v
Search
Advanced

Publications > Journals > Journal of Translational Gastroenterology> Article Full Text

  • OPEN ACCESS

Understanding Inflammatory Bowel Disease: An Integrative Framework of Microbiome, Metabolome, and Immunological Biomarkers

  • Prithvi S. Prabhu,
  • Rija Kalita,
  • Vanshika Sharma and
  • Tulika Prakash* 
 Author information 

Abstract

Inflammatory bowel disease (IBD) is characterized by chronic inflammation of the gastrointestinal tract and primarily includes ulcerative colitis and Crohn’s disease. As the number of patients suffering from IBD increases, diagnosis and treatment have become pressing yet challenging tasks. A major challenge is that patients with IBD often do not exhibit characteristic symptoms, making it difficult to distinguish IBD from other intestinal abnormalities. Endoscopy is the most conventional method used to diagnose IBD; however, this technique is invasive and costly. Therefore, there is a need to develop affordable, non-invasive diagnostic methods, which underscores the importance of identifying biomarkers specific to IBD. It is now well established that the gut microbiome plays a significant role in the development of IBD, and changes in the abundance of various gut organisms have been widely studied to identify microbial signatures associated with the disease. This review discusses the current state of knowledge regarding biomarkers in IBD, with a primary focus on the gut microbiome, associated metabolic signatures, and their links with immunological biomarkers. These biomarkers will help propose an integrative model to better understand the pathophysiology of this complex disease. Such an integrated approach also offers insights into potential therapeutic targets for designing more effective treatment strategies for patients.

Keywords

Inflammatory bowel disease, Microbial biomarkers, Metabolomic biomarkers, Immunological biomarkers, Integrated biomarker framework, Crohn’s disease, Ulcerative colitis

Introduction

Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal (GI) tract, subdivided into ulcerative colitis (UC) and Crohn’s disease (CD).1 IBD is associated with a range of complications that can significantly impact quality of life and health outcomes.2 The predominant symptoms—such as diarrhea, abdominal pain, GI bleeding, weight loss, malnutrition, and fatigue—can have substantial psychosocial consequences.3 Other health complications include strictures, fistulas, abscesses, and an increased risk of colorectal cancer, particularly in patients with long-standing disease or extensive colonic involvement.4 Extraintestinal manifestations, such as arthritis, skin disorders, and liver disease, further contribute to the burden of IBD, underscoring the need for comprehensive management strategies.5

Endoscopy remains the primary method for diagnosing IBD; however, it is invasive and time-consuming.6 Technologies such as artificial intelligence (AI), endocytoscopy, and molecular imaging have significantly enhanced endoscopic examinations, providing more accurate and detailed insights into IBD detection.7 AI improves risk prediction, genetic data analysis, and disease severity assessment through image analysis.8 Endoscopic molecular imaging offers valuable insights by assessing disease severity, predicting treatment outcomes, and detecting dysplasia even in inflamed tissue.6 While endocytoscopy provides highly detailed, real-time microscopic views of tissue during an endoscopy, with magnification up to 1,400 times,9 these advances still highlight the need for non-invasive diagnostic methods.

In IBD, immune system dysregulation drives disease progression through inflammatory mediators that perpetuate local inflammation, alter gut permeability, and result in gut dysbiosis.10 Employing gut microbiota and their metabolite biomarkers, which are characteristic of the disease, appears to be a promising non-invasive approach for diagnosis and monitoring. This strategy can aid in early detection and prediction of IBD, enabling timely interventions and reducing the risk of complications.11 This highlights the crucial, multifunctional role of various biomarkers in IBD, which can be utilized for diagnosis, treatment planning, and assessing mucosal healing in patients.12 This review examines the complex interplay among gut dysbiosis, metabolite alterations, and immunological responses in IBD. It seeks to clarify the mechanisms through which alterations in the gut microbiome and its related metabolites contribute to IBD development, emphasizing specific microbial signatures, metabolomic changes, and immune biomarkers associated with disease activity, progression, and treatment response. Furthermore, it discusses the emerging roles of AI and machine learning (ML) in the diagnosis, management, and personalized treatment of IBD. Through this integrative analysis, the article aims to enhance our understanding of IBD’s multifactorial nature and identify potential avenues for future research and clinical application.

Global burden and prevalence

Over the past few decades, the global burden of IBD has been rising, largely driven by changes in environmental, genetic, and lifestyle factors. Between 1990 and 2019, the global IBD population increased substantially, rising from 3.3 million to 4.9 million,13 with China and the United States reporting the highest numbers of cases.14 A 2019 study revealed that rates of IBD occurrence, deaths, and disability-adjusted life years were higher in older individuals compared to younger ones. Interestingly, men generally exhibited higher rates for these indicators than women until approximately 85 years of age, after which women had higher rates.15 The study also found that the highest number of IBD cases occurred in the 50–54-year age group for women and the 60–64-year age group for men. Additionally, the mortality rate associated with IBD was highest among individuals aged 95 years and older.13 Individuals with a family history of IBD, particularly CD, have a significantly increased risk of developing the condition. Siblings of CD patients are 13 to 36 times more likely to develop IBD, while those with a sibling affected by UC have a seven- to seventeen-fold higher risk. The presence of multiple affected family members further elevates this risk, especially in children. Offspring of two IBD-affected parents face the highest risk, ranging from 33% to 52%.16 A comprehensive analysis of 491 studies demonstrated a dramatic increase in both CD and UC rates over the past century. Incidence rates for CD and UC surged from fewer than one to over nine and fourteen cases per 100,000 people, respectively, with significantly rising prevalence trends.17 In 2021, a study in Iran analyzed the cost of illness, including medical treatments and other expenses, for IBD and found the annual cost per patient to be $1,077 for UC and $1,608 for CD. Patients over 40 years of age incurred the highest costs, with nationwide totals reaching $8.2 million for UC and $7.1 million for CD.18 A 2022 European study involving 3,687 IBD patients across 12 countries found that disease costs varied based on factors such as disease type, activity, comorbidities, age, gender, country, and healthcare system characteristics. UC patients generally had higher costs, particularly for medication.19

Factors affecting IBD

The etiology of IBD involves a complex interplay among genetic, environmental, and microbial factors (Fig. 1).20 Among the genetic factors, human leukocyte antigen (HLA) genes, particularly HLA class II molecules like HLA-DRB1-1502 and HLA-DRB1-1501, are strongly associated with UC, while HLA-DR4 is linked to CD in certain populations. These genes influence immune responses by presenting antigens to T cells, playing a critical role in disease susceptibility. Ethnic variability and clinical heterogeneity further contribute to the diverse genetic associations observed in IBD. Polymorphisms in cytokine genes, such as TNFα and LTα in the MHC class III region, regulate immune system activity and are linked to both UC and CD, although findings are often inconsistent. The interleukin (IL)-1 receptor antagonist gene allele 2 is associated with increased disease severity in UC and outcomes like pouchitis, highlighting its potential role in modulating inflammation. Family linkage studies also identify genetic predispositions, suggesting linkage disequilibrium near disease susceptibility loci. Other immune-related genes, such as ICAM-1, complement C3, and T-cell receptor genes, show conflicting associations, underscoring the complexity of IBD genetics. Collectively, these findings illustrate the intricate interplay between genetic predisposition and immune dysregulation in the development of IBD.21 Environmental factors, primarily diet, play a more significant role than genetics in shaping the microbiome; however, specific genotype–microbiome interactions remain important in IBD. For instance, the NOD2 gene variant is associated with an increased abundance of Enterobacteriaceae. A subset of microbiome taxa or functions, such as oxidative stress resistance, may be more directly linked to IBD despite environmental factors driving most microbiome variations.22 Given this complex array of influences, it is crucial to investigate the interplay among these factors for a more detailed understanding of the causes and potential treatments. This review discusses the gut microbiome and metabolome in detail, as well as their integration with immune-related biomarkers in IBD.

Integrated view of the factors associated with IBD.
Fig. 1  Integrated view of the factors associated with IBD.

IBD, inflammatory bowel disease.

Microbiome signatures in IBD

The microorganisms, including a variety of bacteria, fungi, and viruses, in the lower GI tract form an enormous and complex ecosystem.23 Beyond changes in the gut bacteriome of IBD patients, recent studies have reported remarkable alterations in the gut mycobiome and virome.23 This suggests that non-bacterial microbes, such as fungi and viruses, might also play unique and important roles in IBD pathogenesis and disease activity. Most gut microbes are beneficial and exert immunoprotective effects by regulating host immune cells. However, due to disease conditions or imbalances in the host system, alterations in the gut microbial ecosystem may occur, leading to microbial dysbiosis. Such disruptions may contribute to chronic intestinal inflammation and impaired gut barrier function, as seen in IBD. In this context, there is a shift in microbial populations, alongside inflammation or infection caused by contact between microbes and the damaged intestinal lining.24 Microbial dysbiosis has been demonstrated in both UC and CD.23

Compared to healthy individuals, the structure of the gut microbiota is significantly altered in IBD across different taxonomic levels.25 Changes in gut microbiota composition can occur early in IBD development.26 Thus, both diversity and composition of the gut microbiota are critical factors in disease progression. IBD significantly impacts alpha and beta diversity of the gut microbiome—where alpha diversity refers to diversity within local communities (habitats), and beta diversity refers to the spatial variation in species composition between communities.27 Studies show that patients with IBD exhibit reduced alpha diversity compared to healthy controls.28 This reduction renders the microbiota functionally less capable and less redundant, thereby increasing vulnerability to perturbations.29

Gut bacteria

More than 99% of the healthy gut bacteriome consists of species from four phyla: Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria.23 In IBD, there is a significant reduction in beneficial bacteria such as Faecalibacterium prausnitzii (F. prausnitzii) and members of the Ruminococcaceae family,30,31 with the exception of Ruminococcus gnavus,32 members of the Leuconostocaceae family,30 and Bifidobacterium species (Table 1).32–68 Conversely, pathogenic bacteria such as Campylobacter concisus, enterotoxigenic Bacteroides fragilis, Escherichia coli (E. coli), Bacteroides fragilis, Fusobacterium nucleatum, and Mycobacterium avium subspecies paratuberculosis are increased.35

Table 1

Organisms of the gut microbiome, their metabolites, and the immune factors they affect

Microbiome list
Gut bacteria in IBD
BacteriaAbundanceAssociated metaboliteMetabolite levelsEffect on immune componentReferences
F. prausnitziiDecreaseButyrateDecreaseTh17/Treg balance disrupted. Blocks IL-6/STAT3/IL-17 pathway and promotes pro- inflammatory effect37,38
EnterobacteriaceaeIncreaseLPSIncreaseReduced IL-10. Increased IL-8, tumor necrosis factor (TNF)-α, and IL-1β36
LactobacilliDecreaseIndole-3-lactic acidDecreaseImpairs CD8+ T cells and IL-12a production3941
Exopolysaccharides (EPS)DecreasesIncreased pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6). Decreased anti-inflammatory cytokines (IL-10)
Conjugated linoleic acidDecreaseProduction of colonic IL-6 and TNF-α
BifidobacteriaDecreaseIndole-3-lactic acidDecreaseIncreased production of IL-8 and TNF-α, and pro-inflammatory cytokines42,43
FlavobacteriumDecreaseCitric acidDecreaseProduction of TNF-α, IL-6, IL-12 and promotion of pro- inflammation44,45
Trimethylamine-N-oxide (TMAO)IncreaseImpacts ATG16L1-induced autophagy. Activates NLRP3 inflammasome. Promotes inflammation44,46
BacteroidetesDecreaseSCFADecreaseReduced Treg cells47
E. coliIncreaseLPSIncreaseIncreased IL-8 and other pro-inflammatory cytokines35,48,49
Mycobacterial speciesIncreaseSCFADecreasesReduced suppression of NF-κB; Reduced activation of inflammasomes; Reduced Treg cells. Reduction in anti-inflammatory mediators (TGF-β and IL-10)35,50,51
AnaerostipesDecreaseButyrateDecreasesIncrease in NF-κB-induced pro-inflammatory cytokines TNFα and IL652,53
MethanobrevibacterDecreaseMethaneDecreasesIncreases IL-6, TNF-α, IL-1β, IFN-γ, NF-κB5456
ChristensenellaceaeDecreaseAcetate, ButyrateDecreasesIncrease in IL-8, NF-κB activation57
Ruminococcus gnavusIncreaseGlucorhamnan polysaccharideIncreasesIncreases TNFα32
Prevotella copriDecreaseValerate and other SCFADecreasesIncreases TNFα5861
Clostridium leptum group (IV)DecreaseButyrateDecreasesReduced Treg differentiation. Reduced IL-22 production. Promotes inflammation through LPS-induced NF-kβ activation. Increased pro-inflammatory factors62,63
Gut mycobiome in IBD
C. albicansIncreaseSCFADecreaseIncreases IL-17 and IL-23 production64,65
MalasseziaIncreaseFFAs such as oleic acid, cell wall carbohydrates, indolesIncreaseIncrease production of IL-17 IL-18, IL-8, and IL-6 and Th22 chemokines64,66
KluyveromycesDecreaseβ-glucanDecreasesReduction in Treg and IL-1064,67,68

Short-chain fatty acids (SCFAs), produced by gut microbiome members, are important for GI tract homeostasis.69 The association between SCFAs and IBD was suggested when alterations in associated microbiota, such as Bifidobacteria, were observed to affect SCFA levels.33 In UC patients, a reduction of Bifidobacteria in the colon was observed, which is an important SCFA producer.34,70 SCFAs and gut microbiota in IBD influence reactive oxygen species, whose increased levels can damage the mucosal layer of the GI tract.71 This damage may increase intestinal permeability, resulting in a leaky gut. Microbes such as Streptococcus, Bifidobacterium, and Lactobacillus, often administered as probiotics, help inhibit reactive oxygen species production and maintain a healthy intestinal microbiota.72 Disturbances in gut microbiota composition and reduced fermentation of dietary fibers in IBD might lead to altered SCFA profiles.71 In a study of IBD patients, reduced tryptophan metabolism was observed, presumed due to altered bacterial communities.73 Dysbiosis in IBD is marked by an increase in Enterobacteriaceae,74 facultative anaerobes that utilize electron acceptors, such as nitrate, to generate energy. These taxa may impair gut barrier function through inflammatory cytokine production. Consequently, increased Enterobacteriaceae alter bile acid metabolism and decrease tight junction integrity, resulting in loss of impermeability in the intestinal epithelium.36 The beneficial bacterium Lactobacillus is decreased in IBD and has shown anti-inflammatory effects in mouse models.75 In CD, a lower abundance of F. prausnitzii signals potential intestinal health issues in adults.76F. prausnitzii plays an essential physiological role by providing mucosal protection and anti-inflammatory functions.76 Gut bacterial signatures have been employed as biomarkers, demonstrating associations with mucosal state and related symptoms in UC patients.77 Certain bacteria, such as Enterobacteriaceae, Klebsiella, and some Lachnospiraceae species, are more abundant in UC patients experiencing symptoms like frequent bowel movements.77

Probiotic administration has shown beneficial effects on gut microbiota and therapeutic benefits in various diseases. For example, rectal infusion of Lactobacillus reuteri ATCC 55730 improved mucosal inflammation in pediatric patients with distal active UC and altered cytokine expression involved in IBD pathogenesis.78 Another study found that administration of Lactobacillus delbrueckii and Lactobacillus fermentum decreased inflammatory cytokines, suggesting probiotics may help prevent UC.79 The major anaerobic bacterial species in the colon, Bacteroides, is reduced in UC; however, administering Lactobacilli and Bifidobacteria prior to experimental UC induction helped stabilize Bacteroides levels, reducing inflammation and tissue damage.80 A correlation between gut bacterial composition and potential future development of CD was revealed using ML techniques, implying a role for gut bacteria in disease pathogenesis. Based on this, a microbiome risk score was developed, assigning risk scores to individuals based on their gut bacterial profiles, particularly in healthy first-degree relatives of CD patients.81 These studies demonstrate the potential for using microbial profiles as distinctive markers. By longitudinally tracking changes in these microbial signatures, researchers may predict disease flares or treatment responses.

Extracellular vesicles (EVs) in IBD

EVs are tiny spheres enclosed by lipid bilayers that play a crucial role in the release and transport of various substances, including carbohydrates, lipids, cell wall components, proteins, DNA, RNA, and signaling molecules.82 Intestinal EVs engage in direct or indirect interactions with immune cells, intestinal epithelial cells, and the gut microbiota, actively participating in the regulation of anti-inflammatory responses, restoration of mucosal barrier integrity, and reconstitution of microbiota composition.83 EVs are released by various types of cells, such as intestinal epithelial cells, immune cells (including macrophages, T cells, B cells, NK cells, polymorphonuclear neutrophils, and dendritic cells), and the microbiota.83–85 EVs derived from intestinal epithelial cells maintain gut homeostasis and modulate immune responses.83 EVs from T cells are involved in intercellular communication and immune modulation.85 EVs from B cells also contribute to immune responses.86 By stimulating macrophages, EVs containing inflammasomes can activate the NF-κB pathway, thereby amplifying inflammatory signaling.87 EVs derived from the microbiota can influence host immune responses and inflammation.88 Several mechanisms of EV release have been described and warrant further study. For example, plasma membrane budding involves the direct outward budding of the plasma membrane and requires GTPases and the ESCRT complex.84 EVs can also be released through the fusion of plasma membranes with multivesicular bodies, leading to the release of exosomes.84 Another mechanism is calcium-dependent release, in which calcium functions as a second messenger and regulator of EV secretion.89 EVs facilitate intercellular communication by carrying a variety of bioactive molecules, including nucleic acids, proteins, lipids, and metabolites, which can initiate intracellular signaling pathways.90,91 These cargoes can include tricarboxylic acid (TCA) cycle intermediates, steroid hormones, sterols, enzymes, signaling proteins, surface receptors, mRNAs, microRNAs (miRNAs), sphingolipids, and phospholipids. EVs can also elicit responses in recipient cells by initiating intracellular signaling pathways.90,91

Recent studies have found that circulating miRNAs contained in EVs are small, non-coding RNA molecules that can serve as biomarkers for diseases.92 In the context of IBD, miRNAs are of particular interest as potential biomarkers. For example, elevated expression of circulating miRNAs has been detected in EVs isolated from the serum, plasma, or peripheral blood of individuals with IBD.93 Proteomic analyses comparing EVs from individuals with IBD and healthy controls have identified certain proteins, including ANXA1 and PSMA7, that are enriched in the EVs of IBD patients.94,95 Notably, levels of EV-associated PSMA7 were found to be lower in IBD patients in remission compared to those with active disease, highlighting the potential utility of EV-based biomarkers for monitoring disease progression in IBD.95 By addressing various aspects, such as release mechanisms, signaling pathways, and the role of EVs as biomarkers, a comprehensive understanding can be developed of how EVs contribute to the pathophysiology of IBD.

Gut virome

Viruses are increasingly recognized as integral components of the human microbiome, serving diverse ecological functions, including preying on bacteria, stimulating the immune system, facilitating genetic diversity, enabling horizontal gene transfer, fostering microbial interactions, and enhancing metabolic processes.96 The interaction between bacteria and viruses in the gut highlights the role of viruses in maintaining gut equilibrium and influencing pathological states.97 The healthy human gut virome includes members of the Malgrandaviricetes (spherical ssDNA) and Caudoviricetes (tailed dsDNA) phage classes.98 Although the bacterial hosts of many of these viruses remain largely unidentified, Caudoviricetes are presumed to infect a diverse array of bacterial phyla, including Bacteroidetes, Verrucomicrobia, Proteobacteria, Firmicutes, and Actinobacteria.99

Recently, important associations have been observed between the gut virome and IBD. Virome dysbiosis has been implicated in IBD pathogenesis, with elevated levels of phages infecting Clostridiales, Alteromonadales, and Clostridium acetobutylicum detected in individuals with IBD compared to healthy subjects.100 A study on Chinese cohorts of IBD patients and healthy controls identified 139 IBD-associated viral OTUs, noting a higher abundance of the Retroviridae family in IBD patients101 Additionally, IBD patients showed increased evenness and richness in their eukaryotic virome compared to healthy controls. Genomoviridae and Retroviridae were two eukaryotic viral families found to be enriched in IBD patients. In contrast, the prokaryotic virome in IBD patients displayed significantly decreased diversity. Families such as Siphoviridae and Myoviridae were enriched in patients, while crAss-like phages and Quimbyviridae were decreased. At the OTU level, numerous IBD-enriched Siphoviridae and Myoviridae viral OTUs were identified, which infect bacteria such as Escherichia, Klebsiella, and other opportunistic pathogens known to induce inflammation and trigger disease. Moreover, fecal virome transplantation in mouse models has shown that colonization by these IBD-associated viruses can modulate experimental colitis.101 However, more comprehensive and focused research is required to achieve a detailed understanding of the virome’s role in IBD (Table 2).101–104

Table 2

Changes in the abundance of the gut virome in IBD

Gut viromeAbundanceRoleReferences
MyoviridaeIncreaseThese are temperate viruses belonging to the order Caudovirales, but their function in IBD virome remains mostly unclear101
MicroviridaeDecreaseBelonging to the order Petitivirales, impacts on bacterial dysbiosis102
SiphoviridaeIncreaseTemperate viruses belonging to the order Caudovirales, impacts on bacterial dysbiosis101,103
QuimbyviridaeDecreaseRole in IBD remains unclear101
GenomoviridaeIncreaseBelonging to order Geplafuvirales, the role remains unclear101
AnelloviridaeIncreaseThey are useful for reporting reduced immune surveillance and the effectiveness of immunosuppression104
RetroviridaeIncreaseThe overgrowth of this family has been linked to several diseases, including CD, but its exact role remains unclear101

Gut mycobiome

Fungi make up approximately 0.1% of the gut microbiome and have been identified in the GI tract of around 70% of healthy individuals. They interact with both viruses and bacteria in the gut, exhibiting antagonistic and synergistic relationships.105 The healthy human GI tract mycobiome is primarily composed of three major phyla: Basidiomycota, Ascomycota, and Chytridiomycota.105 Dysbiosis in the fungal community plays a crucial role in IBD by altering gut microbiota composition or promoting the production of pro-inflammatory cytokines.106 A distinctive feature of IBD is an increased Basidiomycota-to-Ascomycota ratio (Table 3).64,66,107–110 An increase in the abundance of fungi such as Candida species, which can exacerbate inflammation, and a decrease in Saccharomyces have been observed in IBD cases.108–110 Another report indicated elevated abundances of Sterigmatomyces, Aspergillus, Candida, and Wickerhamomyces, along with lower abundances of Penicillium, Exophiala, Alternaria, Acremonium, Trametes, Epicoccum, and Emericella in patients with UC.111 In a clinical study involving colon biopsies from 10 IBD patients and 18 healthy controls, Pseudomonas was elevated, and the opportunistic pathogen Malasseziales was found to be the most abundant in UC. An increased Basidiomycota-to-Ascomycota ratio was also observed in UC compared to CD, due to the higher abundance of Malasseziales, which may serve as an indicator of UC.112 Fungal dysbiosis has been shown to promote IBD by enhancing CD4+ T cell responses in a mouse model, as well as in human colonic and CD4+ T cell samples from healthy donors, UC patients, and CD patients. In this report, Candida albicans was found to increase pro-inflammatory cytokine production, and slower progression of IBD was observed when terbinafine was used to deplete fungi.113

Table 3

Changes in the abundance of the gut mycobiome in IBD

Abundance
IncreasedDecreasedReferences
C. albicans; Malassezia; Filobasidiaceae;D. hansenii; Xeromyces; Rhodosporium; Lipomyces; Yamadazyma; Yamadazyma friedrichii; Lipomyces doorenjongiiSaccharomyces cerevisae; Saccharomyces boulardii; Kluyveromyces;C. tropicalis; Zygomycota; Aspergillus; Debaromyces; Cladosporium; Microdochium; Phaeosphaeria; A. rubrobrunneus64,66,107110

Correlating microbial signatures with disease outcome and treatment response

When discussing microbial signatures in IBD, it is important to highlight the specific compositional changes in the gut microbiota associated with disease activity, progression, and treatment response. In patients with IBD, the intestinal microbiota is dysregulated compared to that of healthy individuals, showing decreased bacterial diversity—particularly reduced abundances of Firmicutes and Bacteroidetes—and an increase in Proteobacteria.114 Differences also exist between CD and UC. Patients with CD exhibit a higher level of dysregulation, characterized by reduced microbial diversity and stability, which can be considered a specific signature of CD.115 Microbial biomarkers, such as specific bacterial strains or metabolites, can indicate whether a patient is likely to respond to a particular treatment. For instance, in patients with CD treated with the anti-integrin therapy vedolizumab, those who achieved remission had a gut microbiome enriched with Roseburia inulinivorans and a Burkholderiales species compared to non-responders.116 Similarly, in pediatric IBD patients, a higher absolute abundance of Bifidobacteriales and a lower abundance of Actinomycetales at baseline were associated with a rapid response to infliximab therapy.117 Additional studies like these will help us better understand the microbial signatures associated with IBD and support the development of personalized treatment strategies, ultimately improving patient outcomes.

Metabolome signatures in IBD

The metabolome refers to the pool of small metabolites present in a biological sample under specific conditions at a particular time.118 Important metabolites include lipids, amino acids, and TCA cycle intermediates, among others. A diverse range of biosamples, including easily accessible fluids such as blood, urine, serum, feces, and saliva, as well as less accessible and more invasive samples such as organs, tissues, or cells, has been used to identify metabolomic markers in IBD.119 These samples provide varying degrees of information: for example, urine provides a comprehensive overview of endogenous and exogenous metabolism; stool gives insight into digestive metabolism; blood offers a systemic perspective; tissue samples provide direct information on localized pathology; and less conventional samples like breath may reflect dynamic metabolic processes.120

Several studies have reported variations in metabolites from different types of biosamples in patients with IBD compared to healthy controls. For example, in a large cohort of 117 individuals with CD, increased levels of 1-octen-3-ol, 6-methyl-2-heptanone, 2-piperidinone, and heptanal were observed in fecal samples from the active CD group compared to healthy controls. Conversely, reduced quantities of methanethiol, 3-methyl-phenol, SCFAs, and ester derivatives were observed in CD patients.121 Another study found that fecal metabolites could distinguish between UC and CD, as well as between healthy controls and UC. Fecal samples from CD patients showed enrichment in lactate, succinate, alanine, and tyrosine, while UC patients exhibited higher levels of leucine, alanine, and tyrosine.122 In yet another study, 53.6% of lipid metabolites were significantly altered in CD compared to controls, whereas only five lipid-related metabolites were decreased in UC. Both CD and UC exhibited consistent decreases in various fatty acids compared to controls. Interestingly, glycerol levels were notably reduced in CD, indicating lipolysis. Essential acylcarnitine metabolites were also reduced in CD compared to both UC and control groups. Bile acid pathways were significantly altered in IBD, with increased primary and secondary bile acids observed in CD, while UC displayed reduced primary bile acids and altered secondary bile acids. TCA cycle intermediates, including citrate, aconitate, α-ketoglutarate, succinate, fumarate, and malate, were significantly decreased in CD. Additionally, β-hydroxybutyrate, derived from acetyl-CoA, showed the most substantial reduction in CD, being 11 times lower than in controls and 18 times lower than in UC subjects.123

Metabolic dysfunction is defined as a series of abnormal or disrupted metabolic processes occurring in the body, particularly those related to energy production, nutrient utilization, or the regulation of various molecules. In IBD, this dysfunction is marked by decreased levels of trimethylamine-N-oxide, reduced SCFAs like butyrate, lower hippurate levels, and alterations in primary and secondary bile acid profiles.120 This dysbiosis can contribute to inflammation by influencing metabolic pathways or the immune system.124 Several metabolites in the gut are derived from the resident microbiota. Therefore, microbial dysbiosis is expected to be associated with metabolic imbalance. For example, in IBD, there is an imbalance in microbial composition, with an increase in Proteobacteria and a decrease in Firmicutes. Thus, the roles of specific bacteria, such as E. coli and the butyrate-producing F. prausnitzii, become particularly important. This suggests that combining microbiome and metabolome analyses may provide valuable insights for understanding, diagnosing, and treating IBD.120 Supporting this idea, Lijun Ning et al. (2023) identified unique biomarkers related to IBD that consist of specific gut bacteria and metabolites. These biomarkers are expected to have a low likelihood of being incorrectly identified in both GI and non-GI diseases, suggesting their potential as valuable disease-specific markers for IBD with diagnostic utility.125

Metabolomic profiling has also proven valuable in predicting treatment responses in IBD patients by identifying specific metabolites associated with therapeutic outcomes. Key metabolites such as bile acids, glycine, linoleic acid, N-acetylserotonin, and methylglutaric acid have been linked to responses to therapies like anti-TNF and infliximab, with distinct profiles observed between responders and non-responders. Notably, bile acids, along with urinary cysteine and bile acids measured in various bodily fluids, have emerged as potential indicators of treatment efficacy. Furthermore, fecal lipid profiles have shown higher predictive accuracy than serum profiles. These findings emphasize the potential of metabolomic analyses, particularly of fecal samples, to enhance personalized treatment strategies and improve understanding of IBD’s metabolic alterations.126

To explore metabolite profiles, it is essential to implement highly sensitive techniques for metabolomics analysis. The two key analytical techniques used in metabolomics are nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry (MS). NMR provides insights into metabolite structure and concentration by detecting energy changes in nuclei under a magnetic field. It includes 1D-NMR, which is commonly used in high-throughput studies, and 2D-NMR, which resolves overlapping peaks for complex metabolite characterization using techniques such as COSY and TOCSY.127 While NMR faces limitations such as low sensitivity and overlapping proton peaks, it is expected to remain a vital tool in healthcare for at least the next decade due to advantages including minimal sample preparation, high reproducibility, and the ability to analyze an entire sample in a single measurement.128

MS, on the other hand, measures the mass-to-charge ratio (m/z) of ionized metabolites, offering high sensitivity and specificity. MS is often coupled with chromatographic separation (e.g., liquid chromatography-MS or gas chromatography-MS) to reduce sample complexity and improve accuracy.127 However, structural elucidation in untargeted analyses presents significant challenges, particularly with stability and reproducibility during large-scale sample runs using untargeted liquid chromatography-MS. These issues underscore the need for advancements that enhance reproducibility and improve the accuracy of structure elucidation.128

In IBD research, untargeted metabolomics is particularly valuable as it captures a holistic metabolic profile without predefined targets. The workflow includes spectral processing to generate metabolic features, data analysis to explore associations with phenotypic traits, biomarker discovery for diagnostics, and pathway analysis to connect metabolites with biological processes. Advanced bioinformatics tools are essential for managing the complexity of metabolomics data, enabling multi-omics integration, and identifying biomarkers and pathways relevant to IBD. Together, these techniques provide a comprehensive understanding of disease mechanisms and support the development of personalized therapeutic strategies.129

Immunological biomarkers in IBD

The need for precise diagnostic tools and effective treatment strategies for IBD highlights the importance of immunological biomarkers. Intensive research aimed at identifying these biomarkers, coupled with the vast amount of related data being generated, now enables speculation about the dynamics between immunological components and the gut microbiome. Understanding how dysbiosis in the gut microbiome can affect the regulation of various immune system components is crucial for elucidating the mechanisms by which the disease progresses. Numerous studies have identified different immunological biomarkers, including various types of T cells, receptors, antibodies, ILs, and cytokines. These biomarkers can be classified according to their clinical applications: diagnosis, prognosis prediction, and treatment monitoring.

Diagnosis

Immunological biomarkers are essential for the accurate diagnosis of IBD, helping to differentiate it from other GI disorders, particularly in cases where clinical symptoms are ambiguous. Specific biomarkers, such as anti-Saccharomyces cerevisiae antibodies and perinuclear anti-neutrophil cytoplasmic antibodies, are commonly used to distinguish between CD and UC, respectively.130 These markers provide less invasive diagnostic options and complement traditional methods like endoscopy. Elevated levels of pro-inflammatory cytokines, including IL-1β and IL-8, have been identified in IBD patients, aiding in the identification of active inflammation. IL-1β is a biomarker noted for its elevated levels in the serum of patients with IBD, in those experiencing relapse, and in pediatric cases.131,132 IL-1β alone is not capable of exacerbating IBD, but its effect is mediated through fluctuations in the levels of inflammasomes like NLRP3, an important source of mucosal IL-1β.133 Lower levels of the NLRP3 inflammasome, and consequently lower levels of IL-1β, lead to a decreased state of colitis in mice.134 Furthermore, in mice, aggravated intestinal inflammation is linked to bacterial members of the genus Prevotella, mainly Prevotella intestinalis, which can reduce the production of SCFAs, especially acetate.135 Interestingly, acetate is now known to suppress NLRP3 inflammasome-mediated production of IL-1β.136 From these studies, it can be understood that in severe cases of intestinal inflammation, Prevotella species reduce acetate production, which in turn increases IL-1β production and promotes inflammation. Similarly, IL-8 levels are elevated in the serum of patients with IBD relapse and in pediatric cases.131,132 The increase in IL-8 levels in CD and UC can be explained by the higher abundance of various Enterobacteriaceae, such as E. coli, and the reduced abundance of Bifidobacteria and Lactobacillus species in the gut.137–139 A study using IBD isolates showed that flagellin shed by mucosa-associated E. coli induces IL-8 expression through a MAPK-dependent pathway.139Bifidobacterium species are also known to bind and neutralize lipopolysaccharides (LPS) from E. coli, which, if not neutralized, can lead to LPS-induced increases in IL-8 levels.140 It is possible that due to the increased levels of E. coli and lower levels of Bifidobacteria, LPS produced by E. coli is not effectively neutralized, leading to more LPS-induced IL-8 production. Bifidobacterium is also an SCFA-producing bacterium, and these SCFAs exert an anti-inflammatory effect by blocking the NF-κB signaling pathway. Thus, low levels of Bifidobacteria in IBD, and consequently lower production of SCFAs, can result in increased pro-inflammatory consequences.33

Similarly, an experiment in IBD patients demonstrated that an increase in Lactobacillus species, and thus increased butyric acid production, leads to decreased levels of IL-8 and other pro-inflammatory molecules.141 From this, we can infer that lower levels of Lactobacillus may contribute to elevated IL-8 levels. Dysbiosis in the gut microbiome, characterized by altered levels of bacterial species such as Prevotella intestinalis and E. coli, further influences the production of these cytokines, reinforcing their diagnostic relevance.

Prognosis prediction

Immunological biomarkers are instrumental in predicting the progression of IBD and identifying patients at risk for severe disease or complications. For instance, Oncostatin M (OSM), which belongs to the IL-6 family of cytokines, has been shown to be an important biomarker in IBD.142 It is rapidly released during degranulation and can initiate other signaling pathways, such as the JAK-STAT and PI3K-Akt pathways, which promote disease progression.142 Mucosal OSM levels have been found to be upregulated in patients newly diagnosed with IBD and in those experiencing relapse, even after surgery.142 High levels of OSM have been identified in patients with both CD and UC, and have also been shown to predict non-responsiveness to anti-TNFα therapy.143 Dysbiosis in CD has been associated with a decreased abundance of Roseburia intestinalis, a gut microbiome member, and reduced levels of this species are linked to increased OSM levels in CD patients.144 Interestingly, normal R. intestinalis abundance can suppress intestinal inflammation by downregulating pro-inflammatory cytokines and increasing anti-inflammatory cytokines and regulatory T cells (Tregs).144 This effect can be explained by the fact that R. intestinalis is an SCFA-producing bacterium capable of synthesizing butyrate, which has multiple effects on immune regulation, including promoting the proliferation of Tregs.145 From this, it can be understood that in IBD, dysbiosis results in decreased R. intestinalis abundance, leading to reduced butyrate production, downregulation of Tregs and anti-inflammatory factors, and upregulation of pro-inflammatory mediators, including OSM.

Treatment monitoring

Biomarkers also play a critical role in monitoring treatment response and guiding therapeutic decisions in IBD. A recent study was performed to identify IBD remission- or relapse-specific biomarkers. A list of ILs, cytokines, and other immunological factors, including Galectin-1, IL-15, IL-21, IL-25, IL-13, IFN-β, CXCL11, CXCL9, CXCL10, and G-CSF, whose levels are elevated in patients with relapse.132 The possibility that higher levels of IL-15 could result from changes in the levels and composition of the Bacteroidetes and Firmicutes phyla, as well as decreased abundance of butyrate-producing bacteria (leading to reduced butyrate and other SCFA levels), has already been highlighted.146 The link between lower levels of butyrate and other SCFAs with elevated IL-15 levels may be similar to the mechanisms observed for IL-8 and IL-1β. An experiment performed on patients with hepatocellular carcinoma showed that increased levels of gram-negative bacteria and decreased levels of gram-positive bacteria, such as Firmicutes, are associated with elevated IL-25 levels.147 The increased levels of IL-25 observed in IBD may similarly result from dysbiosis. In IBD, significantly higher levels of the bacteriophage Caudovirales have been observed; combined with bacterial infection (where these phages replicate within bacteria), this can trigger pro-inflammatory effects, heightened T cell immune responses, and IFN-β production, while suppressing phagocytosis and TNF production, thereby maintaining gut inflammation.148 It is now known that CXCL9, CXCL10, and CXCL11 have strong bactericidal activity, and their increased levels may be linked to the higher abundance of E. coli in IBD, potentially resulting in infections by bacteria such as Listeria monocytogenes and Bacillus anthracis.149–151

Another study of CD and UC patient samples using mass cytometry identified immunological molecules characteristic of IBD samples, including CXCR3+ plasmablasts, HLA-DR+CD38+ T cells, and IL1B+ macrophages and monocytes. Specifically in UC samples, elevated IL17A+ CD161+ effector memory T cells, HLA-DR+CD56+ granulocytes, and reduced type 3 innate lymphoid cells were observed. In CD samples, IL1B+ dendritic cells, IL1B+TNF+IFNG+ naïve B cells, IL1B+HLA-DR+CD38+ T cells, and IL1B+ plasmacytoid dendritic cells were identified.152 How gut dysbiosis influences the levels of these immune components is not yet fully understood and warrants further exploration. Much remains unknown about how changes in the abundance and composition of the gut microbiome lead to higher levels of these biomarkers.

Integration of the gut-microbiome, metabolome, and immunological aspects in IBD

The sections thus far have discussed three types of biomarkers—the gut microbiome, the gut metabolome, and immunological biomarkers—independently. However, it is important to consider the interplay between gut dysbiosis, alterations in metabolite levels, and the consequent increase in pro-inflammatory factors and reduction of anti-inflammatory factors. As already established, IBD is characterized by gut dysbiosis, which constitutes the first group of biomarkers. This imbalance in the abundance of different gut microbiota leads to a corresponding imbalance in the levels of various associated metabolites. Broadly, SCFAs and LPS represent two common groups of metabolites whose levels decrease and increase, respectively, due to gut dysbiosis, forming the second group of biomarkers. Various cytokine levels are affected by these changes in metabolite concentrations. The cytokines whose levels change significantly and form the third group of biomarkers include IL-12, IL-10, IL-8, IL-6, IL-1β, and TNF-α. An overall representation of this integrated concept of the three biomarkers is shown in Figure 2. This concept can be further illustrated by examining F. prausnitzii, whose abundance in the gut decreases in IBD. This bacterium is a major butyrate producer, and its reduced abundance leads to a corresponding decrease in butyrate levels.153 Butyrate, an SCFA, impacts the host immune system, supporting the observation that reduced butyrate levels disrupt the Th17/Treg balance. It also inhibits the IL-6/STAT3/IL-17 pathway and promotes pro-inflammatory effects, ultimately exacerbating the disease.144 This is just one example of a mechanism linking all three types of biomarkers seen in IBD. Other such mechanisms can be similarly understood from Table 1 and are visually summarized in Figure 3. Considering the interactions mentioned above and those outlined in Table 1, it may be hypothesized that: i) alterations in gut microbiome composition directly or indirectly influence the production of immunological biomarkers in IBD patients; ii) the metabolomic signatures associated with dysbiosis in IBD correlate with specific immunological responses and disease severity; and iii) modulating the gut microbiome may lead to measurable changes in both immunological biomarkers and metabolomic profiles, resulting in improved clinical outcomes for IBD patients. The interplay between the gut microbiome, metabolome, and immune factors in IBD presents numerous complexities and unresolved questions. One significant uncertainty lies in the causal relationships between specific microorganisms, metabolites, and immune responses. While associations have been established, the directionality of these relationships remains unclear. For instance, it is uncertain whether dysbiosis leads to altered immune responses or if inflammation modifies microbiome composition. Alterations in metabolite profiles, as discussed above, have been observed in IBD patients, yet the specific metabolic pathways affected and their implications for immune function are not fully elucidated. The role of metabolites produced by gut bacteria in modulating immune responses involves complex interactions that warrant further investigation. Individual variability in immune responses to microbial and metabolic signals further complicates our understanding of pathogenesis. Factors such as genetics, environmental influences, and prior exposures can significantly affect how the immune system interacts with the microbiome. To address these uncertainties, several research methods and technical approaches could be employed. Conducting longitudinal studies that track changes in microbiome composition, metabolomic profiles, and immunological biomarkers over time could help establish causal relationships. This approach would allow researchers to observe how shifts in one domain influence the others. Utilizing an integrative multiomics approach (combining genomics, transcriptomics, proteomics, and metabolomics) can provide comprehensive insights into the interactions between microorganisms, metabolites, and immune factors. Advanced computational models could analyze this data to identify key pathways involved in IBD pathogenesis. Developing animal models that mimic human IBD can facilitate controlled experiments to test specific hypotheses about microbial influence on immune responses. For example, germ-free mice could be colonized with specific bacterial strains to observe subsequent changes in immune activation and metabolite production. Designing clinical trials that monitor immunological biomarkers alongside microbiome and metabolome changes during treatment could provide insights into how therapies influence these interactions. This could lead to personalized treatment strategies based on individual biomarker profiles. These uncertainties need to be addressed through targeted research methods to deepen our understanding of IBD pathogenesis and potentially uncover new therapeutic targets for managing this complex disease.

Dynamics between gut microbiome, metabolome, and immunological components in IBD.
Fig. 2  Dynamics between gut microbiome, metabolome, and immunological components in IBD.

Created with Biorender.com. IBD, inflammatory bowel disease; SCFA, short-chain fatty acids; LPS, lipopolysaccharide; IL-6, interleukin-6; IL-8, interleukin-8; IL-10, interleukin-10; IL-12, interleukin-12; TNF-α, tumor necrosis factor-alpha; IL-1β, interleukin-1 beta.

Visual representation of the interactions between gut microbiome, metabolome, and immunological markers summarized from .
Fig. 3  Visual representation of the interactions between gut microbiome, metabolome, and immunological markers summarized from Table 1 and Table 2.

Created with Biorender.com. Text in red and green indicates a decrease and increase in levels, respectively. SCFA, short-chain fatty acids; FFA, free fatty acids; LPS, lipopolysaccharide; IL-10, interleukin-10; Treg cells, regulatory T cells; IL-1β, Interleukin-1 beta; IL-8, interleukin-8; IL-17, interleukin-17; NF-κB, nuclear factor kappa B; TNF-α, tumor necrosis factor-alpha.

AI-ML-based advancements

As our understanding of IBD progresses and we strive to enhance clinical trial outcomes and treatment goals, AI and ML have emerged as promising tools to improve diagnostic processes and treatment outcomes. Various ML algorithms, such as Random Forest and Support Vector Machines, have demonstrated efficacy in predicting patient responses to therapies and assessing disease severity.8 These algorithms utilize high-dimensional data ranging from clinical genomics to microbiome data, enabling a better understanding of individual patient profiles. A study proposes the Holistic AI in Medicine framework, which uses multimodal inputs such as tabular data, time series, text, and images to enhance predictive modeling in healthcare. The integration of multimodal data using AI techniques has been shown to improve diagnostic accuracy.154 However, many challenges remain unresolved in this field. One major issue is the lack of diversity in patient samples, which can lead to biased predictions. This highlights the need to develop robust AI models that generalize well across populations. Research has shown that AI models trained predominantly on data from specific demographics can result in imbalances in healthcare outcomes, with less accurate algorithms for underrepresented racial or ethnic groups.155 Moreover, the increasing applications of AI in managing IBD present exciting prospects. AI may not only predict how individuals respond to biological therapies but also contribute to refining the standard of care. This sets the groundwork for personalized treatment in the future, with the potential to reduce costs and improve overall disease management.156 The integration of AI in the treatment and diagnostics of IBD offers significant potential for enhancing patient care through improved and personalized strategies. To recognize and address these challenges, future research must focus on validating AI systems in real clinical environments using diverse data to optimize the models.

Future research directions

Future research on understanding IBD should focus on conducting large-scale longitudinal studies to explore the intricate relationships between immunological biomarkers, the microbiome, and the metabolome.157 These studies should aim to recruit a diverse cohort of participants, including those diagnosed with CD and UC, alongside healthy controls for baseline comparisons. Inclusion criteria must consider age diversity and disease duration to effectively evaluate temporal changes in biomarkers, with a target sample size of 500–1,000 participants to ensure robust data analysis.

The study design should adopt a prospective cohort framework, facilitating data collection at multiple time points, such as baseline, six months, and annually thereafter. Regular follow-ups will be essential for gathering biological samples (blood and feces) and clinical data, including symptom diaries and medication use. An intervention group receiving dietary modifications or probiotics could provide valuable insights into their impact on biomarkers over time.

Data collection methods must encompass comprehensive biological sampling for immunological analysis and microbiome sequencing, alongside standardized clinical assessments to evaluate disease activity. Advanced statistical analyses and ML approaches will be crucial for identifying patterns that predict disease flares or treatment responses based on microbiome and metabolomic changes.158 Ethical considerations are paramount; thus, obtaining informed consent and ethics approval from relevant boards is essential. Finally, dissemination of findings through publication in peer-reviewed journals and engagement with healthcare providers will enhance awareness of potential biomarkers and their clinical relevance.159 By implementing these structured strategies, researchers can gain valuable insights into the complex dynamics of IBD, ultimately informing future therapeutic strategies and improving patient outcomes.

Conclusions

In this review, we have summarized biomarkers of IBD with a major focus on signatures in the gut microbiome. We discussed the characteristic variation in the levels of different organisms in the gut, how this variation causes fluctuations in the levels of metabolites produced by these organisms, and finally, how the imbalance of these metabolites can induce altered levels of various immune system components. Thus, this review provides a comprehensive outlook on microbial, metabolic, and immunological signatures and their interrelations. Although extensive research is being conducted to identify biomarkers for IBD, the ones identified so far are not ideal. Most reported biomarkers are invasive, lack specificity to IBD, and are not highly sensitive. Moreover, biomarkers reported across different studies are inconsistent due to variations in study protocols, sample sizes, populations, environmental influences, experimental bias, and other factors. This implies that there is no standardized panel of biomarkers that can be universally applied across all populations and stages of IBD. The biomarkers known so far need to be validated through longitudinal studies across diverse, larger populations using various study designs and heterogeneous patient samples. This will help establish standard biomarkers usable in all IBD cases. These biomarkers can then be used for timely prognosis, accurate diagnosis, and can also inform personalized treatment strategies for patients.

Declarations

Acknowledgement

None.

Funding

None.

Conflict of interest

The authors have no conflict of interest related to this publication.

Authors’ contributions

Study concept and design (PSP, RK, VS, TP), creating tables and figures (RK, PSP, VS), drafting of the manuscript (PSP, RK, VS), critical revision of the manuscript for important intellectual content (TP), and study supervision (TP). All authors have made significant contributions to this study and have approved the final manuscript.

References

  1. McDowell C, Farooq U, Haseeb M. Inflammatory bowel disease.[updated 2022 Jun 27]. StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 PubMed/NCBI
  2. Vivan TK, Santos BM, dos Santos CHM. Quality of life of patients with inflammatory bowel disease. J Coloproctol 2017;37(04):279-284 View Article
  3. Ghosh S, Mitchell R. Impact of inflammatory bowel disease on quality of life: Results of the European Federation of Crohn’s and Ulcerative Colitis Associations (EFCCA) patient survey. J Crohns Colitis 2007;1(1):10-20 View Article PubMed/NCBI
  4. Fumery M, Yzet C, Chatelain D, Yzet T, Brazier F, LeMouel JP, et al. Colonic Strictures in Inflammatory Bowel Disease: Epidemiology, Complications, and Management. J Crohns Colitis 2021;15(10):1766-1773 View Article PubMed/NCBI
  5. Levine JS, Burakoff R. Extraintestinal manifestations of inflammatory bowel disease. Gastroenterol Hepatol (N Y) 2011;7(4):235-241 PubMed/NCBI
  6. Ham NS, Myung SJ. Endoscopic molecular imaging in inflammatory bowel disease. Intest Res 2021;19(1):33-44 View Article PubMed/NCBI
  7. Testoni SGG, Albertini Petroni G, Annunziata ML, Dell’Anna G, Puricelli M, Delogu C, et al. Artificial Intelligence in Inflammatory Bowel Disease Endoscopy. Diagnostics (Basel) 2025;15(7):905 View Article PubMed/NCBI
  8. Gubatan J, Levitte S, Patel A, Balabanis T, Wei MT, Sinha SR. Artificial intelligence applications in inflammatory bowel disease: Emerging technologies and future directions. World J Gastroenterol 2021;27(17):1920-1935 View Article PubMed/NCBI
  9. Nardone OM, Cannatelli R, Ghosh S, Iacucci M. New endoscopic tools in inflammatory bowel disease. United European Gastroenterol J 2022;10(10):1103-1112 View Article PubMed/NCBI
  10. Underhill DM, Braun J. Fungal microbiome in inflammatory bowel disease: a critical assessment. J Clin Invest 2022;132(5):e155786 View Article PubMed/NCBI
  11. Wang X, Peng J, Cai P, Xia Y, Yi C, Shang A, et al. The emerging role of the gut microbiota and its application in inflammatory bowel disease. Biomed Pharmacother 2024;179:117302 View Article PubMed/NCBI
  12. Lewis JD. The utility of biomarkers in the diagnosis and therapy of inflammatory bowel disease. Gastroenterology 2011;140(6):1817-1826.e2 View Article PubMed/NCBI
  13. Wang R, Li Z, Liu S, Zhang D. Global, regional and national burden of inflammatory bowel disease in 204 countries and territories from 1990 to 2019: a systematic analysis based on the Global Burden of Disease Study 2019. BMJ Open 2023;13(3):e065186 View Article PubMed/NCBI
  14. Li CJ, Wang YK, Zhang SM, Ren MD, He SX. Global burden of inflammatory bowel disease 1990-2019: A systematic examination of the disease burden and twenty-year forecast. World J Gastroenterol 2023;29(42):5751-5767 View Article PubMed/NCBI
  15. Zhou JL, Bao JC, Liao XY, Chen YJ, Wang LW, Fan YY, et al. Trends and projections of inflammatory bowel disease at the global, regional and national levels, 1990-2050: a bayesian age-period-cohort modeling study. BMC Public Health 2023;23(1):2507 View Article PubMed/NCBI
  16. Borowitz SM. The epidemiology of inflammatory bowel disease: Clues to pathogenesis?. Front Pediatr 2022;10:1103713 View Article PubMed/NCBI
  17. Gorospe J, Windsor J, Hracs L, Coward S, Buie M, Quan J, et al. Trends in Inflammatory Bowel Disease Incidence and Prevalence across Epidemiologic Stages: A Global Systematic Review with Meta-Analysis. Inflamm Bowel Dis 2024;30(Suppl 1):S00 View Article
  18. Pakdin M, Zarei L, Bagheri Lankarani K, Ghahramani S. The cost of illness analysis of inflammatory bowel disease. BMC Gastroenterol 2023;23(1):21 View Article PubMed/NCBI
  19. Holko P, Kawalec P, Sajak-Szczerba M, Avedano L, Mossakowska M. Out-of-pocket expenses of patients with inflammatory bowel disease: a comparison of patient-reported outcomes across 12 European countries. Eur J Health Econ 2023;24(7):1073-1083 View Article PubMed/NCBI
  20. Jarmakiewicz-Czaja S, Zielińska M, Sokal A, Filip R. Genetic and Epigenetic Etiology of Inflammatory Bowel Disease: An Update. Genes (Basel) 2022;13(12):2388 View Article PubMed/NCBI
  21. Bing X, Crusius JBA, Meuwissen SGM, Peña AS. Inflammatory bowel disease: definition, epidemiology, etiologic aspects, and immunogenetic studies. World J Gastroenterol 1998;4(5):446-458 View Article PubMed/NCBI
  22. Knights D, Silverberg MS, Weersma RK, Gevers D, Dijkstra G, Huang H, et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med 2014;6(12):107 View Article PubMed/NCBI
  23. Andoh A, Nishida A. Alteration of the Gut Microbiome in Inflammatory Bowel Disease. Digestion 2023;104(1):16-23 View Article PubMed/NCBI
  24. Zehrh I, Habiba U, Picco MR, Bashir SH, Rehman UA, Haider O, et al. Metagenomics and Machine Learning-Based Precision Medicine Approaches for Autoimmune Diseases. Preprints 2023
  25. Alam MT, Amos GCA, Murphy ARJ, Murch S, Wellington EMH, Arasaradnam RP. Microbial imbalance in inflammatory bowel disease patients at different taxonomic levels. Gut Pathog 2020;12:1 View Article PubMed/NCBI
  26. Qiu P, Ishimoto T, Fu L, Zhang J, Zhang Z, Liu Y. The Gut Microbiota in Inflammatory Bowel Disease. Front Cell Infect Microbiol 2022;12:733992 View Article PubMed/NCBI
  27. Enkhtur K, Brehm G, Boldgiv B, Pfeiffer M. Alpha and beta diversity patterns of macro-moths reveal a breakpoint along a latitudinal gradient in Mongolia. Sci Rep 2021;11(1):15018 View Article PubMed/NCBI
  28. Pisani A, Rausch P, Bang C, Ellul S, Tabone T, Marantidis Cordina C, et al. Dysbiosis in the Gut Microbiota in Patients with Inflammatory Bowel Disease during Remission. Microbiol Spectr 2022;10(3):e0061622 View Article PubMed/NCBI
  29. Sommer F, Anderson JM, Bharti R, Raes J, Rosenstiel P. The resilience of the intestinal microbiota influences health and disease. Nat Rev Microbiol 2017;15(10):630-638 View Article PubMed/NCBI
  30. Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 2012;13(9):R79 View Article PubMed/NCBI
  31. Cao Y, Shen J, Ran ZH. Association between Faecalibacterium prausnitzii Reduction and Inflammatory Bowel Disease: A Meta-Analysis and Systematic Review of the Literature. Gastroenterol Res Pract 2014;2014:872725 View Article PubMed/NCBI
  32. Henke MT, Kenny DJ, Cassilly CD, Vlamakis H, Xavier RJ, Clardy J. Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn’s disease, produces an inflammatory polysaccharide. Proc Natl Acad Sci U S A 2019;116(26):12672-12677 View Article PubMed/NCBI
  33. Yao S, Zhao Z, Wang W, Liu X. Bifidobacterium Longum: Protection against Inflammatory Bowel Disease. J Immunol Res 2021;2021:8030297 View Article PubMed/NCBI
  34. Kennedy JM, De Silva A, Walton GE, Gibson GR. A review on the use of prebiotics in ulcerative colitis. Trends Microbiol 2024;32(5):507-515 View Article PubMed/NCBI
  35. Bajaj A, Markandey M, Kedia S, Ahuja V. Gut bacteriome in inflammatory bowel disease: An update on recent advances. Indian J Gastroenterol 2024;43(1):103-111 View Article PubMed/NCBI
  36. Baldelli V, Scaldaferri F, Putignani L, Del Chierico F. The Role of Enterobacteriaceae in Gut Microbiota Dysbiosis in Inflammatory Bowel Diseases. Microorganisms 2021;9(4):697 View Article PubMed/NCBI
  37. Miquel S, Leclerc M, Martin R, Chain F, Lenoir M, Raguideau S, et al. Identification of metabolic signatures linked to anti-inflammatory effects of Faecalibacterium prausnitzii. mBio 2015;6(2):e00300-15 View Article PubMed/NCBI
  38. Zhou L, Zhang M, Wang Y, Dorfman RG, Liu H, Yu T, et al. Faecalibacterium prausnitzii Produces Butyrate to Maintain Th17/Treg Balance and to Ameliorate Colorectal Colitis by Inhibiting Histone Deacetylase 1. Inflamm Bowel Dis 2018;24(9):1926-1940 View Article PubMed/NCBI
  39. Najafi S, Sotoodehnejadnematalahi F, Amiri MM, Pourshafie MR, Rohani M. Decreased mucosal adhesion of Lactobacillus species in patients with inflammatory bowel disease. Caspian J Intern Med 2022;13(4):713-720 View Article PubMed/NCBI
  40. Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun 2018;9(1):3294 View Article PubMed/NCBI
  41. Guo W, Mao B, Tang X, Zhang Q, Zhao J, Zhang H, et al. Improvement of inflammatory bowel disease by lactic acid bacteria-derived metabolites: a review. Crit Rev Food Sci Nutr 2025;65(7):1261-1278 View Article PubMed/NCBI
  42. Wang W, Chen L, Zhou R, Wang X, Song L, Huang S, et al. Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. J Clin Microbiol 2014;52(2):398-406 View Article PubMed/NCBI
  43. Ehrlich AM, Pacheco AR, Henrick BM, Taft D, Xu G, Huda MN, et al. Indole-3-lactic acid associated with Bifidobacterium-dominated microbiota significantly decreases inflammation in intestinal epithelial cells. BMC Microbiol 2020;20(1):357 View Article PubMed/NCBI
  44. Santoru ML, Piras C, Murgia A, Palmas V, Camboni T, Liggi S, et al. Cross sectional evaluation of the gut-microbiome metabolome axis in an Italian cohort of IBD patients. Sci Rep 2017;7(1):9523 View Article PubMed/NCBI
  45. Jeong HY, Choi YS, Lee JK, Lee BJ, Kim WK, Kang H. Anti-Inflammatory Activity of Citric Acid-Treated Wheat Germ Extract in Lipopolysaccharide-Stimulated Macrophages. Nutrients 2017;9(7):730 View Article PubMed/NCBI
  46. Yue C, Yang X, Li J, Chen X, Zhao X, Chen Y, et al. Trimethylamine N-oxide prime NLRP3 inflammasome via inhibiting ATG16L1-induced autophagy in colonic epithelial cells. Biochem Biophys Res Commun 2017;490(2):541-551 View Article PubMed/NCBI
  47. Nomura K, Ishikawa D, Okahara K, Ito S, Haga K, Takahashi M, et al. Bacteroidetes Species Are Correlated with Disease Activity in Ulcerative Colitis. J Clin Med 2021;10(8):1749 View Article PubMed/NCBI
  48. Fang X, Monk JM, Mih N, Du B, Sastry AV, Kavvas E, et al. Escherichia coli B2 strains prevalent in inflammatory bowel disease patients have distinct metabolic capabilities that enable colonization of intestinal mucosa. BMC Syst Biol 2018;12(1):66 View Article PubMed/NCBI
  49. Kim JM, Oh YK, Kim YJ, Youn J, Ahn MJ. Escherichia coli up-regulates proinflammatory cytokine expression in granulocyte/macrophage lineages of CD34 stem cells via p50 homodimeric NF-kappaB. Clin Exp Immunol 2004;137(2):341-350 View Article PubMed/NCBI
  50. Duan H, Wang L, Huangfu M, Li H. The impact of microbiota-derived short-chain fatty acids on macrophage activities in disease: Mechanisms and therapeutic potentials. Biomed Pharmacother 2023;165:115276 View Article PubMed/NCBI
  51. Santana PT, Rosas SLB, Ribeiro BE, Marinho Y, de Souza HSP. Dysbiosis in Inflammatory Bowel Disease: Pathogenic Role and Potential Therapeutic Targets. Int J Mol Sci 2022;23(7):3464 View Article PubMed/NCBI
  52. Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, 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 Article PubMed/NCBI
  53. Ma J, Wang K, Wang J, Zeng Q, Liu K, Zheng S, et al. Microbial Disruptions in Inflammatory Bowel Disease: A Comparative Analysis. Int J Gen Med 2024;17:1355-1367 View Article PubMed/NCBI
  54. Huang C, Zhang W, Sun A, Zhang X, Guo J, Ji R, et al. Methane Ameliorates Lipopolysaccharide-Induced Acute Orchitis by Anti-inflammatory, Antioxidative, and Antiapoptotic Effects via Regulation of the PK2/PKR1 Pathway. Oxid Med Cell Longev 2020;2020:7075836 View Article PubMed/NCBI
  55. Ghavami SB, Rostami E, Sephay AA, Shahrokh S, Balaii H, Aghdaei HA, et al. Alterations of the human gut Methanobrevibacter smithii as a biomarker for inflammatory bowel diseases. Microb Pathog 2018;117:285-289 View Article PubMed/NCBI
  56. Houshyar Y, Massimino L, Lamparelli LA, Danese S, Ungaro F. Going Beyond Bacteria: Uncovering the Role of Archaeome and Mycobiome in Inflammatory Bowel Disease. Front Physiol 2021;12:783295 View Article PubMed/NCBI
  57. Kropp C, Le Corf K, Relizani K, Tambosco K, Martinez C, Chain F, et al. The Keystone commensal bacterium Christensenella minuta DSM 22607 displays anti-inflammatory properties both in vitro and in vivo. Sci Rep 2021;11(1):11494 View Article PubMed/NCBI
  58. Akhtar M, Chen Y, Ma Z, Zhang X, Shi D, Khan JA, et al. Gut microbiota-derived short chain fatty acids are potential mediators in gut inflammation. Anim Nutr 2022;8:350-360 View Article PubMed/NCBI
  59. Wenzel TJ, Gates EJ, Ranger AL, Klegeris A. Short-chain fatty acids (SCFAs) alone or in combination regulate select immune functions of microglia-like cells. Mol Cell Neurosci 2020;105:103493 View Article PubMed/NCBI
  60. Zang X, Xiao M, Yu L, Chen Y, Duan H, Zhang C, et al. Prevotella copri—a potential next-generation probiotic. Food Front 2024;5(4):1391-1409 View Article
  61. Guggeis MA, Harris DM, Welz L, Rosenstiel P, Aden K. Microbiota-derived metabolites in inflammatory bowel disease. Semin Immunopathol 2025;47(1):19 View Article PubMed/NCBI
  62. Kabeerdoss J, Sankaran V, Pugazhendhi S, Ramakrishna BS. Clostridium leptum group bacteria abundance and diversity in the fecal microbiota of patients with inflammatory bowel disease: a case-control study in India. BMC Gastroenterol 2013;13:20 View Article PubMed/NCBI
  63. Ota S, Sakuraba H. Uptake and advanced therapy of butyrate in inflammatory bowel disease. Immuno 2022;2(4):692-702 View Article
  64. Pandey H, Jain D, Tang DWT, Wong SH, Lal D. Gut microbiota in pathophysiology, diagnosis, and therapeutics of inflammatory bowel disease. Intest Res 2024;22(1):15-43 View Article PubMed/NCBI
  65. Guinan J, Wang S, Hazbun TR, Yadav H, Thangamani S. Antibiotic-induced decreases in the levels of microbial-derived short-chain fatty acids correlate with increased gastrointestinal colonization of Candida albicans. Sci Rep 2019;9(1):8872 View Article PubMed/NCBI
  66. Yang Q, Ouyang J, Pi D, Feng L, Yang J. Malassezia in Inflammatory Bowel Disease: Accomplice of Evoking Tumorigenesis. Front Immunol 2022;13:846469 View Article PubMed/NCBI
  67. Youn HY, Kim HJ, Kim H, Seo KH. A comparative evaluation of the kefir yeast Kluyveromyces marxianus A4 and sulfasalazine in ulcerative colitis: anti-inflammatory impact and gut microbiota modulation. Food Funct 2024;15(12):6717-6730 View Article PubMed/NCBI
  68. Cristofori F, Dargenio VN, Dargenio C, Miniello VL, Barone M, Francavilla R. Anti-Inflammatory and Immunomodulatory Effects of Probiotics in Gut Inflammation: A Door to the Body. Front Immunol 2021;12:578386 View Article PubMed/NCBI
  69. Caetano MAF, Castelucci P. Role of short chain fatty acids in gut health and possible therapeutic approaches in inflammatory bowel diseases. World J Clin Cases 2022;10(28):9985-10003 View Article PubMed/NCBI
  70. Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol 2019;16(10):605-616 View Article PubMed/NCBI
  71. Shin Y, Han S, Kwon J, Ju S, Choi TG, Kang I, et al. Roles of Short-Chain Fatty Acids in Inflammatory Bowel Disease. Nutrients 2023;15(20):4466 View Article PubMed/NCBI
  72. Ashique S, Mishra N, Garg A, Sibuh BZ, Taneja P, Rai G, et al. Recent updates on correlation between reactive oxygen species and synbiotics for effective management of ulcerative colitis. Front Nutr 2023;10:1126579 View Article PubMed/NCBI
  73. Lamas B, Richard ML, Leducq V, Pham HP, Michel ML, Da Costa G, et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med 2016;22(6):598-605 View Article PubMed/NCBI
  74. Lemons JMS, Conrad M, Tanes C, Chen J, Friedman ES, Roggiani M, et al. Enterobacteriaceae Growth Promotion by Intestinal Acylcarnitines, a Biomarker of Dysbiosis in Inflammatory Bowel Disease. Cell Mol Gastroenterol Hepatol 2024;17(1):131-148 View Article PubMed/NCBI
  75. Li C, Peng K, Xiao S, Long Y, Yu Q. The role of Lactobacillus in inflammatory bowel disease: from actualities to prospects. Cell Death Discov 2023;9(1):361 View Article PubMed/NCBI
  76. Miquel S, Martín R, Rossi O, Bermúdez-Humarán LG, Chatel JM, Sokol H, et al. Faecalibacterium prausnitzii and human intestinal health. Curr Opin Microbiol 2013;16(3):255-261 View Article PubMed/NCBI
  77. Kim S, Jung Y, Lee SB, Oh HS, Hong SN. Gut microbial signatures in clinically stable ulcerative colitis according to the mucosal state and associated symptoms. J Gastroenterol Hepatol 2024;39(2):319-327 View Article PubMed/NCBI
  78. Oliva S, Di Nardo G, Ferrari F, Mallardo S, Rossi P, Patrizi G, et al. Randomised clinical trial: the effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis. Aliment Pharmacol Ther 2012;35(3):327-334 View Article PubMed/NCBI
  79. Hegazy SK, El-Bedewy MM. Effect of probiotics on pro-inflammatory cytokines and NF-kappaB activation in ulcerative colitis. World J Gastroenterol 2010;16(33):4145-4151 View Article PubMed/NCBI
  80. Nanda Kumar NS, Balamurugan R, Jayakanthan K, Pulimood A, Pugazhendhi S, Ramakrishna BS. Probiotic administration alters the gut flora and attenuates colitis in mice administered dextran sodium sulfate. J Gastroenterol Hepatol 2008;23(12):1834-1839 View Article PubMed/NCBI
  81. Raygoza Garay JA, Turpin W, Lee SH, Smith MI, Goethel A, Griffiths AM, et al. Gut Microbiome Composition Is Associated With Future Onset of Crohn’s Disease in Healthy First-Degree Relatives. Gastroenterology 2023;165(3):670-681 View Article PubMed/NCBI
  82. Rodríguez-Díaz C, Martín-Reyes F, Taminiau B, Ho-Plágaro A, Camargo R, Fernandez-Garcia F, et al. The Metagenomic Composition and Effects of Fecal-Microbe-Derived Extracellular Vesicles on Intestinal Permeability Depend on the Patient’s Disease. Int J Mol Sci 2023;24(5):4971 View Article PubMed/NCBI
  83. Shen Q, Huang Z, Yao J, Jin Y. Extracellular vesicles-mediated interaction within intestinal microenvironment in inflammatory bowel disease. J Adv Res 2022;37:221-233 View Article PubMed/NCBI
  84. Beer KB, Wehman AM. Mechanisms and functions of extracellular vesicle release in vivo-What we can learn from flies and worms. Cell Adh Migr 2017;11(2):135-150 View Article PubMed/NCBI
  85. Kumar MA, Baba SK, Sadida HQ, Marzooqi SA, Jerobin J, Altemani FH, et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct Target Ther 2024;9(1):27 View Article PubMed/NCBI
  86. Mittal S, Gupta P, Chaluvally-Raghavan P, Pradeep S. Emerging Role of Extracellular Vesicles in Immune Regulation and Cancer Progression. Cancers (Basel) 2020;12(12):3563 View Article PubMed/NCBI
  87. Zhang Y, Liu F, Yuan Y, Jin C, Chang C, Zhu Y, et al. Inflammasome-Derived Exosomes Activate NF-κB Signaling in Macrophages. J Proteome Res 2017;16(1):170-178 View Article PubMed/NCBI
  88. Brown L, Wolf JM, Prados-Rosales R, Casadevall A. Through the wall: extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi. Nat Rev Microbiol 2015;13(10):620-630 View Article PubMed/NCBI
  89. Jin Y, Ma L, Zhang W, Yang W, Feng Q, Wang H. Extracellular signals regulate the biogenesis of extracellular vesicles. Biol Res 2022;55(1):35 View Article PubMed/NCBI
  90. Ginini L, Billan S, Fridman E, Gil Z. Insight into Extracellular Vesicle-Cell Communication: From Cell Recognition to Intracellular Fate. Cells 2022;11(9):1375 View Article PubMed/NCBI
  91. Berumen Sánchez G, Bunn KE, Pua HH, Rafat M. Extracellular vesicles: mediators of intercellular communication in tissue injury and disease. Cell Commun Signal 2021;19(1):104 View Article PubMed/NCBI
  92. Mori MA, Ludwig RG, Garcia-Martin R, Brandão BB, Kahn CR. Extracellular miRNAs: From Biomarkers to Mediators of Physiology and Disease. Cell Metab 2019;30(4):656-673 View Article PubMed/NCBI
  93. Masi L, Capobianco I, Magrì C, Marafini I, Petito V, Scaldaferri F. MicroRNAs as Innovative Biomarkers for Inflammatory Bowel Disease and Prediction of Colorectal Cancer. Int J Mol Sci 2022;23(14):7991 View Article PubMed/NCBI
  94. Leoni G, Neumann PA, Kamaly N, Quiros M, Nishio H, Jones HR, et al. Annexin A1-containing extracellular vesicles and polymeric nanoparticles promote epithelial wound repair. J Clin Invest 2015;125(3):1215-1227 View Article PubMed/NCBI
  95. Zheng X, Chen F, Zhang Q, Liu Y, You P, Sun S, et al. Salivary exosomal PSMA7: a promising biomarker of inflammatory bowel disease. Protein Cell 2017;8(9):686-695 View Article PubMed/NCBI
  96. Stockdale SR, Shkoporov AN, Khokhlova EV, Daly KM, McDonnell SA, O’ Regan O, et al. Interpersonal variability of the human gut virome confounds disease signal detection in IBD. Commun Biol 2023;6(1):221 View Article PubMed/NCBI
  97. Tun HM, Peng Y, Massimino L, Sin ZY, Parigi TL, Facoetti A, et al. Gut virome in inflammatory bowel disease and beyond. Gut 2024;73(2):350-360 View Article PubMed/NCBI
  98. Jansen D, Matthijnssens J. The Emerging Role of the Gut Virome in Health and Inflammatory Bowel Disease: Challenges, Covariates and a Viral Imbalance. Viruses 2023;15(1):173 View Article PubMed/NCBI
  99. Benler S, Yutin N, Antipov D, Rayko M, Shmakov S, Gussow AB, et al. Thousands of previously unknown phages discovered in whole-community human gut metagenomes. Microbiome 2021;9(1):78 View Article PubMed/NCBI
  100. Ungaro F, Massimino L, D’Alessio S, Danese S. The gut virome in inflammatory bowel disease pathogenesis: From metagenomics to novel therapeutic approaches. United European Gastroenterol J 2019;7(8):999-1007 View Article PubMed/NCBI
  101. Tian X, Li S, Wang C, Zhang Y, Feng X, Yan Q, et al. Gut virome-wide association analysis identifies cross-population viral signatures for inflammatory bowel disease. Microbiome 2024;12(1):130 View Article PubMed/NCBI
  102. Li X-L, Megdadi M, Quadri HS. Interaction between gut virome and microbiota on inflammatory bowel disease. World J Methodol 2025;15(3) View Article
  103. Hetta HF, Ramadan YN, Alharbi AA, Alsharef S, Alkindy TT, Alkhamali A, et al. Gut Microbiome as a Target of Intervention in Inflammatory Bowel Disease Pathogenesis and Therapy. Immuno 2024;4(4):400-425 View Article
  104. Liang G, Conrad MA, Kelsen JR, Kessler LR, Breton J, Albenberg LG, et al. Dynamics of the Stool Virome in Very Early-Onset Inflammatory Bowel Disease. J Crohns Colitis 2020;14(11):1600-1610 View Article PubMed/NCBI
  105. Li J, Chen D, Yu B, He J, Zheng P, Mao X, et al. Fungi in Gastrointestinal Tracts of Human and Mice: from Community to Functions. Microb Ecol 2018;75(4):821-829 View Article PubMed/NCBI
  106. Iliev ID, Leonardi I. Fungal dysbiosis: immunity and interactions at mucosal barriers. Nat Rev Immunol 2017;17(10):635-646 View Article PubMed/NCBI
  107. Balderramo DC, Romagnoli PA, Granlund AVB, Catalan-Serra I. Fecal Fungal Microbiota (Mycobiome) Study as a Potential Tool for Precision Medicine in Inflammatory Bowel Disease. Gut Liver 2023;17(4):505-515 View Article PubMed/NCBI
  108. Sokol H, Leducq V, Aschard H, Pham HP, Jegou S, Landman C, et al. Fungal microbiota dysbiosis in IBD. Gut 2017;66(6):1039-1048 View Article PubMed/NCBI
  109. Catalán-Serra I, Thorsvik S, Beisvag V, Bruland T, Underhill D, Sandvik AK, 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 2024;30(7):1164-1177 View Article PubMed/NCBI
  110. Chehoud C, Albenberg LG, Judge C, Hoffmann C, Grunberg S, Bittinger K, et al. Fungal Signature in the Gut Microbiota of Pediatric Patients With Inflammatory Bowel Disease. Inflamm Bowel Dis 2015;21(8):1948-1956 View Article PubMed/NCBI
  111. Qiu X, Ma J, Jiao C, Mao X, Zhao X, Lu M, et al. Alterations in the mucosa-associated fungal microbiota in patients with ulcerative colitis. Oncotarget 2017;8(64):107577-107588 View Article PubMed/NCBI
  112. Acar C, Celik SK, Ozdemirel HO, Tuncdemir BE, Alan S, Mergen H. Composition of the colon microbiota in the individuals with inflammatory bowel disease and colon cancer. Folia Microbiol (Praha) 2024;69(2):333-345 View Article PubMed/NCBI
  113. Yu M, Ding H, Gong S, Luo Y, Lin H, Mu Y, et al. Fungal dysbiosis facilitates inflammatory bowel disease by enhancing CD4+ T cell glutaminolysis. Front Cell Infect Microbiol 2023;13:1140757 View Article PubMed/NCBI
  114. Zheng J, Sun Q, Zhang J, Ng SC. The role of gut microbiome in inflammatory bowel disease diagnosis and prognosis. United European Gastroenterol J 2022;10(10):1091-1102 View Article PubMed/NCBI
  115. Pascal V, Pozuelo M, Borruel N, Casellas F, Campos D, Santiago A, et al. A microbial signature for Crohn’s disease. Gut 2017;66(5):813-822 View Article PubMed/NCBI
  116. Collij V, Klaassen MAY, Weersma RK, Vila AV. Gut microbiota in inflammatory bowel diseases: moving from basic science to clinical applications. Hum Genet 2021;140(5):703-708 View Article PubMed/NCBI
  117. Höyhtyä M, Korpela K, Saqib S, Junkkari S, Nissilä E, Nikkonen A, et al. Quantitative Fecal Microbiota Profiles Relate to Therapy Response During Induction With Tumor Necrosis Factor α Antagonist Infliximab in Pediatric Inflammatory Bowel Disease. Inflamm Bowel Dis 2023;29(1):116-124 View Article PubMed/NCBI
  118. Tebani A, Afonso C, Bekri S. Advances in metabolome information retrieval: turning chemistry into biology. Part I: analytical chemistry of the metabolome. J Inherit Metab Dis 2018;41(3):379-391 View Article PubMed/NCBI
  119. Aldars-García L, Gisbert JP, Chaparro M. Metabolomics Insights into Inflammatory Bowel Disease: A Comprehensive Review. Pharmaceuticals (Basel) 2021;14(11):1190 View Article PubMed/NCBI
  120. Gallagher K, Catesson A, Griffin JL, Holmes E, Williams HRT. Metabolomic Analysis in Inflammatory Bowel Disease: A Systematic Review. J Crohns Colitis 2021;15(5):813-826 View Article PubMed/NCBI
  121. Ahmed I, Greenwood R, Costello B, Ratcliffe N, Probert CS. Investigation of faecal volatile organic metabolites as novel diagnostic biomarkers in inflammatory bowel disease. Aliment Pharmacol Ther 2016;43(5):596-611 View Article PubMed/NCBI
  122. Lins Neto MÁF, Verdi GMX, Veras AO, Veras MO, Caetano LC, Ursulino JS. USE OF METABOLOMICS TO THE DIAGNOSIS OF INFLAMMATORY BOWEL DISEASE. Arq Gastroenterol 2020;57(3):311-315 View Article PubMed/NCBI
  123. Scoville EA, Allaman MM, Brown CT, Motley AK, Horst SN, Williams CS, et al. Alterations in Lipid, Amino Acid, and Energy Metabolism Distinguish Crohn’s Disease from Ulcerative Colitis and Control Subjects by Serum Metabolomic Profiling. Metabolomics 2018;14(1):17 View Article PubMed/NCBI
  124. Sultan S, El-Mowafy M, Elgaml A, Ahmed TAE, Hassan H, Mottawea W. Metabolic Influences of Gut Microbiota Dysbiosis on Inflammatory Bowel Disease. Front Physiol 2021;12:715506 View Article PubMed/NCBI
  125. Ning L, Zhou YL, Sun H, Zhang Y, Shen C, Wang Z, et al. Microbiome and metabolome features in inflammatory bowel disease via multi-omics integration analyses across cohorts. Nat Commun 2023;14(1):7135 View Article PubMed/NCBI
  126. Chen R, Zheng J, Li L, Li C, Chao K, Zeng Z, et al. Metabolomics facilitate the personalized management in inflammatory bowel disease. Therap Adv Gastroenterol 2021;14:17562848211064489 View Article PubMed/NCBI
  127. Emwas AH, Roy R, McKay RT, Tenori L, Saccenti E, Gowda GAN, et al. NMR Spectroscopy for Metabolomics Research. Metabolites 2019;9(7):123 View Article PubMed/NCBI
  128. Boye TL, Hammerhøj A, Nielsen OH, Wang Y. Metabolomics for enhanced clinical understanding of inflammatory bowel disease. Life Sci 2024;359:123238 View Article PubMed/NCBI
  129. Vich Vila A, Zhang J, Liu M, Faber KN, Weersma RK. Untargeted faecal metabolomics for the discovery of biomarkers and treatment targets for inflammatory bowel diseases. Gut 2024;73(11):1909-1920 View Article PubMed/NCBI
  130. Alghoul Z, Yang C, Merlin D. The Current Status of Molecular Biomarkers for Inflammatory Bowel Disease. Biomedicines 2022;10(7):1492 View Article PubMed/NCBI
  131. Tatsuki M, Hatori R, Nakazawa T, Ishige T, Hara T, Kagimoto S, et al. Serological cytokine signature in paediatric patients with inflammatory bowel disease impacts diagnosis. Sci Rep 2020;10(1):14638 View Article PubMed/NCBI
  132. Kessel C, Lavric M, Weinhage T, Brueckner M, de Roock S, Däbritz J, et al. Serum biomarkers confirming stable remission in inflammatory bowel disease. Sci Rep 2021;11(1):6690 View Article PubMed/NCBI
  133. Mao L, Kitani A, Strober W, Fuss IJ. The Role of NLRP3 and IL-1β in the Pathogenesis of Inflammatory Bowel Disease. Front Immunol 2018;9:2566 View Article PubMed/NCBI
  134. Bauer C, Duewell P, Mayer C, Lehr HA, Fitzgerald KA, Dauer M, et al. Colitis induced in mice with dextran sulfate sodium (DSS) is mediated by the NLRP3 inflammasome. Gut 2010;59(9):1192-1199 View Article PubMed/NCBI
  135. Iljazovic A, Roy U, Gálvez EJC, Lesker TR, Zhao B, Gronow A, et al. Perturbation of the gut microbiome by Prevotella spp. enhances host susceptibility to mucosal inflammation. Mucosal Immunol 2021;14(1):113-124 View Article PubMed/NCBI
  136. Xu M, Jiang Z, Wang C, Li N, Bo L, Zha Y, et al. Acetate attenuates inflammasome activation through GPR43-mediated Ca(2+)-dependent NLRP3 ubiquitination. Exp Mol Med 2019;51(7):1-13 View Article PubMed/NCBI
  137. Bai AP, Ouyang Q. Probiotics and inflammatory bowel diseases. Postgrad Med J 2006;82(968):376-382 View Article PubMed/NCBI
  138. Imaoka A, Shima T, Kato K, Mizuno S, Uehara T, Matsumoto S, et al. Anti-inflammatory activity of probiotic Bifidobacterium: enhancement of IL-10 production in peripheral blood mononuclear cells from ulcerative colitis patients and inhibition of IL-8 secretion in HT-29 cells. World J Gastroenterol 2008;14(16):2511-2516 View Article PubMed/NCBI
  139. Subramanian S, Rhodes JM, Hart CA, Tam B, Roberts CL, Smith SL, et al. Characterization of epithelial IL-8 response to inflammatory bowel disease mucosal E. coli and its inhibition by mesalamine. Inflamm Bowel Dis 2008;14(2):162-175 View Article PubMed/NCBI
  140. Park MS, Kim MJ, Ji GE. Assessment of lipopolysaccharide-binding activity of Bifidobacterium and its relationship with cell surface hydrophobicity, autoaggregation, and inhibition of interleukin-8 production. J Microbiol Biotechnol 2007;17(7):1120-1126 PubMed/NCBI
  141. Kim H, Venancio VP, Fang C, Dupont AW, Talcott ST, Mertens-Talcott SU. Mango (Mangifera indica L.) polyphenols reduce IL-8, GRO, and GM-SCF plasma levels and increase Lactobacillus species in a pilot study in patients with inflammatory bowel disease. Nutr Res 2020;75:85-94 View Article PubMed/NCBI
  142. Verstockt S, Verstockt B, Machiels K, Vancamelbeke M, Ferrante M, Cleynen I, et al. Oncostatin M Is a Biomarker of Diagnosis, Worse Disease Prognosis, and Therapeutic Nonresponse in Inflammatory Bowel Disease. Inflamm Bowel Dis 2021;27(10):1564-1575 View Article PubMed/NCBI
  143. Giachero F, Jenke A, Zilbauer M. Improving prediction of disease outcome for inflammatory bowel disease: progress through systems medicine. Expert Rev Clin Immunol 2021;17(8):871-881 View Article PubMed/NCBI
  144. Shen Z, Zhu C, Quan Y, Yang J, Yuan W, Yang Z, et al. Insights into Roseburia intestinalis which alleviates experimental colitis pathology by inducing anti-inflammatory responses. J Gastroenterol Hepatol 2018;33(10):1751-1760 View Article PubMed/NCBI
  145. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013;504(7480):451-455 View Article PubMed/NCBI
  146. Meisel M, Mayassi T, Fehlner-Peach H, Koval JC, O’Brien SL, Hinterleitner R, et al. Interleukin-15 promotes intestinal dysbiosis with butyrate deficiency associated with increased susceptibility to colitis. ISME J 2017;11(1):15-30 View Article PubMed/NCBI
  147. Li Q, Ma L, Shen S, Guo Y, Cao Q, Cai X, et al. Intestinal dysbacteriosis-induced IL-25 promotes development of HCC via alternative activation of macrophages in tumor microenvironment. J Exp Clin Cancer Res 2019;38(1):303 View Article PubMed/NCBI
  148. Li Y, Handley SA, Baldridge MT. The dark side of the gut: Virome-host interactions in intestinal homeostasis and disease. J Exp Med 2021;218(5):e20201044 View Article PubMed/NCBI
  149. Azimi T, Nasiri MJ, Chirani AS, Pouriran R, Dabiri H. The role of bacteria in the inflammatory bowel disease development: a narrative review. APMIS 2018;126(4):275-283 View Article PubMed/NCBI
  150. Lightfoot YL, Yang T, Sahay B, Zadeh M, Cheng SX, Wang GP, et al. Colonic immune suppression, barrier dysfunction, and dysbiosis by gastrointestinal bacillus anthracis Infection. PLoS One 2014;9(6):e100532 View Article PubMed/NCBI
  151. Wei J, Zhang C, Gao Y, Li Y, Zhang Q, Qi H, et al. Gut Epithelial-derived CXCL9 Maintains Gut Homeostasis Through Preventing Overgrown E. coli. J Crohns Colitis 2022;16(6):963-977 View Article PubMed/NCBI
  152. Mitsialis V, Wall S, Liu P, Ordovas-Montanes J, Parmet T, Vukovic M, et al. 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 View Article PubMed/NCBI
  153. Lopez-Siles M, Duncan SH, Garcia-Gil LJ, Martinez-Medina M. Faecalibacterium prausnitzii: from microbiology to diagnostics and prognostics. ISME J 2017;11(4):841-852 View Article PubMed/NCBI
  154. Soenksen LR, Ma Y, Zeng C, Boussioux L, Villalobos Carballo K, Na L, et al. Integrated multimodal artificial intelligence framework for healthcare applications. NPJ Digit Med 2022;5(1):149 View Article PubMed/NCBI
  155. Mittermaier M, Raza MM, Kvedar JC. Bias in AI-based models for medical applications: challenges and mitigation strategies. NPJ Digit Med 2023;6(1):113 View Article PubMed/NCBI
  156. Ahmad HA, East JE, Panaccione R, Travis S, Canavan JB, Usiskin K, et al. Artificial intelligence in inflammatory bowel disease: implications for clinical practice and future directions. Intest Res 2023;21(3):283-294 View Article PubMed/NCBI
  157. Witges K, Targownik LE, Haviva C, Walker JR, Graff LA, Sexton KA, et al. Living With Inflammatory Bowel Disease: Protocol for a Longitudinal Study of Factors Associated With Symptom Exacerbations. JMIR Res Protoc 2018;7(11):e11317 View Article PubMed/NCBI
  158. Liñares-Blanco J, Fernandez-Lozano C, Seoane JA, López-Campos G. Machine Learning Based Microbiome Signature to Predict Inflammatory Bowel Disease Subtypes. Front Microbiol 2022;13:872671 View Article PubMed/NCBI
  159. Kyriakos N, Papaefthymiou A, Giakoumis M, Iatropoulos G, Mantzaris G, Liatsos C. Informed consent in inflammatory bowel disease: a necessity in real-world clinical practice. Ann Gastroenterol 2021;34(4):466-475 View Article PubMed/NCBI

About this Article

Cite this article
Prabhu PS, Kalita R, Sharma V, Prakash T. Understanding Inflammatory Bowel Disease: An Integrative Framework of Microbiome, Metabolome, and Immunological Biomarkers. J Transl Gastroenterol. 2025;3(1):24-38. doi: 10.14218/JTG.2024.00030.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
September 10, 2024 November 26, 2024 February 20, 2025 March 25, 2025
DOI http://dx.doi.org/10.14218/JTG.2024.00030
  • Journal of Translational Gastroenterology
  • eISSN 2994-8754
Back to Top

Understanding Inflammatory Bowel Disease: An Integrative Framework of Microbiome, Metabolome, and Immunological Biomarkers

Prithvi S. Prabhu, Rija Kalita, Vanshika Sharma, Tulika Prakash
  • Reset Zoom
  • Download TIFF