Skip to main content
United European Gastroenterology Journal logoLink to United European Gastroenterology Journal
. 2022 Dec 3;10(10):1091–1102. doi: 10.1002/ueg2.12338

The role of gut microbiome in inflammatory bowel disease diagnosis and prognosis

Jiaying Zheng 1,2,3, Qianru Sun 1,2,3, Jingwan Zhang 1,2,3,, Siew C Ng 1,2,3,
PMCID: PMC9752296  PMID: 36461896

Abstract

Inflammatory bowel disease (IBD) is a chronic immune‐mediated intestinal disease consisting of ulcerative colitis and Crohn's disease. Inflammatory bowel disease is believed to be developed as a result of interactions between environmental, immune‐mediated and microbial factors in a genetically susceptible host. Recent advances in high‐throughput sequencing technologies have aided the identification of consistent alterations of the gut microbiome in patients with IBD. Preclinical and murine models have also shed light on the role of beneficial and pathogenic bacteria in IBD. These findings have stimulated interest in development of non‐invasive microbial and metabolite biomarkers for predicting disease risk, disease progression, recurrence after surgery and responses to therapeutics. This review briefly summarizes the current evidence on the role of gut microbiome in IBD pathogenesis and mainly discusses the latest literature on the utilization of potential microbial biomarkers in disease diagnosis and prognosis.

Keywords: biomarker, Crohn's disease, inflammatory bowel disease, microbiome, ulcerative colitis


Microbiome‐associated biomarkers in IBD diagnosis and prognosis. Fecal and mucosal microbiome, including bacteria, fungi, virus, are useful in IBD diagnosis, classification, disease activity, disease course, recurrence after surgery and responses to therapeutics. Bacteria derived metabolites, and serum and fecal microbe‐associated proteins are applied in IBD determination and classification.

graphic file with name UEG2-10-1091-g001.jpg

INTRODUCTION

Inflammatory bowel disease (IBD) is a chronic relapsing inflammatory disease which typically includes two subtypes, Crohn's disease (CD) and ulcerative colitis (UC). Over the past few decades, the incidence of IBD in the West including Europe and North America has stabilized whereas the incidence in newly industrialized countries has continued to climb at a rapid rate. 1 , 2 , 3 Although the etiology of IBD is not completely understood, it has been reported that disease pathogenesis is related to changes in host genetics, 4 , 5 mucosa immunity, 6 , 7 environmental factors 8 and the gut microbiome. 9 , 10 Epidemiological studies have shown that environmental exposures such as diet, cigarette smoking, hygiene status, antibiotic use, mode of birth and breastfeeding 11 may contribute to disease pathogenesis of IBD in part via alterations of the gut microbiota. 12 , 13 , 14 , 15 , 16 , 17 Recent advances in high‐throughput sequencing with more rapid and less costly microbial sequencing of biosamples have allowed more comprehensive delineation of microbial gene and pathway composition as well as integrative network analyses between different microbial communities. 18

Several studies have shown that patients with IBD have perturbed and dysregulated intestinal microbiota compared with healthy subjects. 19 , 20 , 21 Microbial biomarkers are emerging as promising non‐invasive tools to predict disease risk, disease activity, disease course, recurrence after surgery and responses to therapeutics. 11 This review briefly summarizes the current evidence on the role of gut microbiome in IBD pathogenesis (Figure 1) and mainly discusses the latest literature on the utilization of potential microbial biomarkers in disease diagnosis and prognosis (Figure 2).

FIGURE 1.

FIGURE 1

Microbial alterations in IBD. The gut microbiome in IBD patients is generally characterized by a decrease in bacterial diversity, decrease in abundance of Firmicutes and Bacteroidetes, and an increase in Proteobacteria. Alteared bacteria at genus and species level, fungi, virus are shown in the figure. IBD, inflammatory bowel disease

FIGURE 2.

FIGURE 2

Microbiome‐associated biomarkers in IBD diagnosis and prognosis. Fecal and mucosal microbiome, including bacteria, fungi, virus, are useful in IBD diagnosis, classification, disease activity, disease course, recurrence after surgery and responses to therapeutics. Bacteria derived metabolites, and serum and fecal microbe‐associated proteins are applied in IBD determination and classification. IBD, inflammatory bowel disease

THE ROLE OF GUT MICROBIOME IN IBD

Preclinical and clinical studies have shown an important role of gut dysbiosis in IBD pathogenesis. In animal studies, a germ‐free environment prevents development of inflammation in genetically susceptible mice, 22 whereas the transfer of proinflammatory microbiota from diseased mice into healthy mice induced inflammation. 23 Furthermore, colonization of mice with fecal microbiota from patient with IBD worsened colitis by altering the gut microbiota. 24 In human subjects, disease activity is most obvious in areas where bacterial populations are highest (the colon) and where there is stasis of feces (terminal ileum and rectum). In addition, fecal diversion improves inflammation but restoration of bowel continuity leads to disease recurrence. 25 , 26 Antibiotics have proven to be effective in some patients with CD and specific bacteria have been reported to drive or suppress intestinal inflammation. 27

To date, fecal samples are the most commonly used sample type to depict the gut microbiome as optimized methods to process mucosal microbiota has been more challenging. The gut microbiome of IBD patients is generally characterized by reduced diversity, decrease in abundance of Firmicutes and Bacteroidetes, and an increase in Proteobacteria. At the genus level, patients with IBD commonly lack beneficial bacteria, such as Roseburia, Faecalibacterium, Dorea, Blautia, Christensenellaceae, Collinsella, Ruminococcus and other butyrate‐producing bacteria. 28 Conversely, bacterial groups such as Enterobacteriaceae, Fusobacterium, Enterococcus, Megasphaera, Campylobacter and sulfate‐reducing Gammaproteobacteria and Deltaproteobacteria were shown to be expanded in stool and mucosa of patients with IBD. 9 , 29

Within the class Clostridia, several studies have reported a decrease in the Clostridium leptum groups, particularly Faecalibacterium prausnitzii. It has been reported that F. prausnitzii with anti‐inflammatory properties is depleted in both mucosal and fecal samples in patients with UC and CD. 30 , 31 Roseburia, a clade of Clostridia XIVa group, is an acetate utilizer and butyrate producer. Roseburia hominis was found to be negatively correlated with disease activity in UC patients. Roseburia intestinalis has been shown to produce butyrate and induce anti‐inflammatory responses to alleviate experimental colitis. 32 Within Verrucomicrobiota phylum, Akkermansia muciniphila, a mucus‐degrader that was found to colonize the gut could induce the production of homeostatic IgG and prevent pathogenic bacteria from multiplying. 33

Adherent‐invasive Escherichia coli and Shigella have been consistently reported to be increased in fecal and mucosal samples of IBD patients. 34 Ruminococcus gnavus and Ruminococcus torques, commonly known as colon‐associated mucolytic bacteria, were abundant in the gut of IBD patients. 35 , 36 Ruminococcus gnavus produced inflammatory polysaccharide and induced the secretion of inflammatory cytokines such as TNF‐α by dendritic cells. 37 Fusobacterium nucleatum was frequently found in the gut of IBD patients and their presence were associated with reduced overall fecal microbial diversity, and their abundance also correlated with disease activity. Studies have indicated that F. nucleatum can damage intestinal epithelium and trigger inflammation in the gut. 38 , 39 Bacteroides fragilis, especially the enterotoxigenic B. fragilis, were found to be of higher prevalence and abundance in fecal samples of patients with IBD than that in healthy controls. 40 , 41 Recently, a potential pathobiont, Proteus mirabilis is positively correlated with Crohn's disease activity score (CDAI) and associated with Crohn's disease pathogenesis by activating pro‐inflammatory pathways in germ‐free mice. 42 The prevalence and abundance of P. mirabilis were also higher in fecal and mucosal samples of patients with CD than that in controls. A recent study 43 , 44 found that Klebsiella pneumoniae (Kp) was enriched in the gut microbiota of IBD patients across geography. Isolated Kp strains induced severe intestinal inflammation and tissue damage, suggesting that the Kp strains may contribute to worsening IBD.

The composition and diversity of fungi and viruses in the gut microbiome are also altered in patients with IBD. 45 , 46 Candida albicans was reported to be enriched while Saccharomyces cerevisiae was depleted in the faeces of IBD patients and the mucosa of CD patients. 47 , 48 , 49 Malassezia restricta, 50 the common skin resident fungus, is significantly increased in the mucosal samples of patients with CD. Fecal samples analysis showed that the gut virome of patients with IBD was associated with an expansion of Caudovirales bacteriophages 51 and rectal samples in patients with UC had an increased abundance of Caudovirales bacteriophages with a reduction in bacterial diversity which correlated with intestinal inflammation. 52 In addition, the microbiome of patients with ileal CD showed an increase in fungi at the expense of bacteria, whereas patients with UC and those with CD without ileal involvement exhibited reduced fungal diversity. 47

ROLE OF GUT MICROBIOME IN IBD DIAGNOSIS

Recently, researchers have explored different approaches, such as machine learning (ML) methods to target specific microbiome signatures either the bacteria genes or species for disease classification. 53 , 54 To further support the impact of the gut microbiome in patients with IBD, recent studies have demonstrated the role of specific microbes in driving or suppressing inflammation, predicting response to therapy and determining the risk of recurrence after surgery. Herein we aimed to highlight the specific role of bacterial species and discuss the potential of utilizing these microbes as biomarkers for IBD diagnosis and prognosis (Table 1).

TABLE 1.

Gut microbial biomarkers for IBD diagnosis, classification and activity assessment

Study Population Sample size Platform Sample type Marker Purpose Performance
Lopez‐Siles et al. (2016) Spanish 45 CD, 25 UC, 31 controls qPCR Biopsy Faecalibacterium prausnitzii phylogroup I Diagnose CD AUC = 0.851
Diagnose UC AUC = 0.763
Guo et al. (2019) Chinese 176 IBD (95 CD, 81 UC), 65 IBS, 105 controls qPCR Stool
  • Fecal markers

  • Fusobacterium nucleatum (Fn)

  • F. prausnitzii (Fp)

Distinguish CD from controls Fn: AUC = 0.841
Fp: AUC = 0.811
Fn + Fp: AUC = 0.867
Distinguish CD from IBS Fn: AUC = 0.767
Fp: AUC = 0.658
Fn + Fp: AUC = 0.771
Pascal et al. (2017) European (Spain, Belgium, UK and Germany)
  • Discovery cohort: 34 HC, 33 UC, 34 CD

  • Validation cohort: 1247 HC, 339 CD, 158 UC, 202 IBS and 99 anorexia

16S sequencing Stool 8 genera: Faecalibacterium, an unknown Peptostreptococcaceae, Anaerostipes, Methanobrevibacter, an unknown Christensenellaceae, Collinsella and Fusobacterium, Escherichia Discriminate CD from UC, IBS, anorexia, and healthy control
  • Sensitivity: 80%

  • Specificity: 90%–95%

Chamorro et al. (2021) Chilean and Spanish
  • Chilean: 20 UC, 21 CD, 5 controls

  • Spanish: 13 CD; 7 controls

16S sequencing Biopsy 20 OPU (operational phylogenetic unit) Discriminate dysbiosis IBD from eubiosis IBD (similar to controls) AUC = 0.96–0.99
11 OPU Discriminate UC from CD AUC = 0.83
Manandhar et al. (2021) America 729 IBD, 700 non‐IBD 16S sequencing Stool 50 bacterial taxa Diagnose IBD AUC = 0.80
331 CD, 141 UC 117 bacterial taxa Discriminate UC from CD AUC >0.90
Clooney et al. (2021) Irish and Canadian 303 CD, 228 UC, 161 controls 16S sequencing Stool 715 species Discriminate CD from controls AUC = 0.88
732 species Discriminate UC from controls AUC = 0.88
Papa et al. (2012) America
  • Discovery cohort: 23 CD, 43 UC, 1 undefined IBD, 24 non‐IBD controls (pediatric patients)

  • Validation cohort: 25 CD, 30 UC, 13 controls (children and young adults)

16S sequencing Stool Selected features Diagnose IBD Discovery cohort: AUC = 0.83 (80.3% sensitivity and 69.7% specificity)
Validation cohort: AUC = 0.84 (92% sensitivity, 58.5% specificity)
Discriminate UC from CD AUC = 0.76
Assess the activity of IBD AUC = 0.72
Gevers et al. (2014) North America 447 pediatric CD, and 221 control subjects (RISK) 16S sequencing Biopsy and stool Microbial dysbiosis index (MD‐index) Diagnose CD Terminal ileum biopsies: AUC = 0.85
Rectum biopsies: AUC = 0.78
Stool: AUC = 0.66
Franzosa et al. (2019) USA (PRISM) 68 CD, 53 UC, 34 controls Metagenomics Stool Microbial species Diagnose IBD AUC = 0.90
AUC = 0.86
Netherlands 20 CD, 23 UC, 22 controls LC‐MS Metabolites AUC = 0.92
AUC = 0.89
Serrano‐Gomezal et al. (2021) America, Spanish, Belgian 65 CD, 38 UC, 27 controls (America); 34 CD, 33 UC, 67 controls (Spanish); 49 CD (Belgian) Metagenomics Stool 26 species Diagnose CD AUC = 0.938
9 species Diagnose UC AUC = 0.646
11 species Predict CD relapse AUC = 0.769
Zuo et al. (2022) Caucasian 7 pediatric UC, 8 controls 16S sequencing Stool Genus Diagnose UC AUC = 0.869
Metagenomics Species AUC = 0.763
Metagenomics Pathway AUC = 0.764
Kolho et al. (2015) Finland 68 pediatric IBD, 26 controls Phylogenetic microarray Stool 9 bacterial groups Assess the activity of IBD AUC = 0.85
Tedjo et al. (2016) Netherlands 71 CD (97 active and 97 remission samples) 16S sequencing Stool 50 OTUs Assess the activity of CD AUC = 0.82
El Mouzan et al. (2018) Saudi Arabia 15 pediatric CD, 20 controls ITS sequencing Stool Selected features Diagnose CD AUC = 0.85
Biopsy AUC = 0.71
Sarrabayrouse et al. (2021) Spanish and Belgian 65 IBD, 28 controls qPCR Stool Microbial load data + demographic and standard laboratory data Diagnose IBD AUC = 0.842

Abbreviations: AUC, area under the curve; CD, Crohn's disease; HC, healthy control; IBD, inflammatory bowel disease; IBS, irritable bowel syndrome; ITS, internal transcribed spacer; LC‐MS, liquid chromatograph mass spectrometer; OPU, operational phylogenetic unit; qPCR, quantitative polymerase chain reaction; UC, ulcerative colitis.

Disease diagnosis

Specific microbial signatures can be used to diagnose IBD especially if they were consistently present in higher levels in cases than in controls, and they may also have a role in differentiating CD from UC. Faecalibacterium prausnitzii is commonly found to be depleted in patients with IBD. Lopez‐Siles et al. measured the abundance of F. prausnitzii and its two Phylogroups (I and II) in the intestinal mucosa of patients with IBD and controls using quantitative polymerase chain reaction (qPCR) assay. They found that F. prausnitzii phylogroup I had the best performance in discriminating healthy subjects from subjects with CD (area under the curve [AUC] = 0.851) and UC (AUC = 0.763). 55 In a Chinese cohort, the presence of F. prausnitzii and F. nucleatum in stool samples based on qPCR assay showed a good performance with an AUC of 0.841 and 0.811, respectively. 56 In a study of 2045 non‐IBD and IBD patients from four European countries, an algorithm based on eight selected genera from stool samples (Faecalibacterium, an unknown Peptostreptococcaceae, Anaerostipes, Methanobrevibacter, an unknown Christensenellaceae, Collinsella and Fusobacterium, Escherichia) performed well in discriminating CD from UC, irritable bowel syndrome, anorexia, and healthy control. 57 By assessing 20 bacteria markers in mucosal samples from Chilean and Spanish patients with IBD, Chamorro et al. 58 were able to discriminate dysbiosis and eubiosis in IBD patients, with an AUC ranging from 0.96 to 0.99.

Others have selected more bacterial markers as biomarkers for IBD determination using various feature selection methods and ML models. 59 , 60 , 61 Manandhar et al. 53 selected 50 fecal bacterial taxa for disease diagnosis in a large American cohort. By using five different supervised ML algorithms, their classifiers attained an AUC of around 0.80. A recent Irish and Canadian study 62 identified a large number of species from subjects' stool samples for differentiating IBD subtype and controls, achieving an AUC of 0.88 for separating UC and CD from controls. Among them, Eubacteria rectale and Clostridium cluster XIVa, which were both decreased in CD and UC patients, were the most contributing operational taxonomic units (OTUs) in disease diagnostic models. In another large pediatric cohort, Gevers et al. 63 calculated the microbial dysbiosis index (MD‐index) for early diagnosis of CD using biopsies and stool samples, showing the best performance in terminal ileum biopsies and marginally worse results in rectum biopsies.

Metagenomic sequencing, which has a higher taxonomy resolution that enables better identification of specific bacterial species or strains related to disease development, is increasingly applied for the discovery of microbial markers. Franzosa et al. 64 trained a random forest classifier on selected fecal microbial species and found a consistent diagnostic accuracy with AUC of 0.90 in the discovery cohort from USA and AUC of 0.86 in a validation cohort from the Netherlands. Serrano‐Gomez et al. 65 used the 26 species and nine species for predicting CD with an AUC of 0.938 and predicting UC with an AUC = 0.646. Notably, Veillonella parvula, E. coli, R. gnavus and Clostridium clostridioforme were significantly enriched species in CD compared with UC and healthy controls, while F. prausnitzii is the most depleted species in CD. Interestingly, Zuo et al. 44 demonstrated that although the 16s data shows similar results as shotgun sequencing data in terms of alpha diversity and beta diversity, 16S genus data (AUC = 0.869) achieved higher pediatric UC prediction performance than shotgun species data (AUC = 0.763) and pathway data (AUC = 0.764).

Alterations in the gut mycobiome have also been explored for disease diagnosis and ecological processes. 66 EI Mouzan et al. 67 reported fungal dysbiosis in mucosa and stool of patients with CD and found that the performance of the classifier based on stool samples was significantly higher (AUC = 0.85) than that of mucosal samples (AUC = 0.71). Sarrabayrouse et al. 68 examined the role of fungal and bacterial loads in predicting IBD, IBD subtypes, and disease flare. They showed that combined with the demographic and standard laboratory data, and microbial load data from stool samples improved the performance of the random forest models for IBD diagnosis (AUC = 0.842). However, the potential role of fungal and viral dysbiosis in the diagnosis of IBD have not been studied extensively, due to the low abundance of fungal and viral DNA relative to bacterial DNA and the limited available genome references. But with the development of detection method and the improvement of fungal and viral database, gut mycobiome and virome will be completely captured and explored. The disease diagnostic performance will be further enhanced by combining the new‐found fungal and viral biomarkers with the existing bacterial biomarkers.

However, most of these studies were based on the sequencing results. Although the NGS technologies facilitate the microbiome analysis by providing the composition and relative abundance of microbes, they also have disadvantages, including high cost, complicated operations, sophisticated results interpretation, and low detection sensitivity. The use of qPCR assays for microbial markers to screen colorectal cancer provides a new direction for disease diagnosis and management. 69 qPCR detection is a cheap, easy‐to‐use, and multiplexed technique. Therefore, the development of qPCR‐based microbial marker detection methods or other affordable methods 70 to realize the diagnosis and prognosis of IBD has great potentials. This will simultaneously lead to the problem that the numbers of microbiome markers used for IBD diagnosis and prognosis should be balanced with the diagnosis and prediction performance. Selecting several most contributing biomarkers for detection would be more cost‐effective.

Besides, the above studies mostly based on the cohorts from Europe and North America, only few Asian cohorts were included. This may be attributed to the different incidence rate of IBD in difference region. Studies have revealed that the geographical, diet and lifestyle variations have a great impact on the human microbiome. In this regard, it is important to find out the similarities and differences in gut microbial variations between people of different races. Universal and region‐specific microbial markers should be developed to achieve a more accurate diagnosis performance.

Overall, the development of microbial biomarkers for disease prediction and diagnosis appeared promising and may complement current more invasive diagnostic modalities. However, further validation of each marker or a combination of markers needs to be performed in different populations and inter‐individual variations of microbial markers should be studied.

Disease classification

Although symptoms of CD and UC are relatively similar, their treatment, outcomes and need for surgery differ. Thus, it is necessary to differentiate the subtypes of IBD accurately. Studies have used fecal or mucosal bacteria signatures to distinguish UC from CD. Manandhar et al. 53 identified 117 differential bacterial taxa from stool samples for discriminating CD with UC which showed excellent performance (AUC >0.90). Using only 11 mucosal bacteria, Chamorro et al. 58 could differentiate UC from CD with an AUC of 0.83. These findings suggest the possibility of using a specific set of microbes for IBD subtype classification. However, a recent study that included Irish and Canadian patients with IBD showed a lower AUC value of 0.60–0.70 in differentiating CD from UC. Interestingly, the most important OTU in this model was F. prausnitzii. 62

Disease activity

Since IBD is a chronic disease with long‐term therapeutic strategies, a better non‐invasive tool for disease assessment is needed. Currently, fecal calprotectin is commonly used, but levels can be raised in any inflammatory conditions and may not be specific to IBD. 71 , 72 , 73 Effective and better monitoring of disease activity can help clinicians to assess the disease status, and tailor treatment more efficiently. Association between bacterial markers and disease severity scores have raised the possibility of using them as indicators of disease status. 74 , 75 Kolho et al. 76 performed a phylogenetic microarray with 9 bacterial groups to assess the activity of IBD, leading to an AUC of 0.85 when using 100 μg/g as the cutoff of fecal calprotectin levels. Tedjo et al. 75 also identified a discriminatory panel of fecal microbes to differentiate between active and inactive CD with an AUC of 0.82. These data highlight the potential of fecal microbial signatures in monitoring disease activity.

ROLE OF GUT MICROBIOME IN DISEASE PROGNOSIS AND RESPONSE TO THERAPY

Disease recurrence

IBD comes with natural courses following with periods of remission and relapse. Studies have showed that the gut microbiome dysbiosis may be involved in the disease activity of IBD. 77 A systematic review showed that C. leptum, F. prausnitzii and Bifidobacterium decreased in CD and UC patients with active disease status when compared to patients in remission. But lower abundance of Clostridium coccoides was only found in active UC patients but not CD patients. 78 Streptococcus levels were also found to be more abundant in samples of patients with postoperative CD recurrence. 79 The profile of mucosa‐related gut microbiota in the ileum of patients with CD exhibited significant alterations following surgery. Sokol et al. 80 reported that reduction in alpha diversity and an increase in the Proteobacteria phylum were linked to endoscopic recurrence of CD, with an accompanying decrease of members from Lachnospiraceae and the Ruminococcaceae families within the Firmicutes phylum. The alteration of gut microbiota at the time of surgery can predict endoscopic recurrence with the AUC of 0.81. The most contributing taxa in this model were Streptococcus, R. gnavus group and Gammaproteobacteria. Patients with CD who had recurrence after surgery had elevated Proteus genera and reduced Faecalibacterium in their mucosa, and recurrence was also associated with a history of smoking. 81 Moreover, in this study, a model comprising the Proteus genera, the abundance of Faecalibacterium, and smoking status has been developed to predict postoperative CD recurrence and showed moderate accuracy (AUC = 0.740). 81 Machiels et al. 82 showed there were a different mucosal microbiome after ileocecal resection between recurrence and non‐recurrence CD patients. Fusobacteria was thought to be the most prominent player driving early postoperative disease recurrence. Based on the mucosa‐associated genera Ralstonia, Haemophilus, Gemella, and Phasolarctobacterium at the time of resection, the AUC of predicting postoperative endoscopic recurrence was 0.739. And the AUC is 0.875 when using the fecal microbiome (Coprobacilus, unidentified Lachnospiraceae genus, and Dorea) at the time of resection. Serrano‐Gomez et al. 65 used 11 species based on stool metagenomic data for predicting CD relapse with the AUC of 0.769. R. torques, Fusicatenibacter saccharivorans, Clostridium bolteae were the significant differentially abundant species between CD remission and relapse.

Therapeutic response

The gut microbiota and specific bacteria taxonomic features have also been shown to influence drug response and outcome. 83 , 84 , 85 , 86 In a prospective study of serial fecal samplings of patients with CD who were initiating anti‐integrin inhibitors, Ananthakrishnan et al. 87 found that patients achieving remission had a higher α‐diversity and higher abundance of Roseburia inulinivorans and Burkholderiales species at baseline, as well as branched‐chain amino acid biosynthesis pathways compared to those who did not achieve remission, and the predictive ability using the microbial taxa (AUC = 0.715) performed better than utilizing clinical data alone (AUC = 0.619) in predicting remission.

Zhou et al. 77 showed that microbial taxonomy, mainly Clostridiales, have great high prediction ability in predicting response to infliximab treatment with 86.5% accuracy alone and with 93.8% accuracy when combined with calprotecin levels and CDAI. A multi‐omics study combining fecal metagenomic, serum metabolomic, and proteomic markers has identified several markers that predict differential response to IBD biologic therapy. 88 The performance of model using clinical and metagenomic features in classifying remission at week 14 among patients taking anti‐cytokine therapy was higher (AUC = 0.849) than using the clinical variables only (AUC = 0.624). The abundances of nine bacterial species at baseline were correlated to earlier remission, among which included Phascolarctobacteriaum faecium, Agathobaculum butyriciproducens, and Clostridium citroneae, and these bacteria have been previously reported to have anti‐inflammatory effects. 89 Following validation, six of the nine species markers were linked to anti‐cytokine response. Interestingly, a study analyzed the gut mycobiota and found that C. albicans was more abundant in non‐responders than responders to the anti‐TNF agent, Infliximab. 90 In a systematic review of 19 studies, increased baseline gut bacteria α‐diversity was observed in subjects with IBD who achieved response with exclusive enteral nutrition, Infliximab, Ustekinumab or Vedolizumab. Moreover, an increase in the abundance of F. prausnitzii was noted in subjects who responded to aminosalicylates, anti‐TNF medications and Ustekinumab. 91 Overall, these data support the importance of gut microbiome composition in determining treatment response in IBD.

ROLE OF MICROBIAL‐DERIVED METABOLITES IN IBD

The gut microbiota can produce a variety of bioactive metabolites that can be absorbed into the enterohepatic circulation and then into the host circulatory system. 92 These bacteria metabolites and derivatives affect host energy homeostasis, inflammation, endocrine regulation, and regulate host metabolism. Metagenomic analysis, plus targeted and untargeted metabolomics analysis have been utilized to investigate the function of microbiota‐derived metabolites and the importance and variation of metabolites from feces, urine, and serum between IBD patients and healthy controls. 93 , 94

Some bacterial‐associated metabolites, including short‐chain fatty acids, 64 medium‐chain fatty acids, 95 tryptophan, 96 , 97 bile acid 98 and sphingolipid, 99 have shown great potential as new biomarkers in IBD diagnosis and prediction. Marchesi et al. 100 first characterized the fecal extracts from CD and UC patients and found a decrease in butyrate, acetate, methylamine, and trimethylamine, as well as an increase in amino acids. Based on principal component analysis, they showed distinct clustering that separated CD patients from healthy controls, and UC patients from healthy controls, suggesting that metabolite profiling may help discriminate patients with IBD from healthy individuals. In an Italian cohort of IBD patients, biogenic amines, amino acids and lipids were significantly increased in their stool, while two B group vitamins were decreased compared to healthy subjects. 101 orthogonal partial least square‐discriminant analysis could separate both CD patients from healthy controls and UC patients from healthy controls. 101 In a study of 155 subjects from the USA cohort and 65 subjects from Netherlands, metabolomics analysis showed that eight fecal metabolites were significantly increased in CD patients compared with controls, the most prominent of which were sphingolipids, carboximidic acids and bile acids. Compared with the control group, the levels of seven metabolites were significantly increased in fecal samples of UC patients, while the level of phenylacetamides was increased, but the difference was not statistically significant. In addition, levels of metabolites such as chenodeoxycholate, C22:0‐sphingomyelin, 2‐hydroxymyristic acid, C54:6 TAG, lactate and pantothenate were significantly different between CD, UC and controls. The classifiers based on selected metabolites features achieved high accuracy (AUC = 0.92 in USA cohort, AUC = 0.89 in Netherlands cohort) in predicting IBD. 64

ROLE OF MICROBIOTA‐ASSOCIATED PROTEINS IN IBD

Meta‐proteomic studies, a vital complement of metagenomics, offer new possibilities for the characterization of proteins from host or microbes associated with IBD and understanding the functional roles and interactions of microbes in communities. 102 , 103 , 104 , 105 With the use of untargeted and targeted proteomics, 12 bacterial proteins and one human protein were found to be over‐represented or under‐represented in patients with CD compared with healthy subjects. 106 In another study, 107 seven serum proteins analyzed by enzyme‐linked immunosorbent assay (ELISA) could easily distinguish controls from patients with IBD with an AUC of 0.785. When additional serum biomarkers of the gut barrier, such as matrix metalloproteinase (MMP)‐9, MMP‐14, and tissue inhibitor of metalloproteinases 1 were included in the test set, the AUC increased to 0.904. MMP‐9 and MMP‐14 were also important contributors in the model for discriminating UC patients from controls, and CD patients from controls. In classifying CD and UC, the model using a set of selected proteins showed good performance with an AUC of 0.9. A recent study 108 also reported that stool proteins identified by an aptamer‐based screen and validated by ELISA could distinguish patients with UC from controls or patients with CD from controls. Some of these proteomic stool biomarkers showed a stronger correlation with the disease activity in patients with UC who were followed up longitudinally. Their performance in disease monitoring and prediction was also higher when compared with fecal calprotectin alone. However, studies that have identified protein markers were mainly based on a very small sample size, and subtle differences between the disease and healthy group have been ignored. Hence, large cohorts are required to explore useful, reproducible and reliable protein markers for IBD prediction and prognosis in the future.

CONCLUSION

In summary, over the past few decades, an increasing number of animal and human studies have shown consistent alterations in gut microbiome composition that contributes to IBD pathogenesis. Emerging data have focused not only on the gut bacteria taxa but also on other microbial communities including the fungi, viruses, microbial metabolites and microbe‐associated proteins. These data have shed light on the potential role of microbiome biomarkers in IBD diagnosis and prediction. Recent advances in sequencing technology and analytical platforms with validation across different disease phenotypes and populations will improve our understanding of perturbations of the microbiome‐metabolome interface in IBD, as well as drive further identification of potential diagnostic markers and therapeutic targets.

CONFLICT OF INTEREST

The authors have no conflicts of interest to declare.

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):1091–102. 10.1002/ueg2.12338

Contributor Information

Jingwan Zhang, Email: wendyjwzhang@cuhk.edu.hk.

Siew C. Ng, Email: siewchienng@cuhk.edu.hk.

DATA AVAILABILITY STATEMENT

No data was created in this review.

REFERENCES

  • 1. Roda G, Chien Ng S, Kotze PG, Argollo M, Panaccione R, Spinelli A, et al. Crohn’s disease. Nat Rev Dis Prim. 2020;6(1):22. 10.1038/s41572-020-0156-2 [DOI] [PubMed] [Google Scholar]
  • 2. Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat Rev Gastroenterol Hepatol. 2015;12(12):720–7. 10.1038/nrgastro.2015.150 [DOI] [PubMed] [Google Scholar]
  • 3. Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population‐based studies. Lancet. 2017;390(10114):2769–78. 10.1016/s0140-6736(17)32448-0 [DOI] [PubMed] [Google Scholar]
  • 4. Mirkov MU, Verstockt B, Cleynen I. Genetics of inflammatory bowel disease: beyond NOD2. Lancet Gastroenterol Hepatol. 2017;2(3):224–34. 10.1016/s2468-1253(16)30111-x [DOI] [PubMed] [Google Scholar]
  • 5. Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47(9):979–86. 10.1038/ng.3359 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Neurath MF. Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat Immunol. 2019;20(8):970–9. 10.1038/s41590-019-0415-0 [DOI] [PubMed] [Google Scholar]
  • 7. Chang JT. Pathophysiology of inflammatory bowel diseases. N Engl J Med. 2020;383(27):2652–64. 10.1056/nejmra2002697 [DOI] [PubMed] [Google Scholar]
  • 8. Wark G, Samocha‐Bonet D, Ghaly S, Danta M. The role of diet in the pathogenesis and management of inflammatory bowel disease: a review. Nutrients. 2020;13(1):135. 10.3390/nu13010135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Metwaly A, Reitmeier S, Haller D. Microbiome risk profiles as biomarkers for inflammatory and metabolic disorders. Nat Rev Gastroenterol Hepatol. 2022;19(6):383–97. 10.1038/s41575-022-00581-2 [DOI] [PubMed] [Google Scholar]
  • 10. Lee M, Chang EB. Inflammatory bowel diseases (IBD) and the microbiome‐searching the crime scene for clues. Gastroenterology. 2021;160(2):524–37. 10.1053/j.gastro.2020.09.056 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Guo X, Huang C, Xu J, Xu H, Liu L, Zhao H, et al. Gut microbiota is a potential biomarker in inflammatory bowel disease. Front Nutr. 2021;8:818902. 10.3389/fnut.2021.818902 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Mentella MC, Scaldaferri F, Pizzoferrato M, Gasbarrini A, Miggiano GAD. Nutrition, IBD and gut microbiota: a review. Nutrients. 2020;12(4):944. 10.3390/nu12040944 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Papoutsopoulou S, Satsangi J, Campbell BJ, Probert CS. Review article: impact of cigarette smoking on intestinal inflammation‐direct and indirect mechanisms. Aliment Pharmacol Ther. 2020;51(12):1268–85. 10.1111/apt.15774 [DOI] [PubMed] [Google Scholar]
  • 14. Panelli S, Epis S, Cococcioni L, Perini M, Paroni M, Bandi C, et al. Inflammatory bowel diseases, the hygiene hypothesis and the other side of the microbiota: parasites and fungi. Pharmacol Res. 2020;159:104962. 10.1016/j.phrs.2020.104962 [DOI] [PubMed] [Google Scholar]
  • 15. Ramirez J, Guarner F, Bustos Fernandez L, Maruy A, Sdepanian VL, Cohen H. Antibiotics as major disruptors of gut microbiota. Front Cell Infect Microbiol. 2020;10:572912. 10.3389/fcimb.2020.572912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Mitchell CM, Mazzoni C, Hogstrom L, Bryant A, Bergerat A, Cher A, et al. Delivery mode affects stability of early infant gut microbiota. Cell Rep Med. 2020;1(9):100156. 10.1016/j.xcrm.2020.100156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. van den Elsen LWJ, Garssen J, Burcelin R, Verhasselt V. Shaping the gut microbiota by breastfeeding: the gateway to allergy prevention? Front Pediatr. 2019;7:47. 10.3389/fped.2019.00047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Wei LQ, Cheong IH, Yang GH, Li XG, Kozlakidis Z, Ding L, et al. The application of high‐throughput technologies for the study of microbiome and cancer. Front Genet. 2021;12:699793. 10.3389/fgene.2021.699793 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Zheng L, Wen XL. Gut microbiota and inflammatory bowel disease: the current status and perspectives. World J Clin Cases. 2021;9(2):321–33. 10.12998/wjcc.v9.i2.321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Khan I, Ullah N, Zha L, Bai Y, Khan A, Zhao T, et al. Alteration of gut microbiota in inflammatory bowel disease (IBD): cause or consequence? IBD treatment targeting the gut microbiome. Pathogens. 2019;8(3):126. 10.3390/pathogens8030126 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Sankarasubramanian J, Ahmad R, Avuthu N, Singh AB, Guda C. Gut microbiota and metabolic specificity in ulcerative colitis and Crohn's disease. Front Med. 2020;7:606298. 10.3389/fmed.2020.606298 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Veltkamp C, Tonkonogy SL, de Jong YP, Albright C, Grenther WB, Balish E, et al. Continuous stimulation by normal luminal bacteria is essential for the development and perpetuation of colitis in Tgϵ26 mice. Gastroenterology. 2001;120(4):900–13. 10.1053/gast.2001.22547 [DOI] [PubMed] [Google Scholar]
  • 23. Ohkusa T, Okayasu I, Ogihara T, Morita K, Ogawa M, Sato N. Induction of experimental ulcerative colitis by Fusobacterium varium isolated from colonic mucosa of patients with ulcerative colitis. Gut. 2003;52(1):79–83. 10.1136/gut.52.1.79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Britton GJ, Contijoch EJ, Mogno I, Vennaro OH, Llewellyn SR, Ng R, et al. Microbiotas from humans with inflammatory bowel disease alter the balance of gut Th17 and RORγt+ regulatory T cells and exacerbate colitis in mice. Immunity. 2019;50(1):212–24.e4. 10.1016/j.immuni.2018.12.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Feagan BG, Panaccione R, Sandborn WJ, D'Haens GR, Schreiber S, Rutgeerts PJ, et al. Effects of adalimumab therapy on incidence of hospitalization and surgery in Crohn's disease: results from the CHARM study. Gastroenterology. 2008;135(5):1493–9. 10.1053/j.gastro.2008.07.069 [DOI] [PubMed] [Google Scholar]
  • 26. Rutgeerts P, Peeters M, Hiele M, Vantrappen G, Pennincx F, Aerts R, et al. Effect of faecal stream diversion on recurrence of Crohn's disease in the neoterminal ileum. Lancet. 1991;338(8770):771–4. 10.1016/0140-6736(91)90663-a [DOI] [PubMed] [Google Scholar]
  • 27. Thia KT, Mahadevan U, Feagan BG, Wong C, Cockeram A, Bitton A, et al. Ciprofloxacin or metronidazole for the treatment of perianal fistulas in patients with Crohn's disease: a randomized, double‐blind, placebo‐controlled pilot study. Inflamm Bowel Dis. 2009;15(1):17–24. 10.1002/ibd.20608 [DOI] [PubMed] [Google Scholar]
  • 28. Manichanh C, Borruel N, Casellas F, Guarner F. The gut microbiota in IBD. Nat Rev Gastroenterol Hepatol. 2012;9(10):599–608. 10.1038/nrgastro.2012.152 [DOI] [PubMed] [Google Scholar]
  • 29. Schirmer M, Garner A, Vlamakis H, Xavier RJ. Microbial genes and pathways in inflammatory bowel disease. Nat Rev Microbiol. 2019;17(8):497–511. 10.1038/s41579-019-0213-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez‐Humarán LG, Gratadoux J‐J, et al. Faecalibacterium prausnitzii is an anti‐inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci USA. 2008;105(43):16731–6. 10.1073/pnas.0804812105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, et al. A decrease of the butyrate‐producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014;63(8):1275–83. 10.1136/gutjnl-2013-304833 [DOI] [PubMed] [Google Scholar]
  • 32. 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):1751–60. 10.1111/jgh.14144 [DOI] [PubMed] [Google Scholar]
  • 33. Neurath MF. Host‐microbiota interactions in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020;17(2):76–7. 10.1038/s41575-019-0248-1 [DOI] [PubMed] [Google Scholar]
  • 34. Palmela C, Chevarin C, Xu Z, Torres J, Sevrin G, Hirten R, et al. Adherent‐invasive Escherichia coli in inflammatory bowel disease. Gut. 2018;67(3):574–87. 10.1136/gutjnl-2017-314903 [DOI] [PubMed] [Google Scholar]
  • 35. Willing BP, Dicksved J, Halfvarson J, Andersson AF, Lucio M, Zheng Z, et al. A pyrosequencing study in Twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology. 2010;139(6):1844–54.e1. 10.1053/j.gastro.2010.08.049 [DOI] [PubMed] [Google Scholar]
  • 36. Joossens M, Huys G, Cnockaert M, De Preter V, Verbeke K, Rutgeerts P, et al. Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives. Gut. 2011;60(5):631–7. 10.1136/gut.2010.223263 [DOI] [PubMed] [Google Scholar]
  • 37. Henke Matthew T, Kenny Douglas J, Cassilly Chelsi D, Vlamakis H, Xavier Ramnik J, Clardy J. Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn’s disease, produces an inflammatory polysaccharide. Proc Natl Acad Sci USA. 2019;116(26):12672–7. 10.1073/pnas.1904099116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Huh J‐W, Roh T‐Y. Opportunistic detection of Fusobacterium nucleatum as a marker for the early gut microbial dysbiosis. BMC Microbiol. 2020;20(1):208. 10.1186/s12866-020-01887-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Liu H, Hong XL, Sun TT, Huang XW, Wang JL, Xiong H. Fusobacterium nucleatum exacerbates colitis by damaging epithelial barriers and inducing aberrant inflammation. J Dig Dis. 2020;21(7):385–98. 10.1111/1751-2980.12909 [DOI] [PubMed] [Google Scholar]
  • 40. Prindiville TP, Sheikh RA, Cohen SH, Tang YJ, Cantrell MC, Silva J Jr. Bacteroides fragilis enterotoxin gene sequences in patients with inflammatory bowel disease. Emerg Infect Dis. 2000;6(2):171–4. 10.3201/eid0602.000210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Zamani S, Hesam Shariati S, Zali MR, Asadzadeh Aghdaei H, Sarabi Asiabar A, Bokaie S, et al. Detection of enterotoxigenic Bacteroides fragilis in patients with ulcerative colitis. Gut Pathog. 2017;9(1):53. 10.1186/s13099-017-0202-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Zhang J, Hoedt EC, Liu Q, Berendsen E, Teh JJ, Hamilton A, et al. Elucidation of Proteus mirabilis as a key bacterium in Crohn's disease inflammation. Gastroenterology. 2021;160(1):317–30.e11. 10.1053/j.gastro.2020.09.036 [DOI] [PubMed] [Google Scholar]
  • 43. Federici S, Kredo‐Russo S, Valdes‐Mas R, Kviatcovsky D, Weinstock E, Matiuhin Y, et al. Targeted suppression of human IBD‐associated gut microbiota commensals by phage consortia for treatment of intestinal inflammation. Cell. 2022;185(16):2879–98.e24. 10.1016/j.cell.2022.07.003 [DOI] [PubMed] [Google Scholar]
  • 44. Zuo W, Wang B, Bai X, Luan Y, Fan Y, Michail S, et al. 16S rRNA and metagenomic shotgun sequencing data revealed consistent patterns of gut microbiome signature in pediatric ulcerative colitis. Sci Rep. 2022;12(1):6421. 10.1038/s41598-022-07995-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Underhill DM, Braun J. Fungal microbiome in inflammatory bowel disease: a critical assessment. J Clin Invest. 2022;132(5). 10.1172/jci155786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Guzzo GL, Andrews JM, Weyrich LS. The neglected gut microbiome: fungi, Protozoa, and bacteriophages in inflammatory bowel disease. Inflamm Bowel Dis. 2022;28(7):1112–22. 10.1093/ibd/izab343 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Sokol H, Leducq V, Aschard H, Pham H‐P, Jegou S, Landman C, et al. Fungal microbiota dysbiosis in IBD. Gut. 2017;66(6):1039–48. 10.1136/gutjnl-2015-310746 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Zhang L, Zhan H, Xu W, Yan S, Ng SC. The role of gut mycobiome in health and diseases. Therap Adv Gastroenterol. 2021;14:17562848211047130. 10.1177/17562848211047130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Lam S, Zuo T, Ho M, Chan FKL, Chan PKS, Ng SC. Review article: fungal alterations in inflammatory bowel diseases. Aliment Pharmacol Ther. 2019;50(11‐12):1159–71. 10.1111/apt.15523 [DOI] [PubMed] [Google Scholar]
  • 50. Limon JJ, Tang J, Li D, Wolf AJ, Michelsen KS, Funari V, et al. Malassezia is associated with Crohn's disease and exacerbates colitis in mouse models. Cell Host Microbe. 2019;25(3):377–88.e6. 10.1016/j.chom.2019.01.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Norman Jason M, Handley Scott A, Baldridge Megan T, Droit L, Liu Catherine Y, Keller Brian C, et al. Disease‐specific alterations in the enteric virome in inflammatory bowel disease. Cell. 2015;160(3):447–60. 10.1016/j.cell.2015.01.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Zuo T, Lu X‐J, Zhang Y, Cheung CP, Lam S, Zhang F, et al. Gut mucosal virome alterations in ulcerative colitis. Gut. 2019;68(7):1169–79. 10.1136/gutjnl-2018-318131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Manandhar I, Alimadadi A, Aryal S, Munroe PB, Joe B, Cheng X. Gut microbiome‐based supervised machine learning for clinical diagnosis of inflammatory bowel diseases. Am J Physiol Gastrointest Liver Physiol. 2021;320(3):G328–G37. 10.1152/ajpgi.00360.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Marcos‐Zambrano LJ, Karaduzovic‐Hadziabdic K, Loncar Turukalo T, Przymus P, Trajkovik V, Aasmets O, et al. Applications of machine learning in human microbiome studies: a review on feature selection, biomarker identification, disease prediction and treatment. Front Microbiol. 2021;12:634511. 10.3389/fmicb.2021.634511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Lopez‐Siles M, Martinez‐Medina M, Suris‐Valls R, Aldeguer X, Sabat‐Mir M, Duncan SH, et al. Changes in the abundance of Faecalibacterium prausnitzii phylogroups I and II in the intestinal mucosa of inflammatory bowel disease and patients with colorectal cancer. Inflamm Bowel Dis. 2016;22(1):28–41. 10.1097/mib.0000000000000590 [DOI] [PubMed] [Google Scholar]
  • 56. Guo S, Lu Y, Xu B, Wang W, Xu J, Zhang G. A simple fecal bacterial marker panel for the diagnosis of Crohn’s disease. Front Microbiol. 2019;10. 10.3389/fmicb.2019.01306 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. 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):813–22. 10.1136/gutjnl-2016-313235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Chamorro N, Montero DA, Gallardo P, Farfan M, Contreras M, De la Fuente M, et al. Landscapes and bacterial signatures of mucosa‐associated intestinal microbiota in Chilean and Spanish patients with inflammatory bowel disease. Microb Cell. 2021;8(9):223–38. 10.15698/mic2021.09.760 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Papa E, Docktor M, Smillie C, Weber S, Preheim SP, Gevers D, et al. Non‐invasive mapping of the gastrointestinal microbiota identifies children with inflammatory bowel disease. PLoS One. 2012;7(6):e39242. 10.1371/journal.pone.0039242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Linares‐Blanco J, Fernandez‐Lozano C, Seoane JA, Lopez‐Campos G. Machine learning based microbiome signature to predict inflammatory bowel disease subtypes. Front Microbiol. 2022;13:872671. 10.3389/fmicb.2022.872671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Kubinski R, Djamen‐Kepaou JY, Zhanabaev T, Hernandez‐Garcia A, Bauer S, Hildebrand F, et al. Benchmark of data processing methods and machine learning models for gut microbiome‐based diagnosis of inflammatory bowel disease. Front Genet. 2022;13:784397. 10.3389/fgene.2022.784397 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Clooney AG, Eckenberger J, Laserna‐Mendieta E, Sexton KA, Bernstein MT, Vagianos K, et al. Ranking microbiome variance in inflammatory bowel disease: a large longitudinal intercontinental study. Gut. 2021;70(3):499–510. 10.1136/gutjnl-2020-321106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Gevers D, Kugathasan S, Denson Lee A, Vázquez‐Baeza Y, Van Treuren W, Ren B, et al. The treatment‐naive microbiome in new‐onset Crohn’s disease. Cell Host Microbe. 2014;15(3):382–92. 10.1016/j.chom.2014.02.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Franzosa EA, Sirota‐Madi A, Avila‐Pacheco J, Fornelos N, Haiser HJ, Reinker S, et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat Microbiol. 2019;4(2):293–305. 10.1038/s41564-018-0306-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Serrano‐Gomez G, Mayorga L, Oyarzun I, Roca J, Borruel N, Casellas F, et al. Dysbiosis and relapse‐related microbiome in inflammatory bowel disease: a shotgun metagenomic approach. Comput Struct Biotechnol J. 2021;19:6481–9. 10.1016/j.csbj.2021.11.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Iliev ID. Mycobiota‐host immune interactions in IBD: coming out of the shadows. Nat Rev Gastroenterol Hepatol. 2022;19(2):91–2. 10.1038/s41575-021-00541-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. El Mouzan MI, Korolev KS, Al Mofarreh MA, Menon R, Winter HS, Al Sarkhy AA, et al. Fungal dysbiosis predicts the diagnosis of pediatric Crohn's disease. World J Gastroenterol. 2018;24(39):4510–6. 10.3748/wjg.v24.i39.4510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Sarrabayrouse G, Elias A, Yáñez F, Mayorga L, Varela E, Bartoli C, et al. Fungal and bacterial loads: noninvasive inflammatory bowel disease biomarkers for the clinical setting. mSystems. 2021;6(2):e01277‐20. 10.1128/msystems.01277-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Liang JQ, Li T, Nakatsu G, Chen YX, Yau TO, Chu E, et al. A novel faecal Lachnoclostridium marker for the non‐invasive diagnosis of colorectal adenoma and cancer. Gut. 2020;69(7):1248–57. 10.1136/gutjnl-2019-318532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Takahashi MK, Tan X, Dy AJ, Braff D, Akana RT, Furuta Y, et al. A low‐cost paper‐based synthetic biology platform for analyzing gut microbiota and host biomarkers. Nat Commun. 2018;9(1):3347. 10.1038/s41467-018-05864-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Khaki‐Khatibi F, Qujeq D, Kashifard M, Moein S, Maniati M, Vaghari‐Tabari M. Calprotectin in inflammatory bowel disease. Clin Chim Acta. 2020;510:556–65. 10.1016/j.cca.2020.08.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Rokkas T, Portincasa P, Koutroubakis IE. Fecal calprotectin in assessing inflammatory bowel disease endoscopic activity: a diagnostic accuracy meta‐analysis. J Gastrointestin Liver Dis. 2018;27(3):299–306. 10.15403/jgld.2014.1121.273.pti [DOI] [PubMed] [Google Scholar]
  • 73. van Rheenen PF, Van de Vijver E, Fidler V. Faecal calprotectin for screening of patients with suspected inflammatory bowel disease: diagnostic meta‐analysis. BMJ. 2010;341(jul15 1):c3369. 10.1136/bmj.c3369 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Barberio B, Facchin S, Patuzzi I, Ford AC, Massimi D, Valle G, et al. A specific microbiota signature is associated to various degrees of ulcerative colitis as assessed by a machine learning approach. Gut Microb. 2022;14(1):2028366. 10.1080/19490976.2022.2028366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Tedjo DI, Smolinska A, Savelkoul PH, Masclee AA, van Schooten FJ, Pierik MJ, et al. The fecal microbiota as a biomarker for disease activity in Crohn’s disease. Sci Rep. 2016;6(1):35216. 10.1038/srep35216 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Kolho KL, Korpela K, Jaakkola T, Pichai MV, Zoetendal EG, Salonen A, et al. Fecal microbiota in pediatric inflammatory bowel disease and its relation to inflammation. Am J Gastroenterol. 2015;110(6):921–30. 10.1038/ajg.2015.149 [DOI] [PubMed] [Google Scholar]
  • 77. Zhou Y, Xu ZZ, He Y, Yang Y, Liu L, Lin Q, et al. Gut microbiota offers universal biomarkers across ethnicity in inflammatory bowel disease diagnosis and infliximab response prediction. mSystems. 2018;3(1). 10.1128/msystems.00188-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Prosberg M, Bendtsen F, Vind I, Petersen AM, Gluud LL. The association between the gut microbiota and the inflammatory bowel disease activity: a systematic review and meta‐analysis. Scand J Gastroenterol. 2016;51(12):1407–15. 10.1080/00365521.2016.1216587 [DOI] [PubMed] [Google Scholar]
  • 79. Goel RM, Prosdocimi EM, Amar A, Omar Y, Escudier MP, Sanderson JD, et al. Streptococcus salivarius: a potential salivary biomarker for orofacial granulomatosis and Crohn's disease? Inflamm Bowel Dis. 2019;25(8):1367–74. 10.1093/ibd/izz022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Sokol H, Brot L, Stefanescu C, Auzolle C, Barnich N, Buisson A, et al. Prominence of ileal mucosa‐associated microbiota to predict postoperative endoscopic recurrence in Crohn's disease. Gut. 2020;69(3):462–72. 10.1136/gutjnl-2019-318719 [DOI] [PubMed] [Google Scholar]
  • 81. Wright EK, Kamm MA, Wagner J, Teo SM, Cruz P, Hamilton AL, et al. Microbial factors associated with postoperative Crohn's disease recurrence. J Crohns Colitis. 2017;11(2):191–203. 10.1093/ecco-jcc/jjw136 [DOI] [PubMed] [Google Scholar]
  • 82. Machiels K, Pozuelo del Río M, Martinez‐De la Torre A, Xie Z, Pascal Andreu V, Sabino J, et al. Early postoperative endoscopic recurrence in Crohn’s disease is characterised by distinct microbiota recolonisation. J Crohns Colitis. 2020;14(11):1535–46. 10.1093/ecco-jcc/jjaa081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Doherty MK, Ding T, Koumpouras C, Telesco SE, Monast C, Das A, et al. Fecal microbiota signatures are associated with response to Ustekinumab therapy among Crohn's disease patients. mBio. 2018;9(2). 10.1128/mbio.02120-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Crouwel F, Buiter HJC, de Boer NK. Gut microbiota‐driven drug metabolism in inflammatory bowel disease. J Crohns Colitis. 2020;15(2):307–15. 10.1093/ecco-jcc/jjaa143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Franzin M, Stefančič K, Lucafò M, Decorti G, Stocco G. Microbiota and drug response in inflammatory bowel disease. Pathogens. 2021;10(2):211. 10.3390/pathogens10020211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Caenepeel C, Sadat Seyed Tabib N, Vieira‐Silva S, Vermeire S. Review article: how the intestinal microbiota may reflect disease activity and influence therapeutic outcome in inflammatory bowel disease. Aliment Pharmacol Ther. 2020;52(9):1453–68. [DOI] [PubMed] [Google Scholar]
  • 87. Ananthakrishnan AN, Luo C, Yajnik V, Khalili H, Garber JJ, Stevens BW, et al. Gut microbiome function predicts response to anti‐integrin biologic therapy in inflammatory bowel diseases. Cell Host Microbe. 2017;21(5):603–10.e3. 10.1016/j.chom.2017.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Lee JWJ, Plichta D, Hogstrom L, Borren NZ, Lau H, Gregory SM, et al. Multi‐omics reveal microbial determinants impacting responses to biologic therapies in inflammatory bowel disease. Cell Host Microbe. 2021;29(8):1294–304.e4. 10.1016/j.chom.2021.06.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Watanabe Y, Nagai F, Morotomi M. Characterization of Phascolarctobacterium succinatutens sp. nov., an asaccharolytic, succinate‐utilizing bacterium isolated from human feces. Appl Environ Microbiol. 2012;78(2):511–8. 10.1128/aem.06035-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Ventin‐Holmberg R, Eberl A, Saqib S, Korpela K, Virtanen S, Sipponen T, et al. Bacterial and fungal profiles as markers of infliximab drug response in inflammatory bowel disease. J Crohns Colitis. 2021;15(6):1019–31. 10.1093/ecco-jcc/jjaa252 [DOI] [PubMed] [Google Scholar]
  • 91. Radhakrishnan ST, Alexander JL, Mullish BH, Gallagher KI, Powell N, Hicks LC, et al. Systematic review: the association between the gut microbiota and medical therapies in inflammatory bowel disease. Aliment Pharmacol Ther. 2022;55(1):26–48. 10.1111/apt.16656 [DOI] [PubMed] [Google Scholar]
  • 92. Wu J, Wang K, Wang X, Pang Y, Jiang C. The role of the gut microbiome and its metabolites in metabolic diseases. Protein Cell. 2021;12(5):360–73. 10.1007/s13238-020-00814-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Lavelle A, Sokol H. Gut microbiota‐derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020;17(4):223–37. 10.1038/s41575-019-0258-z [DOI] [PubMed] [Google Scholar]
  • 94. Lloyd‐Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila‐Pacheco J, Poon TW, et al. Multi‐omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569(7758):655–62. 10.1038/s41586-019-1237-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. De Preter V, Machiels K, Joossens M, Arijs I, Matthys C, Vermeire S, et al. Faecal metabolite profiling identifies medium‐chain fatty acids as discriminating compounds in IBD. Gut. 2015;64(3):447–58. 10.1136/gutjnl-2013-306423 [DOI] [PubMed] [Google Scholar]
  • 96. Agus A, Planchais J, Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe. 2018;23(6):716–24. 10.1016/j.chom.2018.05.003 [DOI] [PubMed] [Google Scholar]
  • 97. Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun. 2018;9(1):3294. 10.1038/s41467-018-05470-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Duboc H, Rajca S, Rainteau D, Benarous D, Maubert MA, Quervain E, et al. Connecting dysbiosis, bile‐acid dysmetabolism and gut inflammation in inflammatory bowel diseases. Gut. 2013;62(4):531–9. 10.1136/gutjnl-2012-302578 [DOI] [PubMed] [Google Scholar]
  • 99. Abdel Hadi L, Di Vito C, Riboni L. Fostering inflammatory bowel disease: sphingolipid strategies to join forces. Mediators Inflamm. 2016;2016:3827684–13. 10.1155/2016/3827684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Marchesi JR, Holmes E, Khan F, Kochhar S, Scanlan P, Shanahan F, et al. Rapid and noninvasive metabonomic characterization of inflammatory bowel disease. J Proteome Res. 2007;6(2):546–51. 10.1021/pr060470d [DOI] [PubMed] [Google Scholar]
  • 101. 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. 10.1038/s41598-017-10034-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Torres J, Petralia F, Sato T, Wang P, Telesco SE, Choung RS, et al. Serum biomarkers identify patients who will develop inflammatory bowel diseases up to 5 years before diagnosis. Gastroenterology. 2020;159(1):96–104. 10.1053/j.gastro.2020.03.007 [DOI] [PubMed] [Google Scholar]
  • 103. Bennike T, Birkelund S, Stensballe A, Andersen V. Biomarkers in inflammatory bowel diseases: current status and proteomics identification strategies. World J Gastroenterol. 2014;20(12):3231–44. 10.3748/wjg.v20.i12.3231 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Henry C, Bassignani A, Berland M, Langella O, Sokol H, Juste C. Modern metaproteomics: a unique tool to characterize the active microbiome in health and diseases, and pave the road towards new biomarkers‐example of Crohn's disease and ulcerative colitis flare‐ups. Cells. 2022;11(8):1340. 10.3390/cells11081340 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Chen C‐S, Sullivan S, Anderson T, Tan AC, Alex PJ, Brant SR, et al. Identification of novel serological biomarkers for inflammatory bowel disease using Escherichia coli proteome chip. Mol Cell Proteomics. 2009;8(8):1765–76. 10.1074/mcp.m800593-mcp200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Juste C, Kreil DP, Beauvallet C, Guillot A, Vaca S, Carapito C, et al. Bacterial protein signals are associated with Crohn's disease. Gut. 2014;63(10):1566–77. 10.1136/gutjnl-2012-303786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Coufal S, Galanova N, Bajer L, Gajdarova Z, Schierova D, Jiraskova Zakostelska Z, et al. Inflammatory bowel disease types differ in markers of inflammation, gut barrier and in specific anti‐bacterial response. Cells. 2019;8(7):719. 10.3390/cells8070719 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Soomro S, Venkateswaran S, Vanarsa K, Kharboutli M, Nidhi M, Susarla R, et al. Predicting disease course in ulcerative colitis using stool proteins identified through an aptamer‐based screen. Nat Commun. 2021;12(1):3989. 10.1038/s41467-021-24235-0 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No data was created in this review.


Articles from United European Gastroenterology Journal are provided here courtesy of Wiley

RESOURCES