Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Pancreatic Cancer (Jul 2025)
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • Sex Differences in Medicine (Sep 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
Top
  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal
  • Top
  • Abstract
  • Introduction
  • Role of the microbiome in IBD across contexts
  • Utilizing microbiome context to improve MBIs
  • Conclusions
  • Acknowledgments
  • Footnotes
  • References
  • Version history
  • Article usage
  • Citations to this article

Advertisement

Review Series Open Access | 10.1172/JCI193969

Emerging concepts and shifting paradigms for understanding the microbial basis of inflammatory bowel diseases

Megan S. Kennedy and Eugene B. Chang

Department of Medicine, University of Chicago, Chicago, Illinois, USA.

Address correspondence to: Eugene B. Chang, Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA. [email protected].

Find articles by Kennedy, M. in: PubMed | Google Scholar

Department of Medicine, University of Chicago, Chicago, Illinois, USA.

Address correspondence to: Eugene B. Chang, Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA. [email protected].

Find articles by Chang, E. in: PubMed | Google Scholar |

Published September 2, 2025 - More info

Published in Volume 135, Issue 17 on September 2, 2025
J Clin Invest. 2025;135(17):e193969. https://doi.org/10.1172/JCI193969.
© 2025 Kennedy et al. This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
Published September 2, 2025 - Version history
View PDF
Abstract

Inflammatory bowel diseases (IBDs) are complex immune disorders that arise at the intersection of genetic susceptibility, environmental exposures, and dysbiosis of the gut microbiota. Our understanding of the role of the microbiome in IBD has greatly expanded over the past few decades, although efforts to translate this knowledge into precision microbiome-based interventions for the prevention and management of disease have thus far met limited success. Here we survey and synthesize recent primary research in order to propose an updated conceptual framework for the role of the microbiome in IBD. We argue that accounting for gut microbiome context — elements such disease regionality, phase of disease, diet, medication use, and patient lifestyle — is essential for the development of a clear and mechanistic understanding of the microbiome’s contribution to pathogenesis or health. Armed with better mechanistic and contextual understanding, we will be better prepared to translate this knowledge into effective and precise strategies for microbiome restitution.

Introduction

Inflammatory bowel diseases (IBDs) encompass a range of often debilitating chronic gastrointestinal (GI) diseases that impose immense quality-of-life and health-care costs on individuals and society (1). It is increasingly appreciated that among other factors like genetic predisposition and environmental exposures, the gut microbiome plays an essential role in the pathophysiology of IBD. For instance, studies in germ-free mice have demonstrated that gut microbes are required for the development of colitis in several models of genetic susceptibility (2–4) and that transplant of an inflamed gut microbiome into an otherwise healthy gut can induce inflammation (5–7). An abundance of human studies have identified correlations between IBD status and various microbiome omics profiles (8, 9). However, we still lack effective microbiome-based interventions (MBIs) for prevention, management, and treatment of IBD, in spite of the microbiome’s clear contributory role in the etiopathogenesis of IBD.

In this Review, we draw on recent primary evidence to formulate an updated conceptual framework for the role of the microbiome in IBD. Although the gut virome and mycobiome, the respective viral and fungal counterparts to the bacterial microbiome, have also recently been highlighted as important contributors to the complex dynamics of the gut ecosystem (10, 11), here we focus on the more extensively characterized bacterial component. In particular, we argue that in order to effectively harness the bacterial microbiome for therapeutic gain, we must take into consideration the basic contextual drivers of gut microbial health and function — that is, the shifting conditions of regional gut ecosystems across distinct phases of disease, diets, microbial network structures, comorbid conditions, medications, and patient lifestyles (Figure 1).

Contextual drivers of the microbiome in IBD.Figure 1

Contextual drivers of the microbiome in IBD. The specific role played by the microbiome in IBD varies across a wide variety of contextual factors. Sleep and circadian rhythms impact the rhythmicity and functional output of the microbiota. Diet affects composition and function of the microbiota. Medications can affect microbiome composition, and the microbiota can reciprocally impact drug metabolism. Across stages of disease from diagnosis to relapse, microbiota composition varies in ways that may help to predict disease activity. Different IBD subtypes are associated with distinct microbiome signature. Distinct regions of the GI tract harbor distinct microbiota profiles both in health and in disease. Exercise and lifestyle impact the microbiota both directly and via effects on motility of the GI tract. Early-life exposures may alter tolerance to commensal microbes, and even the healthy microbiota varies over the human lifespan.

This Review is divided into two main sections: First, we discuss what is currently known about the role of the microbiome in IBD, and how accounting for various aspects of microbiome context can refine that understanding. Second, we discuss existing MBIs and how more deliberate control over microbiome context might improve their efficacy. With this approach, we acknowledge and embrace the complexity of the host-microbe ecosystem as it applies to this multifactorial chronic disease, mining that complexity for meaningful ways to stratify subjects and inform interventional strategies. In so doing, we hope to inspire more incisive research that will bring MBIs into an era of clinical utility.

Role of the microbiome in IBD across contexts

Current understanding of the microbiome in IBD. IBDs, comprising ulcerative colitis (UC) and Crohn’s disease (CD), are complex immune disorders that arise from genetic susceptibility; environmental exposures like diet, toxins, infectious stimuli, etc.; and an imbalance or disruption in the composition and function of the gut microbiome (dysbiosis) — each necessary, but not sufficient, to cause disease. For example, a patient whose immune system is genetically programmed to respond at a lower stimulus threshold might mount a proinflammatory response against a benign ingested or microbial antigen passing through the GI tract (12). In patients with CD, this hyperresponsiveness could be caused by mutations in genes like NOD2, which is involved in intracellular processing of bacterial components (13, 14), or ATG16L1 or IRGM, which are involved in autophagy and clearance of intracellular pathogens (15). In UC or CD, various mutations in IL-23 pathway genes, including IL23R, IL12B, STAT3, and JAK2, serve to induce a hyperactive Th17 cell response (16). The resultant inflammation may damage the epithelial barrier, allowing the commensal microbes to penetrate the gut mucosa and come into contact with deeper tissues of the GI tract that are laden with immune cells (17).

Importantly, the microbes that trigger the immune system need not be the initiating cause of inflammation, nor do they even need to be strictly pathogenic organisms (18). The once-predominant view that a single pathogenic microbe could be isolated as the causative agent of disease has been almost wholly unsupported in the IBD-microbiome literature (19). Study after study has failed to consistently identify a single causative agent in patients with active IBD, and even in the handful of studies that could link one organism to disease (20–22), these organisms more often take the form of opportunistic “pathobionts,” or otherwise commensal organisms that become pathogenic only under specific disease-promoting conditions (18, 23).

Rather, an extensive body of literature now shows that the gut microbiome of IBD patients often has few unique community members compared with that of healthy control individuals, but instead exhibits skewed proportions of the same “healthy” taxa, resulting in altered overall community composition and function (19, 24). Thus, microbiome “eubiosis” or “dysbiosis” can be better understood as emergent properties of the community as a whole, rather than the work of individual “good” or “bad” microbes.

How exactly a healthy microbiome transitions toward dysbiosis, and whether that is cause or consequence of inflammation, remains the subject of intense research and debate (19, 25). In all likelihood, the answer depends on context. Genetics, environment, and microbiome interact in a complex web of feedback responses to tip the balance toward health or inflammation. An otherwise benign antigen (26) or commensal microbe (27, 28) might trigger a genetically hyperresponsive immune system. Lifestyle factors like diet or cigarette smoking might shift the microbiome toward dysbiosis that predisposes even a stable immune system to inflammation (29, 30). The task at hand is to identify which elements of microbiome context best enable us to understand and manage disease or to maintain states of remission.

Disease subtype context: the wide range of IBDs. A diverse array of semi-overlapping disease presentations fall under the broad umbrella of IBD. Most commonly, these are subcategorized into UC or CD. The microbiome profile of patients with either subtype is altered compared with that of healthy control individuals, with IBD patients exhibiting reduced diversity; increased levels of Enterobacteriaceae; loss of putatively beneficial taxa like Ruminococcaceae and Lachnospiraceae; and an altered metagenome and metabolome with enrichment of sphingolipids and primary bile acids (8, 31–33). But depending on which GI tissues are involved, the microbiota of UC and CD are also distinguishable from each other (31, 34), to the extent that disease subtype can be accurately predicted from microbiome signature alone (35).

Even within these major branches of IBD, however, there is significant variability in disease presentation, prompting efforts to further subclassify and stratify patients. For CD, subclasses have been proposed along lines of disease behavior (perforating, stricturing, creeping fat, etc.) (36), age of onset (36, 37), disease localization (ileal, colonic, ileocolonic, perianal, perioral, etc.) (36, 38), immune cell profiles (26, 39), molecular markers (40), and genetic risk variants (41, 42). UC has similarly been subtyped by localization (proctitis, left-sided UC, pancolitis) (43, 44), disease severity (45), genetic factors (41), and immune profile (46–48). These factors could all be reasonably expected to modify the physical and immunological environment experienced by the gut microbiota. Indeed, the microbiota of healthy individuals is known to vary by GI region (49), age (50), genetics (51), and immune profile (52). Correspondingly, many subtyping schemes have been shown to correlate with distinct microbiota states, likely reflecting shared underlying pathophysiology that can help us to more effectively deduce mechanisms of disease.

An excellent example is CD with or without creeping fat. “Creeping fat” refers to hyperplastic mesenteric adipose tissue that wraps around inflamed intestinal lesions in CD, particularly in the antimesenteric region, worsening inflammation, inducing fibrosis, and promoting the development of strictures that predispose patients to bowel obstruction (53). Researchers determined that individuals with creeping fat exhibited a distinct microbiome localization signature — that is, in patients with creeping fat, members of the microbiome migrate via transmural lesions of the bowel wall into the surrounding fatty tissues (54). Once in the mesenteric adipose, these microbes interface with the immune system to induce inflammation, tissue remodeling, hyperplasia, and fibrosis (55). Thus, for this disease subtype, a precise mechanism was traced from aberrant microbiome localization to disease manifestation.

Other schemes of disease subclassification have similarly shown correlations with microbiome state. For example, Moustafa et al. identified a modest correlation between IBD genetic risk variants and microbiome composition (56), and Imhann et al. showed that even healthy individuals with IBD genetic risk factors have a microbiome distinct from those without genetic risk factors (57). Ghosh et al. identified distinct microbiome signatures for IBD among different age groups (58), Kedia et al. showed that the UC microbiome varies across categories of disease severity (59), and multiple groups have shown associations between CD flare localization and microbiota composition (38, 60). Additional experimental follow-up is needed to transition from correlative findings to mechanistic understanding of the microbiome’s role in these scenarios.

GI regional context: biogeography of the bowel. Regarding methods of subclassifying UC and CD, there is mounting evidence in support of stratifying patients according to which GI tissues are affected by inflammation, particularly for CD, which spans a greater diversity of tissues than UC (38, 61). It has been well established that the gut environment varies along the rostro-caudal axis anatomically and physiologically, with biochemical gradients in factors like pH (62), oxygen level (63), mucus thickness (64), and bile acid pool (65). The immune system is likewise distinct across regions of the GI tract that serve different physiological purposes (66). In healthy individuals, these factors all contribute to differences in microbial biomass, composition, and function across each region of the gut (49, 67).

It is hardly surprising, then, that disease location impacts microbiome signature and that patients with ileum-only versus colon-only CD exhibit distinct microbiome profiles (38, 60, 68). Whether cause or consequence of inflammation and immune activation, the pool of microbes available to participate in pathologic processes is distinct across GI regions, and therefore we can expect that GI region will be a pivotal factor in determining microbial involvement in disease. This premise is supported by recent work that combined host and microbiome multiomics data, and found that the multiomics profile of colon-only CD is more similar to that of UC than to ileal CD, with relative enrichment of neutrophil-related proteins and the strict anaerobe Bacteroides vulgatus in the colon (60). These similarities across colonic CD and UC could suggest a convergent microbial response to inflammatory conditions in the colon. More fundamentally, these data illustrate the importance of evaluating region-specific disease presentations separately.

An important corollary of the notion that GI regional context matters for understanding microbial drivers of disease is that study of fecal samples is not sufficient to gain insight into local disease processes in other parts of the GI tract. Microbial activity in the small bowel is simply not captured by fecal samples (69), and although the duodenum, terminal ileum, and colon can be reached and sampled via labor-intensive endoscopic approaches, the remaining approximately 20 feet of small bowel is largely inaccessible. Tools like CapScan or the SIMBA Capsule, ingestible pill-shaped sampling devices, are in development to enable relatively noninvasive luminal sampling throughout the upper and lower GI tract (70, 71). Several studies have shown that even within the lower GI tract, the fecal microbiome is distinct from the microbiome of mucosal scrapings or luminal contents of the distal colon (72, 73). Thus, researchers must carefully weigh the costs and benefits of mucosal versus fecal sampling when designing studies.

Temporal context: initial presentation, remission, and relapse. IBDs are fundamentally characterized as chronic, relapsing and remitting diseases. Much of the work evaluating the multifactorial causes of IBD has relied on cross-sectional studies comparing healthy control with affected individuals. However, this approach limits our ability to infer time-course dynamics, and inappropriate aggregation of data across phases of disease may obscure microbial signatures associated with the onset of inflammation.

Because we cannot yet predict when IBD will initially present, longitudinal cohort studies with true pre- and postdiagnosis samples are rare. These prospective studies typically enroll unaffected relatives of patients with IBD, given their high risk for eventually developing IBD (74, 75). In one such study with pre- and postdiagnosis samples from patients with UC, researchers found that although major reshaping of the microbiome happens after diagnosis, subclinical changes in gut microbiome composition, metagenome, and proteolytic activity do precede disease onset. Follow-up experiments in germ-free mice showed that the pre-UC microbiota, but not that of healthy control individuals, was sufficient to increase proteolytic activity and increase the abundance of polymorphonuclear immune cells. Together, these results suggest that while the microbiota responds robustly to inflammation, it may also play an early role in causing inflammation.

Another approach to resolving the temporal dynamics of the microbiome in IBD is to compare samples taken from the same patients during active disease and during remission. Several studies now suggest that the microbiome of patients with IBD generally fluctuates more over time than that of healthy control individuals (31, 76, 77), although there is no clear consensus regarding whether specific microbiome compositional shifts precede or correlate with states of disease flare or remission (78, 79). A notable shortcoming of these studies is the variable (and poorly reported) number of days between “remission” and “active disease” sampling. Among the studies reviewed here, the shortest reported interval between remission and flare samples was one month (31). Given that microbiome composition can shift on a timescale of days to weeks in response to host and environmental stimuli (80), sampling on a timescale of months may not adequately capture the dynamics that precede and contribute to disease, especially given the particularly dynamic nature of the IBD microbiome.

Patients who undergo bowel resection for refractory IBD provide unique insight into the dynamics of disease recurrence. After surgical removal of inflamed bowel, “clean slate” samples are taken from the remaining healthy tissues and are compared with inflamed tissues as disease recurs. For CD, the postoperative, healthy ileal microbiota of individuals whose disease eventually recurred is distinct from the microbiota of those who maintain remission (81–83). Several such studies independently found that, at the postoperative time point, those who went on to develop recurrence had relative increases in the abundance of facultative anaerobes like Fusobacterium and various Proteobacteria genera, with losses of butyrate-producing taxa from the Lachnospiraceae or Ruminococcaceae families (81–83).

For UC, postoperative recurrence has been studied with the pouchitis model: after total colectomy with ileal pouch anal anastomosis, nearly 50% of individuals will eventually experience UC-like inflammation in the previously unaffected ileal tissue of the J-pouch (84). In the healthy pouch, specific, distinct microbiome changes have been documented among those who go on to develop pouchitis compared with those who do not, including overall loss of diversity and changes in abundances of specific anaerobes (85–87).

Collectively, these time course studies indicate that although the microbiome certainly responds to inflammatory processes and abrupt changes in host physiology like those encountered after surgery, microbiome changes themselves may precede, perpetuate, or exacerbate inflammation in a feedback cycle with the host and immune system. That is, microbial dysbiosis can be both cause and consequence of inflammation.

Developmental context: consequences of early-life exposures. On a broader timescale, recent evidence suggests that IBD pathophysiology and corresponding microbiome changes may depend on developmental cues from early life. Multiple mouse studies show that after antibiotic treatment of pregnant dams, the dysbiotic microbiome inherited by offspring predisposes them to spontaneous colitis later in life (88, 89). Miyoshi et al. further demonstrated that early-life exposure to a diverse array of microbes promoted the development of immune cell subsets that imparted tolerance to those microbes upon reexposure, whereas mice that inherited a low-diversity, dysbiotic microbiota developed inflammation when first exposed to those otherwise benign commensal taxa at a later stage in life (88). Similar results were identified in humans by Gevers et al., who found that early-life exposure to antibiotics amplified CD-associated microbiome dysbiosis (90). Thus, for autoimmune pathologies like IBD, understanding not only the peri-disease microbiome but also the broader history of the microbiome may shed light on disease pathogenesis.

Lifestyle context: diet, medications, and sleep. Perhaps the most expansive and daunting aspect of microbiome context that is nevertheless essential to understanding the role of the microbiome in IBD is host lifestyle. The bacteria that colonize the human gut can adapt and evolve in response to new stimuli on an extraordinarily rapid timescale (91, 92). This means that behavioral changes in factors like diet (80, 92), medication use (93), sleep habits (94, 95), and exercise (96) can have a nearly immediate impact on microbiome community composition and function. When ignored, these factors will undoubtedly confound our search for microbial determinants of disease. However, when properly accounted for, they may even may even be exploited as some of the lowest-risk, least-invasive, and least-expensive approaches to modifying the course of disease via the microbiome.

Among the most salient lifestyle drivers of microbiome change in IBD is diet. The microbiome responds robustly to changes in diet, with more recent work identifying specific responses to dietary components like saturated versus unsaturated fats, preservatives, processed sugars, and specific fiber sources (or their absence) (97–99). For example, several murine studies have shown that a high-fat diet can induce proinflammatory dysbiosis (100, 101). The role of fiber and complex carbohydrates in protecting against inflammation is also increasingly appreciated: work from Desai et al. showed that in the absence of microbiota-accessible carbohydrates, microbes preferentially consume host-derived mucosal glycoproteins, invading and thinning the protective mucosal barrier in the colon and predisposing to inflammation (102).

On a molecular level, the byproducts of microbial metabolism, which depend profoundly on host diet, can also interface directly with the immune system to modulate inflammation. For instance, short-chain fatty acids (SCFAs) like acetate, propionate, and butyrate are produced by microbial fermentation of dietary fiber. These compounds, and especially butyrate, can directly act via G protein–coupled receptors GPCR41 and GPCR43 to promote the differentiation of antiinflammatory regulatory T cells (103). Low-fiber diets with correspondingly low levels of butyrate have been linked to the development of IBD (104). Similarly, diet impacts host production and circulation of primary bile acids, which serve to aid in the digestion and absorption of dietary fat. Secondary bile acids are produced exclusively by bacterial transformation of primary bile acids, and beyond their role in lipid absorption can act via the FXR and TGR5 receptors to modify intestinal permeability and barrier integrity (105) as well as to directly stimulate antigen-presenting cells (106). Imbalances in bile acid profile have been associated with the development of both UC and CD (8, 106, 107). Indole metabolites resulting from microbial transformation of dietary tryptophan, by contrast, are protective against inflammation (108).

Medication use is another lifestyle factor with profound influence on the microbiome and predisposition to IBD. Antibiotics, for example, have played a complex role in IBD management. On one hand, antibiotic treatment has been therapeutic for some cases of IBD and is first-line for pouchitis (109). On the other hand, antibiotic-induced dysbiosis is a risk factor for the development of IBD, particularly with antibiotics are used in multiple times early in life (110–112). Mainstays of IBD treatment like corticosteroids have also been shown to impact the colonic microenvironment and the gut microbiota in murine models (113). Other nonantibiotic medications including proton pump inhibitors and metformin have well-documented effects on microbiome composition and function (114–116). Thus, IBD patients whose symptom management or comorbidities require a broad and multimodal treatment regimen may ultimately be at greater risk of microbiome dysbiosis and disease recurrence as a consequence of those very interventions.

Other lifestyle factors like sleep (117) and circadian rhythms (94, 95) or exercise and general activity level (118) affect gut function and motility both directly and via the microbiome. There are nearly countless ways that host lifestyle choices could affect the microbiome. In addition to continuing dedicated exploration of the potentially causal effects of these factors on IBD pathogenesis via the microbiome, future work must at minimum collect appropriate metadata on lifestyle elements like diet, medication use, activity level, and sleep behaviors so that their impacts can be analytically disentangled and accounted for.

Utilizing microbiome context to improve MBIs

Quantifying success: from molecular change to clinical outcomes. First and foremost, a discussion of how to quantify successful use of MBIs in the treatment of IBD is warranted (Figure 2). The vast majority of clinical research employing MBIs for the treatment of IBD evaluates exclusively clinical outcomes: disease remission or symptom improvement (119). Of course, clinical outcomes are most important for patients, but empiric clinical measures alone leave little room to evaluate subclinical microbiome change resulting from an intervention. Many studies have at least perfunctorily examined microbiome composition via 16S sequencing. However, mechanistic work in preclinical models has increasingly shown that taxonomic data may have limited value in predicting the functionality of a microbial community, as there is extensive functional redundancy among taxonomically disparate microbes (120, 121).

Metrics to quantify the success of microbiome-based interventions.Figure 2

Metrics to quantify the success of microbiome-based interventions. Methods of quantifying the impact of MBIs span diverse metrics ranging from empiric clinical outcomes to quantification of the microbiome itself to the evaluation of the functional outputs of the microbiome. Clinical outcomes like the Mayo endoscopic score for UC or the CD activity index are used to gauge disease activity based on endoscopic findings and clinical symptomatology. Clinical biomarkers like fecal calprotectin and serum C-reactive protein provide less-invasive objective measures of inflammation severity. Fecal 16S rRNA sequencing provides a low-resolution readout of microbiome composition, while shotgun metagenomics evaluates the genomic content and functional capacity of a sample, and metatranscriptomics quantifies mRNA to quantify gene expression and evaluate functional activity of the microbiota. Metabolomics and metaproteomics evaluate the functional outputs of the gut microbiome (i.e., metabolic byproducts and proteins produced), which can often directly interact with the host. These strategies each address different elements of the IBD phenotype and are often most informative when evaluated together.

Many researchers have recently turned toward downstream functional metrics of microbiome community function like metabolomics or metaproteomics (122, 123). Products of microbial metabolism like SCFAs, conjugated bile acids, tryptophan metabolites, and various amino acids have been proposed as salient readouts of microbiome function (124–128). Thus, with the establishment of healthy reference ranges for these metabolites, they could provide clues into specific mechanistic shortcomings of the microbiome and, perhaps, how to fix them.

Importantly, even reliable, mechanistically informed metrics of microbiome functional change may not shift immediately or completely following intervention. Although the microbiome can respond very rapidly to intervention, pursuing the right trajectory of microbiome change may require successive or iterative cycles of intervention. Thus, to monitor the success of MBIs, we may require repeat, quantitative measurements over time, much in the way trends in standard blood chemistry tests like a complete blood count or basic metabolic panel are used in clinical practice (124). This can help us to identify scenarios in which subtle microbiome change may precede macro-scale clinical change.

With better metrics for quantifying the impact of MBIs across scales from molecular change to clinical response, we can more effectively connect the mechanistic dots between intervention and outcome.

Strategies for microbiome modulation: ablation, replacement, and support. Current strategies for microbiome manipulation can be broadly characterized as microbiome ablation, replacement, or support (Figure 3 and Table 1).

Strategies for MBIs.Figure 3

Strategies for MBIs. The “Untreated” panel schematically depicts a microbial community. “Ablation” refers to the eradication of the endogenous microbiome via methods like antibiotic treatment or via more targeted approaches like phage therapy. “Replacement” refers to strategies that introduce exogenous microbes into the gut, such as probiotics, synbiotics, FMT, or live biotherapeutics. “Support” represents strategies that involve changing the resource environment to support microbial growth, such as dietary interventions or prebiotics, which provide fermentable fiber sources that are selectively metabolized by gut microbes.

Table 1

Summary of microbiome-based intervention strategies

Antibiotics differentially ablate members of the native microbiome. Their impact can vary from person to person (129) based on factors like initial microbiome composition (130), treatment duration, and dosage (131) or confounding elements like host diet (132), and they provide no subsequent intervention to guide recovery. In the context of IBD, antibiotic use is limited primarily to the management of infectious complications (e.g., abscess, fistula, bowel perforation, or perioperative) (133–135) and pouchitis (109). Other promising ablative strategies under development include viral phage therapy, in which bacteriophages are engineered to target specific bacterial taxa and introduced into the gut for precision removal of undesired microbes (136, 137).

Among existing MBIs, microbiome replacement or transplant strategies have commanded the vast majority of clinical, academic, and commercial interest. Replacement approaches encompass probiotics, synbiotics, fecal microbiota transplant (FMT), and live biotherapeutics (Table 1) and generally aim to replace “disease-promoting” microbes with different, putatively “health-promoting” taxa. Probiotics are live bacteria most often derived from fermented food products. These bacteria are typically not well-adapted to survive in the human gut and have shown limited ability to engraft in their recipient community (138, 139) or shape a sustainable trajectory of microbiome change (140). “Synbiotics” denotes a pairing of probiotic and resource supplement to promote the growth of that probiotic (141). This premise is rational, and there is evidence supporting the idea that specifically tailored metabolic support can promote the growth and engraftment of exogenous microbes (142), but in practice, most synbiotics are composed of fermented food–derived probiotics with a carbohydrate resource that is not selectively metabolized by the probiotic over other members of the endogenous microbiota (141).

FMT and live biotherapeutics transition away from food-derived microbes and instead introduce taxa derived from a human gut. FMT refers to the bulk transplant of stool from a healthy human donor into an unhealthy individual. “Live biotherapeutic products” is a new term developed by the US Food and Drug Administration to categorize products containing live organisms like bacteria that have proven clinical efficacy in treating disease, and they are regulated as drugs (143). Although FMT is very effective for the treatment of refractory Clostridioides difficile infection (rCDI) (144), its success rate in IBD has been inconsistent, hovering near 40% under most protocols (145). Only two live biotherapeutic products are currently approved in the US, both for the treatment of rCDI (146, 147) and each derived from a human fecal community. Three new live biotherapeutic products are in phase II clinical trials for the treatment of CD and UC (148–150).

Finally, interventions to support microbial growth by changing the gut resource environment include prebiotics and dietary interventions. Prebiotics seek to guide microbiome change by providing resources selectively utilized by gut microbes in order to support their growth (151). Most prebiotics are made of compounds like inulin or fructooligosaccharides, which are carbohydrate compounds that can be fermented into SCFAs by a broad range of bacteria, but not the host (151, 152). While they may generally support the growth of commensal bacterial, there is little evidence to suggest that they promote compositional shifts or supplement the growth of specific, potentially beneficial taxa. Although dietary change can drastically affect the microbiota, it is usually relatively untargeted, and adherence can be difficult for patients.

“Postbiotics” refers to downstream products of microbial metabolism or components of microbes themselves, which are introduced into the gut without any live organisms. These are used not so much as a method of microbiome change as a means of sidestepping the microbiome altogether by introducing microbially derived molecules that can act directly on the host (153). Interestingly, early evidence from experiments using postbiotics have shown that these products can nevertheless act to shift microbiome composition and function (154, 155).

Each of these strategies derives from reasonable hypotheses about how to enact microbiome change. However, the specific details of their execution, especially with regard to microbiome context, must be examined to improve their efficacy.

Future directions: embracing context to improve MBIs. Fundamentally, most existing MBIs neglect microbiome context and have been consequently plagued by inconsistency (156, 157). Microbiome ablation strategies like antibiotics, for instance, rarely take into account host diet, which can fundamentally alter the trajectory of community re-assembly following a perturbation (132). Studies of prebiotics and synbiotics fail to determine whether resource supplements will selectively promote the growth of one or multiple endogenous microbes versus an introduced probiotic. FMTs are conducted without standardized administration protocols (158, 159), without consideration of where inflammation is most severe, which microbes can thrive in that region and whether they are present in the transplanted community, and without any real characterization of host and recipient communities (160).

These problems can be addressed through rationally designed intervention in which a specific goal is approached via a tailored strategy, taking into account contextual information as available. For instance, if we would like to address microbial dysbiosis in the small bowel, we should consider MBIs that promote small bowel microbiota (161). If we would like to promote the growth of specific microbial taxa, we should identify those taxa’s metabolic needs and target them specifically and selectively through prebiotics or dietary change (142, 162). If we would like a transplanted microbial community to serve a particular function, we should either formulate a synthetic community to achieve those functional goals or at least screen donor communities, bioinformatically or in vitro, for functional criteria. We should moreover carefully consider how that community might change under the dietary or inflammatory conditions of the recipient, and in competition with the recipient microbiome. This could guide decisions about recipient diet or supplementation, optimal timing of MBI relative to disease flare and immunomodulatory interventions, and whether antibiotic pretreatment of the recipient may be of benefit.

Given the temporal context of microbial community change and disease course, effective MBIs may need to span multiple rounds of targeted interventions. In macroecology, “secondary succession” refers to a process of gradual community change as the ecosystem undergoes iterative cycles of environmental change and community adaptation and response (163). For instance, dysbiotic conditions after a disturbance like a forest fire permit the growth of only tough, weedy grasses and pioneer species; but these species modify the soil in a way that allows the growth of bushes and shrubs, which change the environment to enable the growth of larger trees, in a process that progresses step by step until the climax community is achieved (164). Microbiome transplant after a disturbance like the severe inflammation of IBD may successfully introduce some pioneer taxa, but the gut environment has not yet changed enough to accommodate the growth of the climax community. Repeat transplant of whole communites or staged introduction of specific taxa or metabolites, chosen based on quantitative metrics of gut environmental recovery, facilitate a successional process of microbiome reconstitution.

Precision medicine strategies are emerging as a way to integrate individualized contextual data to generate personalized therapeutic interventions (165). For instance, one study developed a machine-learning tool that used individual data on blood markers, dietary habits, anthropometric measurements, physical activity, and gut microbiota to produce personalized dietary recommendations for better glycemic control (166, 167). In the context of IBD, such approaches could integrate information on genetic susceptibility, disease subtype and localization, medication use, prior antibiotic exposure, diet, and microbiome composition and function to predict features like chance of disease progression, drug response, or even optimal interventions for microbiome modulation. Even without personalizing treatments for each individual with IBD, applying this approach to a sizeable cohort could help to identify salient characteristics of patient subsets who respond similarly to certain interventions.

By embracing as much context as is readily available when designing interventions, MBIs will become more precise, consistent, and effective tools in the treatment of IBD.

Multimodal approaches to IBD management. IBD pathogenesis results from the dysregulation of many factors, including the microbiome, the spatial and structural physiology of the epithelium and mucosal barrier, environmental metabolites and oxygen levels, and the immune response. The current standard of care for IBD medical management relies on biologic therapies that target only the immune system, which integrates cues from all these elements (168). There has been minimal research on multimodal approaches that combine, for instance, microbiome interventions, dietary interventions, wound-healing and mucosal-protective strategies, and immunosuppressive therapies. By simultaneously targeting multiple aspects of IBD pathophysiology, we might more effectively be able to tip the scales in the direction of recovery.

More specifically, inflammation itself strongly affects microbiome composition and function (169). Attempting to modify the microbiome without addressing a florid inflammatory response is unlikely to bear fruit, as a transplanted microbiome may itself be changed by the environmental inflammation. Similarly, unaddressed metabolic derangements like bile acid dysregulation in CD involving the terminal ileum may hinder attempts to modify the microbiome. Bile acids are directly bactericidal to many taxa (170), and if imbalances in their abundance profile are not addressed, a microbial transplant will experience these bactericidal effects just as the native community did (171). Restoring a balanced bile acid profile by modifying dietary intake could be one way to lessen this impact, in conjunction with other interventions.

Wound healing remains an understudied component of IBD pathogenesis and therapy. If the heightened immune activity of IBD is thought to result in part from direct contact between submucosal immune effectors and microbes at wounded epithelial surfaces, then wound healing should be a critical element of any rational therapeutic approach (172). Strategies to promote wound healing, which to date include methods like mesenchymal stem cell transplant (173) or administration of intestinal growth factors like teduglutide (174), growth hormone (175), or epidermal growth factor (176), could change the spatial ecology of inflammation. Promising new research is exploring the use of synthetic hydrogels to restore the mucus layer and promote wound healing (177). Interestingly, recent data indicate that gut microbes and their mediators are needed for proper intestinal stem cell development, restoration of gut microenvironment, and revascularization of inflamed tissues, paving the way for healing (178–180).

By simultaneously addressing all of these contextual elements, their impacts and benefits might feed back into each other to deescalate the inflammatory cycle.

Prevention over treatment. Although we do not yet have strong tools to predict new onset of IBD, better monitoring of microbiome dysfunction, possibly via regular measurement of microbiome functional outputs like the metabolome, could provide a means of identifying early subclinical shifts that precede more overt inflammation. Addressing such concerns before the onset of inflammation may enable treatment and prevention by more cost-effective and less invasive means such as dietary intervention.

Conclusions

Research into the role of the microbiome in IBD has expanded tremendously over the past several decades, but translation of this body of work into clinically effective therapies has been slow, and at times frustrating. Rather than be discouraged by the fractal nuance of this complex, multi-scale ecosystem, we should be inspired by the opportunities that the microbiome and its drivers can provide as unique avenues of intervention. The details of microbiome context do matter to the trillions of microscopic eco-evolutionary agents inhabiting our guts. A contextualized, multi-pronged therapeutic strategy — grounded in ecological and mechanistic insight — may be essential to break the cycle of chronic inflammation in IBD.

Acknowledgments

Funding support was received from the NIDDK P30 Center for Interdisciplinary Study of Inflammatory Intestinal Disorders (NIDDK P30 DK042086) and T32DK007074 (MSK); NIGMS T32GM150375 (MSK); NIDDK awards R01DK113788, NIDDK R01DK138072, and R01DK143041; the Hong Kong Innovation and Technology (InnoHK) program; and the Gastrointestinal Research Foundation of Chicago. This work is the result of NIH funding, in whole or in part, and is subject to the NIH Public Access Policy. This work is the result of NIH funding, in whole or in part, and is subject to the NIH Public Access Policy. Through acceptance of this federal funding, the NIH has been given a right to make the work publicly available in PubMed Central.

Address correspondence to: Eugene B. Chang, Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA. [email protected].

Footnotes

Conflict of interest: EBC is a shareholder and board member for 32 Bioscience Inc.

Copyright: © 2025, Kennedy et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.

Reference information: J Clin Invest. 2025;135(17):e193969. https://doi.org/10.1172/JCI193969.

References
  1. Park KT, et al. The cost of inflammatory bowel disease: an initiative from the Crohn’s & Colitis Foundation. Inflamm Bowel Dis. 2020;26(1):1–10.
    View this article via: CrossRef PubMed Google Scholar
  2. Eun CS, et al. Induction of bacterial antigen-specific colitis by a simplified human microbiota consortium in gnotobiotic interleukin-10-/- mice. Infect Immun. 2014;82(6):2239–2246.
    View this article via: CrossRef PubMed Google Scholar
  3. Veltkamp C, et al. Continuous stimulation by normal luminal bacteria is essential for the development and perpetuation of colitis in Tg(epsilon26) mice. Gastroenterology. 2001;120(4):900–913.
    View this article via: CrossRef PubMed Google Scholar
  4. Sellon RK, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66(11):5224–5231.
    View this article via: CrossRef PubMed Google Scholar
  5. Schaubeck M, et al. Dysbiotic gut microbiota causes transmissible Crohn’s disease-like ileitis independent of failure in antimicrobial defence. Gut. 2016;65(2):225–237.
    View this article via: CrossRef PubMed Google Scholar
  6. Ohkusa T, et al. Induction of experimental ulcerative colitis by Fusobacterium varium isolated from colonic mucosa of patients with ulcerative colitis. Gut. 2003;52(1):79–83.
    View this article via: CrossRef PubMed Google Scholar
  7. Chen LA, et al. Unaffected relatives with high IBD polygenic risk scores from multiplex Crohn’s disease families carry microbiomes that protect IL-10 deficient mice against intestinal inflammation. Gastroenterology. 2024;166(3):S95–S96.
    View this article via: CrossRef Google Scholar
  8. Lloyd-Price J, et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569(7758):655–662.
    View this article via: CrossRef PubMed Google Scholar
  9. Ning L, et al. Microbiome and metabolome features in inflammatory bowel disease via multi-omics integration analyses across cohorts. Nat Commun. 2023;14(1):7135.
    View this article via: CrossRef PubMed Google Scholar
  10. Tun HM, et al. Gut virome in inflammatory bowel disease and beyond. Gut. 2024;73(2):350–360.
    View this article via: CrossRef PubMed Google Scholar
  11. Hsu C, et al. Mycobiome and inflammatory bowel disease: role in disease pathogenesis, current approaches and novel nutritional-based therapies. Inflamm Bowel Dis. 2023;29(3):470–479.
    View this article via: CrossRef PubMed Google Scholar
  12. Cho JH. The genetics and immunopathogenesis of inflammatory bowel disease. Nat Rev Immunol. 2008;8(6):458–466.
    View this article via: CrossRef PubMed Google Scholar
  13. Hugot J-P, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature. 2001;411(6837):599–603.
    View this article via: CrossRef PubMed Google Scholar
  14. Abraham C, Cho JH. Functional consequences of NOD2 (CARD15) mutations. Inflamm Bowel Dis. 2006;12(7):641–650.
    View this article via: CrossRef PubMed Google Scholar
  15. Burton PR, et al. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447(7145):661–678.
    View this article via: CrossRef PubMed Google Scholar
  16. Franke A, et al. Replication of signals from recent studies of Crohn’s disease identifies previously unknown disease loci for ulcerative colitis. Nat Genet. 2008;40(6):713–715.
    View this article via: CrossRef PubMed Google Scholar
  17. Odenwald MA, Turner JR. The intestinal epithelial barrier: a therapeutic target? Nat Rev Gastroenterol Hepatol. 2017;14(1):9–21.
    View this article via: CrossRef PubMed Google Scholar
  18. Caruso R, et al. Host-pathobiont interactions in Crohn’s disease. Nat Rev Gastroenterol Hepatol. 2024;22(6):395–414.
    View this article via: CrossRef PubMed Google Scholar
  19. Ni J, et al. Gut microbiota and IBD: causation or correlation? Nat Rev Gastroenterol Hepatol. 2017;14(10):573–584.
    View this article via: CrossRef PubMed Google Scholar
  20. Palmela C, et al. Adherent-invasive Escherichia coli in inflammatory bowel disease. Gut. 2018;67(3):574–587.
    View this article via: CrossRef PubMed Google Scholar
  21. Strauss J, et al. Invasive potential of gut mucosa-derived Fusobacterium nucleatum positively correlates with IBD status of the host. Inflamm Bowel Dis. 2011;17(9):1971–1978.
    View this article via: CrossRef PubMed Google Scholar
  22. Mills RH, et al. Multi-omics analyses of the ulcerative colitis gut microbiome link Bacteroides vulgatus proteases with disease severity. Nat Microbiol. 2022;7(2):262–276.
    View this article via: CrossRef PubMed Google Scholar
  23. Mirsepasi-Lauridsen HC, et al. Escherichia coli pathobionts associated with inflammatory bowel disease. Clin Microbiol Rev. 2019;32(2):e00060-18.
    View this article via: CrossRef PubMed Google Scholar
  24. Glassner KL, et al. The microbiome and inflammatory bowel disease. J Allergy Clin Immunol. 2020;145(1):16–27.
    View this article via: CrossRef PubMed Google Scholar
  25. Sartor RB, Wu GD. Roles for intestinal bacteria, viruses, and fungi in pathogenesis of inflammatory bowel diseases and therapeutic approaches. Gastroenterology. 2017;152(2):327–339.
    View this article via: CrossRef PubMed Google Scholar
  26. Prideaux L, et al. Serological antibodies in inflammatory bowel disease: a systematic review. Inflamm Bowel Dis. 2012;18(7):1340–1355.
    View this article via: CrossRef PubMed Google Scholar
  27. Miyoshi J, et al. Early-life microbial restitution reduces colitis risk promoted by antibiotic-induced gut dysbiosis in interleukin 10-/- mice. Gastroenterology. 2021;161(3):940–952.
    View this article via: CrossRef PubMed Google Scholar
  28. Rakoff-Nahoum S, et al. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118(2):229–241.
    View this article via: CrossRef PubMed Google Scholar
  29. Bolte LA, et al. Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut. 2021;70(7):1287–1298.
    View this article via: CrossRef PubMed Google Scholar
  30. Biedermann L, et al. Smoking cessation induces profound changes in the composition of the intestinal microbiota in humans. PLoS One. 2013;8(3):e59260.
    View this article via: CrossRef PubMed Google Scholar
  31. Halfvarson J, et al. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat Microbiol. 2017;2(5):17004.
    View this article via: CrossRef PubMed Google Scholar
  32. Duvallet C, et al. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat Commun. 2017;8(1):1784.
    View this article via: CrossRef PubMed Google Scholar
  33. Franzosa EA, et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat Microbiol. 2019;4(2):293–305.
    View this article via: CrossRef PubMed Google Scholar
  34. Sankarasubramanian J, et al. Gut microbiota and metabolic specificity in ulcerative colitis and Crohn’s disease. Front Med. 2020;7:606298.
    View this article via: CrossRef PubMed Google Scholar
  35. Zheng J, et al. Noninvasive, microbiome-based diagnosis of inflammatory bowel disease. Nat Med. 2024;30(12):3555–3567.
    View this article via: CrossRef PubMed Google Scholar
  36. Satsangi J, et al. The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut. 2006;55(6):749–753.
    View this article via: CrossRef PubMed Google Scholar
  37. Pigneur B, et al. Natural history of Crohn’s disease: comparison between childhood- and adult-onset disease. Inflamm Bowel Dis. 2010;16(6):953–961.
    View this article via: CrossRef PubMed Google Scholar
  38. Atreya R, Siegmund B. Location is important: differentiation between ileal and colonic Crohn’s disease. Nat Rev Gastroenterol Hepatol. 2021;18(8):544–558.
    View this article via: CrossRef PubMed Google Scholar
  39. VanDussen KL, et al. Genetic variants synthesize to produce paneth cell phenotypes that define subtypes of Crohn’s disease. Gastroenterology. 2014;146(1):200–209.
    View this article via: CrossRef PubMed Google Scholar
  40. Weiser M, et al. Molecular classification of Crohn’s disease reveals two clinically relevant subtypes. Gut. 2018;67(1):36–42.
    View this article via: CrossRef PubMed Google Scholar
  41. Cleynen I, et al. Inherited determinants of Crohn’s disease and ulcerative colitis phenotypes: a genetic association study. Lancet. 2016;387(10014):156–167.
    View this article via: CrossRef PubMed Google Scholar
  42. Jostins L, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119–124.
    View this article via: CrossRef PubMed Google Scholar
  43. Shin DS, et al. Extensive disease subtypes in adult patients with ulcerative colitis: non-pancolitis versus pancolitis. Dig Dis Sci. 2018;63(11):3097–3104.
    View this article via: CrossRef PubMed Google Scholar
  44. Albayrak NE, Polydorides AD. Characteristics and outcomes of left-sided ulcerative colitis with a cecal/periappendiceal patch of inflammation. Am J Surg Pathol. 2022;46(8):1116–1125.
    View this article via: CrossRef PubMed Google Scholar
  45. Feuerstein JD, et al. AGA clinical practice guidelines on the management of moderate to severe ulcerative colitis. Gastroenterology. 2020;158(5):1450–1461.
    View this article via: CrossRef PubMed Google Scholar
  46. Mo S, et al. A precise molecular subtyping of ulcerative colitis reveals the immune heterogeneity and predicts clinical drug responses. J Transl Med. 2023;21(1):466.
    View this article via: CrossRef PubMed Google Scholar
  47. Sun S, et al. Biological characteristics of molecular subtypes of ulcerative colitis characterized by ferroptosis and neutrophil infiltration. Sci Rep. 2024;14(1):9510.
    View this article via: CrossRef PubMed Google Scholar
  48. Wang J, et al. Crohn’s disease and ulcerative colitis share 2 molecular subtypes with different mechanisms and drug responses. J Crohns Colitis. 2024;19(3):jjae152.
    View this article via: CrossRef PubMed Google Scholar
  49. Kennedy MS, Chang EB. The microbiome: composition and locations. Prog MolBiol Transl Sci. 2020;176:1–42.
    View this article via: CrossRef PubMed Google Scholar
  50. Yatsunenko T, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227.
    View this article via: CrossRef PubMed Google Scholar
  51. Goodrich JK, et al. Human genetics shape the gut microbiome. Cell. 2014;159(4):789–799.
    View this article via: CrossRef PubMed Google Scholar
  52. Levy M, et al. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17(4):219–232.
    View this article via: CrossRef PubMed Google Scholar
  53. Mao R, 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.
    View this article via: CrossRef PubMed Google Scholar
  54. Batra A, et al. Mesenteric fat - control site for bacterial translocation in colitis? Mucosal Immunol. 2012;5(5):580–591.
    View this article via: CrossRef PubMed Google Scholar
  55. Ha CWY, et al. Translocation of viable gut microbiota to mesenteric adipose drives formation of creeping fat in humans. Cell. 2020;183(3):666–683.
    View this article via: CrossRef PubMed Google Scholar
  56. Moustafa A, et al. Genetic risk, dysbiosis, and treatment stratification using host genome and gut microbiome in inflammatory bowel disease. Clin Transl Gastroenterol. 2018;9(1):e132.
    View this article via: CrossRef PubMed Google Scholar
  57. Imhann F, et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut. 2018;67(1):108–119.
    View this article via: CrossRef PubMed Google Scholar
  58. Ghosh TS, et al. Adjusting for age improves identification of gut microbiome alterations in multiple diseases. Elife. 2020;9:e50240.
    View this article via: CrossRef PubMed Google Scholar
  59. Kedia S, et al. Gut microbiome diversity in acute severe colitis is distinct from mild to moderate ulcerative colitis. J Gastroenterol Hepatol. 2021;36(3):731–739.
    View this article via: CrossRef PubMed Google Scholar
  60. Gonzalez CG, et al. Location-specific signatures of Crohn’s disease at a multi-omics scale. Microbiome. 2022;10(1):133.
    View this article via: CrossRef PubMed Google Scholar
  61. Dulai PS, et al. Should we divide crohn’s disease into ileum-dominant and isolated colonic diseases? Clin Gastroenterol Hepatol. 2019;17(13):2634–2643.
    View this article via: CrossRef PubMed Google Scholar
  62. Evans DF, et al. Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut. 1988;29(8):1035–1041.
    View this article via: CrossRef PubMed Google Scholar
  63. Zheng L, et al. Physiologic hypoxia and oxygen homeostasis in the healthy intestine. A review in the theme: cellular responses to hypoxia. Am J Physiol Cell Physiol. 2015;309(6):C350–C360.
    View this article via: CrossRef PubMed Google Scholar
  64. Johansson MEV, et al. The gastrointestinal mucus system in health and disease. Nat Rev Gastroenterol Hepatol. 2013;10(6):352–361.
    View this article via: CrossRef PubMed Google Scholar
  65. Ridlon JM, et al. Bile acids and the gut microbiome. Curr Opin Gastroenterol. 2014;30(3):332–338.
    View this article via: CrossRef PubMed Google Scholar
  66. Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14(10):667–685.
    View this article via: CrossRef PubMed Google Scholar
  67. Martinez-Guryn K, et al. Regional diversity of the gastrointestinal microbiome. Cell Host Microbe. 2019;26(3):314–324.
    View this article via: CrossRef PubMed Google Scholar
  68. Tyler AD, et al. Microbiome heterogeneity characterizing intestinal tissue and inflammatory bowel disease phenotype. Inflamm Bowel Dis. 2016;22(4):807–816.
    View this article via: CrossRef PubMed Google Scholar
  69. Leite GGS, et al. Mapping the segmental microbiomes in the human small bowel in comparison with stool: a REIMAGINE study. Dig Dis Sci. 2020;65(9):2595–2604.
    View this article via: CrossRef PubMed Google Scholar
  70. Shalon D, et al. Profiling the human intestinal environment under physiological conditions. Nature. 2023;617(7961):581–591.
    View this article via: CrossRef PubMed Google Scholar
  71. Wang G, et al. Spatially and temporally precise microbiome profiling in the small intestine using the SIMBA capsule with X-ray tracking. Front Microbiomes. 2024;3:1321624.
    View this article via: CrossRef Google Scholar
  72. Ringel Y, et al. High throughput sequencing reveals distinct microbial populations within the mucosal and luminal niches in healthy individuals. Gut Microbes. 2015;6(3):173–181.
    View this article via: CrossRef PubMed Google Scholar
  73. Kim D, et al. Comparison of sampling methods in assessing the microbiome from patients with ulcerative colitis. BMC Gastroenterol. 2021;21(1):396.
    View this article via: CrossRef PubMed Google Scholar
  74. Galipeau HJ, et al. Novel fecal biomarkers that precede clinical diagnosis of ulcerative colitis. Gastroenterology. 2021;160(5):1532–1545.
    View this article via: CrossRef PubMed Google Scholar
  75. Rausch P, et al. First insights into microbial changes within an inflammatory bowel disease family cohort study [preprint]. https://doi.org/10.1101/2024.07.23.24310327 Posted on bioRxiv July 24, 2024.
  76. Clooney AG, et al. Ranking microbiome variance in inflammatory bowel disease: a large longitudinal intercontinental study. Gut. 2021;70(3):499–510.
    View this article via: CrossRef PubMed Google Scholar
  77. Martinez C, et al. Unstable composition of the fecal microbiota in ulcerative colitis during clinical remission. Am J Gastroenterol. 2008;103(3):643–648.
    View this article via: CrossRef PubMed Google Scholar
  78. Öhman L, et al. Fecal microbiota dynamics during disease activity and remission in newly diagnosed and established ulcerative colitis. Sci Rep. 2021;11(1):8641.
    View this article via: CrossRef PubMed Google Scholar
  79. Rajca S, et al. Alterations in the intestinal microbiome (dysbiosis) as a predictor of relapse after infliximab withdrawal in Crohn’s disease. Inflamm Bowel Dis. 2014;20(6):978–986.
    View this article via: PubMed CrossRef Google Scholar
  80. David LA, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–563.
    View this article via: CrossRef PubMed Google Scholar
  81. Sokol H, et al. Prominence of ileal mucosa-associated microbiota to predict postoperative endoscopic recurrence in Crohn’s disease. Gut. 2020;69(3):462–472.
    View this article via: CrossRef PubMed Google Scholar
  82. Hamilton AL, et al. Luminal microbiota related to Crohn’s disease recurrence after surgery. Gut Microbes. 2020;11(6):1713–1728.
    View this article via: CrossRef PubMed Google Scholar
  83. Mondot S, et al. Structural robustness of the gut mucosal microbiota is associated with Crohn’s disease remission after surgery. Gut. 2016;65(6):954–962.
    View this article via: CrossRef PubMed Google Scholar
  84. Barnes EL, et al. Incidence, risk factors, and outcomes of pouchitis and pouch-related complications in patients with ulcerative colitis. Clin Gastroenterol Hepatol. 2021;19(8):1583–1591.
    View this article via: CrossRef PubMed Google Scholar
  85. Maharshak N, et al. Alterations of enteric microbiota in patients with a normal ileal pouch are predictive of pouchitis. J Crohns Colitis. 2017;11(3):314–320.
    View this article via: CrossRef PubMed Google Scholar
  86. Reshef L, et al. Pouch inflammation is associated with a decrease in specific bacterial taxa. Gastroenterology. 2015;149(3):718–727.
    View this article via: CrossRef PubMed Google Scholar
  87. Young VB, et al. Multiphasic analysis of the temporal development of the distal gut microbiota in patients following ileal pouch anal anastomosis. Microbiome. 2013;1(1):9.
    View this article via: CrossRef PubMed Google Scholar
  88. Miyoshi J, et al. Peripartum antibiotics promote gut dysbiosis, loss of immune tolerance, and inflammatory bowel disease in genetically prone offspring. Cell Rep. 2017;20(2):491–504.
    View this article via: CrossRef PubMed Google Scholar
  89. Schulfer AF, et al. Intergenerational transfer of antibiotic-perturbed microbiota enhances colitis in susceptible mice. Nat Microbiol. 2018;3(2):234–242.
    View this article via: CrossRef PubMed Google Scholar
  90. Gevers D, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15(3):382–392.
    View this article via: CrossRef PubMed Google Scholar
  91. Kennedy MS, et al. Dynamic genetic adaptation of Bacteroides thetaiotaomicron during murine gut colonization. Cell Rep. 2023;42(8):113009.
    View this article via: CrossRef PubMed Google Scholar
  92. Carmody RN, et al. Diet dominates host genotype in shaping the murine gut microbiota. Cell Host Microbe. 2015;17(1):72–84.
    View this article via: CrossRef PubMed Google Scholar
  93. Nagata N, et al. Population-level metagenomics uncovers distinct effects of multiple medications on the human gut microbiome. Gastroenterology. 2022;163(4):1038–1052.
    View this article via: CrossRef PubMed Google Scholar
  94. Leone V, et al. Effects of diurnal variation of gut microbes and high-fat feeding on host circadian clock function and metabolism. Cell Host Microbe. 2015;17(5):681–689.
    View this article via: CrossRef PubMed Google Scholar
  95. Zarrinpar A, et al. Diet and feeding pattern affect the diurnal dynamics of the gut microbiome. Cell Metab. 2014;20(6):1006–1017.
    View this article via: CrossRef PubMed Google Scholar
  96. Lamoureux EV, et al. Moderate exercise has limited but distinguishable effects on the mouse microbiome. mSystems. 2017;2(4):e00006-17.
    View this article via: CrossRef PubMed Google Scholar
  97. Loayza JJJ, et al. Effect of food additives on key bacterial taxa and the mucosa-associated microbiota in Crohn’s disease. The ENIGMA study. Gut Microbes. 2023;15(1):2172670.
    View this article via: CrossRef PubMed Google Scholar
  98. Suez J, et al. Personalized microbiome-driven effects of non-nutritive sweeteners on human glucose tolerance. Cell. 2022;185(18):3307–3328.
    View this article via: CrossRef PubMed Google Scholar
  99. Tanes C, et al. Role of dietary fiber in the recovery of the human gut microbiome and its metabolome. Cell Host Microbe. 2021;29(3):394–407.
    View this article via: CrossRef PubMed Google Scholar
  100. Devkota S, et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice. Nature. 2012;487(7405):104–108.
    View this article via: CrossRef PubMed Google Scholar
  101. Qiao Y, et al. Alterations of the gut microbiota in high-fat diet mice is strongly linked to oxidative stress. Appl Microbiol Biotechnol. 2013;97(4):1689–1697.
    View this article via: CrossRef PubMed Google Scholar
  102. Desai MS, et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell. 2016;167(5):1339–1353.
    View this article via: CrossRef PubMed Google Scholar
  103. Smith PM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569–573.
    View this article via: CrossRef PubMed Google Scholar
  104. Parada Venegas D, et al. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10:277.
    View this article via: CrossRef PubMed Google Scholar
  105. Cipriani S, et al. The bile acid receptor GPBAR-1 (TGR5) modulates integrity of intestinal barrier and immune response to experimental colitis. PLoS One. 2011;6(10):e25637.
    View this article via: CrossRef PubMed Google Scholar
  106. Lee MH, et al. How bile acids and the microbiota interact to shape host immunity. Nat Rev Immunol. 2024;24(11):798–809.
    View this article via: CrossRef PubMed Google Scholar
  107. Gentry EC, et al. Reverse metabolomics for the discovery of chemical structures from humans. Nature. 2024;626(7998):419–426.
    View this article via: CrossRef PubMed Google Scholar
  108. Dodd D, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551(7682):648–652.
    View this article via: CrossRef PubMed Google Scholar
  109. Bar N, et al. Long-term antibiotic treatment in pouchitis-patterns of use and safety. Inflamm Bowel Dis. 2022;28(7):1027–1033.
    View this article via: CrossRef PubMed Google Scholar
  110. Nguyen LH, et al. Antibiotic use and the development of inflammatory bowel disease: a national case-control study in Sweden. Lancet Gastroenterol Hepatol. 2020;5(11):986–995.
    View this article via: CrossRef PubMed Google Scholar
  111. Mark-Christensen A, et al. Early-life exposure to antibiotics and risk for Crohn’s disease: a nationwide Danish birth cohort study. Inflamm Bowel Dis. 2022;28(3):415–422.
    View this article via: CrossRef PubMed Google Scholar
  112. Faye AS, et al. Antibiotic use as a risk factor for inflammatory bowel disease across the ages: a population-based cohort study. Gut. 2023;72(4):663–670.
    View this article via: CrossRef PubMed Google Scholar
  113. Huang EY, et al. Using corticosteroids to reshape the gut microbiome: implications for inflammatory bowel diseases. Inflamm Bowel Dis. 2015;21(5):963–972.
    View this article via: CrossRef PubMed Google Scholar
  114. Imhann F, et al. Proton pump inhibitors affect the gut microbiome. Gut. 2016;65(5):740–748.
    View this article via: CrossRef PubMed Google Scholar
  115. Forslund K, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015;528(7581):262–266.
    View this article via: CrossRef PubMed Google Scholar
  116. Maier L, et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555(7698):623–628.
    View this article via: CrossRef PubMed Google Scholar
  117. Smith RP, et al. Gut microbiome diversity is associated with sleep physiology in humans. PLoS One. 2019;14(10):e0222394.
    View this article via: CrossRef PubMed Google Scholar
  118. Dalton A, et al. Exercise influence on the microbiome-gut-brain axis. Gut Microbes. 2019;10(5):555–568.
    View this article via: CrossRef PubMed Google Scholar
  119. Knox NC, et al. The gut microbiome as a target for IBD treatment: are we there yet? Curr Treat Options Gastroenterol. 2019;17(1):115–126.
    View this article via: CrossRef PubMed Google Scholar
  120. Louca S, et al. Function and functional redundancy in microbial systems. Nat Ecol Evol. 2018;2(6):936–943.
    View this article via: CrossRef PubMed Google Scholar
  121. Wilmanski T, et al. From taxonomy to metabolic output: what factors define gut microbiome health? Gut Microbes. 2021;13(1):1–20.
    View this article via: CrossRef PubMed Google Scholar
  122. Zierer J, et al. The fecal metabolome as a functional readout of the gut microbiome. Nat Genet. 2018;50(6):790–795.
    View this article via: CrossRef PubMed Google Scholar
  123. Wang Y, et al. Metaproteomics: a strategy to study the taxonomy and functionality of the gut microbiota. J Proteomics. 2020;219:103737.
    View this article via: CrossRef PubMed Google Scholar
  124. DeLeon O, et al. State of health of the gut microbiome assessed through a quantitative metabolomic-based marker panel [preprint]. https://doi.org/10.1101/2025.02.14.24319815 Posted on bioRxiv February 18, 2025.
  125. Mann ER, et al. Short-chain fatty acids: linking diet, the microbiome and immunity. Nat Rev Immunol. 2024;24(8):577–595.
    View this article via: CrossRef PubMed Google Scholar
  126. Ogilvie LA, Jones BV. Dysbiosis modulates capacity for bile acid modification in the gut microbiomes of patients with inflammatory bowel disease: a mechanism and marker of disease? Gut. 2012;61(11):1642–1643.
    View this article via: CrossRef PubMed Google Scholar
  127. Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun. 2018;9(1):3294.
    View this article via: CrossRef PubMed Google Scholar
  128. Wu J, et al. Changes in amino acid concentrations and the gut microbiota composition are implicated in the mucosal healing of ulcerative colitis and can be used as noninvasive diagnostic biomarkers. Clin Proteomics. 2024;21(1):62.
    View this article via: CrossRef PubMed Google Scholar
  129. Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A. 2011;108(suppl 1):4554–4561.
    View this article via: CrossRef PubMed Google Scholar
  130. Raymond F, et al. The initial state of the human gut microbiome determines its reshaping by antibiotics. ISME J. 2016;10(3):707–720.
    View this article via: CrossRef PubMed Google Scholar
  131. Lekang K, et al. Effects of different amoxicillin treatment durations on microbiome diversity and composition in the gut. PLoS One. 2022;17(10):e0275737.
    View this article via: CrossRef PubMed Google Scholar
  132. Kennedy MS, et al. Diet outperforms microbial transplant to drive microbiome recovery in mice. Nature. 2025;642(8068):747–755.
    View this article via: CrossRef PubMed Google Scholar
  133. Fansiwala K, et al. Oral vs intravenous discharge antibiotic regimens in the management of intra-abdominal abscesses in penetrating Crohn’s disease. Inflamm Bowel Dis. 2024;30(12):2280–2288.
    View this article via: CrossRef PubMed Google Scholar
  134. Singh A, et al. Management of perianal fistulizing Crohn’s disease. Inflamm Bowel Dis. 2024;30(9):1579–1603.
    View this article via: CrossRef PubMed Google Scholar
  135. Olson S, et al. Perioperative considerations for the surgical treatment of Crohn’s disease with discussion on surgical antibiotics practices and impact on the gut microbiome. Antibiotics (Basel). 2024;13(4):317.
    View this article via: CrossRef PubMed Google Scholar
  136. Federici S, 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.
    View this article via: CrossRef PubMed Google Scholar
  137. Titécat M, et al. Safety and efficacy of an AIEC-targeted bacteriophage cocktail in a mice colitis model. J Crohns Colitis. 2022;16(10):1617–1627.
    View this article via: CrossRef PubMed Google Scholar
  138. Grimm V, et al. Colonization of C57BL/6 mice by a potential probiotic Bifidobacterium bifidum strain under germ-free and specific pathogen-free conditions and during experimental colitis. PLoS One. 2015;10(10):e0139935.
    View this article via: CrossRef PubMed Google Scholar
  139. Maldonado-Gómez MX, et al. Stable engraftment of Bifidobacterium longum AH1206 in the human gut depends on individualized features of the resident microbiome. Cell Host Microbe. 2016;20(4):515–526.
    View this article via: CrossRef PubMed Google Scholar
  140. Limketkai BN, et al. Probiotics for induction of remission in Crohn’s disease. Cochrane Database Syst Rev. 2020;7(7):CD006634.
    View this article via: PubMed CrossRef Google Scholar
  141. Kolida S, Gibson GR. Synbiotics in health and disease. Annu Rev Food Sci Technol. 2011;2(1):373–393.
    View this article via: CrossRef PubMed Google Scholar
  142. Shepherd ES, et al. An exclusive metabolic niche enables strain engraftment in the gut microbiota. Nature. 2018;557(7705):434–438.
    View this article via: CrossRef PubMed Google Scholar
  143. Cordaillat-Simmons M, et al. Live biotherapeutic products: the importance of a defined regulatory framework. Exp Mol Med. 2020;52(9):1397–1406.
    View this article via: CrossRef PubMed Google Scholar
  144. Nood E van, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013;368(5):407–415.
    View this article via: CrossRef PubMed Google Scholar
  145. Colman RJ, Rubin DT. Fecal microbiota transplantation as therapy for inflammatory bowel disease: a systematic review and meta-analysis. J Crohns Colitis. 2014;8(12):1569–1581.
    View this article via: CrossRef PubMed Google Scholar
  146. Khanna S, et al. Efficacy and safety of RBX2660 in PUNCH CD3, a phase III, randomized, double-blind, placebo-controlled trial with a bayesian primary analysis for the prevention of recurrent clostridioides difficile infection. Drugs. 2022;82(15):1527–1538.
    View this article via: CrossRef PubMed Google Scholar
  147. Feuerstadt P, et al. SER-109, an oral microbiome therapy for recurrent Clostridioides difficile infection. N Engl J Med. 2022;386(3):220–229.
    View this article via: CrossRef PubMed Google Scholar
  148. Vedanta Biosciences Inc. Randomized, Double-Blind, Placebo-Controlled, Phase 2 Study of VE202 in Patients With Mild-to-Moderate Ulcerative Colitis. https://clinicaltrials.gov/study/NCT05370885 Accessed July 15, 2025.
  149. Guangzhou Zhiyi Biotechnology Co Ltd. A Phase 2, Multi-center, Randomized, Double-blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety of Live SK08 Powder (Bacteroides Fragilis) in Adult Patients with Active Mild to Moderate Ulcerative Colitis. https://clinicaltrials.gov/study/NCT06725017?cond=bacteroides&viewType=Table&checkSpell=&rank=3 Accessed July 15, 2025.
  150. Bezault M. A Phase 2 Multicentre, Randomised, Parallel-arm, Placebo-controlled, Double-blind Study to Evaluate the Safety and Target Engagement of EXL01 in the Prevention of Post-operative Recurrence of Crohn’s Disease. https://clinicaltrials.gov/study/NCT06925061?term=EXL01&rank=5 Accessed July 15, 2025.
  151. Fernández J, et al. Colon microbiota fermentation of dietary prebiotics towards short-chain fatty acids and their roles as anti-inflammatory and antitumour agents: a review. J Funct Foods. 2016;25:511–522.
    View this article via: CrossRef Google Scholar
  152. Davani-Davari D, et al. Prebiotics: definition, types, sources, mechanisms, and clinical applications. Foods. 2019;8(3):92.
    View this article via: CrossRef PubMed Google Scholar
  153. Salminen S, et al. The international scientific association of probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat Rev Gastroenterol Hepatol. 2021;18(9):649–667.
    View this article via: CrossRef PubMed Google Scholar
  154. Levy M, et al. Microbiota-modulated metabolites shape the intestinal microenvironment by regulating NLRP6 inflammasome signaling. Cell. 2015;163(6):1428–1443.
    View this article via: CrossRef PubMed Google Scholar
  155. Zhang T, et al. Stronger gut microbiome modulatory effects by postbiotics than probiotics in a mouse colitis model. NPJ Sci Food. 2022;6(1):53.
    View this article via: CrossRef PubMed Google Scholar
  156. Ianiro G, et al. Variability of strain engraftment and predictability of microbiome composition after fecal microbiota transplantation across different diseases. Nat Med. 2022;28(9):1913–1923.
    View this article via: CrossRef PubMed Google Scholar
  157. Danne C, et al. Recipient factors in faecal microbiota transplantation: one stool does not fit all. Nat Rev Gastroenterol Hepatol. 2021;18(7):503–513.
    View this article via: CrossRef PubMed Google Scholar
  158. Keshteli AH, et al. Pretreatment with antibiotics may enhance the efficacy of fecal microbiota transplantation in ulcerative colitis: a meta-analysis. Mucosal Immunol. 2017;10(2):565–566.
    View this article via: CrossRef PubMed Google Scholar
  159. Borody TJ, et al. Fecal microbiota transplantation: indications, methods, evidence, and future directions. Curr Gastroenterol Rep. 2013;15(8):337.
    View this article via: CrossRef PubMed Google Scholar
  160. Woodworth MH, et al. Challenges in fecal donor selection and screening for fecal microbiota transplantation: a review. Gut Microbes. 2017;8(3):225–237.
    View this article via: CrossRef PubMed Google Scholar
  161. DeLeon O, et al. Microbiome mismatches from microbiota transplants lead to persistent off-target metabolic and immunomodulatory effects. [published online May 30, 2025]. Cell. https://doi.org/10.1016/j.cell.2025.05.014.
    View this article via: PubMed Google Scholar
  162. Kearney SM, et al. Orthogonal dietary niche enables reversible engraftment of a gut bacterial commensal. Cell Rep. 2018;24(7):1842–1851.
    View this article via: CrossRef PubMed Google Scholar
  163. Prach K, Walker LR. Four opportunities for studies of ecological succession. Trends Ecol Evol. 2011;26(3):119–123.
    View this article via: CrossRef PubMed Google Scholar
  164. Hanes TL. Succession after fire in the chaparral of Southern California. Ecol Monogr. 1971;41(1):27–52.
    View this article via: CrossRef Google Scholar
  165. Ratiner K, et al. Utilization of the microbiome in personalized medicine. Nat Rev Microbiol. 2024;22(5):291–308.
    View this article via: CrossRef PubMed Google Scholar
  166. Zeevi D, et al. Personalized nutrition by prediction of glycemic responses. Cell. 2015;163(5):1079–1094.
    View this article via: CrossRef PubMed Google Scholar
  167. Rein M, et al. Effects of personalized diets by prediction of glycemic responses on glycemic control and metabolic health in newly diagnosed T2DM: a randomized dietary intervention pilot trial. BMC Med. 2022;20(1):56.
    View this article via: CrossRef PubMed Google Scholar
  168. Kapizioni C, et al. Biologic therapy for inflammatory bowel disease: real-world comparative effectiveness and impact of drug sequencing in 13 222 patients within the UK IBD BioResource. J Crohns Colitis. 2024;18(6):790–800.
    View this article via: CrossRef PubMed Google Scholar
  169. Borton MA, et al. Chemical and pathogen-induced inflammation disrupt the murine intestinal microbiome. Microbiome. 2017;5(1):47.
    View this article via: CrossRef PubMed Google Scholar
  170. Watanabe M, et al. Comprehensive evaluation of the bactericidal activities of free bile acids in the large intestine of humans and rodents. J Lipid Res. 2017;58(6):1143–1152.
    View this article via: CrossRef PubMed Google Scholar
  171. Tian Y, et al. The microbiome modulating activity of bile acids. Gut Microbes. 2020;11(4):979–996.
    View this article via: CrossRef PubMed Google Scholar
  172. Villablanca EJ, et al. Mechanisms of mucosal healing: treating inflammatory bowel disease without immunosuppression? Nat Rev Gastroenterol Hepatol. 2022;19(8):493–507.
    View this article via: CrossRef PubMed Google Scholar
  173. Ouboter L, et al. DOP64 Endoscopically injected allogeneic mesenchymal stromal cells alter the mucosal immune cell compartment in patients with ulcerative proctitis. J Crohns Colitis. 2021;15(suppl_1):S098–S099.
    View this article via: CrossRef Google Scholar
  174. Al Draiweesh S, et al. Teduglutide in patients with active Crohn’s disease and short bowel syndrome. Inflamm Bowel Dis. 2019;25(9):e109.
    View this article via: CrossRef PubMed Google Scholar
  175. Slonim AE, et al. A preliminary study of growth hormone therapy for Crohn’s disease. N Engl J Med. 2000;342(22):1633–1637.
    View this article via: CrossRef PubMed Google Scholar
  176. Sinha A, et al. Epidermal growth factor enemas with oral mesalamine for mild-to-moderate left-sided ulcerative colitis or proctitis. N Engl J Med. 2003;349(4):350–357.
    View this article via: CrossRef PubMed Google Scholar
  177. Sun C, et al. Sustainable conversion of husk into viscoelastic hydrogels for value-added biomedical applications. Matter. 2025;8(3):102002.
    View this article via: CrossRef PubMed Google Scholar
  178. Cham CM, et al. Intestinal epithelial heat shock protein 25/27 integrates host and microbial drivers of mucosal restitution following inflammatory injury [preprint]. https://doi.org/10.1101/2022.06.30.498349 Posted on bioRxiv July 2, 2022.
  179. Stappenbeck TS, et al. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc Natl Acad Sci U S A. 2002;99(24):15451–15455.
    View this article via: CrossRef PubMed Google Scholar
  180. Liu T-C, et al. Western diet induces Paneth cell defects through microbiome alterations and farnesoid X receptor and type I interferon activation. Cell Host Microbe. 2021;29(6):988–1001.
    View this article via: CrossRef PubMed Google Scholar
  181. Rahimi R, et al. A meta-analysis of antibiotic therapy for active ulcerative colitis. Dig Dis Sci. 2007;52(11):2920–2925.
    View this article via: CrossRef PubMed Google Scholar
  182. Ledder O, Turner D. Antibiotics in IBD: still a role in the biological era? Inflamm Bowel Dis. 2018;24(8):1676–1688.
    View this article via: CrossRef PubMed Google Scholar
  183. Yassine F, et al. The role of probiotics, prebiotics, and synbiotics in the treatment of inflammatory bowel diseases: an overview of recent clinical trials. Front Syst Biol. 2025;5:1561047.
    View this article via: CrossRef Google Scholar
  184. Saez-Lara MJ, et al. The role of probiotic lactic acid bacteria and bifidobacteria in the prevention and treatment of inflammatory bowel disease and other related diseases: a systematic review of randomized human clinical trials. Biomed Res Int. 2015;2015:505878.
    View this article via: CrossRef PubMed Google Scholar
  185. Hedin C, et al. Evidence for the use of probiotics and prebiotics in inflammatory bowel disease: a review of clinical trials. Proc Nutr Soc. 2007;66(3):307–315.
    View this article via: CrossRef PubMed Google Scholar
  186. Costello SP, et al. Effect of fecal microbiota transplantation on 8-week remission in patients with ulcerative colitis: a randomized clinical trial. JAMA. 2019;321(2):156–164.
    View this article via: CrossRef PubMed Google Scholar
  187. Rossen NG, et al. Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis. Gastroenterology. 2015;149(1):110–118.
    View this article via: CrossRef PubMed Google Scholar
  188. El Hage Chehade N, et al. Efficacy of fecal microbiota transplantation in the treatment of active ulcerative colitis: a systematic review and meta-analysis of double-blind randomized controlled trials. Inflamm Bowel Dis. 2023;29(5):808–817.
    View this article via: CrossRef PubMed Google Scholar
  189. Fehily SR, et al. Fecal microbiota transplantation therapy in Crohn’s disease: systematic review. J Gastroenterol Hepatol. 2021;36(10):2672–2686.
    View this article via: CrossRef PubMed Google Scholar
  190. Zhang X-F, et al. Clinical effects and gut microbiota changes of using probiotics, prebiotics or synbiotics in inflammatory bowel disease: a systematic review and meta-analysis. Eur J Nutr. 2021;60(5):2855–2875.
    View this article via: CrossRef PubMed Google Scholar
  191. Cohen SA, et al. Clinical and mucosal improvement with specific carbohydrate diet in pediatric Crohn disease. J Pediatr Gastroenterol Nutr. 2014;59(4):516–521.
    View this article via: CrossRef PubMed Google Scholar
  192. Gibson PR. Use of the low-FODMAP diet in inflammatory bowel disease. J Gastroenterol Hepatol. 2017;32(Suppl 1):40–42.
    View this article via: CrossRef PubMed Google Scholar
  193. De Filippis F, et al. Dietary interventions to modulate the gut microbiome-how far away are we from precision medicine. Inflamm Bowel Dis. 2018;24(10):2142–2154.
    View this article via: CrossRef PubMed Google Scholar
  194. Firoozi D, et al. Effects of short chain fatty acid-butyrate supplementation on the disease severity, inflammation, and psychological factors in patients with active ulcerative colitis: a double-blind randomized controlled trial. J Nutr Metab. 2025;2025(1):3165876.
    View this article via: CrossRef PubMed Google Scholar
  195. Hamer HM, et al. Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther. 2007;27(2):104–119.
    View this article via: CrossRef PubMed Google Scholar
  196. Truyens M, et al. Effect of 5-hydroxytryptophan on fatigue in quiescent inflammatory bowel disease: a randomized controlled trial. Gastroenterology. 2022;163(5):1294–1305.
    View this article via: CrossRef PubMed Google Scholar
  197. Guo S, et al. Effects of postbiotics on chronic diarrhea in young adults: a randomized, double-blind, placebo-controlled crossover trial assessing clinical symptoms, gut microbiota, and metabolite profiles. Gut Microbes. 2024;16(1):2395092.
    View this article via: CrossRef PubMed Google Scholar
Version history
  • Version 1 (September 2, 2025): Electronic publication

Article tools

  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal

Review Series

Microbiome in Health and Disease

  • Assessing the health of the gut microbial organ: why and how?
    Orlando DeLeon et al.
  • The microbiome as a modulator of neurological health across the maternal-offspring interface
    Stephanie B. Orchanian et al.
  • Skin microbiome and dermatologic disorders
    Tiffany C. Scharschmidt et al.
  • Microbial influencers: the airway microbiome’s role in asthma
    Young Jin Kim et al.
  • Diet’s impact on gut microbial assemblage in health and disease
    Carolina Koletic et al.
  • The gut microbiome and cancer response to immune checkpoint inhibitors
    Francesca S. Gazzaniga et al.
  • Social, microbial, and immune factors linking bacterial vaginosis and infectious diseases
    Nicole M. Gilbert et al.
  • Present and future of microbiome-targeting therapeutics
    Lauren E. Lynch et al.
  • Emerging concepts and shifting paradigms for understanding the microbial basis of inflammatory bowel diseases
    Megan S. Kennedy et al.

Metrics

  • Article usage
  • Citations to this article

Go to

  • Top
  • Abstract
  • Introduction
  • Role of the microbiome in IBD across contexts
  • Utilizing microbiome context to improve MBIs
  • Conclusions
  • Acknowledgments
  • Footnotes
  • References
  • Version history
Advertisement
Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts