ArticlePDF AvailableLiterature Review

Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: A systematic review of randomized controlled trials

Authors:

Abstract and Figures

Background The effects of probiotic supplementation on fecal microbiota composition in healthy adults have not been well established. We aimed to provide a systematic review of the potential evidence for an effect of probiotic supplementation on the composition of human fecal microbiota as assessed by high-throughput molecular approaches in randomized controlled trials (RCTs) of healthy adults. Methods The survey of peer-reviewed papers was performed on 17 August 2015 by a literature search through PubMed, SCOPUS, and ISI Web of Science. Additional papers were identified by checking references of relevant papers. Search terms included healthy adult, probiotic, bifidobacterium, lactobacillus, gut microbiota, fecal microbiota, intestinal microbiota, intervention, and (clinical) trial. RCTs of solely probiotic supplementation and placebo in healthy adults that examined alteration in composition of overall fecal microbiota structure assessed by shotgun metagenomic sequencing, 16S ribosomal RNA sequencing, or phylogenetic microarray methods were included. Independent collection and quality assessment of studies were performed by two authors using predefined criteria including methodological quality assessment of reports of the clinical trials based on revised tools from PRISMA/Cochrane and by the Jadad score. Results Seven RCTs investigating the effect of probiotic supplementation on fecal microbiota in healthy adults were identified and included in the present systematic review. The quality of the studies was assessed as medium to high. Still, no effects were observed on the fecal microbiota composition in terms of α-diversity, richness, or evenness in any of the included studies when compared to placebo. Only one study found that probiotic supplementation significantly modified the overall structure of the fecal bacterial community in terms of β-diversity when compared to placebo. Conclusions This systematic review of the pertinent literature demonstrates a lack of evidence for an impact of probiotics on fecal microbiota composition in healthy adults. Future studies would benefit from pre-specifying the primary outcome and transparently reporting the results including effect sizes, confidence intervals, and P values as well as providing a clear distinction of between-group and within-group comparisons.
Content may be subject to copyright.
R E S E A R C H Open Access
Alterations in fecal microbiota composition
by probiotic supplementation in healthy
adults: a systematic review of randomized
controlled trials
Nadja B. Kristensen
*
, Thomas Bryrup, Kristine H. Allin, Trine Nielsen, Tue H. Hansen and Oluf Pedersen
Abstract
Background: The effects of probiotic supplementation on fecal microbiota composition in healthy adults have not
been well established. We aimed to provide a systematic review of the potential evidence for an effect of probiotic
supplementation on the composition of human fecal microbiota as assessed by high-throughput molecular approaches
in randomized controlled trials (RCTs) of healthy adults.
Methods: The survey of peer-reviewed papers was performed on 17 August 2015 by a literature search through
PubMed, SCOPUS, and ISI Web of Science. Additional papers were identified by checking references of relevant
papers. Search terms included healthy adult, probiotic, bifidobacterium, lactobacillus, gut microbiota, fecal microbiota,
intestinal microbiota, intervention, and (clinical) trial. RCTs of solely probiotic supplementation and placebo in healthy
adults that examined alteration in composition of overall fecal microbiota structure assessed by shotgun metagenomic
sequencing, 16S ribosomal RNA sequencing, or phylogenetic microarray methods were included. Independent collection
and quality assessment of studies were performed by two authors using predefined criteria including methodological
quality assessment of reports of the clinical trials based on revised tools from PRISMA/Cochrane and by the Jadad score.
Results: Seven RCTs investigating the effect of probiotic supplementation on fecal microbiota in healthy adults were
identified and included in the present systematic review. The quality of the studies was assessed as medium to high. Still,
no effects were observed on the fecal microbiota composition in terms of α-diversity, richness, or evenness in any of the
included studies when compared to placebo. Only one study found that probiotic supplementation significantly modified
theoverallstructureofthefecal bacterial community in terms of β-diversity when compared to placebo.
Conclusions: This systematic review of the pertinent literature demonstrates a lack of evidence for an impact of
probiotics on fecal microbiota composition in healthy adults. Future studies would benefit from pre-specifying
the primary outcome and transparently reporting the results including effect sizes, confidence intervals, and Pvalues as
well as providing a clear distinction of between-group and within-group comparisons.
Background
The human gut microbiota refers to the microbes that
reside inside the gut and partake in several functions
beneficial to the host including fermentation of other-
wise indigestible dietary fibers and other food items [1],
synthesis of vitamins and amino acids [2], prevention of
pathogen colonization [3], maturation and regulation of
the immune system [4], modulation of gastrointestinal
hormone release, and regulation of brain behavior
through bidirectional neuronal signaling as part of the gut-
brain axis [5]. The development of culture-indpendent,
high-throughput molecular techniques has enabled the
identification of previously unknown bacterial species,
thereby providing novel insights into the compositional
diversity and functional capacity of fecal microbiota. As
a result, studies have suggested that disorders such as
colorectal cancer, rheumatoid arthritis, type 2 diabetes,
and obesity are associated with disease-specific dysbiotic
* Correspondence: nadja@sund.ku.dk
The Novo Nordisk Foundation Center for Basic Metabolic Research, Section
of Metabolic Genetics, Faculty of Health and Medical Sciences, University of
Copenhagen, Universitetsparken 1, 2nd floor, Copenhagen Ø 2100, Denmark
© 2016 Kristensen et al. Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Kristensen et al. Genome Medicine (2016) 8:52
DOI 10.1186/s13073-016-0300-5
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
shifts in fecal microbiota [611]. Consequently, in recent
years the gut microbiota as a potential modifiable risk fac-
tor for disease development has received massive atten-
tion. One common approach applied to convey health
benefits by way of modifying the gut microbiota has been
the use of probiotic supplementation. Probiotics are de-
fined as live microorganisms that, when administered in
adequate amounts, confer a health benefit on the host in a
safe and efficacious manner [12]. Suggested mechanisms
by which probiotics may benefit the gut environment and
the health of the host include improvement of the intes-
tinal barrier function through effects on the epithelium
and mucus lining, production of anti-microbial substances,
competition with pathogenic bacteria, and regulation of
luminal acidity (reviewed in [13, 14]).
The therapeutic effect of probiotic supplementation
has been studied in a broad range of diseases, particu-
larly in regard to gastrointestinal and metabolic disor-
ders where results have supported the potential use of
probiotics as therapeutic agents (reviewed in [15, 16]).
Common to both sets of disorders is a multitude of readily
available, clinically relevant outcome measures (e.g. body
mass index, fat mass, insulin resistance, severity of gastro-
intestinal symptoms) by which to measure treatment ef-
fect. The effect of probiotics in disease-free individuals is,
however, not as easily assessed. Interpretation of an effect
on the composition of fecal microbiota in healthy indi-
viduals may be particularly complicated due to the lack
of an internationally accepted consensus definition of a
normal or a healthy fecal microbial community [17, 18].
Terms such as ecological stability, idealized compos-
ition, or favorable functional profile have been suggested
as hallmarks of a healthy gut microbiota [17]. These are
all very unspecific concepts and the compositional and
functional characteristics of a healthy gut microbial
community remain to be defined. Furthermore, an ef-
fect of probiotics on the composition of the gut micro-
biota is an intermediate outcome only and should be
interpreted with caution in regard to implications for
the health of the host. Despite these limitations, several
probiotic interventions aiming to observe alterations in
fecal microbiota composition have been performed in
healthy adults [1928]. Results from these studies have
the potential to provide insights into the underlying
mechanisms of probiotics and fecal microbiota. Cur-
rently, no systematic review has addressed the effects of
probiotics on fecal microbiota composition using high-
throughput metagenomic methods (i.e. phylogenetic
microarrays, 16S ribosomal RNA (rRNA) sequencing,
or shotgun metagenomic sequencing) in healthy adults.
In the context of a billion dollar market for probiotic
supplements [29] with products being marketed, in
part, toward healthy individuals by stating effects on
gastrointestinal health, alluding to the fecal microbial
community, an overview of the current evidence is
warranted.
The objective of the present systematic review was to
explore in healthy adults the current evidence for an
effect of probiotic supplementation compared to pla-
cebo on the composition of human fecal microbiota as
assessed by high-throughput molecular approaches in
randomized controlled trials (RCTs).
Methods
We undertook a systematic review of the possible effects
of probiotic intervention on the composition of fecal
microbiota in healthy adults. The available literature was
identified and examined as a systematic review and not a
meta-analysis due to the heterogeneity of the study designs
and methods. The results are reported in accordance with
the PRISMA statement guidelines (Preferred Reporting
Items for Systematic Reviews and Meta-Analyses) [30].
The study followed an a priori established protocol.
Eligibility criteria
The criteria for eligibility were healthy adults as study
population, probiotics and placebo as intervention, alter-
ation in composition of fecal microbiota assessed by
shotgun metagenomic sequencing, 16S rRNA sequen-
cing or phylogenetic microarray methods as the primary
outcome, and RCT as the study design with no criteria
on study duration. No limits were applied for language
or time. Studies not exploiting the randomized con-
trolled design and providing only within-group results
(i.e. results before and after the intervention in the pro-
biotic group only) were not included. Moreover, only
studies assessing the overall bacterial ecology were in-
cluded. Accordingly, studies investigating survival of the
probiotic strains only were considered ineligible. Studies
with interventions combining probiotics with other sup-
plements (e.g. prebiotics, antibiotics, medications) were
excluded. If studies had more than two arms, only the com-
parison of probiotics to placebo was considered. Studies
examining both healthy and unhealthy participants were
excluded.
Information sources, search strategy, and study selection
The identification of papers involved four sequential
processes performed by two independent reviewers
(NBK and TB). On 17 August 2015, a literature search
was conducted through multiple electronic databases
(PubMed, SCOPUS, and ISI Web of Science) to capture
as many relevant citations as possible. The search
phrase used was:
Healthy adult AND (probiotic OR bifidobac* OR
lactobac*) AND (gut microbio* OR f*cal microbio* OR
intestinal microbio*) AND (clinical trial OR interven-
tion OR trial).
Kristensen et al. Genome Medicine (2016) 8:52 Page 2 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
In PubMed, Specieswas limited to include only
humans and Article typeswas limited to cover Clinical
trial,”“Comparative study,”“Controlled clinical trial,
Journal article,and Randomized controlled trial.
In ISI Web of Science, Document typeswas limited to
contain Article,”“Clinical trial,”“Other,and Abstract.
In Scopus, Source type and document typewas limited
to comprise Journals and article,”“Short survey,and
Erratum.For Subject area,”“Agricultural and biological
sciences,”“Nursing,”“Pharmacology,”“Toxicology and
pharmaceutics,”“Environmental science,”“Veterinary,
Chemistry,and Neurosciencewere excluded.
Full reports were obtained and screened for all titles
appearing to meet the inclusion criteria or in case of
any uncertainty. References in 31 full-text articles were
also assessed for inclusion in the present review.
Screening and eligibility assessment by title and ab-
stract resulted in 1373 citations (Fig. 1). The assessment
was performed independently in an unblinded standard-
ized manner by NBK and TB resulting in seven included
studies. Any disagreements between reviewers were re-
solved by consensus.
Data collection process
Independent data collection was performed by two authors
(NBK and TB). Corresponding authors of the following
studies were contacted in order to acquire missing infor-
mation on allocation concealment or other measures of
risk of bias: Lahti et al., Rampelli et al., Ferrario et al., Bjerg
et al., Hanifi et al., and Simon et al. [19, 2124, 27]. Unpub-
lished information about blinding was obtained from Lahti
et al., Bjerg et al., and Hanifi et al. [19, 21, 22] and the rea-
son for excluding data from three participants from the
intervention group (missing fecal samples) was obtained
from Hanifi et al. [21].
Fig. 1 Flow chart of literature selection process [30]
Kristensen et al. Genome Medicine (2016) 8:52 Page 3 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Data items
Information extracted from each included RCT was: (1)
participant characteristics (including age and gender);
(2) intervention (including probiotic strain and dosage
as well as mode of administration); (3) design (including
study design and duration); and (4) outcome measure
(including the microbiomics and intervention effect on
overall fecal microbiota structure).
Quality assessment
The methodological quality assessment of reports of the
clinical trials was performed independently by NBK and
TB using a three-item instrument (the Jadad score) that
evaluates likelihood of bias in research reports [31].
The three items evaluated by a five-point scale are
quality of randomization, quality of blinding, and rea-
sons for withdrawal/drop-out (0 = worst, 5 = best). Risk
of bias was further assessed regarding concealment of
randomization, early termination of trial, blinding of
patients, healthcare providers, data collectors and out-
come assessors, reporting of drop-out or withdrawal,
selective outcome reporting, and other potential biases
[32, 33].
Summary measures
Intervention effects on the overall fecal microbiota struc-
ture, that is, richness, abundance, evenness, α-diversity or
compositional dissimilarity (β-diversity), were the primary
measures of treatment effects.
Results
Study selection
A total of 1368 citations were identified through the
search in PubMed, SCOPUS, and ISI Web of Science
and an additional five were identified through checking
the references of relevant papers. After the removal of
duplicates, 1287 citations were left. NBK and TB
screened the initial search results using abstracts and
1256 citations were excluded as irrelevant for one or
more of the following reasons: animal study, meta-
analysis/review, non-healthy or non-adult participants, no
probiotic intervention, or no assessment of fecal micro-
biota composition. The full papers of the remaining 31
citations and references therein were assessed to select
studies for inclusion using the abovementioned criteria,
resulting in the exclusion of 24 studies due to one or a
combination of the following reasons: no assessment of
fecal microbiota composition, assessment of single-strain
survival only, inclusion of non-healthy participants, non-
randomized controlled design, provision of only within-
group results, and combined intervention of probiotic
with prebiotics or other foods. Following the selection
process (Fig. 1), seven studies [1924, 27] remained
(five of which were identified by checking the references
of relevant papers) and were included in the present sys-
tematic review.
Study characteristics
All seven studies were published in English language
journals between February 2013 and October 2015
(EPub June 2015). One study was performed in Finland
[22], two in Italy [23, 27], two in Denmark [19, 20], one
in the United States [21], and one in Germany [24]. An
overview of the study characteristics and main results
are presented in Table 1. The studies were designed as
RCTs, one of which used a cross-over design [27]. Six
studies were double-blinded, whereas one was single-
blinded [20]. Participants were all healthy adults (range,
1988 years) with a proportion of female participants in
the range of 50100 %. The total number of included in-
dividuals was in the range of 2181. The intervention
received was probiotics belonging to the genus Lactoba-
cillus (n = 5) [19, 20, 22, 24, 27], Bifidobacterium and
Lactobacillus combined (n = 1) [23], or Bacillus (n = 1)
[21] which was provided in biscuits (n = 1) [23], milk-
based drinks (n = 1) [22], sachets (n = 1) [20], or capsules
(n = 4) [19, 21, 24, 27] administered at a dose of ~10
9
to
10
11
colony-forming units (CFU) for 2142 days. Three
of the studies collected additional samples 13 weeks
after the intervention had ended [19, 21, 22]. Compli-
ance was assessed by pill count or screening for the pro-
biotic in fecal microbiota and evaluated as sufficient in
most of the studies. However, Rampelli et al. found only
a trend towards enrichment of the probiotic strain [23].
Habitual diet was assessed in two studies [20, 27]. In the
present review, the primary outcome of interest is alter-
ations in fecal microbiota composition, which was assessed
by either microarray hybridization (HITchip (n = 1) [22],
HTF-Microbi.Array (n = 1) [23]), or next-generation se-
quencing methods (16S rRNA sequencing on Ion Torrent
PGM (n = 1) [27], Illumina MiSeq platforms (n = 1) [24], or
454 pyrosequencing (n = 2) [19, 21]), or metagenomics on
a SOLiD 5500×l platform (n = 1) [20]. Of the studies using
a 16S rRNA-based approach, one did not report which
hypervariable region of the 16S rRNA gene was targeted
and no studies targeted the same set of regions. The data-
bases used for mapping the sequences were GreenGenes
version 13.5 (n = 1) [27], RDP (MultiClassifier 1.1 or not
specified) (n = 2) [19, 24], or both (versions not specified)
(n = 1) [21], while two did not report the database used.
The study by Brahe et al. [20] used metagenomics and
mapped reads to a reference catalogue of 3.3 million bac-
terial genes [34].
Risk of bias
Seven studies were identified and evaluated as medium
to high quality by the Jadad score (35) as presented in
Table 2. The quality of the included studies is generally
Kristensen et al. Genome Medicine (2016) 8:52 Page 4 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Table 1 Characteristics of the studies reviewed
Study Participant
characteristics
Intervention Design Microbiomics Sample size
calculation
Probiotic effects on fecal
microbiota compared to placebo
Lahti et al.
2013 [22]
25/25 (72 %)
2355 years
Finland
L. rhamnosus GG ATCC53103
(1.55 × 10
10
CFU) in 250 mL
milk-based fruit drink
Double-blinded, parallel,
two-armed, placebo (drink)
controlled (21 days)
16S rRNA (regions V1 and V6);
HITchip based characterization of
>1000 microbial species-like phylotypes
Post hoc Composition of the fecal microbiota
Stability of the fecal microbiota
quantified by inter-individual and
intra-individual correlations
within and between time points
Rampelli
et al. 2013
[23]
32/32 (59 %)
7188 years
Italy
B. longum Bar33 and
L. helveticus Bar13 (10
9
CFU)
in biscuit
Double-blinded, parallel,
two-armed, placebo (biscuit)
controlled (30 days)
16S rRNA (region unknown);
HTF-Microbi.Array based characterization
of 31 phylogenetically related groups
No Relative abundance of 31
phylogenetically related groups
Ferrario
et al. 2014
[27]
22/34* (56 %)
2355 years
Italy
L. paracasei DG
(>2.4 × 10
10
CFU) in capsules
Double-blinded, cross-over, placebo
(capsules) controlled (two 28-day
intervention periods with a
28-day wash-out)
16S rRNA (region V3) sequencing on Ion
Torrent platform
No α-diversity
Modified βdiversity (with absolute
distances higher for the probiotic
than for the placebo treatments)
Bjerg et al.
2015 [19]
20/64* (50 %)
2045 years
Denmark
L. casei W8® (10
10
CFU)
in capsules
Double-blinded, parallel, two-armed,
placebo (capsules) controlled
(28 days)
16S rRNA (regions V3 and V4) sequencing
on Roche 454 pyrosequencing platform
No α- and β-diversity
Brahe et al.
2015 [20]
34/58*
(100 %)
4070 years
Denmark
L. paracasei F19 (9.4 × 10
10
CFU)
or flaxseed mucilage (10 g)
in sachets
Single-blinded, parallel, three-armed,
placebo (sachets) controlled (42 days)
Quantitative metagenomics
on a SOLiD 5500×l platform
Yes Bacterial gene count (richness)
and abundance of specific bacterial
genes compared to placebo
Hanifi et al.
2015 [21]
37/81* (52 %)
1949 years
United States
Bacillus subtilis R0179
(0.1 × 10
9
, 1.0 × 10
9
, and
10 × 10
9
CFU, respectively)
in capsules
Double-blinded, parallel, four-armed,
placebo (capsules) controlled
(28 days)
16S rRNA (regions V1 to V3)
pyrosequencing
No β-diversity and OTU based richness
Simon et al.
2015 [24]
21/21 (52 %)
4065 years
Germany
L. reuteri SD5865 (2 × 10
10
viable cells)
in capsules
Double-blinded, parallel, two-armed,
placebo (capsules) controlled
(28 days)
16S rRNA (regions V5 and V6)
sequencing on Illumina MiSeq platform
Yes α- and β-diversity
Participant characteristics are number of participants with microbiome data/number of participants included in the study. Participant characteristics (% women and age range of participants in years) are based upon
number of participants included in the study. CFU, colony-forming units. OTU, operational taxonomic unit. indicates no difference between the probiotic group and placebo. indicates an increase in the probiotic
group compared to placebo. indicates a decrease in the probiotic group compared to placebo. Pvalues are unadjusted for multiple testing. *Performed next-generation sequencing on fecal samples from a subgroup
of the include d individuals. In the study by Ferrario et al., 22 participa nts (of 34) completed the stu dy. Bjerg et al. sel ected 10 (of 32) placebo-t reated and 10 (of 32) probiotic-treated participa nts with the high est
triacylglycerol concentration in the blood at week 0. The stu dy by Brahe et al . had a third int erventio n arm not relev ant for the present study where the numb er of partic ipants were approximately two-t hirds of
the total number of participants included in th e study. Hanifi et al. selected 20 (a ll) placebo-treated and 17 (of 20 ) probiotic-treate d participants from the group with the highest dose (10 × 10
9
CFU) of the
probiotic tr eatment
Kristensen et al. Genome Medicine (2016) 8:52 Page 5 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Table 2 Assessment of risk of bias in the studies reviewed
Study Concealment of
randomization
RCT stopped
early
Patients
blinded
Healthcare providers
blinded
Data collectors
blinded
Outcome assessors
blinded
Reporting drop-out
or withdrawal
Other
potential bias
Selective outcome
reporting
Jadad
score
Lahti et al.
2013 [22]
Yes No Yes Yes Yes No Yes No No 4
Rampelli et al.
2013 [23]
Yes No Yes Unclear
a
Unclear
a
Unclear
a
Unclear Yes
c
Yes
c
3
Ferrario et al.
2014 [27]
Yes No Yes Unclear
a
Unclear
a
Unclear
a
Yes No No 3
Bjerg et al.
2015 [19]
Yes No Yes Yes Yes Yes Yes Yes
b
No 5
Brahe et al.
2015 [20]
Yes No Yes No No No Yes No No 4
Hanifi et al.
2015 [21]
Yes No Yes Yes Yes Yes Yes Yes
b
No 5
Simon et al.
2015 [24]
Yes No Yes Unclear
a
Unclear
a
Unclear
a
Yes No No 4
Based on PRISMA (and Cochrane)s tools for assessing risk of bias. The Jadad score is a three-item instrument that evaluates likelihood of bias in terms of quality of randomization, quality of blinding, and reasons for
withdrawal/drop-out. It is compiled of the following quality items from the table: Concealment of randomization, Patients blinded, Healthcare providers blinded, Data collectors blinded, Outcome assessors blinded,
and Reporting drop-out or withdrawal
a
Double-blinded study but unclear whether healthcare providers, data collectors, and outcome assessors were all blinded
b
Performed next-gene ration seq uencing on fe cal samples from a subg roup of the in cluded indi viduals . Bjerg et al. se lected 10 (o f 32) placebo-treated and 10 (of 32) probiotic-treated participants with the highest
triacylglycerol concentrat ion in the bloo d at week 0. Ha nifi et al. selected 20 ( all) placeb o-treat ed and 17 (of 20 ) probiotic-treated participants from th e group wit h the highes t dose (10 × 10
9
colony-forming units)
of the probi otic treatment
c
No direct comparison between treatment groups was made for the age-related dysbiosis
Kristensen et al. Genome Medicine (2016) 8:52 Page 6 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
high in regard to risk of bias and the methods of asses-
sing fecal microbiota configuration. However, blinding of
healthcare providers, data collectors, and outcome asses-
sors were either not performed or unclearly reported in
three of the seven included studies, which may have
caused performance and detection bias. Two studies only
investigated the effect of the probiotic treatment on a
subgroup of participants, which is also a potential source
of bias.
Results of individual studies
In terms of richness, evenness, or α-diversity measures,
no effects were observed on the fecal microbiota com-
position in any of the included studies when compared
to placebo and only in the study by Ferrario et al. [27]
was it found that probiotic treatment significantly modi-
fied the compositional dissimilarity (β-diversity).
In the study by Lahti et al. [22], the temporal stability
of fecal microbiota, quantified by the correlation of the
fecal microbiota profiles among three time points, did
not differ between the probiotic (L. rhamnosus GG
ATCC53103) and the placebo group.
In the study by Rampelli et al. [23], there was no effect
of probiotic supplementation (B. longum Bar33 and L.
helveticus Bar13) on the relative abundance of 31 phylo-
genetically related groups when compared to placebo. In
the same study, the effect of probiotic supplementation on
age-related dysbiosis was also evaluated. The probiotic
intervention reverted an age-related increase of Clostridium
cluster Xi, C. difficile,C. perfringens,Enterococcus Faecium,
and Campylobacter when comparing the probiotic and the
placebo group to a common reference of eight young,
healthy adults; but no direct comparison was made be-
tween treatment groups.
In the study by Ferrario et al. [27], the α-diversity re-
ported as Chao1 and Shannon coefficients and number
of detected genera did not change as a result of the pro-
biotic intervention (L. paracasei DG) when compared to
placebo. Yet, the β-diversity between the probiotic and
the placebo group was modified with absolute distances
significantly higher for the probiotic than for the placebo
treatments when assessed by principal coordinate ana-
lysis (PCoA) of weighted UniFrac distances. Accordingly,
the relative abundance of Proteobacteria (P= 0.006) and
Clostridiales genus Coprococcus (P= 0.009) were inc-
reased and the Clostridiales genus Blautia (P= 0.036)
was decreased in the probiotic group when compared
to placebo. Additionally, analyses of predicted functional
profiles revealed changes in eight Kyoto Encyclopedia of
Genes and Genomes modules related to bacterial path-
ways in membrane transport, amino acid metabolism,
energy metabolism, and metabolism of cofactors and vita-
mins (P<0.05).
In the study by Bjerg et al. [19], the β-diversity was
not affected by the probiotic intervention (L. casei W8®)
compared to placebo when assessed by PCoA of species
and genus level Operational Taxonomic Unit (OTU)
based UniFrac distances. Furthermore, no difference in
α-diversity (Chao1 and Shannon index) or species rich-
ness was observed between the probiotic and the pla-
cebo group.
In the study by Brahe et al. [20], fecal microbiota was
assessed by shotgun-sequencing-based metagenomics.
The bacterial gene count (richness) did not change
within the probiotic group (L. paracasei F19) compared
to placebo. Alterations in the abundance of individual
bacterial genes (2493 genes assigned to two metage-
nomic species) were observed after the intervention in
the probiotic group. However, fewer alterations were ob-
served in the intervention group compared to the pla-
cebo group (7436 genes assigned to six metagenomic
species). Yet, no direct comparison between the groups
is explicitly stated.
In the study by Hanifi et al. [21], no difference in com-
positional dissimilarity (β-diversity) between the treat-
ment groups (Bacillus subtilis R0179 in different doses)
and placebo was shown when analyzed using PCoA
based on the UniFrac metric. Sequence reads binned to
multiple OTUs assigned to the genus Ruminococcus in-
creased in the probiotic group (with the highest dose
(10 × 10
9
CFU, Table 2) compared to placebo (P< 0.01).
In the study by Simon et al. [24], the overall compos-
ition of fecal microbiota was unaffected by probiotic
supplementation (L. reuteri SD5865) both in terms of α-
(Chao1, Shannon, and Simpson indices) and β-(Bray-
Curtis, Morisita-Horn, and weighted UniFrac) diversity.
Discussion
Overall, this systematic review demonstrates that there is
no convincing evidence for consistent effects of probiotics
on fecal microbiota composition in healthy adults.
No effects were observed on the fecal microbiota com-
position in terms of α-diversity, richness, or evenness in
any of the included studies when compared to placebo.
Only in the paper by Ferrario et al. [27] was it reported
that probiotic supplementation significantly modified the
overall structure of the fecal bacterial community in
terms of compositional dissimilarity (β-diversity) when
compared to placebo.
Study design and reporting of results
Overall, the reporting of the analyses and results was non-
transparent and difficult to assess with very few effect
sizes, confidence intervals, and Pvalues reported. This is
possibly due to the fact that fecal microbiomics is a rela-
tively new research area that currently relies heavily on
non-parametric statistics and lacks an internationally
Kristensen et al. Genome Medicine (2016) 8:52 Page 7 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
accepted standard approach of reporting results. Unfortu-
nately, this impedes the comparison of results in the
present review. As the only study, Ferrario et al. [27] used
a cross-over design, which may not be the ideal design to
assess the effects of a probiotic intervention due to the risk
of carry-over effects [35]. In the study the probiotic cell
count was decreased after the 4-week wash-out period
compared with baseline count, suggesting that wash-out
was effective. However, a carry-over effect at the outcome
level cannot be excluded. Only two studies provided a
priori sample size calculations [20, 24], of which two calcu-
lated statistical power based on alterations in fecal micro-
biota composition [20, 22]. Thus, several of the studies
may well have been underpowered, with an inherent
risk of unequal distribution of confounding factors. A
potential confounder in the studies reviewed is habitual
diet. Human studies have revealed that short-term and
long-term changes in diet (such as plant-based vs. animal-
based, amount of dietary fibers and fat) influence the fecal
microbiota composition and function [3638]. Hence, the
enormous inter-individual variation in the dietary intake
and its effect on the fecal microbiota may mask the true
picture of the impact of a probiotic treatment. Only one
of the included studies monitored habitual diet with the
aim of accounting for differences in dietary habits, specif-
ically considering prebiotic fibers, during the intervention
period [27]. A major limitation of most included studies is
an unclear, inexplicit statement of the pre-specified pri-
mary outcome and delimitation of secondary outcomes.
Only one study [24] is explicitly labeled as a pilot trial,
reporting a multitude of outcomes, only in part addressing
multiple testing. Two studies do not address the issue of
multiple testing [23, 27], while others report multiple pri-
mary outcomes or make no distinction between pri-
mary and secondary outcomes [1922]. Reporting of
the results is generally unclear, with between-group
comparisons on primary outcomes intermixed with re-
sults on secondary outcomes and within-group compar-
isons of differences between baseline and post-
intervention measures.
Heterogeneity
Although study participants in the included trials were
all healthy adults, the demographic makeup varied
widely among studies. Rampelli et al. [23] included only
elderly individuals, who may respond differently to pro-
biotics than young individuals and Brahe et al. [20] in-
cluded postmenopausal women only.
Considering that the impact on the fecal microbiota
may differ among strains of the same bacterial species
[39], despite close phylogenetic relationships, a potential
source of heterogeneity is the use of various probiotic
agents. Six studies used single-strain interventions with
probiotic products belonging to the genera Lactobacillus
[19, 20, 22, 24, 27] or Bacillus [21]. One study used a
double-strain probiotic mixture of bacteria belonging
to the genera Lactobacillus and Bifidobacterium [23].
Whereas the use of different probiotic agents makes it
difficult to draw any meta-analytical conclusions, the
choice between single-strain and multi-strain interven-
tion is probably of less importance. In most cases, inert
bacteria are administered and within a few hours are
entering a diverse ecosystem where they are numerically a
minority. So while additive or synergistic effects might be
observed in vitro, the opportunity for metabolically active
strains delivered in combination to result in similar effects
in vivo may not present itself.
None of the studies included in the present review
comment on the rationale behind their choice of dosage.
The International Scientific Association for Probiotics
and Prebiotics provides a list of dosages ranging from
1×10
8
to 1.8 × 10
12
CFU twice daily depending on strain
and disease, based on at least one well designed clinical
trial showing a beneficial effect for a health promoting
or therapeutic outcome [40]. However, the list covers
gastrointestinal disorders only and does not address fecal
microbiota in healthy participants. In general, different
dosages should be assessed to facilitate an interpretation
of the dose-response relationship of probiotic consump-
tion on relevant outcomes, rather than on safety and via-
bility alone. The information provided by such studies
would enable the identification of the dosage needed to
observe an impact on the relevant outcome and add to
the likelihood that an observed association is causal [41].
Hanifi et al. [21] examined and detected oral dose-
response relationships, but for tolerance and gastrointes-
tinal viability only. As of now, it is impossible to draw
any conclusions on the ideal dosage regarding effects on
the fecal microbiota composition. Likewise, the optimal
duration of intervention remains elusive.
Mode of administration may also contribute to the
observed lack of impact on fecal microbiota. Ingested
probiotics must survive hostile environments including
acidic, protease and bile-salt rich conditions during their
passage through the gastrointestinal tract [42, 43]. Cur-
rently, screening feces is the only way to assess whether the
probiotics have survived through the gastrointestinal tract.
Yet, the site of action may be proximal to the colon and it
is not necessarily possible to conclude on the degree of
colonization or even the amount of bacteria that produce
the effect [44]. In contrast to the findings in five of the in-
cluded studies, the study by Rampelli et al. [23] showed
only a trend towards enrichment of the probiotic strain,
perhaps due to the use of biscuits as the mode of adminis-
tration, yet, another reason may be low compliance. This
may add to the explanation why little effect of probiotics
was found in Rampelli et al. [23]. Compliance was evalu-
ated as sufficient in the remaining studies [1922, 24, 27].
Kristensen et al. Genome Medicine (2016) 8:52 Page 8 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Further adding to the heterogeneity between studies
is the application of different methods for assessing
fecal microbiota. Even though all studies applied high-
throughput metagenomics approaches, the resolution
and specificity levels varied tremendously and no studies
used the same methodological approach. Only one study
examined the fecal microbiota by an untargeted metage-
nomic approach using shotgun sequencing and thereby
providing information on microbial gene and derived
functional levels, free of bias introduced by amplification
of a specific genomic region as is the case in targeted 16S
rRNA sequencing and array-based analyses [45]. Com-
positional information can be achieved by mapping the
reads to a microbial gene reference catalogue [34], as
was done by Brahe et al. [20]. Still, only a fraction of se-
quencing reads can be mapped to the existing reference
catalogues. The targeted 16S rRNA approach provides
information at the taxonomic level in the form of abun-
dance and phylogenetic relationship, but the method has
pitfalls in PCR amplification steps [46] and cross-platform
comparison is not straightforward. Of the included stud-
ies, two use 454 pyrosequencing [19, 21], one uses Illu-
mina MiSeq [24], another uses Ion Torrent sequencing
[27], and two use phylogenetic microarrays [22, 23]. The
sequencing platforms differ in costs, coverage, and length
of reads with the Illumina platform becoming more widely
used [45]. Community profiles from HITChip correlate
well with pyrosequencing-based compositions (Pearson
correlations at phylum (average r = 0.94), class (0.93),
order (0.94) and family levels (0.77)) and the HTF
microbe.array has demonstrated good reproducibility by
cluster analysis of the phylogenetic fingerprint in samples
from the same participant [47, 48]. In general, using
phylogenic microarray approaches have the advantages of
being cost-efficient for compositional characterization;
however cross-hybridization may occur and only taxa that
are covered by the reference sequences can be detected
[47]. Another well-known source of bias in 16S rRNA-
based studies is the targeted hypervariable region of the
16S rRNA gene. The region used for analysis in the in-
cluded studies applying 16S rRNA-based methods varies
with one study using V1 and V6 [22], one study using V3
[27], one study using V3 and V4 [19], one study using V5
and V6 [24], and one study does not specify [23]. Several
studies have examined the effects of region choice when
evaluating fecal microbiota composition with no current
international consensus [47, 4951].
Probiotics in health and disease
In a recent systematic review including 29 trials studying
healthy adults with undisturbed microbiota (using
non-high-throughput molecular techniques) only ~20 %
showed an effect of probiotic treatment on fecal micro-
biota. It is concluded that there is little, if any, evidence of
an effect of probiotic treatment in circumstances where
the microbiota is unperturbed by pathophysiological pro-
cesses or pharmaceutical treatment (antibiotics or chemo-
therapy), either concurrent with or prior to intervention.
However, where dysbiosis is present or where the micro-
biota is perturbed, there is some evidence for a restorative
or protective effect of certain strains of probiotics, both on
the fecal microbial community itself, but more import-
antly, also on host physiology, e.g. alleviation of gastro-
intestinal symptoms [18].
In the case of dysbiotic or perturbed microbial commu-
nities, any restorative or protective effect on the micro-
biota alone, without any measurable beneficial effect for
the host, would predominantly be of academic interest by
improving our understanding of the intestinal ecosystem.
In the case of undisturbed microbiota, any inference of
health benefit from changes to the microbiota alone would
be highly speculative without a direct linkage to relevant
host phenotypes. Ideally, hard endpoint data would deter-
mine the effects of probiotics in healthy individuals, but
considering the time perspective of generating such data
this may be long in coming. Until such studies are avail-
able, any statement on health benefits of probiotic supple-
mentation in healthy participants would rely on observed
effects on biomarkers or other intermediate outcomes.
Limitations
Limitations of this review include the search terms used
to identify relevant papers. In addition to probio*, we
specifically searched on bifido* and lacto*, but other
search terms such as Bacillus and Saccharomyces could
have been relevant. Publication bias is a well-known
challenge within the field of systematic reviews and
meta-analyses; however, the majority of the studies in-
cluded in the present review provide null findings, indi-
cating that this concern may be settled to some extent.
Language bias cannot be ruled out since our search was
exclusively based on English language dominated sources.
Conclusions
Based on our review of the available RCTs, we find there
is a lack of evidence to conclude whether or not there is
an effect of probiotics on fecal microbiota composition
in healthy adults, as assessed by high-throughput mo-
lecular techniques. A number of issues blur the conclu-
sions that can be drawn from the studies, including
small sample sizes with lack of statistical power, low
resolution-methods of assessing fecal microbiota com-
position, inter-individual variation in susceptibility to-
ward the probiotic, use of different probiotic strains
either in isolation or in combination, variations in dos-
age and administration mode of probiotics, duration of
intervention, or variation in the habitual diet of partici-
pants. Future research on the impact of probiotics on
Kristensen et al. Genome Medicine (2016) 8:52 Page 9 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
fecal microbiota configuration and function should in-
volve statistically well-powered RCTs in well-phenotyped
individuals. Importantly, future studies would also benefit
from pre-specifying the primary outcome and transpar-
ently reporting the results including effect sizes, confi-
dence intervals, and Pvalues as well as providing a clear
distinction of within-group and between-group com-
parisons. For the purpose of demonstrating health ben-
efits of probiotic supplementation, effects should be
demonstrated on relevant host phenotypes, which is
non-trivial in healthy participants. Studies with micro-
biome features as the primary outcome should be reserved
for improving our understanding of biology in general and
our insight into microbial interactionsinvivoinparticular.
Abbreviations
CFU: Colony forming units; OTU: Operational taxonomic unit; PCoA: Principal
coordinate analysis; RCT: Randomized controlled trial; rRNA: Ribosomal RNA.
Competing interests
The authors declare that they have no competing interests.
Authorscontributions
NBK performed the literature search, quality assessment of the included
studies, and wrote the manuscript. TB performed the literature search and
quality assessment of the included studies. KA, TN, THH, and OP supervised
the literature search and quality assessment of the included studies and
revised the manuscript critically. All authors approved the final manuscript.
Funding
The Novo Nordisk Foundation Center for Basic Metabolic Research is an
independent research center at the Faculty of Health and Medical Sciences,
University of Copenhagen and is partially funded by an unrestricted grant
from the Novo Nordisk Foundation (http://www.metabol.ku.dk). The funders
played no role in study design, collection, analysis, interpretation of data,
writing of the report, or in the decision to submit the paper for publication.
Received: 15 January 2016 Accepted: 8 April 2016
References
1. Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al. The gut
microbiota as an environmental factor that regulates fat storage. P Natl
Acad Sci USA. 2004;101(44):1571823.
2. LeBlanc JG, Milani C, de Giori GS, Sesma F, van Sinderen D, Ventura M.
Bacteria as vitamin suppliers to their host: a gut microbiota perspective.
Curr Opin Biotechnol. 2013;24(2):1608.
3. Endt K, Stecher B, Chaffron S, Slack E, Tchitchek N, Benecke A, et al. The
microbiota mediates pathogen clearance from the gut lumen after non-
typhoidal Salmonella diarrhea. PLoS Pathog. 2010;6(9):e1001097.
4. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune
responses during health and disease. Nat Rev Immunol. 2009;9(5):31323.
5. Collins SM, Surette M, Bercik P. The interplay between the intestinal
microbiota and the brain. Nat Rev Microbiol. 2012;10(11):73542.
6. Feng Q, Liang S, Jia H, Stadlmayr A, Tang L, Lan Z, et al. Gut microbiome
development along the colorectal adenoma-carcinoma sequence. Nat
Commun. 2015;6:6528.
7. Yu J, Feng Q, Wong SH, Zhang D, Liang QY, Qin Y, et al. Metagenomic
analysis of faecal microbiome as a tool towards targeted non-invasive
biomarkers for colorectal cancer. Gut. 2015. doi:10.1136/gutjnl-2015-309800.
8. Zhang X, Zhang D, Jia H, Feng Q, Wang D, Liang D, et al. The oral and gut
microbiomes are perturbed in rheumatoid arthritis and partly normalized
after treatment. Nat Med. 2015;21(8):895905.
9. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al.
Richness of human gut microbiome correlates with metabolic markers.
Nature. 2013;500(7464):5416.
10. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide
association study of gut microbiota in type 2 diabetes. Nature. 2012;
490(7418):5560.
11. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S,
et al. Disentangling type 2 diabetes and metformin treatment signatures in
the human gut microbiota. Nature. 2015;528(7581):2626.
12. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert
consensus document: The International Scientific Association for Probiotics
and Prebiotics consensus statement on the scope and appropriate use of
the term probiotic. Nat Rev Gastroenterol Hepatol. 2014;11(8):50614.
13. Gerritsen J, Smidt H, Rijkers GT, de Vos WM. Intestinal microbiota in human
health and disease: the impact of probiotics. Genes Nutr. 2011;6(3):20940.
14. Rijkers GT, Bengmark S, Enck P, Haller D, Herz U, Kalliomaki M, et al. Guidance
for substantiating the evidence for beneficial effects of probiotics: current
status and recommendations for future research. J Nutr. 2010;140(3):671S6S.
15. Ford AC, Quigley EM, Lacy BE, Lembo AJ, Saito YA, Schiller LR, et al. Efficacy
of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and
chronic idiopathic constipation: Systematic review and meta-analysis.
Am J Gastroenterol. 2014;109(10):154761.
16. Le Barz M, Anhê FF, Varin TV, Desjardins Y, Levy E, Roy D, et al. Probiotics as
complementary treatment for metabolic disorders. Diabetes Metab J. 2015;
39(4):291303.
17. Bäckhed F, Fraser CM, Ringel Y, Sanders ME, Sartor RB, Sherman PM, et al.
Defining a healthy human gut microbiome: current concepts, future
directions, and clinical applications. Cell Host Microbe. 2012;12(5):61122.
18. McFarland LV. Use of probiotics to correct dysbiosis of normal microbiota
following disease or disruptive events: a systematic review. BMJ Open. 2014;
4(8):e005047.
19. Bjerg AT, Sørensen MB, Krych L, Hansen LH, Astrup A, Kristensen M, et al.
The effect of Lactobacillus paracasei subsp. paracasei L. casei W8® on blood
levels of triacylglycerol is independent of colonisation. Benef Microbes.
2015;6(3):2639.
20. Brahe LK, Le Chatelier E, Prifti E, Pons N, Kennedy S, Blædel T, et al. Dietary
modulation of the gut microbiotaa randomised controlled trial in obese
postmenopausal women. Br J Nutr. 2015;114(03):40617.
21. Hanifi A, Culpepper T, Mai V, Anand A, Ford A, Ukhanova M, et al. Evaluation
of Bacillus subtilis R0179 on gastrointestinal viability and general wellness: a
randomised, double-blind, placebo-controlled trial in healthy adults. Benef
microbes. 2014;6(1):1927.
22. Lahti L, Salonen A, Kekkonen RA, Salojärvi J, Jalanka-Tuovinen J, Palva A,
et al. Associations between the human intestinal microbiota, Lactobacillus
rhamnosus GG and serum lipids indicated by integrated analysis of high-
throughput profiling data. PeerJ. 2013;1:e32.
23. Rampelli S, Candela M, Severgnini M, Biagi E, Turroni S, Roselli M, et al. A
probiotics-containing biscuit modulates the intestinal microbiota in the
elderly. J Nutr Health Aging. 2013;17(2):16672.
24. Simon MC, Strassburger K, Nowotny B, Kolb H, Nowotny P, Burkart V, et al.
Intake of Lactobacillus reuteri improves incretin and insulin secretion in
glucose-tolerant humans: a proof of concept. Diabetes Care. 2015;38(10):
182734.
25. McNulty NP, Yatsunenko T, Hsiao A, Faith JJ, Muegge BD, Goodman AL,
et al. The impact of a consortium of fermented milk strains on the gut
microbiome of gnotobiotic mice and monozygotic twins. Sci Transl Med.
2011;3(106):106ra106.
26. Zhang J, Wang L, Guo Z, Sun Z, Gesudu Q, Kwok L, et al. 454 pyrosequencing
reveals changes in the faecal microbiota of adults consuming Lactobacillus
casei Zhang. FEMS Microbiol Ecol. 2014;88(3):61222.
27. Ferrario C, Taverniti V, Milani C, Fiore W, Laureati M, De Noni I, et al. Modulation
of fecal Clostridiales bacteria and butyrate by probiotic intervention with
Lactobacillus paracasei DG varies among healthy adults. J Nutr. 2014;144(11):
178796.
28. Plaza-Diaz J, Fernandez-Caballero JA, Chueca N, Garcia F, Gomez-Llorente C,
Saez-Lara MJ, et al. Pyrosequencing analysis reveals changes in intestinal
microbiota of healthy adults who received a daily dose of
immunomodulatory probiotic strains. Nutrients. 2015;7(6):39994015.
29. marketsandmarkets.com. Probiotic Ingredients Market by Function (Regular,
Preventative, Therapy), Application (Food & Beverage, Dietary Supplements
& Animal Probiotics), Ingredient (Bacteria & Yeast), and by Region - Global
Trends & Forecast to 2020. Available at: http://www.marketsandmarkets.
com/Market-Reports/probiotic-market-advanced-technologies-and-global-
market-69.html (accessed 14 Mar 2016).
Kristensen et al. Genome Medicine (2016) 8:52 Page 10 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
30. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for
systematic reviews and meta-analyses: the PRISMA statement. Ann Intern
Med. 2009;151(4):2649.
31. Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJM, Gavaghan DJ,
et al. Assessing the quality of reports of randomized clinical trials: is blinding
necessary? Control Clin Trials. 1996;17(1):112.
32. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gotzsche PC, Ioannidis JP, et al.
The PRISMA statement for reporting systematic reviews and meta-analyses
of studies that evaluate health care interventions: explanation and
elaboration. PLoS Med. 2009;6(7):e1000100.
33. Higgins JPT, Green S. Cochrane Handbook for Systematic Reviews of
Interventions Version 5.1.0 [updated March 2011]. The Cochrane
Collaboration; 2011. Available at: http://handbook.cochrane.org/.
34. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human
gut microbial gene catalogue established by metagenomic sequencing.
Nature. 2010;464(7285):5965.
35. Haller D, Antoine JM, Bengmark S, Enck P, Rijkers GT, Lenoir-Wijnkoop I.
Guidance for substantiating the evidence for beneficial effects of probiotics:
probiotics in chronic inflammatory bowel disease and the functional
disorder irritable bowel syndrome. J Nutr. 2010;140(3):690s7s.
36. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE,
et al. Diet rapidly and reproducibly alters the human gut microbiome.
Nature. 2014;505(7484):55963.
37. Shoaie S, Ghaffari P, Kovatcheva-Datchary P, Mardinoglu A, Sen P, Pujos-
Guillot E, et al. Quantifying diet-induced metabolic changes of the human
gut microbiome. Cell Metab. 2015;22(2):32031.
38. Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A, et al.
Personalized nutrition by prediction of glycemic responses. Cell. 2015;163(5):
107994.
39. Menard O, Butel MJ, Gaboriau-Routhiau V, Waligora-Dupriet AJ. Gnotobiotic
mouse immune response induced by Bifidobacterium sp. strains isolated
from infants. Appl Environ Microbiol. 2008;74(3):6606.
40. Guarner F, Khan AG, Garisch J, Eliakim R, Gangl A, Thomson A, et al. World
Gastroenterology Organisation Global Guidelines: probiotics and prebiotics
October 2011. J Clin Gastroenterol. 2012;46(6):46881.
41. Hill AB. The Environment and disease: association or causation? Proc R Soc
Med. 1965;58:295300.
42. Cook MT, Tzortzis G, Charalampopoulos D, Khutoryanskiy VV.
Microencapsulation of probiotics for gastrointestinal delivery. J Control
Release. 2012;162(1):5667.
43. Charteris WP, Kelly PM, Morelli L, Collins JK. Development and application of
an in vitro methodology to determine the transit tolerance of potentially
probiotic Lactobacillus and Bifidobacterium species in the upper human
gastrointestinal tract. J Appl Microbiol. 1998;84(5):75968.
44. Farnworth ER. The evidence to support health claims for probiotics. J Nutr.
2008;138(6):1250S4S.
45. Weinstock GM. Genomic approaches to studying the human microbiota.
Nature. 2012;489(7415):2506.
46. Rastogi G, Sani RK. Molecular techniques to assess microbial community
structure, function, and dynamics in the environment. In: Ahmad I, Ahmad F,
Pichtel J, Rastogi G, Sani R, editors. Microbes and Microbial Technology. New
York: Springer; 2011. p. 2957.
47. Claesson MJ, OSullivan O, Wang Q, Nikkila J, Marchesi JR, Smidt H, et al.
Comparative analysis of pyrosequencing and a phylogenetic microarray for
exploring microbial community structures in the human distal intestine.
PLoS One. 2009;4(8):e6669.
48. Candela M, Consolandi C, Severgnini M, Biagi E, Castiglioni B, Vitali B, et al.
High taxonomic level fingerprint of the human intestinal microbiota by ligase
detection reactionuniversal array approach. BMC Microbiol. 2010;10:116.
49. Mizrahi-Man O, Davenport ER, Gilad Y. Taxonomic classification of bacterial
16S rRNA genes using short sequencing reads: evaluation of effective study
designs. PLoS One. 2013;8(1):e53608.
50. Sundquist A, Bigdeli S, Jalili R, Druzin ML, Waller S, Pullen KM, et al. Bacterial
flora-typing with targeted, chip-based Pyrosequencing. BMC Microbiol.
2007;7:108.
51. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid
assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ
Microbiol. 2007;73(16):52617.
We accept pre-submission inquiries
Our selector tool helps you to find the most relevant journal
We provide round the clock customer support
Convenient online submission
Thorough peer review
Inclusion in PubMed and all major indexing services
Maximum visibility for your research
Submit your manuscript at
www.biomedcentral.com/submit
Submit your next manuscript to BioMed Central
and we will help you at every step:
Kristensen et al. Genome Medicine (2016) 8:52 Page 11 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Moreover, the long-term efficacy of probiotics in functionally altering the gut microbiota remains controversial, with most studies demonstrating no substantial changes in overall microbial diversity following probiotic supplementation in humans 108 . Since PJIs can occur anytime during the life of an arthroplasty recipient, the therapy's inability to be efficacious over a patient's lifespan could be a limitation [108][109][110] . If probiotics fail, FMTor other microbial therapeutics (prebiotic, phage, defined microbial consortia, fermented food, and fermentation products) may be explored as an alternative; FMT has been shown to produce persistent changes in the recipient microbial signature 69 . ...
... Given the contributions of the gut microbiota to bone health, probiotics are a potential future therapeutic option in populations of patients requiring or living with hip and knee implants. How-ever, dosages and therapeutic timelines are far from being elucidated, and the ability of probiotic interventions to cause longterm changes in the gut microbiota remains controversial [108][109][110]113 . Finally, nonspecific probiotic treatment may worsen gut health in certain circumstances, highlighting the need for personalized therapies in the future 115 . ...
Article
Full-text available
Background The prevalence of revision surgery due to aseptic loosening and periprosthetic joint infection (PJI) following total hip and knee arthroplasty is growing. Strategies to prevent the need for revision surgery and its associated health-care costs and patient morbidity are needed. Therapies that modulate the gut microbiota to influence bone health and systemic inflammation are a novel area of research. Methods A literature review of preclinical and clinical peer-reviewed articles relating to the role of the gut microbiota in bone health and PJI was performed. Results There is evidence that the gut microbiota plays a role in maintaining bone mineral density, which can contribute to osseointegration, osteolysis, aseptic loosening, and periprosthetic fractures. Similarly, the gut microbiota influences gut permeability and the potential for bacterial translocation to the bloodstream, increasing susceptibility to PJI. Conclusions Emerging evidence supports the role of the gut microbiota in the development of complications such as aseptic loosening and PJI after total hip or knee arthroplasty. There is a potential for microbial therapies such as probiotics or fecal microbial transplantation to moderate the risk of developing these complications. However, further investigation is required. Clinical Relevance Modulation of the gut microbiota may influence patient outcomes following total joint arthroplasty.
... These are live microorganisms which when administered in sufficient amount can bring health benefits to the host body. Few studies that have been done till date demonstrate the potential advantages of probiotics on cancer treatments and recent research has concentrated on the effects of probiotics to prevent the formation and metagenesis of tumors or on the toxicity connected to cancer therapy [79]. A placebo-controlled, double-blind study with 490 patients when administered with a probiotic preparation consisting of 8 strains of lactic acid producing bacteria (Streptococcus thermophilus, Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus delbrueckii subsp. ...
Article
Full-text available
The immune system plays a pivotal role in the battle against cancer, serving as a formidable guardian in the ongoing fight against malignant cells. To combat these malignant cells, immunotherapy has emerged as a prevalent approach leveraging antibodies and peptides such as anti-PD-1, anti-PD-L1, and anti-CTLA-4 to inhibit immune checkpoints and activate T lymphocytes. The optimization of gut microbiota plays a significant role in modulating the defense system in the body. This study explores the potential of certain gut-resident bacteria to amplify the impact of immunotherapy. Contemporary antibiotic treatments, which can impair gut flora, may diminish the efficacy of immune checkpoint blockers. Conversely, probiotics or fecal microbiota transplantation can help re-establish intestinal microflora equilibrium. Additionally, the gut microbiome has been implicated in various strategies to counteract immune resistance, thereby enhancing the success of cancer immunotherapy. This paper also acknowledges cutting-edge technologies such as nanotechnology, CAR-T therapy, ACT therapy, and oncolytic viruses in modulating gut microbiota. Thus, an exhaustive review of literature was performed to uncover the elusive link that could potentiate the gut microbiome’s role in augmenting the success of cancer immunotherapy. Graphical Abstract
... Probiotics have been suggested to have the potential to treat these diseases by modulating the gut microbiota [78]. However, there are still no clear conclusions as to whether or not probiotics alter the diversity or composition of the gut microbiota [33]. Most studies have shown that probiotics did not significantly affect the diversity of the gut microbiota [79,80]. ...
Article
Full-text available
Probiotics are natural microbial agents with beneficial properties such as bacteriostatic and anti-infective properties. Lactobacillus plantarum Q21, Q25 and QA85, were isolated from the Chinese specialty fermented food “Jiangshui” and proved to be highly resistant to Helicobacter pylori (p < 0.0001). In vitro results showed that Q21, Q25 and QA85 strongly inhibited H. pylori and could specifically co-aggregate H. pylori in vitro (more than 56%). Strains have the potential to adhere to cells and hinder H. pylori colonization (p < 0.0001). To assess the anti-H. pylori efficacy of strains in vivo, volunteers were recruited and a self-controlled study of probiotic intervention was conducted. Compared to pre-probiotics, volunteers who took Q21, Q25 and QA85 for 1 month showed significant improvement in discomfort, a significant reduction in GSRS scores (p < 0.05), and modulation of inflammatory response (p < 0.05). Q21, Q25 and QA85 resulted in a decreasing trend of H. pylori load in volunteers (454.30 ± 327.00 vs. 328.35 ± 237.19, p = 0.06). However, the strains were not significantly effective in modulating the imbalance of the gut microbiota caused by H. pylori infection. In addition, strains affect metabolic pathways by increasing the levels of O-Phosphoethanolamine and other related metabolites, which may ameliorate associated symptoms. Therefore, Lactobacillus plantarum Q21, Q25 and QA85 can be regarded as a candidate probiotic preparation that exerts direct or indirect anti-H. pylori effects by inhibiting H. pylori activity and colonization, reducing inflammation and discomfort, maintaining homeostasis in the internal environment, affecting the metabolic pathways and repairing the body barrier. They can play a role in relieving H. pylori infection.
... In the future, we need more targeted characteristic populations and longer-period interventions to continue exploring the potential efficacy of 207-1 as a promising new probiotic. evenness of the composition of the fecal microbiota [44]. Only one study found that probiotic supplementation significantly altered the overall structure of the fecal bacterial community in terms of beta-diversity compared to placebo [45]. ...
Article
Full-text available
Purpose Our study aimed to explore the efficacy of Bifidobacterium breve 207-1 on specific neurotransmitters and hormones and the ability to regulate lifestyle behaviors in healthy adults. Methods In total, 120 healthy adults with high mental stress, overweight, insomnia, and constipation were randomly assigned to receive low-dose B. breve 207-1 (LD, n = 40), high-dose B. breve 207-1 (HD, n = 40), or placebo (n = 40) for 28 days. Fecal and blood samples were collected and questionnaires were answered before and after the trial. Neurotransmitters and serum hormones were detected using enzyme-linked immunosorbent assay. The gut microbiota composition was assessed using 16 S rRNA sequencing. Short–chain fatty acids (SCFAs) concentrations were determined via gas chromatography–mass spectrometry (GC–MS). Results The primary outcome of our study was changes in mental wellness, including neurotransmitters, the hypothalamic–pituitary–adrena (HPA) axis hormones, and the psychological scales. The results showed that γ-aminobutyric acid (GABA) increased significantly and the HPA axis hormones were suppressed overall in the probiotic groups while 5-hydroxytryptamine (5-HT) did not change significantly. However, there was no significant change in mood scale scores. The secondary outcome focused on the ability of 207-1 to regulate the body and lifestyle of healthy adults (e.g., sleep, diet, exercise, etc.). The PSQI scores in the probiotics groups significantly decreased, indicating improved sleep quality. Meanwhile, the probiotic groups had a slight increase in exercise consumption while dietary intake stabilized. By physical examination, the participants showed weight loss although no statistically significant difference was observed between the groups. Then, validated by gut microbiota, changes in the gut microbiota were observed under the effective intervention of 207-1 while short-chain fatty acids (SCFAs) increased in the LD group, particularly acetic and propionic acids. There was a slight decrease in alpha–diversity in the HD group. Conclusion Bifidobacterium breve 207-1 entered the organism and affected neurotransmitter and the HPA axis hormone levels via the microbiome-gut-brain axis. Meanwhile, 207-1 supplementation improved daily lifestyle behaviors in healthy adults, which may in turn lead to changes in their bodies (e.g. weight and lipid metabolism). However, this study did not find significant mood-modulating efficacy. The mechanism of the overall study is unclear, but we hypothesize that SCFAs may be the key pathway, and more experiments are needed for validation in the future. Trial registration This trial was retrospectively registered in the Chinese Clinical Trial Registry under the accession number ChiCTR2300069453 on March 16, 2023.
... Evidence-based investigations have revealed that probiotics can effectively prevent or treat infectious diar-rhea, inflammatory bowel disease, and other intestinal diseases, by interfering with pathogens, improving intestinal barrier function, immunomodulation and neurotransmi er production [12]. Whether the benefits of probiotics relied on the gut microbiota was still unclear, because some intestinal microbiota did not change after probiotics was given [13] (Table 1). Adequate amounts of a wide range of micronutrients are needed by body tissues to maintain health. ...
Article
Full-text available
It is now widely recognized that gut microbiota plays a critical role not only in the development and progression of diseases, but also in its susceptibility to dietary patterns, food composition, and nutritional intake. In this comprehensive review, we have compiled the latest findings on the effects of food nutrients and bioactive compounds on the gut microbiota. The research indicates that certain components, such as unsaturated fatty acids, dietary fiber, and protein have a significant impact on the composition of bile salts and short-chain fatty acids through catabolic processes, thereby influencing the gut microbiota. Additionally, these compounds also have an effect on the ratio of Firmicutes to Bacteroides, as well as the abundance of specific species like Akkermansia muciniphila. The gut microbiota has been found to play a role in altering the absorption and metabolism of nutrients, bioactive compounds, and drugs, adding another layer of complexity to the interaction between food and gut microbiota, which often requires long-term adaptation to yield substantial outcomes. In conclusion, understanding the relationship between food compounds and gut microbiota can offer valuable insights into the potential therapeutic applications of food and dietary interventions in various diseases and health conditions.
... The end products of metabolism, such as vitamins, polyunsaturated fatty acids, conjugated linoleic acid, contribute to the functionality of the intestine and the probiotic properties of bifidobacteria. The end products of bifidobacterial fermentation of complex carbohydrates in the gastrointestinal tract play a key role in human metabolism, as they contribute to the integrity of the epithelial barrier, produce antibacterial compounds, ferment indigestible fiber, stimulate the absorption of water and sodium, reduce the pH of the lumen and bioavailability of toxic amines, promote the production of amino acids and vitamins, prevent colonization of pathogenic microorganisms [6,7,8,9]. There is evidence that acetate produced by bifidobacteria can enhance intestinal protection by epithelial cells and thereby protect the host from E. coli O157:H7 infection [10]. ...
Article
Full-text available
The article provides a brief justification for the need to create new fermented milk products with probiotic properties based on microbial consortia consisting of thermophilic lactic acid streptococcus, propionic acid bacteria and bifidobacteria. The biochemical activity of consortia has been studied. Data on the synthesis of vitamin B12 in various microbial consortia are presented. A high level of viable cells of propionic acid microorganisms and bifidobacteria was shown when co-cultured with thermophilic streptococcus. The results of the conducted studies have shown the possibility of using microbial consortia consisting of thermophilic lactic acid streptococcus, propionic acid bacteria and bifidobacteria for the production of protein fermented dairy bioproducts.
... Some studies show that probiotics induce changes in the GM by increasing potentially beneficial lactobacilli and bifidobacteria and inhibiting potential pathogens in older adults [12,13]. However, other studies and a meta-analysis conclude that, in general, probiotics do not appear to influence the GM composition to any larger extent in healthy adults [14,15]. Interestingly, even though Eloe-Fadrosh et al. [15] found that Lacticaseibacillus rhamnosus GG supplementation in older adults did not influence the overall GM composition, pronounced and consistent alterations in GM functional dynamics (gene expression) were induced by the probiotic [15]. ...
Article
Full-text available
Aging has been associated with a changed composition and function of the gut microbiota (GM). Here, we investigate the effects of the multi-strain probiotic HOWARU® Restore on GM composition and function in seniors. Ninety-eight healthy adult volunteers aged ≥75 years were enrolled in a randomised, double-blinded intervention (NCT02207140), where they received HOWARU Restore (1010 CFU) or the placebo daily for 24 weeks, with 45 volunteers from each group completing the intervention. Questionnaires monitoring the effects on gastro-intestinal discomfort and bowel movements were collected. Faecal samples for GM characterisation (qPCR, 16S rRNA gene amplicon sequencing) and metabolomics (GC-FID, 1H NMR) were collected at the baseline and after 24 weeks. In the probiotic group, self-reported gastro-intestinal discomfort in the form of flatulence was significantly decreased during the intervention. At the baseline, 151 ‘core species’ (present in ≥95% of samples) were identified. Most core species belonged to the Lachnospiraceae and Ruminococcaceae families. Neither alpha diversity nor beta diversity or faecal metabolites was affected by probiotic intake. On the contrary, we observed high intra-individual GM stability, with ‘individual’ accounting for 72–75% of variation. In conclusion, 24 weeks of HOWARU Restore intake reduced gastro-intestinal discomfort in the form of flatulence in healthy seniors without significantly influencing GM composition or activity.
Article
The gut microbiota has been proposed to grant the athlete a metabolic advantage that might be key when optimising performance. While a taxonomic core set of microorganisms characterising the athlete’s gut microbiota has not been delineated, some compositional features might be associated with improved metabolic efficiency, which appears to be driven by the production of bacterial metabolites, such as short-chain fatty acids. Not only long-term exercise but also dietary patterns associated with high-level sports practice contribute to this microbial environment, yet isolating the impact of individual dietary components is challenging. The present review synthetises the available evidence on the compositional aspects of the athlete’s gut microbiota, discusses mechanisms involved in the bidirectional association between exercise and the gut environment, and evaluates the role of athletes’ diet in this interplay. Additionally, a practical approach to indicators commonly reported in metagenomic and metabolomic analyses is provided to explore how these insights can translate to support dietary protocols.
Article
Full-text available
Systematic reviews should build on a protocol that describes the rationale, hypothesis, and planned methods of the review; few reviews report whether a protocol exists. Detailed, well-described protocols can facilitate the understanding and appraisal of the review methods, as well as the detection of modifications to methods and selective reporting in completed reviews. We describe the development of a reporting guideline, the Preferred Reporting Items for Systematic reviews and Meta-Analyses for Protocols 2015 (PRISMA-P 2015). PRISMA-P consists of a 17-item checklist intended to facilitate the preparation and reporting of a robust protocol for the systematic review. Funders and those commissioning reviews might consider mandating the use of the checklist to facilitate the submission of relevant protocol information in funding applications. Similarly, peer reviewers and editors can use the guidance to gauge the completeness and transparency of a systematic review protocol submitted for publication in a journal or other medium.
Article
Full-text available
In recent years, several associations between common chronic human disorders and altered gut microbiome composition and function have been reported. In most of these reports, treatment regimens were not controlled for and conclusions could thus be confounded by the effects of various drugs on the microbiota, which may obscure microbial causes, protective factors or diagnostically relevant signals. Our study addresses disease and drug signatures in the human gut microbiome of type 2 diabetes mellitus (T2D). Two previous quantitative gut metagenomics studies of T2D patients that were unstratified for treatment yielded divergent conclusions regarding its associated gut microbial dysbiosis. Here we show, using 784 available human gut metagenomes, how antidiabetic medication confounds these results, and analyse in detail the effects of the most widely used antidiabetic drug metformin. We provide support for microbial mediation of the therapeutic effects of metformin through short-chain fatty acid production, as well as for potential microbiota-mediated mechanisms behind known intestinal adverse effects in the form of a relative increase in abundance of Escherichia species. Controlling for metformin treatment, we report a unified signature of gut microbiome shifts in T2D with a depletion of butyrate-producing taxa. These in turn cause functional microbiome shifts, in part alleviated by metformin-induced changes. Overall, the present study emphasizes the need to disentangle gut microbiota signatures of specific human diseases from those of medication.
Article
Full-text available
Assessment and characterization of gut microbiota has become a major research area in human disease, including type 2 diabetes, the most prevalent endocrine disease worldwide. To carry out analysis on gut microbial content in patients with type 2 diabetes, we developed a protocol for a metagenome-wide association study (MGWAS) and undertook a two-stage MGWAS based on deep shotgun sequencing of the gut microbial DNA from 345 Chinese individuals. We identified and validated approximately 60,000 type-2-diabetes-associated markers and established the concept of a metagenomic linkage group, enabling taxonomic species-level analyses. MGWAS analysis showed that patients with type 2 diabetes were characterized by a moderate degree of gut microbial dysbiosis, a decrease in the abundance of some universal butyrate-producing bacteria and an increase in various opportunistic pathogens, as well as an enrichment of other microbial functions conferring sulphate reduction and oxidative stress resistance. An analysis of 23 additional individuals demonstrated that these gut microbial markers might be useful for classifying type 2 diabetes.
Article
OBJECTIVES: Irritable bowel syndrome (IBS) and chronic idiopathic constipation (CIC) are functional bowel disorders. Evidence suggests that disturbance in the gastrointestinal microbiota may be implicated in both conditions. We performed a systematic review and meta-analysis to examine the efficacy of prebiotics, probiotics, and synbiotics in IBS and CIC. METHODS: MEDLINE, EMBASE, and the Cochrane Controlled Trials Register were searched (up to December 2013). Randomized controlled trials (RCTs) recruiting adults with IBS or CIC, which compared prebiotics, probiotics, or synbiotics with placebo or no therapy, were eligible. Dichotomous symptom data were pooled to obtain a relative risk (RR) of remaining symptomatic after therapy, with a 95% confidence interval (CI). Continuous data were pooled using a standardized or weighted mean difference with a 95% CI. RESULTS: The search strategy identified 3,216 citations. Forty-three RCTs were eligible for inclusion. The RR of IBS symptoms persisting with probiotics vs. placebo was 0.79 (95% CI 0.70-0.89). Probiotics had beneficial effects on global IBS, abdominal pain, bloating, and flatulence scores. Data for prebiotics and synbiotics in IBS were sparse. Probiotics appeared to have beneficial effects in CIC (mean increase in number of stools per week=1.49; 95% CI=1.02-1.96), but there were only two RCTs. Synbiotics also appeared beneficial (RR of failure to respond to therapy=0.78; 95% CI 0.67-0.92). Again, trials for prebiotics were few in number, and no definite conclusions could be drawn. CONCLUSIONS: Probiotics are effective treatments for IBS, although which individual species and strains are the most beneficial remains unclear. Further evidence is required before the role of prebiotics or synbiotics in IBS is known. The efficacy of all three therapies in CIC is also uncertain.
Article
Elevated postprandial blood glucose levels constitute a global epidemic and a major risk factor for prediabetes and type II diabetes, but existing dietary methods for controlling them have limited efficacy. Here, we continuously monitored week-long glucose levels in an 800-person cohort, measured responses to 46,898 meals, and found high variability in the response to identical meals, suggesting that universal dietary recommendations may have limited utility. We devised a machine-learning algorithm that integrates blood parameters, dietary habits, anthropometrics, physical activity, and gut microbiota measured in this cohort and showed that it accurately predicts personalized postprandial glycemic response to real-life meals. We validated these predictions in an independent 100-person cohort. Finally, a blinded randomized controlled dietary intervention based on this algorithm resulted in significantly lower postprandial responses and consistent alterations to gut microbiota configuration. Together, our results suggest that personalized diets may successfully modify elevated postprandial blood glucose and its metabolic consequences. VIDEO ABSTRACT.
Article
Objective: To evaluate the potential for diagnosing colorectal cancer (CRC) from faecal metagenomes. Design: We performed metagenome-wide association studies on faecal samples from 74 patients with CRC and 54 controls from China, and validated the results in 16 patients and 24 controls from Denmark. We further validated the biomarkers in two published cohorts from France and Austria. Finally, we employed targeted quantitative PCR (qPCR) assays to evaluate diagnostic potential of selected biomarkers in an independent Chinese cohort of 47 patients and 109 controls. Results: Besides confirming known associations of Fusobacterium nucleatum and Peptostreptococcus stomatis with CRC, we found significant associations with several species, including Parvimonas micra and Solobacterium moorei. We identified 20 microbial gene markers that differentiated CRC and control microbiomes, and validated 4 markers in the Danish cohort. In the French and Austrian cohorts, these four genes distinguished CRC metagenomes from controls with areas under the receiver-operating curve (AUC) of 0.72 and 0.77, respectively. qPCR measurements of two of these genes accurately classified patients with CRC in the independent Chinese cohort with AUC=0.84 and OR of 23. These genes were enriched in early-stage (I-II) patient microbiomes, highlighting the potential for using faecal metagenomic biomarkers for early diagnosis of CRC. Conclusions: We present the first metagenomic profiling study of CRC faecal microbiomes to discover and validate microbial biomarkers in ethnically different cohorts, and to independently validate selected biomarkers using an affordable clinically relevant technology. Our study thus takes a step further towards affordable non-invasive early diagnostic biomarkers for CRC from faecal samples.