Skip Navigation
Skip to contents

Intest Res : Intestinal Research

IMPACT FACTOR

Articles

Page Path
HOME > Intest Res > Volume 16(1); 2018 > Article
Review Immunological pathogenesis of inflammatory bowel disease
Seung Hoon Lee, Jeong eun Kwon, Mi-La Cho
Intestinal Research 2018;16(1):26-42.
DOI: https://doi.org/10.5217/ir.2018.16.1.26
Published online: January 18, 2018

The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea.

Correspondence to Mi-La Cho, The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea. Tel: +82-2-2258-7467, Fax: +82-2-599-4287, iammila@catholic.ac.kr
• Received: March 15, 2017   • Revised: April 18, 2017   • Accepted: April 19, 2017

© Copyright 2018. Korean Association for the Study of Intestinal Diseases.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

prev next
  • 37,678 Views
  • 1,391 Download
  • 411 Web of Science
  • 408 Crossref
  • 438 Scopus
  • Inflammatory bowel disease (IBD) is a chronic inflammatory state of the gastrointestinal tract and can be classified into 2 main clinical phenomena: Crohn's disease (CD) and ulcerative colitis (UC). The pathogenesis of IBD, including CD and UC, involves the presence of pathogenic factors such as abnormal gut microbiota, immune response dysregulation, environmental changes, and gene variants. Although many investigations have tried to identify novel pathogenic factors associated with IBD that are related to environmental, genetic, microbial, and immune response factors, a full understanding of IBD pathogenesis is unclear. Thus, IBD treatment is far from optimal, and patient outcomes can be unsatisfactory. As result of massive studying on IBD, T helper 17 (Th17) cells and innate lymphoid cells (ILCs) are investigated on their effects on IBD. A recent study of the plasticity of Th17 cells focused primarily on colitis. ILCs also emerging as novel cell family, which play a role in the pathogenesis of IBD. IBD immunopathogenesis is key to understanding the causes of IBD and can lead to the development of IBD therapies. The aim of this review is to explain the pathogenesis of IBD, with a focus on immunological factors and therapies.
The gastrointestinal tract is chronically exposed to various antigens found in bacteria and food. In the normal state in the absence of intestinal inflammation, gut homeostasis is maintained by suppressing excessive immune responses to foreign antigens. IBD is an idiopathic disorder caused by chronic and excessive inflammation of the gastrointestinal tract, leading to rectal bleeding and weight loss.12 IBD, a dysregulated immune inflammatory state of the gastrointestinal tract, is classified into 2 archetypal phenotypes, UC and CD. These 2 subtypes of IBD are characterized by chronic inflammation in the gastrointestinal tract and repeated cycles of relapse and remission. Although UC and CD show differences in their clinical presentation, the same risk factors are implicated in the pathogenesis of both subtypes. Phenotypes common to both subtypes include chronic inflammation and a dysregulated immune inflammatory response; therefore, much of the research on IBD pathogenesis has focused on the immune system. The pathogenesis of both UC and CD involve genetic factors, changes in the gut microbiome, and immune response cells including cytokines and immune cells.
Even though the pathogenesis of IBD is complicated, several studies have demonstrated that excessive interleukin (IL)-17 production is involved in the progression of IBD.3 Recently, research on IBD pathogenesis has focused on T helper (Th)17 cells, which secrete IL-17. It is well documented that Th17 inhibition can decrease the development of acute colitis by reducing inflammation.4 Additionally, innate lymphoid cells (ILCs) were recently discovered to be novel pathogenic effector lymphocytes in IBD. In this review, this topic will be discussed primarily in the context of human IBD and experimental IBD animal models. Additionally, current therapeutics targeting Th17 and ILCs will be discussed.
Immune cells secrete products that are actively involved in the initiation and preservation of inflammation, leading to gut tissue damage. In IBD patients, colonic lesions show excessive immune cell infiltration and tissue devastation (Fig. 1A). The expression of STAT3 and IL-17 are also increased in inflamed colon tissue (Fig. 1B and C). Many cytokines and chemokines are associated with IBD development.
1. Cytokines
Several pro-inflammatory cytokines are involved in the progression of IBD. For example, the IL-1 family of cytokines has a key role in IBD pathogenesis.5 In UC, IL-1β promotes inflammation because IL-1 originates from monocytes and macrophages, and active IL-1β is expressed in the colonic mucosa.6 IL-18 is also an IL-1 family member and is increased in the mucosa of CD patients.7 It has been suggested that IL-18 increases the Th1 response.89 However, in CD patients with active disease, IL-10 released from mucosal T cells was decreased by IL-18.10 IL-33, another member of the IL-1 family, stimulates mucus secretion to protect the epithelium and upregulates the expression of IL-5 and IL-13 as part of the Th2 response.11 There is evidence that the expression of IL-33 and its receptor ST2 are increased in UC patients.1213
IL-6 activates signal transducer and activator of transcription 3 (STAT3) and has an important function in the inflammatory response. IL-6 and its soluble IL-6 receptor were increased in UC and CD patients.1415 IL-6 also has a key role in the pathogenesis of UC and the carcinogenesis of colorectal cancers related to UC.16
Tumor necrosis factor α (TNF-α) has a significant function in IBD pathogenesis because IL-1β, IL-6, and IL-33 expression can all be increased by TNF-α.1718 The clinical severity of UC and CD were correlated with TNF-α levels in the serum of IBD patients.
IL-10 is a typical immunosuppressive cytokine that may have therapeutic value for treating chronic IBD.19 Although IL-10 is an anti-inflammatory cytokines, there are inconsistencies of IL-10 concentrations in IBD. A study showed that gut IL-10 expression levels were either the same or higher in IBD patients than in normal controls.20 It is well documented that IL-10 gene expression is higher in the mucosal T cells of UC patients than normal controls.21 Furthermore, IL-10 production is enhanced in the serum of CD patients.22 On the other hand, other investigation demonstrated that IL-10 levels in serum of patients with UC and CD are similar to healthy subjects.23 It is also well documented that downregu-lation of IL-10 promotes disease progression in patients with CD.2425
Transforming growth factor β (TGF-β) has dual activities in the pathogenesis of IBD. It stimulates epithelial compensation and fibrosis and induces tolerance and homeostasis through an impressive immunoregulatory function.26 In the lamina propria, TGF-β1 levels in mononuclear cells were enhanced in UC patients but decreased in CD patients.27 TGF-β improved intestinal inflammation by reducing the expression of IL-33.28
IL-17 is a pro-inflammatory cytokine that activates STAT3, which stimulates a strong chronic immune inflammatory response.2930 Thus, IL-17 is critical in the pathogenesis of IBD. Indeed, IL-17 mRNA levels were enhanced in the inflamed mucosa of patients with IBD, both UC and CD.31 IL-17 has many isoforms, including IL-17A (also known as IL-17), IL-17B, IL-17C, IL-17D, IL-17E (also known as IL-25), and IL-17F.32 Although IL-17A inhibition can attenuate the inflammatory response, whether IL-17A has a pathogenic role in IBD is controversial. It has been suggested that IL-17A inhibition mediated by phosphorylated STAT3 suppression decreases inflammation and the progression of acute colitis,4 whereas IL-17A can improve experimental colitis.33 Additionally, by reinforcing tight junction formation, IL-17 can also protect human intestinal epithelial cells.34 However, IL-17 is recognized as a significant inflammatory factor in CD pathogenesis. Previous studies found higher levels of IL-17 and CD161+ memory cells expressing IL-17 and interferon γ (IFN-γ) in CD patients.3536 It has been reported that IL-17 can increase the recruitment of T cells into the lamina propria during the inflammatory response.37
2. Chemokines
IL-8 is mainly a neutrophil chemoattractant that induces the migration of neutrophils from peripheral blood into inflamed tissue. It is well known that IL-8 production is increased in the tissue of UC patients compared with that of normal controls.38 Moreover, other chemokines are elevated in the mucosa of IBD patients. Various reports have shown that the expression of chemokine (C-C motif) ligand (CCL) 2 (also known as monocyte chemoattractant protein [MCP]-1), CCL3 (also known as macrophage inflammatory proteins [MIP]-1α), CCL4 (also known as MIP-1β), CCL7 (also known as MCP-3), CCL20 (also known as MIP-3α), CX-C motif chemokine (CXCL) 5, CXCL8, CXCL10 and regulated on activation, normal T cell expressed and secreted (RANTES) are upregulated in tissues from IBD patients.39404142
Presently, Th17 cells are considered a main pathogenic factor in IBD. A study found a massive infiltration of Th17 cells in the inflamed intestinal mucosa of IBD patients.43 Cells that release IL-17 and Th17-related cytokines were also increased in inflamed tissue from IBD patients compared with normal tissue.3143
1. Th17 Cell Differentiation in the Intestine
T cells can be differentiated into Th17 cells when they interact with various inflammatory mediators in the intestine. Naive T cells can be differentiated into Th1, Th2, Th17, and regulatory T (Treg) cells through a process controlled by effector cytokines produced by antigen-presenting cells. During Th17 polarization, many cytokine receptors (IL-1R, IL-6R, IL-21R, IL-23R, and TGF-βR) of naive T cells have important roles. Many reports have shown that specific cytokines such as IL-1β, IL-6, IL-21, IL-23, and TGF-β induce Th17 polarization.444546 STAT3 was first identified as a Th17 cell specific transcription factor It has been suggested that STAT3 overexpression can promote Th17 differentiation and proliferation. Consistent with this, loss of STAT3 can suppress the differentiation of naive T cells into Th17 cells.47
2. Th17 Cells and the Microbiota
The gastrointestinal tract contains a large microbial community. Thus, gut microbial settlement in the gastrointestinal tract is crucial to the formation of the immune system.48 The microbiota also plays a key role in the pathogenesis of IBD. Indeed, metagenomics research has revealed that various intestinal microbiota genes are involved in host mRNA expression.49 Destruction of the intestinal mucosal barrier is caused by genetic susceptibility in IBD patients.50 Analyses of biopsy samples from UC and CD patients have shown that changes in the composition of the microbiota may be associated with IBD pathogenesis.515253
Communication of T cells with microbes in the gastrointestinal tract is very important for maintaining intestinal immunity. The process of Th17 cell differentiation is determined by factors such as the composition of the endogenous microbiota in the intestine.5455 Correspondingly, Th17 cell differentiation in the intestine is remarkably decreased in a germ-free system and in the presence of antibiotics.5657 Additionally, many studies have shown that specific gut microbiota drive Th17 cell differentiation.5658
3. Th17 Cells Function in Intestinal Inflammation
Th cells are relevant to the pathogenesis of IBD because they are fundamentally plastic to provocation from the surrounding state.59 Th17 cells releasing IL-17 are a strong pro-inflammatory factor in IBD. In a study on IBD patients, IL-17 expression and IL-17A and IL-17F mRNA levels were higher in the mucosa and serum of IBD patients than healthy controls. 3160 In a subtype of IBD, IL-17 was more abundant in CD patients than UC patients.31 Recently, various cytokines related to Th17 were found to be upregulated in both UC and CD patients compared with normal subjects, but they were higher in the UC patients.61 It has also been documented that UC severity is correlated with IL-17 expression in peripheral blood mononuclear cells.62 Genome-wide association scans showed that various UC and CD susceptibility genes, such as STAT3, were related to Th17 genes; therefore, the Th17 signaling pathway may be involved in the pathogenesis of IBD.6364
Although it is known that Th17 cells play a role in the pathogenesis of IBD, a dextran sulfate sodium (DSS)-induced colitis model showed that IL-17A and IL-17F have contrasting roles. Studies using this model found that antibody-mediated IL-17A protein suppression and gene knockout caused excessive inflammation and damage to the intestinal epithelium.6566 On the other hand, in mice with DSS-induced colitis, it was shown that IL-17F could also have a protective role.67 It has also been reported that loss of IL-17F can improve the intestinal inflammatory response.67 Thus, the focus of current IBD therapies is on blocking IL-17A and IL-17F.68
4. Maintenance of Intestinal Th17 and Treg Cell Proliferation
Generally, Th17 and Treg cells modulate the proliferation of each other to maintain balance. It has been reported that the developmental pathways of Th17 and Tregs are related with their differentiation and Th17/Treg balance is important to maintain immune response in intestine.69 This influences the outcome of immune responses in the context of inflammatory conditions.
Treg cells are associated with the pathogenesis of IBD because the intestinal inflammatory response in IBD is mediated mainly by the T-cell response.70 Treg cells perform a critical role in preserving immune homeostasis and establishing inflammation in response to foreign or non-pathogenic antigens such as commensal bacteria, and failure of Treg cell function can lead to an inflammatory disorder. Indeed, mutations in CD25 and IL-10, which are involved in Treg cell differentiation, lead to aberrant Treg cell function and increased susceptibility to IBD.71 Moreover, loss of IL-10 results in intestinal inflammation, and Treg cells lacking the IL-10 receptor are more susceptible to colitis.7273
As research using IBD mouse models has shown that Treg cells could suppress intestinal inflammation,74 it is said that Treg cells have an anti-colitis effect. Indeed, ablation of Treg cells or impairment of TGF-β1 signaling in Treg cells increased colitis progression.7576 Additionally, there is evidence that IL-10 released from Treg cells can decrease colitis progression.777879 In a mouse model, an increase in Treg cell differentiation downregulated the development of experimental ileitis, and co-transfer of conventional T cells and Treg cells decreased intestinal inflammation in a RAG1 knock-out mouse model.80
As the concentration of T cell differentiation and balance has moved from the Th1/Th2 to that of Th17/Treg, this paradigm has indeed been shown to affect in IBD. Th17 and Treg cells exist primarily in the intestinal mucosa, where they have a significant role in T-cell-mediated immune responses.8182 It is well known that IL-17-releasing Th17 cells are entirely dependent on STAT3 and are primarily pro-inflammatory.83 On the other hand, Foxp3-expressing Treg cells show anti-inflammatory activity that is mediated through the suppression of the Th17 response. In an experimental colitis model, Treg cells prevented intestinal inflammation and reduced the expression of Th17-related cytokines.84
As Th17 cells play a key function in intestinal inflammation, it has been proposed that they may be therapeutic targets to regulate the intestinal inflammatory response.
1. Th17 Cell Blockade
Theoretically, pro-inflammatory cytokines, including IL-6, can increase Th17 cell differentiation and proliferation. Therefore, it has been suggested that new IBD therapies may involve neutralizing monoclonal antibodies that target these cytokines or their receptors. It is well documented that monoclonal antibodies against IL-12/23 p40 can improve colitis severity in murine models.8586 Consistent with this, neutralizing IL-21 antibody treatment downregulated the infiltration of colonic T cells and the expression of pro-inflammatory cytokines such as IL-6 and IL-17A in inflamed intestinal tissue from mice with DSS-induced colitis.87
Potential strategies for IBD treatment include a blockade of pro-inflammatory cytokines related to Th17 cells. Suppression of IL-17 expression using the oral immunosuppressive drug vidofludimus reduced the proliferation of lymphocytes in vitro. Furthermore, the safety and therapeutic efficacy of vidofludimus was demonstrated in a clinical trial involving IBD patients.8889 However, IL-17 inhibition is controversial in IBD therapy. Although genetic deletion of the IL-17 receptor improved intestinal inflammation in mice with trinitrobenzenesulfonic acid (TNBS)-induced colitis,90 IL-17A deficiency exacerbated the intestinal inflammatory response in mice with DSS-induced colitis.67 In a clinical trial involving CD patients, secukinumab, a monoclonal antibody that neutralizes IL-17A, showed no therapeutic effect.91 A blockade of both IL-17A and IL-17F led to reduced colonic inflammation in experimental colitis; therefore, this may be a potential strategy for IBD therapy.6892
2. Inhibition of Specific Transcription Factors Associated with Th17 Cells
Other potential IBD therapies involve blocking the transcription factors associated with Th17 cells. Therefore, these transcription factors have been intensely studied. Transcription factors that are potential pharmacological targets include RAR-related orphan receptor (ROR)γt and STAT3. These transcription factors are essential for the regulation of Th17 proliferation and function. Indeed, one study found that dual inhibition of nuclear factor (NF)-κB and STAT3 attenuated intestinal inflammation.93 Consistent with this, vidofludimus, which can improve experimental colitis by decreasing IL-17A and IL-17F levels via inhibition of NF-κB and STAT3 activation, had a therapeutic effect in IBD clinical trials.8889 Moreover, pioglitazone, a nuclear receptor peroxisome proliferator-activated receptor γ (PPAR-γ) agonist, decreased Th17 cell differentiation through RORγt and improved DSS-induced colitis.9495 Recently, inhibition of STAT3 activation also had a positive therapeutic effect. In an experimental DSS-induced colitis mouse model, overexpression of the STAT3 inhibitors metformin and gene-associated retinoid-interferon-induced mortality (GRIM)-19 ameliorated intestinal inflammation and Th17 cell differentiation.496
1. Innate Lymphoid Cells
ILCs provide host protective immunity in the mucosal tissues. ILCs are a novel family of effector lymphocytes in IBD that produce IBD-relevant cytokines. ILCs are unique in that they lack antigen-specific receptors and phenotypic markers associated with immune cells but do have a lymphoid morphology.97 The ILC family can be subdivided into 3 subsets based on the types of transcription factors they express for lineage differentiation: ILC1, ILC2, and ILC3. The lineage-specific transcription factors expressed in ILC1, ILC2, and ILC3 are T-bet, GATA-3, and RORγt, respectively.9899100 Cytokines secreted by ILCs are the same as those of T effector cells (Fig. 2).
2. ILCs in Intestinal Innate Immunity
ILC3s, in particular, are involved in host defense to extracellular bacteria and fungi.101102103 ILC3s can be further subdivided based on whether they express the natural cytotoxicity receptor (NCR). NCR+ILC3 produces IL-22; however, NCR-ILC3, similar to Th17 cells, produces both IL-22 and IL-17.104 On the other hand, RORγt-dependent NCR+ILC3 downregulated RORγt expression and subsequently differentiated into T-bet-dependent IFN-γ-producing cells under IL-12 effects.98101105 ILCs that produce IL-17 and IFN-γ are implicated in the pathogenesis of IBD. The pathogenicity of ILC3 was shown in a Helicobacter hepaticus IBD model and a Tbx21−/−Rag2−/− UC model.106107108109 Moreover, innate immune cells isolated from IBD patients expressed ILC3 genes (IL17A , IL22, RORC, and IL23R ).110
The levels of T-bet responsive and IFN-γ-producing ILC1 are also higher in CD patients.111112 IL-12- and IL-15-responsive intraepithelial CD103+NKp46+ILC1 and lamina propria NKp46+ ILC1 were increased in CD patients, and it was suggested that they may have a pathogenic role in the ileum.111112113 Meanwhile, ILC2s may contribute to intestinal fibrosis via IL-13 production in the gut. IL-13 producing CD3-KIR+ cells are more abundant in fibrotic areas of the intestine in CD patients.114 Fibrotic lesions have higher levels of IL-13, IL-13Rα2 and collagen expression than non-fibrotic lesions, which is evidence that ILC2s can also aggravate IBD.114
3. Cytokines
Similar to Th17 cells, pathogenic ILC3s are also responsible for IL-23 production, which induces the secretion of IL-17 and IL-22 by ILC3. TNF-α, a key cytokine in IBD pathogenesis, also increased IL-17 production in ILC3s in a mouse model of colitis.107115 IL-12 stimulates the production of ILC1-specific cytokines in synergy with IL-15 and IL-18.111112 IL-12 and IL-23 can also contribute to differentiation to either ILC1 or ILC3. It seems that ILC differentiation and contribution to IBD pathogenesis is orchestrated by a combination of these cytokines.111
4. Interaction of ILCs with Mucosal Cells
Interactions between ILCs and immune and non-immune cells determine how ILCs respond to the environment (Fig. 3). Crosstalk between ILCs and mucosal, epithelial, and dendritic cells contributes to the host immune response via ILCs. Mononuclear phagocytes have an important role in the activation of ILCs in the intestine. CD14+CX3CR1+ mononuclear phagocytes produce IL-23, IL-1β, IL-6, TNF-α, and TL1A, which promote the activation of ILCs.116117118 CX3CR1+ or CD14+ mononuclear phagocytes mediate ILC3 activation, and this contact is important for ILC3 responsiveness to the gut environment.116119
ILCs also interact with Treg cells, which are important for intestinal immune control. Commensal bacteria-responsive, IL-1β-producing mononuclear phagocytes induce GM-CSG secretion by ILC3s, and these ILC3s produce retinoic acid and TGF-β for Treg cell differentiation.120
With the exception of ILC1, ILC2 and ILC3 express major histocompatibility complex (MHC) class II and can influence CD4+T cells. ILC2 activates Th2 cell differentiation through MHC class II, CD80 and CD86.121 ILC3 that is lacking CD80, CD86, and CD40 cause dysregulated T-cell regulation and increased IL-17 secretion, illustrating the immunoregulatory role of ILC3 in gut T cells.122123124 Interactions between ILCs and B cells promote Ig production T-cells-independently. Thus, B-cell activating factor (BAFF), CD40L and Notch ligand delta-like 1 (DLL1) are increased by ILCs interaction in splenic marginal zone and augments antibody secretion by B1 cells.125 ILC3s also produce IL-10 and express the CCL60 receptor, CCR6, for trafficking to Peyer's patches and the intestinal epithelium. These properties of ILC3 are dependent on IL-22 signaling, because a lack of IL-22 causes a loss in tolerance to commensal bacteria and unchecked growth of pathogenic bacteria, which, together, increase the probability of developing colitis.126127128
Although cytokines secreted by ILCs are very similar to T cells, this new population of cytokines has unique property that expresses both receptors for T cells and NK cells. In IBD patients, ILCs are abundant in inflamed lesions of the intestine; therefore, ILCs have a pathological role and should be considered targets in the development of future IBD therapies. Moreover, ILCs mediate environmental signals for T and B cell development. Therefore, ILCs should be validated in IBD patients and may be key targets to treat IBD.
1. Classical Drugs for IBD Treatment
There are 2 main categories of therapeutics for treating IBD: (1) anti-inflammatories or immunosuppressive agents, and (2) biological agents. Classically, anti-inflammatory drugs, such as 5-aminosalicylates (5-ASAs), are used to treat UC. 5-ASAs are effective for maintenance of remission in UC and also reduce tumor development by inhibiting PG synthesis, reducing pro-inflammatory cytokine levels, blocking neutrophil attraction and activating mast cells by inhibiting NF-κB in immune cells, and promoting PPAR-γ expression and nuclear translocation.129130131132 Unfortunately, 5-ASAs have little efficacy in CD patients.133 Corticosteroids are also used for induction of remission in UC patients. Glucocorticoids inactivate NF-κB, activator protein 1 and prevent the production of inflammatory cytokines such as IL-1 and IL-6.134135136
Azathioprine, methotrexate, and cyclosporine-A are classical immunosuppressive drugs used in IBD therapy. Methotrexate is used for maintenance of remission in CD and cyclosporine-A is used to induce remission state of UC, while azathioprine has efficacy in both CD and UC.137138139140141 Cyclosporine-A and methotrexate suppress the secretion of pro-inflammatory cytokines and induce apoptosis.142143144145 Cyclosporine-A suppresses the production of IL-22 and TNF-α by NCR+ILC3.146
TNF-α is the main pathogenic factor that is produced by immune and non-immune cells in the gut of IBD patients. Anti-TNF agents, including infliximab, adalimumab, and golimumab, are classic IBD therapies. Combination therapy with infliximab and azathioprine is very effective for maintenance of remission in both CD and UC.139147148 However, IBD treatments that involve simply blocking or neutralizing the TNF receptor using incomplete antibodies, such as etanercept, are not effective because such antibodies have a short half-life and low efficacy, consistent with the in vivo results of anti-TNF therapy.149150
2. Inhibition of Lymphoid Cell Homing
In the last decade, clinical trials have shown that 30% to 50% of IBD patients do not respond to anti-TNF therapy. Therefore, various strategies are attempted to target IBD mechanism. Natalizumab, an antibody targeting α4 integrin, is a promising new target for CD therapy and works by blocking T-cell recruitment into lesions via α4β7 integrins, 151152153154 however, risk of progressive multifocal leukoencephalopathy in the use of natalizumab was reported.155156 Some ILC subsets also express α4β7 integrins for homing to the gut.98157 Thus, the more specific α4β7 integrin blocker, vedolizumab, was developed and showed good efficacy in both CD and UC.158159160161162 Vedolizumab also can be used in patients who do not respond to anti-TNF agents.163 Starting with anti-α4β7 therapy, subsequent trials involved the inhibition of T-cell homing using antibodies against MAdCM1 and β7 integrin.164165166
3. Inhibition of IBD-Related Lymphoid Cell Survival
Some IBD therapies target signaling pathways specific to lymphoid cell survival. Sphinogosine-1-phosphate (S1P) and G protein-coupled receptor (S1PR) signaling pathways both activate NF-κB and STAT3, resulting in T-cell proliferation and angiogenesis.167 In clinical trials, ozanimod, a S1PR agonist, induced lymphopenia, suppressed experimental colitis, and induced remission of UC.168169170 Preclinical challenges for pathogenic ILCs have been developed too. In a mouse colitis model, the ILC-specific cytokine, IL-7, controlled ILC3 survival and ILC3-specific cytokine production. Additionally, blockade of IL-7R effectively reduced intestinal ILCs.107146 In other studies, CD90 inhibition depleted ILCs and ameliorated IBD in a mouse model.106158
4. Targeting Epithelial Cells
Intestinal epithelial cells can also be targeted to treat IBD. MMP9 (GS-5746) is an enzyme that induces proteolysis, stimulates the infiltration of immune cells into inflamed gut tissue, and increases TNF-α levels. In a clinical trial, however, targeting MMP9 failed to reduce inflammation in UC patients.171 Repeated cycles of inflammation and healing also induce the formation of tissue fibrosis. Small-interfering RNAs mediated silencing of CHST15, a chondroitin synthetic enzyme, resulted in reduced α-SMA levels in fibroblasts and reduced collagen deposition in animal models of colitis. CHST15-specific small-interfering RNA (STNM01) reduced inflammation and fibrosis in CD clinical trials.172
5. Targeting Cytokines in IBD Therapy
Main cytokines related with IBD were classical targets for IBD treatment. Surprisingly, studies showed that anti-IFN-γ and anti-IL-17A antibody therapies aggravated CD, possibly due to unexpected effects or their protective roles in gut epithelial cells.91173174 IL-6 is one of the main pro-inflammatory cytokines that activates immune cells. The IL-6-targeting antibody tocilizumab (previously known as MRA) showed high efficacy in a colitis model and induced remission in CD patients.175176 Considering the role played by IL-6 signaling in the proliferation of gut epithelial cells, the therapeutic value of anti-IL-6 or anti-gp130 (IL-6R) antibodies should be investigated. The pro-inflammatory cytokines IL-23 (heterodimer of p19 and p40) and IL-12 (heterodimer of p19 and p40) cause inflammation by induction of Th17, Th1 or ILCs development in the gut mucosa of CD patients.177178 Therefore, anti-p40 and p19 antibodies targeting IL-23 and IL-12 have been developed. In clinical trials involving p40 blockers, such as ABT-874 and ustekinumab, a high response was induced in CD patients, even among patients who had anti-TNF therapy.179180 Neutralization of IL-23 (p19) also blocked the stimulation of pathogenic ILC3s and the production of IL-17A- and IL-22-producing cells.106107 Clinical trials are currently investigating the efficacy of p19-targeting drugs (BI655066 and AMG139) in CD patients.181 Other targets in IBD therapy include cytokine-related signaling proteins, including Janus kinases (JAK1-3 and TYK2), which can suppress the secretion of cytokines into the mucosa. In another study, the JAK inhibitor tofacitinib was promising in UC but not CD patients.182183 In contrast, the JAK1-specific inhibitor filgotinib stabilized the remission state in CD patients. Phos phodiesterase-4 inhibition by apremilast negatively regulated cyclic adenosine 3′5′-monophosphate (cAMP), a key mediator of inflammation, by suppressing the pro-inflammatory cytokines IFN-γ, TNF-α, IL-12, IL-17, and IL-23.184
Although the pathogenesis of IBD is complicated and involves many pro-inflammatory mediators, it is clear that Th17 cells play a central role in the induction and maintenance of chronic intestinal inflammation in IBD patients. With regard to reducing intestinal inflammation, research has shown that Th17 cells should be the primary targets for IBD therapy. Many studies have shown that Th17 cells have a pathogenic roles in intestinal inflammation; however, there is still much to be investigated. Therefore, a better understanding of Th17 cells and their targeting could lead to the development of an effective IBD therapy. ILCs should be considered for IBD therapy. ILCs have common therapeutic targets with Th17 cells and are abundant in the gut of IBD patients. Further studies on the role of ILCs in gut immunity would lead to the development of better IBD therapies (Fig. 4).
Beyond innate immunity, adaptive immunity also has a direct role in the pathogenesis of IBD. An overwhelming number of effector cells, such as Th17 cells and ILCs, induce self-destructive immunity; therefore, a cure for IBD would involve understanding how immunological balance is controlled.

FINANCIAL SUPPORT: This study was supported by a grant of the Korean Health Technology R&D Project, Ministry for Health & Welfare, Republic of Korea (HI14C3417).

CONFLICT OF INTEREST: No potential conflict of interest relevant to this article was reported.

  • 1. Bernstein CN, Fried M, Krabshuis JH, et al. Inflammatory bowel disease: a global perspective. Global guidelines. Milwaukee: World Gatroenterology Organization, 2009.
  • 2. Baumgart DC, Sandborn WJ. Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet 2007;369:1641–1657.PMID: 17499606.ArticlePubMed
  • 3. Leppkes M, Becker C, Ivanov II, et al. RORgamma-expressing Th17 cells induce murine chronic intestinal inflammation via redundant effects of IL-17A and IL-17F. Gastroenterology 2009;136:257–267.PMID: 18992745.ArticlePubMed
  • 4. Lee SY, Lee SH, Yang EJ, et al. Metformin ameliorates inflammatory bowel disease by suppression of the STAT3 signaling pathway and regulation of the between Th17/Treg balance. PLoS One 2015;10:e0135858. PMID: 10.1371/journal.pone.0135858. PMID: 26360050.ArticlePubMedPMC
  • 5. Dinarello CA. Interleukin-1beta and the autoinflammatory diseases. N Engl J Med 2009;360:2467–2470.PMID: 19494224.ArticlePubMed
  • 6. McAlindon ME, Hawkey CJ, Mahida YR. Expression of interleukin 1 beta and interleukin 1 beta converting enzyme by intestinal macrophages in health and inflammatory bowel disease. Gut 1998;42:214–219.PMID: 9536946.ArticlePubMedPMC
  • 7. Pizarro TT, Michie MH, Bentz M, et al. IL-18, a novel immunoregulatory cytokine, is up-regulated in Crohn's disease: expression and localization in intestinal mucosal cells. J Immunol 1999;162:6829–6835.PMID: 10352304.ArticlePubMed
  • 8. Dinarello CA. IL-18: a TH1-inducing, proinflammatory cytokine and new member of the IL-1 family. J Allergy Clin Immunol 1999;103:11–24.PMID: 9893178.ArticlePubMed
  • 9. Kanai T, Watanabe M, Okazawa A, et al. Macrophage-derived IL-18-mediated intestinal inflammation in the murine model of Crohn's disease. Gastroenterology 2001;121:875–888.PMID: 11606501.ArticlePubMed
  • 10. Maerten P, Shen C, Colpaert S, et al. Involvement of interleukin 18 in Crohn's disease: evidence from in vitro analysis of human gut inflammatory cells and from experimental colitis models. Clin Exp Immunol 2004;135:310–317.PMID: 14738461.ArticlePubMedPMC
  • 11. Schmitz J, Owyang A, Oldham E, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 2005;23:479–490.PMID: 16286016.ArticlePubMed
  • 12. Beltran CJ, Nunez LE, Diaz-Jimenez D, et al. Characterization of the novel ST2/IL-33 system in patients with inflammatory bowel disease. Inflamm Bowel Dis 2010;16:1097–1107.PMID: 20014018.ArticlePubMed
  • 13. Kobori A, Yagi Y, Imaeda H, et al. Interleukin-33 expression is specifically enhanced in inflamed mucosa of ulcerative colitis. J Gastroenterol 2010;45:999–1007.PMID: 20405148.ArticlePubMed
  • 14. Mitsuyama K, Toyonaga A, Sasaki E, et al. Soluble interleukin-6 receptors in inflammatory bowel disease: relation to circulating interleukin-6. Gut 1995;36:45–49.PMID: 7890234.ArticlePubMedPMC
  • 15. Reinisch W, Gasché C, Tillinger W, et al. Clinical relevance of serum interleukin-6 in Crohn's disease: single point measurements, therapy monitoring, and prediction of clinical relapse. Am J Gastroenterol 1999;94:2156–2164.PMID: 10445543.ArticlePubMed
  • 16. Li Y, de Haar C, Chen M, et al. Disease-related expression of the IL6/STAT3/SOCS3 signalling pathway in ulcerative colitis and ulcerative colitis-related carcinogenesis. Gut 2010;59:227–235.PMID: 19926618.ArticlePubMed
  • 17. Murch SH, Braegger CP, Walker-Smith JA, MacDonald TT. Location of tumour necrosis factor alpha by immunohistochemistry in chronic inflammatory bowel disease. Gut 1993;34:1705–1709.PMID: 8031350.ArticlePubMedPMC
  • 18. Sanchez-Munoz F, Dominguez-Lopez A, Yamamoto-Furusho JK. Role of cytokines in inflammatory bowel disease. World J Gastroenterol 2008;14:4280–4288.PMID: 18666314.ArticlePubMedPMC
  • 19. Li MC, He SH. IL-10 and its related cytokines for treatment of inflammatory bowel disease. World J Gastroenterol 2004;10:620–625.PMID: 14991925.ArticlePubMedPMC
  • 20. Schreiber S, Heinig T, Thiele HG, Raedler A. Immunoregulatory role of interleukin 10 in patients with inflammatory bowel disease. Gastroenterology 1995;108:1434–1444.PMID: 7729636.ArticlePubMed
  • 21. Melgar S, Yeung MM, Bas A, et al. Over-expression of interleukin 10 in mucosal T cells of patients with active ulcerative colitis. Clin Exp Immunol 2003;134:127–137.PMID: 12974765.ArticlePubMedPMC
  • 22. Wang AH, Lam WJ, Han DY, et al. The effect of IL-10 genetic variation and interleukin 10 serum levels on Crohn's disease susceptibility in a New Zealand population. Hum Immunol 2011;72:431–435.PMID: 21354456.ArticlePubMed
  • 23. Nielsen OH, Køppen T, Rüdiger N, Horn T, Eriksen J, Kirman I. Involvement of interleukin-4 and -10 in inflammatory bowel disease. Dig Dis Sci 1996;41:1786–1793.PMID: 8794795.ArticlePubMed
  • 24. Mitsuyama K, Tomiyasu N, Takaki K, et al. Interleukin-10 in the pathophysiology of inflammatory bowel disease: increased serum concentrations during the recovery phase. Mediators Inflamm 2006;2006:26875. PMID: 17392581.ArticlePubMedPMCPDF
  • 25. Ljuca F, Gegic A, Salkic NN, Pavlovic-Calic N. Circulating cytokines reflect mucosal inflammatory status in patients with Crohn's disease. Dig Dis Sci 2010;55:2316–2326.PMID: 19834804.ArticlePubMed
  • 26. Li MO, Flavell RA. TGF-beta: a master of all T cell trades. Cell 2008;134:392–404.PMID: 18692464.ArticlePubMedPMC
  • 27. Del Zotto B, Mumolo G, Pronio AM, Montesani C, Tersigni R, Boirivant M. TGF-beta1 production in inflammatory bowel disease: differing production patterns in Crohn's disease and ulcerative colitis. Clin Exp Immunol 2003;134:120–126.PMID: 12974764.ArticlePubMedPMC
  • 28. Rani R, Smulian AG, Greaves DR, Hogan SP, Herbert DR. TGF-beta limits IL-33 production and promotes the resolution of colitis through regulation of macrophage function. Eur J Immunol 2011;41:2000–2009.PMID: 21469118.ArticlePubMedPMC
  • 29. Gu FM, Li QL, Gao Q, et al. IL-17 induces AKT-dependent IL-6/JAK2/STAT3 activation and tumor progression in hepatocellular carcinoma. Mol Cancer 2011;10:150PMID: 22171994.ArticlePubMedPMC
  • 30. Wruck CJ, Fragoulis A, Gurzynski A, et al. Role of oxidative stress in rheumatoid arthritis: insights from the Nrf2-knockout mice. Ann Rheum Dis 2011;70:844–850.PMID: 21173018.ArticlePubMed
  • 31. Fujino S, Andoh A, Bamba S, et al. Increased expression of interleukin 17 in inflammatory bowel disease. Gut 2003;52:65–70.PMID: 12477762.ArticlePubMedPMC
  • 32. Fort MM, Cheung J, Yen D, et al. IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologies in vivo. Immunity 2001;15:985–995.PMID: 11754819.ArticlePubMed
  • 33. O'Connor W Jr, Kamanaka M, Booth CJ, et al. A protective function for interleukin 17A in T cell-mediated intestinal inflammation. Nat Immunol 2009;10:603–609.PMID: 19448631.ArticlePubMedPMCPDF
  • 34. Kinugasa T, Sakaguchi T, Gu X, Reinecker HC. Claudins regulate the intestinal barrier in response to immune mediators. Gastroenterology 2000;118:1001–1011.PMID: 10833473.ArticlePubMed
  • 35. Sakuraba A, Sato T, Kamada N, Kitazume M, Sugita A, Hibi T. Th1/Th17 immune response is induced by mesenteric lymph node dendritic cells in Crohn's disease. Gastroenterology 2009;137:1736–1745.PMID: 19632232.ArticlePubMed
  • 36. Kleinschek MA, Boniface K, Sadekova S, et al. Circulating and gut-resident human Th17 cells express CD161 and promote intestinal inflammation. J Exp Med 2009;206:525–534.PMID: 19273624.ArticlePubMedPMC
  • 37. Strober W, Zhang F, Kitani A, Fuss I, Fichtner-Feigl S. Proinflammatory cytokines underlying the inflammation of Crohn's disease. Curr Opin Gastroenterol 2010;26:310–317.PMID: 20473158.ArticlePubMedPMC
  • 38. Mahida YR, Ceska M, Effenberger F, Kurlak L, Lindley I, Hawkey CJ. Enhanced synthesis of neutrophil-activating peptide-1/interleukin-8 in active ulcerative colitis. Clin Sci (Lond) 1992;82:273–275.PMID: 1312411.ArticlePubMed
  • 39. Grimm MC, Doe WF. Chemokines in inflammatory bowel disease mucosa: expression of RANTES, macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and gamma-interferon-inducible protein-10 by macrophages, lymphocytes, endothelial cells, and granulomas. Inflamm Bowel Dis 1996;2:88–96.PMID: 23282513.ArticlePubMed
  • 40. Uguccioni M, Gionchetti P, Robbiani DF, et al. Increased expression of IP-10, IL-8, MCP-1, and MCP-3 in ulcerative colitis. Am J Pathol 1999;155:331–336.PMID: 10433925.ArticlePubMedPMC
  • 41. Kaser A, Ludwiczek O, Holzmann S, et al. Increased expression of CCL20 in human inflammatory bowel disease. J Clin Immunol 2004;24:74–85.PMID: 14997037.ArticlePubMed
  • 42. Mitsuyama K, Toyonaga A, Sasaki E, et al. IL-8 as an important chemoattractant for neutrophils in ulcerative colitis and Crohn's disease. Clin Exp Immunol 1994;96:432–436.PMID: 8004812.ArticlePubMedPMC
  • 43. Zenewicz LA, Antov A, Flavell RA. CD4 T-cell differentiation and inflammatory bowel disease. Trends Mol Med 2009;15:199–207.PMID: 19362058.ArticlePubMed
  • 44. Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol 2007;8:942–949.PMID: 17676045.ArticlePubMedPDF
  • 45. Zhou L, Ivanov II, Spolski R, et al. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol 2007;8:967–974.PMID: 17581537.ArticlePubMedPDF
  • 46. Gálvez J. Role of Th17 cells in the pathogenesis of human IBD. ISRN Inflamm 2014;2014:928461. PMID: 25101191.ArticlePubMedPMCPDF
  • 47. Yang XO, Panopoulos AD, Nurieva R, et al. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem 2007;282:9358–9363.PMID: 17277312.ArticlePubMed
  • 48. Lathrop SK, Bloom SM, Rao SM, et al. Peripheral education of the immune system by colonic commensal microbiota. Nature 2011;478:250–254.PMID: 21937990.ArticlePubMedPMCPDF
  • 49. Venema K. Role of gut microbiota in the control of energy and carbohydrate metabolism. Curr Opin Clin Nutr Metab Care 2010;13:432–438.PMID: 20531179.ArticlePubMed
  • 50. Asquith M, Powrie F. An innately dangerous balancing act: intestinal homeostasis, inflammation, and colitis-associated cancer. J Exp Med 2010;207:1573–1577.PMID: 20679404.ArticlePubMedPMC
  • 51. 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;55:205–211.PMID: 16188921.ArticlePubMedPMC
  • 52. Lepage P, Häsler R, Spehlmann ME, et al. Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology 2011;141:227–236.PMID: 21621540.ArticlePubMed
  • 53. Guarner F, Bourdet-Sicard R, Brandtzaeg P, et al. Mechanisms of disease: the hygiene hypothesis revisited. Nat Clin Pract Gastroenterol Hepatol 2006;3:275–284.PMID: 16673007.ArticlePubMedPDF
  • 54. Ivanov II, Atarashi K, Manel N, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009;139:485–498.PMID: 19836068.ArticlePubMedPMC
  • 55. Wu S, Rhee KJ, Albesiano E, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med 2009;15:1016–1022.PMID: 19701202.ArticlePubMedPMCPDF
  • 56. 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;4:337–349.PMID: 18854238.ArticlePubMedPMC
  • 57. Atarashi K, Nishimura J, Shima T, et al. ATP drives lamina propria T(H)17 cell differentiation. Nature 2008;455:808–812.PMID: 18716618.ArticlePubMed
  • 58. Kamada N, Núñez G. Role of the gut microbiota in the development and function of lymphoid cells. J Immunol 2013;190:1389–1395.PMID: 23378581.ArticlePubMedPMC
  • 59. Murphy KM, Stockinger B. Effector T cell plasticity: flexibility in the face of changing circumstances. Nat Immunol 2010;11:674–680.PMID: 20644573.ArticlePubMedPMCPDF
  • 60. Seiderer J, Elben I, Diegelmann J, et al. Role of the novel Th17 cytokine IL-17F in inflammatory bowel disease (IBD): upregulated colonic IL-17F expression in active Crohn's disease and analysis of the IL17F p.His161Arg polymorphism in IBD. Inflamm Bowel Dis 2008;14:437–445.PMID: 18088064.ArticlePubMed
  • 61. Kobayashi T, Okamoto S, Hisamatsu T, et al. IL23 differentially regulates the Th1/Th17 balance in ulcerative colitis and Crohn's disease. Gut 2008;57:1682–1689.PMID: 18653729.ArticlePubMed
  • 62. Raza A, Shata MT. Letter: pathogenicity of Th17 cells may differ in ulcerative colitis compared with Crohn's disease. Aliment Pharmacol Ther 2012;36:204PMID: 22703461.ArticlePubMed
  • 63. Thompson AI, Lees CW. Genetics of ulcerative colitis. Inflamm Bowel Dis 2011;17:831–848.PMID: 21319274.ArticlePubMed
  • 64. Biancheri P, Powell N, Monteleone G, Lord G, MacDonald TT. The challenges of stratifying patients for trials in inflammatory bowel disease. Trends Immunol 2013;34:564–571.PMID: 24035478.ArticlePubMed
  • 65. Ogawa A, Andoh A, Araki Y, Bamba T, Fujiyama Y. Neutralization of interleukin-17 aggravates dextran sulfate sodium-induced colitis in mice. Clin Immunol 2004;110:55–62.PMID: 14962796.ArticlePubMed
  • 66. Garrido-Mesa N, Utrilla P, Comalada M, et al. The association of minocycline and the probiotic Escherichia coli Nissle 1917 results in an additive beneficial effect in a DSS model of reactivated colitis in mice. Biochem Pharmacol 2011;82:1891–1900.PMID: 21930116.ArticlePubMed
  • 67. Yang XO, Chang SH, Park H, et al. Regulation of inflammatory responses by IL-17F. J Exp Med 2008;205:1063–1075.PMID: 18411338.ArticlePubMedPMC
  • 68. Wedebye Schmidt EG, Larsen HL, Kristensen NN, et al. TH17 cell induction and effects of IL-17A and IL-17F blockade in experimental colitis. Inflamm Bowel Dis 2013;19:1567–1576.PMID: 23689808.ArticlePubMed
  • 69. Omenetti S, Pizarro TT. The Treg/Th17 axis: a dynamic balance regulated by the gut microbiome. Front Immunol 2015;6:639PMID: 26734006.ArticlePubMedPMC
  • 70. Brand S. Crohn's disease: Th1, Th17 or both? The change of a paradigm: new immunological and genetic insights implicate Th17 cells in the pathogenesis of Crohn's disease. Gut 2009;58:1152–1167.PMID: 19592695.ArticlePubMed
  • 71. Boden EK, Snapper SB. Regulatory T cells in inflammatory bowel disease. Curr Opin Gastroenterol 2008;24:733–741.PMID: 19125486.ArticlePubMed
  • 72. Wirtz S, Neurath MF. Mouse models of inflammatory bowel disease. Adv Drug Deliv Rev 2007;59:1073–1083.PMID: 17825455.ArticlePubMed
  • 73. Chaudhry A, Samstein RM, Treuting P, et al. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 2011;34:566–578.PMID: 21511185.ArticlePubMedPMC
  • 74. Geem D, Harusato A, Flannigan K, Denning TL. Harnessing regulatory T cells for the treatment of inflammatory bowel disease. Inflamm Bowel Dis 2015;21:1409–1418.PMID: 25793328.ArticlePubMedPMC
  • 75. Boehm F, Martin M, Kesselring R, et al. Deletion of Foxp3+ regulatory T cells in genetically targeted mice supports development of intestinal inflammation. BMC Gastroenterol 2012;12:97PMID: 22849659.ArticlePubMedPMCPDF
  • 76. Huber S, Schramm C, Lehr HA, et al. Cutting edge: TGF-beta signaling is required for the in vivo expansion and immunosuppressive capacity of regulatory CD4+CD25+ T cells. J Immunol 2004;173:6526–6531.PMID: 15557141.ArticlePubMed
  • 77. Rubtsov YP, Rasmussen JP, Chi EY, et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 2008;28:546–558.PMID: 18387831.ArticlePubMed
  • 78. Maloy KJ, Salaun L, Cahill R, Dougan G, Saunders NJ, Powrie F. CD4+CD25+ T(R) cells suppress innate immune pathology through cytokine-dependent mechanisms. J Exp Med 2003;197:111–119.PMID: 12515818.ArticlePubMedPMC
  • 79. Uhlig HH, Coombes J, Mottet C, et al. Characterization of Foxp3+CD4+CD25+ and IL-10-secreting CD4+CD25+ T cells during cure of colitis. J Immunol 2006;177:5852–5860.PMID: 17056509.ArticlePubMedPMC
  • 80. Collins CB, Aherne CM, McNamee EN, et al. Flt3 ligand expands CD103+ dendritic cells and FoxP3+ T regulatory cells, and attenuates Crohn's-like murine ileitis. Gut 2012;61:1154–1162.PMID: 22068168.ArticlePubMed
  • 81. Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006;441:235–238.PMID: 16648838.ArticlePubMedPDF
  • 82. Xu L, Kitani A, Fuss I, Strober W. Cutting edge: regulatory T cells induce CD4+CD25−Foxp3−T cells or are self-induced to become Th17 cells in the absence of exogenous TGF-beta. J Immunol 2007;178:6725–6729.PMID: 17513718.ArticlePubMed
  • 83. de Beaucoudrey L, Puel A, Filipe-Santos O, et al. Mutations in STAT3 and IL-12RB1 impair the development of human IL-17-producing T cells. J Exp Med 2008;205:1543–1550.PMID: 18591412.ArticlePubMedPMC
  • 84. Ogino H, Nakamura K, Ihara E, Akiho H, Takayanagi R. CD4+CD25+ regulatory T cells suppress Th17-responses in an experimental colitis model. Dig Dis Sci 2011;56:376–386.PMID: 20521112.ArticlePubMed
  • 85. Becker C, Dornhoff H, Neufert C, et al. Cutting edge: IL-23 cross-regulates IL-12 production in T cell-dependent experimental colitis. J Immunol 2006;177:2760–2764.PMID: 16920909.ArticlePubMed
  • 86. 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;116:1310–1316.PMID: 16670770.ArticlePubMedPMC
  • 87. Stolfi C, Rizzo A, Franzè E, et al. Involvement of interleukin-21 in the regulation of colitis-associated colon cancer. J Exp Med 2011;208:2279–2290.PMID: 21987656.ArticlePubMedPMC
  • 88. Fitzpatrick LR, Small JS, Doblhofer R, Ammendola A. Vidofludimus inhibits colonic interleukin-17 and improves hapten-induced colitis in rats by a unique dual mode of action. J Pharmacol Exp Ther 2012;342:850–860.PMID: 22691298.ArticlePubMed
  • 89. Herrlinger KR, Diculescu M, Fellermann K, et al. Efficacy, safety and tolerability of vidofludimus in patients with inflammatory bowel disease: the ENTRANCE study. J Crohns Colitis 2013;7:636–643.PMID: 23078909.ArticlePubMedPDF
  • 90. Zhang Z, Zheng M, Bindas J, Schwarzenberger P, Kolls JK. Critical role of IL-17 receptor signaling in acute TNBS-induced colitis. Inflamm Bowel Dis 2006;12:382–388.PMID: 16670527.ArticlePubMed
  • 91. 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;61:1693–1700.PMID: 22595313.ArticlePubMedPMC
  • 92. McLean LP, Cross RK, Shea-Donohue T. Combined blockade of IL-17A and IL-17F may prevent the development of experimental colitis. Immunotherapy 2013;5:923–925.PMID: 23998727.ArticlePubMed
  • 93. Fitzpatrick LR. Inhibition of IL-17 as a pharmacological approach for IBD. Int Rev Immunol 2013;32:544–555.PMID: 23886112.ArticlePubMed
  • 94. Klotz L, Burgdorf S, Dani I, et al. The nuclear receptor PPAR gamma selectively inhibits Th17 differentiation in a T cell-intrinsic fashion and suppresses CNS autoimmunity. J Exp Med 2009;206:2079–2089.PMID: 19737866.ArticlePubMedPMC
  • 95. Hontecillas R, Horne WT, Climent M, et al. Immunoregulatory mechanisms of macrophage PPAR-gamma in mice with experimental inflammatory bowel disease. Mucosal Immunol 2011;4:304–313.PMID: 21068720.ArticlePubMedPDF
  • 96. Kim JK, Lee SH, Lee SY, et al. Grim19 attenuates DSS induced colitis in an animal model. PLoS One 2016;11:e0155853. PMID: 10.1371/journal.pone.0155853. PMID: 27258062.ArticlePubMedPMC
  • 97. Spits H, Cupedo T. Innate lymphoid cells: emerging insights in development, lineage relationships, and function. Annu Rev Immunol 2012;30:647–675.PMID: 22224763.ArticlePubMed
  • 98. Klose CS, Flach M, Möhle L, et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 2014;157:340–356.PMID: 24725403.ArticlePubMed
  • 99. Fallon PG, Ballantyne SJ, Mangan NE, et al. Identification of an interleukin (IL)-25-dependent cell population that provides IL-4, IL-5, and IL-13 at the onset of helminth expulsion. J Exp Med 2006;203:1105–1116.PMID: 16606668.ArticlePubMedPMC
  • 100. Reynders A, Yessaad N, Vu Manh TP, et al. Identity, regulation and in vivo function of gut NKp46+RORgammat+ and NKp46+RORgammat-lymphoid cells. EMBO J 2011;30:2934–2947.PMID: 21685873.ArticlePubMedPMC
  • 101. Satoh-Takayama N, Vosshenrich CA, Lesjean-Pottier S, et al. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity 2008;29:958–970.PMID: 19084435.ArticlePubMed
  • 102. Gladiator A, Wangler N, Trautwein-Weidner K, LeibundGut-Landmann S. Cutting edge: IL-17-secreting innate lymphoid cells are essential for host defense against fungal infection. J Immunol 2013;190:521–525.PMID: 23255360.ArticlePubMed
  • 103. Sonnenberg GF, Monticelli LA, Elloso MM, Fouser LA, Artis D. CD4(+) lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity 2011;34:122–134.PMID: 21194981.ArticlePubMed
  • 104. Takatori H, Kanno Y, Watford WT, et al. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med 2009;206:35–41.PMID: 19114665.ArticlePubMedPMC
  • 105. Sanos SL, Bui VL, Mortha A, et al. RORgammat and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat Immunol 2009;10:83–91.PMID: 19029903.ArticlePubMedPDF
  • 106. Buonocore S, Ahern PP, Uhlig HH, et al. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 2010;464:1371–1375.PMID: 20393462.ArticlePubMedPMCPDF
  • 107. Powell N, Walker AW, Stolarczyk E, et al. The transcription factor T-bet regulates intestinal inflammation mediated by interleukin-7 receptor+ innate lymphoid cells. Immunity 2012;37:674–684.PMID: 23063332.ArticlePubMedPMC
  • 108. Garrett WS, Lord GM, Punit S, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell 2007;131:33–45.PMID: 17923086.ArticlePubMedPMC
  • 109. Garrett WS, Punit S, Gallini CA, et al. Colitis-associated colorectal cancer driven by T-bet deficiency in dendritic cells. Cancer Cell 2009;16:208–219.PMID: 19732721.ArticlePubMedPMC
  • 110. Geremia A, Arancibia-Cárcamo CV, Fleming MP, et al. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J Exp Med 2011;208:1127–1133.PMID: 21576383.ArticlePubMedPMC
  • 111. Bernink JH, Peters CP, Munneke M, et al. Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat Immunol 2013;14:221–229.PMID: 23334791.ArticlePubMedPDF
  • 112. Fuchs A, Vermi W, Lee JS, et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-gamma-producing cells. Immunity 2013;38:769–781.PMID: 23453631.ArticlePubMedPMC
  • 113. Vonarbourg C, Mortha A, Bui VL, et al. Regulated expression of nuclear receptor RORgmamat confers distinct functional fates to NK cell receptor-expressing RORgammat(+) innate lymphocytes. Immunity 2010;33:736–751.PMID: 21093318.ArticlePubMedPMC
  • 114. Bailey JR, Bland PW, Tarlton JF, et al. IL-13 promotes collagen accumulation in Crohn's disease fibrosis by down-regulation of fibroblast MMP synthesis: a role for innate lymphoid cells? PLoS One 2012;7:e52332. PMID: 10.1371/journal.pone.0052332. PMID: 23300643.ArticlePubMedPMC
  • 115. Ermann J, Staton T, Glickman JN, de Waal Malefyt R, Glimcher LH, et al. Nod/Ripk2 signaling in dendritic cells activates IL-17A-secreting innate lymphoid cells and drives colitis in T-bet−/−.Rag2−/− (TRUC) mice. Proc Natl Acad Sci U S A 2014;111:E2559–E2566.PMID: 24927559.ArticlePubMedPMC
  • 116. 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;211:1571–1583.PMID: 25024136.ArticlePubMedPMC
  • 117. 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;118:2269–2280.PMID: 18497880.PubMedPMC
  • 118. Ogino T, Nishimura J, Barman S, et al. Increased Th17-inducing activity of CD14+ CD163 low myeloid cells in intestinal lamina propria of patients with Crohn's disease. Gastroenterology 2013;145:1380–1391.e1.PMID: 23993972.ArticlePubMed
  • 119. Mizuno S, Mikami Y, Kamada N, et al. Cross-talk between RORgammat+ innate lymphoid cells and intestinal macrophages induces mucosal IL-22 production in Crohn's disease. Inflamm Bowel Dis 2014;20:1426–1434.PMID: 24991784.ArticlePubMed
  • 120. Mortha A, Chudnovskiy A, Hashimoto D, et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 2014;343:1249288. PMID: 24625929.ArticlePubMedPMC
  • 121. Neill DR, Wong SH, Bellosi A, et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 2010;464:1367–1370.PMID: 20200518.ArticlePubMedPMCPDF
  • 122. Hepworth MR, Monticelli LA, Fung TC, et al. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature 2013;498:113–117.PMID: 23698371.ArticlePubMedPMCPDF
  • 123. Sonnenberg GF, Fouser LA, Artis D. Border patrol: regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat Immunol 2011;12:383–390.PMID: 21502992.ArticlePubMedPDF
  • 124. Goto Y, Panea C, Nakato G, et al. Segmented filamentous bacteria antigens presented by intestinal dendritic cells drive mucosal Th17 cell differentiation. Immunity 2014;40:594–607.PMID: 24684957.ArticlePubMedPMC
  • 125. Magri G, Miyajima M, Bascones S, et al. Innate lymphoid cells integrate stromal and immunological signals to enhance antibody production by splenic marginal zone B cells. Nat Immunol 2014;15:354–364.PMID: 24562309.ArticlePubMedPMCPDF
  • 126. Cella M, Fuchs A, Vermi W, et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature 2009;457:722–725.PMID: 18978771.ArticlePubMed
  • 127. Goto Y, Obata T, Kunisawa J, et al. Innate lymphoid cells regulate intestinal epithelial cell glycosylation. Science 2014;345:1254009. PMID: 25214634.ArticlePubMedPMC
  • 128. Pham TA, Clare S, Goulding D, et al. Epithelial IL-22RA1-mediated fucosylation promotes intestinal colonization resistance to an opportunistic pathogen. Cell Host Microbe 2014;16:504–516.PMID: 25263220.ArticlePubMedPMC
  • 129. Desreumaux P, Romano O. 5-Aminosalicylates and colorectal cancer: preventive role in chronic inflammatory bowel disease? Gastroenterol Clin Biol 2004;28:509PMID: 15243337.ArticlePubMed
  • 130. Rousseaux C, Lefebvre B, Dubuquoy L, et al. Intestinal antiinflammatory effect of 5-aminosalicylic acid is dependent on peroxisome proliferator-activated receptor-gamma. J Exp Med 2005;201:1205–1215.PMID: 15824083.ArticlePubMedPMC
  • 131. Allgayer H. Review article: mechanisms of action of mesalazine in preventing colorectal carcinoma in inflammatory bowel disease. Aliment Pharmacol Ther 2003;18(Suppl 2): 10–14.PMID: 12950415.ArticlePubMed
  • 132. Velayos FS, Terdiman JP, Walsh JM. Effect of 5-aminosalicylate use on colorectal cancer and dysplasia risk: a systematic review and metaanalysis of observational studies. Am J Gastroenterol 2005;100:1345–1353.PMID: 15929768.ArticlePubMed
  • 133. Lim WC, Wang Y, MacDonald JK, Hanauer S. Aminosalicylates for induction of remission or response in Crohn's disease. Cochrane Database Syst Rev 2016;7:CD008870. PMID: 10.1002/14651858.CD008870. PMID: 27372735.ArticlePubMed
  • 134. Danese S, Fiocchi C. Ulcerative colitis. N Engl J Med 2011;365:1713–1725.PMID: 22047562.ArticlePubMed
  • 135. Oakley RH, Cidlowski JA. The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol 2013;132:1033–1044.PMID: 24084075.ArticlePubMedPMC
  • 136. Rezaie A, Kuenzig ME, Benchimol EI, et al. Budesonide for induction of remission in Crohn's disease. Cochrane Database Syst Rev 2015;(6): CD000296. PMID: 10.1002/14651858.CD000296.pub4. PMID: 26039678.ArticlePubMedPMC
  • 137. Tiede I, Fritz G, Strand S, et al. CD28-dependent Rac1 activation is the molecular target of azathioprine in primary human CD4+ T lymphocytes. J Clin Invest 2003;111:1133–1145.PMID: 12697733.ArticlePubMedPMC
  • 138. D'Haens G, Geboes K, Ponette E, Penninckx F, Rutgeerts P. Healing of severe recurrent ileitis with azathioprine therapy in patients with Crohn's disease. Gastroenterology 1997;112:1475–1481.PMID: 9136824.ArticlePubMed
  • 139. Colombel JF, Sandborn WJ, Reinisch W, et al. Infliximab, azathioprine, or combination therapy for Crohn's disease. N Engl J Med 2010;362:1383–1395.PMID: 20393175.ArticlePubMed
  • 140. Feagan BG, Fedorak RN, Irvine EJ, et al. North American Crohn's Study Group Investigators. A comparison of methotrexate with placebo for the maintenance of remission in Crohn's disease. N Engl J Med 2000;342:1627–1632.PMID: 10833208.ArticlePubMed
  • 141. Feuerstein JD, Akbari M, Tapper EB, Cheifetz AS. Systematic review and meta-analysis of third-line salvage therapy with infliximab or cyclosporine in severe ulcerative colitis. Ann Gastroenterol 2016;29:341–347.PMID: 27366036.ArticlePubMedPMC
  • 142. Steiner S, Daniel C, Fischer A, et al. Cyclosporine A regulates pro-inflammatory cytokine production in ulcerative colitis. Arch Immunol Ther Exp (Warsz) 2015;63:53–63.PMID: 25155925.ArticlePubMed
  • 143. Matsuda S, Koyasu S. Mechanisms of action of cyclosporine. Immunopharmacology 2000;47:119–125.PMID: 10878286.ArticlePubMed
  • 144. Nielsen CH, Albertsen L, Bendtzen K, Baslund B. Methotrexate induces poly(ADP-ribose) polymerase-dependent, caspase 3-independent apoptosis in subsets of proliferating CD4+ T cells. Clin Exp Immunol 2007;148:288–295.PMID: 17286800.ArticlePubMedPMC
  • 145. Wessels JA, Huizinga TW, Guchelaar HJ. Recent insights in the pharmacological actions of methotrexate in the treatment of rheumatoid arthritis. Rheumatology (Oxford) 2008;47:249–255.PMID: 18045808.ArticlePubMedPDF
  • 146. Glatzer T, Killig M, Meisig J, et al. RORgammat+ innate lymphoid cells acquire a proinflammatory program upon engagement of the activating receptor NKp44. Immunity 2013;38:1223–1235.PMID: 23791642.ArticlePubMed
  • 147. Hanauer SB, Feagan BG, Lichtenstein GR, et al. Maintenance infliximab for Crohn's disease: the ACCENT I randomised trial. Lancet 2002;359:1541–1549.PMID: 12047962.ArticlePubMed
  • 148. Panaccione R, Ghosh S, Middleton S, et al. Combination therapy with infliximab and azathioprine is superior to monotherapy with either agent in ulcerative colitis. Gastroenterology 2014;146:392–400.e3.PMID: 24512909.ArticlePubMed
  • 149. Biancheri P, Brezski RJ, Di Sabatino A, et al. Proteolytic cleavage and loss of function of biologic agents that neutralize tumor necrosis factor in the mucosa of patients with inflammatory bowel disease. Gastroenterology 2015;149:1564–1574.e3.PMID: 26170138.ArticlePubMed
  • 150. Van den Brande JM, Koehler TC, Zelinkova Z, et al. Prediction of antitumour necrosis factor clinical efficacy by real-time visualisation of apoptosis in patients with Crohn's disease. Gut 2007;56:509–517.PMID: 17082252.ArticlePubMed
  • 151. Ghosh S, Goldin E, Gordon FH, et al. Natalizumab for active Crohn's disease. N Engl J Med 2003;348:24–32.PMID: 12510039.ArticlePubMed
  • 152. Sandborn WJ, Colombel JF, Enns R, et al. Natalizumab induction and maintenance therapy for Crohn's disease. N Engl J Med 2005;353:1912–1925.PMID: 16267322.ArticlePubMed
  • 153. Targan SR, Feagan BG, Fedorak RN, et al. Natalizumab for the treatment of active Crohn's disease: results of the ENCORE trial. Gastroenterology 2007;132:1672–1683.PMID: 17484865.ArticlePubMed
  • 154. Van Assche G, Van Ranst M, Sciot R, et al. Progressive multifocal leukoencephalopathy after natalizumab therapy for Crohn's disease. N Engl J Med 2005;353:362–368.PMID: 15947080.ArticlePubMed
  • 155. Warnke C, Menge T, Hartung HP, et al. Natalizumab and progressive multifocal leukoencephalopathy: what are the causal factors and can it be avoided? Arch Neurol 2010;67:923–930.PMID: 20697042.ArticlePubMedPMC
  • 156. Bloomgren G, Richman S, Hotermans C, et al. Risk of natalizumab-associated progressive multifocal leukoencephalopathy. N Engl J Med 2012;366:1870–1880.PMID: 22591293.ArticlePubMed
  • 157. Munneke JM, Bjorklund AT, Mjosberg JM, et al. Activated innate lymphoid cells are associated with a reduced susceptibility to graft-versus-host disease. Blood 2014;124:812–821.PMID: 24855210.ArticlePubMed
  • 158. Fischer A, Zundler S, Atreya R, et al. Differential effects of alpha4beta7 and GPR15 on homing of effector and regulatory T cells from patients with UC to the inflamed gut in vivo. Gut 2016;65:1642–1664.PMID: 26209553.ArticlePubMed
  • 159. Feagan BG, Rutgeerts P, Sands BE, et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2013;369:699–710.PMID: 23964932.ArticlePubMed
  • 160. Sandborn WJ, Feagan BG, Rutgeerts P, et al. Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med 2013;369:711–721.PMID: 23964933.ArticlePubMed
  • 161. Wyant T, Leach T, Sankoh S, et al. Vedolizumab affects antibody responses to immunisation selectively in the gastrointestinal tract: randomised controlled trial results. Gut 2015;64:77–83.PMID: 24763133.ArticlePubMed
  • 162. Lam MC, Bressler B. Vedolizumab for ulcerative colitis and Crohn's disease: results and implications of GEMINI studies. Immunotherapy 2014;6:963–971.PMID: 25341118.ArticlePubMed
  • 163. Stallmach A, Langbein C, Atreya R, et al. Vedolizumab provides clinical benefit over 1 year in patients with active inflammatory bowel disease: a prospective multicenter observational study. Aliment Pharmacol Ther 2016;44:1199–1212.PMID: 27714831.ArticlePubMed
  • 164. Zundler S, Schillinger D, Fischer A, et al. Blockade of alpha-Ebeta7 integrin suppresses accumulation of CD8+ and Th9 lymphocytes from patients with IBD in the inflamed gut in vivo. Gut 2017;66:1936–1948.PMID: 27543429.ArticlePubMed
  • 165. Vermeire S, O'Byrne S, Keir M, et al. Etrolizumab as induction therapy for ulcerative colitis: a randomised, controlled, phase 2 trial. Lancet 2014;384:309–318.PMID: 24814090.ArticlePubMed
  • 166. Rutgeerts PJ, Fedorak RN, Hommes DW, et al. Randomised phase I study of etrolizumab (rhuMAb beta7) in moderate to severe ulcerative colitis. Gut 2013;62:1122–1130.PMID: 22717454.ArticlePubMed
  • 167. Gonzalez-Cabrera PJ, Brown S, Studer SM, Rosen H. S1P signaling: new therapies and opportunities. F1000Prime Rep 2014;6:109PMID: 25580263.ArticlePubMedPMC
  • 168. Deguchi Y, Andoh A, Yagi Y, et al. The S1P receptor modulator FTY720 prevents the development of experimental colitis in mice. Oncol Rep 2006;16:699–703.PMID: 16969482.ArticlePubMed
  • 169. Degagné E, Saba JD. S1pping fire: sphingosine-1-phosphate signaling as an emerging target in inflammatory bowel disease and colitis-associated cancer. Clin Exp Gastroenterol 2014;7:205–214.PMID: 25061328.ArticlePubMedPMC
  • 170. Sandborn WJ, Feagan BG, Wolf DC, et al. Ozanimod induction and maintenance treatment for ulcerative colitis. N Engl J Med 2016;374:1754–1762.PMID: 27144850.ArticlePubMed
  • 171. Gilead terminates phase 2/3 study of GS-5745 in patients with ulcerative colitis. Gilead Science, Inc., Accessed July 12, 2017. http://www.gilead.com/news/press-releases/2016/9/gilead-terminates-phase-23-study-of-gs5745-in-patients-with-ulcerative-colitis.
  • 172. Suzuki K, Yokoyama J, Kawauchi Y, et al. Phase 1 clinical study of siRNA targeting carbohydrate sulphotransferase 15 in Crohn's disease patients with active mucosal lesions. J Crohns Colitis 2017;11:221–228.PMID: 27484097.ArticlePubMedPDF
  • 173. Reinisch W, Hommes DW, Van Assche G, et al. A dose escalating, placebo controlled, double blind, single dose and multidose, safety and tolerability study of fontolizumab, a humanised anti-interferon gamma antibody, in patients with moderate to severe Crohn's disease. Gut 2006;55:1138–1144.PMID: 16492717.ArticlePubMedPMC
  • 174. Maxwell JR, Zhang Y, Brown WA, et al. Differential roles for interleukin-23 and interleukin-17 in intestinal immunoregulation. Immunity 2015;43:739–750.PMID: 26431947.ArticlePubMed
  • 175. Atreya R, Mudter J, Finotto S, et al. Blockade of interleukin 6 trans signaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: evidence in Crohn disease and experimental colitis in vivo. Nat Med 2000;6:583–588.PMID: 10802717.ArticlePubMedPDF
  • 176. Ito H, Takazoe M, Fukuda Y, et al. A pilot randomized trial of a human anti-interleukin-6 receptor monoclonal antibody in active Crohn's disease. Gastroenterology 2004;126:989–996.PMID: 15057738.ArticlePubMed
  • 177. 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;12:9–15.PMID: 16374252.ArticlePubMed
  • 178. Monteleone G, Biancone L, Marasco R, et al. Interleukin 12 is expressed and actively released by Crohn's disease intestinal lamina propria mononuclear cells. Gastroenterology 1997;112:1169–1178.PMID: 9098000.ArticlePubMed
  • 179. Sandborn WJ, Feagan BG, Fedorak RN, et al. A randomized trial of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn's disease. Gastroenterology 2008;135:1130–1141.PMID: 18706417.ArticlePubMed
  • 180. Sandborn WJ, Gasink C, Gao LL, et al. Ustekinumab induction and maintenance therapy in refractory Crohn's disease. N Engl J Med 2012;367:1519–1528.PMID: 23075178.ArticlePubMed
  • 181. Kock K, Pan WJ, Gow JM, et al. Preclinical development of AMG 139, a human antibody specifically targeting IL-23. Br J Pharmacol 2015;172:159–172.PMID: 25205227.ArticlePubMed
  • 182. Sandborn WJ, Ghosh S, Panes J, et al. A phase 2 study of tofacitinib, an oral Janus kinase inhibitor, in patients with Crohn's disease. Clin Gastroenterol Hepatol 2014;12:1485–1493.e2.PMID: 24480677.ArticlePubMed
  • 183. Sandborn WJ, Ghosh S, Panes J, et al. Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N Engl J Med 2012;367:616–624.PMID: 22894574.ArticlePubMed
  • 184. Mazur M, Karczewski J, Lodyga M, Żaba R, Adamski Z. Inhibitors of phosphodiesterase 4 (PDE 4): a new therapeutic option in the treatment of psoriasis vulgaris and psoriatic arthritis. J Dermatolog Treat 2015;26:326–328.PMID: 25424050.ArticlePubMed
Fig. 1

Histology of colon tissue from IBD patients. (A) H&E of colon tissue from UC and CD patients (H&E, ×40). (B, C) Signal transducer and activator of transcription 3 (STAT3) and interleukin 17 (IL-17) expression in colon tissue from UC and CD patients (immunohistochemistry, ×200).

ir-16-26-g001.jpg
Fig. 2

Pathogenic innate lymphoid cells (ILCs) and T cells in mucosal cells from IBD patients. ILCs have common properties with T effector cells. Lineage-specific transcription factors expressing ILCs or a subset of T cells produce the same cytokine. Natural cytotoxicity receptor (NCR)-expressing ILCs are classified differently from T cells. T-bet, T-box expressed in T cells; IFN, interferon; TNF, tumor necrosis factor; IL, interleukin; RORγt, retinoic acid receptor-related orphan receptor γt.

ir-16-26-g002.jpg
Fig. 3

Present IBD therapeutic strategies that involve prevention of T cell and innate lymphoid cells (ILC) production or their inhibition. T cells and ILCs have a common therapeutic target. Compared with classical IBD therapeutic agents, new therapeutic strategies may involve T cells; ILCs such as interleukin (IL)-23 and IL-12-, tumor necrosis factor (TNF)-, and integrin-targeting agents; and signal transducer and activator of transcription (STAT) inhibitors. NF, nuclear factor; AP-1, activator protein 1; cAMP, cyclic adenosine 3′:5′-monophosphate.

ir-16-26-g003.jpg
Fig. 4

Reciprocal balance for intestinal immune homeostasis and inflammation. The normal state is mediated by a reciprocal balance between immune cells (Treg and Breg vs. Th17 and ILC1) and cytokines that are secreted to maintain the conditions in the intestine. However, an imbalance in immune cells leads to the destruction of intestinal epithelial cells and the invasion of commensal microbiota. This situation leads to the uncontrolled release of cytokines, which is a key event in the pathogenesis of IBD. Treg, regulatory T; Breg, regulatory B; ILC, innate lymphoid cells; DC, dendritic cell; IL, interleukin; NKT, natural killer T; Th17, T helper 17.

ir-16-26-g004.jpg

Figure & Data

REFERENCES

    Citations

    Citations to this article as recorded by  
    • Zinc and Inflammatory Bowel Disease: From Clinical Study to Animal Experiment
      Xi Peng, Yingxiang Yang, Rao Zhong, Yuexuan Yang, Fang Yan, Na Liang, Shibin Yuan
      Biological Trace Element Research.2025; 203(2): 624.     CrossRef
    • Macadamia oil alleviates dextran sulfate sodium-induced ulcerative colitis in mice via activating the Nrf2/Ho-1 pathway
      Chunlan Shan, Ting Liu, Fujun Miao, Gangjun Guo
      Food Science and Biotechnology.2025; 34(4): 1027.     CrossRef
    • Unleashing the potential of extracellular vesicles for ulcerative colitis and Crohn's disease therapy
      George Chigozie Njoku, Cathal Patrick Forkan, Fumie Mitani Soltysik, Peter Lindberg Nejsum, Flemming Pociot, Reza Yarani
      Bioactive Materials.2025; 45: 41.     CrossRef
    • Sanguinarine chloride hydrate mitigates colitis symptoms in mice through the regulation of the intestinal microbiome and metabolism of short-chain fatty acids
      Jige Xin, Lin He, Yanlin Li, Qiqi Pu, Xuan Du, Fuze Ban, Diangang Han
      Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease.2025; 1871(2): 167579.     CrossRef
    • IgG4 in the gut: Gastrointestinal IgG4-related disease or a new subtype of inflammatory bowel disease
      Sarah Bencardino, Cosimo Simone Matichecchia, Jacopo Fanizza, Laurent Peyrin-Biroulet, Emanuel Della-Torre, Silvio Danese, Ferdinando D’Amico
      Autoimmunity Reviews.2025; 24(2): 103720.     CrossRef
    • Novel targets for mucosal healing in inflammatory bowel disease therapy
      Pardis Mansouri, Pegah Mansouri, Esmaeil Behmard, Sohrab Najafipour, Amin Kouhpayeh, Akbar Farjadfar
      International Immunopharmacology.2025; 144: 113544.     CrossRef
    • Decursinol angelate relieves inflammatory bowel disease by inhibiting the ROS/TXNIP/NLRP3 pathway and pyroptosis
      Yudi Wang, Jiamin Wang, Yonghu Chen, Xuezheng Li, Zhe Jiang
      Frontiers in Pharmacology.2025;[Epub]     CrossRef
    • Phillyrin ameliorates DSS-induced colitis in mice via modulating the gut microbiota and inhibiting the NF-κB/MLCK pathway
      Tong Li, Guiqiu Hu, Shoupeng Fu, Di Qin, Zheyu Song, Cassio Almeida-da-Silva
      Microbiology Spectrum.2025;[Epub]     CrossRef
    • Joint analysis of genome‐wide cross‐trait and multi‐omics reveals molecular mechanisms of inflammatory bowel disease and nominates its novel therapeutic genes
      Zijun Zhu, Rongxing Wei, Hailong Li, Xin Wang, Guanzhi He, Meiyu Du, Senwei Tan, Liang Cheng
      The FASEB Journal.2025;[Epub]     CrossRef
    • Association between allergic diseases and mucosal healing in ulcerative colitis
      Yasunori Yamamoto, Shinya Furukawa, Osamu Yoshida, Teruki Miyake, Kana Shiraishi, Yu Hashimoto, Kazuhiro Tange, Shogo Kitahata, Tomoyuki Ninomiya, Sen Yagi, Hanayama Masakazu, Seiyuu Suzuki, Naozumi Shibata, Hidehiro Murakami, Katsuhisa Ohashi, Hideomi To
      Scientific Reports.2025;[Epub]     CrossRef
    • Unfolded protein response: An essential element of intestinal homeostasis and a potential therapeutic target for inflammatory bowel disease
      Miriam Di Mattia, Michele Sallese, Loris Riccardo Lopetuso
      Acta Physiologica.2025;[Epub]     CrossRef
    • Targeting gut microbiota dysbiosis in inflammatory bowel disease: a systematic review of current evidence
      Asmaa Farah, Pradipta Paul, Anfal Sher Khan, Aparajita Sarkar, Sa’ad Laws, Ali Chaari
      Frontiers in Medicine.2025;[Epub]     CrossRef
    • Tissue factor-dependent colitogenic CD4+ T cell thrombogenicity is regulated by activated protein C signalling
      Gemma Leon, Paula A. Klavina, Aisling M. Rehill, Sarah E. J. Cooper, Anna Dominik, Shrikanth Chomanahalli Basavarajappa, James S. O’Donnell, Seamus Hussey, Patrick T. Walsh, Roger J. S. Preston
      Nature Communications.2025;[Epub]     CrossRef
    • Exploring the potential active components and mechanisms of Tetrastigma hemsleyanum against ulcerative colitis based on network pharmacology in LPS-induced RAW264.7 cells
      Qiang Zhang, Tinghui Feng, Qinxiang Chang, Dongfeng Yang, Yuan li, Yujie Shang, Wenxin Gao, Jiayan Zhao, Xiaohu Li, Lei Ma, Zongsuo Liang
      Journal of Ethnopharmacology.2025; 342: 119384.     CrossRef
    • Cardiovascular disease: extraintestinal manifestation of inflammatory bowel disease
      Samridhi Lakhanpal, Kanishk Aggarwal, Harmanjit Kaur, Kunal Kanwar, Vasu Gupta, Jill Bhavsar, Rohit Jain
      Intestinal Research.2025; 23(1): 23.     CrossRef
    • Beyond the skin: endocrine, psychological and nutritional aspects in women with hidradenitis suppurativa
      Anna Dattolo, Monica Torres, Evelyn Frias-Toral, Alessia Paganelli, Mariana Zhang, Stefania Madonna, Laura Mercurio, Gabriela Cucalón, Federico Garbarino, Cristina Albanesi, Emanuele Scala
      Journal of Translational Medicine.2025;[Epub]     CrossRef
    • The role of efferocytosis in inflammatory bowel disease
      Yi-Qian Liu, Zhan-Zhan Li, Yong-Li Han, Qing-Bo Wang
      Frontiers in Immunology.2025;[Epub]     CrossRef
    • Network pharmacology and experimental verification in vivo reveal the mechanism of Zhushao Granules against ulcerative colitis
      Benjiao Gong, Chenglin Zhang, Shaofei Hu, Xueying Zhang, Hui Zou, Jiayao Li, Jiahui Wang, Yanlei Kao, Fujun Liu
      Biological Procedures Online.2025;[Epub]     CrossRef
    • The role of BATF in immune cell differentiation and autoimmune diseases
      Xiaomeng Wang, Yue Hong, Jinmei Zou, Bo Zhu, Chao Jiang, Liwei Lu, Jie Tian, Jing Yang, Ke Rui
      Biomarker Research.2025;[Epub]     CrossRef
    • Progressing advanced therapies for inflammatory bowel disease: Current status including dual biologic therapy and discontinuation of biologics
      Kazuo Yashima, Hiroki Kurumi, Naoyuki Yamaguchi, Hajime Isomoto
      Expert Review of Gastroenterology & Hepatology.2025; 19(3): 291.     CrossRef
    • Anti-inflammatory agents design via the fragment hybrid strategy in the discovery of compound c1 for treating ALI and UC
      Mi Guo, Yu Zou, Ke Dong, Nan Huang, Zhichao Chen, Chenhui Sun, Pan Chen, Qi Chen, Luxiao Zhu, Yuehua Lv, Kaixin Zhang, Miao Jiang, Yitian Gao, Young-Chang Cho, Qidong Tang, Guang Liang, Di Wu
      European Journal of Medicinal Chemistry.2025; 289: 117431.     CrossRef
    • Immunomodulatory effects and clinical application of exosomes derived from mesenchymal stem cells
      Yang-Fei Yi, Zi-Qi Fan, Can Liu, Yi-Tong Ding, Yao Chen, Jie Wen, Xiao-Hong Jian, Yu-Fei Li
      World Journal of Stem Cells.2025;[Epub]     CrossRef
    • The gut-skin axis: a bi-directional, microbiota-driven relationship with therapeutic potential
      Maira Jimenez-Sanchez, Larissa S. Celiberto, Hyungjun Yang, Ho Pan Sham, Bruce A. Vallance
      Gut Microbes.2025;[Epub]     CrossRef
    • Ginsenoside Rg1 alleviated experimental colitis in obesity mice by regulating memory follicular T cells via Bcl-6/Blimp-1 pathway
      Zeyun Zhang, Jiaqi Huang, Xiyan Zhu, Bailin Deng, Haimei Zhao, Haiyan Wang, Duanyong Liu
      The Journal of Nutritional Biochemistry.2025; 140: 109880.     CrossRef
    • A systematic review on the role of gut microbiome in inflammatory bowel disease: Spotlight on virome and plant metabolites
      Md. Mizanur Rahaman, Phurpa Wangchuk, Subir Sarker
      Microbial Pathogenesis.2025; 205: 107608.     CrossRef
    • Cholesteryl Ester Species but Not Serum Proprotein Convertase Subtilisin/Kexin Type 9 Levels Decline in Male Patients with Active Inflammatory Bowel Disease
      Angelika Hettenbach, Tanja Elger, Muriel Huss, Gerhard Liebisch, Marcus Höring, Johanna Loibl, Arne Kandulski, Martina Müller, Hauke Christian Tews, Christa Buechler
      Pathophysiology.2025; 32(2): 13.     CrossRef
    • Glycyrrhiza uralensis extract supplementation mitigated the negative effects of prolonged low-dose exposure to Deoxynivalenol and Zearalenone on growth performance and intestinal health of broiler chickens
      Yan Chen, Guohua Zhang, Jiawei Li, Ximei Li, Susu Jiang, Yingpai Zha Xi, Yanli Guo, Jianxiong Lu
      Frontiers in Veterinary Science.2025;[Epub]     CrossRef
    • Serum Maresin-1 and Resolvin-D1 Levels as Non-Invasive Biomarkers for Monitoring Disease Activity in Ulcerative Colitis
      Selim Demirci, Semih Sezer
      Diagnostics.2025; 15(7): 834.     CrossRef
    • Resveratrol targeting MDM2/P53/PUMA axis to inhibit colonocyte apoptosis in DSS-induced ulcerative colitis mice
      Rui Tang, Ling Jiang, Quan Ji, Pengyuan Kang, Yuan Liu, Pengyu Miao, Xiaofan Xu, Mingxi Tang
      Frontiers in Pharmacology.2025;[Epub]     CrossRef
    • Predictors of Immunogenicity and Loss of Response to ANTI-TNFα Therapy in Crohn Disease—A Systematic Review
      Simona Grad, Radu A. Farcas, Dan L. Dumitrascu, Teodora Surdea-Blaga, Abdulrahman Ismaiel, Stefan Popa
      American Journal of Therapeutics.2025; 32(3): e262.     CrossRef
    • Prevalence and clinical characteristics of children with coexisting coeliac disease and inflammatory bowel disease
      Christopher Bakewell, James John Ashton, Bhumita Vadgama, R Mark Beattie, Akshay Batra
      Archives of Disease in Childhood.2025; : archdischild-2025-328470.     CrossRef
    • Features of interleukin-17 and interleukin-23 expression in patients with inflammatory bowel diseases
      Yu.M. Stepanov, M.V. Stoikevych, O.M. Tatarchuk, T.S. Tarasova
      GASTROENTEROLOGY.2025; 59(1): 44.     CrossRef
    • From traditional to artificial intelligence-driven approaches: Revolutionizing personalized and precision nutrition in inflammatory bowel disease
      Simone Baldi, Dilara Sarikaya, Sofia Lotti, Francesca Cuffaro, Dorian Fink, Barbara Colombini, Francesco Sofi, Amedeo Amedei
      Clinical Nutrition ESPEN.2025; 68: 106.     CrossRef
    • Particulate matter-shaped Th17 cell plasticity impairs the colonic mucus layer
      Bin Li, Nannan Huang, Jing Wang, Xianan Zhang, Qingtao Meng, Shenshen Wu, Rui Chen
      Ecotoxicology and Environmental Safety.2025; 296: 118219.     CrossRef
    • Biosimilars versus biological therapy in inflammatory bowel disease: challenges and targeting strategies using drug delivery systems
      Ahmed Aljabri, Ghareb M. Soliman, Yasmin N. Ramadan, Mohammed A. Medhat, Helal F. Hetta
      Clinical and Experimental Medicine.2025;[Epub]     CrossRef
    • Bone Broth Benefits: How Its Nutrients Fortify Gut Barrier in Health and Disease
      Ayah Matar, Nada Abdelnaem, Michael Camilleri
      Digestive Diseases and Sciences.2025;[Epub]     CrossRef
    • Chlorpyrifos Inhibits Intestinal Stem Cell Proliferation and Differentiation at the Acceptable Daily Intake and Disrupts Immune Responses at High Doses
      Xinlei Yuan, Fang Wu, Le Cheng, Tengteng Ji, Chenyan Zheng, Yumeng Ma, Yutong Jin, Jianguo Dong, Yan Jin, Bing Fang
      Journal of Agricultural and Food Chemistry.2025; 73(20): 12455.     CrossRef
    • Immune modulation for the patterns of epithelial cell death in inflammatory bowel disease
      Yuting Jiang, Jie Chen, Yaoyao Du, Minwei Fan, Lan Shen
      International Immunopharmacology.2025; 154: 114462.     CrossRef
    • Comprehensive computational strategies for multi-target drug discovery in inflammatory bowel disease utilizing bioactive compounds
      Pardis Mansouri, Pegah Mansouri, Sohrab Najafipour, Seyed Amin Kouhpayeh, Akbar Farjadfar, Esmaeil Behmard
      Scientific Reports.2025;[Epub]     CrossRef
    • Gut microbiome and lung cancer: mechanisms, interactions, and dietary interventions
      Jayashri Chakraborty, Ngashepam Lanchenba Singh, Bhrigu Kumar Das
      Gut Microbes Reports.2025;[Epub]     CrossRef
    • Anti-inflammatory effects and gut microbiota modulation of synbiotic mulberry in DSS-induced colitis rats
      Atcharaporn Ontawong, Arthid Thim-Uam, Sirinat Pengnet, Narongsuk Munkong, Pairote Wongputtisin, Kullanat Kuntakhut, Prathakphong Riyamongkhol, Dej Mann, Doungporn Amornlerdpison
      Molecular and Cellular Biochemistry.2025;[Epub]     CrossRef
    • A critical review on inflammatory bowel diseases risk factors, dietary nutrients regulation and protective pathways based on gut microbiota during recent 5 years
      Pengkui Xia, Tao Hou, Hong Jin, Yaqi Meng, Jing Li, Fuchao Zhan, Fang Geng, Bin Li
      Critical Reviews in Food Science and Nutrition.2024; 64(24): 8805.     CrossRef
    • Safety and Effectiveness of Vedolizumab in Elderly Patients with Inflammatory Bowel Disease
      Dushyant Singh Dahiya, Saurabh Chandan, Jay Bapaye, Babu P. Mohan, Daryl Ramai, Lena L. Kassab, Ojasvini C. Chandan, Parambir S. Dulai, Gursimran S. Kochhar
      Journal of Clinical Gastroenterology.2024; 58(4): 378.     CrossRef
    • Real-World Experience of Adalimumab Biosimilar (ABP 501) Use in Patients with Inflammatory Bowel Disease in Europe
      Ran Jin, Chidozie Nduka, Delphine Courmier, Hannah Knight, Rachael Meadows, James Piercy, J. R. F. Cummings, Waldemar Radziszewski
      Advances in Therapy.2024; 41(1): 331.     CrossRef
    • Gut-brain axis interacts with immunomodulation in inflammatory bowel disease
      Xianglu Ye, Miao Zhang, Ning Zhang, Hai Wei, Bing Wang
      Biochemical Pharmacology.2024; 219: 115949.     CrossRef
    • Unraveling the gut-Lung axis: Exploring complex mechanisms in disease interplay
      Mariam Wed Eladham, Balachandar Selvakumar, Narjes Saheb Sharif-Askari, Fatemeh Saheb Sharif-Askari, Saleh Mohamed Ibrahim, Rabih Halwani
      Heliyon.2024; 10(1): e24032.     CrossRef
    • Gut-on-chip devices as intestinal inflammation models and their future for studying multifactorial diseases
      Susanne Taavitsainen, Kati Juuti-Uusitalo, Kalle Kurppa, Katri Lindfors, Pasi Kallio, Minna Kellomäki
      Frontiers in Lab on a Chip Technologies.2024;[Epub]     CrossRef
    • Pathogenesis and precision medicine for predicting response in inflammatory bowel disease: advances and future directions
      Robert D Little, Thisun Jayawardana, Sabrina Koentgen, Fan Zhang, Susan J Connor, Alex Boussioutas, Mark G Ward, Peter R Gibson, Miles P Sparrow, Georgina L Hold
      eGastroenterology.2024; 2(1): e100006.     CrossRef
    • Burden of Anxiety, Depression and Perceived Stress in Patients with Inflammatory Bowel Disease: A Cohort Study from North India
      Arshdeep Singh, Arshia Bhardwaj, Ashish Tripathi, Mukesh Kumar Ranjan, Dharmatma Singh, Ashi Sachdeva, Mahima Marwah, Kriti Sood Sadana, Namita Bansal, Ramit Mahajan, Kirandeep Kaur, Vandana Midha, Ajit Sood
      Digestive Diseases and Sciences.2024; 69(3): 775.     CrossRef
    • SARS-CoV-2 vaccination in inflammatory bowel disease patients is not associated with flares: a retrospective single-centre Swiss study
      Laura N. Rossier, Natalie P. Décosterd, Christoph B. Matter, Dominic A. Staudenmann, André Moser, Bernhard Egger, Frank W. Seibold
      Annals of Medicine.2024;[Epub]     CrossRef
    • Chensinin-1b Alleviates DSS-Induced Inflammatory Bowel Disease by Inducing Macrophage Switching from the M1 to the M2 Phenotype
      Yue Sun, Huiyu Li, Xingpeng Duan, Xiaoxiao Ma, Chenxi Liu, Dejing Shang
      Biomedicines.2024; 12(2): 345.     CrossRef
    • Boswellic Acids: A Critical Appraisal of Their Therapeutic and Nutritional Benefits in Chronic Inflammatory Diseases
      Neeta Solanki, Gaurav Gupta, Dinesh Kumar Chellappan, Sachin Kumar Singh, Monica Gulati, Keshav Raj Paudel, Philip M Hansbro, Kamal Dua, Suraj Bhan, Manisha Saini, Harish Dureja
      Endocrine, Metabolic & Immune Disorders - Drug Targets.2024; 24(1): 116.     CrossRef
    • Exploring Chinese herbal medicine for the treatment of inflammatory bowel disease: A comprehensive overview
      Jeetendra Kumar Gupta, Aniruddh Pratap Singh, Yati Sharma
      Pharmacological Research - Modern Chinese Medicine.2024; 10: 100380.     CrossRef
    • Silent information regulator 2 deficiency exacerbates chronic cold exposure-induced colonic injury and p65 activation in mice
      Jingru Guo, Huaixiu Zhang, Huijie Hu, Tianrui Zhao, Hong Ji, Li Ma, Jingjing Lu, Jianbin Yuan, Bin Xu
      Gene.2024; 907: 148276.     CrossRef
    • Identified S100A9 as a target for diagnosis and treatment of ulcerative colitis by bioinformatics analysis
      Lulu Tan, Xin Li, Hong Qin, Qingqing Zhang, Jinfeng Wang, Tao Chen, Chengwu Zhang, Xiaoying Zhang, Yuyan Tan
      Scientific Reports.2024;[Epub]     CrossRef
    • Design, synthesis, in vitro and in vivo biological evaluation of pterostilbene derivatives for anti-inflammation therapy
      Liuzeng Chen, Ke Wang, Xiaohan Liu, Lifan Wang, Hui Zou, Shuying Hu, Lingling Zhou, Rong Li, Shiying Cao, Banfeng Ruan, Quanren Cui
      Journal of Enzyme Inhibition and Medicinal Chemistry.2024;[Epub]     CrossRef
    • Long noncoding RNAs and circular RNAs as potential diagnostic biomarkers of inflammatory bowel diseases: a systematic review and meta-analysis
      Melaku Ashagrie Belete, Selamyhun Tadesse, Mihret Tilahun, Alemu Gedefie, Agumas Shibabaw, Zewudu Mulatie, Muluken Amare Wudu, Saba Gebremichael, Habtu Debash, Mihreteab Alebachew, Ermiyas Alemayehu
      Frontiers in Immunology.2024;[Epub]     CrossRef
    • Peripheral and intestinal T lymphocyte subsets in dogs with chronic inflammatory enteropathy
      Beatriz Agulla, Alejandra Villaescusa, Ángel Sainz, David Díaz‐Regañón, Fernando Rodríguez‐Franco, Lydia Calleja‐Bueno, Patricia Olmeda, Mercedes García‐Sancho
      Journal of Veterinary Internal Medicine.2024; 38(3): 1437.     CrossRef
    • Review of Animal Models of Colorectal Cancer in Different Carcinogenesis Pathways
      Xue Chen, Yirong Ding, Yun Yi, Zhishan Chen, Jiaping Fu, Ying Chang
      Digestive Diseases and Sciences.2024; 69(5): 1583.     CrossRef
    • Association between inflammatory bowel disease and cancer risk: evidence triangulation from genetic correlation, Mendelian randomization, and colocalization analyses across East Asian and European populations
      Di Liu, Meiling Cao, Haotian Wang, Weijie Cao, Chenguang Zheng, Yun Li, Youxin Wang
      BMC Medicine.2024;[Epub]     CrossRef
    • Effects of Shenling Baizhu Powder on Intestinal Short-Chain Fatty Acid Content and Intestinal Protection in Rats with Inflammatory Bowel Disease
      可心 崔
      Advances in Clinical Medicine.2024; 14(03): 1930.     CrossRef
    • Tissue-specific reprogramming leads to angiogenic neutrophil specialization and tumor vascularization in colorectal cancer
      Triet M. Bui, Lenore K. Yalom, Edward Ning, Jessica M. Urbanczyk, Xingsheng Ren, Caroline J. Herrnreiter, Jackson A. Disario, Brian Wray, Matthew J. Schipma, Yuri S. Velichko, David P. Sullivan, Kouki Abe, Shannon M. Lauberth, Guang-Yu Yang, Parambir S. D
      Journal of Clinical Investigation.2024;[Epub]     CrossRef
    • Discontinuation of therapy in inflammatory bowel disease: Current views
      Antonio Meštrović, Marko Kumric, Josko Bozic
      World Journal of Clinical Cases.2024; 12(10): 1718.     CrossRef
    • Colonic Dysregulation of Major Metabolic Pathways in Experimental Ulcerative Colitis
      Ji Yeon Noh, Naser Farhataziz, Michael T. Kinter, Xin Yan, Yuxiang Sun
      Metabolites.2024; 14(4): 194.     CrossRef
    • Cirsimaritin Alleviates Dextran Sodium Sulfate-Induced Acute Colitis in Experimental Animals: A Therapeutic Approach for Inflammatory Bowel Disease
      Abdelrahim Alqudah, Esam Qnais, Omar Gammoh, Yousra Bseiso, Mohammed Wedyan, Mohammad Alqudah, Taher Hatahet
      Preventive Nutrition and Food Science.2024; 29(1): 31.     CrossRef
    • Potential shared pathogenic mechanisms between endometriosis and inflammatory bowel disease indicate a strong initial effect of immune factors
      Haolong Zhang, Yaxin Mo, Ling Wang, Haoling Zhang, Sen Wu, Doblin Sandai, Ahmad Naqib Shuid, Xingbei Chen
      Frontiers in Immunology.2024;[Epub]     CrossRef
    • Level of interleukin 17 in inflammatory bowel disease and its relation with disease activity
      Ayman Menesy, Mohamed Hammad, Salah Aref, Fatma Adel Mourad Abozeid
      BMC Gastroenterology.2024;[Epub]     CrossRef
    • Pregnancy period and early-life risk factors for inflammatory bowel disease: a Northern Finland birth cohort 1966 study
      Timo M. Blomster, Olli-Pekka Koivurova, Ritva Koskela, Karl-Heinz Herzig, Nicholas J. Talley, Jukka Ronkainen
      BMC Public Health.2024;[Epub]     CrossRef
    • Anti-inflammatory and protective effects of Aripiprazole on TNBS-Induced colitis and associated depression in rats: Role of kynurenine pathway
      Afrooz Mohammadgholi-Beiki, Mohammad Sheibani, Majid Jafari-Sabet, Manijeh Motevalian, Parvaneh Rahimi-Moghaddam
      International Immunopharmacology.2024; 133: 112158.     CrossRef
    • Serum Galectin-3 as a Non-Invasive Marker for Primary Sclerosing Cholangitis
      Ganimete Bajraktari, Tanja Elger, Muriel Huss, Johanna Loibl, Andreas Albert, Arne Kandulski, Martina Müller, Hauke Christian Tews, Christa Buechler
      International Journal of Molecular Sciences.2024; 25(9): 4765.     CrossRef
    • Take vedolizumab home: transition from intravenous to subcutaneous treatment
      Kaituo Huang, Lingya Yao, Jing Liu, Qian Cao
      Therapeutic Advances in Chronic Disease.2024;[Epub]     CrossRef
    • Urinary soluble CD163 is a putative non-invasive biomarker for primary sclerosing cholangitis
      Tanja Elger, Tanja Fererberger, Muriel Huss, Stefanie Sommersberger, Patricia Mester, Petra Stoeckert, Stefan Gunawan, Gerhard Liebisch, Johanna Loibl, Arne Kandulski, Martina Müller, Christa Buechler, Hauke Christian Tews
      Experimental and Molecular Pathology.2024; 137: 104900.     CrossRef
    • Bone marrow mesenchymal stromal cells support regeneration of intestinal damage in a colitis mouse model, independent of their CXCR4 expression
      Burcu Pervin, Merve Gizer, Mehmet Emin Şeker, Özgür Doğuş Erol, Sema Nur Gür, Ece Gizem Polat, Bahar Değirmenci, Petek Korkusuz, Fatima Aerts‐Kaya
      Clinical and Translational Science.2024;[Epub]     CrossRef
    • Identification and Molecular Mechanism of Anti-inflammatory Peptides Isolated from Jack Bean Protein Hydrolysates: in vitro Studies with Human Intestinal Caco-2BBe Cells
      Bambang Dwi Wijatniko, Yoshinari Yamamoto, Makoto Hirayama, Takuya Suzuki
      Plant Foods for Human Nutrition.2024; 79(3): 624.     CrossRef
    • Mesenchymal Stromal Cells: New Generation Treatment of Inflammatory Bowel Disease
      Shulin Wei, Mingxing Li, Qin Wang, Yueshui Zhao, Fukuan Du, Yu Chen, Shuai Deng, Jing Shen, Ke Wu, Jiayue Yang, Yuhong Sun, Li Gu, Xiaobing Li, Wanping Li, Meijuan Chen, Xiao Ling, Lei Yu, Zhangang Xiao, Lishu Dong, Xu Wu
      Journal of Inflammation Research.2024; Volume 17: 3307.     CrossRef
    • From Gut to Brain: Uncovering Potential Serum Biomarkers Connecting Inflammatory Bowel Diseases to Neurodegenerative Diseases
      Oliviu-Florentiu Sarb, Adriana-Daniela Sarb, Maria Iacobescu, Irina-Maria Vlad, Mircea-Vasile Milaciu, Lorena Ciurmarnean, Vitalie Vacaras, Alina-Ioana Tantau
      International Journal of Molecular Sciences.2024; 25(11): 5676.     CrossRef
    • Impact of high-salt diet in health and diseases and its role in pursuit of cancer immunotherapy by modulating gut microbiome
      Yuvaraj Balan, Raja Sundaramurthy, Archana Gaur, Sakthivadivel Varatharajan, Gerard Marshall Raj
      Journal of Family Medicine and Primary Care.2024; 13(5): 1628.     CrossRef
    • Integrated analyses reveal the diagnostic and predictive values of COL5A2 and association with immune environment in Crohn’s disease
      Tingting Zhong, Xiaoqing Cheng, Qianru Gu, Guoxiang Fu, Yihong Wang, Yujie Jiang, Jiaqi Xu, Zhinong Jiang
      Genes & Immunity.2024; 25(3): 209.     CrossRef
    • Protection effect of Dioscoreae Rhizoma against ethanol-induced gastric injury in vitro and in vivo: A phytochemical and pharmacological study
      Yujun Xie, Luyao An, Xiaoyan Wang, Yajie Ma, Alamusi Bayoude, Xinxin Fan, Boyang Yu, Renshi Li
      Journal of Ethnopharmacology.2024; 333: 118427.     CrossRef
    • Co-delivery of a curcumin and asafoetida as a bioavailable complex using fenugreek galactomannan hydrogel scaffold alleviates inflammatory bowel disease on experimental animals
      Krishnapriya Anil, Svenia P. Jose, Syam Das S, A. Abdul Vahab
      Pharmacological Research - Natural Products.2024; 4: 100071.     CrossRef
    • The roles of Th cells in myocardial infarction
      Jun Liu, Feila Liu, Tingting Liang, Yue Zhou, Xiaohan Su, Xue Li, Jiao Zeng, Peng Qu, Yali Wang, Fuli Chen, Qian Lei, Gang Li, Panke Cheng
      Cell Death Discovery.2024;[Epub]     CrossRef
    • Monitoring immunE DysregulAtion foLLowing Immune checkpOint-inhibitioN (MEDALLION): protocol for an observational cancer immunotherapy cohort study
      Abigail Gault, Linda Hogarth, Kristian C Williams, Alastair Greystoke, Neil Rajan, Ally Speight, Christopher A Lamb, Alison Bridgewood, Lisa-Jayne Brown-Schofield, Fiona Rayner, John D Isaacs, Jérémie Nsengimana, Christopher J Stewart, Amy E Anderson, Rut
      BMC Cancer.2024;[Epub]     CrossRef
    • A comprehensive immunobiology review of IBD: With a specific glance to Th22 lymphocytes development, biology, function, and role in IBD
      Jing Lv, Yousif Saleh Ibrahim, Alexey Yumashev, Ahmed Hjazi, Ali Faraz, Mohammed Jawad Alnajar, Maytham T. Qasim, Pallavi Ghildiyal, Ahmed Hussein Zwamel, Yasser Fakri Mustafa
      International Immunopharmacology.2024; 137: 112486.     CrossRef
    • Synbiotics containing sea buckthorn polysaccharides ameliorate DSS-induced colitis in mice via regulating Th17/Treg homeostasis through intestinal microbiota and their production of BA metabolites and SCFAs
      Mingyou Yuan, Lili Chang, Pan Gao, Jing Li, Xinyuan Lu, Mingfang Hua, Xiulian Li, Xuebo Liu, Ying Lan
      International Journal of Biological Macromolecules.2024; 276: 133794.     CrossRef
    • FGL2 improves experimental colitis related to gut microbiota structure and bile acid metabolism by regulating macrophage autophagy and apoptosis
      Yuan Zhao, Zheng Xiang, Haoran Pan, Xielin Huang, Weizhen Chen, Zhiming Huang
      Heliyon.2024; 10(14): e34349.     CrossRef
    • The emerging role of oxidative stress in inflammatory bowel disease
      Peter Muro, Li Zhang, Shuxuan Li, Zihan Zhao, Tao Jin, Fei Mao, Zhenwei Mao
      Frontiers in Endocrinology.2024;[Epub]     CrossRef
    • Causal relationship between circulating immune cells and inflammatory bowel disease: A Mendelian randomization analysis
      Shan Li, Dujuan Mao, Quanshui Hao, Lijuan You, Xiufang Li, Yaohua Wu, Lai Wei, Heng Du
      Medicine.2024; 103(30): e39056.     CrossRef
    • Therapeutic role of extracellular vesicles from human umbilical cord mesenchymal stem cells and their wide therapeutic implications in inflammatory bowel disease and other inflammatory disorder
      Muhammad Azhar Ud Din, Aijun Wan, Ying Chu, Jing Zhou, Yongmin Yan, Zhiliang Xu
      Frontiers in Medicine.2024;[Epub]     CrossRef
    • Evaluating the causal effect of circulating proteome on the risk of inflammatory bowel disease-related traits using Mendelian randomization
      Beining Li, Ping Hu, Hongyan Liang, Xingliang Zhao, Aiting Zhang, Yingchong Xu, Bin Zhang, Jie Zhang
      Frontiers in Immunology.2024;[Epub]     CrossRef
    • Mucosal Immunity and Trained Innate Immunity of the Gut
      Tsvetelina Velikova, Issa El Kaouri, Konstantina Bakopoulou, Milena Gulinac, Kremena Naydenova, Martin Dimitrov, Milena Peruhova, Snezhina Lazova
      Gastroenterology Insights.2024; 15(3): 661.     CrossRef
    • Myrrh Essential Oil Improves DSS-Induced Colitis by Modulating the MAPK Signaling Pathway: In vitro and in vivo Studies
      Tiantian Tang, Yujiao Wang, Taotao Li, Ding Liu, Kai Yang, Jing Sun, Yajun Shi, Dongyan Guo, Junbo Zou, Fengyun Bai, Ying Sun, Mei Wang, Xiaofei Zhang
      Journal of Inflammation Research.2024; Volume 17: 5139.     CrossRef
    • Engineering a Novel Probiotic Toolkit in Escherichia coli Nissle 1917 for Sensing and Mitigating Gut Inflammatory Diseases
      Nathalie Weibel, Martina Curcio, Atilla Schreiber, Gabriel Arriaga, Marine Mausy, Jana Mehdy, Lea Brüllmann, Andreas Meyer, Len Roth, Tamara Flury, Valerie Pecina, Kim Starlinger, Jan Dernič, Kenny Jungfer, Fabian Ackle, Jennifer Earp, Martin Hausmann, Ma
      ACS Synthetic Biology.2024; 13(8): 2376.     CrossRef
    • Electroacupuncture at Zusanli regulates the pathological phenotype of inflammatory bowel disease by modulating the NLRP3 inflammasome pathway
      Yanqiang Chen, Miaomiao Cai, Boyuan Shen, Changchang Fan, Xiang Zhou
      Immunity, Inflammation and Disease.2024;[Epub]     CrossRef
    • DJ-X-013 reduces LPS-induced inflammation, modulates Th17/ myeloid-derived suppressor cells, and alters NF-κB expression to ameliorate experimental colitis
      Mousumi Mandal, Ahmed Rakib, Md Abdullah Al Mamun, Santosh Kumar, Frank Park, Dong-Jin Hwang, Wei Li, Duane D. Miller, Udai P. Singh
      Biomedicine & Pharmacotherapy.2024; 179: 117379.     CrossRef
    • Inflammatory Bowel Disease in Children
      Nabila Annisa Harum, Primadita Syahbani, Idznika Nurannisa Wibowo
      Archives of Pediatric Gastroenterology, Hepatology, and Nutrition.2024; 3(3): 38.     CrossRef
    • Shared Pathophysiology of Inflammatory Bowel Disease and Psoriasis: Unraveling the Connection
      Walter Jauregui, Yozahandy A Abarca, Yasmin Ahmadi, Vaishnavi B Menon, Daniela A Zumárraga, Maria Camila Rojas Gomez , Aleeza Basri, Rohitha S Madala, Peter Girgis, Zahra Nazir
      Cureus.2024;[Epub]     CrossRef
    • Recent Insight into Herbal Bioactives-based Novel Approaches for Chronic Intestinal Inflammatory Disorders Therapy
      Ranjit K. Harwansh, Sonia Chauhan, Rohitas Deshmukh, Rupa Mazumder
      Current Pharmaceutical Biotechnology.2024; 25(14): 1835.     CrossRef
    • Air pollution exposure and inflammatory bowel disease: a systematic literature review of epidemiological and mechanistic studies
      Henrik Olstrup, Hani A. S. Mohamed, Julie Honoré, Jörg Schullehner, Torben Sigsgaard, Bertil Forsberg, Anna Oudin
      Frontiers in Environmental Health.2024;[Epub]     CrossRef
    • Zhumeria majdae essential oil attenuates TNBS-induced colitis in rats by regulating inflammatory and apoptotic pathways
      Helia Aghamiri, Afrooz Mohammadgholi-Beiki, Rojin Rashidian, Manijeh Motevalian, Parvaneh Rahimi-Moghaddam, Mohammad Sheibani, Majid Jafari-Sabet
      Inflammopharmacology.2024; 32(6): 3809.     CrossRef
    • Nanobody-as versatile tool emerging in autoimmune diseases
      Ling Wang, Ran Luo, Weilang Zhang, Hanyu Jiang, Yongkang Yu, Wenhu Zhou, Fan Zhang, Jian Ma, Lin Mei
      Smart Materials in Medicine.2024; 5(4): 501.     CrossRef
    • Intracellularly Gelated Macrophages Loaded with Probiotics for Therapy of Colitis
      Siyao Gu, Xiaona Zhao, Fang Wan, Dayong Gu, Weidong Xie, Cheng Gao
      Nano Letters.2024; 24(43): 13504.     CrossRef
    • Dysregulation of T cell response in the pathogenesis of inflammatory bowel disease
      Rucha Chandwaskar, Rajdeep Dalal, Saurabh Gupta, Aishwarya Sharma, Deepak Parashar, Vivek K. Kashyap, Jagdip Singh Sohal, Subhash K. Tripathi
      Scandinavian Journal of Immunology.2024;[Epub]     CrossRef
    • The role of gut microbiota, immune system, and autophagy in the pathogenesis of inflammatory bowel disease: Molecular mechanisms and therapeutic approaches
      Beatrice Garavaglia, Letizia Vallino, Angela Amoruso, Marco Pane, Alessandra Ferraresi, Ciro Isidoro
      Aspects of Molecular Medicine.2024; 4: 100056.     CrossRef
    • Biological Response of Treatment with Saffron Petal Extract on Cytokine-Induced Oxidative Stress and Inflammation in the Caco-2/Human Leukemia Monocytic Co-Culture Model
      Federica De Cecco, Sara Franceschelli, Valeria Panella, Maria Anna Maggi, Silvia Bisti, Arturo Bravo Nuevo, Damiano D’Ardes, Francesco Cipollone, Lorenza Speranza
      Antioxidants.2024; 13(10): 1257.     CrossRef
    • Gut Microbiome as a Target of Intervention in Inflammatory Bowel Disease Pathogenesis and Therapy
      Helal F. Hetta, Yasmin N. Ramadan, Ahmad A. Alharbi, Shomokh Alsharef, Tala T. Alkindy, Alanoud Alkhamali, Abdullah S. Albalawi, Hussein El Amin
      Immuno.2024; 4(4): 400.     CrossRef
    • Candida tropicalis -derived vitamin B3 exerts protective effects against intestinal inflammation by promoting IL-17A/IL-22-dependent epithelial barrier function
      Ha T Doan, Li-Chieh Cheng, Yi-Ling Chiu, Yuan-Kai Cheng, Cheng-Chih Hsu, Yee-Chun Chen, Hsiu-Jung Lo, Hao-Sen Chiang
      Gut Microbes.2024;[Epub]     CrossRef
    • Sturgeon-derived peptide LLLE alleviated colitis via regulating gut microbiota and its metabolites
      Jie Lin, Jiani Yang, Leqi Cui, Ravinder Nagpal, Prashant Singh, Gloria Salazar, Qinchun Rao, Ye Peng, Quancai Sun
      Current Research in Food Science.2024; : 100898.     CrossRef
    • Atopic disease and inflammatory bowel disease: A bidirectional Mendelian randomization study
      Dongyuan Zheng, Qinke Xu, Yingchao Liu
      Medicine.2024; 103(42): e40143.     CrossRef
    • The Therapeutic Effects of Lactic Acid Bacteria Isolated from Spotted Hyena on Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice
      Mengen Xu, Miao Hu, Jingbo Han, Lei Wang, Yuanyuan He, Md. F. Kulyar, Xiaohu Zhang, Yaozhong Lu, Siyang Mu, Hang Su, Jintao Cao, Jiakui Li
      Nutrients.2024; 16(21): 3682.     CrossRef
    • Therapeutic potential of mesenchymal stem cell‐derived extracellular vesicles: A focus on inflammatory bowel disease
      Laura Clua‐Ferré, Roger Suau, Irene Vañó‐Segarra, Iris Ginés, Carolina Serena, Josep Manyé
      Clinical and Translational Medicine.2024;[Epub]     CrossRef
    • Safety and efficacy of S1P receptor modulators for the induction and maintenance phases in inflammatory bowel disease: A systematic review and meta-analysis of randomized controlled trials
      Abdullah Akram, Misha Ahmed, Kanza Farhan, Areeb Omer, Shamama Kaleem, Ali Tahir Khan, Uzma Aslam, Muhammad Abdullah Tahir, Saria Memon, Ayatul Karam, Humam Furqan, Muhammad Umair Anjum, Pratik Bhattarai
      Medicine.2024; 103(36): e39372.     CrossRef
    • Gut‑liver axis in liver disease: From basic science to clinical treatment (Review)
      Jianpeng Wang, Xinyi Wang, Enba Zhuo, Bangjie Chen, Shixin Chan
      Molecular Medicine Reports.2024;[Epub]     CrossRef
    • Microsomal Prostaglandin E Synthase-1 Controls Colonic Prostaglandin E2 Production and Exerts a Protective Effect on Colitis Induced by Trinitrobenzene Sulfonic Acid in Mice
      Fumiaki Kojima, Yuka Hioki, Hiroki Sekiya, Hitoshi Kashiwagi, Yoshiko Iizuka, Kei Eto, Shotaro Maehana, Fumitaka Kawakami, Makoto Kubo, Hitoshi Ishibashi, Takafumi Ichikawa
      International Journal of Molecular Sciences.2024; 25(22): 12326.     CrossRef
    • N-acetyltransferase 10 is implicated in the pathogenesis of cycling T cell-mediated autoimmune and inflammatory disorders in mice
      Wen-ping Li, Xin-tao Mao, Jia-huan Xie, Jie-yu Li, Bao-qin Liu, Le-xi Wu, Bing Yang, Yi-yuan Li, Jin Jin
      Nature Communications.2024;[Epub]     CrossRef
    • Potential diagnostic markers and therapeutic targets for non-alcoholic fatty liver disease and ulcerative colitis based on bioinformatics analysis and machine learning
      Zheng Luo, Cong Huang, Jilan Chen, Yunhui Chen, Hongya Yang, Qiaofeng Wu, Fating Lu, Tian E. Zhang
      Frontiers in Medicine.2024;[Epub]     CrossRef
    • Evaluation of the Effect of Probiotic Supplementation on Intestinal Barrier Integrity and Epithelial Damage in Colitis Disease: A Systematic Review
      Dilek Sivri, Betül Şeref, Melike Şare Bulut, Makbule Gezmen Karadağ
      Nutrition Reviews.2024;[Epub]     CrossRef
    • 18F-FDG-PET and other imaging modalities in the diagnosis and management of inflammatory bowel disease
      Abhijit Bhattaru
      American Journal of Nuclear Medicine and Molecular Imaging.2024; 14(5): 295.     CrossRef
    • In reviewing the emerging biomarkers of human inflammatory bowel disease (IBD): Endothelial progenitor cells (EPC) and their vesicles as potential biomarkers of cardiovascular manifestations and targets for personalized treatments
      Carmela Rita Balistreri
      Mechanisms of Ageing and Development.2024; 222: 112006.     CrossRef
    • Diagnostic value of interleukins 23 and 17 in the assessment of ulcerative colitis severity
      M.V. Stoikevych, O.M. Tatarchuk, T.S. Tarasova
      GASTROENTEROLOGY.2024; 58(4): 253.     CrossRef
    • Ulcerative Colitis, LAIR1 and TOX2 Expression, and Colorectal Cancer Deep Learning Image Classification Using Convolutional Neural Networks
      Joaquim Carreras, Giovanna Roncador, Rifat Hamoudi
      Cancers.2024; 16(24): 4230.     CrossRef
    • Innate immune cells in the pathogenesis of inflammatory bowel disease - from microbial metabolites to immune modulation
      Rabia S. Mousa, Pietro Invernizzi, Hani S. Mousa
      Frontiers in Gastroenterology.2024;[Epub]     CrossRef
    • Protective Potential of Limosilactobacillus fermentum Strains and Their Mixture on Inflammatory Bowel Disease via Regulating Gut Microbiota in Mice
      Jae Yeon Joung, Kayoung Choi, Ju-Hoon Lee, Nam Su Oh
      Journal of Microbiology and Biotechnology.2024;[Epub]     CrossRef
    • A Phase 2b, Randomised, Double-blind, Placebo-controlled, Parallel-arm, Multicenter Study Evaluating the Safety and Efficacy of Tesnatilimab in Patients with Moderately to Severely Active Crohn’s Disease
      Matthieu Allez, Bruce E Sands, Brian G Feagan, Geert D’Haens, Gert De Hertogh, Charles W Randall, Bin Zou, Jewel Johanns, Christopher O’Brien, Mark Curran, Rory Rebuck, Mei-Lun Wang, Nina Sabins, Thomas Baker, Taku Kobayashi
      Journal of Crohn's and Colitis.2023; 17(8): 1235.     CrossRef
    • Angelica oil restores the intestinal barrier function by suppressing S100A8/A9 signalling in mice with ulcerative colitis
      Chang Liu, Yue-Xian He, Jia-Ning Zhang, Fang Yang, Shu-Yuan Wang, Ji-Liang Hu, Yang Yu
      Phytomedicine.2023; 108: 154490.     CrossRef
    • Identification of anti-inflammatory components in Panax ginseng of Sijunzi Decoction based on spectrum-effect relationship
      Hong Kan, Dongxue Zhang, Weijia Chen, Shihan Wang, Zhongmei He, Shifeng Pang, Shuai Qu, Yingping Wang
      Chinese Herbal Medicines.2023; 15(1): 123.     CrossRef
    • Starch from Pueraria lobata and the amylose fraction alleviates dextran sodium sulfate induced colitis in mice
      Yifei Yang, Mingxing Li, Qingsong Liu, Qianyun Zhao, Jiuping Zeng, Qin Wang, Yueshui Zhao, Fukuan Du, Yu Chen, Jing Shen, Haoming Luo, Shengpeng Wang, Wanping Li, Meijuan Chen, Xiaobing Li, Fang Wang, Yuhong Sun, Li Gu, Zhangang Xiao, Yu Du, Xu Wu
      Carbohydrate Polymers.2023; 302: 120329.     CrossRef
    • Basiliximab therapy for immune‐mediated bowel disease in a pediatric heart transplant patient
      Amy L. Kiskaddon, Michael Wilsey, Ignacio Gonzalez‐Gomez, Jessica Laks, Alyssa Miles, Jennifer Carapellucci, Alfred Asante‐Korang
      Pediatric Transplantation.2023;[Epub]     CrossRef
    • Controlling optical properties of small SWCNT using new N-doping chains method: A first-principles study
      R. Takassa, F. Elfatouaki, O. Farkad, S. Hassine, O. Choukri, A.E. Mouncharih, Y. Ijdiyaou, E.A. Ibnouelghazi, D. Abouelaoualim
      Physica E: Low-dimensional Systems and Nanostructures.2023; 148: 115627.     CrossRef
    • Role of perioperative nutritional status and enteral nutrition in predicting and preventing post-operative complications in patients with Crohn’s disease
      Tianyu Jiang, Yongmei Jiang, Qianwen Jin, Shining Xu, Abraham Fingerhut, Yongmei Shi, Minhua Zheng, Zirui He
      Frontiers in Nutrition.2023;[Epub]     CrossRef
    • The Gut–Vascular Barrier as a New Protagonist in Intestinal and Extraintestinal Diseases
      Natalia Di Tommaso, Francesco Santopaolo, Antonio Gasbarrini, Francesca Romana Ponziani
      International Journal of Molecular Sciences.2023; 24(2): 1470.     CrossRef
    • Salvia miltiorrhiza polysaccharide and its related metabolite 5-methoxyindole-3-carboxaldehyde ameliorate experimental colitis by regulating Nrf2/Keap1 signaling pathway
      Yu-Ping Fu, Xi Peng, Chao-Wen Zhang, Quan-Xing Jiang, Cen-Yu Li, Berit Smestad Paulsen, Frode Rise, Chao Huang, Bin Feng, Li-Xia Li, Xing-Fu Chen, Ren-Yong Jia, Yang-Ping Li, Xing-Hong Zhao, Gang Ye, Hua-Qiao Tang, Xiao-Xia Liang, Cheng Lv, Meng-Liang Tia
      Carbohydrate Polymers.2023; 306: 120626.     CrossRef
    • Therapeutic potential of mesenchymal stem/stromal cells (MSCs)-based cell therapy for inflammatory bowel diseases (IBD) therapy
      Mohamed J. Saadh, Maria V. Mikhailova, Soheil Rasoolzadegan, Mojgan Falaki, Roozbeh Akhavanfar, José Luis Arias Gonzáles, Amir Rigi, Bahman Abedi Kiasari
      European Journal of Medical Research.2023;[Epub]     CrossRef
    • Revisiting the “starved gut” hypothesis in inflammatory bowel disease
      Sean P. Colgan, Ruth X. Wang, Caroline H.T. Hall, Geetha Bhagavatula, J. Scott Lee
      Immunometabolism.2023; 5(1): e0016.     CrossRef
    • Hydroxytyrosol and Its Potential Uses on Intestinal and Gastrointestinal Disease
      Alessia Arangia, Ylenia Marino, Daniela Impellizzeri, Ramona D’Amico, Salvatore Cuzzocrea, Rosanna Di Paola
      International Journal of Molecular Sciences.2023; 24(4): 3111.     CrossRef
    • Inhibition of NLRP3 attenuates sodium dextran sulfate-induced inflammatory bowel disease through gut microbiota regulation
      Shi-Le Wang, Man-Man Zhang, Han Zhou, Guo-Qiang Su, Yi Ding, Guang-Hui Xu, Xu Wang, Cheng-Fu Li, Wei-Feng Huang, Li-Tao Yi
      Biomedical Journal.2023; 46(5): 100580.     CrossRef
    • Innate and Adaptive Immunity during SARS-CoV-2 Infection: Biomolecular Cellular Markers and Mechanisms
      Brent Brown, Vanshika Ojha, Ingo Fricke, Suhaila A Al-Sheboul, Chinua Imarogbe, Tanya Gravier, Michael Green, Lori Peterson, Ivoyl P. Koutsaroff, Ayça Demir, Jonatane Andrieu, Chiuan Yee Leow, Chiuan Herng Leow
      Vaccines.2023; 11(2): 408.     CrossRef
    • Nsun2 coupling with RoRγt shapes the fate of Th17 cells and promotes colitis
      Wen-Lan Yang, Weinan Qiu, Ting Zhang, Kai Xu, Zi-Juan Gu, Yu Zhou, Heng-Ji Xu, Zhong-Zhou Yang, Bin Shen, Yong-Liang Zhao, Qi Zhou, Ying Yang, Wei Li, Peng-Yuan Yang, Yun-Gui Yang
      Nature Communications.2023;[Epub]     CrossRef
    • Protective Effect of Lactiplantibacillus plantarum subsp. plantarum SC-5 on Dextran Sulfate Sodium—Induced Colitis in Mice
      Ruoran Shi, Fazheng Yu, Xueyu Hu, Yan Liu, Yuanyuan Jin, Honglin Ren, Shiying Lu, Jian Guo, Jiang Chang, Yansong Li, Zengshan Liu, Xiaoxu Wang, Pan Hu
      Foods.2023; 12(4): 897.     CrossRef
    • Nitric Oxide, Neutrophil/Lymphocyte, and Platelet/Lymphocyte Ratios as Promising Inflammatory Biomarkers in Complicated Crohn’s Disease: Outcomes of Corticosteroids and Anti-TNF-α Therapies
      Imene Soufli, Abdelkrim Hablal, Samia Bessaad, Manel Amri, Moussa Labsi, Rania Sihem Boussa, Fahima Ameur, Houda Belguendouz, Sonia Ait Younes, Nassim Sid Idris, Chafia Touil-Boukoffa
      Inflammation.2023; 46(3): 1091.     CrossRef
    • Inulin: properties and health benefits
      Yu-Qing Qin, Liu-Yan Wang, Xin-Yu Yang, Yi-Jie Xu, Gang Fan, Yan-Ge Fan, Jing-Nan Ren, Qi An, Xiao Li
      Food & Function.2023; 14(7): 2948.     CrossRef
    • In Vitro Evaluation of the Most Active Probiotic Strains Able to Improve the Intestinal Barrier Functions and to Prevent Inflammatory Diseases of the Gastrointestinal System
      Alessandra Fusco, Vittoria Savio, Donatella Cimini, Sergio D’Ambrosio, Adriana Chiaromonte, Chiara Schiraldi, Giovanna Donnarumma
      Biomedicines.2023; 11(3): 865.     CrossRef
    • RNA Editing is a Valuable Biomarker for Predicting Carcinogenesis in Ulcerative Colitis
      Kazutaka Takahashi, Kunitoshi Shigeyasu, Yoshitaka Kondo, Kazuyoshi Gotoh, Shuya Yano, Yuzo Umeda, Toshihiro Inokuchi, Caiming Xu, Kazuhiro Yoshida, Hibiki Umeda, Toshiaki Takahashi, Sho Takeda, Ryuichi Yoshida, Fuminori Teraishi, Hiroyuki Kishimoto, Yosh
      Journal of Crohn's and Colitis.2023; 17(5): 754.     CrossRef
    • Donkey whey proteins ameliorate dextran sulfate sodium-induced ulcerative colitis in mice by downregulating the S100A8-TRAF6-NF-κB axis-mediated inflammatory response
      Mohan Li, Qilong Li, Rayhnigul Abdlla, Jiali Chen, Xiqing Yue, Siew Young Quek
      Food Science and Human Wellness.2023; 12(5): 1809.     CrossRef
    • The oral microbiome in autoimmune diseases: friend or foe?
      Xiaoyan Huang, Xiangyu Huang, Yi Huang, Jiarong Zheng, Ye Lu, Zizhao Mai, Xinyuan Zhao, Li Cui, Shaohong Huang
      Journal of Translational Medicine.2023;[Epub]     CrossRef
    • The Role of Seaweed Polysaccharides in Gastrointestinal Health: Protective Effect against Inflammatory Bowel Disease
      N. M. Liyanage, D. P. Nagahawatta, Thilina U. Jayawardena, You-Jin Jeon
      Life.2023; 13(4): 1026.     CrossRef
    • ABX464 (Obefazimod) Upregulates miR-124 to Reduce Proinflammatory Markers in Inflammatory Bowel Diseases
      Cécile Apolit, Noëlie Campos, Audrey Vautrin, Christina Begon-Pescia, Laure Lapasset, Didier Scherrer, Paul Gineste, Hartmut Ehrlich, Aude Garcel, Julien Santo, Jamal Tazi
      Clinical and Translational Gastroenterology.2023; 14(4): e00560.     CrossRef
    • Dietary protocatechuic acid redistributes tight junction proteins by targeting Rho-associated protein kinase to improve intestinal barrier function
      Bo Ram So, San Kim, Se Hyeon Jang, Min Jeong Kim, Jeong Jae Lee, Soo Rin Kim, Sung Keun Jung
      Food & Function.2023; 14(10): 4777.     CrossRef
    • Experimental Murine Models for Colorectal Cancer Research
      Íris Neto, João Rocha, Maria Manuela Gaspar, Catarina P. Reis
      Cancers.2023; 15(9): 2570.     CrossRef
    • Effectiveness and safety of vedolizumab for ulcerative colitis: a single-center retrospective real-world study in China
      Kaituo Huang, Jing Liu, Wenhao Xia, Chuwen Tian, Lingya Yao, Qian Cao, Haotian Chen
      Frontiers in Pharmacology.2023;[Epub]     CrossRef
    • Reviewing not Homer’s Iliad, but “Kai Bao Ben Cao”: indigo dye—the past, present, and future
      Yusuke Yoshimatsu, Tomohisa Sujino, Takanori Kanai
      Intestinal Research.2023; 21(2): 174.     CrossRef
    • Progress on Regulation of NLRP3 Inflammasome by Chinese Medicine in Treatment of Ulcerative Colitis
      Hao-xian Sun, Ying Zhu
      Chinese Journal of Integrative Medicine.2023; 29(8): 750.     CrossRef
    • Identification of the informational and supportive needs of patients diagnosed with inflammatory bowel disease: a scoping review
      Narges Norouzkhani, Mahbobeh Faramarzi, Sara Ghodousi Moghadam, Mohammad Amin Karimi, Javad Shokri Shirvani, Ali Bahari, Mahdie ShojaeiBaghini, Saeid Eslami, Hamed Tabesh
      Frontiers in Psychology.2023;[Epub]     CrossRef
    • New-Onset Crohn’s Disease following Initiation of Secukinumab: A Case Report and Review of the Role of IL-17 in the Pathogenesis of Crohn’s Disease
      Anas Khouri, Cesar Moreno, Benjamin Niland, Hirotada Akiho
      Case Reports in Gastrointestinal Medicine.2023; 2023: 1.     CrossRef
    • A Holistic Perspective on How Photobiomodulation May Influence Fatigue, Pain, and Depression in Inflammatory Bowel Disease: Beyond Molecular Mechanisms
      E-Liisa Laakso, Tatjana Ewais
      Biomedicines.2023; 11(5): 1497.     CrossRef
    • The Crosstalk between Gut Microbiota and Nervous System: A Bidirectional Interaction between Microorganisms and Metabolome
      Monica Montagnani, Lucrezia Bottalico, Maria Assunta Potenza, Ioannis Alexandros Charitos, Skender Topi, Marica Colella, Luigi Santacroce
      International Journal of Molecular Sciences.2023; 24(12): 10322.     CrossRef
    • Fecal microbiota transplantation improves chicken growth performance by balancing jejunal Th17/Treg cells
      Ziyu Ma, Muhammad Akhtar, Hong Pan, Qiyao Liu, Yan Chen, Xinxin Zhou, Yingting You, Deshi Shi, Huazhen Liu
      Microbiome.2023;[Epub]     CrossRef
    • Methylation-Regulated Long Non-Coding RNA Expression in Ulcerative Colitis
      Christopher G. Fenton, Mithlesh Kumar Ray, Wei Meng, Ruth H. Paulssen
      International Journal of Molecular Sciences.2023; 24(13): 10500.     CrossRef
    • Infliximab trough levels are associated with endoscopic healing but not with transmural healing at one year treatment with infliximab in pediatric patients with Crohn’s disease
      So Yoon Choi, Yiyoung Kwon, Sujin Choi, So Mi Lee, Byung-Ho Choe, Ben Kang
      Frontiers in Immunology.2023;[Epub]     CrossRef
    • Role of Serum/Glucocorticoid-Regulated Kinase 1 (SGK1) in Immune and Inflammatory Diseases
      Xixi Bian, Honglu Xue, Dehuai Jing, Yan Wang, Guangxi Zhou, Fengqin Zhu
      Inflammation.2023; 46(5): 1612.     CrossRef
    • Investigating association between inflammatory bowel disease and rotavirus vaccination in a paediatric cohort in the UK
      Aidan Flatt, Thomas Inns, Kate M. Fleming, Miren Iturriza-Gómara, Daniel Hungerford
      Epidemiology and Infection.2023;[Epub]     CrossRef
    • Diet high in linoleic acid dysregulates the intestinal endocannabinoid system and increases susceptibility to colitis in Mice
      Poonamjot Deol, Paul Ruegger, Geoffrey D. Logan, Ali Shawki, Jiang Li, Jonathan D. Mitchell, Jacqueline Yu, Varadh Piamthai, Sarah H. Radi, Sana Hasnain, Kamil Borkowski, John W. Newman, Declan F. McCole, Meera G. Nair, Ansel Hsiao, James Borneman, France
      Gut Microbes.2023;[Epub]     CrossRef
    • Ameliorative effect of aqueous extract of Carica papaya Linn. leaves on Acetic acid induced Ulcerative Colitis in Male Albino Wistar rats
      Rachana Govind Hublikar, Sadhana N Holla, Cheshmitha Minnamreddigari
      Research Journal of Pharmacy and Technology.2023; : 2147.     CrossRef
    • Protopine Alleviates Dextran Sodium Sulfate-Induced Ulcerative Colitis by Improving Intestinal Barrier Function and Regulating Intestinal Microbiota
      Meishan Yue, Jialu Huang, Xiaolan Ma, Peng Huang, Yisong Liu, Jianguo Zeng
      Molecules.2023; 28(13): 5277.     CrossRef
    • Chlorella vulgaris Modulates Gut Microbiota and Induces Regulatory T Cells to Alleviate Colitis in Mice
      Priyanka Velankanni, Seok-Ho Go, Jong Beom Jin, Jin-Soo Park, Sunhee Park, Su-Bin Lee, Ho-Keun Kwon, Cheol-Ho Pan, Kwang Hyun Cha, Choong-Gu Lee
      Nutrients.2023; 15(15): 3293.     CrossRef
    • Discovery of a novel small molecule with efficacy in protecting against inflammation in vitro and in vivo by enhancing macrophages activation
      Cui Zhang, Zheng Cao, Hehua Lei, Chuan Chen, Ruichen Du, Yuchen Song, Ce Zhang, Jinlin Zhou, Yujing Lu, Luodong Huang, Peihong Shen, Limin Zhang
      Biomedicine & Pharmacotherapy.2023; 165: 115273.     CrossRef
    • Anti-inflammatory potential of lactic acid bacteria from southern Brazil by reducing TNF-α and increasing IL-10 synthesis: an in vitro study
      Angélica Vincenzi, Márcia Inês Goettert, Claucia Fernanda Volken de Souza
      Journal of Applied Microbiology.2023;[Epub]     CrossRef
    • A mannitol-modified emodin nano-drug restores the intestinal barrier function and alleviates inflammation in a mouse model of DSS-induced ulcerative colitis
      Yin-Yue Xu, Min Zhu, Jiang Wu, Long-Biao Luo, Si-jing Dong, Meng-Gai Zhang, Xue Liu, Ke Wang, Hua Luo, Wang-Hui Jing, Lin Wang, Si-Cen Wang
      Chinese Medicine.2023;[Epub]     CrossRef
    • Anti-Inflammatory Effects of Serotonin Receptor and Transient Receptor Potential Channel Ligands in Human Small Intestinal Epithelial Cells
      Gregory Ian Robinson, Dongping Li, Bo Wang, Yeva Zahoruiko, Marta Gerasymchuk, Darryl Hudson, Olga Kovalchuk, Igor Kovalchuk
      Current Issues in Molecular Biology.2023; 45(8): 6743.     CrossRef
    • Clinical Usefulness of Immune Profiling for Differential Diagnosis between Crohn’s Disease, Intestinal Tuberculosis, and Behcet’s Disease
      Ji Won Yoo, Su In Jo, Dong Woo Shin, Ji Won Park, Sung-Eun Kim, Hyun Lim, Ho Suk Kang, Sung-Hoon Moon, Min Kyu Kim, Sang-Yeob Kim, Sung Wook Hwang, Jae Seung Soh
      Diagnostics.2023; 13(18): 2904.     CrossRef
    • Evaluation of serum cytokines and acute phase proteins as possible pharmacodynamic biomarkers to monitor endoscopic remission during ustekinumab therapy in patients with Crohn’s disease
      Nathalie Van den Berghe, Dahham Alsoud, Bram Verstockt, Séverine Vermeire, Paul Declerck, Debby Thomas
      Therapeutic Advances in Gastroenterology.2023;[Epub]     CrossRef
    • miR-369-3p Modulates Intestinal Inflammatory Response via BRCC3/NLRP3 Inflammasome Axis
      Viviana Scalavino, Emanuele Piccinno, Anna Maria Valentini, Nicolò Schena, Raffaele Armentano, Gianluigi Giannelli, Grazia Serino
      Cells.2023; 12(17): 2184.     CrossRef
    • The P2X7 Receptor in Autoimmunity
      Fabio Grassi, Gaia Salina
      International Journal of Molecular Sciences.2023; 24(18): 14116.     CrossRef
    • Vaccination therapy for inflammatory bowel disease
      Yafei Liu, Fei Liao
      Human Vaccines & Immunotherapeutics.2023;[Epub]     CrossRef
    • The Effect of Serotonin Transmission on Depressive and Insomnia Symptoms in Inflammatory Bowel Diseases
      Marcin Sochal, Alicja Witkowska, Agata Binienda, Agata Gabryelska, Piotr Białasiewicz, Jakub Fichna, Renata Talar-Wojnarowska, Ewa Małecka-Wojciesko
      Journal of Clinical Medicine.2023; 12(19): 6353.     CrossRef
    • Cell membrane nanomaterials composed of phospholipids and glycoproteins for drug delivery in inflammatory bowel disease: A review
      Pengyu Lei, Haiyang Yu, Jiahui Ma, Jiao Du, Yimeng Fang, Qinsi Yang, Kun Zhang, Li Luo, Libo Jin, Wei Wu, Da Sun
      International Journal of Biological Macromolecules.2023; 249: 126000.     CrossRef
    • Mechanistic insights into the role of probiotics in modulating immune cells in ulcerative colitis
      Ni Guo, Lu‐lu Lv
      Immunity, Inflammation and Disease.2023;[Epub]     CrossRef
    • A new face of old cells: An overview about the role of senescence and telomeres in inflammatory bowel diseases
      Michał Sienkiewicz, Kamila Sroka, Agata Binienda, Diana Jurk, Jakub Fichna
      Ageing Research Reviews.2023; 91: 102083.     CrossRef
    • The Earlier You Find, the Better You Treat: Red Flags for Early Diagnosis of Inflammatory Bowel Disease
      Laura Cantoro, Rita Monterubbianesi, Giuliano Falasco, Caterina Camastra, Paolo Pantanella, Mariangela Allocca, Rocco Cosintino, Roberto Faggiani, Silvio Danese, Gionata Fiorino
      Diagnostics.2023; 13(20): 3183.     CrossRef
    • Bariatric Surgery and Inflammatory Bowel Disease: National Trends and Outcomes Associated with Procedural Sleeve Gastrectomy vs Historical Bariatric Surgery Among US Hospitalized Patients 2009–2020
      Joseph-Kevin Igwe, Phani Keerthi Surapaneni, Erin Cruz, Cedric Cole, Kingsley Njoku, Jisoo Kim, Ugo Alaribe, Kelechi Weze, Bilal Mohammed
      Obesity Surgery.2023; 33(11): 3472.     CrossRef
    • Kynurenine pathway and its role in neurologic, psychiatric, and inflammatory bowel diseases
      Mohammad Sheibani, Maryam Shayan, Mina Khalilzadeh, Zahra Ebrahim Soltani, Majid Jafari-Sabet, Mehdi Ghasemi, Ahmad Reza Dehpour
      Molecular Biology Reports.2023; 50(12): 10409.     CrossRef
    • APE1/Ref-1 as a Therapeutic Target for Inflammatory Bowel Disease
      Lauren Sahakian, Ainsley M. Robinson, Linda Sahakian, Rhian Stavely, Mark R. Kelley, Kulmira Nurgali
      Biomolecules.2023; 13(11): 1569.     CrossRef
    • Imaging Agents for PET of Inflammatory Bowel Disease: A Review
      Farzaneh Rezazadeh, Aidan P. Kilcline, Nerissa T. Viola
      Journal of Nuclear Medicine.2023; 64(12): 1858.     CrossRef
    • Crosstalk between alternative splicing and inflammatory bowel disease: Basic mechanisms, biotechnological progresses and future perspectives
      Chentao Zou, Xinquan Zan, Zhenyu Jia, Lu Zheng, Yijie Gu, Fei Liu, Ye Han, Chunfang Xu, Airong Wu, Qiaoming Zhi
      Clinical and Translational Medicine.2023;[Epub]     CrossRef
    • Reclassification of Inflammatory Bowel Disease Type Unclassified by Small Bowel Capsule Endoscopy
      Ana-Maria Singeap, Catalin Sfarti, Irina Girleanu, Laura Huiban, Cristina Muzica, Sergiu Timofeiov, Carol Stanciu, Anca Trifan
      Medicina.2023; 59(12): 2064.     CrossRef
    • Cytokine induced inflammatory bowel disease model using organ-on-a-chip technology
      Christine Tataru, Maya Livni, Carrie Marean-Reardon, Maria Clara Franco, Maude David, Masanori A. Murayama
      PLOS ONE.2023; 18(12): e0289314.     CrossRef
    • Psilocybin and Eugenol Reduce Inflammation in Human 3D EpiIntestinal Tissue
      Gregory Ian Robinson, Dongping Li, Bo Wang, Tahiat Rahman, Marta Gerasymchuk, Darryl Hudson, Olga Kovalchuk, Igor Kovalchuk
      Life.2023; 13(12): 2345.     CrossRef
    • Impact of artificial intelligence on prognosis, shared decision-making, and precision medicine for patients with inflammatory bowel disease: a perspective and expert opinion
      Philippe Pinton
      Annals of Medicine.2023;[Epub]     CrossRef
    • Next -generation probiotics as potential therapeutic supplement for gastrointestinal infections
      Nidhi Gupta, Kajal Kachhawaha, Deepak Kumar Behera, Vijay Kumar Verma
      Pharmacological Research - Reports.2023; 1: 100002.     CrossRef
    • Pharmacogenetic Effect of Thiopurine Methyl Transferase (TPMT) Gene Expression and Serum TNF on the Imuran Response in Ulcerative Colitis (UC) Iraqi Patients
      Noor Ali, Rafid Abdulkareem
      Reports of Biochemistry and Molecular Biology.2023; 12(3): 438.     CrossRef
    • Serum Globulin Is Associated with Endoscopic Findings and Mucosal Healing in Japanese Patients with Ulcerative Colitis
      Kana Shiraishi, Shinya Furukawa, Sen Yagi, Kenichirou Mori, Tomoyuki Ninomiya, Keitarou Kawasaki, Yuji Mizukami, Seiyuu Suzuki, Masayoshi Uraoka, Naozumi Shibata, Sanae Nakamura, Satoshi Imamine, Hidehiro Murakami, Katsuhisa Ohashi, Masamoto Torisu, Makot
      Digestive Diseases and Sciences.2022; 67(1): 233.     CrossRef
    • Epigenetic regulation of pediatric and neonatal immune responses
      Jennifer Bermick, Matthew Schaller
      Pediatric Research.2022; 91(2): 297.     CrossRef
    • Seroprevalence of viral infectious diseases and associated factors in Korean patients with inflammatory bowel diseases
      Hee Seung Hong, Jiwon Jung, Sang Hyoung Park, Hwa Jung Kim, Sung Wook Hwang, Dong-Hoon Yang, Jeong-Sik Byeon, Seung-Jae Myung, Suk-Kyun Yang, Byong Duk Ye
      The Korean Journal of Internal Medicine.2022; 37(1): 73.     CrossRef
    • Physical Activity in Patients With Inflammatory Bowel Disease: A Narrative Review
      Maitreyi Raman, Vidya Rajagopalan, Sandeep Kaur, Raylene A Reimer, Christopher Ma, Subrata Ghosh, Jeff Vallance
      Inflammatory Bowel Diseases.2022; 28(7): 1100.     CrossRef
    • Association between IL-10 rs3024505 and susceptibility to inflammatory bowel disease: A systematic review and meta-analysis
      Meiling Liu, Wang Yuan, Sunmin Park
      Cytokine.2022; 149: 155721.     CrossRef
    • Specificity of clinical decision support tools for vedolizumab and ustekinumab in biologically naïve patients with Crohn’s disease
      Ji Eun Kim, Hojeong Won, Young-Ho Kim, Eun Ran Kim, Sung Noh Hong, Dong Kyung Chang
      Scandinavian Journal of Gastroenterology.2022; 57(4): 446.     CrossRef
    • (RS)-bambuterol and its enantiomers: Potential improvement of (R)-bambuterol in mice with colitis
      Liangjun Deng, Shanping Wang, Haihua Guo, Xiaoming Liu, Xinfeng Zou, Rui Zhang, Yue Lin, Wen Tan
      International Immunopharmacology.2022; 103: 108501.     CrossRef
    • Neuroinflammation as an etiological trigger for depression comorbid with inflammatory bowel disease
      Colin F. Craig, Rhiannon T. Filippone, Rhian Stavely, Joel C. Bornstein, Vasso Apostolopoulos, Kulmira Nurgali
      Journal of Neuroinflammation.2022;[Epub]     CrossRef
    • Facilitation of colonic T cell immune responses is associated with an exacerbation of dextran sodium sulfate–induced colitis in mice lacking microsomal prostaglandin E synthase-1
      Fumiaki Kojima, Hiroki Sekiya, Yuka Hioki, Hitoshi Kashiwagi, Makoto Kubo, Masaki Nakamura, Shotaro Maehana, Yoshitaka Imamichi, Koh-ichi Yuhki, Fumitaka Ushikubi, Hidero Kitasato, Takafumi Ichikawa
      Inflammation and Regeneration.2022;[Epub]     CrossRef
    • Protective Effect of Probiotics Isolated from Traditional Fermented Tea Leaves (Miang) from Northern Thailand and Role of Synbiotics in Ameliorating Experimental Ulcerative Colitis in Mice
      Napapan Kangwan, Sarawut Kongkarnka, Nitsara Boonkerd, Kridsada Unban, Kalidas Shetty, Chartchai Khanongnuch
      Nutrients.2022; 14(1): 227.     CrossRef
    • Randomised clinical trial: a phase 1b study of GB004, an oral HIF‐1α stabiliser, for treatment of ulcerative colitis
      Silvio Danese, Barrett G. Levesque, Brian G. Feagan, Alina Jucov, Bal Raj Bhandari, Rish K. Pai, Kristen Taylor Meadows, Brian J. Kirby, Jean‐Marie Bruey, Allan Olson, Robin Osterhout, Courtney Van Biene, Julia Ford, Richard Aranda, Kartik Raghupathi, Wil
      Alimentary Pharmacology & Therapeutics.2022; 55(4): 401.     CrossRef
    • The Role of Innate and Adaptive Immune Cells in the Pathogenesis and Development of the Inflammatory Response in Ulcerative Colitis
      Aleksandra Kałużna, Paweł Olczyk, Katarzyna Komosińska-Vassev
      Journal of Clinical Medicine.2022; 11(2): 400.     CrossRef
    • Anti‐ulcerative effects of wogonin on ulcerative colitis induced by dextran sulfate sodium via Nrf2/TLR4/NF‐κB signaling pathway in BALB/c mice
      Yadong Zhou, Fafu Dou, Huwei Song, Tao Liu
      Environmental Toxicology.2022; 37(4): 954.     CrossRef
    • Mesenchymal stromal cells encapsulated in licensing hydrogels exert delocalized systemic protection against ulcerative colitis via subcutaneous xenotransplantation
      Ainhoa Gonzalez-Pujana, Ana Beloqui, José Javier Aguirre, Manoli Igartua, Edorta Santos-Vizcaino, Rosa Maria Hernandez
      European Journal of Pharmaceutics and Biopharmaceutics.2022; 172: 31.     CrossRef
    • Tea Tree Oil Terpinen-4-ol Protects Gut Barrier Integrity by Upregulation of Tight Junction Proteins via the ERK1/2-Signaling Pathway
      Yanhong Yong, Biao Fang, Yingxin Huang, Junyu Li, Tianyue Yu, Lianyun Wu, Canying Hu, Xiaoxi Liu, Zhichao Yu, Xingbin Ma, Ravi Gooneratne, Sidong Li, A. M. Abd El-Aty, Xianghong Ju
      Frontiers in Nutrition.2022;[Epub]     CrossRef
    • Immunity and Nutrition: The Right Balance in Inflammatory Bowel Disease
      Bartolo Tamburini, Marco Pio La Manna, Lidia La Barbera, Leila Mohammadnezhad, Giusto Davide Badami, Mojtaba Shekarkar Azgomi, Francesco Dieli, Nadia Caccamo
      Cells.2022; 11(3): 455.     CrossRef
    • Hyperactive neutrophil chemotaxis contributes to anti‐tumor necrosis factor‐α treatment resistance in inflammatory bowel disease
      Tung On Yau, Jayakumar Vadakekolathu, Gemma Ann Foulds, Guodong Du, Benjamin Dickins, Christos Polytarchou, Sergio Rutella
      Journal of Gastroenterology and Hepatology.2022; 37(3): 531.     CrossRef
    • The role of diosgenin in crohn’s disease
      Sunday Ogundepo, Adinnu M. Chiamaka, Mercy Olatinwo, David Adepoju, Matthias Taiwo Aladesanmi, Ugwu Obiora Celestine, Kingsley Chijioke Ali, Ogochukwu Jennifer Umezinwa, Janet Olasore, Abdullahi Alausa
      Clinical Phytoscience.2022;[Epub]     CrossRef
    • The Possible Pathogenic Role of IgG4-Producing Plasmablasts in Stricturing Crohn’s Disease
      Omar Bushara, David Joseph Escobar, Samuel Edward Weinberg, Leyu Sun, Jie Liao, Guang-Yu Yang
      Pathobiology.2022; 89(4): 187.     CrossRef
    • Splenic size as a marker for active inflammation in Crohn's disease
      Alla Khashper, Doron Shwartz, Benjamin Hyatt Taragin, Tamar Shalmon
      Clinical Imaging.2022; 84: 164.     CrossRef
    • IBD-associated G protein-coupled receptor 65 variant compromises signalling and impairs key functions involved in inflammation
      Virginie Mercier, Gabrielle Boucher, Dominic Devost, Kyla Bourque, Azadeh Alikashani, Claudine Beauchamp, Alain Bitton, Sylvain Foisy, Philippe Goyette, Guy Charron, Terence E. Hébert, John D. Rioux
      Cellular Signalling.2022; 93: 110294.     CrossRef
    • Cow Milk Extracellular Vesicle Effects on an In Vitro Model of Intestinal Inflammation
      Samanta Mecocci, Alessio Ottaviani, Elisabetta Razzuoli, Paola Fiorani, Daniele Pietrucci, Chiara Grazia De Ciucis, Silvia Dei Giudici, Giulia Franzoni, Giovanni Chillemi, Katia Cappelli
      Biomedicines.2022; 10(3): 570.     CrossRef
    • Phycocyanin ameliorates mouse colitisviaphycocyanobilin-dependent antioxidant and anti-inflammatory protection of the intestinal epithelial barrier
      Wei Guo, Mingyong Zeng, Suqin Zhu, Shiyang Li, Yilin Qian, Haohao Wu
      Food & Function.2022; 13(6): 3294.     CrossRef
    • Role of probiotics in the management of cervical cancer: An update
      Kusuma Kandati, Praveen Belagal, John Sushma Nannepaga, Buddolla Viswanath
      Clinical Nutrition ESPEN.2022; 48: 5.     CrossRef
    • Protective Effect of Nelumbo nucifera Plant on Dextran Sodium Sulfate-Induced Ulcerative Colitis in Rats
      Jignesh I. Patel, Monika Kumbhani, Morvi M. Raval
      Annals of the National Academy of Medical Sciences (India).2022; 58: 78.     CrossRef
    • Serum Albumin to Globulin Ratio is Associated with the Presence and Severity of Inflammatory Bowel Disease
      Yanyan Wang, Chengyong Li, Weiyi Wang, Jiajia Wang, Jinhui Li, Shuangjie Qian, Chao Cai, Yuntao Liu
      Journal of Inflammation Research.2022; Volume 15: 1907.     CrossRef
    • Pro-inflammatory effects of silver nanoparticles in the intestine
      Adelaide Sousa, Tracey D. Bradshaw, Daniela Ribeiro, Eduarda Fernandes, Marisa Freitas
      Archives of Toxicology.2022; 96(6): 1551.     CrossRef
    • Aconitate Decarboxylase 1 Deficiency Exacerbates Mouse Colitis Induced by Dextran Sodium Sulfate
      Ho Won Kim, A-Reum Yu, Ji Won Lee, Hoe Sun Yoon, Byung Soo Lee, Hwan-Woo Park, Sung Ki Lee, Young Ik Lee, Jake Whang, Jong-Seok Kim
      International Journal of Molecular Sciences.2022; 23(8): 4392.     CrossRef
    • Immune–related biomarkers shared by inflammatory bowel disease and liver cancer
      Thong Ba Nguyen, Duy Ngoc Do, Thuy T. P. Nguyen, Truc Ly Nguyen, Tung Nguyen-Thanh, Ha Thi Nguyen, Divijendra Natha Reddy Sirigiri
      PLOS ONE.2022; 17(4): e0267358.     CrossRef
    • Naphthoquinone derivatives as P-glycoprotein inducers in inflammatory bowel disease: 2D monolayers, 3D spheroids, and in vivo models
      Nuha Mahmoud, Mohamed-Elamir F. Hegazy, Walaa Wadie, Mohamed Elbadawi, Edmond Fleischer, Anette Klinger, Gerhard Bringmann, Mohamed T. Khayyal, Thomas Efferth
      Pharmacological Research.2022; 179: 106233.     CrossRef
    • Lactobacillus acidophilus and HKL Suspension Alleviates Ulcerative Colitis in Rats by Regulating Gut Microbiota, Suppressing TLR9, and Promoting Metabolism
      Kasimujiang Aximujiang, Kuerbannaimu Kaheman, Xilinguli Wushouer, Guixia Wu, Abulaiti Ahemaiti, Kurexi Yunusi
      Frontiers in Pharmacology.2022;[Epub]     CrossRef
    • Adipose tissue‐derived mesenchymal stem cells' acellular product extracellular vesicles as a potential therapy for Crohn's disease
      Jessica Altemus, Neda Dadgar, Yan Li, Amy L. Lightner
      Journal of Cellular Physiology.2022; 237(7): 3001.     CrossRef
    • Emerging role of biosimilars in the clinical care of inflammatory bowel disease patients
      Hala Najeeb, Farah Yasmin, Salim Surani
      World Journal of Clinical Cases.2022; 10(14): 4327.     CrossRef
    • Increasing thirty-day readmissions of Crohn’s disease and ulcerative colitis in the United States: A national dilemma
      Dushyant Singh Dahiya, Abhilash Perisetti, Asim Kichloo, Amandeep Singh, Hemant Goyal, Laura Rotundo, Madhu Vennikandam, Hafeez Shaka, Gurdeep Singh, Jagmeet Singh, Sailaja Pisipati, Mohammad Al-Haddad, Madhusudhan R Sanaka, Sumant Inamdar
      World Journal of Gastrointestinal Pathophysiology.2022; 13(3): 85.     CrossRef
    • Melatonin mediated inhibition of EZH2-NOS2 crosstalk attenuates inflammatory bowel disease in preclinical in vitro and in vivo models
      Mohammed Nadim Sardoiwala, Soni Jignesh Mohanbhai, Avinash Chandra Kushwaha, Atul Dev, Liku Biswal, Shyam Sunder Sharma, Subhasree Roy Choudhury, Surajit Karmakar
      Life Sciences.2022; 302: 120655.     CrossRef
    • Pyroptosis and Its Role in Autoimmune Disease: A Potential Therapeutic Target
      Ruixuan You, Xinglan He, Zhuotong Zeng, Yi Zhan, Yangfan Xiao, Rong Xiao
      Frontiers in Immunology.2022;[Epub]     CrossRef
    • Inflammatory auto-immune diseases of the intestine and their management by natural bioactive compounds
      Naoual El Menyiy, Aicha El Allam, Sara Aboulaghras, Imane Jaouadi, Saad Bakrim, Nasreddine El Omari, Mohammad Ali Shariati, Alevtin Miftakhutdinov, Polrat Wilairatana, Mohammad S. Mubarak, Abdelhakim Bouyahya
      Biomedicine & Pharmacotherapy.2022; 151: 113158.     CrossRef
    • Curcumin-based nanotechnology approaches and therapeutics in restoration of autoimmune diseases
      Niloufar Rahiman, Yuliya V. Markina, Prashant Kesharwani, Thomas P. Johnston, Amirhossein Sahebkar
      Journal of Controlled Release.2022; 348: 264.     CrossRef
    • HucMSC-Ex carrying miR-203a-3p.2 ameliorates colitis through the suppression of caspase11/4-induced macrophage pyroptosis
      Yuting Xu, Xiaohua Tang, Anning Fang, Jialai Yan, Dickson Kofi Wiredu Ocansey, Xu Zhang, Fei Mao
      International Immunopharmacology.2022; 110: 108925.     CrossRef
    • Intestinal Barrier Dysfunction in Fatty Liver Disease: Roles of Microbiota, Mucosal Immune System, and Bile Acids
      Biki Gupta, Ravi Rai, Michael Oertel, Reben Raeman
      Seminars in Liver Disease.2022; 42(02): 122.     CrossRef
    • 8-Oxypalmatine, a novel oxidative metabolite of palmatine, exhibits superior anti-colitis effect via regulating Nrf2 and NLRP3 inflammasome
      Juanjuan Cheng, Xingdong Ma, Haitao Zhang, Xiaoyan Wu, Minhua Li, Gaoxiang Ai, Ruoting Zhan, Jianhui Xie, Ziren Su, Xiaoqi Huang
      Biomedicine & Pharmacotherapy.2022; 153: 113335.     CrossRef
    • Deciphering the interdependent labyrinth between gut microbiota and the immune system
      A. Saini, P. Dalal, D. Sharma
      Letters in Applied Microbiology.2022; 75(5): 1122.     CrossRef
    • Null Function of Npr1 Disturbs Immune Response in Colonic Inflammation During Early Postnatal Stage
      Changkun Long, Hongfei Liu, Wenxing Zhan, Liping Chen, Andong Wu, Lin Yang, Shenghan Chen
      Inflammation.2022; 45(6): 2419.     CrossRef
    • Antitumor necrosis factor treatment in patients with inflammatory bowel disease does not promote psoriasis development: A meta-analysis
      Yu Kyung Jun, Joo Young Park, Seong-Joon Koh, Hyunsun Park, Hyoun Woo Kang, Jong Pil Im, Joo Sung Kim
      Medicine.2022; 101(27): e29872.     CrossRef
    • Association between intestinal microbiota and inflammatory bowel disease
      Yunchang Zhang, Xuemeng Si, Ling Yang, Hui Wang, Ye Sun, Ning Liu
      Animal Models and Experimental Medicine.2022; 5(4): 311.     CrossRef
    • Intra-abdominal septic complications after ileocolic resection increases risk for endoscopic and surgical postoperative Crohn’s disease recurrence
      Salam P Bachour, Ravi S Shah, Florian Rieder, Taha Qazi, Jean Paul Achkar, Jessica Philpott, Bret Lashner, Stefan D Holubar, Amy L Lightner, Edward L Barnes, Jordan Axelrad, Miguel Regueiro, Benjamin Click, Benjamin L Cohen
      Journal of Crohn's and Colitis.2022; 16(11): 1696.     CrossRef
    • The Business of T Cell Subsets and Cytokines in the Immunopathogenesis of Inflammatory Bowel Disease
      Shreekant Bharti, Mridushri Bharti
      Cureus.2022;[Epub]     CrossRef
    • A nationwide analysis on the influence of obesity in inflammatory bowel disease hospitalizations
      Dushyant Singh Dahiya, Asim Kichloo, Farah Wani, Jagmeet Singh, Dhanshree Solanki, Hafeez Shaka
      Intestinal Research.2022; 20(3): 342.     CrossRef
    • Hypoxia-Inducible Factor 1-Alpha Stabilizers in the Treatment of Inflammatory Bowel Diseases: Oxygen as a Novel IBD Therapy?
      Shubha Bhat, Florian Rieder
      Journal of Crohn's and Colitis.2022; 16(12): 1924.     CrossRef
    • Mesenchymal (Stem) Stromal Cells Based as New Therapeutic Alternative in Inflammatory Bowel Disease: Basic Mechanisms, Experimental and Clinical Evidence, and Challenges
      Noemi Eiro, Maria Fraile, Alberto González-Jubete, Luis O. González, Francisco J. Vizoso
      International Journal of Molecular Sciences.2022; 23(16): 8905.     CrossRef
    • Cutting-Edge Delivery Systems and Adjuvants in Tolerogenic Vaccines: A Review
      Chiara Puricelli, Elena Boggio, Casimiro Luca Gigliotti, Ian Stoppa, Salvatore Sutti, Roberta Rolla, Umberto Dianzani
      Pharmaceutics.2022; 14(9): 1782.     CrossRef
    • Identification of potential crucial genes and key pathways shared in Inflammatory Bowel Disease and cervical cancer by machine learning and integrated bioinformatics
      Thong Ba Nguyen, Duy Ngoc Do, My-Le Nguyen-Thi, Hiep Hoang-The, Thanh-Thoa Tran, Tung Nguyen-Thanh
      Computers in Biology and Medicine.2022; 149: 105996.     CrossRef
    • Anxiolytic and anti-colitis effects of Moringa oleifera leaf-aqueous extract on acetic acid-induced colon inflammation in rat
      Paul Aimé Noubissi, Queendoline Njilifac, Michel Archange Fokam Tagne, Mireille Sylviane Dongmo Nguepi, Angèle Foyet Fondjo, Nadège Kouémou Emégam, Joseph Ngakou Mukam, Roger Zintchem, Henri Wambe, Gaétan Olivier Fankem, Agathe Fotio Tonfack, Jean Paul To
      Biomedicine & Pharmacotherapy.2022; 154: 113652.     CrossRef
    • Identification of lncRNA DLEU2 as a potential diagnostic biomarker and anti-inflammatory target for ulcerative colitis
      Qiuling Lin, Dingguo Zhang, Jian Zhang, Weixiang Luo, Zhenglei Xu, Jun Yao, Lisheng Wang
      Frontiers in Pharmacology.2022;[Epub]     CrossRef
    • Integrative bioinformatics analysis to identify the effects of circadian rhythm on Crohn’s disease
      Dan Liu, Yin-Yun Chen, Qing-qing Li, Ming Xu, Jiang-Tao Liao, Ben Wang
      Frontiers in Molecular Biosciences.2022;[Epub]     CrossRef
    • Early fecal microbiome transfer after donor defecation determines response in patients with moderate to severe ulcerative colitis
      Arshdeep Singh, Ramit Mahajan, Bhavjeet Kaur Kahlon, Ashvin Singh Dhaliwal, Vandana Midha, Varun Mehta, Namita Bansal, Dharmatma Singh, Ajit Sood
      Indian Journal of Gastroenterology.2022; 41(4): 389.     CrossRef
    • P2X7 receptor as the regulator of T-cell function in intestinal barrier disruption
      Zhi-Feng Jiang, Wei Wu, Han-Bing Hu, Zheng-Yang Li, Ming Zhong, Lin Zhang
      World Journal of Gastroenterology.2022; 28(36): 5265.     CrossRef
    • Prostaglandins and calprotectin are genetically and functionally linked to the Inflammatory Bowel Diseases
      Mohamad Karaky, Gabrielle Boucher, Saraï Mola, Sylvain Foisy, Claudine Beauchamp, Marie-Eve Rivard, Melanie Burnette, Hugues Gosselin, Alain Bitton, Guy Charron, Philippe Goyette, John D. Rioux, Tuuli Lappalainen
      PLOS Genetics.2022; 18(9): e1010189.     CrossRef
    • Application of the Clustering Technique to Multiple Nutritional Factors Related to Inflammation and Disease Progression in Patients with Inflammatory Bowel Disease
      Agnieszka Dąbek-Drobny, Olga Kaczmarczyk, Agnieszka Piątek-Guziewicz, Michał Woźniakiewicz, Paweł Paśko, Justyna Dobrowolska-Iwanek, Aneta Woźniakiewicz, Aneta Targosz, Agata Ptak-Belowska, Paweł Zagrodzki, Małgorzata Zwolińska-Wcisło
      Nutrients.2022; 14(19): 3960.     CrossRef
    • Ulcerative colitis complicated by autoimmune hepatitis-primary biliary cholangitis-primary sclerosing cholangitis overlap syndrome
      Miaoyu Jing, Wenjia Zhang, Min Xia
      Revista Española de Enfermedades Digestivas.2022;[Epub]     CrossRef
    • Atovaquone attenuates experimental colitis by reducing neutrophil infiltration of colonic mucosa
      Laura D. Manzanares, Joseph David, Xingsheng Ren, Lenore K. Yalom, Enzo B. Piccolo, Yalda Dehghan, Aidan J. David, Stephen B. Hanauer, Ronen Sumagin
      Frontiers in Pharmacology.2022;[Epub]     CrossRef
    • Anti-TNFα Drugs and Interleukin Inhibitors: Epidemiological and Pharmacovigilance Investigation in COVID-19 Positive Patients
      Zaira Maraia, Tony Mazzoni, Marco Bruno Luigi Rocchi, Denise Feliciani, Maria Chiara Romani, Giovanna Acciarri, Stefania Rafaiani, Isidoro Mazzoni
      Journal of Personalized Medicine.2022; 12(11): 1770.     CrossRef
    • Treatment with Extracellular Vesicles from Giardia lamblia Alleviates Dextran Sulfate Sodium-Induced Colitis in C57BL/6 Mice
      Hyun Jung Kim, Young-Ju Lee, Seon-Ok Back, Shin-Hyeong Cho, Hee-Il Lee, Myoung-Ro Lee
      The Korean Journal of Parasitology.2022; 60(5): 309.     CrossRef
    • A novel gut inflammatory rat model by laparotomic injection of peptidoglycan from Staphylococcus aureus
      Savita Devi, Rajeev Kapila, Suman Kapila
      Archives of Microbiology.2022;[Epub]     CrossRef
    • CROHN’S DISEASE: A REVIEW ON EPIDEMIOLOGY, DIAGNOSIS AND THERAPEUTIC MANAGEMENT
      Mohammad Mukim, Mohit Chaturvedi, Rakesh Patel, Supriya Roy, Pratishtha Sharma, Varunesh Chaturvedi, Saloni Goyal, Mohsina F. Patewkar
      Indian Drugs.2022; 59(09): 16.     CrossRef
    • The emerging microbiome‐based approaches to IBD therapy: From SCFAs to urolithin A
      Mohammad Rudiansyah, Saade Abdalkareem Jasim, Bakhadir S. Azizov, Vadim Samusenkov, Walid Kamal Abdelbasset, Ghulam Yasin, Hawraa Jabbar Mohammad, Mohammed Abed Jawad, Trias Mahmudiono, Seyed Reza Hosseini‐Fard, Rasoul Mirzaei, Sajad Karampoor
      Journal of Digestive Diseases.2022; 23(8-9): 412.     CrossRef
    • Mitochondrial Control in Inflammatory Gastrointestinal Diseases
      Guo-Yan Sui, Feng Wang, Jin Lee, Yoon Seok Roh
      International Journal of Molecular Sciences.2022; 23(23): 14890.     CrossRef
    • Role of Vitamin D in Celiac Disease and Inflammatory Bowel Diseases
      Claudia Infantino, Roberta Francavilla, Adriana Vella, Sabrina Cenni, Nicola Principi, Caterina Strisciuglio, Susanna Esposito
      Nutrients.2022; 14(23): 5154.     CrossRef
    • TDAG51 deficiency attenuates dextran sulfate sodium-induced colitis in mice
      Hyoeun Jeon, Dulshara Sachini Amarasekara, Nari Lee, Hye-Won Park, Jiyeon Yu, Jaerang Rho
      Scientific Reports.2022;[Epub]     CrossRef
    • Old but Fancy: Curcumin in Ulcerative Colitis—Current Overview
      Aleksandra Pituch-Zdanowska, Łukasz Dembiński, Aleksandra Banaszkiewicz
      Nutrients.2022; 14(24): 5249.     CrossRef
    • Dysregulated haemostasis in thrombo-inflammatory disease
      Paula A. Klavina, Gemma Leon, Annie M. Curtis, Roger J.S. Preston
      Clinical Science.2022; 136(24): 1809.     CrossRef
    • p-Hydroxybenzoic Acid Ameliorates Colitis by Improving the Mucosal Barrier in a Gut Microbiota-Dependent Manner
      Xue Han, Miaomiao Li, Lijun Sun, Xinjuan Liu, Yue Yin, Jianyu Hao, Weizhen Zhang
      Nutrients.2022; 14(24): 5383.     CrossRef
    • The study of tissue IgG4 in the mucous membrane of the colon in patients with inflammatory bowel disease
      Yu.M. Stepanov, T.S. Tarasova, M.V. Stoikevych, Yu.A. Gaydar, N.S. Fedorova
      GASTROENTEROLOGY.2022; 55(4): 246.     CrossRef
    • Relationship between the humoral link of immunity, cytokine regulation with short-chain fatty acids in patients with inflammatory bowel diseases
      Yu.M. Stepanov, M.V. Stoikevych, I.A. Klenina, O.M. Tatarchuk
      GASTROENTEROLOGY.2022; 56(2): 103.     CrossRef
    • Function of intestinal barrier protected by regulating the miR‐199a‐3p in ulcerative colitis: Modulation of IL‐23/IL‐17A axis
      Huixia Zhang, Fangbin Zhang, Wei Li
      Fundamental & Clinical Pharmacology.2021; 35(5): 852.     CrossRef
    • Hyptis suaveolens (L.) Poit protects colon from TNBS-induced inflammation via immunomodulatory, antioxidant and anti-proliferative mechanisms
      Flávia Danniele Frota Machado, Rodrigo de Oliveira Formiga, Gedson Rodrigues de Morais Lima, Neyres Zínia Taveira de Jesus, Edvaldo Balbino Alves Júnior, Alexsandro Fernandes Marinho, Josean Fechine Tavares, Flávia Almeida Santos, Ana Flávia Seraine Custó
      Journal of Ethnopharmacology.2021; 265: 113153.     CrossRef
    • Euterpe oleracea Mart. (Açaí) attenuates experimental colitis in rats: involvement of TLR4/COX-2/NF-ĸB
      Carlos Eduardo da Silva Monteiro, Humberto Barbosa da Costa Filho, Francisca Géssica Oliveira Silva, Maria de Fathima Felipe de Souza, Johnatan Alisson Oliveira Sousa, Álvaro Xavier Franco, Ângela Castro Resende, Roberto Soares de Moura, Marcellus Henriqu
      Inflammopharmacology.2021; 29(1): 193.     CrossRef
    • Bile Acid Signaling in Inflammatory Bowel Diseases
      Stefano Fiorucci, Adriana Carino, Monia Baldoni, Luca Santucci, Emanuele Costanzi, Luigina Graziosi, Eleonora Distrutti, Michele Biagioli
      Digestive Diseases and Sciences.2021; 66(3): 674.     CrossRef
    • Clinical and Mechanistic Characteristics of Current JAK Inhibitors in IBD
      Elleni J Pippis, Bruce R Yacyshyn
      Inflammatory Bowel Diseases.2021; 27(10): 1674.     CrossRef
    • Telehealth and mobile health interventions in adults with inflammatory bowel disease: A mixed‐methods systematic review
      Suja P. Davis, Megan Suzanne Hardin Ross, Reuben Adatorwovor, Holly Wei
      Research in Nursing & Health.2021; 44(1): 155.     CrossRef
    • The rationale for selenium supplementation in inflammatory bowel disease: A mechanism-based point of view
      Moein Ala, Zahedin Kheyri
      Nutrition.2021; 85: 111153.     CrossRef
    • Enteric Viruses and Inflammatory Bowel Disease
      Georges Tarris, Alexis de Rougemont, Maëva Charkaoui, Christophe Michiels, Laurent Martin, Gaël Belliot
      Viruses.2021; 13(1): 104.     CrossRef
    • Comparison of Long-term Outcomes of Infliximab versus Adalimumab in 1,488 Biologic-Naive Korean Patients with Crohn’s Disease
      Yoon Suk Jung, Minkyung Han, Sohee Park, Jae Hee Cheon
      Gut and Liver.2021; 15(1): 92.     CrossRef
    • Immunomodulatory biomaterials and their application in therapies for chronic inflammation-related diseases
      Jayachandra Reddy Nakkala, Ziming Li, Wajiha Ahmad, Kai Wang, Changyou Gao
      Acta Biomaterialia.2021; 123: 1.     CrossRef
    • Self‐Assembling Nanofibers Inhibit Inflammation in a Murine Model of Crohn's‐Disease‐Like Ileitis
      Matthew I. Bury, Natalie J. Fuller, Tristan D. Clemons, Renea M. Sturm, Christopher D. Morrison, Devon C. Lisy‐Snow, Bonnie G. Nolan, Christopher Tarczynski, Emily M. T. Ayello, Amber Boyce, Bridget Muckian, Nida Ahmad, Catherine J. Hunter, Mark R. Karver
      Advanced Therapeutics.2021;[Epub]     CrossRef
    • Update on the therapeutic management of patients with either psoriatic arthritis or ulcerative colitis: focus on the JAK inhibitor tofacitinib
      Maria Sole Chimenti, Paola Conigliaro, Livia Biancone, Roberto Perricone
      Therapeutic Advances in Musculoskeletal Disease.2021;[Epub]     CrossRef
    • The gut-joint axis in spondyloarthritis: immunological, microbial, and clinical insights
      Zoya Qaiyum, Melissa Lim, Robert D. Inman
      Seminars in Immunopathology.2021; 43(2): 173.     CrossRef
    • Extracellular Vesicles from Thapsigargin-Treated Mesenchymal Stem Cells Ameliorated Experimental Colitis via Enhanced Immunomodulatory Properties
      Hansol Joo, Mi-Kyung Oh, Ji Yeon Kang, Hyun Sung Park, Dong-Hoon Chae, Jieun Kim, Jong-Hee Lee, Hee Min Yoo, Uimook Choi, Do-Kyun Kim, Hakmo Lee, Sungjoo Kim, Kyung-Rok Yu
      Biomedicines.2021; 9(2): 209.     CrossRef
    • Very early onset inflammatory bowel disease with compound heterozygous variants in Nuclear Factor of Activated T cell 5
      Nina V. Kirk, Jens M. B. Jensen, Mikkel S. Petersen, A. Al‐Mousawi, Trine H. Mogensen, Mette Christiansen, Carsten S. Larsen
      European Journal of Immunology.2021; 51(4): 999.     CrossRef
    • Redox-active nanoparticles for inflammatory bowel disease
      Qinjuan Ren, Si Sun, Xiao-Dong Zhang
      Nano Research.2021; 14(8): 2535.     CrossRef
    • Extracellular Vesicles and Immune System in Ageing and Immune Diseases
      So Yeong Cheon, Jong Eun Lee
      Experimental Neurobiology.2021; 30(1): 32.     CrossRef
    • Role of vitamin D in Wnt pathway activation for colonic epithelial cell differentiation
      Satrio Wibowo, Krisni Subandiyah, Kusworini Handono, Sri Poeranto
      Journal of Taibah University Medical Sciences.2021; 16(4): 575.     CrossRef
    • The Role of Tumor Necrosis Factor Alpha (TNF-α) in Autoimmune Disease and Current TNF-α Inhibitors in Therapeutics
      Dan-in Jang, A-Hyeon Lee, Hye-Yoon Shin, Hyo-Ryeong Song, Jong-Hwi Park, Tae-Bong Kang, Sang-Ryong Lee, Seung-Hoon Yang
      International Journal of Molecular Sciences.2021; 22(5): 2719.     CrossRef
    • Biological functions of NLRP3 inflammasome: A therapeutic target in inflammatory bowel disease
      Yijie Song, Yuge Zhao, Yueming Ma, Zhicheng Wang, Lan Rong, Bing Wang, Ning Zhang
      Cytokine & Growth Factor Reviews.2021; 60: 61.     CrossRef
    • Innate Lymphoid Cells and Adaptive Immune Cells Cross-Talk: A Secret Talk Revealed in Immune Homeostasis and Different Inflammatory Conditions
      Vijay Kumar
      International Reviews of Immunology.2021; 40(3): 217.     CrossRef
    • Mast Cell Mediated Regulation of Small Intestinal Chloride Malabsorption in SAMP1/YitFc Mouse Model of Spontaneous Chronic Ileitis
      M Motiur Rahman, Sheuli Afroz, Subha Arthur, Uma Sundaram
      Cells.2021; 10(3): 697.     CrossRef
    • Phase 1 study on the safety and efficacy of E6011, antifractalkine antibody, in patients with Crohn's disease
      Katsuyoshi Matsuoka, Makoto Naganuma, Toshifumi Hibi, Hirohito Tsubouchi, Kiyoshi Oketani, Toshinori Katsurabara, Seiichiro Hojo, Osamu Takenaka, Tetsu Kawano, Toshio Imai, Takanori Kanai
      Journal of Gastroenterology and Hepatology.2021; 36(8): 2180.     CrossRef
    • Th17 immune response in inflammatory bowel disease: Future roles and opportunities for lactic acid bacteria and bioactive compounds released in fermented milk
      Lourdes Santiago-López, Adrián Hernández-Mendoza, Belinda Vallejo-Cordoba, Abraham Wall-Medrano, Aarón F. González-Córdova
      Trends in Food Science & Technology.2021; 112: 109.     CrossRef
    • PDE9 Inhibitor PF-04447943 Attenuates DSS-Induced Colitis by Suppressing Oxidative Stress, Inflammation, and Regulating T-Cell Polarization
      Mohammad Nasiruddin Rana, Jie Lu, Enfu Xue, Jingjing Ruan, Yuting Liu, Lejun Zhang, Rana Dhar, Yajun Li, Zhengqiang Hu, Jie Zhou, Wangqian Ma, Huifang Tang
      Frontiers in Pharmacology.2021;[Epub]     CrossRef
    • The effects of flaxseed supplementation on metabolic syndrome parameters, insulin resistance and inflammation in ulcerative colitis patients: An open‐labeled randomized controlled trial
      Nava Morshedzadeh, Mehran Rahimlou, Shabnam Shahrokh, Soheila Karimi, Parvin Mirmiran, Mohammad Reza Zali
      Phytotherapy Research.2021; 35(7): 3781.     CrossRef
    • Edible Bird’s Nest Ameliorates Dextran Sulfate Sodium-Induced Ulcerative Colitis in C57BL/6J Mice by Restoring the Th17/Treg Cell Balance
      Yaohua Fan, Yanqun Fan, Kunfeng Liu, Piyanuch Lonan, Feng Liao, Yuhang Huo, Xiaohua Zhong, Yueliang Liang, Yaxin Wang, Shaozhen Hou, Xiaoping Lai, Geng Li, Weihong Kuang
      Frontiers in Pharmacology.2021;[Epub]     CrossRef
    • Redox imbalance in Crohn's disease patients is modulated by Azathioprine
      Mohammad Javad Tavassolifar, Mostafa Changaei, Zahra Salehi, Fatemeh Ghasemi, Moslem Javidan, Mohammad Hossein Nicknam, Mohammad Reza Pourmand
      Redox Report.2021; 26(1): 80.     CrossRef
    • Immune Protection of a Helminth Protein in the DSS-Induced Colitis Model in Mice
      Shao Rong Long, Ruo Dan Liu, Deepak Vijaya Kumar, Zhong Quan Wang, Chien-Wen Su
      Frontiers in Immunology.2021;[Epub]     CrossRef
    • Effect of Size and Loading of Retinoic Acid in Polyvinyl Butyrate Nanoparticles on Amelioration of Colitis
      Jinting Li, Yunmei Mu, Yiwei Liu, Akihiro Kishimura, Takeshi Mori, Yoshiki Katayama
      Polymers.2021; 13(9): 1472.     CrossRef
    • Growing role of S100B protein as a putative therapeutic target for neurological- and nonneurological-disorders
      Fabrizio Michetti, Gabriele Di Sante, Maria Elisabetta Clementi, Beatrice Sampaolese, Patrizia Casalbore, Cinzia Volonté, Vincenzo Romano Spica, Pier Paolo Parnigotto, Rosa Di Liddo, Susanna Amadio, Francesco Ria
      Neuroscience & Biobehavioral Reviews.2021; 127: 446.     CrossRef
    • Intestinal production of secreted protein acidic and rich in cysteine (SPARC) in patients with ulcerative colitis
      Gabriela Fonseca-Camarillo, Janette Furuzawa-Carballeda, Natalia Razo-López, Rafael Barreto-Zúñiga, Braulio Martínez-Benítez, Jesús K. Yamamoto-Furusho
      Immunobiology.2021; 226(3): 152095.     CrossRef
    • Root Extract of Lindera aggregata (Sims) Kosterm. Modulates the Th17/Treg Balance to Attenuate DSS-Induced Colitis in Mice by IL-6/STAT3 Signaling Pathway
      Huimin Lai, Zhengbiao Yang, Zhaohuan Lou, Feng Li, Feng Xie, Wei Pan, Cong Xu, Lili Zhang, Sheng Zhang, Lijiang Zhang, Mincong Huang
      Frontiers in Pharmacology.2021;[Epub]     CrossRef
    • Bile Acids Activated Receptors in Inflammatory Bowel Disease
      Michele Biagioli, Silvia Marchianò, Adriana Carino, Cristina Di Giorgio, Luca Santucci, Eleonora Distrutti, Stefano Fiorucci
      Cells.2021; 10(6): 1281.     CrossRef
    • Protective effects of potential probiotic Lactobacillus rhamnosus (MTCC-5897) fermented whey on reinforcement of intestinal epithelial barrier function in a colitis-induced murine model
      Harpreet Kaur, Taruna Gupta, Suman Kapila, Rajeev Kapila
      Food & Function.2021; 12(13): 6102.     CrossRef
    • Characterization of terminal-ileal and colonic Crohn’s disease in treatment-naïve paediatric patients based on transcriptomic profile using logistic regression
      Ilkyu Park, Jaeeun Jung, Sugi Lee, Kunhyang Park, Jea-Woon Ryu, Mi-Young Son, Hyun-Soo Cho, Dae-Soo Kim
      Journal of Translational Medicine.2021;[Epub]     CrossRef
    • Interaction between Lipopolysaccharide and Gut Microbiota in Inflammatory Bowel Diseases
      Marcello Candelli, Laura Franza, Giulia Pignataro, Veronica Ojetti, Marcello Covino, Andrea Piccioni, Antonio Gasbarrini, Francesco Franceschi
      International Journal of Molecular Sciences.2021; 22(12): 6242.     CrossRef
    • Fabrication and Evaluation of Celecoxib Oral Oleogel to Reduce the Inflammation of Ulcerative Colitis
      Nermin M. Sheta, Sylvia A. Boshra
      AAPS PharmSciTech.2021;[Epub]     CrossRef
    • Glycine Attenuates Citrobacter rodentium‐Induced Colitis by Regulating ATF6‐Mediated Endoplasmic Reticulum Stress in Mice
      Yunchang Zhang, Da Jiang, Yuhang Jin, Hai Jia, Ying Yang, In Ho Kim, Zhaolai Dai, Jinhua Zhang, Fazheng Ren, Zhenlong Wu
      Molecular Nutrition & Food Research.2021;[Epub]     CrossRef
    • Critical roles of microRNA-196 in normal physiology and non-malignant diseases: Diagnostic and therapeutic implications
      Milad Bastami, Andrea Masotti, Zahra Saadatian, Abdolreza Daraei, Mojtaba Farjam, Ali Ghanbariasad, Sepideh Zununi Vahed, Shirin Eyvazi, Yaser Mansoori, Ziba Nariman-Saleh-Fam
      Experimental and Molecular Pathology.2021; 122: 104664.     CrossRef
    • Prevalence of, and factors associated with, long-term COVID-19 sick leave in working-age patients followed in general practices in Germany
      Louis Jacob, Ai Koyanagi, Lee Smith, Christian Tanislav, Marcel Konrad, Susanne van der Beck, Karel Kostev
      International Journal of Infectious Diseases.2021; 109: 203.     CrossRef
    • Role of Myeloid-derived suppressor cell (MDSC) in autoimmunity and its potential as a therapeutic target
      Ehsan Nourbakhsh, Ali Mohammadi, Mohammad Salemizadeh Parizi, Atena Mansouri, Farnoosh Ebrahimzadeh
      Inflammopharmacology.2021; 29(5): 1307.     CrossRef
    • Translating Treg Therapy for Inflammatory Bowel Disease in Humanized Mice
      Sushmita Negi, Sheetal Saini, Nikunj Tandel, Kiran Sahu, Ravi P.N. Mishra, Rajeev K. Tyagi
      Cells.2021; 10(8): 1847.     CrossRef
    • Effect of resistance exercise training on Crohn’s disease patients
      Konstantinos Papadimitriou
      Intestinal Research.2021; 19(3): 275.     CrossRef
    • Dysbiotic microbiota interactions in Crohn’s disease
      Esther Caparrós, Reiner Wiest, Michael Scharl, Gerhard Rogler, Ana Gutiérrez Casbas, Bahtiyar Yilmaz, Marcin Wawrzyniak, Rubén Francés
      Gut Microbes.2021;[Epub]     CrossRef
    • Cross Talk between Gut Microbiota and Intestinal Mucosal Immunity in the Development of Ulcerative Colitis
      Junfeng Zou, Chen Liu, Shu Jiang, Dawei Qian, Jinao Duan, Anthony R. Richardson
      Infection and Immunity.2021;[Epub]     CrossRef
    • The Role of T Follicular Helper Cells and Interleukin-21 in the Pathogenesis of Inflammatory Bowel Disease
      Lulu Sun, Ruixue Kong, Hua Li, Dashan Wang, Paul A. Rufo
      Gastroenterology Research and Practice.2021; 2021: 1.     CrossRef
    • Role of MicroRNA in Inflammatory Bowel Disease: Clinical Evidence and the Development of Preclinical Animal Models
      Kanika Suri, Jason A. Bubier, Michael V. Wiles, Leonard D. Shultz, Mansoor M. Amiji, Vishnu Hosur
      Cells.2021; 10(9): 2204.     CrossRef
    • Anti-inflammatory effects of DA-9601, an extract of Artemisia asiatica, on aceclofenac-induced acute enteritis
      Ju Hwan Kim, Chang Yell Shin, Sun Woo Jang, Dong-Seok Kim, Wonae Lee, Hyung-Gun Kim, Hak Rim Kim
      The Korean Journal of Physiology & Pharmacology.2021; 25(5): 439.     CrossRef
    • Common contributing factors to COVID-19 and inflammatory bowel disease
      Ronald Neil Kostoff, Michael Brandon Briggs, Darja Kanduc, Darla Roye Shores, Leda Kovatsi, Alexander I. Vardavas, Alan L. Porter
      Toxicology Reports.2021; 8: 1616.     CrossRef
    • Protective Effect of Salvianolic Acid B in Acetic Acid-Induced Experimental Colitis in a Mouse Model
      Mydhili Govindarasu, Mohammad Azam Ansari, Mohammad N. Alomary, Sami AlYahya, Saad Alghamdi, Azzah M. Bannunah, Mazen Almehmadi, Pari Abirami, Ekambaram Gayathiri, Mariyappan Palani, Govindasamy Rajakumar, Muthu Thiruvengadam, Manju Vaiyapuri
      Processes.2021; 9(9): 1589.     CrossRef
    • Active Ingredients and Potential Mechanisms of the Gan Jiang-Huang Qin-Huang Lian-Ren Shen Decoction against Ulcerative Colitis: A Network Pharmacology and Molecular Docking-Based Study
      Ce Zhou, Hang Zhou, Furong Zhang, Liangliang Hao, Jing Guo, Duygu Ağagündüz
      Evidence-Based Complementary and Alternative Medicine.2021; 2021: 1.     CrossRef
    • Short-term tolerability and effectiveness of methotrexate monotherapy in adult patients with Crohn’s disease: a retrospective study
      Hee Seung Hong, Kyuwon Kim, Kyunghwan Oh, Jae Yong Lee, Seung Wook Hong, Jin Hwa Park, Sung Wook Hwang, Dong-Hoon Yang, Byong Duk Ye, Jeong-Sik Byeon, Seung-Jae Myung, Suk-Kyun Yang, Sang Hyoung Park
      Therapeutic Advances in Gastroenterology.2021;[Epub]     CrossRef
    • Role of Digital Health and Artificial Intelligence in Inflammatory Bowel Disease: A Scoping Review
      Kamila Majidova, Julia Handfield, Kamran Kafi, Ryan D. Martin, Ryszard Kubinski
      Genes.2021; 12(10): 1465.     CrossRef
    • Ozanimod as Induction and Maintenance Therapy for Ulcerative Colitis
      William J. Sandborn, Brian G. Feagan, Geert D’Haens, Douglas C. Wolf, Igor Jovanovic, Stephen B. Hanauer, Subrata Ghosh, AnnKatrin Petersen, Steven Y. Hua, Ji Hwan Lee, Lorna Charles, Denesh Chitkara, Keith Usiskin, Jean-Frederic Colombel, Loren Laine, Si
      New England Journal of Medicine.2021; 385(14): 1280.     CrossRef
    • The combined supplementation of omega-3 fatty acids and probiotics decreased the levels of serum polyamines in experimental colitis
      Havvanur Yoldaş İlktaç, Nihal Büyükuslu, Cüneyd Parlayan
      Food and Health.2021; 7(4): 279.     CrossRef
    • High-altitude hypoxia exacerbates dextran sulfate sodium (DSS)-induced colitis by upregulating Th1 and Th17 lymphocytes
      Mohammed Gamah, Murad Alahdal, Yu Zhang, Yiling Zhou, Qiaorong Ji, Zhouyang Yuan, Ying Han, Xiangqun Shen, Yanming Ren, Wei Zhang
      Bioengineered.2021; 12(1): 7985.     CrossRef
    • The incidence of psoriasis among smokers and/or former smokers inflammatory bowel diseases patients treated with tumor necrosis factor antagonist
      Meiqi Yang, Weixin Liu, Qiuping Deng, Zeng Liang, Qin Wang
      Medicine.2021; 100(42): e27510.     CrossRef
    • Host-Related Laboratory Parameters for Leprosy Reactions
      Yuqian Luo, Mitsuo Kiriya, Kazunari Tanigawa, Akira Kawashima, Yasuhiro Nakamura, Norihisa Ishii, Koichi Suzuki
      Frontiers in Medicine.2021;[Epub]     CrossRef
    • The Role of Mitochondria Dysfunction in Inflammatory Bowel Diseases and Colorectal Cancer
      Patrycja Kłos, Siarhei A. Dabravolski
      International Journal of Molecular Sciences.2021; 22(21): 11673.     CrossRef
    • Crohn’s disease at radiological imaging: focus on techniques and intestinal tract
      Giuseppe Cicero, Silvio Mazziotti
      Intestinal Research.2021; 19(4): 365.     CrossRef
    • Interaction Between Commensal Bacteria, Immune Response and the Intestinal Barrier in Inflammatory Bowel Disease
      Yongyan Chen, Wenwen Cui, Xiao Li, Huan Yang
      Frontiers in Immunology.2021;[Epub]     CrossRef
    • Association Between Peripheral Blood Monocyte Count and Mucosal Healing in Japanese Patients With Ulcerative Colitis
      Shinya Furukawa, Yoshio Ikeda, Sen Yagi, Teruki Miyake, Kana Shiraishi, Kazuhiro Tange, Yu Hashimoto, Kenichirou Mori, Tomoyuki Ninomiya, Seiyuu Suzuki, Naozumi Shibata, Hidehiro Murakami, Katsuhisa Ohashi, Aki Hasebe, Hideomi Tomida, Yasunori Yamamoto, E
      Clinical and Translational Gastroenterology.2021; 12(11): e00429.     CrossRef
    • Microscopic colitis and the role of the specialist inflammatory bowel disease nurse: a clinical review
      Cathy Walsh
      Gastrointestinal Nursing.2021; 19(9): 20.     CrossRef
    • Food Habit Associated Mycobiota Composition and Their Impact on Human Health
      Jata Shankar
      Frontiers in Nutrition.2021;[Epub]     CrossRef
    • Immunogenicity profile in African green monkeys of a vaccine candidate based on a mutated form of human Interleukin-15
      Yunier Rodríguez-Álvarez, Lino Gerardo Batista-Roche, Alexey Llopiz-Arzuaga, Pedro Puente-Pérez, Rafael Martínez-Castillo, Jorge Castro-Velazco, Alicia Santos-Savio
      BMC Immunology.2021;[Epub]     CrossRef
    • A Novel Role of Dapagliflozin in Mitigation of Acetic Acid-Induced Ulcerative Colitis by Modulation of Monocyte Chemoattractant Protein 1 (MCP-1)/Nuclear Factor-Kappa B (NF-κB)/Interleukin-18 (IL-18)
      Mohamed Kh. ElMahdy, Samar A. Antar, Ehab Kotb Elmahallawy, Walied Abdo, Hayfa Hussin Ali Hijazy, Ashraf Albrakati, Ahmed E. Khodir
      Biomedicines.2021; 10(1): 40.     CrossRef
    • Anti-inflammatory effects of apocynin: a narrative review of the evidence
      Maryam Boshtam, Shirin Kouhpayeh, Farahnaz Amini, Yadollah Azizi, Malihe Najaflu, Laleh Shariati, Hossein Khanahmad
      All Life.2021; 14(1): 997.     CrossRef
    • Autoimmune profile of rat blood in experimental ulcerative colitis
      E. V. Davydova, M. V. Osikov, A. E. Bakeeva, N. V. Kaigorodtseva
      Russian Journal of Immunology.2021; 24(2): 209.     CrossRef
    • 1
      Yu.M. Stepanov, T.S. Tarasova, M.V. Stoykevich
      GASTROENTEROLOGY.2021; 55(1): 54.     CrossRef
    • Crohn’s disease and ulcerative colitis are associated with different lipid profile disorders: a nationwide population‐based study
      Hosim Soh, Jong Pil Im, Kyungdo Han, Seona Park, Seung Wook Hong, Jeong Min Moon, Eun Ae Kang, Jaeyoung Chun, Hyun Jung Lee, Joo Sung Kim
      Alimentary Pharmacology & Therapeutics.2020; 51(4): 446.     CrossRef
    • Upregulated IL‐17A secretion and CCR6 co‐expression in Treg subsets are related to the imbalance of Treg/Th17 cells in active UC patients
      Yan Long, Xiaotao Zhao, Changsheng Xia, Xiaoxu Li, Chunhong Fan, Chen Liu, Chengbin Wang
      Scandinavian Journal of Immunology.2020;[Epub]     CrossRef
    • Intestinal Epithelial Deletion of Sphk1 Prevents Colitis-Associated Cancer Development by Inhibition of Epithelial STAT3 Activation
      Seung Bin Park, Byung-il Choi, Beom Jae Lee, Nam Joo Kim, Yoon A. Jeong, Moon Kyung Joo, Hyo Jung Kim, Jong-Jae Park, Jae Seon Kim, Yoon-Seok Noh, Hyun Joo Lee
      Digestive Diseases and Sciences.2020; 65(8): 2284.     CrossRef
    • Activation of adenosine A3 receptor inhibits inflammatory cytokine production in colonic mucosa of patients with ulcerative colitis by down‐regulating the nuclear factor‐kappa B signaling
      Tian Hua Ren, Min Min Lv, Xiao Meng An, Wai Keung Leung, Wai‐Kay Seto
      Journal of Digestive Diseases.2020; 21(1): 38.     CrossRef
    • Phenylethyl isothiocyanate induces oxidative damage of porcine kidney cells mediated by reactive oxygen species
      Yuanyuan Zhu, Shuiping Liu, Sisi Yan, Ji Wang, Linyu Zhang, Xin LI, Lixin Wen, Jing Wu
      Journal of Biochemical and Molecular Toxicology.2020;[Epub]     CrossRef
    • Recovery of CD226-TIGIT+FoxP3+ and CD226-TIGIT-FoxP3+ regulatory T cells contributes to clinical remission from active stage in ulcerative colitis patients
      Yan Long, Chengbin Wang, Changsheng Xia, Xiaoxu Li, Chunhong Fan, Xiaotao Zhao, Chen Liu
      Immunology Letters.2020; 218: 30.     CrossRef
    • Comparison of Real-World Outcomes of Infliximab versus Adalimumab in Biologic-Naïve Korean Patients with Ulcerative Colitis: A Population-Based Study
      Minkyung Han, Yoon Suk Jung, Jae Hee Cheon, Sohee Park
      Yonsei Medical Journal.2020; 61(1): 48.     CrossRef
    • Microbial imbalance in inflammatory bowel disease patients at different taxonomic levels
      Mohammad Tauqeer Alam, Gregory C. A. Amos, Andrew R. J. Murphy, Simon Murch, Elizabeth M. H. Wellington, Ramesh P. Arasaradnam
      Gut Pathogens.2020;[Epub]     CrossRef
    • RETRACTED: miRNA-129/FBW7/NF-κB, a Novel Regulatory Pathway in Inflammatory Bowel Disease
      Qinghui Meng, Weihua Wu, Tiemin Pei, Junlin Xue, Peng Xiao, Liang Sun, Long Li, Desen Liang
      Molecular Therapy - Nucleic Acids.2020; 19: 731.     CrossRef
    • Trends in Healthcare Costs for Inflammatory Bowel Disease in South Korea
      Yoon Suk Jung
      Gut and Liver.2020; 14(1): 3.     CrossRef
    • Presence of PTPN2 SNP rs1893217 Enhances the Anti-inflammatory Effect of Spermidine
      Anna Niechcial, Matthias Butter, Salomon Manz, Nicole Obialo, Katharina Bäbler, Lisa van der Lely, Silvia Lang, Claudia Gottier, Declan F McCole, Michael Scharl, Marianne R Spalinger
      Inflammatory Bowel Diseases.2020; 26(7): 1038.     CrossRef
    • Serotonin: A Potent Immune Cell Modulator in Autoimmune Diseases
      Minjie Wan, Lili Ding, Dong Wang, Jiawen Han, Pujun Gao
      Frontiers in Immunology.2020;[Epub]     CrossRef
    • Clinical Features and Outcomes of Tuberculosis in Inflammatory Bowel Disease Patients Treated with Anti-tumor Necrosis Factor Therapy
      Jihye Kim, Jong Pil Im, Jae-Joon Yim, Chang Kyun Lee, Dong Il Park, Chang Soo Eun, Sung-Ae Jung, Jeong Eun Shin, Kang-Moon Lee, Jae Hee Cheon
      The Korean Journal of Gastroenterology.2020; 75(1): 29.     CrossRef
    • Managing Pain and Psychosocial Care in IBD: a Primer for the Practicing Gastroenterologist
      Emily Weaver, Eva Szigethy
      Current Gastroenterology Reports.2020;[Epub]     CrossRef
    • Co-expression of master transcription factors determines CD4+ T cell plasticity and functions in auto-inflammatory diseases
      Bhalchandra Mirlekar
      Immunology Letters.2020; 222: 58.     CrossRef
    • Sestrin3 enhances macrophage-mediated generation of T helper 1 and T helper 17 cells in a mouse colitis model
      Liuqing Ge, Min Xu, Steven R Brant, Shaoping Liu, Chengliang Zhu, Jian Shang, Qiu Zhao, Feng Zhou
      International Immunology.2020; 32(6): 421.     CrossRef
    • Aspects of free radical oxidation in the large bowel in ulcerative colitis and Crohn’s disease
      MV Osikov, EV Davydova, MS Boyko, AE Bakeeva, NV Kaygorodtseva, IR Galeeva, AA Fedosov, MA Ilyinyh, LV Vorgova
      Bulletin of Russian State Medical University.2020; ((3)2020): 58.     CrossRef
    • Similar Clinical Outcomes of Early and Late Anti-TNF Initiation for Ulcerative Colitis: A Nationwide Population-Based Study
      Minkyung Han, Yoon Suk Jung, Jae Hee Cheon, Sohee Park
      Yonsei Medical Journal.2020; 61(5): 382.     CrossRef
    • Elucidation of colon-protective efficacy of diosgenin in experimental TNBS-induced colitis: inhibition of NF-κB/IkB-α and Bax/Caspase-1 signaling pathways
      Xiaobo Tang, Gengzhen Huang, Tao Zhang, Shiqing Li
      Bioscience, Biotechnology, and Biochemistry.2020; 84(9): 1903.     CrossRef
    • Lessons on the Sigma-1 Receptor in TNBS-Induced Rat Colitis: Modulation of the UCHL-1, IL-6 Pathway
      Nikoletta Almási, Szilvia Török, Szabolcs Dvorácskó, Csaba Tömböly, Ákos Csonka, Zoltán Baráth, Zsolt Murlasits, Zsuzsanna Valkusz, Anikó Pósa, Csaba Varga, Krisztina Kupai
      International Journal of Molecular Sciences.2020; 21(11): 4046.     CrossRef
    • Extracts and Marine Algae Polysaccharides in Therapy and Prevention of Inflammatory Diseases of the Intestine
      Natalya N. Besednova, Tatyana S. Zaporozhets, Tatyana A. Kuznetsova, Ilona D. Makarenkova, Sergey P. Kryzhanovsky, Lydmila N. Fedyanina, Svetlana P. Ermakova
      Marine Drugs.2020; 18(6): 289.     CrossRef
    • Making a big impact with small datasets using machine-learning approaches
      May Y Choi, Christopher Ma
      The Lancet Rheumatology.2020; 2(8): e451.     CrossRef
    • Immunotropic effect of rectal suppositories with vitamin D3 in dynamics of experimental ulcer colitis
      M. V. Osikov, E. V. Simonyan, M. S. Boyko, A. M. Bogomolova
      Experimental and Clinical Gastroenterology.2020; (6): 141.     CrossRef
    • Oleoylethanolamide Ameliorates Dextran Sulfate Sodium-Induced Colitis in Rats
      Shinsuke Otagiri, Shunsuke Ohnishi, Masatsugu Ohara, Qingjie Fu, Koji Yamamoto, Keiko Yamamoto, Takehiko Katsurada, Naoya Sakamoto
      Frontiers in Pharmacology.2020;[Epub]     CrossRef
    • Role of mesenchymal stem cell derived extracellular vesicles in autoimmunity: A systematic review
      Jing-Hua Wang, Xiao-Ling Liu, Jian-Mei Sun, Jing-Han Yang, Dong-Hua Xu, Shu-Shan Yan
      World Journal of Stem Cells.2020; 12(8): 879.     CrossRef
    • Impact of early anti-TNF use on clinical outcomes in Crohn’s disease: a nationwide population-based study
      Yoon Suk Jung, Minkyung Han, Sohee Park, Jae Hee Cheon
      The Korean Journal of Internal Medicine.2020; 35(5): 1104.     CrossRef
    • Red light-emitting diode treatment improves tissue recovery in DSS-induced colitis in mice
      Marina de Paula-Silva, Milena Fronza Broering, Pablo Scharf, Gustavo Henrique Oliveira da Rocha, Sandra Farsky, Adriana Lino-dos-Santos-Franco
      Journal of Photochemistry and Photobiology B: Biology.2020; 212: 112018.     CrossRef
    • Nanocomposites-based targeted oral drug delivery systems with infliximab in a murine colitis model
      Jung Min Kim, Da Hye Kim, Hyo Jeong Park, Hyun Woo Ma, I Seul Park, Mijeong Son, So Youn Ro, Seokmann Hong, Hyo Kyung Han, Soo Jeong Lim, Seung Won Kim, Jae Hee Cheon
      Journal of Nanobiotechnology.2020;[Epub]     CrossRef
    • Dietary Supplementation with Spray-Dried Porcine Plasma Attenuates Colon Inflammation in a Genetic Mouse Model of Inflammatory Bowel Disease
      Lluïsa Miró, Concepció Amat, Cristina Rosell-Cardona, Joy M. Campbell, Javier Polo, Anna Pérez-Bosque, Miquel Moretó
      International Journal of Molecular Sciences.2020; 21(18): 6760.     CrossRef
    • Synergistic therapeutic effect of mesenchymal stem cells and tolerogenic dendritic cells in an acute colitis mouse model
      Hajar Abbasi-Kenarsari, Neda Heidari, Kaveh Baghaei, Davar Amani, Mohammad Reza Zali, Sahar Gaffari Khaligh, Abbas Shafiee, Seyed Mahmoud Hashemi
      International Immunopharmacology.2020; 88: 107006.     CrossRef
    • SOD1 suppresses pro-inflammatory immune responses by protecting against oxidative stress in colitis
      Jiyoung Hwang, Jing Jin, Sejin Jeon, Shin Hye Moon, Min Young Park, Do-Young Yum, Jeong Hyun Kim, Ji-Eun Kang, Mi Hee Park, Eui-Joong Kim, Jae-Gu Pan, Oran Kwon, Goo Taeg Oh
      Redox Biology.2020; 37: 101760.     CrossRef
    • Characterization of mucosal cytokine profile in ulcerative colitis patients under conventional and anti-TNF-a treatment
      Loris Riccardo Lopetuso, Maddalena Corbi, Franco Scaldaferri, Valentina Petito, Cristina Graziani, Federica Castri, Matteo Neri, Antonio Gasbarrini, Alessandro Sgambato, Alfredo Papa
      European Journal of Gastroenterology & Hepatology.2020; 32(12): 1527.     CrossRef
    • Gallic and butyric acids modulated NLRP3 inflammasome markers in a co-culture model of intestinal inflammation
      Ivan Luzardo-Ocampo, Guadalupe Loarca-Piña, Elvira Gonzalez de Mejia
      Food and Chemical Toxicology.2020; 146: 111835.     CrossRef
    • Bioinformatics Analysis of Key Candidate Genes and Pathways in Ulcerative Colitis
      Guangya Xu, Xueling Yan, Jie Chen, Xiaoheng Guo, Xiaolan Guo, Yong Tang, Zheng Shi
      Biological and Pharmaceutical Bulletin.2020; 43(11): 1760.     CrossRef
    • Therapeutic Effect of Mesenchymal Stem Cells in Ulcerative Colitis: A Review on Achievements and Challenges
      Seyed-Kazem Hosseini-Asl, Davood Mehrabani, Feridoun Karimi-Busheri
      Journal of Clinical Medicine.2020; 9(12): 3922.     CrossRef
    • Serum Vitamins D, B9 and B12 in Greek Patients with Inflammatory Bowel Diseases
      Aristea Gioxari, Charalampia Amerikanou, Efstathia Papada, Evangelia Zioga, Andreas D. Georgoulis, George Bamias, Andriana C. Kaliora
      Nutrients.2020; 12(12): 3734.     CrossRef
    • İnflamatuar Bağırsak Hastalıkları Olan Bireylerde İlaç Uyumu ve Yaşam Kalitesi
      Nazli ÖZTÜRK, Yasemin YILDIRIM
      Bandırma Onyedi Eylül Üniversitesi Sağlık Bilimleri ve Araştırmaları Dergisi.2020; 2(3): 192.     CrossRef
    • Nutritional Support and Probiotics as a Potential Treatment of IBD
      Aleksandra Tarasiuk, Guido Eibl
      Current Drug Targets.2020; 21(14): 1417.     CrossRef
    • A role for Th1-like Th17 cells in the pathogenesis of inflammatory and autoimmune disorders
      Ali N. Kamali, Seyedeh Masoomeh Noorbakhsh, Haleh Hamedifar, Farhad Jadidi-Niaragh, Reza Yazdani, José M. Bautista, Gholamreza Azizi
      Molecular Immunology.2019; 105: 107.     CrossRef
    • 5‐Hydroxytryptamine (5‐HT)‐exacerbated DSS‐induced colitis is associated with elevated NADPH oxidase expression in the colon
      Shizhen Dong, Menglu Chen, Faliang Dai, Qingxia Xuan, Pan Chen, Dandan Feng, Lei Gao, Chendi Zhu, Yongchao Chang, Fong‐Fong Chu, Qiang Gao
      Journal of Cellular Biochemistry.2019; 120(6): 9230.     CrossRef
    • pVAX1-A20 alleviates colitis in mice by promoting regulatory T cells
      Tianyong Hu, Wenhui Hu, Li Ma, Xianhai Zeng, Jiangqi Liu, Baohui Cheng, Pingchang Yang, Shuqi Qiu, Gui Yang, Donghui Chen, Zhiqiang Liu
      Digestive and Liver Disease.2019; 51(6): 790.     CrossRef
    • Selective JAKinibs: Prospects in Inflammatory and Autoimmune Diseases
      Anniina T. Virtanen, Teemu Haikarainen, Juuli Raivola, Olli Silvennoinen
      BioDrugs.2019; 33(1): 15.     CrossRef
    • Regional variations in the use of biologics and immunomodulators among Korean patients with inflammatory bowel diseases
      Minkyung Han, Yoon Suk Jung, Jae Hee Cheon, Sohee Park
      Journal of Gastroenterology and Hepatology.2019; 34(7): 1166.     CrossRef
    • Red wine extract preserves tight junctions in intestinal epithelial cells under inflammatory conditions: implications for intestinal inflammation
      Carla Nunes, Víctor Freitas, Leonor Almeida, João Laranjinha
      Food & Function.2019; 10(3): 1364.     CrossRef
    • Mesenchymal Stem Cells Improve Rheumatoid Arthritis Progression by Controlling Memory T Cell Response
      Noymar Luque-Campos, Rafael A. Contreras-López, María Jose Paredes-Martínez, Maria Jose Torres, Sarah Bahraoui, Mingxing Wei, Francisco Espinoza, Farida Djouad, Roberto Javier Elizondo-Vega, Patricia Luz-Crawford
      Frontiers in Immunology.2019;[Epub]     CrossRef
    • Risk of Anxiety and Depression in Patients with Inflammatory Bowel Disease: A Nationwide, Population-Based Study
      Kookhwan Choi, Jaeyoung Chun, Kyungdo Han, Seona Park, Hosim Soh, Jihye Kim, Jooyoung Lee, Hyun Lee, Jong Im, Joo Kim
      Journal of Clinical Medicine.2019; 8(5): 654.     CrossRef
    • Real-world incidence of inflammatory bowel disease among patients with other chronic inflammatory diseases treated with interleukin-17a or phosphodiesterase 4 inhibitors
      Bruno Emond, Lorie A. Ellis, Soumya D. Chakravarty, Martin Ladouceur, Patrick Lefebvre
      Current Medical Research and Opinion.2019; 35(10): 1751.     CrossRef
    • Anti-inflammatory effects of apocynin on dextran sulfate sodium-induced mouse colitis model
      Young-Jae Hwang, Seung-Joo Nam, Wanjoo Chun, Song In Kim, Sung Chul Park, Chang Don Kang, Sung Joon Lee, Partha Mukhopadhyay
      PLOS ONE.2019; 14(5): e0217642.     CrossRef
    • Micronutrient Absorption and Related Outcomes in People with Inflammatory Bowel Disease: A Review
      Kyle Kilby, Holly Mathias, Lindsay Boisvenue, Courtney Heisler, Jennifer L. Jones
      Nutrients.2019; 11(6): 1388.     CrossRef
    • Therapeutic ultrasound attenuates DSS-induced colitis through the cholinergic anti-inflammatory pathway
      Natalia Schneider Nunes, Parwathy Chandran, Maggie Sundby, Fernanda Visioli, Fabiany da Costa Gonçalves, Scott Robert Burks, Ana Helena Paz, Joseph Alan Frank
      EBioMedicine.2019; 45: 495.     CrossRef
    • Inflammatory bowel disease - glycomics perspective
      Maja Hanić, Irena Trbojević-Akmačić, Gordan Lauc
      Biochimica et Biophysica Acta (BBA) - General Subjects.2019; 1863(10): 1595.     CrossRef
    • Genetic mapping of cell type specificity for complex traits
      Kyoko Watanabe, Maša Umićević Mirkov, Christiaan A. de Leeuw, Martijn P. van den Heuvel, Danielle Posthuma
      Nature Communications.2019;[Epub]     CrossRef
    • Diet and Nutrition in IBD—Progress and Gaps
      Maitreyi Raman, Subrata Ghosh
      Nutrients.2019; 11(8): 1740.     CrossRef
    • Comorbidities of hidradenitis suppurativa: A review of the literature
      Alexander Cartron, Marcia S. Driscoll
      International Journal of Women's Dermatology.2019; 5(5): 330.     CrossRef
    • Patients with Inflammatory Bowel Disease Are at an Increased Risk of Parkinson’s Disease: A South Korean Nationwide Population-Based Study
      Seona Park, Jihye Kim, Jaeyoung Chun, Kyungdo Han, Hosim Soh, Eun Ae Kang, Hyun Jung Lee, Jong Pil Im, Joo Sung Kim
      Journal of Clinical Medicine.2019; 8(8): 1191.     CrossRef
    • Paeoniflorin Prevents Intestinal Barrier Disruption and Inhibits Lipopolysaccharide (LPS)-Induced Inflammation in Caco-2 Cell Monolayers
      Xi-Xi Wu, Xie-Lin Huang, Ru-Ru Chen, Tang Li, Hua-Jun Ye, Wei Xie, Zhi-Ming Huang, Gao-Zhong Cao
      Inflammation.2019; 42(6): 2215.     CrossRef
    • The perceptions of natural compounds against dipeptidyl peptidase 4 in diabetes: from in silico to in vivo
      Shian-Ren Lin, Chia-Hsiang Chang, May-Jwan Tsai, Henrich Cheng, Jian-Chyi Chen, Max K. Leong, Ching-Feng Weng
      Therapeutic Advances in Chronic Disease.2019;[Epub]     CrossRef
    • Anti‐inflammatory effects of eriocitrin against the dextran sulfate sodium–induced experimental colitis in murine model
      Gang Guo, Wen Shi, Feiyu Shi, Wenqing Gong, Fanni Li, Guangju Zhou, Junjun She
      Journal of Biochemical and Molecular Toxicology.2019;[Epub]     CrossRef
    • Subsets of mononuclear phagocytes are enriched in the inflamed colons of patients with IBD
      Hong Liu, Suryasarathi Dasgupta, Yu Fu, Brandi Bailey, Christian Roy, Eric Lightcap, Benjamin Faustin
      BMC Immunology.2019;[Epub]     CrossRef
    • Mesenchymal Stem Cell-Derived Exosomes and Other Extracellular Vesicles as New Remedies in the Therapy of Inflammatory Diseases
      Carl Randall Harrell, Nemanja Jovicic, Valentin Djonov, Nebojsa Arsenijevic, Vladislav Volarevic
      Cells.2019; 8(12): 1605.     CrossRef
    • Integrating omics for a better understanding of Inflammatory Bowel Disease: a step towards personalized medicine
      Manoj Kumar, Mathieu Garand, Souhaila Al Khodor
      Journal of Translational Medicine.2019;[Epub]     CrossRef
    • Ulcerative colitis outcomes research in Japan: protocol for an observational prospective cohort study of YOURS (YOu and Ulcerative colitis: Registry and Social network)
      Hajime Yamazaki, Katsuyoshi Matsuoka, Jovelle Fernandez, Toshifumi Hibi, Mamoru Watanabe, Tadakazu Hisamatsu, Shunichi Fukuhara
      BMJ Open.2019; 9(9): e030134.     CrossRef
    • Circular RNA expression alterations in colon tissues of Crohn's disease patients
      Yu Qiao, Chen Cai, Jun Shen, Qing Zheng, Zhi Ran
      Molecular Medicine Reports.2019;[Epub]     CrossRef
    • Frondanol, a Nutraceutical Extract from Cucumaria frondosa, Attenuates Colonic Inflammation in a DSS-Induced Colitis Model in Mice
      Sandeep B. Subramanya, Sanjana Chandran, Saeeda Almarzooqi, Vishnu Raj, Aisha Salem Al Zahmi, Radeya Ahmed Al Katheeri, Samira Ali Al Zadjali, Peter D. Collin, Thomas E. Adrian
      Marine Drugs.2018; 16(5): 148.     CrossRef
    • Beyond the TNF-α Inhibitors: New and Emerging Targeted Therapies for Patients with Axial Spondyloarthritis and their Relation to Pathophysiology
      Susanne Juhl Pedersen, Walter P. Maksymowych
      Drugs.2018; 78(14): 1397.     CrossRef
    • Pleiotropic Immune Functions of Chemokine Receptor 6 in Health and Disease
      Ranmali Ranasinghe, Rajaraman Eri
      Medicines.2018; 5(3): 69.     CrossRef
    • T-cell immune response against cytomegalovirus in peripheral blood and colonic mucosa from ulcerative colitis and Crohn's disease patients
      Seung-Joo Nam, Eun Sun Kim, Yoon Tae Jeen
      Intestinal Research.2018; 16(1): 160.     CrossRef
    • CCR6–CCL20-Mediated Immunologic Pathways in Inflammatory Bowel Disease
      Ranmali Ranasinghe, Rajaraman Eri
      Gastrointestinal Disorders.2018; 1(1): 15.     CrossRef
    • Vitamin A supplementation decreases disease activity index in patients with ulcerative colitis: A randomized controlled clinical trial
      Kourosh Masnadi Shirazi, Zeinab Nikniaz, Arman Masnadi Shirazi, Mohammadreza Rohani
      Complementary Therapies in Medicine.2018; 41: 215.     CrossRef
    • CCR6–CCL20 Axis in IBD: What Have We Learnt in the Last 20 Years?
      Ranmali Ranasinghe, Rajaraman Eri
      Gastrointestinal Disorders.2018; 1(1): 57.     CrossRef
    • Increased end-stage renal disease risk in patients with inflammatory bowel disease: A nationwide population-based study
      Seona Park, Jaeyoung Chun, Kyung-Do Han, Hosim Soh, Kookhwan Choi, Ji Hye Kim, Jooyoung Lee, Changhyun Lee, Jong Pil Im, Joo Sung Kim
      World Journal of Gastroenterology.2018; 24(42): 4798.     CrossRef
    • Commensal Bacteria-Specific CD4+ T Cell Responses in Health and Disease
      Chiara Sorini, Rebeca F. Cardoso, Nicola Gagliani, Eduardo J. Villablanca
      Frontiers in Immunology.2018;[Epub]     CrossRef
    • Association of Perianal Fistulas with Clinical Features and Prognosis of Crohn's Disease in Korea: Results from the CONNECT Study
      Jaeyoung Chun, Jong Pil Im, Ji Won Kim, Kook Lae Lee, Chang Hwan Choi, Hyunsoo Kim, Jae Hee Cheon, Byong Duk Ye, Young-Ho Kim, You Sun Kim, Yoon Tae Jeen, Dong Soo Han, Won Ho Kim, Joo Sung Kim
      Gut and Liver.2018; 12(5): 544.     CrossRef
    • Impact of Autophagy of Innate Immune Cells on Inflammatory Bowel Disease
      Tomoya Iida, Yoshihiro Yokoyama, Kohei Wagatsuma, Daisuke Hirayama, Hiroshi Nakase
      Cells.2018; 8(1): 7.     CrossRef
    • IMMUNOREGULATORY PROFILE OF MICROSYMBIONTS OF THE INTESTINAL HUMAN BIOTOPE
      O. V. Bukharin, I. N. Chainikova, E. V. Ivanova, N. B. Perunova, T. A. Bondarenko, A. I. Smolyagin
      Journal of microbiology, epidemiology and immunobiology.2018; 95(4): 42.     CrossRef

    • PubReader PubReader
    • ePub LinkePub Link
    • Cite
      CITE
      export Copy Download
      Close
      Download Citation
      Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      Immunological pathogenesis of inflammatory bowel disease
      Intest Res. 2018;16(1):26-42.   Published online January 18, 2018
      Close
    • XML DownloadXML Download
    Figure
    • 0
    • 1
    • 2
    • 3
    Related articles
    Immunological pathogenesis of inflammatory bowel disease
    Image Image Image Image
    Fig. 1 Histology of colon tissue from IBD patients. (A) H&E of colon tissue from UC and CD patients (H&E, ×40). (B, C) Signal transducer and activator of transcription 3 (STAT3) and interleukin 17 (IL-17) expression in colon tissue from UC and CD patients (immunohistochemistry, ×200).
    Fig. 2 Pathogenic innate lymphoid cells (ILCs) and T cells in mucosal cells from IBD patients. ILCs have common properties with T effector cells. Lineage-specific transcription factors expressing ILCs or a subset of T cells produce the same cytokine. Natural cytotoxicity receptor (NCR)-expressing ILCs are classified differently from T cells. T-bet, T-box expressed in T cells; IFN, interferon; TNF, tumor necrosis factor; IL, interleukin; RORγt, retinoic acid receptor-related orphan receptor γt.
    Fig. 3 Present IBD therapeutic strategies that involve prevention of T cell and innate lymphoid cells (ILC) production or their inhibition. T cells and ILCs have a common therapeutic target. Compared with classical IBD therapeutic agents, new therapeutic strategies may involve T cells; ILCs such as interleukin (IL)-23 and IL-12-, tumor necrosis factor (TNF)-, and integrin-targeting agents; and signal transducer and activator of transcription (STAT) inhibitors. NF, nuclear factor; AP-1, activator protein 1; cAMP, cyclic adenosine 3′:5′-monophosphate.
    Fig. 4 Reciprocal balance for intestinal immune homeostasis and inflammation. The normal state is mediated by a reciprocal balance between immune cells (Treg and Breg vs. Th17 and ILC1) and cytokines that are secreted to maintain the conditions in the intestine. However, an imbalance in immune cells leads to the destruction of intestinal epithelial cells and the invasion of commensal microbiota. This situation leads to the uncontrolled release of cytokines, which is a key event in the pathogenesis of IBD. Treg, regulatory T; Breg, regulatory B; ILC, innate lymphoid cells; DC, dendritic cell; IL, interleukin; NKT, natural killer T; Th17, T helper 17.
    Immunological pathogenesis of inflammatory bowel disease

    Intest Res : Intestinal Research
    Close layer
    TOP