Signatures of Ulcerative Colitis and Crohn's Disease. Gastroenterology 2020; 159:591.
  35. Martin JC, Chang C, Boschetti G, et al. Single-Cell Analysis of Crohn's Disease Lesions Identifies a Pathogenic Cellular Module Associated with Resistance to Anti-TNF Therapy. Cell 2019; 178:1493.
  36. Bain CC, Scott CL, Uronen-Hansson H, et al. Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal Immunol 2013; 6:498.
  37. Zielinski CE, Mele F, Aschenbrenner D, et al. Pathogen-induced human TH17 cells produce IFN-γ or IL-10 and are regulated by IL-1β. Nature 2012; 484:514.
  38. Hand TW, Dos Santos LM, Bouladoux N, et al. Acute gastrointestinal infection induces long-lived microbiota-specific T cell responses. Science 2012; 337:1553.
  39. Rivollier A, He J, Kole A, et al. Inflammation switches the differentiation program of Ly6Chi monocytes from antiinflammatory macrophages to inflammatory dendritic cells in the colon. J Exp Med 2012; 209:139.
  40. Grainger JR, Wohlfert EA, Fuss IJ, et al. Inflammatory monocytes regulate pathologic responses to commensals during acute gastrointestinal infection. Nat Med 2013; 19:713.
  41. Schiering C, Krausgruber T, Chomka A, et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 2014; 513:564.
  42. Mortha A, Chudnovskiy A, Hashimoto D, et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 2014; 343:1249288.
  43. Spencer SP, Wilhelm C, Yang Q, et al. Adaptation of innate lymphoid cells to a micronutrient deficiency promotes type 2 barrier immunity. Science 2014; 343:432.
  44. Moriyama S, Brestoff JR, Flamar AL, et al. β2-adrenergic receptor-mediated negative regulation of group 2 innate lymphoid cell responses. Science 2018; 359:1056.
  45. Yoo BB, Mazmanian SK. The Enteric Network: Interactions between the Immune and Nervous Systems of the Gut. Immunity 2017; 46:910.
  46. Drokhlyansky E, Smillie CS, Van Wittenberghe N, et al. The Human and Mouse Enteric Nervous System at Single-Cell Resolution. Cell 2020; 182:1606.
  47. Teratani T, Mikami Y, Nakamoto N, et al. The liver-brain-gut neural arc maintains the Treg cell niche in the gut. Nature 2020; 585:591.
  48. Podolsky DK, Isselbacher KJ. Composition of human colonic mucin. Selective alteration in inflammatory bowel disease. J Clin Invest 1983; 72:142.
  49. Schultsz C, Van Den Berg FM, Ten Kate FW, et al. The intestinal mucus layer from patients with inflammatory bowel disease harbors high numbers of bacteria compared with controls. Gastroenterology 1999; 117:1089.
  50. Hollander D. Crohn's disease--a permeability disorder of the tight junction? Gut 1988; 29:1621.
  51. Wyatt J, Vogelsang H, Hübl W, et al. Intestinal permeability and the prediction of relapse in Crohn's disease. Lancet 1993; 341:1437.
  52. Camus M, Esses S, Pariente B, et al. Oligoclonal expansions of mucosal T cells in Crohn's disease predominate in NKG2D-expressing CD4 T cells. Mucosal Immunol 2014; 7:325.
  53. Uo M, Hisamatsu T, Miyoshi J, et al. Mucosal CXCR4+ IgG plasma cells contribute to the pathogenesis of human ulcerative colitis through FcγR-mediated CD14 macrophage activation. Gut 2013; 62:1734.
  54. Han X, Uchida K, Jurickova I, et al. Granulocyte-macrophage colony-stimulating factor autoantibodies in murine ileitis and progressive ileal Crohn's disease. Gastroenterology 2009; 136:1261.
  55. Brazil JC, Louis NA, Parkos CA. The role of polymorphonuclear leukocyte trafficking in the perpetuation of inflammation during inflammatory bowel disease. Inflamm Bowel Dis 2013; 19:1556.
  56. Hermiston ML, Gordon JI. Inflammatory bowel disease and adenomas in mice expressing a dominant negative N-cadherin. Science 1995; 270:1203.
  57. Salzman NH, Hung K, Haribhai D, et al. Enteric defensins are essential regulators of intestinal microbial ecology. Nat Immunol 2010; 11:76.
  58. Su L, Shen L, Clayburgh DR, et al. Targeted epithelial tight junction dysfunction causes immune activation and contributes to development of experimental colitis. Gastroenterology 2009; 136:551.
  59. Turner JR. Molecular basis of epithelial barrier regulation: from basic mechanisms to clinical application. Am J Pathol 2006; 169:1901.
  60. Mayer L, Shlien R. Evidence for function of Ia molecules on gut epithelial cells in man. J Exp Med 1987; 166:1471.
  61. Reinecker HC, Podolsky DK. Human intestinal epithelial cells express functional cytokine receptors sharing the common gamma c chain of the interleukin 2 receptor. Proc Natl Acad Sci U S A 1995; 92:8353.
  62. Reinecker HC, MacDermott RP, Mirau S, et al. Intestinal epithelial cells both express and respond to interleukin 15. Gastroenterology 1996; 111:1706.
  63. Sandborn WJ, Feagan BG, Rutgeerts P, et al. Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med 2013; 369:711.
  64. Sands BE, Feagan BG, Rutgeerts P, et al. Effects of vedolizumab induction therapy for patients with Crohn's disease in whom tumor necrosis factor antagonist treatment failed. Gastroenterology 2014; 147:618.
  65. Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunol 2014; 14:329.
  66. Selby WS, Janossy G, Mason DY, Jewell DP. Expression of HLA-DR antigens by colonic epithelium in inflammatory bowel disease. Clin Exp Immunol 1983; 53:614.
  67. Mayer L, Eisenhardt D, Salomon P, et al. Expression of class II molecules on intestinal epithelial cells in humans. Differences between normal and inflammatory bowel disease. Gastroenterology 1991; 100:3.
  68. Strober W, Fuss IJ. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology 2011; 140:1756.
  69. Heller F, Fuss IJ, Nieuwenhuis EE, et al. Oxazolone colitis, a Th2 colitis model resembling ulcerative colitis, is mediated by IL-13-producing NK-T cells. Immunity 2002; 17:629.
  70. Kaser A, Martínez-Naves E, Blumberg RS. Endoplasmic reticulum stress: implications for inflammatory bowel disease pathogenesis. Curr Opin Gastroenterol 2010; 26:318.
  71. Elson CO, Cong Y, McCracken VJ, et al. Experimental models of inflammatory bowel disease reveal innate, adaptive, and regulatory mechanisms of host dialogue with the microbiota. Immunol Rev 2005; 206:260.
  72. Forbes E, Murase T, Yang M, et al. Immunopathogenesis of experimental ulcerative colitis is mediated by eosinophil peroxidase. J Immunol 2004; 172:5664.
  73. Cadwell K, Liu JY, Brown SL, et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature 2008; 456:259.
  74. VanDussen KL, Liu TC, Li D, et al. Genetic variants synthesize to produce paneth cell phenotypes that define subtypes of Crohn's disease. Gastroenterology 2014; 146:200.
  75. Reinecker HC, Steffen M, Witthoeft T, et al. Enhanced secretion of tumour necrosis factor-alpha, IL-6, and IL-1 beta by isolated lamina propria mononuclear cells from patients with ulcerative colitis and Crohn's disease. Clin Exp Immunol 1993; 94:174.
  76. Reimund JM, Wittersheim C, Dumont S, et al. Increased production of tumour necrosis factor-alpha interleukin-1 beta, and interleukin-6 by morphologically normal intestinal biopsies from patients with Crohn's disease. Gut 1996; 39:684.
  77. Kamada N, Hisamatsu T, Okamoto S, et al. Unique CD14 intestinal macrophages contribute to the pathogenesis of Crohn disease via IL-23/IFN-gamma axis. J Clin Invest 2008; 118:2269.
  78. Takayama T, Kamada N, Chinen H, et al. Imbalance of NKp44(+)NKp46(-) and NKp44(-)NKp46(+) natural killer cells in the intestinal mucosa of patients with Crohn's disease. Gastroenterology 2010; 139:882.
  79. Matsuura T, West GA, Youngman KR, et al. Immune activation genes in inflammatory bowel disease. Gastroenterology 1993; 104:448.
  80. Fiocchi C, Battisto JR, Farmer RG. Studies on isolated gut mucosal lymphocytes in inflammatory bowel disease. Detection of activated T cells and enhanced proliferation to Staphylococcus aureus and lipopolysaccharides. Dig Dis Sci 1981; 26:728.
  81. Chott A, Probert CS, Gross GG, et al. A common TCR beta-chain expressed by CD8+ intestinal mucosa T cells in ulcerative colitis. J Immunol 1996; 156:3024.
  82. Kaulfersch W, Fiocchi C, Waldmann TA. Polyclonal nature of the intestinal mucosal lymphocyte populations in inflammatory bowel disease. A molecular genetic evaluation of the immunoglobulin and T-cell antigen receptors. Gastroenterology 1988; 95:364.
  83. Posnett DN, Schmelkin I, Burton DA, et al. T cell antigen receptor V gene usage. Increases in V beta 8+ T cells in Crohn's disease. J Clin Invest 1990; 85:1770.
  84. Spencer J, Choy MY, MacDonald TT. T cell receptor V beta expression by mucosal T cells. J Clin Pathol 1991; 44:915.
  85. Duchmann R, Kaiser I, Hermann E, et al. Tolerance exists towards resident intestinal flora but is broken in active inflammatory bowel disease (IBD). Clin Exp Immunol 1995; 102:448.
  86. Gulwani-Akolkar B, Akolkar PN, Minassian A, et al. Selective expansion of specific T cell receptors in the inflamed colon of Crohn's disease. J Clin Invest 1996; 98:1344.
  87. Panda SK, Colonna M. Innate Lymphoid Cells in Mucosal Immunity. Front Immunol 2019; 10:861.
  88. BROBERGER O, PERLMANN P. Autoantibodies in human ulcerative colitis. J Exp Med 1959; 110:657.
  89. Takahashi F, Das KM. Isolation and characterization of a colonic autoantigen specifically recognized by colon tissue-bound immunoglobulin G from idiopathic ulcerative colitis. J Clin Invest 1985; 76:311.
  90. Das KM, Dasgupta A, Mandal A, Geng X. Autoimmunity to cytoskeletal protein tropomyosin. A clue to the pathogenetic mechanism for ulcerative colitis. J Immunol 1993; 150:2487.
  91. Saxon A, Shanahan F, Landers C, et al. A distinct subset of antineutrophil cytoplasmic antibodies is associated with inflammatory bowel disease. J Allergy Clin Immunol 1990; 86:202.
  92. Ruemmele FM, Targan SR, Levy G, et al. Diagnostic accuracy of serological assays in pediatric inflammatory bowel disease. Gastroenterology 1998; 115:822.
  93. Chapman RW, Cottone M, Selby WS, et al. Serum autoantibodies, ulcerative colitis and primary sclerosing cholangitis. Gut 1986; 27:86.
  94. Leiper K, Martin K, Ellis A, et al. Randomised placebo-controlled trial of rituximab (anti-CD20) in active ulcerative colitis. Gut 2011; 60:1520.
  95. Uzzan M, Ko HM, Rosenstein AK, et al. Efficient long-term depletion of CD20+ B cells by rituximab does not affect gut-resident plasma cells. Ann N Y Acad Sci 2018; 1415:5.
  96. Butcher EC, Picker LJ. Lymphocyte homing and homeostasis. Science 1996; 272:60.
  97. Koizumi M, King N, Lobb R, et al. Expression of vascular adhesion molecules in inflammatory bowel disease. Gastroenterology 1992; 103:840.
  98. Salmi M, Granfors K, MacDermott R, Jalkanen S. Aberrant binding of lamina propria lymphocytes to vascular endothelium in inflammatory bowel diseases. Gastroenterology 1994; 106:596.
  99. Malizia G, Calabrese A, Cottone M, et al. Expression of leukocyte adhesion molecules by mucosal mononuclear phagocytes in inflammatory bowel disease. Gastroenterology 1991; 100:150.
  100. West NR, Hegazy AN, Owens BMJ, et al. Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease. Nat Med 2017; 23:579.
  101. Li C, Kuemmerle JF. Mechanisms that mediate the development of fibrosis in patients with Crohn's disease. Inflamm Bowel Dis 2014; 20:1250.
  102. Feagan BG, Greenberg GR, Wild G, et al. Treatment of active Crohn's disease with MLN0002, a humanized antibody to the alpha4beta7 integrin. Clin Gastroenterol Hepatol 2008; 6:1370.
  103. Feagan BG, Greenberg GR, Wild G, et al. Treatment of ulcerative colitis with a humanized antibody to the alpha4beta7 integrin. N Engl J Med 2005; 352:2499.
  104. Sandborn WJ, Feagan BG, D'Haens G, et al. Ozanimod as Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med 2021; 385:1280.
  105. Padoan A, Musso G, Contran N, Basso D. Inflammation, Autoinflammation and Autoimmunity in Inflammatory Bowel Diseases. Curr Issues Mol Biol 2023; 45:5534.
  106. Kühn R, Löhler J, Rennick D, et al. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 1993; 75:263.
  107. Powrie F, Leach MW, Mauze S, et al. Phenotypically distinct subsets of CD4+ T cells induce or protect from chronic intestinal inflammation in C. B-17 scid mice. Int Immunol 1993; 5:1461.
  108. Fuss IJ, Marth T, Neurath MF, et al. Anti-interleukin 12 treatment regulates apoptosis of Th1 T cells in experimental colitis in mice. Gastroenterology 1999; 117:1078.
  109. Monteleone G, MacDonald TT, Wathen NC, et al. Enhancing Lamina propria Th1 cell responses with interleukin 12 produces severe tissue injury. Gastroenterology 1999; 117:1069.
  110. Powrie F, Leach MW, Mauze S, et al. Inhibition of Th1 responses prevents inflammatory bowel disease in scid mice reconstituted with CD45RBhi CD4+ T cells. Immunity 1994; 1:553.
  111. Berg DJ, Davidson N, Kühn R, et al. Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4(+) TH1-like responses. J Clin Invest 1996; 98:1010.
  112. Targan SR, Hanauer SB, van Deventer SJ, et al. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn's disease. Crohn's Disease cA2 Study Group. N Engl J Med 1997; 337:1029.
  113. Iqbal N, Oliver JR, Wagner FH, et al. T helper 1 and T helper 2 cells are pathogenic in an antigen-specific model of colitis. J Exp Med 2002; 195:71.
  114. Heller F, Florian P, Bojarski C, et al. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology 2005; 129:550.
  115. Ahrens R, Waddell A, Seidu L, et al. Intestinal macrophage/epithelial cell-derived CCL11/eotaxin-1 mediates eosinophil recruitment and function in pediatric ulcerative colitis. J Immunol 2008; 181:7390.
  116. Griseri T, Arnold IC, Pearson C, et al. Granulocyte Macrophage Colony-Stimulating Factor-Activated Eosinophils Promote Interleukin-23 Driven Chronic Colitis. Immunity 2015; 43:187.
  117. Fujino S, Andoh A, Bamba S, et al. Increased expression of interleukin 17 in inflammatory bowel disease. Gut 2003; 52:65.
  118. Schmidt C, Giese T, Ludwig B, et al. Expression of interleukin-12-related cytokine transcripts in inflammatory bowel disease: elevated interleukin-23p19 and interleukin-27p28 in Crohn's disease but not in ulcerative colitis. Inflamm Bowel Dis 2005; 11:16.
  119. Andoh A, Zhang Z, Inatomi O, et al. Interleukin-22, a member of the IL-10 subfamily, induces inflammatory responses in colonic subepithelial myofibroblasts. Gastroenterology 2005; 129:969.
  120. Annunziato F, Cosmi L, Santarlasci V, et al. Phenotypic and functional features of human Th17 cells. J Exp Med 2007; 204:1849.
  121. Fina D, Sarra M, Fantini MC, et al. Regulation of gut inflammation and th17 cell response by interleukin-21. Gastroenterology 2008; 134:1038.
  122. Saruta M, Yu QT, Avanesyan A, et al. Phenotype and effector function of CC chemokine receptor 9-expressing lymphocytes in small intestinal Crohn's disease. J Immunol 2007; 178:3293.
  123. Rovedatti L, Kudo T, Biancheri P, et al. Differential regulation of interleukin 17 and interferon gamma production in inflammatory bowel disease. Gut 2009; 58:1629.
  124. Hovhannisyan Z, Treatman J, Littman DR, Mayer L. Characterization of interleukin-17-producing regulatory T cells in inflamed intestinal mucosa from patients with inflammatory bowel diseases. Gastroenterology 2011; 140:957.
  125. Uhlig HH, McKenzie BS, Hue S, et al. Differential activity of IL-12 and IL-23 in mucosal and systemic innate immune pathology. Immunity 2006; 25:309.
  126. Hue S, Ahern P, Buonocore S, et al. Interleukin-23 drives innate and T cell-mediated intestinal inflammation. J Exp Med 2006; 203:2473.
  127. Damen GM, van Lierop P, de Ruiter L, et al. Production of IL12p70 and IL23 by monocyte-derived dendritic cells in children with inflammatory bowel disease. Gut 2008; 57:1480.
  128. Izcue A, Hue S, Buonocore S, et al. Interleukin-23 restrains regulatory T cell activity to drive T cell-dependent colitis. Immunity 2008; 28:559.
  129. Fuss IJ, Becker C, Yang Z, et al. Both IL-12p70 and IL-23 are synthesized during active Crohn's disease and are down-regulated by treatment with anti-IL-12 p40 monoclonal antibody. Inflamm Bowel Dis 2006; 12:9.
  130. Yen D, Cheung J, Scheerens H, et al. IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6. J Clin Invest 2006; 116:1310.
  131. Elson CO, Cong Y, Weaver CT, et al. Monoclonal anti-interleukin 23 reverses active colitis in a T cell-mediated model in mice. Gastroenterology 2007; 132:2359.
  132. Abraham C, Dulai PS, Vermeire S, Sandborn WJ. Lessons Learned From Trials Targeting Cytokine Pathways in Patients With Inflammatory Bowel Diseases. Gastroenterology 2017; 152:374.
  133. Ahern PP, Schiering C, Buonocore S, et al. Interleukin-23 drives intestinal inflammation through direct activity on T cells. Immunity 2010; 33:279.
  134. Lee Y, Awasthi A, Yosef N, et al. Induction and molecular signature of pathogenic TH17 cells. Nat Immunol 2012; 13:991.
  135. Canavan JB, Scottà C, Vossenkämper A, et al. Developing in vitro expanded CD45RA+ regulatory T cells as an adoptive cell therapy for Crohn's disease. Gut 2016; 65:584.
  136. Bamias G, Martin C 3rd, Marini M, et al. Expression, localization, and functional activity of TL1A, a novel Th1-polarizing cytokine in inflammatory bowel disease. J Immunol 2003; 171:4868.
  137. Jin S, Chin J, Seeber S, et al. TL1A/TNFSF15 directly induces proinflammatory cytokines, including TNFα, from CD3+CD161+ T cells to exacerbate gut inflammation. Mucosal Immunol 2013; 6:886.
  138. Papadakis KA, Prehn JL, Landers C, et al. TL1A synergizes with IL-12 and IL-18 to enhance IFN-gamma production in human T cells and NK cells. J Immunol 2004; 172:7002.
  139. Takedatsu H, Michelsen KS, Wei B, et al. TL1A (TNFSF15) regulates the development of chronic colitis by modulating both T-helper 1 and T-helper 17 activation. Gastroenterology 2008; 135:552.
  140. Bamias G, Mishina M, Nyce M, et al. Role of TL1A and its receptor DR3 in two models of chronic murine ileitis. Proc Natl Acad Sci U S A 2006; 103:8441.
  141. Meylan F, Richard AC, Siegel RM. TL1A and DR3, a TNF family ligand-receptor pair that promotes lymphocyte costimulation, mucosal hyperplasia, and autoimmune inflammation. Immunol Rev 2011; 244:188.
  142. Papadakis KA, Zhu D, Prehn JL, et al. Dominant role for TL1A/DR3 pathway in IL-12 plus IL-18-induced IFN-gamma production by peripheral blood and mucosal CCR9+ T lymphocytes. J Immunol 2005; 174:4985.
  143. Pappu BP, Borodovsky A, Zheng TS, et al. TL1A-DR3 interaction regulates Th17 cell function and Th17-mediated autoimmune disease. J Exp Med 2008; 205:1049.
  144. Hedl M, Abraham C. A TNFSF15 disease-risk polymorphism increases pattern-recognition receptor-induced signaling through caspase-8-induced IL-1. Proc Natl Acad Sci U S A 2014; 111:13451.
  145. Longman RS, Diehl GE, Victorio DA, et al. CX₃CR1⁺ mononuclear phagocytes support colitis-associated innate lymphoid cell production of IL-22. J Exp Med 2014; 211:1571.
  146. Jostins L, Ripke S, Weersma RK, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012; 491:119.
  147. Barrett JC, Hansoul S, Nicolae DL, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet 2008; 40:955.
  148. Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 2007; 447:661.
  149. Yamazaki K, McGovern D, Ragoussis J, et al. Single nucleotide polymorphisms in TNFSF15 confer susceptibility to Crohn's disease. Hum Mol Genet 2005; 14:3499.
  150. Yang SK, Hong M, Zhao W, et al. Genome-wide association study of Crohn's disease in Koreans revealed three new susceptibility loci and common attributes of genetic susceptibility across ethnic populations. Gut 2014; 63:80.
  151. Liu JZ, van Sommeren S, Huang H, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet 2015; 47:979.
  152. Lozupone CA, Stombaugh JI, Gordon JI, et al. Diversity, stability and resilience of the human gut microbiota. Nature 2012; 489:220.
  153. Ursell LK, Haiser HJ, Van Treuren W, et al. The intestinal metabolome: an intersection between microbiota and host. Gastroenterology 2014; 146:1470.
  154. Franzosa EA, Hsu T, Sirota-Madi A, et al. Sequencing and beyond: integrating molecular 'omics' for microbial community profiling. Nat Rev Microbiol 2015; 13:360.
  155. Neish AS, Gewirtz AT, Zeng H, et al. Prokaryotic regulation of epithelial responses by inhibition of IkappaB-alpha ubiquitination. Science 2000; 289:1560.
  156. Swanson PA 2nd, Kumar A, Samarin S, et al. Enteric commensal bacteria potentiate epithelial restitution via reactive oxygen species-mediated inactivation of focal adhesion kinase phosphatases. Proc Natl Acad Sci U S A 2011; 108:8803.
  157. Kelly D, Campbell JI, King TP, et al. Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-gamma and RelA. Nat Immunol 2004; 5:104.
  158. Olszak T, An D, Zeissig S, et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 2012; 336:489.
  159. Bain CC, Bravo-Blas A, Scott CL, et al. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nat Immunol 2014; 15:929.
  160. Ivanov II, Atarashi K, Manel N, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009; 139:485.
  161. Ivanov II, Frutos Rde L, Manel N, et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 2008; 4:337.
  162. Lathrop SK, Bloom SM, Rao SM, et al. Peripheral education of the immune system by colonic commensal microbiota. Nature 2011; 478:250.
  163. Ohnmacht C, Park JH, Cording S, et al. MUCOSAL IMMUNOLOGY. The microbiota regulates type 2 immunity through RORγt⁺ T cells. Science 2015; 349:989.
  164. Stefka AT, Feehley T, Tripathi P, et al. Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci U S A 2014; 111:13145.
  165. Bosurgi L, Bernink JH, Delgado Cuevas V, et al. Paradoxical role of the proto-oncogene Axl and Mer receptor tyrosine kinases in colon cancer. Proc Natl Acad Sci U S A 2013; 110:13091.
  166. Lee J, Mo JH, Katakura K, et al. Maintenance of colonic homeostasis by distinctive apical TLR9 signalling in intestinal epithelial cells. Nat Cell Biol 2006; 8:1327.
  167. Macpherson AJ, McCoy KD, Johansen FE, Brandtzaeg P. The immune geography of IgA induction and function. Mucosal Immunol 2008; 1:11.
  168. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004; 118:229.
  169. Vijay-Kumar M, Sanders CJ, Taylor RT, et al. Deletion of TLR5 results in spontaneous colitis in mice. J Clin Invest 2007; 117:3909.
  170. Bouskra D, Brézillon C, Bérard M, et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature 2008; 456:507.
  171. Sefik E, Geva-Zatorsky N, Oh S, et al. MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce a distinct population of RORγ⁺ regulatory T cells. Science 2015; 349:993.
  172. Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006; 444:1027.
  173. Garrett WS, Lord GM, Punit S, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell 2007; 131:33.
  174. Elinav E, Strowig T, Kau AL, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 2011; 145:745.
  175. Josefowicz SZ, Niec RE, Kim HY, et al. Extrathymically generated regulatory T cells control mucosal TH2 inflammation. Nature 2012; 482:395.
  176. Vaishnava S, Yamamoto M, Severson KM, et al. The antibacterial lectin RegIIIgamma promotes the spatial segregation of microbiota and host in the intestine. Science 2011; 334:255.
  177. Kim KS, Hong SW, Han D, et al. Dietary antigens limit mucosal immunity by inducing regulatory T cells in the small intestine. Science 2016; 351:858.
  178. Wu GD, Chen J, Hoffmann C, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011; 334:105.
  179. Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, et al. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology 2009; 137:1716.
  180. Desai MS, Seekatz AM, Koropatkin NM, et al. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell 2016; 167:1339.
  181. Weinstock JV. Helminths and mucosal immune modulation. Ann N Y Acad Sci 2006; 1072:356.
  182. Margolis DJ, Fanelli M, Hoffstad O, Lewis JD. Potential association between the oral tetracycline class of antimicrobials used to treat acne and inflammatory bowel disease. Am J Gastroenterol 2010; 105:2610.
  183. Shaw SY, Blanchard JF, Bernstein CN. Association between the use of antibiotics and new diagnoses of Crohn's disease and ulcerative colitis. Am J Gastroenterol 2011; 106:2133.
  184. Kronman MP, Zaoutis TE, Haynes K, et al. Antibiotic exposure and IBD development among children: a population-based cohort study. Pediatrics 2012; 130:e794.
  185. Shaw SY, Blanchard JF, Bernstein CN. Association between early childhood otitis media and pediatric inflammatory bowel disease: an exploratory population-based analysis. J Pediatr 2013; 162:510.
  186. Manichanh C, Rigottier-Gois L, Bonnaud E, et al. Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach. Gut 2006; 55:205.
  187. Frank DN, St Amand AL, Feldman RA, et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A 2007; 104:13780.
  188. Barnich N, Carvalho FA, Glasser AL, et al. CEACAM6 acts as a receptor for adherent-invasive E. coli, supporting ileal mucosa colonization in Crohn disease. J Clin Invest 2007; 117:1566.
  189. Gevers D, Kugathasan S, Denson LA, et al. The treatment-naive microbiome in new-onset Crohn's disease. Cell Host Microbe 2014; 15:382.
  190. Haberman Y, Tickle TL, Dexheimer PJ, et al. Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature. J Clin Invest 2014; 124:3617.
  191. Morgan XC, Tickle TL, Sokol H, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 2012; 13:R79.
  192. Atarashi K, Tanoue T, Ando M, et al. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell 2015; 163:367.
  193. Britton GJ, Contijoch EJ, Mogno I, et al. Microbiotas from Humans with Inflammatory Bowel Disease Alter the Balance of Gut Th17 and RORγt+ Regulatory T Cells and Exacerbate Colitis in Mice. Immunity 2019; 50:212.
  194. Sutton CL, Kim J, Yamane A, et al. Identification of a novel bacterial sequence associated with Crohn's disease. Gastroenterology 2000; 119:23.
  195. Lodes MJ, Cong Y, Elson CO, et al. Bacterial flagellin is a dominant antigen in Crohn disease. J Clin Invest 2004; 113:1296.
  196. Targan SR, Landers CJ, Yang H, et al. Antibodies to CBir1 flagellin define a unique response that is associated independently with complicated Crohn's disease. Gastroenterology 2005; 128:2020.
  197. Mow WS, Vasiliauskas EA, Lin YC, et al. Association of antibody responses to microbial antigens and complications of small bowel Crohn's disease. Gastroenterology 2004; 126:414.
  198. Elkadri AA, Stempak JM, Walters TD, et al. Serum antibodies associated with complex inflammatory bowel disease. Inflamm Bowel Dis 2013; 19:1499.
  199. Tyler AD, Knox N, Kabakchiev B, et al. Characterization of the gut-associated microbiome in inflammatory pouch complications following ileal pouch-anal anastomosis. PLoS One 2013; 8:e66934.
  200. Morgan XC, Kabakchiev B, Waldron L, et al. Associations between host gene expression, the mucosal microbiome, and clinical outcome in the pelvic pouch of patients with inflammatory bowel disease. Genome Biol 2015; 16:67.
  201. Reshef L, Kovacs A, Ofer A, et al. Pouch Inflammation Is Associated With a Decrease in Specific Bacterial Taxa. Gastroenterology 2015; 149:718.
  202. Taurog JD, Richardson JA, Croft JT, et al. The germfree state prevents development of gut and joint inflammatory disease in HLA-B27 transgenic rats. J Exp Med 1994; 180:2359.
  203. Contractor NV, Bassiri H, Reya T, et al. Lymphoid hyperplasia, autoimmunity, and compromised intestinal intraepithelial lymphocyte development in colitis-free gnotobiotic IL-2-deficient mice. J Immunol 1998; 160:385.
  204. Bloom SM, Bijanki VN, Nava GM, et al. Commensal Bacteroides species induce colitis in host-genotype-specific fashion in a mouse model of inflammatory bowel disease. Cell Host Microbe 2011; 9:390.
  205. Palm NW, de Zoete MR, Cullen TW, et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 2014; 158:1000.
  206. Atarashi K, Tanoue T, Oshima K, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013; 500:232.
  207. Yan F, Cao H, Cover TL, et al. Colon-specific delivery of a probiotic-derived soluble protein ameliorates intestinal inflammation in mice through an EGFR-dependent mechanism. J Clin Invest 2011; 121:2242.
  208. Wen L, Ley RE, Volchkov PY, et al. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature 2008; 455:1109.
  209. Vijay-Kumar M, Aitken JD, Carvalho FA, et al. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 2010; 328:228.
  210. Hsiao EY, McBride SW, Hsien S, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013; 155:1451.
  211. Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 2008; 453:620.
  212. Maslowski KM, Vieira AT, Ng A, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009; 461:1282.
  213. Smith PM, Howitt MR, Panikov N, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013; 341:569.
  214. Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013; 504:446.
  215. Atarashi K, Nishimura J, Shima T, et al. ATP drives lamina propria T(H)17 cell differentiation. Nature 2008; 455:808.
  216. Schulthess J, Pandey S, Capitani M, et al. The Short Chain Fatty Acid Butyrate Imprints an Antimicrobial Program in Macrophages. Immunity 2019; 50:432.
  217. Norman JM, Handley SA, Baldridge MT, et al. Disease-specific alterations in the enteric virome in inflammatory bowel disease. Cell 2015; 160:447.
  218. Cadwell K, Patel KK, Maloney NS, et al. Virus-plus-susceptibility gene interaction determines Crohn's disease gene Atg16L1 phenotypes in intestine. Cell 2010; 141:1135.
  219. Iliev ID, Funari VA, Taylor KD, et al. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science 2012; 336:1314.
  220. Chehoud C, Albenberg LG, Judge C, et al. Fungal Signature in the Gut Microbiota of Pediatric Patients With Inflammatory Bowel Disease. Inflamm Bowel Dis 2015; 21:1948.
  221. Franke A, McGovern DP, Barrett JC, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn's disease susceptibility loci. Nat Genet 2010; 42:1118.
  222. de Lange KM, Moutsianas L, Lee JC, et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat Genet 2017; 49:256.
  223. Moran CJ, Klein C, Muise AM, Snapper SB. Very early-onset inflammatory bowel disease: gaining insight through focused discovery. Inflamm Bowel Dis 2015; 21:1166.
  224. Ogura Y, Bonen DK, Inohara N, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature 2001; 411:603.
  225. Hugot JP, Chamaillard M, Zouali H, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature 2001; 411:599.
  226. Abraham C, Medzhitov R. Interactions between the host innate immune system and microbes in inflammatory bowel disease. Gastroenterology 2011; 140:1729.
  227. Kobayashi KS, Chamaillard M, Ogura Y, et al. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science 2005; 307:731.
  228. Couturier-Maillard A, Secher T, Rehman A, et al. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J Clin Invest 2013; 123:700.
  229. Hedl M, Li J, Cho JH, Abraham C. Chronic stimulation of Nod2 mediates tolerance to bacterial products. Proc Natl Acad Sci U S A 2007; 104:19440.
  230. Watanabe T, Asano N, Murray PJ, et al. Muramyl dipeptide activation of nucleotide-binding oligomerization domain 2 protects mice from experimental colitis. J Clin Invest 2008; 118:545.
  231. Caruso R, Mathes T, Martens EC, et al. A specific gene-microbe interaction drives the development of Crohn's disease-like colitis in mice. Sci Immunol 2019; 4.
  232. Hampe J, Cuthbert A, Croucher PJ, et al. Association between insertion mutation in NOD2 gene and Crohn's disease in German and British populations. Lancet 2001; 357:1925.
  233. Economou M, Trikalinos TA, Loizou KT, et al. Differential effects of NOD2 variants on Crohn's disease risk and phenotype in diverse populations: a metaanalysis. Am J Gastroenterol 2004; 99:2393.
  234. Lesage S, Zouali H, Cézard JP, et al. CARD15/NOD2 mutational analysis and genotype-phenotype correlation in 612 patients with inflammatory bowel disease. Am J Hum Genet 2002; 70:845.
  235. Brant SR, Wang MH, Rawsthorne P, et al. A population-based case-control study of CARD15 and other risk factors in Crohn's disease and ulcerative colitis. Am J Gastroenterol 2007; 102:313.
  236. Inoue N, Tamura K, Kinouchi Y, et al. Lack of common NOD2 variants in Japanese patients with Crohn's disease. Gastroenterology 2002; 123:86.
  237. Croucher PJ, Mascheretti S, Hampe J, et al. Haplotype structure and association to Crohn's disease of CARD15 mutations in two ethnically divergent populations. Eur J Hum Genet 2003; 11:6.
  238. Ahmad T, Armuzzi A, Bunce M, et al. The molecular classification of the clinical manifestations of Crohn's disease. Gastroenterology 2002; 122:854.
  239. Cuthbert AP, Fisher SA, Mirza MM, et al. The contribution of NOD2 gene mutations to the risk and site of disease in inflammatory bowel disease. Gastroenterology 2002; 122:867.
  240. Hampe J, Grebe J, Nikolaus S, et al. Association of NOD2 (CARD 15) genotype with clinical course of Crohn's disease: a cohort study. Lancet 2002; 359:1661.
  241. Abreu MT, Taylor KD, Lin YC, et al. Mutations in NOD2 are associated with fibrostenosing disease in patients with Crohn's disease. Gastroenterology 2002; 123:679.
  242. Tyler AD, Milgrom R, Stempak JM, et al. The NOD2insC polymorphism is associated with worse outcome following ileal pouch-anal anastomosis for ulcerative colitis. Gut 2013; 62:1433.
  243. Adler J, Rangwalla SC, Dwamena BA, Higgins PD. The prognostic power of the NOD2 genotype for complicated Crohn's disease: a meta-analysis. Am J Gastroenterol 2011; 106:699.
  244. Hedl M, Abraham C. IRF5 risk polymorphisms contribute to interindividual variance in pattern recognition receptor-mediated cytokine secretion in human monocyte-derived cells. J Immunol 2012; 188:5348.
  245. Hedl M, Abraham C. A TPL2 (MAP3K8) disease-risk polymorphism increases TPL2 expression thereby leading to increased pattern recognition receptor-initiated caspase-1 and caspase-8 activation, signalling and cytokine secretion. Gut 2016; 65:1799.
  246. Hedl M, Proctor DD, Abraham C. JAK2 Disease-Risk Variants Are Gain of Function and JAK Signaling Threshold Determines Innate Receptor-Induced Proinflammatory Cytokine Secretion in Macrophages. J Immunol 2016; 197:3695.
  247. Hedl M, Yan J, Witt H, Abraham C. IRF5 Is Required for Bacterial Clearance in Human M1-Polarized Macrophages, and IRF5 Immune-Mediated Disease Risk Variants Modulate This Outcome. J Immunol 2019; 202:920.
  248. Hedl M, Yan J, Abraham C. IRF5 and IRF5 Disease-Risk Variants Increase Glycolysis and Human M1 Macrophage Polarization by Regulating Proximal Signaling and Akt2 Activation. Cell Rep 2016; 16:2442.
  249. Pandey SP, Yan J, Turner JR, Abraham C. Reducing IRF5 expression attenuates colitis in mice, but impairs the clearance of intestinal pathogens. Mucosal Immunol 2019; 12:874.
  250. Sun R, Hedl M, Abraham C. TNFSF15 Promotes Antimicrobial Pathways in Human Macrophages and These Are Modulated by TNFSF15 Disease-Risk Variants. Cell Mol Gastroenterol Hepatol 2021; 11:249.
  251. Hedl M, Sun R, Abraham C. Disease Risk-Associated Genetic Variants in STAT1 and STAT4 Function in a Complementary Manner to Increase Pattern-Recognition Receptor-Induced Outcomes in Human Macrophages. J Immunol 2020; 205:1406.
  252. Hedl M, Sun R, Huang C, Abraham C. STAT3 and STAT5 Signaling Thresholds Determine Distinct Regulation for Innate Receptor-Induced Inflammatory Cytokines, and STAT3/STAT5 Disease Variants Modulate These Outcomes. J Immunol 2019; 203:3325.
  253. Hedl M, Lahiri A, Ning K, et al. Pattern recognition receptor signaling in human dendritic cells is enhanced by ICOS ligand and modulated by the Crohn's disease ICOSLG risk allele. Immunity 2014; 40:734.
  254. Yan J, Hedl M, Abraham C. An inflammatory bowel disease-risk variant in INAVA decreases pattern recognition receptor-induced outcomes. J Clin Invest 2017; 127:2192.
  255. Lahiri A, Hedl M, Yan J, Abraham C. Human LACC1 increases innate receptor-induced responses and a LACC1 disease-risk variant modulates these outcomes. Nat Commun 2017; 8:15614.
  256. Cader MZ, Boroviak K, Zhang Q, et al. C13orf31 (FAMIN) is a central regulator of immunometabolic function. Nat Immunol 2016; 17:1046.
  257. Ranjan K, Hedl M, Abraham C. The E3 ubiquitin ligase RNF186 and RNF186 risk variants regulate innate receptor-induced outcomes. Proc Natl Acad Sci U S A 2021; 118.
  258. Huang C, Hedl M, Ranjan K, Abraham C. LACC1 Required for NOD2-Induced, ER Stress-Mediated Innate Immune Outcomes in Human Macrophages and LACC1 Risk Variants Modulate These Outcomes. Cell Rep 2019; 29:4525.
  259. Kang JW, Yan J, Ranjan K, et al. Myeloid Cell Expression of LACC1 Is Required for Bacterial Clearance and Control of Intestinal Inflammation. Gastroenterology 2020; 159:1051.
  260. Ranjan K, Hedl M, Sinha S, et al. Ubiquitination of ATF6 by disease-associated RNF186 promotes the innate receptor-induced unfolded protein response. J Clin Invest 2021; 131.
  261. Kaser A, Lee AH, Franke A, et al. XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell 2008; 134:743.
  262. Fisher SA, Tremelling M, Anderson CA, et al. Genetic determinants of ulcerative colitis include the ECM1 locus and five loci implicated in Crohn's disease. Nat Genet 2008; 40:710.
  263. Franke A, Balschun T, Karlsen TH, et al. Replication of signals from recent studies of Crohn's disease identifies previously unknown disease loci for ulcerative colitis. Nat Genet 2008; 40:713.
  264. Rioux JD, Daly MJ, Silverberg MS, et al. Genetic variation in the 5q31 cytokine gene cluster confers susceptibility to Crohn disease. Nat Genet 2001; 29:223.
  265. Stoll M, Corneliussen B, Costello CM, et al. Genetic variation in DLG5 is associated with inflammatory bowel disease. Nat Genet 2004; 36:476.
  266. Peltekova VD, Wintle RF, Rubin LA, et al. Functional variants of OCTN cation transporter genes are associated with Crohn disease. Nat Genet 2004; 36:471.
  267. Franke A, Hampe J, Rosenstiel P, et al. Systematic association mapping identifies NELL1 as a novel IBD disease gene. PLoS One 2007; 2:e691.
  268. Plantinga TS, Crisan TO, Oosting M, et al. Crohn's disease-associated ATG16L1 polymorphism modulates pro-inflammatory cytokine responses selectively upon activation of NOD2. Gut 2011; 60:1229.
  269. Lassen KG, Kuballa P, Conway KL, et al. Atg16L1 T300A variant decreases selective autophagy resulting in altered cytokine signaling and decreased antibacterial defense. Proc Natl Acad Sci U S A 2014; 111:7741.
  270. Lahiri A, Hedl M, Abraham C. MTMR3 risk allele enhances innate receptor-induced signaling and cytokines by decreasing autophagy and increasing caspase-1 activation. Proc Natl Acad Sci U S A 2015; 112:10461.
  271. Muise AM, Xu W, Guo CH, et al. NADPH oxidase complex and IBD candidate gene studies: identification of a rare variant in NCF2 that results in reduced binding to RAC2. Gut 2012; 61:1028.
  272. Dhillon SS, Fattouh R, Elkadri A, et al. Variants in nicotinamide adenine dinucleotide phosphate oxidase complex components determine susceptibility to very early onset inflammatory bowel disease. Gastroenterology 2014; 147:680.
  273. Knights D, Silverberg MS, Weersma RK, et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med 2014; 6:107.
  274. Tong M, McHardy I, Ruegger P, et al. Reprograming of gut microbiome energy metabolism by the FUT2 Crohn's disease risk polymorphism. ISME J 2014; 8:2193.
  275. Sarin R, Wu X, Abraham C. Inflammatory disease protective R381Q IL23 receptor polymorphism results in decreased primary CD4+ and CD8+ human T-cell functional responses. Proc Natl Acad Sci U S A 2011; 108:9560.
  276. Di Meglio P, Di Cesare A, Laggner U, et al. The IL23R R381Q gene variant protects against immune-mediated diseases by impairing IL-23-induced Th17 effector response in humans. PLoS One 2011; 6:e17160.
  277. Pidasheva S, Trifari S, Phillips A, et al. Functional studies on the IBD susceptibility gene IL23R implicate reduced receptor function in the protective genetic variant R381Q. PLoS One 2011; 6:e25038.
  278. Willson TA, Kuhn BR, Jurickova I, et al. STAT3 genotypic variation and cellular STAT3 activation and colon leukocyte recruitment in pediatric Crohn disease. J Pediatr Gastroenterol Nutr 2012; 55:32.
  279. Shouval DS, Biswas A, Goettel JA, et al. Interleukin-10 receptor signaling in innate immune cells regulates mucosal immune tolerance and anti-inflammatory macrophage function. Immunity 2014; 40:706.
  280. Zigmond E, Bernshtein B, Friedlander G, et al. Macrophage-restricted interleukin-10 receptor deficiency, but not IL-10 deficiency, causes severe spontaneous colitis. Immunity 2014; 40:720.
  281. Murai M, Turovskaya O, Kim G, et al. Interleukin 10 acts on regulatory T cells to maintain expression of the transcription factor Foxp3 and suppressive function in mice with colitis. Nat Immunol 2009; 10:1178.
  282. Rubtsov YP, Rasmussen JP, Chi EY, et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 2008; 28:546.
  283. Ip WKE, Hoshi N, Shouval DS, et al. Anti-inflammatory effect of IL-10 mediated by metabolic reprogramming of macrophages. Science 2017; 356:513.
  284. Glocker EO, Kotlarz D, Boztug K, et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med 2009; 361:2033.
  285. Kotlarz D, Beier R, Murugan D, et al. Loss of interleukin-10 signaling and infantile inflammatory bowel disease: implications for diagnosis and therapy. Gastroenterology 2012; 143:347.
  286. Moran CJ, Walters TD, Guo CH, et al. IL-10R polymorphisms are associated with very-early-onset ulcerative colitis. Inflamm Bowel Dis 2013; 19:115.
  287. Shouval DS, Biswas A, Kang YH, et al. Interleukin 1β Mediates Intestinal Inflammation in Mice and Patients With Interleukin 10 Receptor Deficiency. Gastroenterology 2016; 151:1100.
  288. Kugathasan S, Baldassano RN, Bradfield JP, et al. Loci on 20q13 and 21q22 are associated with pediatric-onset inflammatory bowel disease. Nat Genet 2008; 40:1211.
  289. Shih DQ, Zheng L, Zhang X, et al. Inhibition of a novel fibrogenic factor Tl1a reverses established colonic fibrosis. Mucosal Immunol 2014; 7:1492.
  290. Hedl M, Zheng S, Abraham C. The IL18RAP region disease polymorphism decreases IL-18RAP/IL-18R1/IL-1R1 expression and signaling through innate receptor-initiated pathways. J Immunol 2014; 192:5924.
  291. Yan J, Pandey SP, Barnes BJ, et al. T Cell-Intrinsic IRF5 Regulates T Cell Signaling, Migration, and Differentiation and Promotes Intestinal Inflammation. Cell Rep 2020; 31:107820.
  292. Spalinger MR, Kasper S, Chassard C, et al. PTPN2 controls differentiation of CD4⁺ T cells and limits intestinal inflammation and intestinal dysbiosis. Mucosal Immunol 2015; 8:918.
  293. Scharl M, Paul G, Weber A, et al. Protection of epithelial barrier function by the Crohn's disease associated gene protein tyrosine phosphatase n2. Gastroenterology 2009; 137:2030.
  294. Liu TC, Kern JT, VanDussen KL, et al. Interaction between smoking and ATG16L1T300A triggers Paneth cell defects in Crohn's disease. J Clin Invest 2018; 128:5110.
  295. Kattah MG, Shao L, Rosli YY, et al. A20 and ABIN-1 synergistically preserve intestinal epithelial cell survival. J Exp Med 2018; 215:1839.
  296. Fujimoto K, Kinoshita M, Tanaka H, et al. Regulation of intestinal homeostasis by the ulcerative colitis-associated gene RNF186. Mucosal Immunol 2017; 10:446.
  297. Luong P, Hedl M, Yan J, et al. INAVA-ARNO complexes bridge mucosal barrier function with inflammatory signaling. Elife 2018; 7.
  298. Sadlack B, Merz H, Schorle H, et al. Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene. Cell 1993; 75:253.
  299. Willerford DM, Chen J, Ferry JA, et al. Interleukin-2 receptor alpha chain regulates the size and content of the peripheral lymphoid compartment. Immunity 1995; 3:521.
  300. Mombaerts P, Mizoguchi E, Grusby MJ, et al. Spontaneous development of inflammatory bowel disease in T cell receptor mutant mice. Cell 1993; 75:274.
  301. Rudolph U, Finegold MJ, Rich SS, et al. Ulcerative colitis and adenocarcinoma of the colon in G alpha i2-deficient mice. Nat Genet 1995; 10:143.
  302. Morteau O, Morham SG, Sellon R, et al. Impaired mucosal defense to acute colonic injury in mice lacking cyclooxygenase-1 or cyclooxygenase-2. J Clin Invest 2000; 105:469.
  303. Nguyen DD, Maillard MH, Cotta-de-Almeida V, et al. Lymphocyte-dependent and Th2 cytokine-associated colitis in mice deficient in Wiskott-Aldrich syndrome protein. Gastroenterology 2007; 133:1188.
  304. Turer EE, Tavares RM, Mortier E, et al. Homeostatic MyD88-dependent signals cause lethal inflamMation in the absence of A20. J Exp Med 2008; 205:451.
  305. Holländer GA, Simpson SJ, Mizoguchi E, et al. Severe colitis in mice with aberrant thymic selection. Immunity 1995; 3:27.
  306. Morrissey PJ, Charrier K, Braddy S, et al. CD4+ T cells that express high levels of CD45RB induce wasting disease when transferred into congenic severe combined immunodeficient mice. Disease development is prevented by cotransfer of purified CD4+ T cells. J Exp Med 1993; 178:237.
  307. Hammer RE, Maika SD, Richardson JA, et al. Spontaneous inflammatory disease in transgenic rats expressing HLA-B27 and human beta 2m: an animal model of HLA-B27-associated human disorders. Cell 1990; 63:1099.
  308. Kosiewicz MM, Nast CC, Krishnan A, et al. Th1-type responses mediate spontaneous ileitis in a novel murine model of Crohn's disease. J Clin Invest 2001; 107:695.
Topic 4077 Version 28.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟