An Increase in The Akkermansia Spp. Population Induced by Metformin Treatment Improves Glucose Homeostasis in Diet-Induced Obese Mice

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/242333891

An increase in the Akkermansia spp. Population induced by metformin


treatment improves glucose homeostasis in diet-induced obese mice

Article  in  Gut · June 2013


DOI: 10.1136/gutjnl-2012-303839 · Source: PubMed

CITATIONS READS

312 1,970

7 authors, including:

Na-Ri Shin Min-Soo Kim


Kyung Hee University Kyungnam University
56 PUBLICATIONS   862 CITATIONS    108 PUBLICATIONS   1,922 CITATIONS   

SEE PROFILE SEE PROFILE

Tae Woong Whon Myung-Shik Lee


Kyung Hee University Yonsei University
56 PUBLICATIONS   857 CITATIONS    233 PUBLICATIONS   9,858 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Autophagy, Metabolism and Chronic Diseases View project

All content following this page was uploaded by Na-Ri Shin on 20 May 2014.

The user has requested enhancement of the downloaded file.


Gut Online First, published on June 26, 2013 as 10.1136/gutjnl-2012-303839
Gut microbiota

ORIGINAL ARTICLE

An increase in the Akkermansia spp. population


induced by metformin treatment improves glucose
homeostasis in diet-induced obese mice
Na-Ri Shin,1 June-Chul Lee,2 Hae-Youn Lee,2 Min-Soo Kim,1 Tae Woong Whon,1
Myung-Shik Lee,2 Jin-Woo Bae1

▸ Additional material is ABSTRACT


published online only. To view Background Recent evidence indicates that the Significance of this study
please visit the journal online
(http://dx.doi.org/10.1136/
composition of the gut microbiota contributes to the
gutjnl-2012-303839). development of metabolic disorders by affecting the
1 physiology and metabolism of the host. Metformin is What is already known about this subject?
Department of Life and
Nanopharmaceutical Sciences one of the most widely prescribed type 2 diabetes (T2D) ▸ Alterations in the gut microbiota composition
and Department of Biology, therapeutic agents. play a role in the development of adipose
Kyung Hee University, Seoul, Objective To determine whether the antidiabetic effect tissue inflammation and in the pathogenesis of
Korea of metformin is related to alterations of intestinal
2
Department of Medicine and
obesity-induced diabetes.
Department of Biotechnology
microbial composition. ▸ The presence of the mucin-degrading
& Bioengineering, Samsung Design C57BL/6 mice, fed either a normal-chow diet or a bacterium, Akkermansia spp., is associated
Medical Center, Sungkyunkwan high-fat diet (HFD), were treated with metformin for with healthy mucosa.
University School of Medicine, 6 weeks. The effect of metformin on the composition of ▸ Immune cells resident in the adipose tissue are
Seoul, Korea
the gut microbiota was assessed by analysing 16S rRNA critical promoters of obesity and insulin
Correspondence to gene sequences with 454 pyrosequencing. Adipose tissue resistance.
Dr Jin-Woo Bae, inflammation was examined by flow cytometric analysis of
Department of Life and the immune cells present in visceral adipose tissue (VAT). What are the new findings?
Nanopharmaceutical Sciences Results Metformin treatment significantly improved the ▸ The microbial community in the intestine of
and Department of Biology, metformin-treated high-fat diet-fed (HFD-Met)
Kyung Hee University, Seoul glycaemic profile of HFD-fed mice. HFD-fed mice treated
130-701, Korea; baejw@khu. with metformin showed a higher abundance of the mucin- mice exhibited a unique microbial consortium
ac.kr degrading bacterium Akkermansia than HFD-fed control that was distinct from that in the HFD-fed
or Dr Myung-Shik Lee, mice. In addition, the number of mucin-producing goblet control mice (HFD-CT) or normal-chow diet
Department of Medicine, (NCD)-fed mice (NCD-CT and NCD-Met).
cells was significantly increased by metformin treatment
Samsung Medical Center, ▸ Marked changes in the abundance of 29
Sungkyunkwan University (p<0.0001). Oral administration of Akkermansia
School of Medicine, Seoul 135- muciniphila to HFD-fed mice without metformin genera, particularly mucin-degrading
710, Korea; mslee0923@skku. significantly enhanced glucose tolerance and attenuated Akkermansia, accounted for the observed
edu adipose tissue inflammation by inducing Foxp3 regulatory differences in the gut microbial communities
T cells (Tregs) in the VAT. following a HFD or metformin treatment.
N-RS, J-CL and H-YL
contributed equally to this Conclusions Modulation of the gut microbiota (by an ▸ The number of mucin-producing goblet cells
work. increase in the Akkermansia spp. population) may contribute increased upon metformin treatment, regardless
to the antidiabetic effects of metformin, thereby providing a of the dietary composition or glucose tolerance.
Received 28 September 2012
new mechanism for the therapeutic effect of metformin in ▸ Akkermansia-administered HFD-fed mice
Revised 15 May 2013 (HFD-Akk) showed improved glucose tolerance
Accepted 30 May 2013 patients with T2D. This suggests that pharmacological
manipulation of the gut microbiota in favour of Akkermansia and an increase in the number of goblet cells
may be a potential treatment for T2D. and adipose tissue-resident CD4 Foxp3
regulatory T cells (Tregs).
How might it impact on clinical practice in
INTRODUCTION
the foreseeable future?
Worldwide prevalence of obesity has increased over
▸ This study provides circumstantial evidence that
the past 30–40 years owing to changes in dietary pat-
modulation of the gut microbial community,
terns and reduced physical activity.1 The condition
either by metformin treatment or by
has now become a pandemic.2 3 Although the
Akkermansia administration, may result in an
primary reason for obesity is an imbalance between
improved metabolic profile in patients with
dietary energy intake and expenditure, it is becoming
type 2 diabetes.
clear that the gut microbiota plays a role in control-
ling energy balance and immune homeostasis.4–11
To cite: Shin N-R, Lee J-C,
Lee H-Y, et al. Gut
The human gut microbiota comprises 10–100 trillion
Published Online First: microorganisms and more than 1000 different bac-
[please include Day Month terial species.12 It also plays important roles in regu- Apart from its beneficial functions for the host,
Year] doi:10.1136/gutjnl- lating the host’s metabolism and extracting energy gut microbiota can potentially take part in patho-
2012-303839 from ingested food.4 8 10 13 physiological interactions with the host, particularly

Copyright
Shin N-R, et Article authordoi:10.1136/gutjnl-2012-303839
al. Gut 2013;0:1–9. (or their employer) 2013. Produced by BMJ Publishing Group Ltd (& BSG) under licence.
1
Gut microbiota

in the case of obesity and related metabolic disorders. Recent microbiota. The results show that metformin induces a pro-
studies have shown that changes in the gut microbiota may play found shift in the composition of the gut microbiota community
a role in the pathogenesis of the obese and diabetic phenotypes. and suggest a mechanism by which this drug exerts its thera-
For example, germ-free mice are protected from high-fat diet peutic effect in patients with diabetes.
(HFD)-induced obesity and metabolic dysfunction, including
glucose intolerance.4 6 In addition, colonisation of germ-free MATERIALS AND METHODS
animals with gut microbiota isolated from conventionally raised Animals
obese donors led to a significant increase in body fat content C57BL/6 mice (4-week-old, n=24; Charles River Laboratories,
and insulin resistance in recipient mice.10 14 Similarly, the trans- L’Arbresle, France) were maintained in groups of no more than
fer of gut microbiota from Toll-like receptor 5-deficient mice to six mice per cage with free access to food and water. Mice were
wild-type germ-free mice also transferred the diabetic pheno- fed either a normal-chow diet (NCD) or a HFD (60% fat, 20%
type of the Toll-like receptor 5-deficient donor to the recipi- protein, 20% carbohydrate (kcal/100 g), #D12492; Research
ent.15 Taken together, these studies suggest that alterations in Diets, New Brunswick, New Jersey, USA). After 8 weeks, the
the gut microbial community increase the capacity of the host to mice were divided into four groups of six and fed the following
extract energy from a given diet, thereby triggering the develop- diets: (1) a NCD without metformin treatment (NCD-CT), (2)
ment of obesity and diabetes. This indicates a link between the a NCD with metformin treatment (NCD-Met), (3) a HFD
gut microbiota and the diabetic phenotype of the host. without metformin treatment (HFD-CT) or (4) a HFD with
Obesity and type 2 diabetes (T2D) are also associated with metformin treatment (HFD-Met). The two groups of
chronic low-grade tissue inflammation.16 17 Numerous studies metformin-treated mice (NCD-Met and HFD-Met) received
suggest that adipose tissue is not just a fat storage depot; it also 300 mg/kg/day of metformin by oral gavage for a period of
controls systemic metabolism by acting as an endocrine organ.18 6 weeks. Body weight was monitored once a week. Akkermansia
Moreover, inflammation within adipose tissue disrupts triglycer- muciniphila MucT (CIP107 961) were cultured anaerobically on
ide storage, and the subsequent excess of free fatty acids induces brain–heart infusion (BBL) agar plates containing porcine mucin
insulin resistance.19 At the cellular level, immune cells present (Sigma) at 37°C. The bacteria were harvested at the late expo-
in adipose tissue, such as neutrophils and macrophages, partici- nential growth phase, suspended in thioglycolate–phosphate-
pate in obesity-induced pathology.20 21 Of the different types of buffered saline (PBS) (4.0×108 cfu) and orally administered to
immune cells that reside in adipose tissue, CD4 Foxp3 regula- HFD-fed mice (HFD-Akk; n=6).
tory T cells (Tregs) control local inflammation by counteracting
the infiltration of M1 macrophages and CD8 T cells, thereby Biochemical analyses
preventing adipose tissue inflammation.22 The protective role Glucose homeostasis
played by these Tregs has also been demonstrated in several Mice were fasted overnight and a glucose tolerance test was per-
animal models of obesity and insulin resistance, such as leptin- formed after an intraperitoneal injection of glucose (1 g/kg body
deficient mice and HFD-fed mice.23 Both the percentage and weight). The homeostasis model assessment of insulin resistance
number of Tregs in the adipose tissue of obesity-induced dia- (HOMA-IR) index was calculated as previously described.35
betic animals are considerably lower than those in lean controls. Blood glucose concentrations were measured with an
Metformin (1,1-dimethylbiguanide hydrochloride) has been Accu-Check glucometer (Roche) both before (0 min) and after
widely used to treat T2D for the past 50 years.24–26 In T2D, (15, 30, 60, 120 and 180 min) the glucose injections.
increased hepatic glucose production is an important cause of
hyperglycaemia. Although the precise mechanism underlying Histology
the glucose-lowering effect of metformin is not fully under- The distal ileum was removed from all the mice, flushed with
stood, the most commonly accepted mechanism involves the PBS, fixed in 4% formaldehyde overnight at room temperature
suppressed transcription of genes involved in gluconeogenesis and paraffin-embedded. The specimens were then cut into 5 μm
via activation of AMP-activated protein kinase (AMPK), an sections and stained with Alcian blue periodic acid–Schiff (PAS)
enzyme that senses cellular energy levels and regulates the avail- (the goblet cells were stained blue). The results were expressed
ability of fuel.27–29 The intestinal microbiota may also control as the number of goblet cells per intestinal villus.
the activation status of AMPK. For example, germ-free mice
that are protected against diet-induced obesity show increased Analysis of gut microbiota and quantification of
levels of AMPK activity in the liver and in muscle tissue.6 Akkermansia muciniphila
Furthermore, the intestines themselves make an important con- The methods used to analyse the gut microbiota and to quantify
tribution to the glucose-lowering effect of metformin.30–32 A muciniphila are described in the online supplementary
Several studies report much higher concentrations of metformin methods (see online supplementary table S1).
in the intestinal mucosa than in other tissues.31 32 These find-
ings raise the possibility that metformin may have both direct Isolation of stromal vascular fraction cells and flow
and indirect effects on the gut microbiota, which may in turn cytometry
contribute to its antidiabetic effects. The visceral adipose tissue (VAT) was excised and minced into
However, no experimental or clinical studies have examined small pieces (>2 mm) and incubated for 20 min in a collagenase
the effects of antidiabetic drugs on the gut microbiota, although solution (1 mg/mL of collagenase type 2 (Worthington) in
gut microbial dysbiosis has been reported in patients with Dulbecco’s modified Eagle’s medium). The digested tissue was
T2D.33 34 Therefore, questions about whether metformin, as an centrifuged at 1000g for 8 min. The pellet containing the
antidiabetic agent, regulates glucose metabolism by modulating stromal vascular fraction (SVF) was resuspended in PBS and fil-
the gut microbiota remain unanswered. This study examined the tered through a 70 μm mesh filter. The cells were then washed
therapeutic effect of metformin on the progression of the dia- with PBS, incubated in red blood cell (RBC) lysis buffer (Sigma)
betic phenotype in HFD-induced obese and diabetic mouse for 3 min and resuspended in fluorescence-activated cell sorting
models and studied the possible contribution of the gut (FACS) buffer (2 mM EDTA, 2% fetal bovine serum in PBS).

2 Shin N-R, et al. Gut 2013;0:1–9. doi:10.1136/gutjnl-2012-303839


Gut microbiota

The isolated cells were then incubated with labelled monoclonal Classification of the sequences was carried out by non-OTU
antibodies (eBioscience and BD Pharmingen) and analysed by (operational taxonomic units)-based assessment using the
flow cytometry using a FACSAria flow cytometer (Becton Ribosomal Database Project classifier. All the sequences were
Dickinson). Data were analysed with FlowJo software (Tree Star, assigned to eight bacterial phyla, comprising three phyla that
Inc). We validated the flow cytometric identification of CD11c dominate the distal gut in both mice and humans: Firmicutes,
F4/80 CD11b (M1) and CD206 F4/80 CD11b (M2) adipose Bacteroidetes and Actinobacteria.8 Although all individuals are
tissue macrophages. The percentage of Tregs was analysed by born with a specific gut microbiome, dietary factors have a sig-
three-colour flow cytometry after staining with antibodies to nificant influence on the development of the gut microbial com-
CD3, CD4 and mouse/rat Foxp3 (FJK-16 s), as previously position.8 10 13 We observed that HFD-CT mice had a greater
described.36 abundance of Firmicutes (p<0.001) and a lower abundance of
Bacteroidetes (p<0.001) than NCD-CT mice (see online supple-
Statistical analysis mentary figure S2A). In addition, HFD-CT mice showed a sig-
Data were expressed as the mean±SEM. Significance of the dif- nificantly lower abundance of the phyla Verrucomicrobia
ferences between the groups of mice was assessed using the (p<0.001) and TM7 ( p<0.01) than NCD-CT mice.
Student t test. For experiments comparing multiple groups, the Thus, we hypothesised that metformin may alter the compos-
differences were analysed by one-way analysis of variance fol- ition of the gut microbiota in parallel with its antidiabetic effect
lowed by Duncan’s post-hoc test. p Values <0.05 were consid- on host glucose metabolism. Metformin treatment resulted in a
ered significant. The nearest shrunken centroid (NSC) method profound shift in the fecal microbial community profiles of the
was used to detect the bacterial genera that were specifically gut microbiota in diabetic mice (see online supplementary figure
over- or under-represented within each category (diet, treatment S2B). The tendency toward phyla-wide changes after metformin
or diet–treatment combinations).37 The amount of shrinkage treatment was similar in NCD- and HFD-fed mice; however,
was chosen to minimise the overall misclassification error. These the changes were more marked in the HFD-fed group. There
analyses allowed the identification of bacterial genera whose were significant differences in the abundance of Firmicutes and
relative abundance was significantly different between the cat- Bacteroidetes between HFD-CT mice and HFD-Met mice
egories.38 The analysis was performed using the Predictive (p<0.001), but there was no significant difference in the abun-
Analysis for Microarrays package under R software. dance of these two phyla between NCD-Met mice and
NCD-CT mice. Importantly, the abundance of Verrucomicrobia
was significantly increased in the HFD group after metformin
RESULTS treatment ( p<0.001), whereas no significant change was seen in
Metformin improves glucose homeostasis under HFD the NCD group. This suggests that metformin-induced modula-
conditions tion of the gut microbiota is diet-dependent.
To assess the effect of metformin on obesity and glucose intoler- The overall composition of the bacterial community in the
ance, we used a glucose tolerance test to examine glucose different groups was characterised by quantifying and interpret-
homeostasis in metformin-treated and non-treated mice fed ing similarities based on phylogenetic distances using UniFrac
either a NCD or a HFD. As expected, in comparison with metric. The principal coordinates analysis of UniFrac-based pair-
NCD-CT mice, HDF-CT mice showed an increase in the area wise comparisons of community structures disclosed an unex-
under the curve (AUC), an increased HOMA-IR index and an pected distribution of the microbial community among the four
increased incidence of fasting hyperglycaemia (figure 1), all of groups of mice. The main finding of the principal coordinates
which suggest glucose intolerance. Total body weight and fat analysis was that different diets promote the development of dif-
pad weight also increased in mice fed a HFD (see online supple- ferent gut microbial communities. Furthermore, HFD-Met mice
mentary figure S1). Compared with HFD-CT mice, HFD-Met formed a cluster that was distinct from that of HFD-CT mice
mice showed a significant improvement in glucose tolerance, and not even intermediate between those of the control
fasting blood glucose levels and the HOMA-IR index (p<0.01) HFD-fed and NCD-fed mice (figure 2). The microbial commu-
(figure 1A–D), demonstrating that metformin has an anti- nities of the NCD-fed groups were, however, closely clustered,
hyperglycaemic effect. Metformin treatment did not affect even in mice treated with metformin; this indicates that metfor-
fasting blood glucose levels, glucose tolerance or the HOMA-IR min has a more marked effect on gut microbial community
index in NCD-fed mice. In contrast to its effects on glucose composition in HFD-fed mice than it does in NCD-fed mice.
metabolism, 6 weeks of metformin treatment had no effect on Taken together, these results indicate that metformin has differ-
body weight or fat pad weight in NCD- or HFD-fed mice (see ent effects on the gut microbial composition of NCD- and
online supplementary figure S1), indicating that the improve- HFD-fed mice.
ments in glucose tolerance seen in the HFD-Met mice are not Next, to determine which bacterial genera contributed to
related to changes in body weight or fat mass. these differences in microbiota composition between the four
groups, we performed a NSC analysis. The specifically defined
Effect of metformin on the composition of the gut microbial comparisons between community characteristics were tabulated
community on a heat map representing the relative abundance of each
Since the composition of the gut microbiota is associated with genus. By combining both statistical and NSC analyses, we
T2D,33 we next determined the effect of metformin on the found that changes in the abundance of 29 genera, belonging to
composition of the gut microbiota using metagenomic analysis six phyla, accounted for the differences in the gut microbial
based on 454 pyrosequencing technology. A total of 121 051 communities seen in mice fed different diet or metformin treat-
raw reads were obtained from 20 fecal samples. After perform- ment (figure 3), which suggests the possibility that the antidia-
ing quality control procedures to remove low-quality sequences, betic effect of metformin may be mediated by a specific subset
63 132 sequences (with an average length of 370±25 bases) of bacterial taxa. The relative abundance of Anaerotruncus,
were subjected to further analysis (see online supplementary Lactococcus, Akkermansia, Parabacteroides, Odoribacter,
table S2). Alistipes, Lawsonia, Blautia and Lactonifactor was altered by a

Shin N-R, et al. Gut 2013;0:1–9. doi:10.1136/gutjnl-2012-303839 3


Gut microbiota

Figure 1 Metformin improves glucose homeostasis in HFD-fed (diabetic) mice. The glucose tolerance test (GTT) (A), the area under the curve
(AUC) for the GTT curves (B), the fasting glucose levels (C) and the homeostasis model assessment of insulin resistance (HOMA-IR) index (D) are
shown for both control (CT) and metformin-treated (Met) mice fed a normal-chow diet (NCD) or a high-fat diet (HFD). All mice were fasted
overnight (16 h) before the GTT. Data are expressed as the mean±SEM (n=6 mice/group). The letters denote the level of statistical significance
( p<0.05) using one-way analysis of variance followed by Duncan’s post-hoc test.

HFD; however, metformin rescued these HFD-induced (HFD-Abx), showed a significant improvement in glucose toler-
changes, restoring the levels to those seen in NCD-fed mice. ance in comparison with HFD-CT mice ( p<0.05) (see online
Despite the low relative abundance (0.3–2.9%) of sequences supplementary figure S3). However, the reduction in the AUC
assigned to Akkermansia, this genus made a large contribution seen for metformin- and antibiotic-treated HFD-fed mice
to the observed differences in the composition of the gut micro- (HFD-Abx+Met) was not significantly different from that seen
bial communities in HFD-CT and HFD-Met mice. Moreover, for HFD-Abx mice. This suggests that antidiabetic effect of met-
Akkermansia was responsible for the greater abundance of formin can be abrogated in the absence of the gut microbiota.
Verrucomicrobia seen in HFD-Met mice compared with
HFD-CT mice. Thus, metformin restored the relative abun- Metformin treatment increases the number of goblet cells
dance of specific bacterial genera in HFD-Met mice to that seen Because metformin significantly increased the relative abun-
in NCD-fed mice, an action that may play a role in its glucose- dance of Akkermansia in HFD-Met mice and Akkermansia uses
lowering effects. mucus as a nutrient source,39 we next examined the ileum in
The unexpected changes in the composition of the gut micro- mice from the different treatment groups and counted the
biota in HFD-Met mice prompted us to examine whether deplet- number of mucin-producing goblet cells. PAS–Alcian blue stain-
ing the microbiota using broad-spectrum antibiotics would ing showed that metformin treatment increased the number of
abrogate the antidiabetic effects of metformin. To examine this, PAS-positive goblet cells (figure 4A–D). To evaluate these
HFD-fed mice were treated with a combination of antibiotics changes further, we counted the number of PAS-positive goblet
(carbenicillin, metronidazole, neomycin and vancomycin) before cells per villus. An average of 6.6±0.3 goblet cells was seen in
metformin treatment. It is noteworthy that HFD-fed mice HFD-CT mice, which was lower than that in NCD-CT mice
treated with antibiotics, but not treated with metformin (7.8±0.2) (figure 4E). Interestingly, metformin treatment

Figure 2 Cluster analysis of control (CT) or metformin-treated (Met) mice fed a normal-chow diet (NCD) or a high-fat diet (HFD). Bacterial
communities were clustered using the principal coordinates analysis (PCoA) of unweighted (A) and weighted (B) UniFrac distance matrices. The first
two principal coordinates (PC1 and PC2) from the PCoA of unweighted and weighted UniFrac are plotted for each sample. The percentage variation
in the plotted principal coordinates is indicated on the axes. Each spot represents one sample and each group of mice is denoted by a different
colour.

4 Shin N-R, et al. Gut 2013;0:1–9. doi:10.1136/gutjnl-2012-303839


Gut microbiota

Figure 3 Striking differences in the


abundance of the different bacterial
genera that comprise the gut microbial
communities in NCD-fed and HFD-fed
mice in CT and Met mice. (A) Nearest
shrunken centroids of the 29 genera
that account for the differences in the
composition of the microbial
communities between the four groups
of mice. For the genera listed in the
centre, those that are over-represented
are designated by rightward-extending
bars. The bars denoting
under-represented genera extend to
the left. The length of the bar indicates
the strength of the effect. (B) Heat
map showing the relative abundance
of each classified genus. Each column
on the heat map represents a sample
of 20 mice (n=5 mice/group) and each
row represents the genus listed in the
centre. A range of colours, from yellow
to black, indicates the prevalence of
each genus. All the genera listed in the
centre showed significant differences
( p<0.05; one-way analysis of variance
followed by Duncan’s post-hoc test).
NCD-CT, normal-chow diet (NCD)-fed
control mice; NCD-Met, NCD-fed
metformin-treated mice; HFD-CT,
high-fat diet (HFD)-fed control mice;
HFD-Met, HFD-fed metformin-treated
mice.

significantly increased the number of goblet cells in both NCD- Administration of A muciniphila improves insulin signalling
and HFD-fed mice (NCD-Met, 9.1±0.2; HFD-Met, 9.5±0.5; by inducing VAT-resident CD4 Foxp3 Tregs
p<0.001), indicating that metformin alters the goblet cell popu- Since we observed both a greater abundance of Akkermansia and
lation regardless of metabolic profile or dietary composition. In an increase in the number of goblet cells in HFD-Met mice, we
addition, Pearson correlation analysis showed that the number next investigated whether a selective increase in the Akkermansia
of goblet cells was positively correlated with the abundance of population had any effect on the metabolic profile or the number
Akkermansia (figure 4F). of goblet cells in HFD-fed mice. Mice were fed a HFD for

Figure 4 The goblet cell population


is affected by metformin treatment.
(A–D) Representative light
photomicrographs showing ileal
sections stained with periodic acid–
Schiff (PAS)/Alcian blue. (A) NCD-CT
mice. (B) NCD-Met mice. (C) HFD-CT
mice. (D) HFD-Met mice. (E) Changes
in goblet cells (blue in A–D) are
expressed as means±SEM of the
number of PAS-positive goblet cells per
villus. The letters above the bars
denote statistically significant
differences ( p<0.05) between the
means (one-way analysis of variance
followed by Duncan’s post-hoc test).
(F) The relationship between the
relative abundance of Akkermansia
and the number of goblet cells
(Pearson’s correlation test; r=0.4996,
p=0.02). NCD-CT, normal-chow diet
(NCD)-fed control mice; NCD-Met,
NCD-fed metformin-treated mice;
HFD-CT, high-fat diet (HFD)-fed control
mice; HFD-Met, HFD-fed
metformin-treated mice.

Shin N-R, et al. Gut 2013;0:1–9. doi:10.1136/gutjnl-2012-303839 5


Gut microbiota

Figure 5 Akkermansia administration


improves glucose tolerance and
modulates the number of goblet cells
in HFD-fed mice. (A) Changes in goblet
cells are expressed as the number of
periodic acid–Schiff (PAS)-positive
goblet cells per villus. (B)
Representative light photomicrographs
showing ileal sections stained with
PAS/Alcian blue. (C) Area under the
curve (AUC) obtained after the glucose
tolerance test (GTT). The results are
representative of three independent
experiments. All data are expressed as
the mean±SEM (n=6 mice/group). p
Values were determined using a
one-tailed unpaired t test. *p<0.05;
**p<0.01; ***p<0.001. NCD-CT,
normal-chow diet (NCD)-fed control
mice; HFD-CT, high-fat diet (HFD)-fed
control mice; HFD-Met, HFD-fed
metformin-treated mice; HFD-Akk,
HFD-fed Akkermansia-administered
mice.

4 weeks, after which they were given 4.0×108 cfu of A mucini- Because changes in intestinal permeability cannot explain the
phila orally every day for a further 6 weeks. Colonisation of improved metabolic profile seen after Akkermansia administra-
Akkermansia was confirmed by calculating the relative fractional tion, we next examined the possibility that Akkermansia might
abundance of Akkermansia by quantitative PCR. Despite differ- exert an antidiabetic effect by reducing SVF inflammation in the
ences in the initial abundance of Akkermansia between HFD-CT VAT, which has been implicated in the aetiology of insulin resist-
and HFD-Akk mice, Akkermansia treatment prevented any ance.21 40 FACS analysis showed a significant increase in the
decrease in abundance in HFD-fed mice as they aged (see online proportion of M1 macrophages and a decrease in the propor-
supplementary figure S4). Consistent with the positive correl- tion of M2 macrophages in the VAT of HFD-CT mice compared
ation between the number of goblet cells and the abundance of with that in NCD-CT mice. However, M1/M2 macrophage
Akkermansia, supplementation with Akkermansia restored both polarisation in the SVF of HFD-fed mice was not affected by
the number ( per villus) and density ( per unit surface area) of Akkermansia or metformin (see online supplementary figure
goblet cells in HFD-fed mice (figure 5A–B and online supple- S8). In addition, we also observed a significant difference in the
mentary figure S5). CD4/CD8 T cell ratio in the SVF of NCD-CT and HFD-CT
Furthermore, HFD-Akk mice showed a significant improve- mice; however, the CD4/CD8 T cell ratio in the SVF of
ment in glucose tolerance ( p<0.01) (figure 5C), similar to that HFD-fed mice was not affected by Akkermansia or metformin
seen in HFD-Met mice. This indicates that Akkermansia has (see online supplementary figure S9).
the potential to ameliorate the glucose tolerance induced by a We next examined the number of Tregs, which are critical
HFD. However, treatment with a lower dose (4.0×106 cfu) of regulators of immune or inflammatory responses in the adipose
live Akkermansia or with the same dose of autoclaved (dead) tissues of obese animals. We observed a significant reduction in
cells did not ameliorate the impaired glucose tolerance (see the proportion of Tregs in the SVF from the VAT of HFD-CT
online supplementary figure S6), suggesting that the bacteria mice compared with that in NCD-CT mice (p<0.05) (figure 6A
must be metabolically active and used at a dose > 4.0×107 cfu and C), consistent with previous reports.23 Interestingly, admin-
to exert their beneficial effect. We next studied the effect of istration of Akkermansia to HFD-fed mice restored the Treg
Akkermansia on metabolic hormones. As expected, metformin proportion to a level comparable to that in NCD-CT mice
significantly reduced the increases in serum insulin and leptin (NCD-CT, 49.5±2.3; HFD-CT, 32.5±5.7; HFD-Akk, 52.47
levels induced by a HFD ( p<0.001 for insulin; p<0.05 for ±2.176; p<0.01; figure 6A and C). There was also a significant
leptin) (see online supplementary figure S7A and B), an effect increase in the overall number of Tregs within the SVF in
that is probably secondary to improvements in glucose homeo- HFD-Met and HFD-Akk mice compared with that in HFD-CT
stasis. Similarly, HFD-Akk mice showed reduced serum concen- mice (figure 6B). When we examined the expression of proin-
trations of insulin and leptin compared with HFD-CT mice, flammatory cytokines that play a direct role in tissue inflamma-
although the differences were not statistically significant (see tion and insulin resistance, we found that the expression of
online supplementary figure S7A and B). The HFD-induced interleukin (IL)-6 and IL-1β mRNA was significantly increased
increases in serum lipopolysaccharide (LPS) levels reduced by in HFD-CT mice compared with that in NCD-CT mice;
metformin or Akkermansia, suggesting that the decreased endo- however, after treatment with Akkermansia, the expression of
toxaemia seen in HFD-Akk mice might be related to improved IL-6 and IL-1β was similar to that in NCD-CT mice (figure 6D
glucose tolerance. However, the differences were not statistic- and E). Thus, a single bacterial species, A muciniphila, can
ally significant compared with HFD-CT mice (see online sup- attenuate VAT inflammation by inducing the generation of Tregs
plementary figure S7C). Furthermore, neither metformin nor and suppressing the production of proinflammatory cytokines in
Akkermansia affected intestinal permeability in HFD-fed mice the SVF, thereby leading to increased insulin signalling in
(see online supplementary figure S7D). HFD-fed mice.

6 Shin N-R, et al. Gut 2013;0:1–9. doi:10.1136/gutjnl-2012-303839


Gut microbiota

Figure 6 The induction of Tregs and


a reduction in the levels of
proinflammatory cytokines attenuate
visceral adipose tissue (VAT)
inflammation in Akkermansia
treatment to HFD-fed mice. (A)
Percentage of Foxp3 Tregs within the
CD3 CD4 T cell population in the
stromal vascular fraction (SVF) of the
VAT. (B) Absolute number of CD3 CD4
Foxp3 Tregs per million cells in the SVF
of the VAT. (C) Representative
fluorescence activated cell sorting plots
showing the gated Foxp3 Tregs within
the total SVF CD4 T cell population in
the VAT of each mouse. (D and E)
Relative expression of interleukin (IL)-6
and IL-1β mRNA in the VAT. All data
are expressed as the mean±SEM (n=6
mice/group). p Values were determined
using a one-tailed unpaired t test.
*p<0.05; **p<0.01; ***p<0.001.
NCD-CT, normal-chow diet (NCD)-fed
control mice; HFD-CT, high-fat diet
(HFD)-fed control mice; HFD-Met,
HFD-fed metformin-treated mice;
HFD-Akk, HFD-fed
Akkermansia-administered mice.

DISCUSSION reduction in the AUC over that seen in HFD-Abx mice.


The gut microbiota is highly associated with obesity and T2D.8–10 33 However, both HFD-Abx and HFD-Abx+Met mice showed a
Metformin is one of the most common drugs used to treat patients significant improvement in glucose tolerance compared with
with T2D.25 41 Here, we used a metagenomic approach to demon- HFD-CT mice. Accordingly, recent studies suggest that antibio-
strate a role for metformin in modulating the composition of the gut tics improve glucose homeostasis of HFD-fed mice.43 44
microbiota in diet-induced obese and diabetic C57BL/6 mice. Furthermore, it is not possible to test whether the antidiabetic
Consistent with previous findings,7 8 10 42 a HFD led to dysbiosis, effects of metformin are abrogated in germ-free animals because
increased fasting glucose levels and glucose intolerance. Metformin HFD-induced dysbiosis is likely to be responsible for impaired
treatment significantly improved the hyperglycaemia in HFD-CT glucose homeostasis.4 6 Despite this, our study supports the
mice; however, the gut microbiota of HFD-Met mice showed an hypothesis that changes in the composition of the gut micro-
altered microbial composition, which was distinct from that in biota induced by antidiabetic drugs contribute to the improved
HFD-CT or NCD-CT mice. Based on these results, we suggest that glucose homeostasis seen in diabetic mice.
the gut microbiota might be a contributing factor to the glucose- Recently, several studies have highlighted the effects of mucus
lowering effect of metformin. degradation on host physiology.45 46 The mucin-degrading bac-
Depending on the diet fed to the mice, metformin had differ- terium, A muciniphila, which belongs to the phylum
ent effects on microbial composition. The gut microbial com- Verrucomicrobia, is more abundant in the mucosa of healthy
munity in HFD-Met mice was distinct from that in NCD-CT subjects than in that of diabetic patients or animals.47–50 Also,
and NCD-Met mice (figure 2). Specifically, NSC analysis identi- the colonisation of germ-free mice with A muciniphila led to
fied several genera that contributed to the observed differences altered host mucosal transcriptome profiles, which showed
in the distribution of bacterial communities between the four balanced immune responses indicative of tolerance to com-
groups (figure 3). Metformin offset the changes in the propor- mensal bacteria.46 We observed that a HFD reduced the abun-
tions of several genera that were induced by a HFD. For dance of Akkermansia, suggesting an unhealthy state in these
example, the significant reductions in the proportions of mice. The abundance of Akkermansia in HFD-Met mice was,
Akkermansia and Alistipes and the increases in the proportions however, significantly higher than that in HFD-CT mice, sug-
of Anaerotruncus, Lactococcus, Parabacteroides, Odoribacter, gesting increased caecal mucin levels.45 In this regard, we
Lawsonia, Blautia and Lactonifactor, seen in HFD-CT mice, observed a greater number of mucin-producing goblet cells in
were reversed by metformin. Consequently, the proportions of both NCD- and HFD-fed mice after metformin treatment. We
these genera were similar in HFD-Met and NCD-CT mice (the also noted a positive correlation between the abundance of
non-diabetic phenotype). Depleting the gut microbiota using a Akkermansia and the number of PAS-positive goblet cells (figure
combination of antibiotics before metformin treatment abro- 4). Additionally, the amelioration of glucose intolerance and an
gated the antidiabetic effects of the drug, as indicated by the increase in the goblet cell number were seen in HFD-Akk mice
finding that HFD-Abx+Met mice did not show any additional (figure 5). Akkermansia is a mucin-degrading bacterium and the

Shin N-R, et al. Gut 2013;0:1–9. doi:10.1136/gutjnl-2012-303839 7


Gut microbiota

proportion of Akkermansia correlates with an increase in the planned the experiments. N-RS, J-CL, M-SK, TWW, M-SL and J-WB wrote the
number of goblet cells; thus, an increase in the number of manuscript. M-SL and J-WB conceived and supervised the study.
goblet cells may underlie the improved glucose profiles seen Funding This work was supported by a grant from the Mid-Career Researcher
after Akkermansia administration. Program (2011–0028854 to J-WB) through the National Research Foundation of
Korea (NRF), funded by the Ministry of Education, Science and Technology (MEST)
In addition to producing components of the mucus layer, and by the Bio R&D Program (2008–04090 to M-SL). M-SL is the recipient of a
goblet cells also produce molecules that are associated with global research laboratory grant from the National Research Foundation of Korea
innate defence mechanisms, such as intestinal trefoil factor and (K21004000003-12A0500-00310).
resistin-like molecule β. Because these molecules augment barrier Competing interests None.
integrity,51 52 an increase in the number of goblet cells after Provenance and peer review Not commissioned; externally peer reviewed.
Akkermansia administration may attenuate glucose intolerance by
reducing LPS translocation across the intestinal barrier. However,
serum LPS levels and gut permeability were not significantly dif- REFERENCES
1 Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin
ferent in HFD-Akk and HFD-CT mice, suggesting that factors resistance and type 2 diabetes. Nature 2006;444:840–6.
other than changes in barrier function are responsible for the 2 Rokholm B, Baker JL, Sorensen TI. The levelling off of the obesity epidemic since
improved metabolic profile found in HFD-Akk mice. the year 1999–a review of evidence and perspectives. Obes Rev 2010;11:835–46.
Of the various T cell subsets, Tregs play a key role in control- 3 Wang Y, Beydoun MA, Liang L, et al. Will all Americans become overweight or
obese? estimating the progression and cost of the US obesity epidemic. Obesity
ling the classic adaptive immune responses and also the innate (Silver Spring) 2008;16:2323–30.
immune responses associated with obesity-induced chronic 4 Bäckhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor
inflammation.53 In this context, we observed a significant that regulates fat storage. Proc Natl Acad Sci USA 2004;101:15718–23.
decrease in the Treg population in HFD-CT mice compared with 5 Bäckhed F, Ley RE, Sonnenburg JL, et al. Host-bacterial mutualism in the human
intestine. Science 2005;307:1915–20.
NCD-CT mice. However, oral administration of Akkermansia to
6 Bäckhed F, Manchester JK, Semenkovich CF, et al. Mechanisms underlying the
HFD-fed mice restored both the percentage and absolute resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci USA
number of Tregs within the total CD4 T cell population in the 2007;104:979–84.
VAT, which was similar to the effect of metformin treatment 7 Cani PD, Bibiloni R, Knauf C, et al. Changes in gut microbiota control metabolic
(figure 6A–C). These results provide evidence that a selective endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in
mice. Diabetes 2008;57:1470–81.
increase in the Akkermansia population improves the metabolic 8 Ley RE, Bäckhed F, Turnbaugh P, et al. Obesity alters gut microbial ecology. Proc
profile of individuals with diet-induced obesity via a mechanism Natl Acad Sci USA 2005;102:11070–5.
that is mediated by the anti-inflammatory activity of VAT-resident 9 Ley RE, Turnbaugh PJ, Klein S, et al. Microbial ecology: human gut microbes
Tregs. In addition, the increases in IL-6 and IL-1β mRNA expres- associated with obesity. Nature 2006;444:1022–3.
10 Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome
sion seen in the VAT after a HFD were abolished by Akkermansia
with increased capacity for energy harvest. Nature 2006;444:1027–31.
treatment (figure 6D and E). This is consistent with the findings 11 Tilg H. Obesity, metabolic syndrome and microbiota: multiple interactions. J Clin
of previous studies suggesting that Akkermansia has anti- Gastroenterol 2010;44(Suppl 1):S16–18.
inflammatory activity.48 54 Recent papers report that goblet cells 12 Dethlefsen L, Huse S, Sogin ML, et al. The pervasive effects of an antibiotic on the
affect intestinal immune homeostasis by delivering antigens from human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol
2008;6:e280.
the lumen to tolerogenic dendritic cells, which then induce 13 Jumpertz R, Le DS, Turnbaugh PJ, et al. Energy-balance studies reveal associations
Tregs.55 Thus, the increase in the number of goblet cells after between gut microbes, caloric load and nutrient absorption in humans. Am J Clin
Akkermansia treatment might also induce Tregs; however, the Nutr 2011;94:58–65.
increased goblet cell population does not appear to modulate sys- 14 Turnbaugh PJ, Bäckhed F, Fulton L, et al. Diet-induced obesity is linked to marked
but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe
temic LPS or intestinal permeability. Also, the increased goblet
2008;3:213–23.
cell population might expedite tolerogenic dendritic cell 15 Vijay-Kumar M, Aitken JD, Carvalho FA, et al. Metabolic syndrome and altered gut
responses to Akkermansia under HFD conditions and dendritic microbiota in mice lacking Toll-like receptor 5. Science 2010;328:228–31.
cell-augmented Treg activity might suppress inflammatory 16 Guilherme A, Virbasius JV, Puri V, et al. Adipocyte dysfunctions linking obesity to
changes in adipose tissues, leading to increased glucose tolerance. insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol 2008;9:367–77.
17 Xu H, Barnes GT, Yang Q, et al. Chronic inflammation in fat plays a crucial role in
In summary, our results show that metformin treatment the development of obesity-related insulin resistance. J Clin Invest
induced an unexpected change in the composition of the gut 2003;112:1821–30.
microbiota in mice receiving a HFD, which may constitute a new 18 Winer S, Winer DA. The adaptive immune system as a fundamental regulator of
mechanism for the antidiabetic activity of metformin. Moreover, adipose tissue inflammation and insulin resistance. Immunol Cell Biol
2012;90:755–62.
these findings reinforce the concept that changes in the gut
19 Furuhashi M, Fucho R, Gorgun CZ, et al. Adipocyte/macrophage fatty acid-binding
microbial community, aimed at increasing the Akkermansia popu- proteins contribute to metabolic deterioration through actions in both macrophages
lation, can rescue the glucose intolerance associated with T2D. and adipocytes in mice. J Clin Invest 2008;118:2640–50.
Although it is still unclear which of the molecular mechanisms or 20 Elgazar-Carmon V, Rudich A, Hadad N, et al. Neutrophils transiently infiltrate
interactions associated with Akkermansia (and which govern intra-abdominal fat early in the course of high-fat feeding. J Lipid Res
2008;49:1894–903.
glucose homeostasis) are involved in increasing the goblet cell 21 Weisberg SP, McCann D, Desai M, et al. Obesity is associated with macrophage
and Treg populations, our pharmacological intervention study accumulation in adipose tissue. J Clin Invest 2003;112:1796–808.
provides circumstantial evidence that modulation of the gut 22 Liu G, Ma H, Qiu L, et al. Phenotypic and functional switch of macrophages
microbial community improves the health of patients with T2D. induced by regulatory CD4+CD25+ T cells in mice. Immunol Cell Biol
2011;89:130–42.
It also supports the suggestion that Akkermansia has a potential
23 Feuerer M, Herrero L, Cipolletta D, et al. Lean, but not obese, fat is enriched for a
role as a probiotic with antidiabetic effects. It would be interest- unique population of regulatory T cells that affect metabolic parameters. Nat Med
ing to undertake a follow-up study to identify the immunomodu- 2009;15:930–9.
latory molecule(s) and/or soluble factors produced by 24 Cusi K, DeFronzo RA. Metformin: A review of its metabolic effects. Diabetes
Akkermansia and their related interactions. Reviews 1998;6:89–131.
25 Donnelly LA, Doney AS, Hattersley AT, et al. The effect of obesity on glycaemic
response to metformin or sulphonylureas in Type 2 diabetes. Diabet Med
Contributors N-RS, J-CL and H-YL contributed equally to this work. N-RS, J-CL 2006;23:128–33.
and H-YL performed the experiments and data analysis. N-RS, J-CL, M-SL and J-WB 26 Witters LA. The blooming of the French lilac. J Clin Invest 2001;108:1105–7.

8 Shin N-R, et al. Gut 2013;0:1–9. doi:10.1136/gutjnl-2012-303839


Gut microbiota

27 Shaw RJ, Lamia KA, Vasquez D, et al. The kinase LKB1 mediates glucose 43 Carvalho BM, Guadagnini D, Tsukumo DM, et al. Modulation of gut microbiota by
homeostasis in liver and therapeutic effects of metformin. Science antibiotics improves insulin signalling in high-fat fed mice. Diabetologia
2005;310:1642–6. 2012;55:2823–34.
28 He L, Sabet A, Djedjos S, et al. Metformin and insulin suppress hepatic 44 Membrez M, Blancher F, Jaquet M, et al. Gut microbiota modulation with
gluconeogenesis through phosphorylation of CREB binding protein. Cell norfloxacin and ampicillin enhances glucose tolerance in mice. FASEB J
2009;137:635–46. 2008;22:2416–26.
29 Zhou G, Myers R, Li Y, et al. Role of AMP-activated protein kinase in mechanism of 45 Van den Abbeele P, Gerard P, Rabot S, et al. Arabinoxylans and inulin differentially
metformin action. J Clin Invest 2001;108:1167–74. modulate the mucosal and luminal gut microbiota and mucin-degradation in
30 Bailey CJ, Mynett KJ, Page T. Importance of the intestine as a site of humanized rats. Environ Microbiol 2011;13:2667–80.
metformin-stimulated glucose utilization. Br J Pharmacol 1994;112:671–5. 46 Derrien M, Van Baarlen P, Hooiveld G, et al. Modulation of Mucosal Immune
31 Wilcock C, Bailey CJ. Accumulation of metformin by tissues of the normal and Response, Tolerance and Proliferation in Mice Colonized by the Mucin-Degrader
diabetic mouse. Xenobiotica 1994;24:49–57. Akkermansia muciniphila. Front Microbiol 2011;2:166.
32 Bailey CJ, Wilcock C, Scarpello JHB. Metformin and the intestine. Diabetologia 47 Liou AP, Paziuk M, Luevano JM Jr., et al. Conserved shifts in the gut microbiota
2008;51:1552–3. due to gastric bypass reduce host weight and adiposity. Sci Transl Med
33 Larsen N, Vogensen FK, van den Berg FW, et al. Gut microbiota in human adults 2013;5:178ra41.
with type 2 diabetes differs from non-diabetic adults. PLoS One 2010;5:e9085. 48 Png CW, Linden SK, Gilshenan KS, et al. Mucolytic bacteria with increased
34 Qin J, Li Y, Cai Z, et al. A metagenome-wide association study of gut microbiota in prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria.
type 2 diabetes. Nature 2012;490:55–60. Am J Gastroenterol 2010;105:2420–8.
35 Cai D, Yuan M, Frantz DF, et al. Local and systemic insulin resistance resulting from 49 Santacruz A, Collado MC, Garcia-Valdes L, et al. Gut microbiota composition is
hepatic activation of IKK-beta and NF-kappaB. Nat Med 2005;11:183–90. associated with body weight, weight gain and biochemical parameters in pregnant
36 Kim DH, Lee JC, Kim S, et al. Inhibition of autoimmune diabetes by TLR2 tolerance. women. Br J Nutr 2010;104:83–92.
J Immunol 2011;187:5211–20. 50 Swidsinski A, Dorffel Y, Loening-Baucke V, et al. Acute appendicitis is characterised
37 Koren O, Spor A, Felin J, et al. Human oral, gut and plaque microbiota in patients by local invasion with Fusobacterium nucleatum/necrophorum. Gut 2011;60:34–40.
with atherosclerosis. Proc Natl Acad Sci USA 2011;108(Suppl 1):4592–8. 51 Artis D, Wang ML, Keilbaugh SA, et al. RELMbeta/FIZZ2 is a goblet cell-specific
38 Knights D, Kuczynski J, Koren O, et al. Supervised classification of microbiota immune-effector molecule in the gastrointestinal tract. Proc Natl Acad Sci USA
mitigates mislabeling errors. ISME J 2011;5:570–3. 2004;101:13596–600.
39 Derrien M, Vaughan EE, Plugge CM, et al. Akkermansia muciniphila gen. nov., sp. 52 Suemori S, Lynch-Devaney K, Podolsky DK. Identification and characterization of rat
nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol intestinal trefoil factor: tissue- and cell-specific member of the trefoil protein family.
2004;54:1469–76. Proc Natl Acad Sci USA 1991;88:11017–21.
40 Nishimura S, Manabe I, Nagasaki M, et al. CD8+ effector T cells contribute to 53 Cipolletta D, Kolodin D, Benoist C, et al. Tissular T(regs): a unique population of
macrophage recruitment and adipose tissue inflammation in obesity. Nat Med adipose-tissue-resident Foxp3+CD4+ T cells that impacts organismal metabolism.
2009;15:914–20. Semin Immunol 2011;23:431–7.
41 Bailey CJ, Turner RC. Metformin. N Engl J Med 1996;334:574–9. 54 Hansen CH, Holm TL, Krych L, et al. Gut microbiota regulates NKG2D ligand
42 Everard A, Lazarevic V, Derrien M, et al. Responses of gut microbiota and glucose expression on intestinal epithelial cells. Eur J Immunol 2013;43:447–57.
and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant 55 McDole JR, Wheeler LW, McDonald KG, et al. Goblet cells deliver luminal antigen
mice. Diabetes 2011;60:2775–86. to CD103+ dendritic cells in the small intestine. Nature 2012;483:345–9.

Shin N-R, et al. Gut 2013;0:1–9. doi:10.1136/gutjnl-2012-303839 9


View publication stats

You might also like