scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Metformin Is Associated With Higher Relative Abundance of Mucin-Degrading Akkermansia muciniphila and Several Short-Chain Fatty Acid-Producing Microbiota in the Gut.

TL;DR: The hypothesis that metformin shifts gut microbiota composition through the enrichment of mucin-degrading A. muciniphila as well as several SCFA-producing microbiota is supported.
Abstract: OBJECTIVE Recent studies suggest the beneficial effects of metformin on glucose metabolism may be microbially mediated. We examined the association of type 2 diabetes, metformin, and gut microbiota in community-dwelling Colombian adults. On the basis of previous research, we hypothesized that metformin is associated with higher levels of short-chain fatty acid (SCFA)–producing and mucin-degrading microbiota. RESEARCH DESIGN AND METHODS Participants were selected from a larger cohort of 459 participants. The present analyses focus on the 28 participants diagnosed with diabetes—14 taking metformin— and the 84 participants without diabetes who were matched (3-to-1) to participants with diabetes by sex, age, and BMI. We measured demographic information, anthropometry, and blood biochemical parameters and collected fecal samples from which we performed 16S rRNA gene sequencing to analyze the composition and structure of the gut microbiota. RESULTS We found an association between diabetes and gut microbiota that was modified by metformin use. Compared with participants without diabetes, participants with diabetes taking metformin had higher relative abundance of Akkermansia muciniphila, a microbiota known for mucin degradation, and several gut microbiota known for production of SCFAs, including Butyrivibrio, Bifidobacterium bifidum, Megasphaera, and an operational taxonomic unit of Prevotella. In contrast, compared with participants without diabetes, participants with diabetes not taking metformin had higher relative abundance of Clostridiaceae 02d06 and a distinct operational taxonomic unit of Prevotella and a lower abundance of Enterococcus casseliflavus. CONCLUSIONS Our results support the hypothesis that metformin shifts gut microbiota composition through the enrichment of mucin-degrading A. muciniphila as well as several SCFA-producing microbiota. Future studies are needed to determine if these shifts mediate metformin’s glycemic and anti-inflammatory properties.

Content maybe subject to copyright    Report

Metformin Is Associated With
Higher Relative Abundance of
Mucin-Degrading
Akkermansia
muciniphila
and Several Short-
Chain Fatty AcidProducing
Microbiota in the Gut
Diabetes Care 2017;40:5462 | DOI: 10.2337/dc16-1324
OBJECTIVE
Recent studies suggest the beneci al effects of metform in on glucose metabolism
may be microbially mediated. We examined the association of type 2 diabetes,
metformin, and gut microbiota in community-dwelling Colombian adults. On the
basis of previous research, we hypothesized that metformin is associ ated with
higher levels of short-chain fatty acid (SCFA)producing and mucin-degrading
microbiota.
RESEARCH DESIGN AND METHODS
Parti cipants were selected from a larger cohor t of 459 partici pants. The present
analyses focus on the 28 participan ts diagnosed with diabetes d14 taking
metformind and t he 8 4 participan ts withou t diab etes who were match ed
(3-t o-1) t o participants w ith diabetes by sex, age, and BMI. We measured de-
mographic information, anthropomet ry, and blood biochemical par ameters and
collected fecal samples from which we performed 16S rRNA gene sequencing to
analyze the composition and structure of the gut microbiota.
RESULTS
We f ound an associat ion between diabetes and gut microbiota that wa s modied
by metformin use. Compared with participants witho ut diabetes, par ticipant s
with diabetes taking metformin had higher relative abundance of Akkermansia
muciniphila, a micr obiota known for mucin degradati on, and several gut micro-
biota known for production of SCFAs, including Butyrivibrio, Bidobacterium
bidum, Megasphaera, and an operational taxonomic unit of Prevotella.In
contrast, compared with participants without diabetes, participants with
diabetes not taking metformin had higher relative abundance of Clostridiaceae
02d06 and a distinct operational taxonomic unit of Prevotella and a lower
abundance of Enterococcus casseliavus.
CONCLUSIONS
Our re sults support the hypothesis that metformin shifts gut microbiota compo-
sition through the enrichment of mucin-degrading A. muciniphila as well as sev-
eral SCFA-producing microbiota. Future studies are needed to determine if these
shifts mediate metformi ns glycemic and anti-inammatory properties.
1
VidariumdNutrition, Health and Wellness Re-
search Center, Grupo Empresarial Nutresa, Me-
dellin, Colombia
2
Department of Epidemiology, Johns Hopkins
Bloomberg School of Public Health, Baltimore,
MD
3
Din
´
amica I.P.S.dEspecialista en Ayudas Diag-
n
´
osticas, Medellin, Colombia
4
EPS y Medicina Prepagada Suramericana S.A.,
Medellin, Colombia
Corresponding author: J uan S. Escobar, jsescobar@
serviciosnutresa.com.
Received 20 June 2016 and accepted 27 Septem-
ber 2016.
This article contains Supplementary Data online
at http ://care.diabetesjournals.org/lookup/
suppl/doi:10.2337/dc16-1324/-/DC1.
© 2017 by the American Diabetes Association.
Readers may use this article as long as the work
is properly cited, the use is educational and not
for prot, and the work is not altered. More infor-
mation is available at http://www.diabetesjournals
.org/content/license.
Jacobo de la Cuesta-Zuluaga,
1
Noel T. Mueller,
2
Vanessa Corrales-Agudelo,
1
Elian a P. Vel
´
asquez-Mej
´
ıa,
1
Jenny A. Carmona,
3
Jos
´
e M. Abad,
4
and
Juan S. Escobar
1
54 Diabetes Care Volume 40, Ja nuary 2017
EMERGING TECHNOLOGIES AND THERAPEUTICS
Downloaded from http://diabetesjournals.org/care/article-pdf/40/1/54/519215/dc161324.pdf by guest on 26 August 2022

Microbial communities (microbiota) and
their associated genes (microbiome)
constitute the interface of our environs
and our cells, and the ir composition is
believed to p lay a deterministic role in
human health and disease. In particular,
the development of typ e 2 diab etes, a
disease rising in prevalence around the
globe, has been linked in nonhuman (1)
and human (25) studies to imbalances
in microbiota of the intestinal tract
(gut). However, the most recent human
study on this topic found that the asso-
ciation was modulated in a potent-
ially b enecial manner by metformin
treatment (6).
Metformin (1,1-dimethylbiguanide
hydrochloride) is the most frequent
medication used to treat type 2 diabet es
(7), and ndings from recent studies
suggest it may also prevent cancer (7)
and cardiovascular events (8). Metfor-
min h as pleiotropic effects, yet the
majority of mechanistic studies have
focused on changes in liver function
(7,9,10). Although metformin certainly
alters hepatic glucose production via
effects on AMPK, there is growing evi-
dence that the ge nesis of its action is in
the gut (1115).
Metformin is ;50% bioavailable , al-
lowing for near-equal intestinal and
plasma exposure, but intestinal accumu-
lation of metformin is 300 times that of
the plasma (16), making the gut the pri-
mary reservoir for metformin in humans.
Unlike oral administration, intravenous
administration of metformin in humans
does not improve glycemia (14). More-
over, in mice, oral admi nistration of a
broad-spectrum antibiotic cocktail with
oral metformin ab rogates metformins
glucose- lowering effect (12). Providing
yet further evidence that the glucose-
lowering effect of metformin may originate
in the lower bowel, a delayed-release oral
metformin, which targets the ileum,
had a similar or greater glucose-lowering
effect than immediate-release or extended-
release metformin, despite the delayed-
release metformin having lower systemic
exposure than the other metformin for-
mulations (15).
Recent studies in animal s (11,12,13)
and hu mans (6,17) provide evidence
that metformin may partially restore
gut dysbiosis associated with type 2 di-
abetes. In mice fed a high-fa t diet, met-
formin treatment increased the relative
abundance of Akkermansia muciniphila
(1113), a m ucin-degrading bac teria
that has been shown to rev erse meta -
bolic disorders (1,12). In humans, partic-
ipants with diabetes taki ng metformin
had similar abundance of Subdoligranu-
lum and, to some extent, Akkermansia
compared with control subjects without
diabetes, suggesting that metformin
may help ameliorate a type 2 diabetes
associated gut microbiome (6). It has also
been shown that people with diabetes
taking metformin had a higher relative
abundance of Adlercreutzia (17), and
metagenomic functiona l analyses dem-
onstrated signicantly enhanced butyrate
and propionate production in people with
diabetes using metformin (6). In contrast,
people with diabetes who were not treat-
ed with metformin had a higher abun-
dance of Eubacterium and Clostridiaceae
SMB53 (17) and lower levels of short-
chain fatty acid (SCFA) producers, such
as Roseburia, Subdoligranulum,anda
cluster of butyrate-producing Clostri-
diales ( 6). These ndings provide evi-
dence that gut microbes may contribute
to the antidiabetes effects of metformin
through pathways that include mucin
degradation and SCFA production.
In this study we aimed to test the gen-
eralizability of previous observations con-
cerning the inuence of metformin on the
association of type 2 diabetes and gut
dysbiosis in a Colombian adult popula-
tion. Given the considerable variation in
the microbiota associated with type 2 di-
abetes and that the gut microbiota of Co-
lombians is different to t hat of other
populations (18), we hypothesized that
the microbial taxa involved in the type 2
diabetes dysbiosis of Colombians are dif-
ferent to those observed in Chinese and
European populations (2,3) but that the
effect of metformin is similar, i.e., through
enrichment of mucin-degrading and SCFA-
producing microbiota.
RESEARCH DESIGN AND METHODS
Study Design
Between July and November 2014, we en-
rolled 459 men and women 1862 years
old, with BMI $18.5 kg/m
2
, living in
the Colombian cities of Medellin, Bogota,
Barranquilla, Bucaramanga, and Cali. All
participants enrolled in the study were in-
sured by the health insurance provider
EPS y Medicina Prepagada Suramericana
S.A. (EPS SURA). We excluded pregnant
women, individuals who consumed anti-
biotics or antiparasitics ,3 months prior
to enrollment, and individuals diagnosed
with Alzhe imer disease, Parkinson dis-
ease, or any other neurodegenerative dis-
eases; current or recent cancer (,1year);
and gastrointestinal diseases (Crohn dis-
ease, ulcerative colitis, short bowel syn-
drome, diverticulosis, or celiac disease).
This study was conducted in accor-
dance with the principles of the Decla-
ration of Helsinki, as revised in 2 008, and
had minimal risk according to the
Colombian Ministry of Health (Resolution
008430 of 1993). All of the partici-
pants were thoroughly informed about
the study and p rocedures. Participants
were assured of anonymity and con-
dentiality. Written informed consent was
obtained from all the participants before
beginning the study. The Bioethics
Committee of Sede de Investigaci
´
on
UniversitariadUniversity of Antioquia re-
viewed the protocol and the consent
forms and approved the procedures de-
scribed here (approbation act 1424588
dated 28 May 2014).
Anthropometric, Clinical, and Dietary
Evaluations
We calculated BMI as weight (kg)/height
squared (m
2
) to clas sify participants as
lean (18.5 # BMI , 25.0 kg/m
2
), over-
weight (25.0 # BMI , 30.0 kg/m
2
), or
obese (BMI $30 kg/m
2
). In addition, val-
ues of HDL, LDL, VLDL, total cholester ol,
triglycerides, apolipoprotein B, fasting
glucose, glycated hemoglobin (HbA
1c
),
fasting insulin, adiponectin, an d hs-CRP
were obtained (collection and measure-
ment explained in Supplementary Data).
Dietary intake was evaluated through
24-h dietary recalls (see Supplementary
Data).
DNA Extraction and Sequence
Analysis
Each participant collect ed their own fe-
cal sample in a hermetically sealed, ster-
ile receptacle provided by t he research
team. Samples were immediately refrig-
erated in household freezers and brought
to an EPS SURA facility in each city within
12 h; receipt of samples occurred exclu-
sively in the morning (6
A.M.12 P.M.). As
such, stools were collected between the
evening of the day before and the morn-
ing of the day of sample receipt. Fecal
sampleswerestoredondryiceand
sent to a central laboratory via next-day
delivery. Before DNA e xtraction, stool
consistency was evaluated by trained lab-
oratory technicians.
care.d iabetesjournals.org de la Cuesta-Zuluaga and Associates 55
Downloaded from http://diabetesjournals.org/care/article-pdf/40/1/54/519215/dc161324.pdf by guest on 26 August 2022

Total microbial DNA was extracted
using the QIAamp DNA Stool Mini Kit
(Qiagen, Hilden, Germany) following
the manuf acturers instructions, with a
slight modication consisting in a bead-
beating step with the lysis buffer (20 s at
15 Hz using a stainless steel bead with a
5-mm diameter). After e xtraction, we
quantied DNA concentration using a
NanoDrop spectrophotometer (Nyxor
Biotech, Paris, France) and sent it to the
Microbial Systems Laboratory, University
of Michigan Medical School (Ann Arbor,
MI). The V4 hypervariable region of the
16S rRNA gene was amplied using the
F515 (59-CA CGGTCGKCGGCGCCATT-39)
and R806 (59-GGACTACHVGGGTWTC
TAAT-39) primers and sequenced usi ng
the Illumina MiSeq sequencing platform
with V2 chemistry and the dual-index
sequencing strategy (19). In addition to
DNA from fecal samples, we sequenced
negative controls (ultrapure water and
the QIAamp DNA Stool Mini KitsEBelu-
ti o n buffer), a DNA extraction blank,
and a mock community (HM-782D, BEI
Resources, Manassas, VA) in each instru-
ments run. Sequences were curated
following the MiSeq standard operating
procedure implemented by Mo thur
v.1.36 (20) (see Supplementary Data). Raw
sequences were deposited at the NCBI and
can be accessed through the BioProject
(accession number PRJNA325931).
Denition of Type 2 Diabetes and
Selection of Control Subjects
We identied 28 participants in our study
that had type 2 diabetes; 26 self-reported
physician-diagnosed diabetes prior to
the beginning of the study and 2 were
diagnosed through laboratory testing
(fasting blood glucose $126 mg/dL
and HbA
1c
$6.5%). Of the 28 participants
with type 2 diabetes, 14 were under met-
formin treatment, 14 were not (1 was
treated with insulin alone, 2 with gliben-
clamide, and 11 were under no pharmaco-
logical treatment for type 2 diabetes,
including the 2 participants unaware of
their diabetes status) (Suppleme ntary
Table 1).
We matched each participant with di-
abetes with three participants without
diabetes in our study based on sex, age
(to the cl osest possible age; maximum
difference between case and control
subjects 6 years; mean 1.5 years; median
1 year), and BMI category (lean, over-
weight, or obese). This left us with a total
analytic sample of 112 study participants
comprising 14 with type 2 diabetes using
metformin (T2D-met
+
), 14 with type 2 di-
abetes not using metformin (T2D-met
2
),
and 84 without diabetes (ND).
Statistical Analyses
Anthropometric and clinical variables
were compared across study groups us-
ing ANOVA and t tests after checking for
homoscedasticity and normal distrib u-
tion of residuals (using Fligner-Killeen
tests of homogeneity of variances and
Shapiro-Wilk normality tests). When
necessary, variables were appropriately
transformed ( natural log for uncon-
strained variables or arcsin square root
for proportions). Sex ratio and s tool con-
sistency were compared using x
2
tests.
Statistical analyses were performed
with R v.3.2.2 (21).
Curated DNA sequences ranged f rom
69 to 102,660 sequences per sample
(median 28,699). To limit the effects of
uneven sampling, we rareed the data
set to 4,091 sequences per sample, result-
ing in the exclusion of one T2D-met
2
par-
ticipant with 69 reads. Although rarefaction
may lead to missing low-abundance data, it
is a powerful way to reduce the likelihood
of detecting false positives, especially
among those operational taxonomic units
(OTUs) with very low abundance.
The gut microbiota structure and com-
position was assessed by quantifying and
interpreting similarities based on intra-
and intergroup diversity analyses (a and
b diversity, respectively). For a diversity,
we calculated Goods coverage and the
number of OTUs of each sample using
Mothur and constructed rarefaction
curves. We compared these indices among
groups of participants using analysis of
similarity (ANOSIM) with 1,000 permuta-
tions using the Vegan package of R (22).
b Diversity was assessed using
phylogeny-based generalized UniFrac dis-
tances (with the a parameter controlling
weight on abundant lineages = 0.5) calcu-
lated with the GUniFrac package of R (23).
For this, we rst reduced the alignment
and the OTU table to one representative
sequence per OTU, then obtained a dis-
tance matrix from uncorrected pairwise
distances between aligned sequences,
and nally constructed a relaxed neighbor-
joining phylogenetic tree using Mothur
and Clearcut. Comparisons among
groups of participants were performed
using the adonis function (ANOVA using
distance matrices) of the permutational
multivariate ANOVA (PERMANOVA) im-
plemented in the Vegan package of R (22).
We next used linear discriminant
analysis (LDA) e ffect size (LEfSe) (24) t o
agnostically identify microbial bio-
markers. LEfSe uses the nonparametric
factorial Krus kal-Wallis sum rank test to
detect individual OTUs with signicant
differential abundance among groups
of participants, then performs a set of
pairwise tests among groups of partici-
pants using the unpaired Wilcoxon rank
sum test, and nally uses LDA to esti-
mate the effect siz e of each differen-
tially abundant OTU (24). The strength
of LEfSe c ompared with standard stat is-
tical approaches is that, in addition to
prov iding P values, it provides an estima-
tion of the magnitude of the association
between each OTU and the grouping cat-
egories (e.g., metformin, type 2 diabetes)
through the LDA score. For stringency,
microbial biomarkers in our study were
retained if they had a P , 0.05 and a
(log10) LDA score $3, i.e., one order of
magnitude greater than LEfSesdefault.
Finally, across groups, we tested for dif-
ferences in relative abundance of the
mucin-degrading A. muciniphila and
major butyrate-producing microbial
genera, including Butyrivibrio, Ros eburia,
Subdoligranulum,andFaecalibacterium.
For th is analysis, we pooled all OTUs
classied in each of these phylotypes
(4 for A. muciniphila,11forButyrivibrio,
4forRoseburia,10forSubdoligranulum,
and 5 for Faecalibacterium) and tested for
differences using ANOVA and t tests on
arcsin square root transformed relative
abundances. T his pooling served to
examine whether differences in relative
abundance of these groups of bacteria
occurred across all OTUs or only in
specic OTUs.
Results from LEfSe and from the pooled
analysis of phylotypes were corrected for
multiple testing using the Bayesian ap-
proach implemented in the qvalue pack-
age of R (25). Tests were considered
signicant if they had a P value # 0.05
andaqvalue#0.1.
RESULTS
In Table 1 we present the characteristics
of T2D-met
+
, T2D-met
2
, and ND partic-
ipants. There were no statis tically signif-
icant differences (all P values . 0.10) in
demographic, anthropometric, or clini-
cal parameters between T2D-met
+
and
56 Type 2 Diabetes, Metformin, and Gut Microbiota Diabetes Care Volume 40, Ja nuary 2017
Downloaded from http://diabetesjournals.org/care/article-pdf/40/1/54/519215/dc161324.pdf by guest on 26 August 2022

Table 1General characteristics of T2D-met
+
, T2D-met
2
, and ND participants among community-dwelling Colombian adult s
Group P*
T2D-met
+
T2D-met
2
ND T2D-met
+
vs. T2D-met
2
T2D-met
+
vs. ND T2D-met
2
vs. ND
n 14 14 84 ddd
Age (years) 50 6 10 44 6 9476 9 0.11 0.33 0.24
Sex (F/M) 0.36 0.50 0.43 0.70 0.83 0.84
Anthropometry
BMI (kg/m
2
) 31.88 6 4.63 32.15 6 6.36 31.11 6 4.53 0.98 0.56 0.62
Body fat (%) 0.41 6 0.05 0.41 6 0.03 0.40 6 0.04 0.96 0.68 0.58
Waist circumference (cm) 104.6 6 9.8 102.8 6 14.2 102.0 6 11.3 0.70 0.39 0.85
Clinical parameters
Total cholesterol (mg/dL) 178 6 51 208 6 44 187 6 30 0.11 0.54 0.10
HDL (mg/dL) 40 6 11 38 6 6446 12 0.77 0.21 0.0136
LDL (mg/dL) 105 6 35 128 6 39 115 6 28 0.11 0.31 0.26
VLDL (mg/dL) 38 6 40 52 6 52 31 6 15 0.19 0.64 0.0471
Apolipoprotein B (mg/dL) 102 6 30 108 6 30 97 6 26 0.56 0.55 0.18
Triglycerides (mg/dL) 176 6 202 244 6 253 154 6 75 0.15 0.97 0.07
Fasting glucose (mg/dL) 127 6 47 145 6 76 90 6 10 0.57 0.0047 0.0060
HbA
1c
[% (mmol/mol)] 6.9 6 1.4 (52.0 6 15.3) 7.1 6 1.7 (54.0 6 18.6) 5.6 6 0.3 (38.0 6 3.3) 0.78 0.0026 0.0047
Fasting insulin (mU/mL) 22.24 6 12.58 24.24 6 12.86 15.20 6 8.79 0.49 0.11 0.0029
Insulin re sistance index 2.9 6 1.5 3.7 6 3.0 1.9 6 1.1 0.39 0.0420 0.0018
Leptin (ng/mL) 7.39 6 6.09 8.34 6 5.40 7.89 6 6.14 0.38 0.91 0.25
Adiponectin (mg/mL) 4.59 6 1.97 4.96 6 3.22 6.79 6 3.90 0.91 0.0195 0.07
hs-CRP (mg/L) 2.70 6 2.5 3 3.35 6 2.76 4.04 6 5.02 0.61 0.21 0.62
Dietary intake
Energy intake (calories) 1,843 6 268 1,865 6 584 1, 945 6 572 0.75 0.61 0.57
Carbohydrate (g) 250 6 37 260 6 86 267 6 88 0.92 0.52 0.68
Protein (g) 74.7 6 9.1 69.7 6 13.1 74.1 6 13.8 0.26 0.85 0.26
Fat (g) 60.7 6 13.0 60.2 6 21.3 62.9 6 17.1 0.64 0.76 0.51
Cholesterol (mg) 336 6 31 330 6 41 347 6 38 0.67 0.28 0.18
Dietary ber (g) 17.9 6 4.8 18.6 6 6. 4 17.5 6 4.8 0.88 0.73 0.64
Stool consistency [n (%)] 0.38 0.58 0.50
Hard 4 (28) 2 (14) 13 (15) ddd
Normal 8 (57) 9 (64) 54 (64) ddd
Mushy 2 (14) 1 (7) 13 (15) ddd
Diarrheic 0 (0) 2 (14) 4 (5) ddd
Values presented as mean 6 SD. *All P values from t tests except in sex and stool consistency (x
2
tests).
care.d iabetesjournals.org de la Cuesta-Zuluaga and Associates 57
Downloaded from http://diabetesjournals.org/care/article-pdf/40/1/54/519215/dc161324.pdf by guest on 26 August 2022

T2D-met
2
participants. Compared with
ND participants, T2D -met
+
participants
had higher fasting glucose, HbA
1c
,andin-
sulin resistance than ND participants and
lower levels of the insulin-sensitizing hor-
mone adiponectin (P , 0.05). No other
demographic, anthropometric, or clinical
parameters were statistically different.
16S rRNA Gene Sequencing
Gut microbiota communities were spe-
cic to each participant with marked in-
tersubject differences (Fig. 1A) (overall
interindividual generalized UniFrac dis-
tance = 0.720 6 0.009). We found high
coverage across all groups of participants
(mean Goods coverage 6 SD = 0.990 6
0.001); 99% of OTUs were detected at
least by two DNA reads, demonstrating
thorough sampling of the gut micro-
biota. We next tested for differences
in the number of observed OTUs across
the groups of participants. We found no
differences between participants with
diabetes and N D participants (ANOSIM
statistic R = 0.005, P value=0.425)or
between T2D-met
+
and T2D-met
2
partic-
ipants (ANOSIM statistic R = 20.018, P =
0.557). The number of observed OTUs
tended to be more similar between T2D-
met
+
and ND than between T2D-met
2
and ND participants (Supplementary Fig.
1); however, these differences were not
statistically signicant (T2D-met
+
vs. ND:
ANOSIM statistic R = 0.018, P = 0.348;
T2D-met
2
vs. ND: ANOSIM statistic R =
0.009, P =0.409).
We observed no signicant differences
in b diversity estimates among the three
groups of participants (PERMANOVA: R
2
=
0.019, P = 0.335) (Fig. 1A) or between
participants with diabetes and ND partic-
ipants (R
2
= 0.009, P = 0.416) (Fig. 1B).
However, the comparison between met-
formin and nonmetformin users reached
signicance (R
2
=0.013,P =0.036)(Fig.
1C), demonstrating differences in the
bacterial community structure associated
with metformin use. The difference was
also s ignicant when comparing T2D-
met
+
and ND participants (R
2
=0.015,
P = 0.036) but not when comparing
T2D-met
2
and ND participants (R
2
=
0.008, P = 0.943). These results suggested
the microbial communities of T2D-met
+
versus T2D-met
2
were modestly phylo-
genetically dissimilar.
We next used LEfSe to examine differ-
ences in the relative abundance of gut
microbiota at the OTU level. Note that
we were only interested in OTUs dis-
playing str ong a sso ciatio ns in the LDA
(represented by OTUs with [log10] LDA
scores $3); such st ringency resulted in
fewer retained, but more likely biologically
relevant, OTUs (19 displayed in Fig. 2 and
PCo2 (6.03%)
PCo1 (10.34%)
ND
R
2
= 0.019
P = 0.34
T2D-met
+
T2D-met
-
PCo2 (6.03%)
R
2
= 0.009
P = 0.42
ND
T2D
PCo1 (10.34%)
PCo2 (6.03%)
met-
R
2
= 0.013
P = 0.036
T2D-met
+
PCo1 (10.34%)
A
B
C
Figure 1Principal coordinates analysis based on generalized UniFrac. A: Comp aris on among the
three groups of participants. B: Comparison between participants with diabetes and ND part ici-
pants. C: Comparison between T2D-met
+
and participants not taking metformin (T2D-met
2
and
ND). Ellipses encompass 75% of data distribution in each group of parti cipants. R
2
and P values from
PERMANOVA.
58 Type 2 Diabetes, Metformin, and Gut Microbiota Diabetes Care Volume 40, Ja nuary 2017
Downloaded from http://diabetesjournals.org/care/article-pdf/40/1/54/519215/dc161324.pdf by guest on 26 August 2022

Citations
More filters
Journal ArticleDOI
TL;DR: How the gut microbiota and derived microbial compounds may contribute to human metabolic health and to the pathogenesis of common metabolic diseases are discussed, and examples of microbiota-targeted interventions aiming to optimize metabolic health are highlighted.
Abstract: Observational findings achieved during the past two decades suggest that the intestinal microbiota may contribute to the metabolic health of the human host and, when aberrant, to the pathogenesis of various common metabolic disorders including obesity, type 2 diabetes, non-alcoholic liver disease, cardio-metabolic diseases and malnutrition. However, to gain a mechanistic understanding of how the gut microbiota affects host metabolism, research is moving from descriptive microbiota census analyses to cause-and-effect studies. Joint analyses of high-throughput human multi-omics data, including metagenomics and metabolomics data, together with measures of host physiology and mechanistic experiments in humans, animals and cells hold potential as initial steps in the identification of potential molecular mechanisms behind reported associations. In this Review, we discuss the current knowledge on how gut microbiota and derived microbial compounds may link to metabolism of the healthy host or to the pathogenesis of common metabolic diseases. We highlight examples of microbiota-targeted interventions aiming to optimize metabolic health, and we provide perspectives for future basic and translational investigations within the nascent and promising research field. In this Review, Fan and Pedersen discuss how the gut microbiota and derived microbial compounds may contribute to human metabolic health and to the pathogenesis of common metabolic diseases, and highlight examples of microbiota-targeted interventions aiming to optimize metabolic health.

1,445 citations

Journal ArticleDOI
TL;DR: It is shown that metformin affected pathways with common biological functions in species from two different phyla, and many of the met formin-regulated genes in these species encoded metalloproteins or metal transporters, which provides support for the notion that altered gut microbiota mediates some of metformIn's antidiabetic effects.
Abstract: Metformin is widely used in the treatment of type 2 diabetes (T2D), but its mechanism of action is poorly defined. Recent evidence implicates the gut microbiota as a site of metformin action. In a double-blind study, we randomized individuals with treatment-naive T2D to placebo or metformin for 4 months and showed that metformin had strong effects on the gut microbiome. These results were verified in a subset of the placebo group that switched to metformin 6 months after the start of the trial. Transfer of fecal samples (obtained before and 4 months after treatment) from metformin-treated donors to germ-free mice showed that glucose tolerance was improved in mice that received metformin-altered microbiota. By directly investigating metformin-microbiota interactions in a gut simulator, we showed that metformin affected pathways with common biological functions in species from two different phyla, and many of the metformin-regulated genes in these species encoded metalloproteins or metal transporters. Our findings provide support for the notion that altered gut microbiota mediates some of metformin's antidiabetic effects.

1,022 citations

Journal ArticleDOI
22 Jun 2018-Gut
TL;DR: Recent evidence of the impact of the gut microbiota on metabolic disorders and focus on selected key mechanisms is discussed and the cases of the bacteria Prevotella copri and Akkermansia muciniphila will be discussed as key examples.
Abstract: The microbiome has received increasing attention over the last 15 years. Although gut microbes have been explored for several decades, investigations of the role of microorganisms that reside in the human gut has attracted much attention beyond classical infectious diseases. For example, numerous studies have reported changes in the gut microbiota during not only obesity, diabetes, and liver diseases but also cancer and even neurodegenerative diseases. The human gut microbiota is viewed as a potential source of novel therapeutics. Between 2013 and 2017, the number of publications focusing on the gut microbiota was, remarkably, 12 900, which represents four-fifths of the total number of publications over the last 40 years that investigated this topic. This review discusses recent evidence of the impact of the gut microbiota on metabolic disorders and focus on selected key mechanisms. This review also aims to provide a critical analysis of the current knowledge in this field, identify putative key issues or problems and discuss misinterpretations. The abundance of metagenomic data generated on comparing diseased and healthy subjects can lead to the erroneous claim that a bacterium is causally linked with the protection or the onset of a disease. In fact, environmental factors such as dietary habits, drug treatments, intestinal motility and stool frequency and consistency are all factors that influence the composition of the microbiota and should be considered. The cases of the bacteria Prevotella copri and Akkermansia muciniphila will be discussed as key examples.

846 citations

Journal ArticleDOI
TL;DR: The major principles underlying effects of dietary constituents on the gut microbiota are reviewed, resolving aspects of the diet–microbiota–host crosstalk, and the promises and challenges of incorporating microbiome data into dietary planning are presented.
Abstract: Since the renaissance of microbiome research in the past decade, much insight has accumulated in comprehending forces shaping the architecture and functionality of resident microorganisms in the human gut. Of the multiple host-endogenous and host-exogenous factors involved, diet emerges as a pivotal determinant of gut microbiota community structure and function. By introducing dietary signals into the nexus between the host and its microbiota, nutrition sustains homeostasis or contributes to disease susceptibility. Herein, we summarize major concepts related to the effect of dietary constituents on the gut microbiota, highlighting chief principles in the diet-microbiota crosstalk. We then discuss the health benefits and detrimental consequences that the interactions between dietary and microbial factors elicit in the host. Finally, we present the promises and challenges that arise when seeking to incorporate microbiome data in dietary planning and portray the anticipated revolution that the field of nutrition is facing upon adopting these novel concepts.

806 citations

Journal ArticleDOI
TL;DR: It is proposed that microbes and microbiomegnosy, or knowledge of the authors' gut microbiome, can become a novel source of future therapies as plants and its related knowledge have been the source for designing drugs over the last century.
Abstract: Metabolic disorders associated with obesity and cardiometabolic disorders are worldwide epidemic Among the different environmental factors, the gut microbiota is now considered as a key player interfering with energy metabolism and host susceptibility to several non-communicable diseases Among the next-generation beneficial microbes that have been identified, Akkermansia muciniphila is a promising candidate Indeed, A muciniphila is inversely associated with obesity, diabetes, cardiometabolic diseases and low-grade inflammation Besides the numerous correlations observed, a large body of evidence has demonstrated the causal beneficial impact of this bacterium in a variety of preclinical models Translating these exciting observations to human would be the next logic step and it now appears that several obstacles that would prevent the use of A muciniphila administration in humans have been overcome Moreover, several lines of evidence indicate that pasteurization of A muciniphila not only increases its stability but more importantly increases its efficacy This strongly positions A muciniphila in the forefront of next-generation candidates for developing novel food or pharma supplements with beneficial effects Finally, a specific protein present on the outer membrane of A muciniphila, termed Amuc_1100, could be strong candidate for future drug development In conclusion, as plants and its related knowledge, known as pharmacognosy, have been the source for designing drugs over the last century, we propose that microbes and microbiomegnosy, or knowledge of our gut microbiome, can become a novel source of future therapies

649 citations


Cites background from "Metformin Is Associated With Higher..."

  • ...Conversely, antidiabetic treatments, such as metformin administration and bariatric surgery were both found to be associated with a marked increase in the abundance of A. muciniphila (Figure 1) (Shin et al., 2014; Forslund et al., 2015; de la Cuesta-Zuluaga et al., 2017)....

    [...]

References
More filters
Journal Article
TL;DR: Copyright (©) 1999–2012 R Foundation for Statistical Computing; permission is granted to make and distribute verbatim copies of this manual provided the copyright notice and permission notice are preserved on all copies.
Abstract: Copyright (©) 1999–2012 R Foundation for Statistical Computing. Permission is granted to make and distribute verbatim copies of this manual provided the copyright notice and this permission notice are preserved on all copies. Permission is granted to copy and distribute modified versions of this manual under the conditions for verbatim copying, provided that the entire resulting derived work is distributed under the terms of a permission notice identical to this one. Permission is granted to copy and distribute translations of this manual into another language, under the above conditions for modified versions, except that this permission notice may be stated in a translation approved by the R Core Team.

272,030 citations

Journal ArticleDOI
TL;DR: M mothur is used as a case study to trim, screen, and align sequences; calculate distances; assign sequences to operational taxonomic units; and describe the α and β diversity of eight marine samples previously characterized by pyrosequencing of 16S rRNA gene fragments.
Abstract: mothur aims to be a comprehensive software package that allows users to use a single piece of software to analyze community sequence data. It builds upon previous tools to provide a flexible and powerful software package for analyzing sequencing data. As a case study, we used mothur to trim, screen, and align sequences; calculate distances; assign sequences to operational taxonomic units; and describe the alpha and beta diversity of eight marine samples previously characterized by pyrosequencing of 16S rRNA gene fragments. This analysis of more than 222,000 sequences was completed in less than 2 h with a laptop computer.

17,350 citations

01 Jan 2007

10,427 citations


"Metformin Is Associated With Higher..." refers methods in this paper

  • ...tions using the Vegan package of R (22)....

    [...]

  • ...using the adonis function (ANOVA using distance matrices) of the permutational multivariate ANOVA (PERMANOVA) implemented in theVegan package of R (22)....

    [...]

Journal ArticleDOI
TL;DR: A new method for metagenomic biomarker discovery is described and validates by way of class comparison, tests of biological consistency and effect size estimation to address the challenge of finding organisms, genes, or pathways that consistently explain the differences between two or more microbial communities.
Abstract: This study describes and validates a new method for metagenomic biomarker discovery by way of class comparison, tests of biological consistency and effect size estimation. This addresses the challenge of finding organisms, genes, or pathways that consistently explain the differences between two or more microbial communities, which is a central problem to the study of metagenomics. We extensively validate our method on several microbiomes and a convenient online interface for the method is provided at http://huttenhower.sph.harvard.edu/lefse/.

9,057 citations


"Metformin Is Associated With Higher..." refers methods in this paper

  • ...pairwise tests among groups of participants using the unpaired Wilcoxon rank sum test, and finally uses LDA to estimate the effect size of each differentially abundant OTU (24)....

    [...]

  • ...We next used linear discriminant analysis (LDA) effect size (LEfSe) (24) to agnostically identify microbial biomarkers....

    [...]

Journal ArticleDOI
TL;DR: This work presents an improved method for sequencing variable regions within the 16S rRNA gene using Illumina's MiSeq platform, which is currently capable of producing paired 250-nucleotide reads and demonstrates that it can provide data that are at least as good as that generated by the 454 platform while providing considerably higher sequencing coverage for a fraction of the cost.
Abstract: Rapid advances in sequencing technology have changed the experimental landscape of microbial ecology. In the last 10 years, the field has moved from sequencing hundreds of 16S rRNA gene fragments per study using clone libraries to the sequencing of millions of fragments per study using next-generation sequencing technologies from 454 and Illumina. As these technologies advance, it is critical to assess the strengths, weaknesses, and overall suitability of these platforms for the interrogation of microbial communities. Here, we present an improved method for sequencing variable regions within the 16S rRNA gene using Illumina's MiSeq platform, which is currently capable of producing paired 250-nucleotide reads. We evaluated three overlapping regions of the 16S rRNA gene that vary in length (i.e., V34, V4, and V45) by resequencing a mock community and natural samples from human feces, mouse feces, and soil. By titrating the concentration of 16S rRNA gene amplicons applied to the flow cell and using a quality score-based approach to correct discrepancies between reads used to construct contigs, we were able to reduce error rates by as much as two orders of magnitude. Finally, we reprocessed samples from a previous study to demonstrate that large numbers of samples could be multiplexed and sequenced in parallel with shotgun metagenomes. These analyses demonstrate that our approach can provide data that are at least as good as that generated by the 454 platform while providing considerably higher sequencing coverage for a fraction of the cost.

5,417 citations

Related Papers (5)