Abstract

Preclinical human inflammatory bowel disease (IBD) mechanisms is one of 5 focus areas of the Challenges in IBD Research 2024 document, which also includes environmental triggers, novel technologies, precision medicine, and pragmatic clinical research. Herein, we provide a comprehensive overview of current gaps in inflammatory bowel diseases research that relate to preclinical research and deliver actionable approaches to address them with a focus on how these gaps can lead to advancements in IBD interception, remission, and restoration. The document is the result of multidisciplinary input from scientists, clinicians, patients, and funders and represents a valuable resource for patient-centric research prioritization. This preclinical human IBD mechanisms section identifies major research gaps whose investigation will elucidate pathways and mechanisms that can be targeted to address unmet medical needs in IBD. Research gaps were identified in the following areas: genetics, risk alleles, and epigenetics; the microbiome; cell states and interactions; barrier function; IBD complications (specifically fibrosis and stricturing); and extraintestinal manifestations. To address these gaps, we share specific opportunities for investigation for basic and translational scientists and identify priority actions.

Lay Summary

To address the unmet medical needs of patients with inflammatory bowel diseases (IBD) and move toward cures, preclinical human-relevant research must center on mechanistic questions pertinent to patients with IBD in the 3 areas of disease interception, remission, and restoration.

Introduction

To address the unmet medical needs of patients with inflammatory bowel diseases (IBDs) and move toward the ultimate goal of cure, preclinical human-relevant research is critical and must center on mechanistic questions relevant to patients with IBD in the 3 areas of disease: interception, remission, and restoration. Gaps previously highlighted in “Challenges in IBD Research 2019” in preclinical human IBD mechanisms included (1) triggers of immune response, (2) intestinal epithelial homeostasis and wound repair, (3) developmental and age-related pathophysiology, (4) biology of complications, (5) biological determinants of disease location, and (6) new therapies and response to treatment.1

Over the past 5 years, significant progress in these areas has been achieved. “Triggers of Immune Response,” called for greater research into the host, diet, and microbiome interactions as instigators for dysregulated immune responses associated with IBD. Significant strides were made in microbiome science through advancements in technology and analytic tools. In conjunction with multi-omic analyses of well-curated IBD patient cohorts,2 studies have shed light on how microbial metabolites shape intestinal homeostasis3 and how microbes influence the efficacy of widely used drugs for IBD4 among a multitude of other important discoveries. The application of single-cell and other -omic technologies to samples from patient cohorts and experimental model systems has advanced our understanding of the other previously identified gaps, specifically the epithelial and mesenchymal cell diversity and function in health and IBD.5–7 We also have gained insight into factors that shape age-related pathophysiology,8–11 the biologic determinants of disease location and complications,12,13 and drivers of resistance to antitumor necrosis factor (anti-TNF) therapy.14,15

Although substantial progress has been achieved, discoveries over the last 5 years have also revealed new gaps in knowledge that form the basis for the identified Challenges in IBD Research 2024 Preclinical Human IBD mechanisms priorities. Along with the other Challenges in IBD Research 2024 Work Groups, we place these gaps in knowledge within the context of disease interception (prevention of progression to the clinical phase), maintenance of remission, and restoration of mucosal homeostasis. The challenges identified herein are guided by the Crohn’s & Colitis Foundation’s mission to cure IBD and improve the quality of life for those affected by these diseases.

The new 2024 gaps encompass 5 areas of focus for IBD preclinical research (Figure 1). They are as follows: (1) genetics, risk alleles, and epigenetics investigations that leverage current knowledge to understand how these factors contribute to IBD development and how they may be targeted for personalized medicine approaches; (2) microbiome studies that are mechanistically oriented and prioritize the identification and validation of microbially rooted, remission-inducing, and resiliency-affording approaches that restore health to the IBD gut; (3) cell states and interactions research centered on understanding the interactions and modulation of immune cell responses and gut barrier function. Specifically, it is critical to close knowledge gaps in the contributions of nonimmune cells (such as fibroblasts and smooth muscle cells), cell states and interactions (immune and stromal), and other cells and tissues (mesentery, nerves, muscle) in initiating and driving pathobiology so that we can address the differences between predisease and remission states in IBD and identify potential points for therapeutic intervention; (4) Barrier function research remains underdeveloped as a target for therapeutic intervention and represents a tremendous opportunity to improve patient outcomes in interception, restoration, and remission. (5) Pathways to complications and extraintestinal manifestations: strictures, fistulas, and extraintestinal manifestations (EIMs) all cause significant morbidity in patients with IBD. A greater understanding of the mechanisms and pathways driving these will shape future interventions and therapies.

Current research gaps and priority actions toward elucidation of pathways and mechanisms that can be targeted to address unmet medical needs in IBD.
Figure 1.

Current research gaps and priority actions toward elucidation of pathways and mechanisms that can be targeted to address unmet medical needs in IBD.

The Challenges in IBD Research 2024 Preclinical Human IBD Mechanisms Work Group also identified 2 important concepts that touch all the gaps described. First, consideration to diversity in gender, ethnicity, age, and race must be a priority of all studies. There is an overarching need for expansion of research pertaining to patients across populations underrepresented in existing IBD cohorts to facilitate personalized medicine approaches. Second, improvements in preclinical models pertaining to characterization, availability, reliability, and consistency are also needed (vertebrate models, organoids, and multicellular cultures). In response to this, we direct attention to the Inflammatory Bowel Diseases journal’s recent call for basic science submissions focused on Translational Preclinical Models of IBD (https://academic.oup.com/ibdjournal/pages/basic-science-cfp).

The following sections will describe in detail the 5 gaps identified by the Challenges in IBD Research 2024 Preclinical Human IBD Mechanisms Work Group and how these gaps pertain to interception, remission, and restoration of IBD when appropriate. Some of these gaps and challenges clearly cross all 3 domains, such as genetics and the microbiome, while others focus primarily on one of the domains. In some cases, the rationale for the domains is similar, and multiple domains are covered together.

Gap 1. Genetics, Risk Alleles, and Epigenetics

Regarding interception, remission, and restoration, there is a need to leverage results from genomic studies to identify how individual or compound genetic variants contribute to IBD development and/or treatment response, as well as impact on restoration and may ultimately be targeted for personalized medicine approaches. Similarly, there are large knowledge gaps in understanding the role of epigenetics in driving IBD development and/or treatment response and the impact on restoration.

Genetics in IBD

Over the past 2 decades, genome-wide association (GWAS) studies have provided key insights into the biological underpinnings of IBD. Over 200 genetic loci have been linked to risk of disease.16–22 Importantly, many of the identified coding variants converge upon common pathways, act through specific cell types, and affect core mechanisms of mucosal immunity and intestinal epithelial barrier function. These points of convergence have become the focus of rigorous mechanistic studies,23 like the role of the NOD2 signaling pathway (including NOD2/CARD15, ATG16L1, CARD9, and RIPK2) in response to microbes and the interleukin (IL)-17/IL-23 immune axis in development of chronic inflammation. Mechanistic studies around GWAS-identified variants in genes such as IL-23R16 contributed to the basis for the development of currently used anti-IL-23 therapies (eg, ustekinumab, risankizumab, mirikizumab) and highlight the importance of preclinical research.

While GWAS studies have given clues into the underlying biology of IBD, the variants identified to date do not account for all cases. Moreover, IBD is a complex, most often polygenic disease. While polygenic risk scores provide a measure of disease risk, the combination of variants necessary to trigger disease remains elusive. Many disease-associated variants are in noncoding regions, making mechanistic studies challenging. As a call to action, human-derived cell culture models and innovative approaches such as massively parallel reporter assays (MPRA)24,25 and clustered regularly interspaced short palindromic repeats (CRISPR)-based technologies should be employed to conduct large-scale genetic screens, using both single-gene modification and combinatorial approaches, to link the hundreds of disease-associated variants, including those in noncoding regions of the genome, to their biological function within the landscape of IBD. Results from such screens will allow for generation of hypotheses that should then be further tested in preclinical cellular and in vivo models of relevance to IBD. This will allow for eventual identification of therapeutic intervention points for interception and to impact remission and restoration.

Population-based differences in disease-associated risk alleles further complicate the role of genetics in IBD. For example, while variants of NOD2 are associated with disease in patients of European ancestry, they are typically not present in patients of African26 or South Asian ancestry.27 The expansion of GWAS studies to populations of non-European ancestry in recent years has uncovered significant differences in disease-associated variants26 outside of NOD2. Variants that are extremely rare and undetectable in patients of European ancestry could be identified in future studies focusing on different patient populations, giving new biological insight into disease and moving us closer to personalized medicine approaches. As a call to action, investigators should determine genetic-based mechanisms involved through analysis of data stemming from diverse patient populations, and through conducting experiments with these patient-derived samples (eg, cells, organoids), genetically engineered cells, and animals to identify specific mechanisms implicated in driving IBD and/or treatment response, restoration, and identification of potential drug targets. Finally, leveraging genetics to understand heterogeneity in disease course and response to treatment remains a significant gap in understanding disease biology. Why do some patients develop fistulizing disease or perianal disease? Why do some patients respond well to treatments while others relapse? A better understanding of how polygenic risk translates into specific disease phenotypes will help stratify patients, improve the use of current therapies, and accelerate the development of novel, personalized diagnostic and therapeutic approaches (see Challenges in IBD 2024 Precision Medicine Work Group Report). As a call to action, researchers should utilize data from precision medicine biomarker studies implicating epigenetic mechanisms and clinical studies implicating genotypic risk groups more likely to experience medication failure or response to particular therapies to generate and test mechanistic hypotheses in preclinical experimental systems.

Epigenetics in IBD

If genetic variants were the only drivers of IBD, one would expect that concordance for identical twins would reach 100%. While twins are more likely to both develop IBD, concordance only reaches 50%,28 suggesting a strong environmental component to disease (see Challenges in IBD 2024 Environmental Triggers Work Group Report). Gene activity can be adjusted throughout life, with no change to the DNA sequence, by modifying the accessibility of genes. These epigenetic modifications can alter host biology in response to the environment and are emerging as key players in human disease, including IBD. Epigenetic modifications fall into 3 classes: histone modifications, expression of noncoding RNAs, and DNA methylation (DNAm). Specific changes in DNAm patterns have been identified in the intestinal epithelium of pediatric IBD patients and correlate with disease outcomes.29 DNA methylation is largely stable in ex vivo patient-derived organoid models,29 enabling the conduct of mechanistic studies on how changes in DNAm affect downstream cellular function. These studies will determine whether epigenetic modifications and genetic risk alleles converge upon common pathways or cellular processes that can be assessed in preclinical models and ultimately therapeutically targeted.

Precision medicine biomarker studies have also shown that DNAm of specific genetic loci is stable in peripheral blood mononuclear cells (PBMCs) of IBD patients.30 Results from such studies should be independently validated and used to generate and subsequently test mechanistic hypotheses in preclinical experimental systems. These studies may be used in the future to help diagnose, stratify, and predict treatment outcomes of patients, moving toward a personalized or precision medicine approach. Ultimately, the identification of the environmental factors causing DNAm changes as well as the pathways and components involved utilizing preclinical model systems will reveal points that can be perturbed to prevent disease in the subset of patients where epigenetics plays an outsized role.

Gap 2. Microbiome

In 2019, the microbiome was contextualized, from the perspective of the Challenges in IBD 2019 Preclinical Work Group, as a trigger of immune responses in IBD.1 Now our current vision has expanded. The microbiome or microbiota, in the context of IBD, is the constellation of archaea, bacteria, fungi, viruses, protists, and helminths that inhabit the body and shape resistance and susceptibility to IBD disease onset, symptoms, flares, remission, and response to treatment. Microbiome science overall and as it is related to IBD has grown tremendously since the Challenges in IBD Research report was published in May 2019.1,31–34 Developments in microbiome studies are driven by expansion of well-curated human cohorts with a gut microbiome focus, advancements in microbiome data generation and analysis tools (especially regarding metabolomics) applied to IBD, and innovations in preclinical models.2–4,35–48 In this gap section, we highlight recent impactful discoveries, existing knowledge gaps, and calls to action regarding IBD and the microbiome and provide ideas and concepts for how the microbiome can be approached using preclinical models, first for IBD interception and then for IBD remission and restoration.

Interception

A knowledge gap persists in the fundamental understanding of how the microbiome influences disease initiation in both Crohn’s disease (CD), ulcerative colitis (UC), and microscopic colitis. Intrinsic to this gap is the need for a more mechanistic understanding of the microbial–host interactome to identify individuals at risk for developing IBD. Is one microbe the culprit, or rather, how does one microbe or many microbes act in a consortium to trigger IBD symptom onset? What are the consequences of particular microbial behaviors (biogeography, metabolite production, interactions with other host or microbial cells) for a host with a particular susceptibility under certain environmental conditions? Are instigating microbes bacterial, fungal, viral, or all of the above, and what are their features, metabolite or structural, that are problematic for some hosts and why? While there has been significant microbiome characterization in newly diagnosed and established IBD patients while they are symptomatic and in remission with appropriate healthy control populations,2,49,50 building cohorts of individuals at risk for IBD prior to symptom development has been more challenging. To begin to resolve this biological complexity, The Crohn’s and Colitis Canada Genetic, Environmental, Microbial (GEM) Project51 built prospective cohorts studying healthy first-degree relatives of CD patients. A recent study from this consortium analyzed stool microbiome data from these individuals prior to the onset of CD to generate microbiome-based IBD risk scores.52 This cohort is and will be immensely useful to IBD researchers and patients alike53–55 and should direct the efforts of basic and translational IBD investigators so that they mechanistically interrogate the microbial–host signaling features and pathways that are inflection points for manifestation of IBD symptoms.

We seek to call investigators to action to identify and validate microbiome bioactive features and their modulators. Our definition of features encompasses specific strains of bacteria, fungi, viruses, microbial pathways, biosynthetic gene clusters, and metabolites that are viable targets for either preventing symptomatic IBD or sustaining health in individuals at risk for IBD. Beyond identification and validation of such features, we also encourage the development of strategies to provision these beneficial features or remediate them. Our working definition of bioactive is holistic and integrated with the other gaps put forward by this group. Bioactivity can be immunoregulatory such as the recently elucidated function of certain bile acid metabolites56,57 or inulin elicited-microbiome metabolites.58 However, bioactivity is not restricted to traditional immune cell populations; effects on the epithelial barrier, specific cell populations within in the epithelium, enteric neurons, and stromal cell populations are welcome areas of investigation when and where robust assays and measures are available.

Specific actions we encourage are development of preclinical pipelines to prioritize microbiome bioactive features that precipitate symptomatic IBD and that can be targeted for interception. Prioritization workflows can encompass advancement in mouse models bearing human IBD risk alleles combined with the use of gnotobiotics and human microbial communities45,46 in the context of microbial feature testing, more reductionist methods, such as organoid-bacterial coculture systems,59,60 that enable the growth of anaerobic bacteria, or computational pipelines paired with cell-based interrogative screening assays of features.

Additional interception approaches we endorse draw from the disciplines of immunology and infectious diseases, specifically vaccinology. Vaccination remains the single most effective strategy to prevent or mitigate the severity of bacterial and viral infections. If the symptoms of IBD or its triggers are rooted in the microbiome (be it bacterial, viral, or fungal), vaccine approaches (either quasi-personalized or bacterial-straining targeting broadly) may be an option, as technology to generate RNA-based vaccines is now within reach of many laboratories. Recently, data analyses from the GEM projects highlighted that Ruminococcus torques is a key contributor to the IBD microbial risk score they identified in first-degree relatives of CD patients.52  Ruminococcus torques is frequently observed in microbiome studies of IBD, as is another member of its genus, R. gnavus, for which certain strains in particular have robust IBD associations.61,62 Beyond these ruminococci, adherent and invasive Escherichia coli have longstanding and well validated associations with ileal CD.63 Rather than enumerate strains associated with risk for IBD or potential vaccine antigens, we encourage vaccine development approaches as an interception or prevention strategy of interest and preclinical models provide ample opportunities for testing such vaccine-based approaches. Adjacent to vaccine strategies would be the further optimization of phage-based approaches in preclinical models for IBD interception.64,65 The opportunity to introduce phages to reverse a pro-inflammatory microbial environment before IBD develops in at-risk individuals is also appealing.

In summary, robust and innovative preclinical systems investigating the microbiome and IBD focused on the topics described previously will be pivotal for effective translation of preventive strategies for IBD interception.

Remission and Restoration

While cure is the Crohn’s & Colitis Foundation’s primary goal, achieving remission and restoration is the penultimate step in that path. Over the past 5 years, advancements in microbiome science as cited previously have identified specific microbes and microbial factors associated with mucosal healing and maintenance of barrier function, yet a gulf remains between these handful of studies and effective translation for therapeutic approaches. Additionally, we still need to identify specific microbes and microbial products that can restore healthy host–microbial interactions in IBD. Fecal microbiome transplant and transfer of consortia have shown promise, and these approaches merit further refinement.66,67 A recent study of the dextran sodium sulfate (DSS) model of colonic injury and inflammation was laudable in its systematic approach for unraveling microbial drivers of the DSS weight loss and histology, providing insight for optimizing this accessible and quick model for more reproducible and robust remission and restoration studies.47 While there are clearly gaps in the knowledge of microbes and features that restore health, there are similar knowledge shortfalls in understanding host factors that influence microbial localization and behaviors that affect effective barrier function. Intelectin-1 influences the localization of Akkermansia mucinphilia, a mucus-loving bacteria, with immunomodulatory and barrier-modifying effects.68 At first glance, intelectin-1 might be seen as a beneficial host defensive factor, but experiments in preclinical models have revealed that its excessive production in IBD results in disadvantageous effects for the epithelial barrier and mucus layers.

When we observe host or microbial features that are differentially abundant in active IBD, it can be difficult to predict whether there is too little or too much of the factor for the host. Preclinical models are pivotal in resolving such Goldilocks zone questions and whether a feature is ultimately beneficial or pathogenic and under what conditions. We encourage preclinical investigators to generate hypotheses from existing and emerging data sets in human cell systems and humanized animal models to identify specific microbes, microbial features, and their host modifiers to restore a healthy host–microbial status. These investigations can be grounded in combining in silico approaches with preclinical in vitro and in vivo models. For example, when one identifies an inflammatory and wound-healing program or cell state signature, as was recently done to better understand stricturing in pediatric Crohn’s disease,69 such signatures can be compared with microbial metabolite cell-screening data sets to identify shared differentially expressed gene signatures. Such approaches can prioritize molecules for additional testing in primary cell cultures or even relatively simple cell line wound-healing assays to winnow the list of bioactives prior to in vivo testing in models humanized from a human genetics or microbiome perspective.

We acknowledge that nonbacterial members of the microbiota have often been understudied in IBD in the context of remission and restoration. A recent study sought to rectify that knowledge gap and uncovered that Debaryomyces hansenii was enriched in intestinal wounds of mice that were incompletely healed and also in inflamed CD patient biopsies.70 The investigators went on to show that D. hansenii impaired wound healing via a macrophage-type 1 interferon-CCL5 program. Additional studies have profiled the mycobiome of well-phenotyped CD and UC patients with extensive outcome data.71 This recent study is quite useful for the IBD community. However, more attention to these fascinating microbes regarding their roles in IBD etiopathogenesis is warranted, specifically with a focus on refining understanding of fungi in IBD for improved remission and restoration outcomes in patients via preclinical investigations that embrace reverse translation methods.72,73 The human virome is also very important, and we anticipate breakthroughs in this area from the NIH-funded initiative on this topic. Regarding research opportunities for preclinical investigators, this Preclinical Work Group prioritizes identification and validation of microbially rooted, remission-inducing, and resiliency-affording approaches that restore health to the IBD gut.

Gap 3. Cell States and Interactions

The maintenance of mucosal homeostasis requires a delicate balance of cell states and cellular interactions of the different components within the intestine including immune, stromal, epithelial, and muscle cells, as well as the enteric nervous system (ENS). Of these, how the dysregulation of the immune and epithelial components contribute to IBD onset, progression, and remission have been the most studied. However, data suggest that other cell types such as stromal cells (fibroblasts, endothelial cells, and smooth muscle cells) are critical to the intricate balance between tolerance and inflammation.6,15,74 Yet, there is limited knowledge about which cell types are important, how they communicate with one another, where they are located, and how they are remodeled during disease. There is also a lack of understanding of how adjacent tissues, such as the mesentery, contribute to IBD. Finally, it is almost entirely unknown whether cell states return to pre-IBD baseline during remission, or alternatively a new baseline is reached after experiencing inflammation. Are there IBD-related epigenetic alterations to these cells? Do these cells return to their pre-inflammatory niches with restoration of the cell-cell communication to homeostatic baseline? Additionally, is there a retained “memory” of the inflammatory state beyond epigenetic regulations and rather through a gut-brain axis as suggested by Koren et al, where areas of the brain store and retrieve memory of immune responses?75 These key questions are further divided later on into those that affect IBD interception, remission, and restoration to identify gaps in our understanding and approaches to address them.

Interception

Several recent studies have used systems biology approaches to understand cell types, their abundances, and interactions in a combination of healthy and IBD tissues from UC and CD patients.6,7,14,15,74,76–85 To further improve our understanding of key drivers of IBD onset, there is a need to apply similar approaches to cohorts beyond those that have already developed IBD, to subjects at risk of developing the disease, as in the GEM project mentioned in Gap 2. This will ensure that states and cellular interactions that predict the onset of IBD can be identified with the goal of modulating these interactions to prevent disease initiation. Furthermore, it is important that these new cohorts are diverse to determine whether mechanisms are conserved between diverse patient populations. Alternatively, can unique signatures be identified that correlate with different ethnicities?

The suspension multi-omic data should be correlated with and complemented by special techniques both at the transcriptional and/or at the protein level. Commercialization of these techniques (Visium by 10X genomics,86 GeoMx87 and CosMx by Nanostring Technologies,88 MERFISH,89 and imaging mass cytometry) has enhanced our understanding of tissue architecture, cellular locations, and cell-cell crosstalk. With the improvement in their resolution and analysis pipelines, it is important to apply these techniques to IBD to identify cellular neighborhoods and interactions that drive IBD onset.

Additionally, the ENS plays important roles in maintaining mucosal homeostasis that could potentially be critical to IBD onset. For example, studies have suggested that certain disease states, such as infectious colitis, drive “neurogenic inflammation” through altering neuropeptides released from neurons and cytokines from pericytes.90 Additional studies have suggested that regulatory T cells (Tregs) are found near and regulated by enteric neurons that themselves receive signals from the microbiome, suggesting a microbiome-ENS-immune circuit.91 Whether these interactions are perturbed in people at risk of IBD is unknown and should be investigated further. It is also known that stress impacts the intestinal system, where IBD flares can be linked to increased stress. An elegant study has recently suggested that the ENS mediates intestinal inflammation during episodes of chronic stress where elevated levels of glucocorticoids promote the generation of inflammatory glia that recruit TNF-producing monocytes to the intestine and additionally cause dysmotility.92

Finally, the mesentery, or the mesenteric fat, has been linked to IBD,93–96 to stricturing CD in particular.97 The mesentery contains numerous immune and nonimmune cell populations (endothelial cells and fibroblasts) that potentially could contribute to IBD pathogenesis and should be investigated further.

Remission

To improve our understanding of how remission can be achieved and maintained, we need to identify drivers of therapeutic responses and treatment failures. Little is known about the contribution of nonimmune cells, cell states and interactions, and other tissues (mesentery) in initiating and driving progression, loss of response, and postsurgical recurrence in patients with IBD. Key questions remain unanswered, such as are there particular “cellular signatures” before the initiation of therapy that could predict individuals’ responses to treatment, and are there signatures in the intestine or other tissue including blood that correlate with remission? As such, there is a need for a deeper understanding of disease states, those that lead to remission and those that predict response failure, to facilitate the identification of new pathways to maintain remission. Some of the recent scRNAseq data sets have begun to do just that and have identified signatures that correlate with TNF failure for example.15 Much more work is needed to identify similar signatures for other biologic and small molecule-based therapies and those that correlate with therapeutic responses and disease remission. This can be accomplished by using multi-omic data sets directly from patients on various medications where the responses to therapies are known. Moreover, there is a need to establish more sophisticated patient-derived organoid cocultured models where epithelial cells are cultured with other intestinal components such as immune cells or fibroblasts to resemble the in vivo system where drug responses can be optimized to establish drivers of therapeutic responses. Both the organoid and patient sample multi-omic data sets should be assessed for signatures that could then lead to hypothesis generation that can be evaluated in preclinical models.

Furthermore, work is required to understand how similar or different the intestinal landscape of remission is to healthy homeostasis. Is “memory” of inflammation retained in the cells of the intestine through epigenetic regulations, so that with certain triggers they are more likely to revert to the inflammatory states? Can these modifications be altered to bring the intestine closer to the “healthy” state?

Finally, it is critical to determine whether there are particular cellular defects in treatment-refractory patients. For example, do these patients have an epithelial defect that leads to intestinal barrier loss, and can this be identified through simple testing in the clinical setting? Please see the barrier dysfunction section for more details.

Restoration

There is also an incomplete understanding of the mechanisms that affect cell populations causing dysregulation and hampering gut restoration. Longitudinal, multi-omic data sets including suspension and spatial data of subjects in various stages of IBD and healthy controls should be used to determine whether restoration, or return to pre-inflammatory “healthy” state, is possible and if there are signatures during onset and remission that can predict full restoration. These data sets would be instrumental in identifying mechanisms affecting key tolerogenic populations such as Tregs and innate lymphoid cells (ILCs) that play a critical role in maintaining homeostasis and could be harnessed to restore balance.

Gap 4. Barrier Function

Decades of preclinical and patient data link intestinal barrier loss to IBD.98 Nevertheless, all available therapies target the immune system, either directly or indirectly. Despite the proliferation of these effective new therapies in recent years, there are tremendous opportunities to improve patient outcomes, and the epithelial barrier is an obvious target that has been under-explored.99

Interception

Nearly 40 years ago, the discovery that increased intestinal permeability (ie, barrier loss) is present in a subset of healthy first-degree relatives of CD patients100 led many to speculate that increased permeability might be a risk factor for IBD development. However, this was only recently validated, when it was shown that, among healthy CD relatives, increased intestinal permeability was associated with a 3-fold greater risk of developing IBD.101 Consistent with this, preclinical studies using mouse IBD models have shown that even partial barrier preservation delays disease.102–104 Together, these clinical and preclinical data suggest that interception (ie, prevention of IBD progression to a clinically evident phase) may be possible in at-risk individuals with increased intestinal permeability.

Remission and Restoration

In patients with established CD, multiple studies demonstrate that increased permeability during clinical remission is a marker of relapse within the next year.105–107 Similarly, data from preclinical studies show that barrier restoration at early time points slows progression and reduces severity of clinically evident disease.102–104,108,109 These data suggest that barrier loss may be a marker of relapse and that barrier restoration may sustain clinical remission. If effective in patients, barrier restoring therapies might also reduce use of biologics and other immune-targeted agents in terms of dose and frequency to lessen associated toxicities.

Clinical and preclinical studies demonstrate that many disease-reducing therapeutic interventions also restore barrier function. In most instances, including the infliximab-induced reductions of intestinal permeability in CD patients,110 effects on inflammatory disease, mucosal repair, and epithelial tight junctions are inextricable. Thus, the contributions of barrier restoration to these therapeutic responses have been impossible to determine. Nearly all studies have relied on probes that cannot distinguish mucosal injury-associated barrier loss from that caused by increased tight junction leak pathway permeability.111 Further, of the 2 routes across the tight junction (the leak and pore pathways), each can be differentially activated by immune signaling, have unique permeability profiles, and are regulated by distinct cellular processes.112 Nevertheless, permeability probes that detect pore pathway flux have only been used rarely in preclinical models and have not been applied to patient-based studies.111–113 Commonly used probes, including 4kD FITC-dextran, lactulose, mannitol, sucralose, fluorescein, and51Cr-EDTA, can detect both tight junction leak and unrestricted (epithelial damage) pathway barrier loss but cannot distinguish between these. Notably, all of these are too large to traverse pore pathway channels, such as those created by claudin-2.113

Despite progress in understanding molecular mechanisms of mucosal barrier loss,98,114–123 drugs that target the epithelium have not been developed. One factor related to this has been the absence of even rudimentary characterization of barrier loss (eg, tight junction-dependent vs epithelial damage-induced) in patients and preclinical animal models. This information is essential because the mechanisms are distinct. Therefore, an agent that promotes mucosal healing may not impact tight junction permeability, while conversely, a tight junction-targeted therapy would likely be ineffective in the setting of severe mucosal damage, where de-epithelialized areas lack intercellular junctions. Tools that discriminate between these and other processes that increase intestinal permeability are, therefore, desperately needed. Ideal probes would be (1) nonmetabolizable by mammals or bacteria; (2) able to efficiently cross the pore, leak, or unrestricted pathways (a pore pathway probe would, of course, also cross via the other 2 routes); (3) freely filtered at the glomerulus (to allow analysis in urine); (4) well-tolerated; and (5) easily analyzed (without complex assays or interference by endogenous materials). No probes that fit this description are available.111 If they were, it would be possible to determine pathways (ie, pore, leak, or unrestricted) responsible for observed changes in barrier function and accurately define and target molecular mechanisms of barrier loss.

Patient, animal, and in vitro data all confirm that barrier function is dynamic.124–127 Yet, normal variation, such as the interaction between diurnal variation and alcohol-induced barrier loss reported in mice,127 has not been considered in human studies or characterized in terms of distinct pathways. The availability of improved probes would, therefore, allow determination of how the specific time of day or other recurrent events, such as phase of the menstrual cycle in women, affect properties of all pathways that define the epithelial barrier. If used to assess at-risk individuals or disease status and therapeutic response in patients, it would be reasonable to ask whether repeated measures improve sensitivity or specificity. If so, how many measures? How often? Use could, for example, be modeled after repeated measurements of any clinical parameter, from blood pressure to fecal calprotectin. Integration of longitudinal permeability studies of healthy, at-risk relatives and IBD patients with parallel analyses of tissue or blood RNA and protein expression, immune markers, microbiome composition, stool metabolomics, other parameters, and clinical outcome may both provide insight into events and pathways associated with initial IBD development, remission failure, CD recurrence after surgery, and pouchitis in UC.

Association studies such as the longitudinal analyses proposed previously would greatly inform other essential studies, including determination of mechanisms that drive barrier loss, such as ER stress, hypoxia, and physiological or pathophysiological stimuli128–130 and developing approaches to restore epithelial function. Such data could ultimately enable personalized approaches that determine which patients are likely to benefit from therapies that reduce permeability of tight junction pore108 or leak104 pathways, limit mucosal damage,34,109,131–135 or augment repair.116,136,137

Overall, there is a critical need to build on and translate existing data to develop a deeper understanding of epithelial dysfunction in IBD. Beyond general descriptions of changes present in preclinical models, this must include detailed analysis of preclinical animal models and patient materials, alongside studies of mechanism-based interventions in preclinical in vitro and in vivo models and, ultimately, patients. Such efforts in the near term will make it possible to achieve the long-term goal of developing new therapies and treatment algorithms that improve outcomes in IBD patients and, potentially, prevent disease development in at-risk populations.

Gap 5. IBD Complications (focus on fibrosis and fistulas) and Extraintestinal Manifestations

IBD Complications

There is a need for a deeper understanding of the pathways and mechanisms involved in driving stricturing and penetrating CD and pathogenic remodeling.

Remission

More than half of CD patients will present with intestinal complications over the course of their disease, including the formation of strictures (fibrostenotic disease) and fistulae (penetrating disease). Fibrostenosis, characterized by bowel wall thickening, luminal narrowing, and prestenotic dilation, is a frequent complication in patients with CD occurring in 30% to 50% of CD patients within 10 years of disease onset.138 Despite the advances in the treatment of CD, current therapies do little to prevent or reverse stricture formation.139 The prevailing view of pathogenesis of fibrostenotic CD is that chronic inflammation and prolonged mucosal injury drives an aberrant repair response, resulting in the accumulation of intestinal myofibroblasts within the bowel wall and increased extracellular matrix (ECM) deposition and organ dysfunction. While aspects of this model may hold true, it does not account for the role of intestinal smooth muscle expansion and newly discovered stromal cell subsets in tissue remodeling. Histological analyses of strictures reveal significant expansion of the smooth muscle near regions with significant ECM deposition.140 The expansion of smooth muscle is reminiscent of tissue remodeling in vascular and airway diseases. Efforts to assess the impact of conserved mechanisms within smooth muscle that regulate growth and phenotype (ie, contractile vs synthetic) should be prioritized to identify new targets to address its expansion in fibrostenotic CD.

Advances in single-cell characterization have demonstrated that the cells of the intestinal stromal compartment demonstrate substantial heterogeneity.6,14 Furthermore, these cell subsets exhibit significant transcriptomic changes in response to inflammation6,14 and in patients with fibrostenotic disease.13 As a call to action, there is a need to characterize these stromal cell subsets and understand their respective roles in health and disease to identify new targets to dampen tissue remodeling processes in IBD.

In addition to the extracellular matrix (ECM)-producing properties of stromal cells, continued efforts are required to understand their role in inflammation. Intestinal myofibroblasts can be activated by a variety of inflammatory mediators and express several innate immune receptors, including toll-like receptors141 and the NOD-like receptors NOD1,141 NOD2,141 and NLRP3.142  NOD2 variants are associated with increased risk of developing fibrostenotic CD;143 however, our understanding of its role in stromal cells is very limited. Recently, NOD2 risk alleles were shown to contribute to aberrant CD14+-derived fibrocyte (circulating fibroblast-like cells) function, wherein accumulation of risk alleles was associated with increased expression of an activated fibroblast gene signature in the context of muramyl dipeptide stimulation.144 These findings highlight the need for continued study into the complex interplay between immune signaling pathways and fibrogenesis in stromal cells.

Significantly less is known about the pathogenesis of penetrating disease. Internal and perianal fistulae occur in 30% to 50% of CD patients. These abnormal passages from the intestine to other regions are often lined with mesenchymal-like cells in place of a typical epithelial layer.145 These cells, termed transitional cells, are thought to arise from the induction of epithelial-to-mesenchymal transition (EMT), in the context of aberrant epithelial repair processes.145 The inflammatory milieu of fistulae consists of TGF-β1/TGF-β2,145 TNF,146,147 and IL-13,148 all of which can induce genes related to EMT. In addition to inflammation, succinate, a microbe-derived metabolite, may contribute to the induction of EMT and fistulae formation.149 Tissue succinate levels and SUCNR1 (succinate receptor) were elevated in patients with penetrating disease compared with samples isolated from fibrostenotic disease and non-IBD controls. Furthermore, the authors demonstrated that succinate could trigger EMT through activation of the Wnt pathway.149 However, it is still unclear how succinate and tissue inflammation contribute to localized EMT and fistula formation. Expanding our understanding of the pathogenesis of fistulae has been significantly hindered by a lack of experimental models and the challenges associated with obtaining samples for primary cell isolation. In vitro models can be used to study the mechanism(s) involved in the induction of EMT; however, this is likely one step in the disease process. To address this gap, models that incorporate primary cells (eg, IECs, stromal cells, immune cells) in coculture (eg, intestinal organoids; gut-chip) along with conditions that mimic the inflammatory and microbial milieu of CD-associated fistulae are urgently needed. In this regard, an important advancement was the development of a personalized intestinal fibrosis model using human intestinal organoids derived from induced pluripotent stem cells that contains mesenchymal and epithelial cells.150 Additional improvements are required to optimize the system for use in therapeutic discovery and furthering our understanding of cell-cell interactions in the context of fibrosis.

Extraintestinal Manifestations

Crohn’s disease and UC are systemic diseases and can affect organ systems beyond the gastrointestinal tract. These extraintestinal manifestations (EIMs) of IBD can significantly impact patient health-related quality of life and may carry significant morbidity. Commonly affected organs include the skin, joints, and eyes, as well as the liver, lung, and pancreas. While some EIMs improve with the treatment of intestinal inflammation (erythema nodosum [EN], peripheral arthritis, episcleritis, and oral ulcers), some persist or progress independently (eg, ankylosing spondylitis [AS], primary sclerosing cholangitis (PSC), and uveitis).151

A panel of experts recently defined EIMs as “an inflammatory pathology in a patient with IBD that is located outside the gut and for which the pathogenesis is either dependent on extension/translocation of immune responses from the intestine, or is an independent inflammatory event perpetuated by IBD or that shares a common environmental or genetic predisposition with IBD.”152 Several important knowledge gaps remain in the mechanisms of EIMs that could lead to improved IBD patient remission. It is of particular interest to define the mechanisms that drive EIMs that progress despite resolution of intestinal inflammation. Novel discoveries may come from the use of advanced ‘omic analyses and single-cell resolution spatial technologies. Other guidance may come from deeper interrogation of existing knowledge. For example, IBD genetic risk factors overlap with AS, uveitis, PSC, pyoderma gangrenosum, and EN. Emerging evidence links the microbiome and microbial factors with AS, EN, and PSC.153 Some EIMs may respond to TNF-α or JAK inhibition but not other therapies, suggesting ties to these inflammatory pathways. Primary sclerosing cholangitis progression on the other hand is not halted by current anti-inflammatory therapies suggesting alternative mechanisms are at play. A few models for EN, spondylarthopathy, and PSC have been described,153–156 and more models of EIMs are needed. Finally, further discovery is needed into more recently described IBD EIMs including fatty liver disease and its complications.157,158 As a call to action, efforts should focus on generating novel or improved preclinical in vitro and in vivo experimental models that will facilitate testing hypotheses based on analysis of data stemming from expanded cohorts with the future goal of identifying potential drug targets that, when modulated, would eliminate the extraintestinal manifestation.

Summary Statement

The depth and breadth of preclinical IBD research discovery have rapidly accelerated in the last decade. In parallel, robust systems and resources for clinical-translational validation of preclinical findings continue to develop and are increasingly accessible. This includes the Crohn’s & Colitis Foundation’s Study of a Prospective Adult Research Cohort with IBD (SPARC-IBD), a curated database of nearly 7000 well-characterized (phenotyped) adult patients, with integrated clinical and self-reported outcome data, as well as serial biospecimens (tissue, blood, and stool).159 Investigators are encouraged to use these resources to investigate deeply, collaborate broadly, work across the gap areas, and integrate/apply discoveries from other inflammatory and neoplastic disease states to IBD. Together, these strategies will propel advancements in the field to advance novel, effective, and targeted interventions for interception and treatment of IBD.

Acknowledgments

The authors thank Brett Sonnenschein for the design of the figure; Orna Ehrlich for coordination with the publisher; Nicole Litwin for facilitating the public comments; Carol Chapman for facilitating the Patient Advisory Committee for the Challenges in IBD Research 2024 document; Robert Barrett, PhD, Peter Higgins, MD, Florian Rieder, MD, Gwendalyn Randolph, PhD, and Brent Polk, MD, for their insightful feedback; the attendees at the Crohn’s & Colitis Foundation Researcher Symposium held September 11-12, 2023, who provided invaluable feedback to inform the direction of our research gaps and priority actions.

Conflicts of Interest

W.S.G. is on the scientific advisory board of Empress Therapeutics, Freya Biosciences, Scipher Medicine, Sail Biosciences, and the Bill and Melinda Gates Foundation, all outside the current work. W.S.G.’s laboratory receives funding from Merck and Astellas. M.A.C. is a consultant or advisory board for Janssen, Pfizer, AbbVie, and Geneoscopy. M.A.C. has also received funding under a sponsored research agreement unrelated to the data in the paper from Incyte, Janssen, and Pfizer. S.A.H. is a scientific advisor for Access Nutrients. A.S. is an employee of Kymera Therapeutics. K.J. is an employee of Generate Biomedicines. A.H-L., C.A.H., A.C.M., and J.L.S. are employees of the Crohn’s & Colitis Foundation. All other authors have no conflicts of interest to disclose.

Supplement Sponsorship

This article appears as part of the supplement “Challenges in IBD Research 2024,” sponsored by the Crohn’s & Colitis Foundation.

References

1.

Pizarro
 
TT
,
Stappenbeck
 
TS
,
Rieder
 
F
, et al.  
Challenges in IBD research: preclinical human IBD mechanisms
.
Inflamm Bowel Dis.
 
2019
;
25
(
Suppl 2
):
S5
-
S12
. doi:10.1093/ibd/izz075

2.

Lloyd-Price
 
J
,
Arze
 
C
,
Ananthakrishnan
 
AN
, et al. ;
IBDMDB Investigators
.
Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases
.
Nature.
 
2019
;
569
(
7758
):
655
-
662
. doi:10.1038/s41586-019-1237-9

3.

Brown
 
EM
,
Ke
 
X
,
Hitchcock
 
D
, et al.  
Bacteroides-derived sphingolipids are critical for maintaining intestinal homeostasis and symbiosis
.
Cell Host Microbe.
 
2019
;
25
(
5
):
668
-
680.e7
. doi:10.1016/j.chom.2019.04.002

4.

Mehta
 
RS
,
Mayers
 
JR
,
Zhang
 
Y
, et al.  
Gut microbial metabolism of 5-ASA diminishes its clinical efficacy in inflammatory bowel disease
.
Nat Med.
 
2023
;
29
(
3
):
700
-
709
. doi:10.1038/s41591-023-02217-7

5.

Nyström
 
EEL
,
Martinez-Abad
 
B
,
Arike
 
L
, et al.  
An intercrypt subpopulation of goblet cells is essential for colonic mucus barrier function
.
Science.
 
2021
;
372
(
6539
):
eabb1590
. doi:10.1126/science.abb1590

6.

Kinchen
 
J
,
Chen
 
HH
,
Parikh
 
K
, et al.  
Structural remodeling of the human colonic mesenchyme in inflammatory bowel disease
.
Cell.
 
2018
;
175
(
2
):
372
-
386.e17
. doi:10.1016/j.cell.2018.08.067

7.

Parikh
 
K
,
Antanaviciute
 
A
,
Fawkner-Corbett
 
D
, et al.  
Colonic epithelial cell diversity in health and inflammatory bowel disease
.
Nature.
 
2019
;
567
(
7746
):
49
-
55
. doi:10.1038/s41586-019-0992-y

8.

Sun
 
Y
,
Yuan
 
S
,
Chen
 
X
, et al.  
The contribution of genetic risk and lifestyle factors in the development of adult-onset inflammatory bowel disease: a prospective cohort study
.
Am J Gastroenterol.
 
2023
;
118
(
3
):
511
-
522
. doi:10.14309/ajg.0000000000002180

9.

Kokkinou
 
E
,
Soini
 
T
,
Pandey
 
RV
, et al.  
The single-cell transcriptional landscape of innate and adaptive lymphocytes in pediatric-onset colitis
.
Cell Rep Med
.
2023
;
4
(
5
):
101038
. doi:10.1016/j.xcrm.2023.101038

10.

Elmentaite
 
R
,
Ross
 
ADB
,
Roberts
 
K
, et al.  
Single-cell sequencing of developing human gut reveals transcriptional links to childhood Crohn’s disease
.
Dev Cell.
 
2020
;
55
(
6
):
771
-
783.e5
. doi:10.1016/j.devcel.2020.11.010

11.

Mortha
 
A
,
Remark
 
R
,
Del Valle
 
DM
, et al.  
Neutralizing anti-granulocyte macrophage-colony stimulating factor autoantibodies recognize post-translational glycosylations on granulocyte macrophage-colony stimulating factor years before diagnosis and predict complicated Crohn’s disease
.
Gastroenterology.
 
2022
;
163
(
3
):
659
-
670
. doi:10.1053/j.gastro.2022.05.029

12.

Akhlaghpour
 
M
,
Haritunians
 
T
,
More
 
SK
, et al. ;
International IBD Genetics Consortium
.
Genetic coding variant in complement factor B (CFB) is associated with increased risk for perianal Crohn’s disease and leads to impaired CFB cleavage and phagocytosis
.
Gut.
 
2023
;
72
(
11
):
2068
-
2080
. doi:10.1136/gutjnl-2023-329689

13.

Mukherjee
 
PK
,
Nguyen
 
QT
,
Li
 
J
, et al.  
Stricturing Crohn’s disease single-cell RNA sequencing reveals fibroblast heterogeneity and intercellular interactions
.
Gastroenterology.
 
2023
;
165
(
5
):
1180
-
1196
. doi:10.1053/j.gastro.2023.07.014

14.

Smillie
 
CS
,
Biton
 
M
,
Ordovas-Montanes
 
J
, et al.  
Intra- and Inter-cellular rewiring of the human colon during ulcerative colitis
.
Cell.
 
2019
;
178
(
3
):
714
-
730.e22
. doi:10.1016/j.cell.2019.06.029

15.

Martin
 
JC
,
Chang
 
C
,
Boschetti
 
G
, et al.  
Single-cell analysis of Crohn’s disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy
.
Cell.
 
2019
;
178
(
6
):
1493
-
1508.e20
. doi:10.1016/j.cell.2019.08.008

16.

Duerr
 
RH
,
Taylor
 
KD
,
Brant
 
SR
, et al.  
A genome-wide association study identifies IL23R as an inflammatory bowel disease gene
.
Science.
 
2006
;
314
(
5804
):
1461
-
1463
. doi:10.1126/science.1135245

17.

Franke
 
A
,
McGovern
 
DPB
,
Barrett
 
JC
, et al.  
Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci
.
Nat Genet.
 
2010
;
42
(
12
):
1118
-
1125
. doi:10.1038/ng.717

18.

Barrett
 
JC
,
Hansoul
 
S
,
Nicolae
 
DL
, et al. ;
NIDDK IBD Genetics Consortium
.
Genome-wide association defines more than 30 distinct susceptibility loci for Crohn’s disease
.
Nat Genet.
 
2008
;
40
(
8
):
955
-
962
. doi:10.1038/ng.175

19.

McGovern
 
DPB
,
Gardet
 
A
,
Törkvist
 
L
, et al. ;
NIDDK IBD Genetics Consortium
.
Genome-wide association identifies multiple ulcerative colitis susceptibility loci
.
Nat Genet.
 
2010
;
42
(
4
):
332
-
337
. doi:10.1038/ng.549

20.

Anderson
 
CA
,
Boucher
 
G
,
Lees
 
CW
, et al.  
Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47
.
Nat Genet.
 
2011
;
43
(
3
):
246
-
252
. doi:10.1038/ng.764

21.

Sazonovs
 
A
,
Stevens
 
CR
,
Venkataraman
 
GR
, et al. ;
Belgium IBD Consortium
.
Large-scale sequencing identifies multiple genes and rare variants associated with Crohn’s disease susceptibility
.
Nat Genet.
 
2022
;
54
(
9
):
1275
-
1283
. doi:10.1038/s41588-022-01156-2

22.

de Lange
 
KM
,
Moutsianas
 
L
,
Lee
 
JC
, et al.  
Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease
.
Nat Genet.
 
2017
;
49
(
2
):
256
-
261
. doi:10.1038/ng.3760

23.

Graham
 
DB
,
Xavier
 
RJ.
 
Pathway paradigms revealed from the genetics of inflammatory bowel disease
.
Nature.
 
2020
;
578
(
7796
):
527
-
539
. doi:10.1038/s41586-020-2025-2

24.

Mouri
 
K
,
Guo
 
MH
,
de Boer
 
CG
, et al.  
Prioritization of autoimmune disease-associated genetic variants that perturb regulatory element activity in T cells
.
Nat Genet.
 
2022
;
54
(
5
):
603
-
612
. doi:10.1038/s41588-022-01056-5

25.

Klein
 
JC
,
Agarwal
 
V
,
Inoue
 
F
, et al.  
A systematic evaluation of the design and context dependencies of massively parallel reporter assays
.
Nat Methods.
 
2020
;
17
(
11
):
1083
-
1091
. doi:10.1038/s41592-020-0965-y

26.

Somineni
 
HK
,
Nagpal
 
S
,
Venkateswaran
 
S
, et al.  
Whole-genome sequencing of African Americans implicates differential genetic architecture in inflammatory bowel disease
.
Am J Hum Genet.
 
2021
;
108
(
3
):
431
-
445
. doi:10.1016/j.ajhg.2021.02.001

27.

Ueda
 
M
,
Oike
 
F
,
Ogura
 
Y
, et al.  
Long-term outcomes of 600 living donor liver transplants for pediatric patients at a single center
.
Liver Transpl.
 
2006
;
12
(
9
):
1326
-
1336
. doi:10.1002/lt.20826

28.

Halme
 
L
,
Paavola-Sakki
 
P
,
Turunen
 
U
, et al.  
Family and twin studies in inflammatory bowel disease
.
World J Gastroenterol.
 
2006
;
12
(
23
):
3668
-
3672
. doi:10.3748/wjg.v12.i23.3668

29.

Howell
 
KJ
,
Kraiczy
 
J
,
Nayak
 
KM
, et al.  
DNA methylation and transcription patterns in intestinal epithelial cells from pediatric patients with inflammatory bowel diseases differentiate disease subtypes and associate with outcome
.
Gastroenterology.
 
2018
;
154
(
3
):
585
-
598
. doi:10.1053/j.gastro.2017.10.007

30.

Joustra
 
V
,
Li Yim
 
AYF
,
Hageman
 
I
, et al.  
Long-term temporal stability of peripheral blood DNA methylation profiles in patients with inflammatory bowel disease
.
Cell Mol Gastroenterol Hepatol
.
2023
;
15
(
4
):
869
-
885
. doi:10.1016/j.jcmgh.2022.12.011

31.

Ho
 
SM
,
Lewis
 
JD
,
Mayer
 
EA
, et al.  
Challenges in IBD research: environmental triggers
.
Inflamm Bowel Dis.
 
2019
;
25
(
Suppl 2
):
S13
-
S23
. doi:10.1093/ibd/izz076

32.

Dhyani
 
M
,
Joshi
 
N
,
Bemelman
 
WA
, et al.  
Challenges in IBD research: novel technologies
.
Inflamm Bowel Dis.
 
2019
;
25
(
Suppl 2
):
S24
-
S30
. doi:10.1093/ibd/izz077

33.

Denson
 
LA
,
Curran
 
M
,
McGovern
 
DPB
, et al.  
Challenges in IBD research: precision medicine
.
Inflamm Bowel Dis.
 
2019
;
25
(
Suppl 2
):
S31
-
S39
. doi:10.1093/ibd/izz078

34.

Scott
 
FI
,
Rubin
 
DT
,
Kugathasan
 
S
, et al.  
Challenges in IBD research: pragmatic clinical research
.
Inflamm Bowel Dis.
 
2019
;
25
(
Suppl 2
):
S40
-
S47
. doi:10.1093/ibd/izz085

35.

Yang
 
C
,
Mogno
 
I
,
Contijoch
 
EJ
, et al.  
Fecal IgA levels are determined by strain-level differences in bacteroides ovatus and are modifiable by gut microbiota manipulation
.
Cell Host Microbe.
 
2020
;
27
(
3
):
467
-
475.e6
. doi:10.1016/j.chom.2020.01.016

36.

Lee
 
JWJ
,
Plichta
 
D
,
Hogstrom
 
L
, et al.  
Multi-omics reveal microbial determinants impacting responses to biologic therapies in inflammatory bowel disease
.
Cell Host Microbe.
 
2021
;
29
(
8
):
1294
-
1304.e4
. doi:10.1016/j.chom.2021.06.019

37.

Mallick
 
H
,
Franzosa
 
EA
,
Mclver
 
LJ
, et al.  
Predictive metabolomic profiling of microbial communities using amplicon or metagenomic sequences
.
Nat Commun.
 
2019
;
10
(
1
):
3136
. doi:10.1038/s41467-019-10927-1

38.

Fornelos
 
N
,
Franzosa
 
EA
,
Bishai
 
J
, et al.  
Growth effects of N-acylethanolamines on gut bacteria reflect altered bacterial abundances in inflammatory bowel disease
.
Nat Microbiol.
 
2020
;
5
(
3
):
486
-
497
. doi:10.1038/s41564-019-0655-7

39.

Quinn
 
RA
,
Melnik
 
AV
,
Vrbanac
 
A
, et al.  
Global chemical effects of the microbiome include new bile-acid conjugations
.
Nature.
 
2020
;
579
(
7797
):
123
-
129
. doi:10.1038/s41586-020-2047-9

40.

Zhang
 
Y
,
Bhosle
 
A
,
Bae
 
S
, et al.  
Discovery of bioactive microbial gene products in inflammatory bowel disease
.
Nature.
 
2022
;
606
(
7915
):
754
-
760
. doi:10.1038/s41586-022-04648-7

41.

Geistlinger
 
L
,
Mirzayi
 
C
,
Zohra
 
F
, et al.  
BugSigDB captures patterns of differential abundance across a broad range of host-associated microbial signatures
.
Nat Biotechnol.
 
2023
. doi:10.1038/s41587-023-01872-y

42.

Ma
 
S
,
Shungin
 
D
,
Mallick
 
H
, et al.  
Population structure discovery in meta-analyzed microbial communities and inflammatory bowel disease using MMUPHin
.
Genome Biol.
 
2022
;
23
(
1
):
208
. doi:10.1186/s13059-022-02753-4

43.

Blanco-Míguez
 
A
,
Beghini
 
F
,
Cumbo
 
F
, et al.  
Extending and improving metagenomic taxonomic profiling with uncharacterized species using MetaPhlAn 4
.
Nat Biotechnol.
 
2023
;
41
(
11
):
1633
-
1644
. doi:10.1038/s41587-023-01688-w

44.

Gentry
 
EC
,
Collins
 
SL
,
Panitchpakdi
 
M
, et al.  
Reverse metabolomics for the discovery of chemical structures from humans
.
Nature.
 
2023
;
626
(
7998
):
419
-
426
. doi:10.1038/s41586-023-06906-8

45.

Lavoie
 
S
,
Conway
 
KL
,
Lassen
 
KG
, et al.  
The Crohn’s disease polymorphism, ATG16L1 T300A, alters the gut microbiota and enhances the local Th1/Th17 response
.
Elife
.
2019
;
8
:
e39982
. doi:10.7554/eLife.39982

46.

Britton
 
GJ
,
Contijoch
 
EJ
,
Spindler
 
MP
, et al.  
Defined microbiota transplant restores Th17/RORγt+ regulatory T cell balance in mice colonized with inflammatory bowel disease microbiotas
.
Proc Natl Acad Sci U S A.
 
2020
;
117
(
35
):
21536
-
21545
. doi:10.1073/pnas.1922189117

47.

Forster
 
SC
,
Clare
 
S
,
Beresford-Jones
 
BS
, et al.  
Identification of gut microbial species linked with disease variability in a widely used mouse model of colitis
.
Nat Microbiol.
 
2022
;
7
(
4
):
590
-
599
. doi:10.1038/s41564-022-01094-z

48.

Poletti
 
M
,
Arnauts
 
K
,
Ferrante
 
M
,
Korcsmaros
 
T.
 
Organoid-based models to study the role of host-microbiota interactions in IBD
.
J Crohns Colitis.
 
2021
;
15
(
7
):
1222
-
1235
. doi:10.1093/ecco-jcc/jjaa257

49.

Kai
 
N
,
Qingsong
 
C
,
Kejia
 
M
, et al.  
An Inflammatory Bowel Diseases Integrated Resources Portal (IBDIRP)
.
Database (Oxford)
.
2024
;
2024
:
baad097
. doi:10.1093/database/baad097

50.

Jangi
 
S
,
Hsia
 
K
,
Zhao
 
N
, et al.  
Dynamics of the gut mycobiome in patients with ulcerative colitis
.
Clin Gastroenterol Hepatol.
 
2023
;
22
(
4
):
821
-
830.e7
. doi:10.1016/j.cgh.2023.09.023

51.

Coward
 
S
,
Benchimol
 
EI
,
Kuenzig
 
ME
, et al.  
The 2023 impact of inflammatory bowel disease in Canada: epidemiology of IBD
.
J. Can. Assoc. Gastroenterol.
 
2023
;
6
(
Suppl 2
):
S9
-
S15
. doi:10.1093/jcag/gwad004

52.

Raygoza Garay
 
JA
,
Turpin
 
W
,
Lee
 
SH
, et al. ;
CCC GEM Project Research Consortium
.
Gut Microbiome composition is associated with future onset of Crohn’s disease in healthy first-degree relatives
.
Gastroenterology.
 
2023
;
165
(
3
):
670
-
681
. doi:10.1053/j.gastro.2023.05.032

53.

Leibovitzh
 
H
,
Lee
 
SH
,
Xue
 
M
, et al. ;
CCC GEM Project Research Consortium
.
Altered gut microbiome composition and function are associated with gut barrier dysfunction in healthy relatives of patients with Crohn’s disease
.
Gastroenterology.
 
2022
;
163
(
5
):
1364
-
1376.e10
. doi:10.1053/j.gastro.2022.07.004

54.

Turpin
 
W
,
Dong
 
M
,
Sasson
 
G
, et al. ;
Crohn’s and Colitis Canada (CCC) Genetic, Environmental, Microbial (GEM) Project Research Consortium
.
Mediterranean-like dietary pattern associations with gut microbiome composition and subclinical gastrointestinal inflammation
.
Gastroenterology.
 
2022
;
163
(
3
):
685
-
698
. doi:10.1053/j.gastro.2022.05.037

55.

Leibovitzh
 
H
,
Lee
 
SH
,
Raygoza Garay
 
JA
, et al. ;
Crohn’s and Colitis Canada (CCC) Genetic, Environmental, Microbial (GEM) Project Research Consortium
.
Immune response and barrier dysfunction-related proteomic signatures in preclinical phase of Crohn’s disease highlight earliest events of pathogenesis
.
Gut.
 
2023
;
72
(
8
):
1462
-
1471
. doi:10.1136/gutjnl-2022-328421

56.

Paik
 
D
,
Yao
 
L
,
Zhang
 
Y
, et al.  
Human gut bacteria produce ΤΗ17-modulating bile acid metabolites
.
Nature.
 
2022
;
603
(
7903
):
907
-
912
. doi:10.1038/s41586-022-04480-z

57.

Hang
 
S
,
Paik
 
D
,
Yao
 
L
, et al.  
Bile acid metabolites control TH17 and Treg cell differentiation
.
Nature.
 
2019
;
576
(
7785
):
143
-
148
. doi:10.1038/s41586-019-1785-z

58.

Arifuzzaman
 
M
,
Won
 
TH
,
Li
 
TT
, et al. ;
JRI IBD Live Cell Bank Consortium
.
Inulin fibre promotes microbiota-derived bile acids and type 2 inflammation
.
Nature.
 
2022
;
611
(
7936
):
578
-
584
. doi:10.1038/s41586-022-05380-y

59.

Angus
 
HC
,
Urbano
 
PC
,
Laws
 
GA
, et al.  
An autologous colonic organoid-derived monolayer model to study immune: bacterial interactions in Crohn’s disease patients
.
Clin Transl Immunology
.
2022
;
11
(
8
):
e1407
. doi:10.1002/cti2.1407

60.

Günther
 
C
,
Winner
 
B
,
Neurath
 
MF
,
Stappenbeck
 
TS.
 
Organoids in gastrointestinal diseases: from experimental models to clinical translation
.
Gut.
 
2022
;
71
(
9
):
1892
-
1908
. doi:10.1136/gutjnl-2021-326560

61.

Hall
 
AB
,
Yassour
 
M
,
Sauk
 
J
, et al.  
A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients
.
Genome Med.
 
2017
;
9
(
1
):
103
. doi:10.1186/s13073-017-0490-5

62.

Henke
 
MT
,
Brown
 
EM
,
Cassilly
 
CD
, et al.  
Capsular polysaccharide correlates with immune response to the human gut microbe Ruminococcus gnavus
.
Proc Natl Acad Sci U S A.
 
2021
;
118
(
20
):
e2007595118
. doi:10.1073/pnas.2007595118

63.

Schirmer
 
M
,
Garner
 
A
,
Vlamakis
 
H
,
Xavier
 
RJ.
 
Microbial genes and pathways in inflammatory bowel disease
.
Nat Rev Microbiol.
 
2019
;
17
(
8
):
497
-
511
. doi:10.1038/s41579-019-0213-6

64.

Federici
 
S
,
Kredo-Russo
 
S
,
Valdés-Mas
 
R
, et al.  
Targeted suppression of human IBD-associated gut microbiota commensals by phage consortia for treatment of intestinal inflammation
.
Cell.
 
2022
;
185
(
16
):
2879
-
2898.e24
. doi:10.1016/j.cell.2022.07.003

65.

Titécat
 
M
,
Rousseaux
 
C
,
Dubuquoy
 
C
, et al.  
Safety and efficacy of an AIEC-targeted bacteriophage cocktail in a mice colitis model
.
J Crohns Colitis.
 
2022
;
16
(
10
):
1617
-
1627
. doi:10.1093/ecco-jcc/jjac064

66.

Lin
 
J
,
Xiong
 
J
,
Jin
 
Y
, et al.  
Fecal microbiota transplantation through transendoscopic enteral tubing for inflammatory bowel disease: high acceptance and high satisfaction
.
J Gastroenterol Hepatol.
 
2023
;
39
(
2
):
328
-
336
. doi:10.1111/jgh.16435

67.

Saleh
 
A
,
Parsa
 
S
,
Garza
 
M
,
Quigley
 
EMM
,
Abraham
 
BP.
 
The role of fecal microbiota transplantation in the induction of remission in ulcerative colitis
.
Dig Dis.
 
2023
;
41
(
4
):
656
-
665
. doi:10.1159/000529591

68.

Matute
 
JD
,
Duan
 
J
,
Flak
 
MB
, et al.  
Intelectin-1 binds and alters the localization of the mucus barrier-modifying bacterium Akkermansia muciniphila
.
J Exp Med.
 
2023
;
220
(
1
):
e20211938
. doi:10.1084/jem.20211938

69.

Haberman
 
Y
,
Minar
 
P
,
Karns
 
R
, et al.  
Mucosal inflammatory and wound healing gene programs reveal targets for stricturing behavior in pediatric Crohn’s disease
.
J Crohns Colitis.
 
2020
;
15
(
2
):
273
-
286
. doi:10.1093/ecco-jcc/jjaa166

70.

Jain
 
U
,
Ver Heul
 
AM
,
Xiong
 
S
, et al.  
Debaryomyces is enriched in Crohn’s disease intestinal tissue and impairs healing in mice
.
Science.
 
2021
;
371
(
6534
):
1154
-
1159
. doi:10.1126/science.abd0919

71.

Catalán-Serra
 
I
,
Thorsvik
 
S
,
Beisvag
 
V
, et al.  
Fungal microbiota composition in inflammatory bowel disease patients: characterization in different phenotypes and correlation with clinical activity and disease course
.
Inflamm Bowel Dis.
 
2023
:
izad289
. doi:10.1093/ibd/izad289

72.

Underhill
 
DM
,
Braun
 
J.
 
Fungal microbiome in inflammatory bowel disease: a critical assessment
.
J Clin Invest.
 
2022
;
132
(
5
):
e155786
. doi:10.1172/JCI155786

73.

Limon
 
JJ
,
Tang
 
J
,
Li
 
D
, et al.  
Malassezia is associated with Crohn’s disease and exacerbates colitis in mouse models
.
Cell Host Microbe.
 
2019
;
25
(
3
):
377
-
388.e6
. doi:10.1016/j.chom.2019.01.007

74.

Nayar
 
S
,
Cho
 
JH.
 
From single-target to cellular niche targeting in Crohn’s disease: intercepting bad communications
.
EBioMedicine
.
2021
;
74
:
103690
. doi:10.1016/j.ebiom.2021.103690

75.

Koren
 
T
,
Yifa
 
R
,
Amer
 
M
, et al.  
Insular cortex neurons encode and retrieve specific immune responses
.
Cell.
 
2021
;
184
(
25
):
6211
. doi:10.1016/j.cell.2021.11.021

76.

Uzzan
 
M
,
Martin
 
JC
,
Mesin
 
L
, et al.  
Ulcerative colitis is characterized by a plasmablast-skewed humoral response associated with disease activity
.
Nat Med.
 
2022
;
28
(
4
):
766
-
779
. doi:10.1038/s41591-022-01680-y

77.

Boland
 
BS
,
He
 
Z
,
Tsai
 
MS
, et al.  
Heterogeneity and clonal relationships of adaptive immune cells in ulcerative colitis revealed by single-cell analyses
.
Sci Immunol.
 
2020
;
5
(
50
):
eabb4432
. doi:10.1126/sciimmunol.abb4432

78.

Corridoni
 
D
,
Antanaviciute
 
A
,
Gupta
 
T
, et al.  
Single-cell atlas of colonic CD8+ T cells in ulcerative colitis
.
Nat Med.
 
2020
;
26
(
9
):
1480
-
1490
. doi:10.1038/s41591-020-1003-4

79.

Jaeger
 
N
,
Gamini
 
R
,
Cella
 
M
, et al.  
Single-cell analyses of Crohn’s disease tissues reveal intestinal intraepithelial T cells heterogeneity and altered subset distributions
.
Nat Commun.
 
2021
;
12
(
1
):
1921
. doi:10.1038/s41467-021-22164-6

80.

Yokoi
 
T
,
Murakami
 
M
,
Kihara
 
T
, et al.  
Identification of a unique subset of tissue-resident memory CD4+ T cells in Crohn’s disease
.
Proc Natl Acad Sci U S A.
 
2023
;
120
(
1
):
e2204269120
. doi:10.1073/pnas.2204269120

81.

Rosati
 
E
,
Rios Martini
 
G
,
Pogorelyy
 
MV
, et al.  
A novel unconventional T cell population enriched in Crohn’s disease
.
Gut.
 
2022
;
71
(
11
):
2194
-
2204
. doi:10.1136/gutjnl-2021-325373

82.

Maddipatla
 
SC
,
Kolachala
 
VL
,
Venkateswaran
 
S
, et al.  
Assessing cellular and transcriptional diversity of ileal mucosa among treatment-naïve and treated Crohn’s disease
.
Inflamm Bowel Dis.
 
2023
;
29
(
2
):
274
-
285
. doi:10.1093/ibd/izac201

83.

Huang
 
B
,
Chen
 
Z
,
Geng
 
L
, et al.  
Mucosal profiling of pediatric-onset colitis and IBD reveals common pathogenics and therapeutic pathways
.
Cell.
 
2019
;
179
(
5
):
1160
-
1176.e24
. doi:10.1016/j.cell.2019.10.027

84.

Mitsialis
 
V
,
Wall
 
S
,
Liu
 
P
, et al. ;
Boston Children’s Hospital Inflammatory Bowel Disease Center
.
Single-Cell analyses of colon and blood reveal distinct immune cell signatures of ulcerative colitis and Crohn’s disease
.
Gastroenterology.
 
2020
;
159
(
2
):
591
-
608.e10
. doi:10.1053/j.gastro.2020.04.074

85.

Swanson
 
E
,
Lord
 
C
,
Reading
 
J
, et al.  
Simultaneous trimodal single-cell measurement of transcripts, epitopes, and chromatin accessibility using TEA-seq
.
Elife
.
2021
;
10
:
e63632
. doi:10.7554/eLife.63632

86.

Ståhl
 
PL
,
Salmén
 
F
,
Vickovic
 
S
, et al.  
Visualization and analysis of gene expression in tissue sections by spatial transcriptomics
.
Science.
 
2016
;
353
(
6294
):
78
-
82
. doi:10.1126/science.aaf2403

87.

Merritt
 
CR
,
Ong
 
GT
,
Church
 
SE
, et al.  
Multiplex digital spatial profiling of proteins and RNA in fixed tissue
.
Nat Biotechnol.
 
2020
;
38
(
5
):
586
-
599
. doi:10.1038/s41587-020-0472-9

88.

He
 
S
,
Bhatt
 
R
,
Brown
 
C
, et al.  
High-plex imaging of RNA and proteins at subcellular resolution in fixed tissue by spatial molecular imaging
.
Nat Biotechnol.
 
2022
;
40
(
12
):
1794
-
1806
. doi:10.1038/s41587-022-01483-z

89.

Xia
 
C
,
Fan
 
J
,
Emanuel
 
G
,
Hao
 
J
,
Zhuang
 
X.
 
Spatial transcriptome profiling by MERFISH reveals subcellular RNA compartmentalization and cell cycle-dependent gene expression
.
Proc Natl Acad Sci U S A.
 
2019
;
116
(
39
):
19490
-
19499
. doi:10.1073/pnas.1912459116

90.

Manion
 
J
,
Musser
 
MA
,
Kuziel
 
GA
, et al.  
C. difficile intoxicates neurons and pericytes to drive neurogenic inflammation
.
Nature.
 
2023
;
622
(
7983
):
611
-
618
. doi:10.1038/s41586-023-06607-2

91.

Yan
 
Y
,
Ramanan
 
D
,
Rozenberg
 
M
, et al.  
Interleukin-6 produced by enteric neurons regulates the number and phenotype of microbe-responsive regulatory T cells in the gut
.
Immunity.
 
2021
;
54
(
3
):
499
-
513.e5
. doi:10.1016/j.immuni.2021.02.002

92.

Schneider
 
KM
,
Blank
 
N
,
Alvarez
 
Y
, et al.  
The enteric nervous system relays psychological stress to intestinal inflammation
.
Cell.
 
2023
;
186
(
13
):
2823
-
2838.e20
. doi:10.1016/j.cell.2023.05.001

93.

Coffey
 
JC
,
O’Leary
 
DP.
 
The mesentery: structure, function, and role in disease
.
Lancet Gastroenterol Hepatol
.
2016
;
1
(
3
):
238
-
247
. doi:10.1016/S2468-1253(16)30026-7

94.

Desreumaux
 
P
,
Ernst
 
O
,
Geboes
 
K
, et al.  
Inflammatory alterations in mesenteric adipose tissue in Crohn’s disease
.
Gastroenterology.
 
1999
;
117
(
1
):
73
-
81
. doi:10.1016/s0016-5085(99)70552-4

95.

Schäffler
 
A
,
Herfarth
 
H.
 
Creeping fat in Crohn’s disease: travelling in a creeper lane of research
?
Gut.
 
2005
;
54
(
6
):
742
-
744
. doi:10.1136/gut.2004.061531

96.

Kredel
 
LI
,
Siegmund
 
B.
 
Adipose-tissue and intestinal inflammation - visceral obesity and creeping fat
.
Front Immunol.
 
2014
;
5
:
462
. doi:10.3389/fimmu.2014.00462

97.

Mao
 
R
,
Kurada
 
S
,
Gordon
 
IO
, et al.  
The mesenteric fat and intestinal muscle interface: creeping fat influencing stricture formation in Crohn’s disease
.
Inflamm Bowel Dis.
 
2019
;
25
(
3
):
421
-
426
. doi:10.1093/ibd/izy331

98.

Horowitz
 
A
,
Chanez-Paredes
 
SD
,
Haest
 
X
,
Turner
 
JR.
 
Paracellular permeability and tight junction regulation in gut health and disease
.
Nat Rev Gastroenterol Hepatol.
 
2023
;
20
(
7
):
417
-
432
. doi:10.1038/s41575-023-00766-3

99.

Danese
 
S
,
Solitano
 
V
,
Jairath
 
V
,
Peyrin-Biroulet
 
L.
 
The future of drug development for inflammatory bowel disease: the need to ACT (advanced combination treatment)
.
Gut.
 
2022
;
71
(
12
):
2380
-
2387
. doi:10.1136/gutjnl-2022-327025

100.

Hollander
 
D
,
Vadheim
 
CM
,
Brettholz
 
E
, et al.  
Increased intestinal permeability in patients with Crohn’s disease and their relatives. A possible etiologic factor
.
Ann Intern Med.
 
1986
;
105
(
6
):
883
-
885
. doi:10.7326/0003-4819-105-6-883

101.

Turpin
 
W
,
Lee
 
SH
,
Raygoza Garay
 
JA
, et al. ;
Crohn’s and Colitis Canada Genetic Environmental Microbial Project Research Consortium
.
Increased intestinal permeability is associated with later development of Crohn’s disease
.
Gastroenterology.
 
2020
;
159
(
6
):
2092
-
2100.e5
. doi:10.1053/j.gastro.2020.08.005

102.

Arrieta
 
MC
,
Madsen
 
K
,
Doyle
 
J
,
Meddings
 
J.
 
Reducing small intestinal permeability attenuates colitis in the IL10 gene-deficient mouse
.
Gut.
 
2009
;
58
(
1
):
41
-
48
. doi:10.1136/gut.2008.150888

103.

Su
 
L
,
Nalle
 
SC
,
Shen
 
L
, et al.  
TNFR2 activates MLCK-dependent tight junction dysregulation to cause apoptosis-mediated barrier loss and experimental colitis
.
Gastroenterology.
 
2013
;
145
(
2
):
407
-
415
. doi:10.1053/j.gastro.2013.04.011

104.

Graham
 
WV
,
He
 
W
,
Marchiando
 
AM
, et al.  
Intracellular MLCK1 diversion reverses barrier loss to restore mucosal homeostasis
.
Nat Med.
 
2019
;
25
(
4
):
690
-
700
. doi:10.1038/s41591-019-0393-7

105.

D’Incà
 
R
,
Di Leo
 
V
,
Corrao
 
G
, et al.  
Intestinal permeability test as a predictor of clinical course in Crohn’s disease
.
Am J Gastroenterol.
 
1999
;
94
(
10
):
2956
-
2960
. doi:10.1111/j.1572-0241.1999.01444.x

106.

Wyatt
 
J
,
Vogelsang
 
H
,
Hübl
 
W
,
Waldhöer
 
T
,
Lochs
 
H.
 
Intestinal permeability and the prediction of relapse in Crohn’s disease
.
Lancet.
 
1993
;
341
(
8858
):
1437
-
1439
. doi:10.1016/0140-6736(93)90882-h

107.

Arnott
 
ID
,
Kingstone
 
K
,
Ghosh
 
S.
 
Abnormal intestinal permeability predicts relapse in inactive Crohn disease
.
Scand J Gastroenterol.
 
2000
;
35
(
11
):
1163
-
1169
. doi:10.1080/003655200750056637

108.

Raju
 
P
,
Shashikanth
 
N
,
Tsai
 
PY
, et al.  
Inactivation of paracellular cation-selective claudin-2 channels attenuates immune-mediated experimental colitis in mice
.
J Clin Invest.
 
2020
;
130
(
10
):
5197
-
5208
. doi:10.1172/JCI138697

109.

Aherne
 
CM
,
Saeedi
 
B
,
Collins
 
CB
, et al.  
Epithelial-specific A2B adenosine receptor signaling protects the colonic epithelial barrier during acute colitis
.
Mucosal Immunol
.
2015
;
8
(
6
):
1324
-
1338
. doi:10.1038/mi.2015.22

110.

Suenaert
 
P
,
Bulteel
 
V
,
Lemmens
 
L
, et al.  
Anti-tumor necrosis factor treatment restores the gut barrier in Crohn’s disease
.
Am J Gastroenterol.
 
2002
;
97
(
8
):
2000
-
2004
. doi:10.1111/j.1572-0241.2002.05914.x

111.

Abraham
 
C
,
Abreu
 
MT
,
Turner
 
JR.
 
Pattern recognition receptor signaling and cytokine networks in microbial defenses and regulation of intestinal barriers: implications for Inflammatory Bowel Disease
.
Gastroenterology.
 
2022
;
162
(
6
):
1602
-
1616.e6
. doi:10.1053/j.gastro.2021.12.288

112.

Turner
 
JR.
 
Intestinal mucosal barrier function in health and disease
.
Nat Rev Immunol.
 
2009
;
9
(
11
):
799
-
809
. doi:10.1038/nri2653

113.

Shashikanth
 
N
,
Rizzo
 
HE
,
Pongkorpsakol
 
P
,
Heneghan
 
JF
,
Turner
 
JR.
 
Electrophysiologic analysis of tight junction size and charge selectivity
.
Curr Protoc
.
2021
;
1
(
6
):
e143
. doi:10.1002/cpz1.143

114.

Zuo
 
L
,
Kuo
 
WT
,
Cao
 
F
, et al.  
Tacrolimus-binding protein FKBP8 directs myosin light chain kinase-dependent barrier regulation and is a potential therapeutic target in Crohn’s disease
.
Gut.
 
2023
;
72
(
5
):
870
-
881
. doi:10.1136/gutjnl-2021-326534

115.

Villablanca
 
EJ
,
Selin
 
K
,
Hedin
 
CRH.
 
Mechanisms of mucosal healing: treating inflammatory bowel disease without immunosuppression
?
Nat Rev Gastroenterol Hepatol.
 
2022
;
19
(
8
):
493
-
507
. doi:10.1038/s41575-022-00604-y

116.

Rana
 
N
,
Privitera
 
G
,
Kondolf
 
HC
, et al.  
GSDMB is increased in IBD and regulates epithelial restitution/repair independent of pyroptosis
.
Cell.
 
2022
;
185
(
2
):
283
-
298.e17
. doi:10.1016/j.cell.2021.12.024

117.

Marchelletta
 
RR
,
Krishnan
 
M
,
Spalinger
 
MR
, et al.  
T cell protein tyrosine phosphatase protects intestinal barrier function by restricting epithelial tight junction remodeling
.
J Clin Invest.
 
2021
;
131
(
17
):
e138230
. doi:10.1172/JCI138230

118.

Sayoc-Becerra
 
A
,
Krishnan
 
M
,
Fan
 
S
, et al.  
The JAK-inhibitor tofacitinib rescues human intestinal epithelial cells and colonoids from cytokine-induced barrier dysfunction
.
Inflamm Bowel Dis.
 
2020
;
26
(
3
):
407
-
422
. doi:10.1093/ibd/izz266

119.

Eftychi
 
C
,
Schwarzer
 
R
,
Vlantis
 
K
, et al.  
Temporally distinct functions of the Cytokines IL-12 and IL-23 drive chronic colon inflammation in response to intestinal barrier impairment
.
Immunity.
 
2019
;
51
(
2
):
367
-
380.e4
. doi:10.1016/j.immuni.2019.06.008

120.

Pott
 
J
,
Kabat
 
AM
,
Maloy
 
KJ.
 
Intestinal epithelial cell autophagy is required to protect against TNF-Induced apoptosis during chronic colitis in mice
.
Cell Host Microbe.
 
2018
;
23
(
2
):
191
-
202.e4
. doi:10.1016/j.chom.2017.12.017

121.

Mohanan
 
V
,
Nakata
 
T
,
Desch
 
AN
, et al.  
C1orf106 is a colitis risk gene that regulates stability of epithelial adherens junctions
.
Science.
 
2018
;
359
(
6380
):
1161
-
1166
. doi:10.1126/science.aan0814

122.

Garcia-Carbonell
 
R
,
Wong
 
J
,
Kim
 
JY
, et al.  
Elevated A20 promotes TNF-induced and RIPK1-dependent intestinal epithelial cell death
.
Proc Natl Acad Sci U S A.
 
2018
;
115
(
39
):
E9192
-
E9200
. doi:10.1073/pnas.1810584115

123.

Castellanos
 
JG
,
Woo
 
V
,
Viladomiu
 
M
, et al.  
Microbiota-induced TNF-like ligand 1A drives group 3 innate lymphoid cell-mediated barrier protection and intestinal T cell activation during colitis
.
Immunity.
 
2018
;
49
(
6
):
1077
-
1089.e5
. doi:10.1016/j.immuni.2018.10.014

124.

Madara
 
JL
,
Pappenheimer
 
JR.
 
Structural basis for physiological regulation of paracellular pathways in intestinal epithelia
.
J Membr Biol.
 
1987
;
100
(
2
):
149
-
164
. doi:10.1007/BF02209147

125.

Turner
 
JR
,
Rill
 
BK
,
Carlson
 
SL
, et al.  
Physiological regulation of epithelial tight junctions is associated with myosin light-chain phosphorylation
.
Am J Physiol.
 
1997
;
273
(
4
):
CC1378
-
CC1385
. doi:10.1152/ajpcell.1997.273.4.c1378

126.

Clayburgh
 
DR
,
Barrett
 
TA
,
Tang
 
Y
, et al.  
Epithelial myosin light chain kinase-dependent barrier dysfunction mediates T cell activation-induced diarrhea in vivo
.
J Clin Invest.
 
2005
;
115
(
10
):
2702
-
2715
. doi:10.1172/JCI24970

127.

Voigt
 
RM
,
Forsyth
 
CB
,
Shaikh
 
M
, et al.  
Diurnal variations in intestinal barrier integrity and liver pathology in mice: implications for alcohol binge
.
Am J Physiol Gastrointest Liver Physiol.
 
2018
;
314
(
1
):
G131
-
G141
. doi:10.1152/ajpgi.00103.2017

128.

Kökten
 
T
,
Gibot
 
S
,
Lepage
 
P
, et al.  
TREM-1 inhibition restores impaired autophagy activity and reduces colitis in mice
.
J Crohns Colitis.
 
2018
;
12
(
2
):
230
-
244
. doi:10.1093/ecco-jcc/jjx129

129.

Grey
 
MJ
,
De Luca
 
H
,
Ward
 
DV
, et al.  
The epithelial-specific ER stress sensor ERN2/IRE1β enables host-microbiota crosstalk to affect colon goblet cell development
.
J Clin Invest.
 
2022
;
132
(
17
):
e153519
. doi:10.1172/JCI153519

130.

Kaser
 
A
,
Lee
 
AH
,
Franke
 
A
, et al.  
XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease
.
Cell.
 
2008
;
134
(
5
):
743
-
756
. doi:10.1016/j.cell.2008.07.021

131.

Taylor
 
SR
,
Ramsamooj
 
S
,
Liang
 
RJ
, et al.  
Dietary fructose improves intestinal cell survival and nutrient absorption
.
Nature.
 
2021
;
597
(
7875
):
263
-
267
. doi:10.1038/s41586-021-03827-2

132.

Peters
 
DE
,
Norris
 
LD
,
Tenora
 
L
, et al.  
A gut-restricted glutamate carboxypeptidase II inhibitor reduces monocytic inflammation and improves preclinical colitis
.
Sci Transl Med.
 
2023
;
15
(
708
):
eabn7491
. doi:10.1126/scitranslmed.abn7491

133.

Kaiko
 
GE
,
Chen
 
F
,
Lai
 
CW
, et al.  
PAI-1 augments mucosal damage in colitis
.
Sci Transl Med.
 
2019
;
11
(
482
):
eaat0852
. doi:10.1126/scitranslmed.aat0852

134.

Brazil
 
JC
,
Sumagin
 
R
,
Cummings
 
RD
,
Louis
 
NA
,
Parkos
 
CA.
 
Targeting of neutrophil lewis X blocks transepithelial migration and increases phagocytosis and degranulation
.
Am J Pathol.
 
2016
;
186
(
2
):
297
-
311
. doi:10.1016/j.ajpath.2015.10.015

135.

Cummins
 
EP
,
Seeballuck
 
F
,
Keely
 
SJ
, et al.  
The hydroxylase inhibitor dimethyloxalylglycine is protective in a murine model of colitis
.
Gastroenterology.
 
2008
;
134
(
1
):
156
-
165
. doi:10.1053/j.gastro.2007.10.012

136.

Xie
 
L
,
Fletcher
 
RB
,
Bhatia
 
D
, et al.  
Robust colonic epithelial regeneration and amelioration of colitis via FZD-Specific activation of Wnt signaling
.
Cell Mol Gastroenterol Hepatol
.
2022
;
14
(
2
):
435
-
464
. doi:10.1016/j.jcmgh.2022.05.003

137.

Lopetuso
 
LR
,
De Salvo
 
C
,
Pastorelli
 
L
, et al.  
IL-33 promotes recovery from acute colitis by inducing miR-320 to stimulate epithelial restitution and repair
.
Proc Natl Acad Sci U S A.
 
2018
;
115
(
40
):
E9362
-
E9370
. doi:10.1073/pnas.1803613115

138.

Schoepfer
 
AM
,
Safroneeva
 
E
,
Vavricka
 
SR
,
Peyrin-Biroulet
 
L
,
Mottet
 
C.
 
Treatment of fibrostenotic and fistulizing Crohn’s disease
.
Digestion.
 
2012
;
86
(
Suppl 1
):
23
-
27
. doi:10.1159/000341961

139.

Li
 
C
,
Kuemmerle
 
JF.
 
Mechanisms that mediate the development of fibrosis in patients with Crohn’s disease
.
Inflamm Bowel Dis.
 
2014
;
20
(
7
):
1250
-
1258
. doi:10.1097/MIB.0000000000000043

140.

Chen
 
W
,
Lu
 
C
,
Hirota
 
C
, et al.  
Smooth muscle hyperplasia/hypertrophy is the most prominent histological change in Crohn’s fibrostenosing bowel strictures: a semiquantitative analysis by using a novel histological grading scheme
.
J Crohns Colitis.
 
2017
;
11
(
1
):
92
-
104
. doi:10.1093/ecco-jcc/jjw126

141.

Otte
 
JM
,
Rosenberg
 
IM
,
Podolsky
 
DK.
 
Intestinal myofibroblasts in innate immune responses of the intestine
.
Gastroenterology.
 
2003
;
124
(
7
):
1866
-
1878
. doi:10.1016/s0016-5085(03)00403-7

142.

Weber
 
S
,
Sitte
 
S
,
Voegele
 
AL
, et al.  
NLRP3 inhibition leads to impaired mucosal fibroblast function in patients with inflammatory bowel diseases
.
J Crohns Colitis.
 
2023
;
18
(
3
):
446
-
461
. doi:10.1093/ecco-jcc/jjad164

143.

Lesage
 
S
,
Zouali
 
H
,
Cézard
 
JP
, et al. ;
EPWG-IBD Group
.
CARD15/NOD2 mutational analysis and genotype-phenotype correlation in 612 patients with inflammatory bowel disease
.
Am J Hum Genet.
 
2002
;
70
(
4
):
845
-
857
. doi:10.1086/339432

144.

Nayar
 
S
,
Morrison
 
JK
,
Giri
 
M
, et al.  
A myeloid-stromal niche and gp130 rescue in NOD2-driven Crohn’s disease
.
Nature.
 
2021
;
593
(
7858
):
275
-
281
. doi:10.1038/s41586-021-03484-5

145.

Bataille
 
F
,
Klebl
 
F
,
Rümmele
 
P
, et al.  
Morphological characterisation of Crohn’s disease fistulae
.
Gut.
 
2004
;
53
(
9
):
1314
-
1321
. doi:10.1136/gut.2003.038208

146.

Frei
 
SM
,
Pesch
 
T
,
Lang
 
S
, et al.  
A role for tumor necrosis factor and bacterial antigens in the pathogenesis of Crohn’s disease-associated fistulae
.
Inflamm Bowel Dis.
 
2013
;
19
(
13
):
2878
-
2887
. doi:10.1097/01.MIB.0000435760.82705.23

147.

Scharl
 
M
,
Weber
 
A
,
Fürst
 
A
, et al.  
Potential role for SNAIL family transcription factors in the etiology of Crohn’s disease-associated fistulae
.
Inflamm Bowel Dis.
 
2011
;
17
(
9
):
1907
-
1916
. doi:10.1002/ibd.21555

148.

Scharl
 
M
,
Frei
 
S
,
Pesch
 
T
, et al.  
Interleukin-13 and transforming growth factor β synergise in the pathogenesis of human intestinal fistulae
.
Gut.
 
2013
;
62
(
1
):
63
-
72
. doi:10.1136/gutjnl-2011-300498

149.

Ortiz-Masiá
 
D
,
Gisbert-Ferrándiz
 
L
,
Bauset
 
C
, et al.  
Succinate activates EMT in intestinal epithelial cells through SUCNR1: a novel protagonist in fistula development
.
Cells
.
2020
;
9
(
5
):
1104
. doi:10.3390/cells9051104

150.

Estrada
 
HQ
,
Patel
 
S
,
Rabizadeh
 
S
, et al.  
Development of a personalized intestinal fibrosis model using human intestinal organoids derived from induced pluripotent stem cells
.
Inflamm Bowel Dis.
 
2022
;
28
(
5
):
667
-
679
. doi:10.1093/ibd/izab292

151.

Rogler
 
G
,
Singh
 
A
,
Kavanaugh
 
A
,
Rubin
 
DT.
 
Extraintestinal manifestations of inflammatory bowel disease: current concepts, treatment, and implications for disease management
.
Gastroenterology.
 
2021
;
161
(
4
):
1118
-
1132
. doi:10.1053/j.gastro.2021.07.042

152.

Hedin
 
CRH
,
Vavricka
 
SR
,
Stagg
 
AJ
, et al.  
The pathogenesis of extraintestinal manifestations: implications for IBD research, diagnosis, and therapy
.
J Crohns Colitis.
 
2019
;
13
(
5
):
541
-
554
. doi:10.1093/ecco-jcc/jjy191

153.

Liao
 
L
,
Schneider
 
KM
,
Galvez
 
EJC
, et al.  
Intestinal dysbiosis augments liver disease progression via NLRP3 in a murine model of primary sclerosing cholangitis
.
Gut.
 
2019
;
68
(
8
):
1477
-
1492
. doi:10.1136/gutjnl-2018-316670

154.

Jatana
 
S
,
Ponti
 
AK
,
Johnson
 
EE
, et al.  
A novel murine model of pyoderma gangrenosum reveals that inflammatory skin-gut crosstalk is mediated by IL-1β-primed neutrophils
.
Front Immunol.
 
2023
;
14
:
1148893
. doi:10.3389/fimmu.2023.1148893

155.

Zhang
 
J
,
Lyu
 
Z
,
Li
 
B
, et al.  
P4HA2 induces hepatic ductular reaction and biliary fibrosis in chronic cholestatic liver diseases
.
Hepatology.
 
2023
;
78
(
1
):
10
-
25
. doi:10.1097/HEP.0000000000000317

156.

Breban
 
M
,
Glatigny
 
S
,
Cherqaoui
 
B
, et al.  
Lessons on SpA pathogenesis from animal models
.
Semin Immunopathol.
 
2021
;
43
(
2
):
207
-
219
. doi:10.1007/s00281-020-00832-x

157.

McHenry
 
S
,
Glover
 
M
,
Ahmed
 
A
, et al.  
NAFLD is associated with quiescent rather than active Crohn’s disease
.
Inflamm Bowel Dis.
 
2023
:
izad129
. doi:10.1093/ibd/izad129

158.

McHenry
 
S
,
Sharma
 
Y
,
Tirath
 
A
, et al.  
Crohn’s disease is associated with an increased prevalence of nonalcoholic fatty liver disease: a cross-sectional study using magnetic resonance proton density fat fraction mapping
.
Clin Gastroenterol Hepatol.
 
2019
;
17
(
13
):
2816
-
2818
. doi:10.1016/j.cgh.2019.02.045

159.

Raffals
 
LE
,
Saha
 
S
,
Bewtra
 
M
, et al.  
The development and initial findings of a study of a prospective adult research cohort with inflammatory bowel disease (SPARC IBD)
.
Inflamm Bowel Dis.
 
2022
;
28
(
2
):
192
-
199
. doi:10.1093/ibd/izab071

Author notes

Matthew A. Ciorba, Liza Konnikova and Wendy S. Garrett Contributed equally

Simon A. Hirota, Elena M. Lucchetta and Jerrold R. Turner Contributed equally

Matthew A. Ciorba and Wendy S. Garrett 2024 Preclinical Human IBD Mechanisms Work Group Co-Chairs

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/pages/standard-publication-reuse-rights)