You are on page 1of 15

Reviews

Microbiome risk profiles as


biomarkers for inflammatory
and metabolic disorders
Amira Metwaly 1
, Sandra Reitmeier2 and Dirk Haller 1,2 ✉

Abstract | The intestine harbours a complex array of microorganisms collectively known as the
gut microbiota. The past two decades have witnessed increasing interest in studying the gut
microbiota in health and disease, largely driven by rapid innovation in high-​throughput multi-
omics technologies. As a result, microbial dysbiosis has been linked to many human pathologies,
including type 2 diabetes mellitus and inflammatory bowel disease. Integrated analyses of multi-
omics data, including metagenomics and metabolomics along with measurements of host
response and cataloguing of bacterial isolates, have identified many bacteria and bacterial
products that are correlated with disease. Nevertheless, insight into the mechanisms through
which microbes affect intestinal health requires going beyond correlation to causation. Current
understanding of the contribution of the gut microbiota to disease causality remains limited,
largely owing to the heterogeneity of microbial community structures, interindividual differences
in disease evolution and incomplete understanding of the mechanisms that integrate microbiota-
derived signals into host signalling pathways. In this Review, we provide a broad insight into the
microbiome signatures linked to inflammatory and metabolic disorders, discuss outstanding
challenges in this field and propose applications of multi-​omics technologies that could lead
to an improved mechanistic understanding of microorganism–host interactions.

Microbiome The human digestive tract harbours a complex array of mellitus (T2DM) as exemplars of microbiota-​associated
All the microorganisms, their microorganisms, including bacteria, archaea, viruses disorders.
genomes and the surrounding and fungi. Trillions of bacteria colonize the gastroin- IBD and T2DM are both recognized to be multi-
host-​shaped environmental testinal tract in a spatially structured manner and their factorial diseases with a global pattern of rising disease
conditions of a given habitat.
The gut microbiome can be
combined genomes contain more than 200 times the incidence following industrialization4–6. Their aetiology
characterized through the number of genes in the human genome. Colonization involves a complex interplay of genetic susceptibil-
application of metagenomics, density exhibits a gradient from the proximal to the ity, environmental triggers and urban lifestyle-​related
metabolomics, metatranscrip- distal part of the gut, such that the highest numbers of factors. Against this shared background, metabolic dis-
tomics and metaproteomics
microorganisms are found in the colon1,2. In contrast to eases (such as T2DM) are additionally characterized by
combined with clinical or
environmental metadata.
the rapid transit of material through the small intestine chronic subclinical inflammation in the liver, adipose
(3–6 h), the comparatively reduced motility of the colon tissue, muscles, pancreas and gut, whereas inflamma-
(20–50 h) results in retention of luminal content, which tory gastrointestinal disorders, such as Crohn’s disease
builds a reservoir of biologically active metabolites. (CD) and ulcerative colitis (UC), are additionally asso-
In addition to microorganisms, the term microbiome ciated with inflammation-​driven metabolic alterations7.
encompasses the environment or niche inhabited by the Genome-​wide association studies have identified large
1
Chair of Nutrition and microbiota, which is shaped by the host. The incorpora- numbers of genetic variants associated with increased
Immunology, Technical tion of host-​related factors provides a broad view of the susceptibility to developing either T2DM (143 loci)8 or
University of Munich,
gut ecosystem, in which bidirectional microorganism– IBD (>240 loci)9. However, these variants collectively
Freising, Germany.
host interactions influence the physical and chemical explain only a small proportion of the heritability of
2
ZIEL Institute for Food &
Health, Technical University of
characteristics of the microbial environment or theatre these disorders: <10% for T2DM, <15% for UC and
Munich, Freising, Germany. of activity3. As the digestive tract and its microbiome <50% for CD10–15. This situation suggests the impor-
✉e-​mail: dirk.haller@tum.de are considered to lie at the intersection of immune tance of environmental triggers, and in particular the
https://doi.org/10.1038/ and metabolic processes, this Review focuses on gut microbiome, as major contributors to the aetiology
s41575-022-00581-2 inflammatory bowel disease (IBD) and type 2 diabetes of these diseases.

nATuRe RevIewS | GaSTROenTeROlOgy & HepaTOlOgy volume 19 | June 2022 | 383

0123456789();:
Reviews

Key points (FMT) have shown clinical efficacy in both T2DM25


and IBD26–30, including UC and, to a lesser extent,
• Several commonalities exist between inflammatory bowel disease (IBD) and type 2 CD. Clinical trials of FMT also provide evidence of a
diabetes mellitus (T2DM), two multifactorial diseases that show a pattern of causal relationship between the gut microbiota and
increasing global incidence following industrialization. other inflammatory, immune or metabolic disorders31.
• Altered gut bacterial composition and changes in host processing of bacteria-​derived For example, FMT is highly effective in treating
metabolites are implicated in IBD and T2DM and provide a shared underlying approximately 90% of patients with Clostridioides
pathogenetic mechanism.
difficile (formerly Clostridium difficile) infection 32
• A causal link between dysbiotic microbial communities and IBD or T2DM has been and has been assessed as a treatment for obesity33 and
established through gnotobiotic mouse experiments and integrative multi-​omics
graft-​versus-​host disease34. In four randomized clin-
studies.
ical trials, FMT induced clinical remission in 28% of
• Challenges in disease-​specific biomarker discovery include determining the causality
patients with UC28,35–37. Few clinical trials have exam-
of observed changes, understanding their functional redundancy in disease
mechanisms and the geographic and ethnic heterogeneity of gut microbiota.
ined the efficacy of FMT in patients with CD and the
results have been rather heterogeneous. In one study of
• Big data refinement, testing and validation of specific bacterial strains, their encoded
genes and metabolic byproducts are necessary to identify disease biomarkers.
174 patients with CD treated with FMT, clinical remis-
sion was achieved in 20% of patients and, overall, 43% of
patients achieved a clinical response38. A separate rand-
Despite great advances in disease risk profiling driven omized controlled trial found no effect of FMT on rates
by data from genome-​wide association studies and of clinical remission in patients with CD, but increased
multi-​omics approaches, the identification of susceptible engraftment of donor microbiota was associated with
individuals is still difficult and validated diagnostic or maintenance of remission27.
prognostic markers are lacking. However, multiple anal- Conversely (and despite a multitude of studies
yses of large population studies and cohorts of patients demonstrating that either dysbiosis specifically or par-
with IBD or T2DM have identified microbiome signatures ticular microbiota profiles are linked to metabolic dis-
linked to specific disease phenotypes16–19, the risk of orders), evidence of a benefit of FMT in patients with
relapse20 and response to treatment21. metabolic diseases is less well established. Landmark
In this Review, we summarize current knowledge studies demonstrated metabolic improvements as well
on the involvement of the gut microbiome in IBD and as beneficial changes in the intestinal microbiome in
T2DM. We critically assess the status of currently avail- patients with metabolic syndrome who received FMT
able disease-​associated microbiome signatures and dis- from lean, healthy donors29. However, these effects were
cuss the limitations that define their clinical use. Finally, inconsistent and transient, which could be explained by
we discuss how ‘big data’ generated by multi-​omics limited engraftment of the donor microbiota29 or by var-
approaches and analysed in an integrative framework iations in donor faecal microbial diversity at baseline25.
can be used to disentangle the complex pathology of Intriguingly, supplementation with low-​fermentable
these diseases. In particular, we focus on the mecha- fibre following oral FMT led to improved insulin sen-
nisms underlying interactions between bacterial strains sitivity, increased microbial diversity and richness, and
and gut-​derived metabolites that promote processes prolonged donor microbial engraftment in patients with
involved in inflammatory and metabolic diseases such both obesity and metabolic syndrome33. These data
as IBD and T2DM. emphasize the value of microbial modulation therapy
Microbiota in reversing metabolic dysfunction. In line with these
The microorganisms (bacteria, Common features of IBD and T2DM findings, FMT from metabolically compromised donors
fungi, archaea and viruses)
of a defined environment.
Both IBD and T2DM are associated with characteris- with obesity transiently worsened insulin sensitivity in
tic microbial alterations, notably reduced community recipients with metabolic syndrome, whereas FMT from
Microbiome signatures richness with the reduction of beneficial microbes and healthy donors after gastric bypass induced a minimal
Unique patterns of microbiome expansion of pathobionts22. The challenge in understand- increase in insulin sensitivity in recipients with meta-
configuration that can stratify
ing the role of microbial alterations in disease initiation bolic syndrome39. This study provides evidence for the
defined physiological and
pathological conditions and and progression lies in the timing of such changes (that transmissibility of donor metabolic profiles by FMT.
can be used to predict the risk is, whether they are a cause or consequence of the dis-
of disease development or ease), the functional redundancy of some changes Microbial dysbiosis in IBD. Several large cohort
progression. (meaning that they result in a similar signal or effect studies (Tables 1,2) have investigated gut microbiota
Pathobionts
on disease-​related mechanisms) and the fluctuations in alterations in patients with IBD by profiling their
Microorganisms that are linked these changes occurring during the disease course (such luminal and mucosal microbial communities. Overall,
to chronic inflammatory fluctuations can only be observed by longitudinal sam- active IBD is associated with an overabundance of
conditions but are harmless pling, which is rarely available). Despite the differences certain bacterial groups such as Enterobacteriaceae,
to the host under normal
in their pathology, IBD and T2DM share several mech- Fusobacterium, Ruminococcus gnavus, Streptococcus
conditions.
anistic features. The metabolic features of T2DM are anginosus, Enterococcus, Megasphaera, Campylobacter
Faecal microbiota accompanied by chronic low-​grade inflammation and and sulfate-​reducing Gammaproteobacteria and Delta­
transplantation disruption of the gut barrier, and recurrent inflamma- proteobacteria. Conversely, IBD is linked to the loss
(FMT). A treatment based on tory flares in patients with IBD coevolve with metabolic of beneficial taxa such as Faecalibacterium prausnitzii,
transfer of microorganisms
from the stool of a healthy
alterations at the cellular and systemic level23,24. Christensenellaceae, Collinsella, Roseburia, Ruminococcus
donor into a patient’s Therapy for these complex diseases remains challeng- and other butyrate-​producing bacteria17,20,21,40–44. Shotgun
digestive system. ing, but controlled trials of faecal microbiota transplantation metagenomics of stool samples has provided a more

384 | June 2022 | volume 19 www.nature.com/nrgastro

0123456789();:
Reviews

Table 1 | Gut microbiome signatures associated with IBD


Study population Methodology Risk signature Ref.
Increased Decreased
67 patients with IBD (34 CD, 33 UC); 16S rRNA gene Fusobacterium, Escherichia coli Faecalibacterium, 17

diagnoses confirmed by endoscopic sequencing of V4 (loss of beneficial microorganisms Peptostreptococcaceae,


and histological criteria, in remission region; multivariate more strongly associated with CD Anaerostipes, Methanobrevibacter,
≥3 months as defined by CAI for UC and analysis than gain of pathogenic ones) Christensenellaceae, Collinsella
CDAI for CD; 111 HCa
235 patients with IBD; diagnoses based 16S rRNA gene Ruminococcus gnavus (ileal Ruminococcus, Coprococcus, Blautia, 40

on clinical, radiological, endoscopic and sequencing of CD); Streptococcus anginosus Eubacterium, Dorea (IBD); Roseburia,
histological criteria; 38 HC V3–V5 region; (IBD); Aggregatibacter segnis, Faecalibacterium, Dorea, Blautia
multivariate Actinobacillusb (in IBD flare vs (IBD flare); Anaerostipes (in IBD,
analysis remission); disease-​specific fungal particularly IBD flare and ileal CD)
microbiota dysbiosis
85 patients with IBD (43 UC, 42 CD); DNA shotgun Streptococcus salivarium Relative abundance of Roseburia 21

disease activity assessed by HBI for sequencing; neural (in patients with UC not achieving inulinivorans vs Burkholderiales at
CD and SCCAI for UC network algorithms remission at week 14) baseline: Bifidobacterium longum,
Eggerthella, R. gnavus, R. inulinivorans,
Veillonella parvula (in patients with
CD achieving remission at week 14)
123 patients with IBD (72 CD, 51 UC); 16S rRNA gene Streptococcus, Proteobacteria, Roseburia, Coprococcus, Clostridiales 71

diagnoses based on standard sequencing of Enterococcaceae (CD);


endoscopic, radiographic and V4 region; random Bacteroidia, Pseudomonadaceae
histological criteria; 73 HC forest models (UC)
447 newly diagnosed (treatment-​naive) 16S rRNA gene Pasteurellaceae, Veillonellaceae, Bacteroides, Clostridiales, 43

paediatric patients with CD; 221 HC sequencing of V4 Neisseriaceae, Fusobacteriaceae Faecalibacterium, Roseburia, Blautia,
region; multivariate and E. coli Ruminococcus, Lachnospiraceae
analysis
29 patients with CD receiving 16S rRNA gene Sulfate-​reducing Akkermansia, Barnesiella, 20

HSCT; diagnoses based on standard sequencing of Gammaproteobacteria Oscillibacter, Roseburia, Odoribacter


endoscopic, radiographic and V3–V4 region; and Deltaproteobacteria,
histological criteria random forest butyrate-​producing Clostridiales,
models Enterococcus, Megasphaera,
Campylobacter, Fusobacterium
300 patients with UC and 213 with CD 16S rRNA gene Firmicutes, Proteobacteria Dorea 158

assessed by HBI for CD and SCCAI for sequencing of


UC; 30 HC V3 region
62 individuals (11 healthy Lithuanian 16S rRNA gene Lachnospiraceae, Actinobacteria, Proteobacteria 101

dizygotic twin pairs, 7 healthy Lithuanian sequencing of Ruminococcaceae


monozygotic twin pairs, 8 German V4–V5 region
monozygotic twin pairs discordant
for UC and 10 German unrelated
HC); disease activity assessed by CAI,
endoscopic appearance and continuity
of inflammation, histology, and proven
exclusive involvement of the colon
CAI, colitis activity index; CD, Crohn’s disease; CDAI, Crohn’s disease activity index; HBI, Harvey–Bradshaw Index; HC, healthy controls; HSCT, haematopoietic stem
cell transplantation; IBD, inflammatory bowel disease; SCCAI, Simple Clinical Colitis Activity Index; UC, ulcerative colitis. aWithout a history of chronic disease.
b
Two members of the Pasteurellaceae family.

comprehensive view of functional dysbiosis and the by a decrease in taxa belonging to Lachnospiraceae and
perturbations of metabolic pathways occurring in Ruminococcaceae and an increase in taxa belonging to
IBD18,43,45–47. These studies showed that metabolic path- Enterobacteriaceae, whereas a decrease in taxa belong-
ways involved in the synthesis of sulfur-​containing ing to Bacteroidaceae and an increase in taxa belonging
amino acids, riboflavin metabolism, glutathione trans- to Lachnospiraceae was observed for UC47. In concord-
porters, oxidative stress and nutrient transport were all ance with this heterogeneity, changes in only a few spe-
upregulated18,43,45–47. One study with the ability to resolve cies were shared across different IBD studies48, which
microbial communities down to the level of individ- suggests that interindividual differences persist within
ual strains within a species revealed increased strain groups of patients with CD despite similar disease
Dysbiosis diversity of pathobionts and reduced strain diversity phenotypes and disease courses.
(Also known as pathobiome). of beneficial microbes in stool samples from patients One of the first pieces of clinical evidence for a patho-
An altered microbial with IBD or irritable bowel syndrome (IBS) com- genetic role of the intestinal microbiota in IBD origi-
community composition that pared with healthy controls47. In-​depth analysis showed nated from experiments showing that diversion of the
affects the host immune
response and leads to the
that 219 taxa (including 152 species) were associated faecal stream from an inflamed segment of the small
emergence and outbreak with CD and 102 taxa (including 93 species) were asso- intestine in patients with CD led to improvements in
of pathogens. ciated with UC47. CD was predominantly characterized disease symptoms49,50. Restoration of the faecal stream

nATuRe RevIewS | GaSTROenTeROlOgy & HepaTOlOgy volume 19 | June 2022 | 385

0123456789();:
Reviews

Table 2 | Functional risk profiling using shotgun metagenomics signatures


Study population Functional risk signature Ref.
Increased Decreased
IBD
A subset of 4 patients with CD confirmed by Modules involved in glycolysis and carbohydrate Abundance of genes involved in lipid 45

endoscopic, pathological or radiographic transport and metabolism (nutrient uptake): metabolism and catabolism; global
findings and 7 HC subjected to shotgun metabolism of sulfur-​containing amino acids and decrease in nicotinamide, purine and
metagenomics to confirm 16S-​predicted cysteine (compounds advantageous for oxidative pyrimidine nucleotide biosynthesis in IBD
functional pathways generated from stress), riboflavin metabolism and glutathione
121 patients with CD, 75 with UC and 27 HC transporters increased in IBD
366 childrena with CD; disease activity Most predictive pathways: sulfur relay systems, Most predictive pathways: glycerophos- 46

assessed by PCDAI galactose metabolism, biosynthesis of siderophores, pholipid metabolism, aminobenzoate


glycolipid metabolism, glutamine and/or glutamate degradation, sulfur relay systems and
metabolism, glutathione metabolism and biosynthesis selenocompound metabolism
of siderophores
266 tissue samples obtained from Facultative anaerobe abundance (Streptococcus Blautia obeum, Alistipes putredinis (IBD) 77

32 people (20 IBD, 12 HC); disease salivarius, Streptococcus parasanguinis), Ruminococcus


activity assessed by CDAI or HBI for gnavus (IBD); upregulation of genes involved in
CD and SCCAI for UC oxidative stress (NADH oxidase and peroxiredoxin),
cysteine biosynthesis, acquisition of iron and 13 genes
involved in sugar utilization and transport
1,792 participants (33 IBD, 412 IBS, 1,025 219 taxa (152 species) associated with CD; 102 taxa Taxa belonging to Lachnospiraceae and 133

HC); diagnoses confirmed by endoscopic, (93 species) associated with UC; taxa belonging to Ruminococcaceae (CD); taxa belonging
pathological or radiographic findings Enterobacteriaceae (CD); perturbations in multiple to Bacteroidaceae (UC)
functional pathways, including pathways involved in
amino acid synthesis, vitamins, neurotransmitters and
short-​chain fatty acid synthesis (IBD); taxa belonging to
Lachnospiraceae (UC)
155 individuals (68 CD, 53 UC, 34 Unclassified Roseburia species (CD, UC); Bifidobacterium Roseburia hominis, Doreaformicigenerans 103

non-​IBD controls); diagnoses confirmed by breve, Clostridium symbiosum uniquely enriched (UC); and Ruminococcus obeum strongly
endoscopic, pathological or radiographic 12 species enriched, including R. gnavus, Escherichia coli enriched in controls without IBD
findings and Clostridium clostridioforme (CD)
Longitudinal sample of 1,638 stool samples Faecalibacterium prausnitzii and R. hominis; nicotinuric E. coli, Ruminococcus torques, R. gnavus, 18

from 132 patients with IBD; disease activity acid exclusively found (IBD) Clostridium hathewayi, Clostridium
assessed by HBI for CD or SCCAI for UC bolteae; pantothenate and nicotinate
(vitamins B5 and B3) depleted (IBD)
T2DM
46 patients with T2DM, 442 with 118 species with significantly altered metagenomics NR 136

prediabetes and 53 HC; assessed by in two-​component systems: fructose and mannose


OGTT and a Finnish diabetes risk score metabolism; pentose phosphate pathway;
(FINDRISC) branched-​chain amino acid synthesis and B group
vitamin (specifically vitamin B7) metabolism
53 patients with T2DM, 49 with impaired Four members of Lactobacillus; metagenomic gene Five members of Clostridium 62

glucose tolerance and 43 HC; assessed by cluster model identified Roseburia and F. prausnitzii as
fasting blood glucose and HbA1c highly discriminant for T2DM; 7 of the T2DM-​enriched
KEGG orthologue markers; starch and glucose
metabolism; fructose and mannose metabolism; ABC
transporters for amino acids; ions and simple sugars;
cysteine and methionine metabolism
1,179 patients from the LL-​DEEP study; GABA degradation and pathway PWY-5022 activity NR 159

assessed by OGTT
77 patients with T2DM, 80 individuals with The most discriminatory metagenomic linkage group for Higher levels of four antimicrobial 61

prediabetes and 97 HC; assessed by GABA separating treatment-​naive T2DM and normal glucose peptides in HC; IQGAP1 and
degradation and pathway PWY-5022 tolerance was Akkermansia muciniphila; F. prausnitzii unconventional MYO1C were uniquely
activity and E. coli were important in distinguishing prediabetes identified in the HC group; lower levels
from T2DM and HC; trimethylamine N-​oxide-​producing of proteases (trypsin and chymotrypsin
enzyme, dimethylaniline monooxygenase; higher and their precursors) and lipases
amylase 1 levels; antimicrobial cathepsin G
71 patients with T2DM and 74 HC Bacteroides caccae, C. hathewayi, Clostridium ramosum, Clostridiales sp. SS3/4, Eubacterium 160

Clostridium symbiosum, Eggerthella lenta, E. coli; rectale, F. prausnitzii, Roseburia intestinalis,


membrane sugar transport; branched-​chain amino Roseburia inulinivorans, Haemophilus
acid transport; methane metabolism; xenobiotic parainfluenzae; bacterial chemotaxis,
degradation and metabolism; sulfate reduction flagellar assembly, butyrate biosynthesis
and metabolism of cofactors and vitamins
ABC, ATP-​binding cassette; CD, Crohn’s disease; CDAI, CD activity index; GABA, γ-​aminobutyric acid; HbA1c, glycated haemoglobin; HBI, Harvey–Bradshaw index;
HC, healthy controls; IBD, inflammatory bowel disease; IBS, irritable bowel syndrome; IQGAP1, RAS GTPase-​activating-​like protein IQGAP1; KEGG, Kyoto
Encyclopedia of Genes and Genomes; MYO1C, myosin IC; NR, not reported; OGTT, oral glucose tolerance test; PCDAI; paediatric CD activity index; SCCAI, simple
clinical colitis activity index; T2DM, type 2 diabetes mellitus; UC, ulcerative colitis. aAged under 22 years.

386 | June 2022 | volume 19 www.nature.com/nrgastro

0123456789();:
Reviews

and postoperative exposure of the neo-​terminal ileum to amino acids compared with that of control individuals.
the gut luminal contents induced inflammation, which These findings suggest that disease-​specific changes in
suggested that the intestinal microbiota triggers postop- the gut microbiome can be detected in individuals with
erative recurrence of CD49,50. Furthermore, the efficacy prediabetes before they transition to T2DM. Differences
of antibiotic treatment in subsets of patients with active in bacterial species and metabolic gene cluster profiles
CD emphasizes the causal link between gut bacteria have been used to identify diabetes status in a set of indi-
and IBD51. viduals with either normal glucose tolerance or T2DM62.
Mechanistic studies in mouse models of acute However, confounding factors including geographic
and chronic intestinal inflammation provide further location, ethnicity, health status and medication history
evidence of a causal relationship between microbial led to inconsistency in identifying microbial alterations
dysbiosis and IBD52,53. For example, FMT from patients associated with T2DM60.
with IBD to germ-​free recipient mice was sufficient to Several studies have provided evidence of a causal
transmit the IBD phenotype20,54 and mice with a genetic link between specific members of the intestinal micro-
predisposition to IBD do not develop spontaneous biota and the pathogenesis of T2DM. For example,
inflammation when kept under germ-​free conditions55. A. muciniphila is among the taxa shown to have a pro-
Additionally, the transfer of dysbiotic microbial commu- tective effect in metabolic disorders in both human
nities from genetically susceptible mice after the onset of and mouse studies63–65. Interestingly, supplementation
IBD is capable of transferring this disease phenotype to with prebiotics normalized the abundance of A. mucin-
germ-​free recipient mice56. The transfer of IBD micro- iphila and improved metabolic health in humans65.
biota into germ-​free, wild-​type mice induced an imbal- Likewise, administration of A. muciniphila to mice
ance in the intestinal T cell response leading to increased fed a high-​fat diet reversed their increased fat mass,
numbers of intestinal T helper 17 (TH17) cells and TH2 metabolic endotoxaemia, adipose tissue inflammation
cells and decreased numbers of RORγt+ (retinoid-​related and insulin resistance66. Despite its high oxygen sensi-
orphan receptor-​γ short isoform) regulatory T (Treg) tivity and need for animal-​derived compounds in the
cells. The proportions of microbiota-​induced TH17 growth medium, A. muciniphila retains its protective
and RORγt+ Treg cells were predictive of human donor effects in mice when this organism is grown in a syn-
disease status in the Rag1−/− mouse model of colitis (in thetic medium compatible with human administration67,
which colitis develops after transfer of naive T cells) which opens avenues for its development as a treatment
when these mice were transplanted with human IBD for human obesity and associated disorders. Further,
microbiota57. Similarly, colonization of germ-​free mice the butyrate-​producing bacterium Anaerobutyricum
by Bacteroides fragilis, a human commensal bacterium soehngenii (previously designated Eubacterium hallii
of the intestinal microbiota, induced the conversion of strain L2-7) showed an increased abundance that cor-
CD4+ T cells into IL-10-​producing FOXP3+ Treg cells58, related with improved peripheral insulin sensitivity in
which suggests the existence of microbiota-​driven recipients of FMT from lean donors29. Administration
disease mechanisms in IBD. of A. soehngenii to patients with T2DM improved their
peripheral insulin sensitivity after 4 weeks of treat-
Microbial dysbiosis in T2DM. Widely variable changes ment, and these benefits were accompanied by an
in the abundance of several bacterial taxa have also altered microbiota composition and changes in bile
been described in T2DM (Tables 2,3). For instance, acid metabolism68.
an increased relative abundance of E. coli, Veillonella,
Blautia, Anaerostipes, Lactobacillus, Faecalibacterium Overlapping microbiome signatures. Curiously, spe-
and Clostridiales (amongst others) has been reported cific bacterial taxa show similar alterations in both
in patients with T2DM. Conversely, a reduced abun- IBD and T2DM, suggesting that common features of
dance of Bacteroides, Bifidobacterium, Parabacteroides, immune-​mediated and metabolic disease lead to simi-
Oscillospira and the mucin-​degrading gut bacterium lar adaptations of the microbiota. Examples include the
Akkermansia muciniphila is associated with improved decreased levels of Clostridium spp., Faecalibacterium,
metabolic health 19,59,60. A metagenomic and metap- Ruminococcus, Akkermansia, Collinsella and Roseburia,
roteomic study published in 2019 analysed the gut and the increased representation of Enterobacteriaceae,
microbiota composition and function in faecal sam- E. coli and Fusobacterium nucleatum spp., which
ples obtained from 254 Chinese individuals, including emphasize the challenges of defining disease-​specific
77 treatment-​naive patients with T2DM, 80 people with markers (Figs 1,2). As an example, the authors of a
prediabetes and 97 control individuals with normal glu- study of 2,045 patients with IBD identified a signature
cose tolerance61. Patients with T2DM and individuals of eight taxa, including unknown members of the fam-
with prediabetes had a lower abundance of bacterial spe- ily Christensenellaceae and the genus Fusobacterium,
cies belonging to Clostridiales and a higher abundance of that could discriminate between patients with CD and
Megasphaera elsdenii compared to metabolically healthy healthy individuals17. However, an increased abundance
controls. Functional differences were observed in the of Christensenellaceae is associated with both low BMI
microbiomes of patients with T2DM and individuals and weight loss69, a catabolic condition that is frequently
with prediabetes. The gut microbiota of individuals with observed in patients with IBD. Similarly, enrichment
prediabetes showed enrichment of pathways involved in of Fusobacterium is considered to be a marker of poor
sugar phosphotransferase systems, bacterial secretion prognosis in patients with metastatic colorectal cancer70.
systems and ATP-​binding cassette (ABC) transporters of Given that patients with IBD have an increased risk of

nATuRe RevIewS | GaSTROenTeROlOgy & HepaTOlOgy volume 19 | June 2022 | 387

0123456789();:
Reviews

developing colorectal cancer, this proposed microbiome environmental data, have enabled detailed charac-
signature could be an epiphenomenon with no causal terization of the functional capacity of the gut micro­
link to the underlying disease mechanisms. biome. Findings from the second phase of the Human
Additional meta-​omics approaches, including shot- Microbiome Project (HMP) immensely improved our
gun metagenomics and metabolomics, together with understanding of microorganism–metabolite interac-
the patient’s treatment history, demographics and tions in T2DM71 and IBD18. Integrative network analysis

Table 3 | Gut microbiome signatures associated with T2DM


Study population Methodology Risk signature Ref.
Increased Decreased
427 patients from the 16S rRNA gene sequencing of Escherichia coli, Veillonella, NR 93

CARE-​In-​DEEP Study, assessed V4 region; random forest Blautia, Anaerostipes


by OGTT models; gene expression analysis
145 women with normal, Shotgun metagenomics; random Lactobacillus gasseri, Desulfurispirillum indicum, Clostridium 62

impaired or diabetic glucose forest models Lactobacillus salivarius beijerinckii, Clostridium eklund,
control, assessed by HbA1c Clostridium botulinum, Pyramidobacter,
Clostridium thermocellum
800 patients with T2DM 16S rRNA gene sequencing Bacteroides thetaiotaomicron, Eubacterium rectale, Parabacteroides 90

of V3–V4 region; stochastic Alistipes putredinis distasonis, Roseburia inulinivorans,


gradient-​boosting regression Eubacterium eligens, Bacteroides vulgatus
60 individuals assessed by 16S rRNA gene sequencing of Bacteroidetes, Verrucomicrobia, Verrucomicrobia, Elusimicrobia, 95

fasting blood glucose levels V3–V4 region Proteobacteria, Elusimicrobia, Methanogenic archaeon
Acidobacteria, Deferribacteres,
Gemmatimonadetes,
Porphyromonadaceae,
Alistipes marseilloanorexic,
Bacillus sporothermodurans,
Staphylococcus, Prevotella
6,896 individuals with 16S rRNA gene sequencing Actinobacteria, Fusobacterium, Akkermansia, Synergistes, 89

metabolic syndromea of V4 region; multivariate Streptococcus, Lactobacillus Methanobrevibacter, Oscillospira,


analysis Roseburia, Bifidobacterium
60 individuals assessed by 16S rRNA gene sequencing Faecalibacterium, Clostridiales, Bifidobacterium, Parabacteroides, 94

fasting blood glucose levels of V3–V4 region; random forest Dorea, Clostridiaceae, Oscillospira, Bacteroides
models Lachnospiraceae
1,832 patients included in 16S rRNA gene sequencing of Lactobacillaceae Mogibacteriaceae, Clostridiaceae, 92

3 cohort studies, assessed V1–V2 region; gradient-boosting Butyrivibrio, Roseburia, Megamonas,


by fasting blood glucose or algorithm Dorea
HbA1c levels
784 patients from the Inter99 Shotgun metagenomics Lactobacillus, E. coli Roseburia, Subdoligranulum, 59

study, assessed by ADA HbA1c Intestinibacter


291 patients from the Africa 16S rRNA gene sequencing Peptostreptococcus, Eubacterium, Collinsella, Adlercreutzia, Anaerostipes, 161

America Diabetes Mellitus of V4 region; gene expression Prevotella, Desulfovibrio Epulopiscium, Clostridium butyricum,
study, assessed by ADA OGTT analysis Ruminococcus, Pediococcus
1,976 patients in the KORA 16S rRNA gene sequencing E. coli Faecalibacterium prausnitzii, 19

cohort assessed by WHO of V3–V4 region; random forest Bifidobacterium longum, Intestinales
OGTT models bartlettii, Coprococcus, Eubacterium
rectale
436 patients from the Popgen 16S rRNA gene sequencing B. thetaiotaomicron Clostridium sensu stricto, 91

cohort and 844 patients of V1–V2 region; multivariate E. coli, Romboutsia, Barnesiella,
from the FoCus cohort with analysis Pseudoflavonifractor, Veillonella,
HOMA-​IR >5.0 Roseburia
977 individuals with 16S rRNA gene sequencing NR Christensenellaceae 69

overweightb or obesityb of V4 region


from the TwinUK cohort
690 patients from the CDC 16S rRNA gene sequencing Gemellaceae, Streptococcus, Parabacteroides, Clostridiaceae, 162

cohort and NYU study, of of V4 region Blautia Lachnospiraceae, Ruminococcaceae,


whom 246 had overweight, Clostridiales, Oscillospira
142 had obesity and 211 had
a healthy weightb
ADA, American Diabetic Association; CDC, Centers for Disease Control and Prevention; HbA1c, glycated haemoglobin; HOMA-​IR, homeostatic model assessment
for insulin resistance; NR, not reported; NYU, New York University; OGTT, oral glucose tolerance test; T2DM, type 2 diabetes mellitus; WHO, World Health
Association. aDefined as waist >90 cm (men) or >85 cm (women), fasting blood glucose ≥6.1 mmol/l (110 mg/dl) or a diagnosis of diabetes mellitus, blood
triglyceride levels ≥1.7 mmol/l (150 mg/dl), high-​density lipoprotein <1.04 mmol/l (40 mg/dl) and blood pressure ≥130/85 mmHg or a previous diagnosis of
hypertension. bDefined as follows: healthy weight, BMI 18.5–24.9 kg/m2; overweight, BMI 25.0–29.9 kg/m2; obesity, BMI ≥30 kg/m2.

388 | June 2022 | volume 19 www.nature.com/nrgastro

0123456789();:
Reviews

1 2 3 4 5
Enriched Enriched Enriched Enriched Decreased Enriched Decreased
Odoribacter Escherichia coli Escherichia coli Ruminococcus Ruminococcus Clostridiales Faecalibacterium
Oscillibacter Veillonella Clostridium gnavus Blautia Enterococcus Peptostreptococcaceae
Pseudoflavonifractor Blautia Romboutsia Streptococcus Coprococcus Megasphaera Anaerostipes
Barnesiella Anaerostipes Barnesiella anginosus Eubacterium Campylobacter Akkermansia
Decreased Pseudoflavonifractor Aggregatibacter Dorea Fusobacterium Methanobrevibacter
Faecalibacterium Veillonella segnis Roseburia Escherichia coli Collinsella
Butyricimonas Roseburia Actinobacillus Faecalibacterium Christensenellaceae
Decreased Pasteurellaceae Anaerostipes Barnesiella
1 Bacteroides Oscillibacter
Enriched Faecalibacterium Roseburia
Pasteurellaceae Bifidobacterium Odoribacter
Veillonellaceae Eubacterium
Neisseriaceae Coprococcus
6
Fusobacteriaceae
Escherichia coli Decreased
Bifidobacterium longum Christensenellaceae
Eggerthella 7,8
Ruminococcus gnavus 6
Roseburia inulinivorans 3 7 8 7 8
4 Enriched Enriched
Veillonella parvula
Streptocccus salivarius 1 5 Firmicutes Lactobacillus
9 11 Proteobacteria Escherichia coli
Decreased 10
Bacteroides 2 Decreased Decreased
Clostridiales Dorea Sulfurospirillum
12 Clostridium
Faecalibacterium
Roseburia inulinivorans Pyramidobacter
Blautia Roseburia
Ruminococcus Subdoligranulum
Lachnospiraceae Intestinibacter
Burkholderiales
Ruminococcus 9 12
Eggerthella
Enriched 10 11 Enriched
Bifidobacterium longum
Peptostreptococcus Enriched Decreased Enriched Decreased Peptostreptococcus
Ruminococcus gnavus
Eubacterium Proteobacteria Verrucomicrobia Actinobacteria Akkermansia Eubacterium
Veillonella parvula
Prevotella Acidobacteria Elusimicrobia Fusobacterium Synergistetes Prevotella
Lactobacillus salivarius
Desulfovibrio Deferribacteres Eubacterium Streptococcus Methanobrevibacter Desulfovibrio
Decreased Bacillus Parabacteroides Lactobacillus Oscillospira Decreased
Collinsella Prevotella Roseburia Faecalibacterium Roseburia Collinsella
Adlercreutzia Bacteroides Bacteroides vulgatus Clostridiales Bifidobacterium Adlercreutzia
Anaerostipes Thetaiotaomicron Dorea Parabacteroides Anaerostipes
Epulopiscium Alistipes Lachnospiraceae Mogibacteriaceae Epulopiscium
IBD Clostridium Gemmatimonadetes Lactobacillaceae Megamonas Clostridium butyricum
Ruminococcus Porphyromonadaceae Butyrivibrio Ruminococcus
T2DM Pediococcus Staphylococcus Dorea Pediococcus

Fig. 1 | Bacterial risk signatures in the gut microbiomes of patients with IBD or T2DM. Box colours reflect enriched
and decreased taxa in inflammatory bowel disease (IBD, blue) and type 2 diabetes mellitus (T2DM, purple) in diverse
populations across the globe.

of microbiome, metabolome and transcriptome data pharmacologic responses to a therapeutic interven-


sets from 132 individuals identified disease-​associated tion”72. An ideal clinical biomarker should be rapid,
network hubs that connect key bacterial taxa (namely quantitative, objective, reproducible, non-​invasive and
F. prausnitzii, unclassified Subdoligranulum, Alistipes, exhibit high accuracy in predicting disease states across
E. coli and Roseburia) to certain metabolites (short-​chain multiple populations or ethnicities73. The identifica-
fatty acids, octanoyl carnitine and several lipids). tion of microbiome biomarkers and their use for the
Interestingly, differences between study participants classification of disease entities require extensive use
with and without IBD were more apparent in the faecal of computational and statistical tools to identify net-
metabolome than in the faecal metagenome, faecal meta- works of bacterial taxa that can accurately discriminate
transcriptome or faecal proteome18. In the Integrated between different disease phenotypes (such as healthy
Personal Omics Profiling Study (iPOP), plasma metab- individuals from patients with IBD, or individuals with
olites were strongly correlated with insulin resistance in prediabetes from patients with T2DM) as well as dis-
longitudinal samples from 106 individuals, which sug- tinguish closely related disease entities (such as IBD
gests that interactions between the host metabolome and and IBS). Profiles of microbial biomarkers also require
the gut microbiome are perturbed in insulin-​resistant validation in large, independent, population-​b ased
individuals. cohorts to confirm their diagnostic or prognostic utility.
In the following sections, we review the progress made
Biomarkers of gut dysbiosis towards the development of microbiome-​based bio-
A National Institutes of Health working group has markers for disease risk profiling. These biomarkers
defined a biomarker as “a characteristic that is objec- increase in complexity from single indicator bacterial
tively measured and evaluated as an indicator of nor- strains or taxa through dysbiotic microbial communities
mal biological processes, pathological processes, or to multi-​omics-​based biomarkers (Box 1).

nATuRe RevIewS | GaSTROenTeROlOgy & HepaTOlOgy volume 19 | June 2022 | 389

0123456789();:
Reviews

Bacillus sporothermodurans
Staphylococcus
Lactobacillus gasseri
Eubacterium
Prevotella Pseudomonadaceae
Alistipes marseiloanorexi Actinobacillus
Lactobacillus salivarius Aggregatibacter segnis
Alistipes putredinis Dorea
Desulfovibrionales Megasphaera
Bacteroides thetaiotaomicron Enterococcus
Blautia Streptococcus salivarius
Parabacteroides
Ruminococcus Streptococcus anginosus
Megamonas Akkermansia Peptostreptococcus
Pediococcus Methanobrevibacter Fusobacterium
Aminicella Collinsella Campylobacter
Intestinibacter Roseburia Neisseriaceae
Clostridium butyricum
Clostridium botulinum Escherichia coli Ruminococcus gnavus
Clostridium beijerinckii Eggerthella
Subdoligranulum Veillonella
Oscillospira Clostridiales
Clostridium thermocellum Anaerostipes Oscillibacter
Epulopiscium Faecalibacterium Coprococcus
Eubacterium eligens Butyrivibrio
Eubacterium rectale Bifidobacterium longum Barnesiella
Desulfurispirillum Burkholderiales
Synergistetes
Pyramidobacter
Adlercreutzia
Bacteroides vulgatus

IBD T2DM Increased in T2DM Decreased in T2DM Increased in IBD Decreased in IBD

Fig. 2 | Overlapping global risk profiles in IBD and T2DM. Bacterial genera signatures associated with inflammatory
bowel disease (IBD) or type 2 diabetes mellitus (T2DM) and common to both diseases based on the results of 16S rRNA
gene sequencing microbiome association studies summarized in Tables 1–3.

Indicator strains and taxa. Many studies have inves- genes involved in improved bacterial colonization.
tigated the utility of microbial alterations as disease These genes involve functions such as oxidative stress
biomarkers, particularly in patients with CD or UC. responses, bacterial adhesion, iron acquisition and
Initial efforts attempted to define single bacterial taxa host mucus utilization77. Similarly, different strains of
that could act as indicators of disease activity. For B. fragilis showed functional divergence that resulted in
instance, F. prausnitzii (a butyrate-​producing Firmicutes) different levels of IgA induction in IBD-​related mouse
is depleted in patients with CD41. A reduced abundance models78. These genetically distinct B. fragilis strains also
of this bacterium in ileal mucosa biopsy samples from showed differential colitogenic and immunomodulatory
patients with CD strongly correlated with an increased effects when inoculated into recipient mice78.
risk of endoscopic recurrence after ileal resection. In a study that aimed to define key dysbiotic taxa for
Conversely, an increased abundance of adherent inva- use in monitoring of disease activity in patients with
sive E. coli correlated with ileal CD74. Most of these early IBD, the absolute abundance ratios of F. prausnitzii and
studies used 16S rRNA amplicon sequences to achieve E. coli (also known as the F–E index) were computed
family-​level or genus-​level taxonomic classification; using quantitative PCR. Use of the F–E index improved
only rarely were species-​level associations identified. the discrimination of patients with UC and IBS from
However, as most bacterial species consist of many those with CD and helped to discriminate colorectal
individual strains that can exhibit considerable genomic cancer from other gut disorders. However, the F–E index
and proteomic variation, strain diversity is of great func- could not discriminate between IBD subtypes79,80, which
tional importance, particularly in determining patho- suggests that single-​taxa indicators have limited util-
genicity. For instance, subspecies of both R. gnavus and ity for classifying subtypes of a disease. Gut dysbiosis
E. coli have been associated with increased severity of indices have mostly included bacterial species only;
IBD75,76. Moreover, a particular subspecies of R. gnavus however, one study used the differential abundance
that shows increased abundance in faecal samples from of two fungal phyla, Basidiomycota and Ascomycota,
IBD patients has been shown to contain strain-​specific which robustly discriminated samples originating from

390 | June 2022 | volume 19 www.nature.com/nrgastro

0123456789();:
Reviews

healthy individuals from samples obtained from patients had a limited capacity to classify IBD disease subtypes,
with IBD of different disease phenotypes40. In addition, and also showed overlap between patients with IBD and
reduced fungal diversity and an increased abundance of healthy individuals43.
Candida taxa have been found in the gut microbiota A study from the second phase of the integra-
of paediatric patients with CD81. Interestingly, fungal and tive HMP (iHMP) used a dysbiosis score based on
bacterial loads differ markedly between healthy relatives Bray–Curtis dissimilarities to non-​IBD metagenomes
of patients with IBD and unrelated healthy controls as to identify samples from patients with IBD that had
well as between patients with different IBD subtypes. highly dysbiotic metagenomic microbial structures18.
These observations demonstrate that fungi as well In addition to the microbial response to inflammation,
as bacteria contribute to IBD-​related gut dysbiosis82. this dysbiosis score encompassed other host-​related
factors (such as transcriptomic regulation) and bio-
Dysbiosis scores. Several studies have evaluated the per- chemical interactions between serum metabolites and
formance of gut dysbiosis indices that describe increas- chemokines, which provided a comprehensive view of
ingly complex co-​o ccurrences of specific bacterial systemic changes and microorganism–host interactions
abundances in disease classification. For instance, in IBD18. Nevertheless, we emphasize that dysbiosis is
one study found that the microbiota of patients with a poorly defined condition and thus different dysbio-
IBD fluctuates more dramatically than that of healthy sis indices will differ according to the methodology
individuals83. The authors of this study identified upper used to derive them, the disease entity investigated,
and lower limits for the normal variation in the gut and their performance in different cohorts or groups of
microbiome of healthy individuals and termed the inter- individuals.
val between these limits the “healthy plane”83. The dis-
tance to the healthy plane varied over time in patients Large-​scale biomarker profiling. Several studies have
with IBD but did not necessarily correlate with disease used machine learning algorithms to validate complex
activity83. In a large cohort of paediatric patients with microbiome signatures in cross-​sectional and longitu-
early-​onset CD (the RISK study), a microbiome dys- dinal patient cohorts. For example, a study published
biosis index (defined as the log of the total abundance in 2017 used 16S rRNA microbiota profiling to analyse
of organisms that are increased in individuals with faecal samples from a large cohort of patients with IBD
IBD divided by the total abundance of organisms that and control individuals without IBD18. The researchers
are decreased in patients with IBD) strongly corre- used a sequence clustering algorithm to identify a CD-​
lated with disease severity and could be used to stratify specific microbial signature based on the abundance of
patients43. Nevertheless, this microbiome dysbiosis index eight bacterial taxa18. Additionally, another group showed
that characteristics of the gut microbiome composition
at baseline could predict the response of patients with
Box 1 | Gut microbiome biomarker discovery IBD to anti-​integrin therapy at 14 weeks after treatment
The following processes are ordered by increasing data complexity. initiation. A microbiome signature generated by a deep
neural network had an area under the receiver operating
Single taxa as indicator markers for disease
characteristic curve (AUC) of 0.87, compared to an AUC
Expansion of pathobionts (for example, Escherichia coli, Ruminococcus gnavus).
of 0.62 for a model based on clinical covariates21.
Reduction of beneficial microbes (for example, Faecalibacterium prausnitzii).
Our group has also assessed the utility of microbiome
Methodology:
signatures as biomarkers of IBD and T2DM. In our first
• 16S rRNA gene sequencing: genus level study, we examined disease activity and response to ther-
• Shotgun metagenomics: strain level apy in a unique cohort of 29 patients with CD who had
• Quantitative PCR for validation: co-​abundances of F. prausnitzii and E. coli (F–E index, undergone autologous haematopoietic stem cell trans-
B/F ratio) plantation. Integration of microbiome and metabolome
Microbiome dysbiotic index risk profiles from human donors and humanized mice
A measure quantifying the deviation (shift) of microbiome composition from a healthy improved the performance of predictive models of dis-
microbiome baseline. ease outcomes from AUC 0.79 to 0.96 and identified a
A ratio of the total abundance of organisms increased in inflammatory bowel disease network of disease-​associated bacteria and metabolites
over the total abundance of organisms decreased in inflammatory bowel disease. involved in sulfur metabolism20. Although these findings
Methodology: sound promising, it is important to acknowledge that
• 16S rRNA gene sequencing and shotgun metagenomics microbiome risk profiles are based on predictive models
• Dissimilarity metric (e.g. β-​diversity, Bray–Curtis) describing microbiome dissimilarity derived from populations or groups of patients included
between healthy and diseased states in prospective cohort studies, and hence could be more
accurate for groups of similar patients (either popula-
Multi-​omics based biomarkers
tions or cohorts) than they are for individuals. Therefore,
The concurrent application of omics applications to achieve a global view of changes in
genetic, metabolic and biochemical processes in the gut microbiome. Using machine
it is important to keep in mind that the predicted risks
learning, a subset of the features (candidate biomarkers) is generated and used to build might not translate directly to individual patients, pos-
predictive models that can effectively discriminate between disease subtypes. sibly owing to limited generalizability of the risk profile
Methodology: in heterogeneous settings.
Our second study investigated metabolic health and
• Metagenomics, metatranscriptomics, metaproteomics, metabolomics
the diurnal rhythmicity of gut microbiota in a German
• Machine learning methods
population-​based cohort of 1,976 individuals. Faecal

nATuRe RevIewS | GaSTROenTeROlOgy & HepaTOlOgy volume 19 | June 2022 | 391

0123456789();:
Reviews

microbiota profiling identified a risk signature of strain diversity. In this context, 16S rRNA and deep
13 microbial taxa that showed disrupted diurnal rhyth- shotgun metagenomic sequencing has been performed
micity in patients with T2DM. A predictive model based on stool samples collected from individuals living in
on this arrhythmic risk signature successfully identified Thailand and the USA, including first-​generation and
individuals at risk of developing T2DM and had an AUC second-​generation Thai immigrants to the USA before
of 0.78 when BMI was included in the model19. These and after immigration88. Intriguingly, US immigration
examples, among others, provide evidence for a role of was associated with substantial alterations to the gut
microbiome signatures in biomarker discovery for diag- microbiome, including loss of diversity, loss of bacterial
nostic and therapeutic purposes. However, it is impor- strains, functional loss of fibre degradation and a shift
tant to note that dysbiosis indices are not standalone from Prevotella to Bacteroides enterotypes. These per-
measurements and need to be integrated into additional turbations were intensified by obesity and increased with
host-​derived and clinical data. Standardization and val- each successive generation88.
idation of these indices require large-​scale studies that The changes reported in selected microbiome asso-
include longitudinal assessment of potential biomark- ciation studies from countries around the world reveal
ers and consider possible confounding variables such important connections between geographical loca-
as diet, age, ethnicity, medical history and lastly time of tion and gut microbial dysbiosis in individuals with
defecation, all of which have been proven to influence IBD and T2DM87,88. Owing to the ready availability of
microbiome alterations. 16S rRNA gene-​sequencing data sets, many of these
cohort studies have used 16S microbial profiling despite
Inconsistency of disease biomarkers variability between studies in the sequenced 16S vari-
Studies that compared populations from industrial- able regions or the sequencing platform used (Fig. 1).
ized countries with rural or non-​industrialized popu­ In the IBD stool-​d erived microbiome, Firmicutes,
lations have identified dramatic differences in their Proteobacteria and genera including Fusobacterium,
gut microbiome characteristics. A 2015 study com- E. coli, R. gnavus and Streptococcus, consistently showed
pared the faecal gut microbiota of individuals from an increased relative abundance that correlated with
two non-​industrialized regions in Papua New Guinea IBD activity17,21,40,43. By contrast, Roseburia, Blautia
with those of individuals resident in the USA 84. and Faecalibacterium consistently showed a decreased
Interestingly, gut microbiome profiles in the Papua relative abundance in these cohorts17,21,40,43. Most stud-
New Guinea residents showed much higher microbial ies of T2DM showed a decrease in the relative abun-
diversity and lower interindividual variation compared dance of A. muciniphila, Clostridium, Roseburia and
to those from US residents84. Many bacterial species Faecalibacterium89–92. However, substantial divergence
were common to both Papua New Guinea and US res- is evident in the disease-​associated microbiome profiles
idents but showed different abundance levels, which of individuals from different geographical and ethnic
were explained by decreased bacterial dispersal rates groups59,62,89,90,93–95 (Supplementary Tables 1,2). The rel-
in Western populations84. In another pioneering study evance of these differentially represented taxa to the
aimed at identifying a global core microbiome in healthy pathogenesis of IBD has been validated in several clini-
populations, the number of core genera decreased from cal and translational gnotobiotic experiments as we have
17 to 14 when gut microbiome profiles from rural popu­ already discussed29,58,63,67,80,96,97.
lations in Papua New Guinea, Peru and Tanzania were Collectively, these data indicate the necessity for
compared with those from populations in industrialized further global studies of human microbiota in differ-
countries, including Flemish and Dutch cohorts as well ent geographic locations across continents to rule out
as UK and US populations85. This loss of resident micro- region-​associated confounding factors and define spe-
bial species, termed the “disappearing microbes”86, might cific and individualized microbiome signatures. Studies
help to explain the rising incidence of chronic diseases included within the iHMP are exploring the link between
in industrialized countries. gut microbiome alterations and the development of
In a robust study conducted to validate the generaliz- IBD or T2DM in large population-​based cohorts18,71.
ability of several microbiota-​based classifying models of Nevertheless, to date, these studies have predominantly
metabolic health, the gut microbiota of 7,009 individuals focused on populations in industrialized countries
from 14 districts in a single Chinese province were char- and most data have been obtained from the USA and
acterized to test the effect of geographic location on the Europe, which represent ~17% of the world’s popula-
predictive power of the included models60. Interestingly, tion. A few national and international studies have been
host location showed the strongest association with initiated to characterize gut microbiome variations in
variations in gut microbiome60. Furthermore, in a large non-​European populations and ethnic groups, including
longitudinal intercontinental study of 531 patients with in Africa, Asia, South America and the Middle East98.
IBD from Ireland and Canada, geographic location In all these studies, the authors have tried to identify
was the major determinant of microbiome variations, disease-​associated microbial risk profiles that contribute
although most (90.3%) of the variance in microbiome to the development of either IBD or T2DM. However,
composition remained unexplained87. The importance achieving a consensus on which disease-​related bac-
of geographic factors and related environmental expo- terial taxa have utility in the diagnosis of these two
sures are also illustrated in migration studies, which diseases remains challenging. Despite the observed het-
clearly demonstrate strong associations between migra- erogeneity, a few bacterial taxa are indeed commonly
tion and both microbiome functional diversity and implicated across different studies (Fig. 1). For instance,

392 | June 2022 | volume 19 www.nature.com/nrgastro

0123456789();:
Reviews

an increased abundance of Akkermansia, Eubacterium metabolite biomarkers that have attracted research atten-
rectale, Alistipes and F. prausnitzii is correlated with tion owing to their associations with the development of
improved metabolic health in multiple reports62,91,94,99,100. inflammatory and metabolic disease in both human and
Similarly, an overabundance of E. coli, Enterococcus, mouse studies122–125. Tryptophan is an essential amino
Fusobacterium, R. gnavus and Streptococcus, and/or acid acquired from the diet and mainly absorbed in the
a reduction in F. prausnitzii, Roseburia and members small intestine, although a small fraction is catabolized
of the Ruminococcus genus, are common features of to indole metabolites in the colon126. Tryptophan metab-
IBD-​ a ssociated dysbiosis 16–18,20,40,43,101. Intriguingly, olism and downstream cellular signalling have been
some taxa show similar trends in both IBD and T2DM. extensively described in other reports126–128 and are not
For instance, an overabundance of Christensenellaceae discussed in this Review. One longitudinal cohort study
and E. coli has been linked to both CD-​associated and confirmed that high baseline fasting serum tryptophan
T2DM-​associated dysbiosis17,19,94, which raises ques- concentrations were associated with an increased risk of
tions about the specificity of the available microbiome developing T2DM in 213 Chinese individuals followed
signatures for discriminating between different dis- up for 10 years (comprising 51 people who went on to
ease entities. Looking at overlapping IBD-​associated develop T2DM and 162 individuals who remained met-
and T2DM-​associated core signatures showed similar abolically healthy)124. Moreover, the predictive power of
or opposite trends. The results of these 16S studies are tryptophan levels as a biomarker was comparable to that
summarized in Tables 1–3. Additional detailed informa- of five existing amino acids in discriminating between
tion on these 16S studies is available in Supplementary individuals who did and did not develop T2DM124.
Tables 1 and 2. Bacterial signatures associated with each Of note, prior reports showed that serum levels of sev-
disease entity are referred to by the highest reported tax- eral amino acids could identify patients with T2DM
onomic level included (bacterial phylum, family, genus from different populations with varying accuracy. For
and species) (Fig. 2). example, phenylalanine and valine performed best in
American populations118, whereas tyrosine was most
Metabolomics-​based biomarker discovery accurate in South Asian populations129. These findings
In the search for disease biomarkers, metabolites serve point to the importance of identifying region-​specific
as the most proximal indicators of disease activity biomarkers in achieving optimal diagnostic accuracy.
and are strongly linked to the regulatory signals that
underlie and modulate disease mechanisms. In fact, Clinical role of microbiome biomarkers. A valuable bio-
the metabolome and microbiome both fluctuate in marker must contribute additional classification power
relation to endogenous and exogenous factors such as to that derived from clinical information. As such, faecal
diet, environment, ageing and overall health status102. biomarkers are an obvious source of diagnostic mark-
Numerous studies have reported substantial altera- ers of mucosal diseases given that the faecal stream
tions in the gut metabolite profiles of patients with is in direct contact with the intestinal mucosa. Faecal
IBD18,20,103–105 or T2DM71,106–108. For instance, reduced lev- calprotectin, a granulocyte-​derived cytosolic protein
els of medium-​chain fatty acids, such as pentanoate and that can be detected in stool, is the most widely utilized
hexanoate109, and reduced levels of B vitamins110 have faecal biomarker for inflammatory disorders owing to
been found in the faecal metabolome of patients with the strength of the correlation between inflammation
IBD. Conversely, increased levels of amino acids, amines severity and faecal calprotectin levels130. Two reports
and carnitines were reported in the faeces and sera of confirmed the ability of faecal calprotectin levels to
adult and paediatric patients with IBD, respectively105,111. detect endoscopic inflammation, with a reported
The results of a landmark study showed that metabo- sensitivity of 70–100% and specificity of 44–100%131,132.
lite profiling could discriminate patients with IBD The wide ranges of these values are explained by varia-
from healthy individuals112. This report was followed tions in the cut-​off thresholds applied in each study131,132.
by numerous others that consistently showed that the Nevertheless, elevated levels of faecal calprotectin are
metabolite phenotype of patients with IBD differs from not specific for IBD but rather reflect inflammatory
that of healthy individuals 103,104,113,114. Interestingly, conditions in the gut, which are also associated with
metabolite profiling could also discriminate between other intestinal and metabolic pathologies (including
different forms of IBD, such as CD and UC112,114, and IBS, gastrointestinal malignancies, obesity and T2DM).
could further classify patients with CD as having either For instance, shotgun metagenomic profiling of the gut
ileal or colonic inflammation113. microbiota in stool samples from 1,792 individuals was
Similarly, metabolite profiling of patients with able to distinguish IBD from IBS, and machine learning
T2DM demonstrated altered activity of metabolic algorithms markedly improved the accuracy of these
Specificity pathways115,116. Serum levels of branched-​chain and aro- prediction models (AUC 0.91) compared to that of faecal
The proportion of individuals
matic amino acids, such as leucine, isoleucine, valine, calprotectin levels alone (AUC 0.80)133. Importantly, the
who genuinely do not have the
disease or condition and phenylalanine, tyrosine and tryptophan, were associated model with the highest prediction accuracy (AUC 0.93)
receive negative test results. with insulin resistance, obesity and the risk of develop- included faecal calprotectin levels as well as metagen-
ing T2DM in multiple reports117–119. Metabolite profiling omic profiling of the top 20 selected taxa133. These results
Sensitivity of patients with T2DM also revealed strong associations suggest that the integration of clinical and microbial
The proportion of individuals
who genuinely have the
between the levels of specific bacterial metabolites biomarkers improves diagnostic accuracy. Such an inte-
disease or condition and and disease onset71,107,120,121. As an example, the trypto- grated approach has been used to predict the response
receive positive test results. phan metabolism pathway includes several candidate to therapy in patients with IBD21. In this study, baseline

nATuRe RevIewS | GaSTROenTeROlOgy & HepaTOlOgy volume 19 | June 2022 | 393

0123456789();:
Reviews

Identification of Validated
microbiome signatures microbiome signatures

Correlation Causation

Microbiome

function
Composition
D
Identification of environmental st isea Intervention studies

ns
io
factors at se Diet, FMT, post-biotics

ct
e

in xa
ra
Ta
Geography, diet

te
Multi-omics Microbial ecosystem therapeutics
Clinical relevance integration Identification and isolation of microbes
Cross-sectional and longitudinal Immune Geographical and metabolites, building functional
population and cohort studies status location consortia
ctors

Enviro
Microbe and host profiling ic D Gnotobiotic mouse models
net ha ieta
st fa

Microbiota (metagenome, metabolome) Ge eup

nme
k bi ry
ma ts
Ho

Host (phenotypes, genome,

Me sures

nt
exp
metabolome, immune response) Se e
Ag

dica
x

o
tion
High-throughput omics analyses Functional characterization
Biomarkers

Fig. 3 | Gut microbiome signature discovery in IBD and T2DM. Once candidate microbiome-​based biomarkers are identified using
Investigation of the causative role of the gut microbiota in the pathogenesis complex computational and machine learning tools, functional validation
of complex diseases, such as inflammatory bowel disease (IBD) and type 2 of candidate biomarkers is achieved through comprehensive in vivo, in vitro
diabetes mellitus (T2DM), requires large cross-​sectional or longitudinal and preclinical studies. Isolation and identification of candidate bacterial
population-​b ased and cohort studies. Microbial, environmental taxa are performed for biobanking and for subsequent mechanistic studies
and host-​related factors need to be considered when establishing in gnotobiotic mice. Colonization of germ-​free mice with single bacterial
correlations between disease entities and the structure or function of the taxa or with synthetic minimal microbial consortia could give insight into
gut microbiome. Multi-​omics data are derived from high-​throughput the causative role of specific microbes and the underlying microorganism–
analysis of samples collected from the host (phenotype, genotype, host interactions. The clinical relevance of identified microbiome
metabolome, transcriptome) as well as luminal or mucosa-​associated gut signatures requires validation in faecal microbiota transfer (FMT) or dietary
microbiomes (composition, metabolic and genetic functional pathways). intervention studies.

clinical data (including serological, endoscopic and clin- and the highest degree of insulin resistance in the val-
ical biomarkers) were insufficiently predictive of remis- idation cohort yielded an AUC of 0.78, which suggests
sion (AUC 0.62), whereas the addition of taxonomic that the gut microbiota is an important modifier of
and metabolic profiles improved the diagnostic power T2DM progression136. In fact, although a broad range
to AUC 0.72 and AUC 0.74, respectively21. Further, faecal of diagnostic biomarkers has been proposed for T2DM,
calprotectin levels alone were able to distinguish patients most of them fail to capture the complexity of this dis-
with pouchitis from those without pouchitis (AUC ease or to grasp both microbial and metabolic altera-
0.63)134. By contrast, a microbiome species model (with tions. In this regard, metabolite biomarkers have been
or without faecal calprotectin level as an additional pre- used in combination with established clinical risk factors
dictor) achieved an AUC of 0.78, confirming that micro- to substantially improve disease classification137,138.
bial profiling had a superior diagnostic performance to
faecal calprotectin levels alone in the identification of Implications of microbiome signatures. Understanding of
pouchitis134. the functional role and specificity of alterations in single
Serological biomarkers of impaired glucose metab- bacterial taxa (pathobionts)139,140 and/or complex micro-
olism used in the diagnosis of T2DM include fasting bial communities (dysbiosis)141–143 is essential to resolve
plasma glucose levels, 2 h plasma glucose levels after the pathogenetic mechanisms of microorganism–host
a 75 g oral glucose challenge (the oral glucose tol- interactions in IBD or T2DM (Fig. 3). In this context,
Diagnostic biomarkers erance test) and levels of glycated haemoglobin135. functional alterations of the gut microbiome potentially
Characteristics that are directly
Combinations of serological and microbiome biomark- represent the consequences of host adaptations.
linked to the aetiology of the
disease such as elevated blood ers that predicted T2DM progression were identified in A causal link of gut microbiota to multiple dis-
glucose concentration (which is a study that used data from two Swedish cohorts136. eases has been demonstrated in gnotobiotic mouse
diagnostic of type 2 diabetes In this study, multivariate analyses demonstrated a experiments20,54,56,99,142,144–146. Germ-​free mouse models
mellitus) and markers that strong correlation between the degree of insulin resist- can be selectively colonized with single bacterial strains,
show a strong correlation with
inflammation in inflammatory
ance and microbiome variations136. Interestingly, the minimal bacterial consortia or defined complex gut
bowel disease (such as faecal use of a microbiome-​based machine learning model microbial ecosystems derived from human stool or other
calprotectin levels). to distinguish between individuals with the lowest donor material to study their impact on host phenotype.

394 | June 2022 | volume 19 www.nature.com/nrgastro

0123456789();:
Reviews

In IBD, single-​strain colonization of germ-​free mouse Nevertheless, the complexity and multifactorial patho-
models with a variety of commensal bacteria, including genesis of most of these diseases, as well as the existence
E. coli, Enterococcus faecalis, Bacteroides vulgatus and of a variety of confounding factors in human studies,
Bilophila wadsworthia has enabled us to understand continue to present major challenges to the clinical
some of the underlying mechanisms of disease initiation implementation of microbiome signatures in diagnosis,
or protection147–149. The generation of well-​characterized predicting prognosis and treatment decision-​making.
minimal bacterial consortia (such as SIHUMI150 and In this context, the identification, isolation and cultiva-
Oligo MM 12,151) provided the means to investigate tion of functionally relevant bacterial strains is needed,
more-​complex mechanisms of host–microorganism and along with isolation and identification of their metabo-
microorganism–microorganism crosstalk under both lites. As such, defining the functional capacity of a given
physiological and pathological conditions. In addition, microbial signature can be achieved by metagenomics
germ-​free mice colonized with human faecal micro- and metabolomics approaches. Strain-​level shotgun
biota (also known as human microbiome-​associated metagenomic sequencing can be used to identify the
mice) have been used extensively to investigate the microbial strains included in these signatures, including
underlying pathogenetic mechanisms of complex dis- keystone species that are present in low abundance yet
eases, including IBD, T2DM, obesity99,142, asthma and have an important role in disease development, and to
malnutrition20,54,99,142,144–146,152–154. infer bacterial metabolic pathways, microbial interac-
Our research work has shown that gut bacteria tions and microbial metabolites that affect host physiol-
are required to drive inflammation in a mouse model ogy. The establishment and use of well-​defined in vivo
of colitis, and that these same bacteria are associated gnotobiotic mouse models are expected to provide
with the risk of relapse in patients with CD20. Despite fundamental information on the effect of microbiome
the known limitations of incomplete human bacterial composition on host physiology and disease suscepti-
transfer into germ-​free mice155, our model captured bility. To address the observed heterogeneity and inter-
key features of the disease-​associated microbiome sig- individual variation in microbiome signatures, dense
natures. The transfer of gut microbiota into germ-​free microbiome sampling and disease modelling across
mice resulted in the successful transfer of several disease populations and ethnic groups should be performed to
states, thereby revealing shared functional metabolic improve the generalizability of predictive models. Thus,
pathways implicated in inflammation20. These shared the stratification of population and patient cohorts is
microbiome signatures could lead to improvements in necessary to improve individual assessments of disease
the classification of disease activity. Similarly, previ- risk. To ensure the reproducibility and between-​study
ous work showing that glucose tolerance156 and insulin comparability of the findings of microbiome studies,
resistance142,157 are influenced by the composition of the the specificity and sensitivity of microbiome signatures
gut microbiome has been verified by a series of FMT must be assessed and validated in multicentre studies of
studies in gnotobiotic mouse models142,143. well-​characterized cohorts of patients.
Current microbiome research is moving beyond the
Conclusions mere description of microbial community structures and
Over the past two decades, evidence from human and disease associations and progressing towards understand-
mouse studies has revealed a fundamental role of the ing the causative roles of gut bacteria in the pathogenesis
intestinal microbiome in the pathogenesis of inflam- of complex chronic disorders. These collective efforts are
matory and metabolic diseases such as IBD and T2DM. expected to enhance microbiome modelling and advance
Changes in the structure and function of the gut microbial the development of microbiota-​based signatures and/or
ecosystem, also termed dysbiotic microbiome signatures, risk profiles that can be utilized in clinical settings.
have been associated with disease activity, risk of relapse
or response to therapy in patients with these diseases. Published online 21 February 2022

1. Backhed, F., Ley, R. E., Sonnenburg, J. L., bacteria in metabolic disease: perpetrators or analysis for the Global Burden of Disease Study 2013.
Peterson, D. A. & Gordon, J. I. Host–bacterial bystanders? Nutrients 12, 1082 (2020). Lancet 387, 251–272 (2016).
mutualism in the human intestine. Science 307, 8. Xue, A. et al. Genome-​wide association analyses 14. Momozawa, Y. et al. IBD risk loci are enriched
1915–1920 (2005). identify 143 risk variants and putative regulatory in multigenic regulatory modules encompassing
2. Sender, R., Fuchs, S. & Milo, R. Are we really mechanisms for type 2 diabetes. Nat. Commun. 9, putative causative genes. Nat. Commun. 9, 2427
vastly outnumbered? Revisiting the ratio of 2941 (2018). (2018).
bacterial to host cells in humans. Cell 164, 337–340 9. de Lange, K. M., Moutsianas, L. & Lee, J. C. 15. Qi, Q., Wang, X., Strizich, G. & Wang, T. Genetic
(2016). Genome-wide association study implicates immune determinants of type 2 diabetes in Asians. Int. J.
3. Berg, G. et al. Microbiome definition re-​visited: old activation of multiple integrin genes in inflammatory Diabetol. Vasc. Dis. Res. https://doi.org/10.19070/
concepts and new challenges. Microbiome 8, 103 bowel disease. Nat. Genet. 49, 256–261 (2017). 2328-353X-​SI01001 (2015).
(2020). 10. Billings, L. K. & Florez, J. C. The genetics of type 2 16. Yilmaz, B. et al. Microbial network disturbances in
4. Kaplan, G. G. The global burden of IBD: from 2015 diabetes: what have we learned from GWAS? Ann. NY relapsing refractory Crohn’s disease. Nat. Med. 25,
to 2025. Nat. Rev. Gastroenterol. Hepatol. 12, Acad. Sci. 1212, 59–77 (2010). 323–336 (2019).
720–727 (2015). 11. Yang, S. K. et al. Genome-​wide association study 17. Pascal, V. et al. A microbial signature for Crohn’s
5. Saeedi, P. et al. Global and regional diabetes of Crohn’s disease in Koreans revealed three new disease. Gut 66, 813–822 (2017).
prevalence estimates for 2019 and projections for susceptibility loci and common attributes of genetic 18. Lloyd-​Price, J. et al. Multi-​omics of the gut microbial
2030 and 2045: results from the International susceptibility across ethnic populations. Gut 63, ecosystem in inflammatory bowel diseases. Nature
Diabetes Federation Diabetes Atlas, 9th edition. 80–87 (2014). 569, 655–662 (2019).
Diabetes Res. Clin. Pract. 157, 107843 (2019). 12. Franke, A. et al. Genome-​wide meta-​analysis 19. Reitmeier, S. et al. Arrhythmic gut microbiome
6. Blüher, M. Obesity: global epidemiology and increases to 71 the number of confirmed Crohn’s signatures predict risk of type 2 diabetes. Cell Host
pathogenesis. Nat. Rev. Endocrinol. 15, 288–298 disease susceptibility loci. Nat. Genet. 42, Microbe 28, 258–272.e6 (2020).
(2019). 1118–1125 (2010). 20. Metwaly, A. et al. Integrated microbiota and
7. Chakaroun, R. M., Massier, L. & Kovacs, P. 13. Zhou, M. et al. Cause-​specific mortality for 240 causes metabolite profiles link Crohn’s disease to sulfur
Gut microbiome, intestinal permeability, and tissue in China during 1990–2013: a systematic subnational metabolism. Nat. Commun. 11, 4322 (2020).

nATuRe RevIewS | GaSTROenTeROlOgy & HepaTOlOgy volume 19 | June 2022 | 395

0123456789();:
Reviews

21. Ananthakrishnan, A. N. et al. Gut microbiome function 46. Lewis, J. D. et al. Inflammation, antibiotics, and diet 72. Biomarkers Definitions Working Group. Biomarkers
predicts response to anti-​integrin biologic therapy in as environmental stressors of the gut microbiome and surrogate endpoints: preferred definitions and
inflammatory bowel diseases. Cell Host Microbe 21, in pediatric Crohn’s disease. Cell Host Microbe 18, conceptual framework. Clin. Pharmacol. Ther. 69,
603–610.e3 (2017). 489–500 (2015). 89–95 (2001).
22. Round, J. L. & Mazmanian, S. K. The gut microbiome 47. Vila, A. V. et al. Gut microbiota composition and 73. Califf, R. M. Biomarker definitions and their
shapes intestinal immune responses during health functional changes in inflammatory bowel disease applications. Exp. Biol. Med. 243, 213–221 (2018).
and disease. Nat. Rev. Immunol. 9, 313–323 (2009). and irritable bowel syndrome. Sci. Transl Med. 10, 74. Darfeuille-​Michaud, A. et al. High prevalence of
23. Jurjus, A. et al. Inflammatory bowel disease, eaap8914 (2019). adherent-​invasive Escherichia coli associated with ileal
colorectal cancer and type 2 diabetes mellitus: 48. Schirmer, M., Garner, A., Vlamakis, H. & Xavier, R. J. mucosa in Crohn’s disease. Gastroenterology 127,
the links. BBA Clin. 5, 16–24 (2015). Microbial genes and pathways in inflammatory bowel 412–421 (2004).
24. Verdugo-​Meza, A., Ye, J., Dadlani, H., Ghosh, S. disease. Nat. Rev. Microbiol. 17, 497–511 (2019). 75. Joossens, M. et al. Dysbiosis of the faecal microbiota
& Gibson, D. L. Connecting the dots between 49. D’Haens, G. R. et al. Early lesions of recurrent Crohn’s in patients with Crohn’s disease and their unaffected
inflammatory bowel disease and metabolic syndrome: disease caused by infusion of intestinal contents in relatives. Gut 60, 631–637 (2011).
a focus on gut-​derived metabolites. Nutrients 12, excluded ileum. Gastroenterology 114, 262–267 76. Fang, X. et al. Metagenomics-​based, strain-​level
1434 (2020). (1998). analysis of Escherichia coli from a time-​series of
25. Kootte, R. S. et al. Improvement of insulin sensitivity 50. Rutgeerts, P. et al. Effect of faecal stream diversion microbiome samples from a Crohn’s disease patient.
after lean donor feces in metabolic syndrome is driven on recurrence of Crohn’s disease in the neoterminal Front. Microbiol. 9, 2559 (2018).
by baseline intestinal microbiota composition. ileum. Lancet 338, 771–774 (1991). 77. Hall, A. B. et al. A novel Ruminococcus gnavus clade
Cell Metab. 26, 611–619.e6 (2017). 51. Perencevich, M. & Burakoff, R. Use of antibiotics enriched in inflammatory bowel disease patients.
26. Wang, H. et al. Promising treatment for type 2 in the treatment of inflammatory bowel disease. Genome Med. 9, 103 (2017).
diabetes: fecal microbiota transplantation reverses Inflamm. Bowel Dis. 12, 651–664 (2006). 78. Palm, N. W. et al. Immunoglobulin A coating identifies
insulin resistance and impaired islets. Front. Cell. 52. Hörmannsperger, G., Schaubeck, M. & Haller, D. colitogenic bacteria in inflammatory bowel disease.
Infect. Microbiol. 9, 455 (2020). Intestinal microbiota in animal models of inflammatory Cell 158, 1000–1010 (2014).
27. Sokol, H. et al. Fecal microbiota transplantation diseases. ILAR J. 56, 179–191 (2015). 79. Lopez-​Siles, M., Duncan, S. H., Garcia-​Gil, L. J. &
to maintain remission in Crohn’s disease: a pilot 53. Ahmed, M., Metwaly, A. & Haller, D. Modeling microbe- Martinez-​Medina, M. Faecalibacterium prausnitzii:
randomized controlled study. Microbiome 8, 12 (2020). host interaction in the pathogenesis of Crohn’s from microbiology to diagnostics and prognostics.
28. Costello, S. P. et al. Systematic review with meta-​ disease. Int. J. Med. Microbiol. 311, 151489 (2021). ISME J. 11, 841–852 (2017).
analysis: faecal microbiota transplantation for the 54. Nagao-​Kitamoto, H. et al. Functional characterization 80. Lopez-​Siles, M. et al. Mucosa-​associated
induction of remission for active ulcerative colitis. of inflammatory bowel disease-​associated gut dysbiosis Faecalibacterium prausnitzii and Escherichia coli
Aliment. Pharmacol. Ther. 46, 213–224 (2017). in gnotobiotic mice. Cell. Mol. Gastroenterol. Hepatol. co-abundance can distinguish irritable bowel syndrome
29. Vrieze, A. et al. Transfer of intestinal microbiota from 2, 468–481 (2016). and inflammatory bowel disease phenotypes.
lean donors increases insulin sensitivity in individuals 55. Sellon, R. K. et al. Resident enteric bacteria are Int. J. Med. Microbiol. 304, 464–475 (2014).
with metabolic syndrome. Gastroenterology 143, necessary for development of spontaneous colitis and 81. Chehoud, C. et al. Fungal signature in the gut
913–916.e7 (2012). immune system activation in interleukin-10-deficient microbiota of pediatric patients with inflammatory
30. Paramsothy, S., Kamm, M. & Kaakoush, N. Multidonor mice. Infect. Immun. 66, 5224–5231 (1998). bowel disease. Inflamm. Bowel Dis. 21, 1948–1956
intensive faecal microbiota transplantation for active 56. Schaubeck, M. et al. Dysbiotic gut microbiota causes (2015).
ulcerative colitis: a randomised placebo-​controlled transmissible Crohn’s disease-​like ileitis independent 82. Sarrabayrouse, G. et al. Fungal and bacterial loads:
trial. Lancet 389, 1218–1228 (2017). of failure in antimicrobial defence. Gut 65, 225–237 noninvasive inflammatory bowel disease biomarkers
31. Gupta, A. & Khanna, S. Fecal microbiota (2016). for the clinical setting. mSystems 6, e01277-20
transplantation. JAMA 318, 102 (2017). 57. Britton, G. J. et al. Microbiotas from humans with (2021).
32. Quraishi, M. N. et al. Systematic review with inflammatory bowel disease alter the balance of gut 83. Halfvarson, J. et al. Dynamics of the human gut
meta-analysis: the efficacy of faecal microbiota Th17 and RORγt+ regulatory T cells and exacerbate microbiome in inflammatory bowel disease.
transplantation for the treatment of recurrent and colitis in mice. Immunity 50, 212–224.e4 (2019). Nat. Microbiol. 2, 17004 (2017).
refractory Clostridium difficile infection. Aliment. 58. Round, J. L. & Mazmanian, S. K. Inducible Foxp3+ 84. Martínez, I. et al. The gut microbiota of rural Papua
Pharmacol. Ther. 46, 479–493 (2017). regulatory T-​cell development by a commensal New Guineans: composition, diversity patterns, and
33. Mocanu, V. et al. Fecal microbial transplantation and bacterium of the intestinal microbiota. Proc. Natl ecological processes. Cell Rep. 11, 527–538 (2015).
fiber supplementation in patients with severe obesity Acad. Sci. USA 107, 12204–12209 (2010). 85. Falony, G. et al. Population-​level analysis of gut
and metabolic syndrome: a randomized double-​blind, 59. Forslund, K. et al. Disentangling type 2 diabetes and microbiome variation. Science 352, 560–564 (2016).
placebo-​controlled phase 2 trial. Nat. Med. 27, metformin treatment signatures in the human gut 86. Blaser, M. J. & Falkow, S. What are the consequences
1272–1279 (2021). microbiota. Nature 528, 262–266 (2015). of the disappearing human microbiota? Nat. Rev.
34. van Lier, Y. F. et al. Donor fecal microbiota 60. He, Y. et al. Regional variation limits applications of Microbiol. 7, 887–894 (2009).
transplantation ameliorates intestinal graft-​versus-host healthy gut microbiome reference ranges and disease 87. Clooney, A. G. et al. Ranking microbiome variance
disease in allogeneic hematopoietic cell transplant models. Nat. Med. 24, 1532–1535 (2018). in inflammatory bowel disease: a large longitudinal
recipients. Sci. Transl Med. 12, eaaz8926 (2020). 61. Zhong, H. et al. Distinct gut metagenomics intercontinental study. Gut 70, 499–510 (2021).
35. Rossen, N. G. et al. Findings from a randomized and metaproteomics signatures in prediabetics and 88. Vangay, P. et al. US immigration westernizes the
controlled trial of fecal transplantation for patients treatment-​naive type 2 diabetics. EBioMedicine 47, human gut microbiome. Cell 175, 962–972.e10
with ulcerative colitis. Gastroenterology 149, 373–383 (2019). (2018).
110–118.e4 (2015). 62. Karlsson, F. H. et al. Gut metagenome in European 89. He, Y. et al. Linking gut microbiota, metabolic syndrome
36. Moayyedi, P. et al. Fecal microbiota transplantation women with normal, impaired and diabetic glucose and economic status based on a population-​level
induces remission in patients with active ulcerative control. Nature 498, 99–103 (2013). analysis. Microbiome 6, 172 (2018).
colitis in a randomized controlled trial. Gastroenterology 63. Shin, N.-R. et al. An increase in the Akkermansia spp. 90. Zeevi, D. et al. Personalized nutrition by prediction
149, 102–109.e6 (2015). population induced by metformin treatment improves of glycemic responses. Cell 163, 1079–1094 (2015).
37. Sood, A. et al. Acceptability, tolerability, and safety glucose homeostasis in diet-​induced obese mice. 91. Thingholm, L. B. et al. Obese individuals with and
of fecal microbiota transplantation in patients with Gut 63, 727–735 (2014). without type 2 diabetes show different gut microbial
active ulcerative colitis (AT&S Study). J. Gastroenterol. 64. Org, E. et al. Genetic and environmental control functional capacity and composition. Cell Host Microbe
Hepatol. 35, 418–424 (2020). of host-​gut microbiota interactions. Genome Res. 25, 26, 252–264.e10 (2019).
38. Xiang, L. et al. Efficacy of faecal microbiota 1558–1569 (2015). 92. Zhang, F. et al. Response of gut microbiota in type 2
transplantation in Crohn’s disease: a new target 65. Depommier, C. et al. Supplementation with diabetes to hypoglycemic agents. Endocrine 66,
treatment? Microb. Biotechnol. 13, 760–769 (2020). Akkermansia muciniphila in overweight and obese 485–493 (2019).
39. de Groot, P. et al. Donor metabolic characteristics human volunteers: a proof-​of-concept exploratory 93. Chávez-​Carbajal, A. et al. Characterization of the gut
drive effects of faecal microbiota transplantation on study. Nat. Med. 25, 1096–1103 (2019). microbiota of individuals at different T2D stages
recipient insulin sensitivity, energy expenditure and 66. Everard, A. et al. Cross-​talk between Akkermansia reveals a complex relationship with the host.
intestinal transit time. Gut 69, 502–512 (2020). muciniphila and intestinal epithelium controls diet-​ Microorganisms 8, 94 (2020).
40. Sokol, H. et al. Fungal microbiota dysbiosis in IBD. induced obesity. Proc. Natl Acad. Sci. USA 110, 94. Li, Q. et al. Implication of the gut microbiome
Gut 66, 1039–1048 (2017). 9066–9071 (2013). composition of type 2 diabetic patients from northern
41. Sokol, H. et al. Faecalibacterium prausnitzii is an 67. Plovier, H. et al. A purified membrane protein from China. Sci. Rep. 10, 5450 (2020).
anti-inflammatory commensal bacterium identified Akkermansia muciniphila or the pasteurized bacterium 95. Ahmad, A. et al. Analysis of gut microbiota of obese
by gut microbiota analysis of Crohn disease patients. improves metabolism in obese and diabetic mice. individuals with type 2 diabetes and healthy individuals.
Proc. Natl Acad. Sci. USA 105, 16731–16736 (2008). Nat. Med. 23, 107–113 (2017). PLoS ONE 14, e0226372 (2020).
42. Schirmer, M. et al. Compositional and temporal 68. Gilijamse, P. W. et al. Treatment with Anaerobutyricum 96. Lopez-​Siles, M. et al. Cultured representatives of two
changes in the gut microbiome of pediatric ulcerative soehngenii: a pilot study of safety and dose–response major phylogroups of human colonic Faecalibacterium
colitis patients are linked to disease course. Cell Host effects on glucose metabolism in human subjects with prausnitzii can utilize pectin, uronic acids, and host-​
Microbe 24, 600–610.e4 (2018). metabolic syndrome. NPJ Biofilms Microbiomes 6, 16 derived substrates for growth. Appl. Environ. Microbiol.
43. Gevers, D. et al. The treatment-​naive microbiome (2020). 78, 420–428 (2012).
in new-​onset Crohn’s disease. Cell Host Microbe 15, 69. Goodrich, J. K. et al. Human genetics shape the gut 97. Duncan, S. H., Hold, G. L., Barcenilla, A., Stewart, C. S.
382–392 (2014). microbiome. Cell 159, 789–799 (2014). & Flint, H. J. Roseburia intestinalis sp. nov., a novel
44. Sokol, H. & Seksik, P. The intestinal microbiota in 70. Zeller, G. et al. Potential of fecal microbiota for early-​ saccharolytic, butyrate-​producing bacterium from
inflammatory bowel diseases: time to connect with the stage detection of colorectal cancer. Mol. Syst. Biol. human faeces. Int. J. Syst. Evol. Microbiol. 52,
host. Curr. Opin. Gastroenterol. 26, 327–331 (2010). 10, 766 (2014). 1615–1620 (2002).
45. Morgan, X. C. et al. Dysfunction of the intestinal 71. Zhou, W. et al. Longitudinal multi-​omics of host– 98. Moossavi, S. The necessity for an Iranian gut
microbiome in inflammatory bowel disease and microbe dynamics in prediabetes. Nature 569, microbiome initiative. Middle East J. Dig. Dis. 6,
treatment. Genome Biol. 13, R79–R79 (2012). 663–671 (2019). 109–110 (2014).

396 | June 2022 | volume 19 www.nature.com/nrgastro

0123456789();:
Reviews

99. Turnbaugh, P. J. et al. An obesity-​associated gut 125. Hisamatsu, T. et al. Novel, objective, multivariate faecalis in a gnotobiotic mouse model of chronic,
microbiome with increased capacity for energy biomarkers composed of plasma amino acid profiles immune-​mediated colitis. Front. Immunol. 10, 1420
harvest. Nature 444, 1027–1031 (2006). for the diagnosis and assessment of inflammatory (2019).
100. Qin, J. et al. A metagenome-​wide association study bowel disease. PLoS ONE 7, e31131 (2012). 149. Kim, S. C., Tonkonogy, S. L., Karrasch, T., Jobin, C.
of gut microbiota in type 2 diabetes. Nature 490, 126. Roager, H. M. & Licht, T. R. Microbial tryptophan & Balfour Sartor, R. Dual-​association of gnotobiotic
55–60 (2012). catabolites in health and disease. Nat. Commun. 9, IL-10–/– mice with 2 nonpathogenic commensal
101. Lepage, P. et al. Twin study indicates loss of interaction 3294 (2018). bacteria induces aggressive pancolitis. Inflamm. Bowel
between microbiota and mucosa of patients with 127. Lavelle, A. & Sokol, H. Gut microbiota-​derived Dis. 13, 1457–1466 (2007).
ulcerative colitis. Gastroenterology 141, 227–236 metabolites as key actors in inflammatory bowel 150. Eun, C. S. et al. Induction of bacterial antigen-​specific
(2011). disease. Nat. Rev. Gastroenterol. Hepatol. 17, colitis by a simplified human microbiota consortium in
102. Cho, I. & Blaser, M. J. The human microbiome: at the 223–237 (2020). gnotobiotic interleukin-10–/– mice. Infect. Immun. 82,
interface of health and disease. Nat. Rev. Genet. 13, 128. Sorgdrager, F. J. H., Naudé, P. J. W., Kema, I. P., 2239–2246 (2014).
260–270 (2012). Nollen, E. A. & De Deyn, P. P. Tryptophan metabolism 151. Brugiroux, S. et al. Genome-​guided design of a defined
103. Franzosa, E. A. et al. Gut microbiome structure and in inflammaging: from biomarker to therapeutic mouse microbiota that confers colonization resistance
metabolic activity in inflammatory bowel disease. target. Front. Immunol. 10, 2565 (2019). against Salmonella enterica serovar typhimurium.
Nat. Microbiol. 4, 293–305 (2019). 129. Tillin, T. et al. Diabetes risk and amino acid profiles: Nat. Microbiol. 2, 16215 (2016).
104. Jacobs, J. P. et al. A disease-​associated microbial cross-​sectional and prospective analyses of ethnicity, 152. Lepage, P. et al. Twin study indicates loss of interaction
and metabolomics state in relatives of pediatric amino acids and diabetes in a South Asian and between microbiota and mucosa patients with ulcerative
inflammatory bowel disease patients. Cell. Mol. European cohort from the SABRE (Southall And Brent colitis. Gastroenterology 141, 227–236 (2011).
Gastroenterol. Hepatol. 2, 750–766 (2016). Revisited) Study. Diabetologia 58, 968–979 (2015). 153. Rehman, A. et al. Geographical patterns of the
105. Kolho, K.-L., Pessia, A., Jaakkola, T., de Vos, W. M. 130. Schoepfer, A. M. et al. Fecal calprotectin correlates standing and active human gut microbiome in health
& Velagapudi, V. Faecal and serum metabolomics more closely with the simple endoscopic score for and IBD. Gut 65, 238–248 (2016).
in paediatric inflammatory bowel disease. J. Crohns Crohn’s disease (SES-​CD) than CRP, blood leukocytes, 154. Turnbaugh, P. J. & Gordon, J. I. The core gut
Colitis 11, 321–334 (2017). and the CDAI. Am. J. Gastroenterol. 105, 162–169 microbiome, energy balance and obesity. J. Physiol.
106. Guasch-​Ferré, M. et al. Metabolomics in prediabetes (2010). 587, 4153–4158 (2009).
and diabetes: a systematic review and meta-​analysis. 131. Lewis, J. D. The utility of biomarkers in the diagnosis 155. Walter, J., Armet, A. M., Finlay, B. B. & Shanahan, F.
Diabetes Care 39, 833–846 (2016). and therapy of inflammatory bowel disease. Establishing or exaggerating causality for the gut
107. Tam, Z. Y. et al. Metabolite profiling in identifying Gastroenterology 140, 1817–1826.e2 (2011). microbiome: lessons from human microbiota-​associated
metabolic biomarkers in older people with late-​onset 132. Sipponen, T. & Kolho, K.-L. Fecal calprotectin in rodents. Cell 180, 221–232 (2020).
type 2 diabetes mellitus. Sci. Rep. 7, 4392 (2017). diagnosis and clinical assessment of inflammatory 156. Brown, K. et al. Prolonged antibiotic treatment
108. Urpi-​Sarda, M. et al. Metabolomics for biomarkers bowel disease. Scand. J. Gastroenterol. 50, 74–80 induces a diabetogenic intestinal microbiome that
of type 2 diabetes mellitus: advances and nutritional (2015). accelerates diabetes in NOD mice. ISME J. 10,
intervention trends. Curr. Cardiovasc. Risk Rep. 9, 12 133. Vich Vila, A. et al. Gut microbiota composition and 321–332 (2016).
(2015). functional changes in inflammatory bowel disease 157. Cani, P. D. et al. Metabolic endotoxemia initiates
109. Preter, V. D. et al. Faecal metabolite profiling identifies and irritable bowel syndrome. Sci. Transl Med. 10, obesity and insulin resistance. Diabetes 56,
medium-​chain fatty acids as discriminating compounds eaap8914 (2018). 1761–1772 (2007).
in IBD. Gut 64, 447–458 (2015). 134. Dubinsky, V. et al. Dysbiosis in metabolic genes of the 158. Vester-​Andersen, M. K. et al. Increased abundance
110. Santoru, M. L. et al. Cross sectional evaluation of the gut microbiomes of patients with an ileo-​anal pouch of proteobacteria in aggressive Crohn’s disease seven
gut-​microbiome metabolome axis in an Italian cohort resembles that observed in Crohn’s disease. mSystems years after diagnosis. Sci. Rep. 9, 13473 (2019).
of IBD patients. Sci. Rep. 7, 9523 (2017). 6, e00984-20 (2021). 159. Sanna, S. et al. Causal relationships among the gut
111. Ni, J. et al. A role for bacterial urease in gut dysbiosis 135. Long, J. et al. Metabolite biomarkers of type 2 microbiome, short-​chain fatty acids and metabolic
and Crohn’s disease. Sci. Transl Med. 9, eaah6888 diabetes mellitus and pre-​diabetes: a systematic diseases. Nat. Genet. 51, 600–605 (2019).
(2017). review and meta-​analysis. BMC Endocr. Disord. 20, 160. Kishikawa, T. et al. A metagenome-​wide association
112. Marchesi, J. R. et al. Rapid and noninvasive 174 (2020). study of gut microbiome in patients with multiple
metabonomic characterization of inflammatory bowel 136. Wu, H. et al. The gut microbiota in prediabetes and sclerosis revealed novel disease pathology. Front. Cell.
disease. J. Proteome Res. 6, 546–551 (2007). diabetes: a population-​based cross-​sectional study. Infect. Microbiol. 10, 585973 (2020).
113. Jansson, J. et al. Metabolomics reveals metabolic Cell Metab. 32, 379–390.e3 (2020). 161. Doumatey, A. P. et al. Gut microbiome profiles are
biomarkers of Crohn’s disease. PLoS ONE 4, e6386 137. Floegel, A. et al. Identification of serum metabolites associated with type 2 diabetes in urban Africans.
(2009). associated with risk of type 2 diabetes using a targeted Front. Cell. Infect. Microbiol. 10, 63 (2020).
114. Le Gall, G. et al. Metabolomics of fecal extracts detects metabolomic approach. Diabetes 62, 639–648 (2013). 162. Peters, B. A. et al. A taxonomic signature of obesity
altered metabolic activity of gut microbiota in ulcerative 138. Lu, Y. et al. Metabolic signatures and risk of type 2 in a large study of American adults. Sci. Rep. 8, 9749
colitis and irritable bowel syndrome. J. Proteome Res. diabetes in a Chinese population: an untargeted (2018).
10, 4208–4218 (2011). metabolomics study using both LC-​MS and GC-​MS.
115. Bain, J. R. et al. Metabolomics applied to diabetes Diabetologia 59, 2349–2359 (2016). Acknowledgements
research: moving from information to knowledge. 139. Buttó, L. F., Schaubeck, M. & Haller, D. Mechanisms The authors’ research is funded by the Deutsche
Diabetes 58, 2429–2443 (2009). of microbe-​host interaction in Crohn’s disease: Forschungsgemeinschaft (DFG, German Research Foundation) –
116. Menni, C. et al. Biomarkers for type 2 diabetes dysbiosis vs. pathobiont selection. Front. Immunol. project number 395357507 (SFB 1371, Microbiome
and impaired fasting glucose using a nontargeted 6, 555 (2015). Signatures) — and has received funding from the European
metabolomics approach. Diabetes 62, 4270–4276 140. Jochum, L. & Stecher, B. Label or concept — what is Union’s Horizon 2020 research and innovation programme
(2013). a pathobiont? Trends Microbiol. 28, 789–792 (2020). under grant agreement number 964590.
117. Batch, B. C. et al. Branched chain amino acids are 141. Buttó, L. F. & Haller, D. Dysbiosis in Crohn’s disease —
novel biomarkers for discrimination of metabolic joint action of stochastic injuries and focal inflammation Author contributions
wellness. Metabolism 62, 961–969 (2013). in the gut. Gut Microbes 8, 53–58 (2017). A.M. and D.H. contributed equally to all aspects of the article.
118. Wang, T. J. et al. Metabolite profiles and the risk 142. Ridaura, V. K. et al. Gut microbiota from twins In addition, S.R. researched data for the article, contributed
of developing diabetes. Nat. Med. 17, 448–453 discordant for obesity modulate metabolism in mice. substantially to discussion of its content, and reviewed and/or
(2011). Science 341, 1241214 (2013). edited the manuscript before submission.
119. Würtz, P. et al. Branched-​chain and aromatic amino 143. Turnbaugh, P. J. et al. A core gut microbiome in obese
acids are predictors of insulin resistance in young and lean twins. Nature 457, 480–484 (2009). Competing interests
adults. Diabetes Care 36, 648–655 (2013). 144. Arrieta, M., Sadarangani, M., Brown, E. M., The authors declare no competing interests.
120. Salihovic, S. et al. Non-​targeted urine metabolomics Russell, S. L. & Nimmo, M. A humanized microbiota
and associations with prevalent and incident type 2 mouse model of ovalbumin-​induced lung inflammation. Peer review information
diabetes. Sci. Rep. 10, 16474 (2020). Gut Microbes 7, 342–352 (2016). Nature Reviews Gastroenterology & Hepatology thanks
121. Wang-​Sattler, R. et al. Novel biomarkers for pre-​ 145. Blanton, L.V. et al. Gut bacteria that prevent Herbert Tilg, Chaysavanh Manichanh and the other, anony-
diabetes identified by metabolomics. Mol. Syst. Biol. growth impairments transmitted by microbiota from mous, reviewer(s) for their contribution to the peer review of
8, 615 (2012). malnourished children. Science 351, aad3311 (2016). this work.
122. Lamas, B. et al. CARD9 impacts colitis by altering 146. Kostic, A. D. et al. The dynamics of the human infant
gut microbiota metabolism of tryptophan into aryl gut microbiome in development and in progression Publisher’s note
hydrocarbon receptor ligands. Nat. Med. 22, towards type 1 diabetes. Cell Host Microbe 17, Springer Nature remains neutral with regard to jurisdictional
598–605 (2016). 260–273 (2016). claims in published maps and institutional affiliations.
123. Nikolaus, S. et al. Increased tryptophan metabolism 147. Steck, N. et al. Enterococcus faecalis metalloprotease
is associated with activity of inflammatory bowel compromises epithelial barrier and contributes Supplementary information
diseases. Gastroenterology 153, 1504–1516.e2 to intestinal inflammation. Gastroenterology 141, The online version contains supplementary material available
(2017). 959–971 (2011). at https://doi.org/10.1038/s41575-022-00581-2.
124. Chen, T. et al. Tryptophan predicts the risk for future 148. Lengfelder, I. et al. Complex bacterial consortia
type 2 diabetes. PLoS ONE 11, e0162192 (2016). reprogram the colitogenic activity of Enterococcus © Springer Nature Limited 2022

nATuRe RevIewS | GaSTROenTeROlOgy & HepaTOlOgy volume 19 | June 2022 | 397

0123456789();:

You might also like