ArticlePDF AvailableLiterature Review

Abstract and Figures

The importance of the gut microbiome in human health and disease is fully acknowledged. A perturbation in the equilibrium among the different microbial populations living in the gut (dysbiosis) has been associated with the development of several types of diseases. Modulation of the gut microbiome through dietary intervention is an emerging therapeutic and preventive strategy for many conditions. Nevertheless, interpersonal differences in response to therapeutic treatments or dietary regimens are often observed during clinical trials, and recent research has suggested that subject-specific features of the gut microbiota may be responsible. In this review, we summarize recent findings in personalized nutrition, highlighting how individualized characterization of the microbiome may assist in designing ad hoc tailored dietary intervention for disease treatment and prevention. Moreover, we discuss the limitations and challenges encountered in integrating patient-specific microbial data into clinical practice.
Content may be subject to copyright.
Inflamm Bowel Dis
• Volume 24, Number 10, October 2018 2142
CliniCal Review aRtiCle
Dietary Interventions to Modulate the Gut Microbiome—How
Far Away Are We From Precision Medicine
FrancescaDe Filippis, PhD,*, PaolaVitaglione, PhD,*, RosarioCuomo, MD,,
RobertoBerni Canani, PhD,,§,, and DaniloErcolini, PhD*,
The importance of the gut microbiome in human health and disease is fully acknowledged. Aperturbation in the equilibrium among the different
microbial populations living in the gut (dysbiosis) has been associated with the development of several types of diseases. Modulation of the gut
microbiome through dietary intervention is an emerging therapeutic and preventive strategy for many conditions. Nevertheless, interpersonal
differences in response to therapeutic treatments or dietary regimens are often observed during clinical trials, and recent research has suggested
that subject-specic features of the gut microbiota may be responsible. In this review, we summarize recent ndings in personalized nutrition,
highlighting how individualized characterization of the microbiome may assist in designing ad hoc tailored dietary intervention for disease treat-
ment and prevention. Moreover, we discuss the limitations and challenges encountered in integrating patient-specic microbial data into clinical
practice.
Key Words: personalized nutrition, gut microbiome, dietary intervention, clinical trials
INTRODUCTION
The human body is home to at least 100 trillion (1014)
microorganisms, most of them inhabiting the human gut. This
community includes taxa from the 3 domains of life (Bacteria,
Eukarya, and Archaea) and viruses, whose combined genome
harbors at least 100 times as many genes as our own genome.1
The consortium of microbial symbionts that resides in and
on the human body collectively constitutes our “microbiota”;
when also considering their pool of genes and the functions
for which they encode, we refer to it as the “microbiome.”
The human body can be considered the result of human and
microbial cells, with our genetic and metabolic potential repre-
senting the arrangement of what comes from our genome and
microbiome. For these reasons, our gut microbiome has earned
the appellative of our “other genome.”2
Several studies have focused on the exploration of the
composition and functionality of the gut microbiome, with an
emphasis on its bacterial members.3 In healthy adults, 2 bac-
terial phyla dominate, namely Firmicutes and Bacteroidetes,
which vary in their proportions across the population.
Proteobacteria, Actinobacteria, and Verrucomicrobia are
present at lower levels.4 A categorization of individuals in 3
groups (“enterotypes”) based on the prevalence of Prevotella,
Bacteroides, or Ruminococcus in the gut microbiota was pro-
posed in an attempt to simplify the complexity of the gut
microbiome.5 This classication, although appealing for under-
standing microbial changes in health and disease, has recently
been criticized, as it may lead to an oversimplied vision of the
gut microbiome, whereas the existence of smooth gradients in
the abundance of dominant taxa is now considered to be more
plausible.6, 7
In this review, we focus on the role of diet in inuenc-
ing the composition and functions of the microbiome with
a particular emphasis on inammatory bowel disease (IBD)
and irritable bowel syndrome (IBS). We also discuss the most
recent evidence for the role of microbiome-targeted dietary
interventions for promoting host health.
MICROBIAL METABOLITES IN THEGUT
Microbial symbionts interact among themselves and
with the host, impacting human physiology and health. They
relate to the host immune system, promoting the maturation
of immune cells and driving the development of immune
functions.8, 9 Moreover, gut microbes perform a wide range of
useful activities, such as fermenting and absorbing undigested
compounds and synthesizing vitamins.10, 11 They can inuence
host health through the production of benecial or detrimen-
tal metabolites. Such compounds can be derived from both
metabolic intermediates of the host and dietary precursors.
Therefore, diet plays a major role in affecting the metabolic
Received for publications October 20, 2017; Editorial Decision January 8, 2018.
From the *Department of Agricultural Sciences, Division of Microbiology,
Task Force on Microbiome Studies, Department of Clinical Medicine and
Surgery, §Department of Translational Medical Science, European Laboratory for
Investigation on Food Induced Diseases, and Ceinge Advanced Biotechnologies,
University of Naples Federico II, Naples, Italy
© 2018 Crohn’s & Colitis Foundation.
Published by Oxford University Press. All rights reserved.
For permissions, please e-mail: journals.permissions@oup.com.
Conicts of interest: The authors have no conicts of interest to declare.
Supported by: The study was in part supported by the grant DINAMIC (Diet-
induced Arrangement of the gut Microbiome for Improvement of Cardiometabolic
health) funded within the Joint Programming Initiative “A Healthy Diet for a Healthy
life” (JPI HDHL) - Joint Action “Intestinal Microbiomics”.
Correspondence: Danilo Ercolini, PhD, Department of Agricultural Sciences,
Division of Microbiology, University of Naples Federico II, via Università, 100 -
80055 Portici, Italy (ercolini@unina.it).
doi: 10.1093/ibd/izy080
Published online 13 April 2018
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2143
Dietary Interventions to Modulate the Gut Microbiome
potential of gut microbes. Some specic gut microbiota mem-
bers can produce short-chain fatty acids (SCFAs) from degrad-
ation of complex plant-derived polysaccharides. SCFAs, mainly
butyrate, propionate, and acetate, exert recognized health-pro-
moting functions, such as anti-inammatory, anticarcino-
genic, and immune-regulatory functions.12, 13 Nevertheless,
gut microbiota can also be responsible for the production of
harmful molecules associated with detrimental effects for the
host and related to the development of several diseases.12, 13
Ahigh-protein diet leads to the accumulation of several amino
acid–derived products in the colon, such as branched-chain
fatty acids, phenols, p-cresol, and phenylacetic acid, previously
reported to be associated with pro-inammatory and carcino-
genic effects.13, 14 Sulfate-reducing bacteria (eg,, Desulfovibrio
spp.) produce suldes through the catabolism of sulfur amino
acids and taurine, which are toxic to colonocytes.13 N-Nitroso
compounds (NOCs) produced by nitration of amines derived
from microbial fermentation of proteins may exert carcinogenic
and mutagenic effects and are correlated with the incidence of
colorectal cancer.13 In addition, a diet rich in fat attracts more
bile in the colon, where bile acids may be converted by micro-
bial enzymes into secondary bile acids, mainly deoxycholic and
lithocholic acids. These can be involved in processes linked to
colorectal carcinogenesis, such as apoptosis, cell proliferation,
and DNA damage induction.14
MICROBIOMERELATED DISORDERS
In recent years, the “1 microbe–1 disease” model has
been regarded as extremely simplistic and obsolete, whereas
an increasing number of studies of the human microbiome
have highlighted that many diseases may be a consequence of
an overall dysbiosis status of the gut microbiome. Several gut
commensal microbes may be considered pathobionts; that is,
they normally inhabit the gut but may act as pathogens and
cause disease when the normal gut microbial community is
altered.15 Indeed, perturbation of the equilibrium in the gut
microbiome has been linked to the development of several
disorders. As they are extensively reviewed elsewhere,16–18 we
provide a short overview below, with particular emphasis on
IBD andIBS.
Obesity: Gut microbiota dysbiosis in obesity is often associ-
ated with increased Firmicutes-to-Bacteroidetes ratio19 and
persistent inammation triggered by increased systemic levels
of lipopolysaccharides arising from Gram-negative bacterial
cells (metabolic endotoxemia).20, 21 Indeed, the perturbation
in the gut microbiota composition caused by the consump-
tion of a high-fat diet leads to amplied gut permeability,
resulting in endotoxemia.21
Type 2 diabetes and metabolic syndrome: The gut micro-
biome plays an important role in modulating the glycemic
response, and thus in the development of type 2 diabetes
(T2D). The gut microbiota of T2D patients is usually
depleted of ber-degrading and SCFA-producing bacteria,
such as Roseburia, Eubacterium, and Faecalibacterium.22
• Cardiovascular diseases: Gut microbiota metabolism of
choline, phosphatidylcholine, and carnitine leads to trimeth-
ylamine (TMA), which is oxidized in the liver to trimethyl-
amine oxide (TMAO) and seems to be associated with the
development of atherosclerotic plaques.23, 24
• Immune-mediated adverse food reactions: food allergy and
celiac disease. These conditions are characterized by an
abnormal response of the immune system to specic food
antigens that do not affect the normal population. Agenetic
predisposition is involved in the development of these con-
ditions, but environmental factors acting at least in part on
gut microbiota could be also implicated.25 Current evidence
suggests that the gut microbiota and its metabolites, together
with exposure to dietary factors in early life, critically inu-
ence the immunology of T-cells.26
Colorectal cancer (CRC): Some bacterial species are suspected
to be involved in CRC development.27 Although the exact
mechanisms remain unknown, the production of pro-inam-
matory or pro-oxidative metabolites and/or toxins, such as
Bacteroides fragilis BFT toxin,27 and the transformation of
primary bile acids into secondary bile acids13 may be involved.
Neurological disorders: Gut microbiota may interact with
the nervous system through the production of several neu-
ro-active molecules and modulate brain development, func-
tions, human mood, and behavior.28, 29 The complexity of
these interactions is implied by the term “gut-brain axis.”30
Moreover, recent evidence suggests that the gut microbiome
is implicated in the etiology of autism spectrum disorder,
Parkinson’s, Alzheimer’s, and other neurodegenerative dis-
eases, although the mechanisms underlying these diseases
remain unclear.31–33
GUT MICROBIOME IN IBD ANDIBS
Inammatory Bowel Diseases
Inammatory bowel diseases are chronic intestinal dis-
eases characterized by inammation of the bowel, including
Crohn’s disease (CD) and ulcerative colitis (UC). UC is man-
ifested exclusively in colonic mucosa, whereas CD affects all
areas of the gastrointestinal (GI) tract, with features such as
granulomas and intestinal brosis. IBD affects up to 0.5% of
the population, with the highest incidence in North America
and Europe.34 The incidence is increasing, especially in devel-
oping countries, with the rate of CD rising faster than that of
UC.35 This phenomenon seems to be related to changes in the
Western lifestyle, suggesting that environmental factors, par-
ticularly diet, play a role in the development and progression
of IBD.36
Furthermore, eating habits also seem to be a key factor
in the clinical care of patients with IBD. Indeed, acute CD
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2144
De Filippis etal
treatment with an elemental diet resulted in a remission com-
parable to treatment with corticosteroids.37 As dietary and
bacterial antigens are the most common types of luminal anti-
gen, it is reasonable to suppose that dietary factors may play
an important role in the pathogenesis of IBD, possibly inter-
acting with the gut microbiota and the mucosal immune sys-
tem.38 Gut microbiota dysbiosis was often observed in IBD
patients, although a causal relationship has not been estab-
lished.39 Some studies reported a decrease of Firmicutes in
IBD patients, in particular the butyrate-producing Roseburia
hominis and Faecalibacterium prausnitzii.40, 41 Conversely,
Proteobacteria, particularly Escherichia coli,40, 42 are com-
monly increased in patients with IBD, whereas the implication
of some pathogenic bacteria, such as Mycobacterium avium
paratuberculosis (MAP), adherent invasive Escherichia coli
(AIEC), Clostridium difcile, Campylobacter, and Salmonella,
is still controversial.39 In addition, CD patients also showed a
higher proportion of fungi compared with bacteria, with an
increased Basidiomycota:Ascomycota ratio, a decreased propor-
tion of Saccharomyces cerevisiae, and an increased abundance
of Candida albicans.43, 44 However, whether the dysbiosis has a
causative role in the onset of the inammation that drives IBD
development or inammation arises independently and leads to
dysbiosis is a matter of ongoing debate.39, 45
Irritable Bowel Syndrome
IBS is a chronic disorder characterized by abdominal pain
related to defecation or changes in bowel habits.46 Several anal-
yses reported a global prevalence of 14% among females and
9% among males.47 IBS is commonly categorized into subtypes
according to the predominant bowel habit: diarrhea-predomi-
nant, constipation-predominant, or mixed/alternating.46 Several
factors seem to be involved in the development of IBS: visceral
hypersensitivity, altered brain-gut signaling, immune dysreg-
ulation, psychosocial factors, and microbiota modication.48
However, most IBS patients report symptoms being triggered
by specic foods; this causes them to limit or exclude these food
items, with consequent changes in dietary habits for the manage-
ment of IBS.49 IBS patients often show an increase in Firmicutes,
particularly unclassied Clostridium cluster IV and XIV, known
SCFA producers.50 Abnormal levels of butyrate can promote vis-
ceral hypersensitivity and atypical intestinal contractions, which
are the primary clinical manifestations of IBS.50, 51 Moreover,
higher levels of mucin-degrading bacteria, such as Ruminococcus
torques and Akkermansia muciniphila, were found. These spe-
cies can degrade the intestinal mucus barrier and therefore may
explain the gut inammatory status in these subjects.50
Microbiome anddiet
The gut microbiome may be inuenced by several factors,
among which diet may be considered the most important. The
long-term habitual diet seems to be the primary factor inu-
encing the gut microbiota. Several recent studies focused on
the co-evolution between humans and their gut microbiota to
understand to what extent the Westernization of diet and life-
style have impacted our microbial symbionts and how this has
affected human health. To this end, it is fundamental to study
rural and traditional African or South American populations,
whose lifestyles likely resemble those of Paleolithic or Neolithic
humans.14, 52–54 Consistent differences in the gut microbiome have
been found. Westernization induced a loss of microbial diver-
sity and the disappearance of specic taxa, with consequent
reduction in the capability to degrade complex polysaccharides
and to produce benecial metabolites from ber utilization.55
Agrarian populations around the world, habitually consuming
a diet rich in fruit, vegetables, and brous tubers and lacking
in animal products, often show an enrichment in ber-degrad-
ing bacteria, such as Prevotella, Lachnospira, Treponema, and
Xylanibacter.14, 52, 54 These changes in gut microbiota compos-
ition are reected in its functionality. Indeed, agrarians also
showed a different metabolome compared with Western sub-
jects, with higher levels of benecial metabolites of microbial
origin, for example, SCFA.14, 52, 54 Consistently, the consumption
of a diet rich in ber in Western subjects (vegetarian/vegan or
Mediterranean-style diet) promotes higher levels of Prevotella
and Lachnospira in the gut microbiota, boosting the production
of benecial SCFA.56 Nevertheless, studies also highlighted the
possibility of inducing changes in the gut microbiome through
a dietary intervention, although these changes are often tran-
sitory, and the gut microbiota tends to revert to the original
condition.57 The possibility of modulation of gut microbiota
composition and activity through ad hoc dietary interventions
is not only fascinating but also a promising research avenue in
the prevention and treatment of disease.
DIET, GUT MICROBIOTA, AND IBDIBS
TREATMENT
Several studies conrmed an association between high
animal protein and fat intake and increased risk of develop-
ing IBD, both in children and in adults,45, 58 whereas a diet rich
in olive oil, sh, vegetables, fruits, grains, and nuts has been
inversely associated with CD in children.59 Moreover, several
recent studies showed a protective role of breastfeeding against
the risk of IBD in pediatric patients, although the mechanism
is largely unclear.60
Based on the dietary recommendations of the National
Institute for Health and Care Excellence (NICE) and the
British Dietetic Association,61, 62 the “traditional” IBS diet is
based on “healthy eating,” reducing fats, caffeine, and excessive
ber intake, and avoiding soft drinks and gas-producing foods.
Furthermore, patients are advised to eat slowly and to chew
meticulously.61, 62
Several dietary therapies have been proposed for the
treatment of IBD/IBS patients or symptom relief; these have
been extensively and recently reviewed.45, 63 However, their ef-
cacy is still controversial, and little is known about the effects
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2145
Dietary Interventions to Modulate the Gut Microbiome
of these dietary treatments on the gut microbiome and thus on
human health.
Specic CarbohydrateDiet
SCD is based on the restriction of complex carbohy-
drates and the exclusion of rened sugars from the diet, based
on the rationale that the sugars and complex carbohydrates are
badly absorbed and could promote intestinal inammation.64
SCD may contain almond, nut, and coconut ours and exclude
grains (wheat, rice, corn); most dairy products are avoided,
except for homemade yogurt fermented for 24 hours to deplete
lactose. Two retrospective studies that assessed SCD as the sole
method of treatment in CD children showed that the use of
SCD for 5 to 30months had positive effects on inammatory
markers and clinical presentation of the disease. The direct
reason for the improvement was not known, but modication
of the intestinal microbiome was proposed as one of the rea-
sons.65, 66
Exclusive Enteral Nutrition
Exclusive enteral nutrition (EEN), is likely to have sub-
stantial effects on gut microbiota, in part due to carbohydrate
composition. EEN therapy with elemental, semi-elemental,
or polymeric formula diets has been widely studied; it is the
only dietary intervention that induced remission of Crohn’s
disease and is therefore the rstline therapy in many parts of
the world. In addition to reducing symptoms, EEN has been
associated with mucosal healing, which may be a superior pre-
dictor of long-term outcome.67 The results about the effect of
EEN on gut microbiota are still preliminary and conicting.
Lewis and coworkers44 showed that EEN induces changes in the
gut microbiome of CD children, but these changes did not lead
to a microbiome resembling that of healthy controls, whereas
Shiga and collaborators68 reported a reduction of Bacteroides
fragilis group, commonly found in inammatory lesions of
CD patients. Markers commonly associated with health, such
as microbial diversity or abundance of butyrate-producing
microbes (eg,, Faecalibacterium prausnitzii) and butyrate levels
were decreased during EEN intervention.69, 70 Another inter-
esting observation was the higher efcacy of EEN in ileal CD
compared with colonic CD or UC.71 However, identifying the
exact role of the diet is hampered by interindividual variabil-
ity in the microbial community and response to intervention.
The role of specic bacteria in disease improvement is yet to be
conrmed, as conicting data have been reported on the protec-
tive effect of Faecalibacterium prausnitzii and other microbes in
pediatric patients.72, 73
Low-FODMAPsDiet
A recent dietary strategy proposed for IBD/IBS man-
agement is the low–fermentable oligo-, di- and monosaccha-
rides and polyols (FODMAPs) diet.45, 63, 74 Diets rich in highly
fermentable but poorly absorbed short-chain carbohydrates
and polyols are hypothesized to trigger symptoms in patients
with IBS.75 The digestibility of carbohydrates varies according
to the absence, or reduced production, of hydrolase enzymes
needed for their digestion. When these sugars arrive undigested
in the colon, they cause an osmotic effect that attracts water
in the small intestine. Moreover, they can be fermented by the
colonic microbiota, with accumulation of gases, including
hydrogen and methane. Both water and gas increase intestinal
volume, which, together with visceral hypersensitivity, causes
pain. However, other mechanisms are evoked in the injurious
effect of FODMAPs. Indeed, these molecules seem to increase
GI motility, which further reduces absorption and increases
potential colonic fermentation.76 A low-FODMAPs diet can
reduce the production of SCFAs as a result of limited avail-
ability of fermentable substrates and decreased levels of taxa
involved in SCFA production.77 Because SCFAs induce vis-
ceral hypersensitivity, the decrease in their luminal levels may
represent another mechanism accounting for the efcacy of a
low-FODMAPs diet.78, 79
FODMAPs might be considered “fast food” for bac-
teria and can lead to the expansion of specic bacterial pop-
ulations in the distal small intestine. This bacterial overgrowth
is similar in patients with IBS, celiac disease, and Crohns dis-
ease.80, 81 Bacterial overgrowth in the small intestine has been
associated with increased small intestinal permeability. Fructo-
oligosaccharides increase intestinal permeability also in the
colon, and in rats experimentally infected with Salmonella,
FODMAPs predispose to severe colitis, in comparison with a
mild colonic inammation occurring in controls.82 It is worth
emphasizing that rapidly fermentable ber compromises the
proximal large bowel, whereas concurrent ingestion of slowly
fermentable or nonfermentable substrates decreases the rate
of fermentation, shifting it more distally in the colon.83 This
mechanism could also be explained by the different localiza-
tion of the inammatory processes. Hence, the pathogenic
hypothesis of inammation development in IBD considers an
initial increase in fermentable sugar (eg,, FODMAPs) con-
sumption, which determines bacterial overgrowth and a con-
sequent increase in intestinal permeability. Bacterial or antigen
translocation triggers an immunologic response and acti-
vation of the inammatory cascade. Data from the literature
suggest that a low-FODMAPs diet decreases gut symptoms in
IBD and IBS patients, also promoting a shift in the gut micro-
biota.45, 48, 83 In a single-blind randomized clinical trial (RCT) in
patients with IBS, a low-FODMAPs diet promoted an increased
richness of Actinobacteria, Firmicutes, and Clostridiales
and a decreased abundance of bidobacteria compared with
a high-FODMAPs diet.83 In the same study, a higher abun-
dance of the hydrogen-consuming genus Adlercreutzia was
found in the low-FODMAPs group, possibly contributing
to a reduction of symptoms, due to the consumption of the
accumulated gas in the intestine.83 Consistently, another study
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2146
De Filippis etal
reported a decreased proportion of bidobacteria in patients
with IBS after a low-FODMAPs diet.75 In patients with Crohn’s
disease, FODMAPs intake is also associated with changes in
fecal microbiota. Indeed, an Australian diet rich in FODMAPs
boosted the levels of butyrate-producing Clostridium cluster
XIVa and mucus-associated Akkermansia muciniphila. This diet
seems to increase IBS-like symptoms.75 Moreover, Chumpitazi
and coworkers84 observed that children with IBS showing a
positive response to a 2-day low-FODMAP intervention had
higher baseline abundance of Sporobacter and Subdoligranum
and decreased Bacteroides compared with nonresponders.
Modulation of FODMAPs intake seems to be a promising
strategy for treating abdominal symptoms in patients with
IBD and IBS. However, potential hazards do exist, such as the
decrease in bidobacteria and other ber-degrading bacteria,
recognized for their health-promoting benets, along with the
consequent reduction of benecial SCFA levels.85 Therefore,
the routine utilization of this strategy for IBD/IBS treatment
should be applied carefully, ideally coupled with gut microbiota
monitoring, to avoid induction of dysbiosis. Considering these
factors, the addition of probiotics during a low-FODMAPs
diet may be a possible solution to optimize clinical manage-
ment and prevent the detrimental effects of a low-FODMAPs
diet on the gut microbiome.
PERSONALIZED NUTRITION AND GUT
MICROBIOME
Key Factors in a Dietary Intervention for
Personalized Clinical Applications
Clinical interventions cannot be based solely on clinical
experience and observational studies but need to be based on
evidence provided by RCTs.86, 87 Despite their cost and labor-in-
tensiveness, RCTs are the most valid research design for evi-
dence-based medicine; nutrition is a eld in need of better
clinical research.88 This gap is the focus of the European Clinical
Research Infrastructures Network (ECRIN) Integrating
Activity (IA; http://www.ecrin.org/activities/projects), which
has identied barriers to good clinical research and offered
solutions to improve evidence-based clinical practice.89–91
Critical Factors in Nutritional Studies
The main difculty in identifying a causal relationship
between the consumption of a specic diet, food, or nutrient
and a health outcome is the coexistence of several factors that
interact and may possibly lead to biased results if not appropri-
ately considered. These factors include population characteris-
tics and lifestyle; the bioavailability of the nutrient (which is in
turn inuenced by aspects related to the technology of produc-
tion and method of consumption); the timing, frequency, and
duration of nutrient exposition; and contextual factors (eg,,
foods, supplements, medications, and diseases that can aid or
hinder absorption).
Therefore, to demonstrate a causal relationship of a
specic nutrient/diet on a health outcome, a multidisciplinary
and multisystem approach is necessary. This approach requires
simultaneously considering nutrition, genetics, the microbi-
ome, and environment (exposome) in the implementation of
nutritional intervention studies, as was recently suggested in
an epidemiological study.92 This strategy could have multiple
spillover effects, including being able to explain and overcome
the discrepancies present in the scientic literature for the ef-
cacy of some nutrients/diets, achieving stronger evidence and
consensus in the scientic community, and producing practical
and valuable applications in the elds of personalized nutri-
tion, preventive and precision medicine, and functional food
development.
Critical factors in the implementation of nutritional
intervention studies and opportunities beyond nutrition are
summarized in Figure1; a systems-based approach could work
in nutritional interventions and specically in nutritional RCTs.
The scheme highlights 4 critical points in RCTs, namely, study
design, subjects, biomarkers, and data analysis. Each of them
must be carefully considered during the implementation of the
trial tailored to nutrient/diet and targeted health outcomes.
Studydesign
The type and form of nutrient, whether provided as a
supplement or included in a food, can considerably inuence
the study design. Indeed, in the case of a supplement, a placebo
(a pill/powder/tablet/liquid similar to the experimental one but
without the putative bioactive compound) can be provided and
a double-blind trial (which is the gold standard design because
it guarantees the blinding of both subjects and investigators)
can be performed. Conversely, in the case of a nutrient included
in a food or a diet, nding the correct control intervention can
be a remarkably critical point. This affects the study design and
the data analysis. In the frame of study design, blinding can be
compromised because, without a placebo, a double-blind trial
is impossible and the dose/frequency of food consumption and
the duration of the intervention must be carefully considered
in view of the bioavailability of the putative bioactive nutrient
in the food.
Sample size calculation and studypower
Power analysis for sample size calculation is usually car-
ried out based on variations in clinical parameters. Calculating
sample size based on microbiome features, although possible,
may often be impracticable, as we do not know the effect that
a specic dietary intervention may have on the complex con-
sortium of gut microorganisms, with diverse taxa responding
in different ways to the treatment. Focusing on the abundance
of only a few taxa may be also unsuccessful. Moreover, due
to the high interpersonal variability in gut microbiome com-
position, the resulting sample size would be excessively high,
making the study unfeasible. As many of the studies found
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2147
Dietary Interventions to Modulate the Gut Microbiome
in the literature are based on a small number of subjects, the
effects on the microbiome may be hidden by high interpersonal
variability, and this may explain the often inconclusive results
obtained in terms of the modulatory effect on the microbiome.
Tools for sample size calculation in microbiome studies have
been recently developed; however, their use remains limited.93, 94
Subjects selection
Subjects’ characteristics, such as age, nutritional and
health status, genetics, the gut microbiome, and behavioral
and lifestyle factors may enormously inuence the metabolic
response and the health effect of a nutrient/diet. Thus, having
clear and strict inclusion and exclusion criteria in RCTs may
allow investigators to have interventions with smaller numbers
of subjects while maintaining a good power of the study to
reduce costs and time of the trials and to enhance the probabil-
ity of clarifying the mechanisms underlying the health effect.
The latter aspect is also inuenced by biomarkers of effective-
ness monitored during the study. In the case of clinical trials
involving patients, the evaluation of markers of the specic
FIGURE1. Critical factors in the implementation of nutritional intervention studies and opportunities beyond nutrition. Randomized clinical trials
represent the gold standard to achieve evidence-based medicine. Eective nutritional RCTs must be tailored to the specic nutrient/diet and health
outcome of interest. The scientic validity of evidence achieved by RCTs depends on the critical management of 4 main points, namely study design,
subjects, biomarkers of responsiveness, and data analysis. Numerous factors coexist and interact between each other to inuence the eect of nutri-
ent/diet on a specic health outcome. These factors mainly include population characteristics and lifestyle, dose and bioavailability of the nutrient,
timing, frequency, and duration of nutrient exposition, and contextual factors. Due to the involvement of the gut microbiome in the ne interplay
between nutrient/diet-subjects-health, its consideration at multiple steps of RCTs (ie, at selection of subjects, biomarkers, and data analysis) may
be crucial. The proper consideration and management of all the critical factors allows scientists to achieve evidence that can nd application in the
elds of personalized nutrition, preventive and precision medicine, and functional food development.
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2148
De Filippis etal
disease under investigation is necessary. In addition, other bio-
markers that are usually considered in subjects at a predisease
stage may also be monitored. These include inammatory,
nutritional, and metabolic biomarkers, and gut microbiota
composition. Most belong to the ne net of signaling medi-
ators involving the metabolic, neuroendocrine, and immune
systems; orchestrating the response to a nutrient/diet; and the
organ-by-organ communication and homeostasis processes
within the body.11, 95, 96 Finally, to monitor changes in the gut
microbiome, antibiotic or probiotic assumption should be con-
sidered as an exclusion criterion, or, when this is not possible
(eg, when intervention is addressed to patients), investigators
should consider it a possible confounding factor.
Biomarker evaluation and data analysis
In the frame of data analysis, the evaluation of individ-
ual compliance is the critical factor. Indeed, in the presence of
a supplement/placebo, the count of not-consumed doses may
be sufcient, whereas in the case of a food/diet-based interven-
tion, the evaluation of individual dietary intake (through food
frequency questionnaires, 24-hour recall, weighed food record,
etc.) or biomarkers of food/diet intake in biological uids
(through metabolomics analysis) is necessary. Although univo-
cal biomarkers of intake have not been recognized for many
foods, metabolomics of biological uids is inestimably valuable
for RCTs because it can show the impact of the intervention on
individual nutrient metabolism in relation to the subjects’ char-
acteristics being considered in the study. This aspect is funda-
mental in targeted nutrition and medicine. Mounting evidence
shows that through metabotyping—that is, grouping metabol-
ically similar individuals—personalized nutritional and phar-
macological strategies may be achieved.92, 97 On the other hand,
the approach of grouping similar individuals for some features
can be applied in RCTs both at the stage of subject selection
and during data analysis.
Overall, a multisystem approach in clinical trials is the
most valuable way to achieve evidence-based medicine and to
reduce the gap currently existing between research and clin-
ical ndings with possible practical applications in the elds of
personalized nutrition, preventive and precision medicine, and
functional food development.
Dietary Interventions for the Modulation of the
Gut Microbiome
Several studies highlighted the effect of a dietary inter-
vention on the gut microbiome, focusing on the addition of a
specic supplement to the diet (mainly different types of diet-
ary ber) or administering diets enriched in carbohydrates, pro-
teins, or fats (Table1). Adding prebiotic ber to the diet may
lead to the enrichment of ber-degrading bacteria in the gut
and improved metabolic health, but results reported in the cur-
rent literature have been conicting. Studies are often limited to
a small number of subjects (20 per treatment group or fewer in
many cases) (Table1).
Moreover, comparison of results across studies is not
always possible, as many confounding factors exist, making
it necessary to have standardized procedures for sample col-
lection, storage, and subsequent analyses,115 such as those
proposed by the International Human Microbiome Standard
Consortium (http://www.microbiome-standards.org).
Although host genomics may also be implicated, inter-
subject variation in gut microbiota composition may explain
the different metabolic responses observed with the same treat-
ment. Interpersonal differences in response to the same type of
drug have been often observed in therapeutic routines. The gut
microbiome may be involved in this process through bio-trans-
formation of bioactive compounds contained in administered
drugs, reducing or sometimes enhancing their effects.10, 116
Statins, commonly used to reduce plasma low-density lipopro-
tein (LDL) levels, are an example of microbiome-driven person-
alized response to drugs. Indeed, subjects positively responding
to statin treatment showed increased levels of specic secondary
bile acids of microbial origin.10 Therefore, the design of thera-
peutic treatments should consider personalized microbiome
features and their effects on drug metabolism, toxicity, and ef-
cacy. Accordingly, recent studies highlighted that subject-spe-
cic gut microbiome traits cannot be disregarded if diet must
be used to benecially modulate microbiome activities for ther-
apeutic approaches (Fig.2). Indeed, the gut microbiota may
be responsible for unpredictable results in intervention studies.
Features of the gut microbiome before starting the intervention
(eg, overall microbial diversity or abundance of specic taxa,
such as Prevotella copri or Akkermansia muciniphila) were pre-
viously suggested to be discriminant factors between subjects
showing a metabolic improvement (eg, decrease in LDL chol-
esterol, inammatory markers, or insulin resistance) to a diet-
ary intervention and those who were not benecially impacted
(Fig.2). For example, 3-day consumption of barley kernel ber
led to improved glucose metabolism, reducing postprandial gly-
cemic response (PPGR) and insulin, but only in a subset of the
cohort studied.108 Separating responders from nonresponders,
the authors observed higher Prevotella copri concentrations in
responders. Moreover, gavaging mice with live P.copri cells, they
conrmed the positive effect exerted on glucose metabolism,
possibly due to a promotion of hepatic glycogen storage.108 In
contrast, Pedersen and coworkers117 suggested that P.copri may
be responsible for branched-chain amino acid production and
induce insulin resistance, demonstrating that the association
of a whole microbial genus or species with a specic metabolic
outcome may be an oversimplication.118, 119
Indeed, it must be noted that high variability at the strain
level exists in the gut microbiome, and in any other complex
environment, although this point was largely overlooked until
recently. Every species may be represented by several different
strains, with intersubject variability.120 Strains belonging to
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2149
Dietary Interventions to Modulate the Gut Microbiome
TABLE1: Main Clinical Trials Assessing the Gut Microbiome and Metabolic Responses After Dietary Intervention
Study
Intervention Population Outcomes
Description No. Description Microbiota Bioclinical Variables
Bennet etal.,
201798
4 weeks of low-FODMAPs
or traditional IBS
67 IBS patients
BMI 24.0±6.0kg/m2
17% male
No change in the microbiota
after traditional IBS diet both
in responders and nonrespond-
ers; >Bacteroides stercoris,
Ruminococcus gnavus, Dorea,
and several Enterobacteriaceae
at baseline in nonresponders
compared with responders to
low-FODMAPs diet
Decrease in IBS symp-
toms Severity score
≥50 in responders
Candela etal.,
201699
3 weeks of macrobiotic diet
(Ma-Pi; enriched in ber,
hypocaloric) or control diet
with the same energy intake
40 Type 2 diabetes
BMI 34.3±6.5kg/m2
50% male
>Akkermansia, Ruminococcus,
Faecalibacterium, Blautia after
both the dietary intervention;
<Collinsella, Streptococcus, and
>Akkermansia after Ma-Pi com-
pared with control diet
Decrease of postpran-
dial glucose, LDL
cholesterol, insulin
resistance, inamma-
tory markers after
Ma-Pi diet
Chumpitazi
etal., 2015100
2days of low-FODMAPs or
typical American diet
33 IBS pediatric patients
(age 7–17years)
BMI and sex not
provided
>Bacteroides, Ruminococcaceae,
Dorea, Faecalibacterium praus-
nitzii at baseline in responders
Decrease in abdominal
pain and gastroin-
testinal symptoms in
responders
Costabile etal.,
2008101
3 weeks of supplement with
48g of whole grain (WG)
or wheat bran
31 BMI 25.0±5.0kg/m2
48% male
>Bidobacterium, Lactobacillus
after WG
Increase of phenolic
acids in WG, no
changes in blood
lipids, cholesterol, glu-
cose, insulin
Cotillard etal.,
2013102
6 weeks of hypocaloric,
high-protein diet
(1200 and 1500 Kcal/d for
men and women, respec-
tively); 6 weeks of stabi-
lization diet (20% caloric
increase)
49 BMI 33.2±0.5kg/m2
16% male
Divided into low (LGC;
n=20) and high
(HGC; n=29) gene
count
>diversity in LGC after the dietary
intervention
Decrease of insulin
resistance, triglycer-
ides, inammatory
markers in HGC after
intervention
Dao etal.,
2016103
6 weeks of hypocaloric,
high-protein, low-carbohy-
drate diet enriched in ber;
6 weeks of weight mainte-
nance diet
49 BMI 32.5±1.0kg/m2
17% male
Divided into high
(HAk; n=25) and
low (LAk; n=24)
Akkermansia mucin-
iphila abundance
<Akkermansia muciniphila after
the intervention in HAk but still
higher than LAk
Higher decrease of LDL
cholesterol, plasma
glucose, tryglycerides,
insulin resistance in
subjects with higher
baseline levels of
Akkermansia (HAk)
Hald etal.,
2016104
4 weeks of Western-style diet
or high-carbohydrate diet
(enriched in arabinoxylan
and resistant starch)
19 BMI 30.0±2.0kg/m2
Sex not provided
>Bidobacterium and
<Bacteroides, Odoribacter,
Desulfovibrionaceae,
Ruminococcus (Lachnospiraceae)
-
Haro etal.,
2016105
12months of Mediterranean-
style diet (MD; 35 fat—
22% monounsaturated;
6% polyunsaturated and
7% saturated) or low-fat,
high-carbohydrate diet
(LFHCD; 28% fat—12%
monounsaturated; 8%
polyunsaturated and 8%
saturated)
20 BMI 32.2±0.5kg/m2
100% male
>Roseburia, Oscillospira, and
<Prevotella in MD
>Prevotella, Faecalibacterium
prausnitzii, and <Roseburia in
LFHCD
Increase of insulin sensi-
tivity in both MD and
LFHCD
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2150
De Filippis etal
Study
Intervention Population Outcomes
Description No. Description Microbiota Bioclinical Variables
Hjorth etal.,
2017106
6months of ad libitum New
Nordic Diet (NDD; rich in
fruit, vegetables, and whole
grains) or Average Danish
Diet
62 BMI 30.0±1.0kg/m2
33% male
Divided into high
(n=28) and low
(n=34) Prevotella-
to-Bacteroides ratio
groups
- Higher body fat and
waist circumference
loss after NDD in
high Prevotella-to-
Bacteroides group
Korem etal.,
2017107
1 week of supplement with
145g/d of sourdough-leav-
ened whole grain bread
or 110g/d of industrial
white bread, separated by 2
weeks of washout
20 BMI 27.9±4.0kg/m2
45% male
No effect detected Interpersonal varia-
tion in the glycemic
response after bread
consumption is
dependent on baseline
microbiome features
Kovatcheva-
Datchary
etal., 2015108
3days of supplement with
barley kernel bread (BKB)
or white bread
20 BMI 25±3.0kg/m2
10% male
Divided into responders
(n=10) and non-
responders (n=10)
based on metabolic
response to BKB
intervention
>Prevotella in responders com-
pared with nonresponders;
>Prevotella copri and methano-
genic Archaea after BKB inter-
vention only in responders
Decrease of postpran-
dial blood glucose
and insulin after BKB
intervention only in
responders
Louis etal.,
2016109
3months of very low-calorie
diet (800 Kcal/d, enriched
in inulin); 9months of
gradual reintroduction of a
normal diet; 12months of
weight maintenance diet
16 BMI 43.0±7.0kg/m2
44% male
Persistent success group
(PS; >10% weight
loss after 24months,
n=9); no persistent
success (NS; <10%
weight loss after
24months, n=7)
Different microbiota at baseline in
PS and NS groups
>Akkermansia, Alistipes,
Clostridium leptum, and
<Bacteroides in PS at baseline
Only Akkermansia stable after
2years
Decrease of insulin
resistance
Pedersen etal.,
2016110
12 weeks of supplement with
galacto-oligosaccharyde
mixture or placebo
29 Type 2 diabetes
BMI 28.0±6.5kg/m2
100% male
No effect detected No effect detected
Roager etal.,
2017111
8 weeks of intervention with
a whole grain–enriched diet
and 8 weeks of intervention
with a rened grain diet,
separated by a washout
period of 6 weeks
60 High risk of metabolic
syndrome
BMI 25–35kg/m2
Sex not provided
>Faecalibacterium prausnitzii and
Prevotella copri after whole grain
diet
Bacteroides thetaiotaomicron
increased after rened grain diet
Decrease of inamma-
tory markers
Weight loss
Vanegas etal.,
2017112
2 weeks of run-in consum-
ing a Western-style diet,
followed by 6 weeks of
weight-maintaining diet
supplemented with 8g/1000
Kcal of rened grains (RG)
or 16g/1000 Kcal WG
81 BMI 26±0.47kg/m2
63% and 59% male
in RG and WG,
respectively
>Lachnospira, Roseburia, and
<Enterobacteriaceae after WG
intervention compared with RG
No effect on inam-
matory and immune
markers
Vitaglione etal.,
2015113
8 weeks of WG (70g/d) or
rened grain (RF; 60g/d)
68 BMI 30±0.9kg/m2
31% and 37% male in
WG and RF groups,
respectively
>Prevotella and
<Dialister, Bidobacterium,
Blautia, Collinsella in WG
Decrease of inamma-
tory markers, increase
of phenolic acids
Walker etal.,
2011114
1 week of weight mainte-
nance, 3 weeks of non-
starch polysaccharides,
followed by 3 weeks of
resistant starch (RS) and
3 weeks of high-protein,
hypocaloric diet
14 BMI 39.4±1.5kg/m2
100% male
>Eubacterium rectale,
Ruminococcus bromii in RS
-
TABLE1: Continued
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2151
Dietary Interventions to Modulate the Gut Microbiome
the same species may harbor a signicantly different genomic
repertoire and may respond in different ways to dietary com-
ponents,118, 119 making it even more difcult to demonstrate a
causative role of diet in the modulation of the gut microbiome.
Different subjects may have distinctive metabolic
responses to the same food. Subjects with a higher Prevotella-to-
Bacteroides ratio (P/B) had higher weight loss after consuming
a 6-month high-ber diet compared with the low P/B group,106
whereas Dao and coworkers103 observed that obese subjects
with higher levels of Akkermansia muciniphila showed better
metabolic outcomes (lower insulin resistance, LDL choles-
terol) compared with those with a lower baseline concentration
of this microbe, when treated with a hypocaloric, high-pro-
tein and -ber diet (Table1). In addition, it was demonstrated
that a meal cannot be considered “good” for everybody.121 The
authors integrated gut microbiota features with anthropomet-
ric and metabolic measures, dietary habits, physical activity,
and lifestyle in a machine-learning algorithm that accurately
predicted personalized PPGR to real-life meals. This predictive
strategy was then used to personalize dietary intervention and
modify postprandial glucose response. They demonstrated that
baseline microbiota composition may be implicated in the sub-
ject-specic response to a dietary intervention and that consid-
ering these differences may help in designing tailored meals for
improving metabolic health.121 Accordingly, the same authors
suggested that the glycemic index of a food alone cannot always
be useful in predicting the glycemic response. They found the
PPGR to white or whole grain bread to be person-specic and
used microbiome composition to predict which type of bread
resulted in the best glycemic response.107 Indeed, subject-spe-
cic signatures in the gut microbiome may be responsible for
weight regain after a dietary intervention, and the extent of
such regain may be predicted by integrating gut microbiome
features in a machine-learning algorithm.122
Although many studies focused on the administra-
tion of specic food supplements, the habitual consump-
tion of a healthy and diverse diet, such as that based on the
Mediterranean model, is recognized to shape the gut micro-
biome and to promote the production of benecial metabo-
lites.56 Although with a limited number of subjects, Haro and
coworkers105 demonstrated that dietary treatment for 1year
with a Mediterranean-style diet improved insulin sensitivity
and increased the abundance of SCFA-producing microbes
(Table1). These examples suggest that following a healthy and
diverse dietary pattern and lifestyle can contribute to main-
taining a “healthy” microbiome without necessarily adopting
microbiome-targeted nutritional interventions.
Most of the studies available on targeted dietary modu-
lation of the gut microbiome have focused on metabolic disor-
ders (Table1). However, the same approach may be useful for
FIGURE2. Subject-specic gut microbiota features may aect the response to a dietary intervention. The metabolic response to a dietary interven-
tion is person-specic, and the same type of food or dietary pattern may produce dierent eects. Dividing responders and nonresponders, recent
studies showed dierences in gut microbiota composition and that this may be the cause of the personalized response to a dietary intervention,
thus highlighting the necessity of personally tailored nutrition based on gut microbiota composition for therapeutic and preventive purposes.
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2152
De Filippis etal
other pathologies, such as IBD and IBS. Indeed, Bennet and
coworkers98 found different baseline microbiota composition in
adult IBS patients responding or not to a low-FODMAP diet
(ie, showing a decrease of the IBS Symptoms Severity Score).
Nonresponders had higher baseline abundance of several taxa,
eg, Bacteroides stercoris, Pseudomonas, Acinetobacter, and
Ruminococcus gnavus, compared with responders. Baseline
microbiota features were implemented in a Random Forest
model, which was used to predict the probability of a posi-
tive response to the dietary treatment. This pioneer study rst
highlighted the possibility to choose the most appropriate diet-
ary treatment for IBS patients, based on their gut microbiota
composition.
Based on these observations, it is plausible that in the near
future, subjects will be stratied based on their gut microbiome
features and study enrollment will be performed considering
their baseline microbiome. As we know some associations of
specic microbial genera/species with diet and/or diseases, we
can speculate on the possibility of stratifying subjects accord-
ing to the abundance of microbial taxa recognized for certain
functional activities (eg, ber-degrading) or for the production
of benecial/detrimental metabolites. Although considering
the microbiota as a stratication factor for subject enrollment is
promising, several issues arise, including increased enrollment
costs and the intrasubject variability of the gut microbiota even
in a short time frame. The possibility of stratifying the study
population a posteriori for the similarity of the microbiome
or microbial metabolite prole during data analysis is also an
alternative. Nevertheless, it must be pointed out that these sec-
ondary analyses do not preserve the benets of randomization.
In addition, such stratications are less likely to be reproduci-
ble due to the risk of chance ndings related to multiple test-
ing. Despite the abovementioned limitations, modulating and
manipulating the gut microbiome with a personally designed
dietary intervention to induce changes in its composition and
functions is surely a promising application for both therapeutic
and preventive clinical strategies.
CONCLUSIONS
Microbiome-targeted dietary interventions constitute a
powerful and tantalizing tool for the prevention and treatment
of different diseases. We are still quite far from microbiome-tar-
geted precision medicine, but we are surely on the right scien-
tic path to developing an exhaustive set of tools and clinical
knowledge to ll the current gaps. Personalized nutrition based
on microbiome features is currently being attempted, although
its real impact and benets suffer from the aforementioned lim-
itations. Most of the studies available are observational, and
controlled clinical trials targeting the microbiome are still too
scarce to draw denitive conclusions or to propose a stand-
ardized protocol for modulating the gut microbiome through
the diet for therapeutic purposes. Future intervention studies
will surely provide new knowledge and will help in overcoming
the current issues associated with such types of interventions.
Indeed, interindividual variations in gut microbiome compos-
ition and functions, along with the need to study the effect of
dietary changes on microbiome functions at the strain level,
make the use of such interventions still only exploratory in clin-
ical practice.
REFERENCES
1. Bäckhed F, Ley RE, Sonnenburg JL, et al. Host-bacterial mutualism in the
human intestine. Science. 2005;307:1915–20.
2. Zhao L. Genomics: the tale of our other genome. Nature. 2010;465:879–80.
3. Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota
on human health: an integrative view. Cell. 2012;148:1258–70.
4. Lozupone CA, Stombaugh JI, Gordon JI, etal. Diversity, stability and resilience
of the human gut microbiota. Nature. 2012;489:220–30.
5. Arumugam M, Raes J, Pelletier E, etal; MetaHIT Consortium. Enterotypes of
the human gut microbiome. Nature. 2011;473:174–80.
6. Jeffery IB, Claesson MJ, O’Toole PW, Shanahan F. Categorization of the gut
microbiota: enterotypes or gradients? Nat Rev Microbiol. 2012;10:591–2.
7. Knights D, Ward TL, McKinlay CE, etal. Rethinking “enterotypes.” Cell Host
Microbe. 2014;16:433–7.
8. Honda K, Littman DR. The microbiota in adaptive immune homeostasis and
disease. Nature. 2016;535:75–84.
9. Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity.
Nature. 2016;535:65–74.
10. Derrien M, Veiga P. Rethinking diet to aid human-microbe symbiosis. Trends
Microbiol. 2017;25:100–12.
11. Shanahan F, van Sinderen D, O’Toole PW, Stanton C. Feeding the microbiota:
transducer of nutrient signals for the host. Gut. 2017;66:1709–17.
12. Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colo-
rectal cancer. Nat Rev Microbiol. 2014;12:661–72.
13. O’Keefe SJ. Diet, microorganisms and their metabolites, and colon cancer. Nat
Rev Gastroenterol Hepatol. 2016;13:691–706.
14. Ou J, Carbonero F, Zoetendal EG, etal. Diet, microbiota, and microbial metab-
olites in colon cancer risk in Rural Africans and African Americans. Am J Clin
Nutr. 2013;98:111–20.
15. Kamada N, Chen GY, Inohara N, Núñez G. Control of pathogens and pathobi-
onts by the gut microbiota. Nat Immunol. 2013;14:685–90.
16. Parekh PJ, Balart LA, Johnson DA. The inuence of the gut microbiome on obe-
sity, metabolic syndrome and gastrointestinal disease. Clin Transl Gastroenterol.
2015;6:e91.
17. Cho I, Blaser MJ. The human microbiome: at the interface of health and disease.
Nat Rev Genet. 2012;13:260–70.
18. Shreiner AB, Kao JY, Young VB. The gut microbiome in health and in disease.
Curr Opin Gastroenterol. 2015;31:69–75.
19. Le Chatelier E, Nielsen T, Qin J, etal; MetaHIT Consortium. Richness of human
gut microbiome correlates with metabolic markers. Nature. 2013;500:541–6.
20. Boulangé CL, Neves AL, Chilloux J, etal. Impact of the gut microbiota on
inammation, obesity, and metabolic disease. Genome Med. 2016;8:42.
21. Cani PD, Amar J, Iglesias MA, etal. Metabolic endotoxemia initiates obesity and
insulin resistance. Diabetes. 2007;56:1761–72.
22. Qin J, Li Y, Cai Z, etal. A metagenome-wide association study of gut microbiota
in type 2 diabetes. Nature. 2012;490:55–60.
23. Wang Z, Klipfell E, Bennett BJ, etal. Gut ora metabolism of phosphatidylcho-
line promotes cardiovascular disease. Nature. 2011;472:57–63.
24. Tang WH, Wang Z, Levison BS, etal. Intestinal microbial metabolism of phos-
phatidylcholine and cardiovascular risk. N Engl J Med. 2013;368:1575–84.
25. Abrahamsson TR, Wu RY, Jenmalm MC. Gut microbiota and allergy: the impor-
tance of the pregnancy period. Pediatr Res. 2015;77:214–9.
26. Kim KS, Hong SW, Han D, etal. Dietary antigens limit mucosal immunity by
inducing regulatory T cells in the small intestine. Science. 2016;351:858–63.
27. Gagnière J, Raisch J, Veziant J, etal. Gut microbiota imbalance and colorectal
cancer. World J Gastroenterol. 2016;22:501–18.
28. Dinan TG, Cryan JF. Brain–gut–microbiota axis—mood, metabolism and behav-
ior. Nat Rev Gastroenterol Hepatol. 2017;14:69–70.
29. Guida F, Turco F, Iannotta M, etal. Antibiotic-induced microbiota perturbation
causes gut endocannabinoidome changes, hippocampal neuroglial reorganization
and depression in mice. Brain Behav Immun. 2018;67:230–45.
30. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interac-
tions between enteric microbiota, central and enteric nervous systems. Ann
Gastroenterol. 2015;28:203–9.
31. De Angelis M, Francavilla R, Piccolo M, et al. Autism spectrum disorders and
intestinal microbiota. Gut Microbes. 2015;6:207–13.
32. Jiang C, Li G, Huang P, etal. The gut microbiota and Alzheimer’s disease. J
Alzheimers Dis. 2017;58:1–15.
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2153
Dietary Interventions to Modulate the Gut Microbiome
33. Klingelhoefer L, Reichmann H. Pathogenesis of parkinson disease–the gut-brain
axis and environmental factors. Nat Rev Neurol. 2015;11:625–36.
34. Molodecky NA, Soon IS, Rabi DM, etal. Increasing incidence and prevalence
of the inammatory bowel diseases with time, based on systematic review.
Gastroenterology. 2012;142:46–54.e42; quiz e30.
35. Ng SC, Shi HY, Hamidi N, etal. Worldwide incidence and prevalence of inam-
matory bowel disease in the 21st century: a systematic review of population-based
studies. Lancet. 2018;390:2769–78.
36. Hou JK, Abraham B, El-Serag H. Dietary intake and risk of developing inam-
matory bowel disease: a systematic review of the literature. Am J Gastroenterol.
2011;106:563–73.
37. O’Moráin C, Segal AW, Levi AJ. Elemental diet as primary treatment of acute
Crohn’s disease: a controlled trial. Br Med J (Clin Res Ed). 1984;288:1859–62.
38. Chapman-Kiddell CA, Davies PS, Gillen L, Radford-Smith GL. Role of
diet in the development of inammatory bowel disease. Inamm Bowel Dis.
2010;16:137–51.
39. Ni J, Wu GD, Albenberg L, Tomov VT. Gut microbiota and IBD: causation or
correlation? Nat Rev Gastroenterol Hepatol. 2017;14:573–84.
40. Wright EK, Kamm MA, Teo SM, etal. Recent advances in characterizing the
gastrointestinal microbiome in Crohn’s disease: a systematic review. Inamm
Bowel Dis. 2015;21:1219–28.
41. Machiels K, Joossens M, Sabino J, etal. A decrease of the butyrate-producing
species Roseburia hominis and Faecalibacterium prausnitzii denes dysbiosis in
patients with ulcerative colitis. Gut. 2014;63:1275–83.
42. Man SM, Kaakoush NO, Mitchell HM. The role of bacteria and pattern-recogni-
tion receptors in Crohn’s disease. Nat Rev Gastroenterol Hepatol. 2011;8:152–68.
43. Sokol H, Leducq V, Aschard H, etal. Fungal microbiota dysbiosis in IBD. Gut.
2017;66:1039–48.
44. Lewis JD, Chen EZ, Baldassano RN, etal. Inammation, antibiotics, and diet as
environmental stressors of the gut microbiome in pediatric Crohn’s disease. Cell
Host Microbe. 2015;18:489–500.
45. Lewis JD, Abreu MT. Diet as a trigger or therapy for inammatory bowel dis-
eases. Gastroenterology. 2017;152:398–414.e6.
46. Mearin F, Lacy BE, Chang L, et al. Bowel disorders. Gastroenterology.
2016;150:1393–407.
47. Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel
syndrome: a meta-analysis. Clin Gastroenterol Hepatol. 2012;10:712–21.e4.
48. Staudacher HM, Whelan K. The low FODMAP diet: recent advances in under-
standing its mechanisms and efcacy in IBS. Gut. 2017;66:1517–27.
49. Monsbakken KW, Vandvik PO, Farup PG. Perceived food intolerance in subjects
with irritable bowel syndrome– etiology, prevalence and consequences. Eur J Clin
Nutr. 2006;60:667–72.
50. Jeffery IB, Quigley EM, Öhman L, etal. The microbiota link to irritable bowel
syndrome: an emerging story. Gut Microbes. 2012;3:572–6.
51. Jeffery IB, O’Toole PW, Öhman L, etal. An irritable bowel syndrome subtype
dened by species-specic alterations in faecal microbiota. Gut. 2012;61:997–1006.
52. Gomez A, Petrzelkova KJ, Burns MB, etal. Gut microbiome of coexisting baaka
pygmies and bantu reects gradients of traditional subsistence patterns. Cell Rep.
2016;14:2142–53.
53. Martínez I, Stegen JC, Maldonado-Gómez MX, et al. The gut microbiota of
rural Papua New Guineans: composition, diversity patterns, and ecological pro-
cesses. Cell Rep. 2015;11:527–38.
54. Obregon-Tito AJ, Tito RY, Metcalf J, etal. Subsistence strategies in traditional
societies distinguish gut microbiomes. Nat Commun. 2015;6:6505.
55. Segata N. Gut microbiome: westernization and the disappearance of intestinal
diversity. Curr Biol. 2015;25:R611–R613.
56. De Filippis F, Pellegrini N, Vannini L, et al. High-level adherence to a
Mediterranean diet benecially impacts the gut microbiota and associated metab-
olome. Gut. 2016;65:1812–21.
57. David LA, Maurice CF, Carmody RN, etal. Diet rapidly and reproducibly alters
the human gut microbiome. Nature. 2014;505:559–63.
58. Wędrychowicz A, Zając A, Tomasik P. Advances in nutritional therapy in inam-
matory bowel diseases: review. World J Gastroenterol. 2016;22:1045–66.
59. D’Souza S, Levy E, Mack D, etal. Dietary patterns and risk for Crohn’s disease
in children. Inamm Bowel Dis. 2008;14:367–73.
60. Barclay AR, Russell RK, Wilson ML, etal. Systematic review: the role of breast-
feeding in the development of pediatric inammatory bowel disease. J Pediatr.
2009;155:421–6.
61. Dalrymple J, Bullock I. Diagnosis and management of irritable bowel syndrome
in adults in primary care: summary of NICE guidance. BMJ. 2008;336:556–8.
62. McKenzie YA, Alder A, Anderson W, etal. British Dietetic Association system-
atic review and evidence-based practice guidelines for the dietary management of
irritable bowel syndrome in adults. J Hum Nutr Diet. 2016;29:549–75.
63. Gibson PR. Use of the low-FODMAP diet in inammatory bowel disease. J
Gastroenterol Hepatol. 2017;32(Suppl 1):40–2.
64. Haas SV, Haas MP. The treatment of celiac disease with the specic carbohydrate
diet; report on 191 additional cases. Am J Gastroenterol. 1955;23:344–60.
65. Cohen SA, Gold BD, Oliva S, etal. Clinical and mucosal improvement with spe-
cic carbohydrate diet in pediatric Crohn disease. J Pediatr Gastroenterol Nutr.
2014;59:516–21.
66. Suskind DL, Wahbeh G, Gregory N, etal. Nutritional therapy in pediatric Crohn
disease: the specic carbohydrate diet. J Pediatr Gastroenterol Nutr. 2014;58:87–91.
67. Lee D, Albenberg L, Compher C, etal. Diet in the pathogenesis and treatment of
inammatory bowel diseases. Gastroenterology. 2015;148:1087–06.
68. Shiga H, Kajiura T, Shinozaki J, etal. Changes of faecal microbiota in patients
with Crohn’s disease treated with an elemental diet and total parenteral nutrition.
Dig Liver Dis. 2012;44:736–42.
69. Gerasimidis K, Bertz M, Hanske L, et al. Decline in presumptively protective
gut bacterial species and metabolites are paradoxically associated with disease
improvement in pediatric Crohn’s disease during enteral nutrition. Inamm Bowel
Dis. 2014;20:861–71.
70. Tjellström B, Högberg L, Stenhammar L, et al. Effect of exclusive enteral
nutrition on gut microora function in children with Crohn’s disease. Scand J
Gastroenterol. 2012;47:1454–9.
71. Afzal NA, Davies S, Paintin M, etal. Colonic crohn’s disease in children does not
respond well to treatment with enteral nutrition if the ileum is not involved. Dig
Dis Sci. 2005;50:1471–5.
72. Gerasimidis K, Russell R, Hansen R, etal. Role of Faecalibacterium prausnit-
zii in Crohn’s disease: friend, foe, or does not really matter? Inamm Bowel Dis.
2014;20:E18–E19.
73. Sokol H, Langella P. Benecial effects of exclusive enteral nutrition in Crohn’s
disease are not mediated by Faecalibacterium prausnitzii. Inamm Bowel Dis.
2014;20:E18.
74. Staudacher HM, Irving PM, Lomer MC, Whelan K. Mechanisms and ef-
cacy of dietary FODMAP restriction in IBS. Nat Rev Gastroenterol Hepatol.
2014;11:256–66.
75. Staudacher HM, Lomer MC, Anderson JL, et al. Fermentable carbohydrate
restriction reduces luminal bidobacteria and gastrointestinal symptoms in
patients with irritable bowel syndrome. J Nutr. 2012;142:1510–8.
76. Madsen JL, Linnet J, Rumessen JJ. Effect of nonabsorbed amounts of a fruc-
tose-sorbitol mixture on small intestinal transit in healthy volunteers. Dig Dis Sci.
2006;51:147–53.
77. Duncan SH, Holtrop G, Lobley GE, etal. Contribution of acetate to butyrate
formation by human faecal bacteria. Br J Nutr. 2004;91:915–23.
78. Bourdu S, Dapoigny M, Chapuy E, etal. Rectal instillation of butyrate provides
a novel clinically relevant model of noninammatory colonic hypersensitivity in
rats. Gastroenterology. 2005;128:1996–2008.
79. Tana C, Umesaki Y, Imaoka A, et al. Altered proles of intestinal microbiota
and organic acids may be the origin of symptoms in irritable bowel syndrome.
Neurogastroenterol Motil. 2010;22:512–519, e114.
80. Castiglione F, Rispo A, Di Girolamo E, etal. Antibiotic treatment of small bowel
bacterial overgrowth in patients with Crohn’s disease. Aliment Pharmacol Ther.
2003;18:1107–12.
81. O’Leary C, Quigley EM. Small bowel bacterial overgrowth, celiac disease, and
IBS: what are the real associations? Am J Gastroenterol. 2003;98:720–2.
82. Bovee-Oudenhoven IM, ten Bruggencate SJ, Lettink-Wissink ML, van der Meer
R. Dietary fructo-oligosaccharides and lactulose inhibit intestinal colonisation
but stimulate translocation of salmonella in rats. Gut. 2003;52:1572–8.
83. McIntosh K, Reed DE, Schneider T, et al. Fodmaps alter symptoms and
the metabolome of patients with IBS: a randomised controlled trial. Gut.
2017;66:1241–51.
84. Chumpitazi BP, Hollister EB, Oezguen N, etal. Gut microbiota inuences low
fermentable substrate diet efcacy in children with irritable bowel syndrome. Gut
Microbes. 2014;5:165–75.
85. Scott KP, Antoine JM, Midtvedt T, van Hemert S. Manipulating the gut microbi-
ota to maintain health and treat disease. Microb Ecol Health Dis. 2015;26:25877.
86. Ioannidis JP. We need more randomized trials in nutrition-preferably large, long-
term, and with negative results. Am J Clin Nutr. 2016;103:1385–6.
87. Maki KC, Slavin JL, Rains TM, Kris-Etherton PM. Limitations of observational
evidence: implications for evidence-based dietary recommendations. Adv Nutr.
2014;5:7–15.
88. Garattini S, Jakobsen JC, Wetterslev J, etal. Evidence-based clinical practice:
overview of threats to the validity of evidence and how to minimise them. Eur J
Intern Med. 2016;32:13–21.
89. Djurisic S, Rath A, Gaber S, etal. Barriers to the conduct of randomised clinical
trials within all disease areas. Trials. 2017;18:360.
90. Neugebauer EAM, Rath A, Antoine SL, etal. Specic barriers to the conduct of
randomised clinical trials on medical devices. Trials. 2017;18:427.
91. Laville M, Segrestin B, Alligier M, etal. Evidence-based practice within nutrition:
what are the barriers for improving the evidence and how can they be dealt with?
Trials. 2017;18:425.
92. Kaddurah-Daouk R, Weinshilboum RM; Pharmacometabolomics Research
Network. Pharmacometabolomics: implications for clinical pharmacology and
systems pharmacology. Clin Pharmacol Ther. 2014;95:154–67.
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
Inflamm Bowel Dis
Volume 24, Number 10, October 2018
2154
De Filippis etal
93. Kelly BJ, Gross R, Bittinger K, etal. Power and sample-size estimation for micro-
biome studies using pairwise distances and PERMANOVA. Bioinformatics.
2015;31:2461–8.
94. Mattiello F, Verbist B, Faust K, et al. A web application for sample size
and power calculation in case-control microbiome studies. Bioinformatics.
2016;32:2038–40.
95. Hotamisligil GS. Inammation, metaammation and immunometabolic disor-
ders. Nature. 2017;542:177–85.
96. Schroeder BO, Bäckhed F. Signals from the gut microbiota to distant organs in
physiology and disease. Nat Med. 2016;22:1079–89.
97. Riedl A, Gieger C, Hauner H, etal. Metabotyping and its application in targeted
nutrition: an overview. Br J Nutr. 2017;117:1631–44.
98. Bennet SMP, Böhn L, Störsrud S, etal. Multivariate modelling of faecal bacterial
proles of patients with IBS predicts responsiveness to a diet low in FODMAPs.
Gut. 2017; doi:10.1136/gutjnl-2016-313128.
99. Candela M, Biagi E, Soverini M, etal. Modulation of gut microbiota dysbioses
in type 2 diabetic patients by macrobiotic ma-pi 2 diet. Br J Nutr. 2016;116:80–93.
100. Chumpitazi BP, Cope JL, Hollister EB, et al. Randomised clinical trial: gut
microbiome biomarkers are associated with clinical response to a low FODMAP
diet in children with the irritable bowel syndrome. Aliment Pharmacol Ther.
2015;42:418–27.
101. Costabile A, Klinder A, Fava F, et al. Whole-grain wheat breakfast cereal has
a prebiotic effect on the human gut microbiota: a double-blind, placebo-con-
trolled, crossover study. Br J Nutr. 2008;99:110–20.
102. Cotillard A, Kennedy SP, Kong LC, etal; ANR MicroObes consortium. Dietary
intervention impact on gut microbial gene richness. Nature. 2013;500:585–8.
103. Dao MC, Everard A, Aron-Wisnewsky J, et al; MICRO-Obes Consortium.
Akkermansia muciniphila and improved metabolic health during a dietary inter-
vention in obesity: relationship with gut microbiome richness and ecology. Gut.
2016;65:426–36.
104. Hald S, Schioldan AG, Moore ME, etal. Effects of arabinoxylan and resistant
starch on intestinal microbiota and short-chain fatty acids in subjects with meta-
bolic syndrome: a randomised crossover study. PLoS One. 2016;11:e0159223.
105. Haro C, Montes-Borrego M, Rangel-Zúñiga OA, etal. Two healthy diets mod-
ulate gut microbial community improving insulin sensitivity in a human obese
population. J Clin Endocrinol Metab. 2016;101:233–42.
106. Hjorth MF, Roager HM, Larsen TM, etal. Pre-treatment microbial Prevotella-
to-Bacteroides ratio, determines body fat loss success during a 6-months rand-
omized controlled diet intervention. Int J Obes. 2017; doi:10.1038/ijo.2017.220.
107. Korem T, Zeevi D, Zmora N, et al. Bread affects clinical parameters and
induces gut microbiome-associated personal glycemic responses. Cell Metab.
2017;25:1243–53.e5.
108. Kovatcheva-Datchary P, Nilsson A, Akrami R, et al. Dietary ber-induced
improvement in glucose metabolism is associated with increased abundance of
Prevotella. Cell Metab. 2015;22:971–82.
109. Louis S, Tappu RM, Damms-Machado A, etal. Characterization of the gut
microbial community of obese patients following a weight-loss intervention
using whole metagenome shotgun sequencing. PLoS One. 2016;11:e0149564.
110. Pedersen C, Gallagher E, Horton F, et al. Host-microbiome interactions in
human type 2 diabetes following prebiotic bre (galacto-oligosaccharide) intake.
Br J Nutr. 2016;116:1869–77.
111. Roager HM, Vogt JK, Kristensen M, etal. Whole grain-rich diet reduces body
weight and systemic low-grade inammation without inducing major changes
of the gut microbiome: a randomised cross-over trial. Gut. 2017; doi:10.1136/
gutjnl-2017-314786.
112. Vanegas SM, Meydani M, Barnett JB, et al. Substituting whole grains for
rened grains in a 6-wk randomized trial has a modest effect on gut microbi-
ota and immune and inammatory markers of healthy adults. Am J Clin Nutr.
2017;105:635–50.
113. Vitaglione P, Mennella I, Ferracane R, etal. Whole-grain wheat consumption
reduces inammation in a randomized controlled trial on overweight and obese
subjects with unhealthy dietary and lifestyle behaviors: role of polyphenols
bound to cereal dietary ber. Am J Clin Nutr. 2015;101:251–61.
114. Walker AW, Ince J, Duncan SH, etal. Dominant and diet-responsive groups of
bacteria within the human colonic microbiota. Isme J. 2011;5:220–30.
115. Costea PI, Zeller G, Sunagawa S, etal. Towards standards for human fecal sam-
ple processing in metagenomic studies. Nat Biotechnol. 2017;35:1069–76.
116. Alexander JL, Wilson ID, Teare J, etal. Gut microbiota modulation of chemo-
therapy efcacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14:356–65.
117. Pedersen HK, Gudmundsdottir V, Nielsen HB, etal; MetaHIT Consortium.
Human gut microbes impact host serum metabolome and insulin sensitivity.
Nature. 2016;535:376–81.
118. De Filippis F, Pellegrini N, Laghi L, etal. Unusual sub-genus associations of
faecal Prevotella and Bacteroides with specic dietary patterns. Microbiome.
2016;4:57.
119. Ley RE. Gut microbiota in 2015: Prevotella in the gut: choose carefully. Nat Rev
Gastroenterol Hepatol. 2016;13:69–70.
120. Truong DT, Tett A, Pasolli E, etal. Microbial strain-level population structure
and genetic diversity from metagenomes. Genome Res. 2017;27:626–38.
121. Zeevi D, Korem T, Zmora N, etal. Personalized nutrition by prediction of glyce-
mic responses. Cell. 2015;163:1079–94.
122. Thaiss CA, Itav S, Rothschild D, et al. Persistent microbiome alterations
modulate the rate of post-dieting weight regain. Nature. 2016; doi:10.1038/
nature20796.
Downloaded from https://academic.oup.com/ibdjournal/article-abstract/24/10/2142/4970097 by Biblioteca Centrale Agraria user on 17 January 2019
... (David et al., 2014). O consumo regular de pão fermentado naturalmente tem sido associado a uma melhoria significativa na composição e diversidade da microbiota intestinal (Marco et al., 2017;De Filippis et al., 2018). As bactérias ácido láticas presentes na massa do pão podem atuar como probióticos, promovendo não apenas o crescimento, mas também o equilíbrio de bactérias benéficas no intestino (Katina et al., 2006). ...
... Além disso, a fermentação natural do pão pode resultar na produção de metabólitos bioativos, como ácidos orgânicos e peptídeos antimicrobianos, que contribuem para a saúde intestinal e geral (Marco et al., 2017;Katina et al., 2006). Esses metabólitos exercem efeitos antioxidantes e anti-inflamatórios, ajudando a proteger contra o estresse oxidativo e a inflamação crônica associados a diversas doenças (De Filippis et al., 2018). Essa associação benéfica pode ser atribuída, em parte, aos compostos bioativos produzidos durante a fermentação. ...
... Por exemplo, as proteínas presentes na farinha são quebradas em aminoácidos durante o processo de fermentação, o que não só facilita sua digestão, mas também aumenta sua absorção pelo intestino delgado (De Filippis et al., 2018). Isso é especialmente importante para pessoas com intolerância ao glúten ou sensibilidade ao trigo, pois a fermentação pode reduzir a presença de proteínas desencadeadoras de reações adversas, como a gliadina (Dieterich e Zopf, 2019). ...
Article
A fermentação natural do pão é um processo ancestral e complexo, no qual microrganismos como leveduras e bactérias desempenham papéis cruciais na transformação de ingredientes simples em um produto final singular e excepcional. Esta mini revisão explora os processos microbiológicos envolvidos nessa fermentação, destacando a contribuição das leveduras, especialmente as pertencentes a espécie Saccharomyces cereviseae, na produção de dióxido de carbono e etanol, fundamentais para a textura e aroma do pão. Além disso, discute-se o papel das bactérias ácido láticas, como Lactobacillus e Leuconostoc, na conferência de sabor, acidez e estabilidade microbiológica ao pão. As inter-relações complexas entre leveduras e bactérias também são abordadas, assim como seus impactos na estrutura, textura, aroma e sabor do pão. Por fim, são explorados os benefícios para a saúde associados à fermentação natural do pão, evidenciando como esse alimento tradicional não apenas agrada ao paladar, mas também promove a saúde intestinal e biodisponibilidade de nutrientes.
... Alternative strategies for modulating the microbiota include probiotics, prebiotics, synbiotics, postbiotics, microbiome-targeted diets, short-chain fatty acid (SCFA) supplementation, bacteriophage therapy and polyphenol supplementation [84][85][86][87][88][89][90][91][92]. In their systematic review of randomized clinical trials, Tenorio-Jiménez et al. found modest improvements in components of metabolic syndrome with the use of various probiotics, such as Lactobacillus plantarum and Bifidobacterium lactis [84,93,94]. ...
... However, the effects of different dietary plans vary between individuals due to their unique intestinal microbiota profiles, which influence how they respond to the same types of food. For instance, individuals with a higher Prevotella-to-Klebsiella ratio may experience greater weight loss on a high-fiber diet compared to those with a lower ratio of these bacteria [88]. In relation to short-chain fatty acids, acetate serves as an example. ...
Article
Full-text available
Over time, extensive research has underscored the pivotal role of gut microbiota in the onset and progression of various diseases, with a particular focus on fecal microbiota transplantation (FMT) as a potential therapeutic approach. The practice of transferring fecal matter from a healthy donor to a patient provides valuable insights into how alterations in gut microbiota can impact disease development and how rectifying dysbiosis may offer therapeutic benefits. Re-establishing a balanced symbiotic relationship in the gastrointestinal tract has shown positive results in managing both intestinal and systemic conditions. Currently, one of the most pressing global health issues is metabolic syndrome—a cluster of conditions that includes insulin resistance, lipid imbalances, central obesity and hypertension. In this context, FMT has emerged as a promising strategy for addressing key components of metabolic syndrome, such as improving insulin sensitivity, body weight and lipid profiles. However, further well-structured studies are needed to refine treatment protocols and establish the long-term safety and efficacy of this intervention.
... Sin embargo aun no está claro la relación de la disbiosis intestinal con el cáncer colorrectal a pesar de que se ha aislado ciertos microorganismos que prefieren los sitios del tumor, por lo que seguramente en el futuro tendremos mas evidencia que demuestre la relación de la disbiosis con el cáncer colorrectal la cual es muy probable que exista basándose en hipótesis actuales. (De Filippis et al., 2018) ...
... Estas pueden presentarse en forma de probióticos o trasplante de microbiota fecal, hay un creciente interés en terapias emergentes y futuras direcciones para el tratamiento de la disbiosis intestinal. Esto incluye el desarrollo de terapias microbianas personalizadas, el uso de técnicas avanzadas de secuenciación para modular la microbiota con mayor precisión (De Filippis et al., 2018) Las poblaciones rurales del mundo, en quienes normalmente su dieta es rica en verduras legumbres tubérculos, muestran un mayor enriquecimiento de su flora y muestran mayor capacidad de digerir carbohidratos complejos y fibra, la occidentalización ha inducido una perdida de la diversidad y una disminución de la capacidad para degradar carbohidratos complejos y la capacidad de producir ácidos grasos de cadena corta. En pacientes con enfermedades como la enfermedad de Crohn se ha visto que una dieta rica en aceite de oliva, pescado, verduras, frutas, cereales y frutos secos mejora la inflamación intestinal. ...
... Although the standardized treatment protocols can improve the operability and scalability of microbiota modulation to a certain extent, personalized treatment remains key to enhancing therapeutic outcomes. There may be significant differences in the composition of the gut microbiota and the status of immune function among different patients, thus necessitating the formulation of individualized treatment plans based on individual differences (89). Formulating a personalized treatment plan requires the consideration of a multitude of factors, including the patient's gut microbiota composition, immune function status, disease type and severity (90). ...
Article
Full-text available
The intervention of microbiota modulation in the treatment of infection complications after allogeneic hematopoietic stem cell transplantation in pediatric patients with hematological malignancies has shown potential benefits. Through the use of probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT), these interventions modulate the gut microbiota and enhance immune function to prevent and treat infections. They have been shown to reduce the incidence of diarrhea and intestinal infections, mitigate the issue of antibiotic resistance, and promote the recovery of gut microbiota. Future research is needed to further assess the safety and efficacy of these interventions and to establish standardized treatment protocols.
... • The personalization of synbiotic interventions could be especially beneficial for patients who have not responded adequately to standard treatments or who experience adverse effects from pharmacological therapies. Collaboration among healthcare professionals is essential to effectively implement personalized strategies [74,79,80]. ...
Article
Full-text available
Background/Objectives: Polycystic ovary syndrome is a common endocrine disorder in women of reproductive age characterized by insulin resistance and hormonal imbalances. Recent research suggests that probiotics and synbiotics may improve these parameters by modulating the gut microbiota. This study systematically reviewed randomized clinical trials evaluating the impact of probiotic, prebiotic, and synbiotic supplementation on insulin resistance and hormonal parameters in women with PCOS. Methods: Exhaustive searches were conducted in PubMed, Cochrane CENTRAL, Scopus, Web of Science, and Embase, following PRISMA guidelines. Randomized trials assessing supplementation with probiotics, prebiotics, or synbiotics for at least 8 weeks in women diagnosed with PCOS according to the Rotterdam criteria were included. Data on participants, interventions, and outcomes related to insulin resistance and hormones were extracted. Results: Eleven studies from Iran involving overweight or obese women aged 15 to 48 were included. Probiotic and synbiotic supplementation showed significant improvements in insulin resistance (reductions in HOMA-IR, fasting glucose, and insulin), lipid profiles (decreased LDL and triglycerides; increased HDL), and hormonal balance (increased SHBG, decreased total testosterone). Synbiotics had more pronounced effects than probiotics or prebiotics alone. Adherence was high, and side effects were minimal. Conclusions: Despite promising results, limitations such as small sample sizes, homogeneous populations, and short intervention durations limit the generalization of the findings. Larger, longer, multicenter trials with diverse populations and standardized methodologies are needed to confirm the efficacy and safety of synbiotics in managing PCOS. Integrating these interventions could improve clinical management and quality of life for affected women, but additional evidence is required to support widespread use.
... The main microbiome-induced mechanisms linked to cancer pathogenesis are altered microbial diversity, weakened immune response, and release of genotoxic or carcinogenic substances. [12][13][14][15] The aim of this review is to provide new information regarding the dietary factors linked to the emergence of CRC. It examines the potential role of the gut microbiome, specifically focusing on how it influences the tumorigenesis processes linked to CRC. ...
Article
Full-text available
Diet plays a critical role in shaping the gut microbiome, which in turn regulates molecular activities in the colonic mucosa. The state and composition of the gut microbiome are key factors in the development of colorectal cancer. An altered gut microbiome, linked to weakened immune responses and the production of carcinogenic substances, is a significant contributor to colorectal cancer pathogenesis. Dietary changes that involve low-fiber and phytomolecule intake, coupled with higher consumption of red meat, can raise the risk of colorectal cancer. Salutary filaments, which reach the colon undigested, are metabolized by the gut microbiome, producing short-chain fatty acids. Short-chain fatty acids possess beneficial anti-inflammatory and antiproliferative properties that promote colon health. A well-balanced microbiome, supported by beneficial fibers and phytochemicals, can regulate the activation of proto-oncogenes and oncogenic pathways, thereby reducing cell proliferation. Recent research suggests that an overabundance of specific microbes, such as Fusobacterium nucleatum, may contribute to adverse changes in the colonic mucosa. Positive lifestyle adjustments have been demonstrated to effectively inhibit the growth of harmful opportunistic organisms. Synbiotics, which combine probiotics and prebiotics, can protect the intestinal mucosa by enhancing immune responses and decreasing the production of harmful metabolites, oxidative stress, and cell proliferation. This narrative review provides a concise understanding of evolving evidence regarding how diet influences the gut microbiome, leading to the restoration of the colonic epithelium. It underscores the importance of a healthy, plant-based diet and associated supplements in preventing colorectal cancer by enhancing gut microbiome health.
Article
The COVID-19 pandemic has highlighted the need for a comprehensive understanding of the factors influencing disease severity and progression. Emerging research indicates that the human microbiota, particularly beneficial bacteria, significantly impacts immune responses and health outcomes in COVID-19 patients. While existing studies provide general insights into the relationship between the microbiota and probiotics with COVID-19, they often lack a detailed exploration of how specific bacterial taxa might be used as adjunctive treatments. This review aims to address this gap by focusing on ten key genera of beneficial bacteria, discussing their roles in COVID-19 and evaluating their potential as probiotics for prevention and treatment. The review covers the impact of these microbes on human health, their population alterations in COVID-19 patients, and their interactions with other viral infections. Among these microbes, several exhibit distinct patterns of abundance in COVID-19 patients, influencing disease outcomes and highlighting their potential roles in infection dynamics. In COVID-19 patients, populations of Akkermansia, Ruminococcus, and Roseburia are consistently reduced, while those of Faecalibacterium show a significant decline in more severe cases. Bacteroides presents varying effects depending on the species involved. Alterations in the abundance of Blautia and Lachnospiraceae are associated with increased inflammation and disease severity. Likewise, the depletion of Lachnospira and Coprococcus populations, both linked to anti-inflammatory effects, may exacerbate symptom severity. Oscillospira, though less studied, is connected to overall health and could have implications for viral infections. This review synthesizes the current understanding of these beneficial microbes to highlight the importance of maintaining a healthy microbiota to alleviate the impact of COVID-19 and contribute to the development of novel therapeutic strategies involving microbiota modulation.
Article
Full-text available
Objective To investigate whether a whole grain diet alters the gut microbiome and insulin sensitivity, as well as biomarkers of metabolic health and gut functionality. Design 60 Danish adults at risk of developing metabolic syndrome were included in a randomised cross-over trial with two 8-week dietary intervention periods comprising whole grain diet and refined grain diet, separated by a washout period of ≥6 weeks. The response to the interventions on the gut microbiome composition and insulin sensitivity as well on measures of glucose and lipid metabolism, gut functionality, inflammatory markers, anthropometry and urine metabolomics were assessed. Results 50 participants completed both periods with a whole grain intake of 179±50 g/day and 13±10 g/day in the whole grain and refined grain period, respectively. Compliance was confirmed by a difference in plasma alkylresorcinols (p<0.0001). Compared with refined grain, whole grain did not significantly alter glucose homeostasis and did not induce major changes in the faecal microbiome. Also, breath hydrogen levels, plasma short-chain fatty acids, intestinal integrity and intestinal transit time were not affected. The whole grain diet did, however, compared with the refined grain diet, decrease body weight (p<0.0001), serum inflammatory markers, interleukin (IL)-6 (p=0.009) and C-reactive protein (p=0.003). The reduction in body weight was consistent with a reduction in energy intake, and IL-6 reduction was associated with the amount of whole grain consumed, in particular with intake of rye. Conclusion Compared with refined grain diet, whole grain diet did not alter insulin sensitivity and gut microbiome but reduced body weight and systemic low-grade inflammation. Trial registration number NCT 01731366; Results.
Article
Full-text available
Technical variation in metagenomic analysis must be minimized to confidently assess the contributions of microbiota to human health. Here we tested 21 representative DNA extraction protocols on the same fecal samples and quantified differences in observed microbial community composition. We compared them with differences due to library preparation and sample storage, which we contrasted with observed biological variation within the same specimen or within an individual over time. We found that DNA extraction had the largest effect on the outcome of metagenomic analysis. To rank DNA extraction protocols, we considered resulting DNA quantity and quality, and we ascertained biases in estimates of community diversity and the ratio between Gram-positive and Gram-negative bacteria. We recommend a standardized DNA extraction method for human fecal samples, for which transferability across labs was established and which was further benchmarked using a mock community of known composition. Its adoption will improve comparability of human gut microbiome studies and facilitate meta-analyses.
Article
Full-text available
Background Medical devices play an important role in the diagnosis, prevention, treatment and care of diseases. However, compared to pharmaceuticals, there is no rigorous formal regulation for demonstration of benefits and exclusion of harms to patients. The medical device industry argues that the classical evidence hierarchy cannot be applied for medical devices, as randomised clinical trials are impossible to perform. This article aims to identify the barriers for randomised clinical trials on medical devices. Methods Systematic literature searches without meta-analysis and internal European Clinical Research Infrastructure Network (ECRIN) communications taking place during face-to-face meetings and telephone conferences from 2013 to 2017 within the context of the ECRIN Integrating Activity (ECRIN-IA) project. ResultsIn addition to the barriers that exist for all trials, we identified three major barriers for randomised clinical trials on medical devices, namely: (1) randomisation, including timing of assessment, acceptability, blinding, choice of the comparator group and considerations on the learning curve; (2) difficulties in determining appropriate outcomes; and (3) the lack of scientific advice, regulations and transparency. Conclusions The present review offers potential solutions to break down the barriers identified, and argues for applying the randomised clinical trial design when assessing the benefits and harms of medical devices.
Article
Full-text available
Background Evidence-based clinical research poses special barriers in the field of nutrition. The present review summarises the main barriers to research in the field of nutrition that are not common to all randomised clinical trials or trials on rare diseases and highlights opportunities for improvements. Methods Systematic academic literature searches and internal European Clinical Research Infrastructure Network (ECRIN) communications during face-to-face meetings and telephone conferences from 2013 to 2017 within the context of the ECRIN Integrating Activity (ECRIN-IA) project. Results Many nutrients occur in multiple forms that differ in biological activity, and several factors can alter their bioavailability which raises barriers to their assessment. These include specific difficulties with blinding procedures, with assessments of dietary intake, and with selecting appropriate outcomes as patient-centred outcomes may occur decennia into the future. The methodologies and regulations for drug trials are, however, applicable to nutrition trials. Conclusions Research on clinical nutrition should start by collecting clinical data systematically in databases and registries. Measurable patient-centred outcomes and appropriate study designs are needed. International cooperation and multistakeholder engagement are key for success. Electronic supplementary material The online version of this article (doi:10.1186/s13063-017-2160-8) contains supplementary material, which is available to authorized users.
Article
Full-text available
Based on the abundance of specific bacterial genera, the human gut microbiota can be divided into two relatively stable groups that might play a role in personalized nutrition. We studied these simplified enterotypes as prognostic markers for successful body fat loss on two different diets. A total of 62 participants with increased waist circumference were randomly assigned to receive an ad libitum New Nordic Diet (NND) high in fiber/wholegrain or an Average Danish Diet (ADD) for 26 weeks. Participants were grouped into two discrete enterotypes by their relative abundance of Prevotella spp. divided by Bacteroides spp. (P/B ratio) obtained by quantitative PCR analysis. Modifications of dietary effects of pre-treatment P/B group were examined by linear mixed models. Among individuals with high P/B the NND resulted in a 3.15 kg (95%CI 1.55;4.76, P<0.001) larger body fat loss compared to ADD whereas no differences was observed among individuals with low P/B (0.88 kg [95% CI -0.61;2.37, P=0.25]). Consequently, a 2.27 kg (95%CI 0.09;4.45, P=0.041) difference in responsiveness to the diets were found between the two groups. In summary, subjects with high P/B-ratio appeared more susceptible to lose body fat on diets high in fiber and wholegrain than subjects with a low P/B-ratio.International Journal of Obesity accepted article preview online, 08 September 2017. doi:10.1038/ijo.2017.220.
Article
Full-text available
Background Randomised clinical trials are key to advancing medical knowledge and to enhancing patient care, but major barriers to their conduct exist. The present paper presents some of these barriers. Methods We performed systematic literature searches and internal European Clinical Research Infrastructure Network (ECRIN) communications during face-to-face meetings and telephone conferences from 2013 to 2017 within the context of the ECRIN Integrating Activity (ECRIN-IA) project. ResultsThe following barriers to randomised clinical trials were identified: inadequate knowledge of clinical research and trial methodology; lack of funding; excessive monitoring; restrictive privacy law and lack of transparency; complex regulatory requirements; and inadequate infrastructures. There is a need for more pragmatic randomised clinical trials conducted with low risks of systematic and random errors, and multinational cooperation is essential. Conclusions The present paper presents major barriers to randomised clinical trials. It also underlines the value of using a pan-European-distributed infrastructure to help investigators overcome barriers for multi-country trials in any disease area.
Article
Background: Inflammatory bowel disease is a global disease in the 21st century. We aimed to assess the changing incidence and prevalence of inflammatory bowel disease around the world. Methods: We searched MEDLINE and Embase up to and including Dec 31, 2016, to identify observational, population-based studies reporting the incidence or prevalence of Crohn's disease or ulcerative colitis from 1990 or later. A study was regarded as population-based if it involved all residents within a specific area and the patients were representative of that area. To be included in the systematic review, ulcerative colitis and Crohn's disease needed to be reported separately. Studies that did not report original data and studies that reported only the incidence or prevalence of paediatric-onset inflammatory bowel disease (diagnosis at age <16 years) were excluded. We created choropleth maps for the incidence (119 studies) and prevalence (69 studies) of Crohn's disease and ulcerative colitis. We used temporal trend analyses to report changes as an annual percentage change (APC) with 95% CI. Findings: We identified 147 studies that were eligible for final inclusion in the systematic review, including 119 studies of incidence and 69 studies of prevalence. The highest reported prevalence values were in Europe (ulcerative colitis 505 per 100 000 in Norway; Crohn's disease 322 per 100 000 in Germany) and North America (ulcerative colitis 286 per 100 000 in the USA; Crohn's disease 319 per 100 000 in Canada). The prevalence of inflammatory bowel disease exceeded 0·3% in North America, Oceania, and many countries in Europe. Overall, 16 (72·7%) of 22 studies on Crohn's disease and 15 (83·3%) of 18 studies on ulcerative colitis reported stable or decreasing incidence of inflammatory bowel disease in North America and Europe. Since 1990, incidence has been rising in newly industrialised countries in Africa, Asia, and South America, including Brazil (APC for Crohn's disease +11·1% [95% CI 4·8-17·8] and APC for ulcerative colitis +14·9% [10·4-19·6]) and Taiwan (APC for Crohn's disease +4·0% [1·0-7·1] and APC for ulcerative colitis +4·8% [1·8-8·0]). Interpretation: At the turn of the 21st century, inflammatory bowel disease has become a global disease with accelerating incidence in newly industrialised countries whose societies have become more westernised. Although incidence is stabilising in western countries, burden remains high as prevalence surpasses 0·3%. These data highlight the need for research into prevention of inflammatory bowel disease and innovations in health-care systems to manage this complex and costly disease. Funding: None.
Article
The microbiota-gut-brain axis (MGBA) regulates the reciprocal interaction between chronic inflammatory bowel and psychiatric disorders. This interaction involves multiple pathways that are highly debated. We examined the behavioural, biochemical and electrophysiological alterations, as well as gut microbiota composition in a model of antibiotic-induced experimental dysbiosis. Inflammation of the small intestine was also assessed. Mice were exposed to a mixture of antimicrobials for 2 weeks. Afterwards, they received Lactobacillus casei DG (LCDG) or a vehicle for up to 7 days via oral gavage. Perturbation of microbiota was accompanied by a general inflammatory state and alteration of some endocannabinoidome members in the gut. Behavioural changes, including increased immobility in the tail suspension test and reduced social recognition were observed, and were associated with altered BDNF/TrkB signalling, TRPV1 phosphorylation and neuronal firing in the hippocampus. Moreover, morphological rearrangements of non-neuronal cells in brain areas controlling emotional behaviour were detected. Subsequent probiotic administration, compared with vehicle, counteracted most of these gut inflammatory, behavioural, biochemical and functional alterations. Interestingly, levels of Lachnospiraceae were found to significantly correlate with the behavioural changes observed in dysbiotic mice. Our findings clarify some of the biomolecular and functional modifications leading to the development of affective disorders associated with gut microbiota alterations.
Article
A general consensus exists that IBD is associated with compositional and metabolic changes in the intestinal microbiota (dysbiosis). However, a direct causal relationship between dysbiosis and IBD has not been definitively established in humans. Findings from animal models have revealed diverse and context-specific roles of the gut microbiota in health and disease, ranging from protective to pro-inflammatory actions. Moreover, evidence from these experimental models suggest that although gut bacteria often drive immune activation, chronic inflammation in turn shapes the gut microbiota and contributes to dysbiosis. The purpose of this Review is to summarize current associations between IBD and dysbiosis, describe the role of the gut microbiota in the context of specific animal models of colitis, and discuss the potential role of microbiota-focused interventions in the treatment of human IBD. Ultimately, more studies will be needed to define host–microbial relationships relevant to human disease and amenable to therapeutic interventions.