ArticlePDF AvailableLiterature Review

Diet and the development of the human intestinal microbiome

Frontiers
Frontiers in Microbiology
Authors:

Abstract and Figures

The important role of the gut microbiome in maintaining human health has necessitated a better understanding of the temporal dynamics of intestinal microbial communities as well as the host and environmental factors driving these dynamics. Genetics, mode of birth, infant feeding patterns, antibiotic usage, sanitary living conditions and long term dietary habits contribute to shaping the composition of the gut microbiome. This review focuses primarily on diet, as it is one of the most pivotal factors in the development of the human gut microbiome from infancy to the elderly. The infant gut microbiota is characterized by a high degree of instability, only reaching a state similar to that of adults by 2-3 years of age; consistent with the establishment of a varied solid food diet. The diet-related factors influencing the development of the infant gut microbiome include whether the child is breast or formula-fed as well as how and when solid foods are introduced. In contrast to the infant gut, the adult gut microbiome is resilient to large shifts in community structure. Several studies have shown that dietary changes induce transient fluctuations in the adult microbiome, sometimes in as little as 24 hours; however, the microbial community rapidly returns to its stable state. Current knowledge of how long-term dietary habits shape the gut microbiome is limited by the lack of long-term feeding studies coupled with temporal gut microbiota characterization. However, long-term weight loss studies have been shown to alter the ratio of the Bacteriodetes and Firmicutes, the two major bacterial phyla residing in the human gastrointestinal tract. With ageing, diet-related factors such as malnutrition are associated with microbiome shifts, although the cause and effect relationship between these factors has not been established. Increased pharmaceutical usage is also more prevalent in the elderly and can contribute to reduced gut microbiota stability and diversity. Foods con
Content may be subject to copyright.
REVIEW ARTICLE
published: 22 September 2014
doi: 10.3389/fmicb.2014.00494
Diet and the development of the human intestinal
microbiome
Noah Voreades, Anne Kozil and Tiffany L. Weir*
Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA
Edited by:
Anton G. Kutikhin, Research Institute
for Complex Issues of Cardiovascular
Diseases under the Siberian Branch of
the Russian Academy of Medical
Sciences, Russia
Reviewed by:
Carl James Yeoman, Montana State
University, USA
Franck Carbonero, University of
Arkansas, USA
*Correspondence:
Tiffany L. Weir, Department of Food
Science and Human Nutrition,
Colorado State University, 1571
Campus Delivery, 210 Gifford
Building, Fort Collins, CO 80523-1571,
USA
e-mail: tiffany.weir@colostate.edu
The important role of the gut microbiome in maintaining human health has necessitated a
better understanding of the temporal dynamics of intestinal microbial communities as well
as the host and environmental factors driving these dynamics. Genetics, mode of birth,
infant feeding patterns, antibiotic usage, sanitary living conditions and long term dietary
habits contribute to shaping the composition of the gut microbiome. This review focuses
primarily on diet, as it is one of the most pivotal factors in the development of the human
gut microbiome from infancy to the elderly. The infant gut microbiota is characterized by
a high degree of instability, only reaching a state similar to that of adults by 2–3 years
of age; consistent with the establishment of a varied solid food diet. The diet-related
factors influencing the development of the infant gut microbiome include whether the
child is breast or formula-fed as well as how and when solid foods are introduced. In
contrast to the infant gut, the adult gut microbiome is resilient to large shifts in community
structure. Several studies have shown that dietary changes induce transient fluctuations
in the adult microbiome, sometimes in as little as 24 h; however, the microbial community
rapidly returns to its stable state. Current knowledge of how long-term dietary habits
shape the gut microbiome is limited by the lack of long-term feeding studies coupled
with temporal gut microbiota characterization. However, long-term weight loss studies
have been shown to alter the ratio of the Bacteroidetes and Firmicutes, the two major
bacterial phyla residing in the human gastrointestinal tract. With aging, diet-related factors
such as malnutrition are associated with microbiome shifts, although the cause and effect
relationship between these factors has not been established. Increased pharmaceutical
usage is also more prevalent in the elderly and can contribute to reduced gut microbiota
stability and diversity. Foods containing prebiotic oligosaccharide components that nurture
beneficial commensals in the gut community and probiotic supplements are being explored
as interventions to manipulate the gut microbiome, potentially improving health status.
Keywords: enterotype, gut microbiome, aging, dietary patterns, colonization
IMPORTANCE OF THE GUT MICROBIOME
The consortium of single-celled organisms residing in our
intestines, the gut microbiome, is rapidly emerging as an impor-
tant determinant of health. Deterrents to proper bacterial col-
onization in early life are hypothesized to contribute to food
sensitivities, allergic reactions, Type I diabetes, and other autoim-
mune disorders (Kelly et al., 2007). Association of the microbiome
to autoimmune diseases has been explained by the “hygiene
hypothesis,” which suggests that the absence of a robust micro-
biome results in defects in development and regulation of the
immune system, resulting in a lack of immune tolerance (Okada
et al., 2010;Rook, 2012). Later in life, strong evidence supports an
important role for intestinal microbiota in weight regulation via
contributions to dietary energy harvest and appetite control (Tilg
and Kaser, 2011). The gut microbiome has also been implicated in
the pathology of several intestinal inflammatory diseases as well
as in the development of colorectal, gastric, and prostate cancers
and cardiometabolic disorders (Sekirov et al., 2010). Mechanisms
giving rise to these conditions include the production of geno-
toxins by bacterial pathogens, microbial metabolism of dietary
components to produce carcinogenic compounds, and inciting
local and systemic inflammatory cascades that result in chronic low
grade inflammation and damage to affected tissues and organs.
While a dysbiotic microbiota can cause disease, a healthymicro-
bial community is vital to assist the host in maintaining optimal
wellness. Thus, there is a need to understand the factors that shape
and alter the microbiome throughout the lifespan of an individ-
ual. Numerous elements, encompassing environmental exposures,
genetics, and other inherent host factors, contribute to the initial
colonization of the microbiome in infants and to the subtle shifts
that occur in adults, occasionally culminating in microbial decline
as observed in frail and unhealthy elderly individuals (Koenig et al.,
2011). However, none of these factors may be as important in the
development of the microbiome as diet. In this review we will
present evidence for the importance of diet in initial coloniza-
tion events and in determining the composition of a stable adult
microbiome. Factors such as malnutrition and pharmaceutical
interventions on the aging gut will also be reviewed. Finally, we
will discuss potential interventions, including dietary changes that
can be used to alter the intestinal microbial community.
www.frontiersin.org September 2014 |Volume 5 |Article 494 |1
Voreades etal. Diet and the gut microbiome
EARLY MICROBIAL COLONIZATION AND ESTABLISHMENT
The infant gut is thought to be sterile at birth, although some
new research characterizing the placental microbiome challenges
that assumption (Aagaard et al., 2014). After birth initial colo-
nization and early establishment of the infant gut is influenced
by whether delivery was vaginal or caesarean, feeding patterns,
sanitary conditions, and antibiotic administration (Marques et al.,
2010). The relative importance of these factors on the long-term
structure of the intestinal microbial community and associated
health outcomes is still debated. It stands to reason that with con-
stant exposure between the microbiome and food components
that diet is one of the primary drivers shaping the changes that
occur during infancy and the structure of the adult microbiome
that eventually establishes. This section will focus on the diet’s role
in shaping the infant gut microbiome from birth to 3yearsof
age. Specifically, the following topics will be explored in detail: (1)
the influence of breast vs. formula-feeding in initial colonization,
(2) changes related to beginning of weaning and introduction of
solid foods, and (3) factors contributing to a stable gut microbiome
profile (Figure 1).
BREAST vs. FORMULA FEEDING
Following birth, the infant gut microbiome is characterized
by low-species diversity and high rates of bacterial flux until
2 or 3 years old (Bergström et al., 2014). Facultative anaerobic
bacteria including Staphylococcus, Streptococcus, Escherichia coli
and Enterobacteria are thought to be the first colonizers of the
gut. Their purpose is to consume oxygen and create an environ-
ment for obligate anaerobes to thrive (Palmer et al., 2007;Jost
et al., 2012). These are later replaced by facultative anaerobes that
dominate the gastrointestinal tract, primarily Actinobacteria and
Firmicutes (Turroni et al., 2012). This change in dominant taxa
representation can be attributed to the introduction of breast
or formula-feeding, signifying the first diet-related colonization
event in the infant gut microbiome (Harmsen et al., 2000;Jost
et al., 2012). In breast-fed infants, the dominant Actinobacteria
are represented by Bifidobacterium species, specifically,B.breve,
B. longum,B. dentium,B. infantis, and B. pseudocatenulatum
(Harmsen et al., 2000;Jost et al., 2012). The Firmicutes phylum
is represented principally by lactic acid bacteria such as Lacto-
bacillus and Enterococcus as well as Clostridium species (Turroni
et al., 2012;Bergström et al., 2014). More than 700 species of bac-
teria have now been identified in human colostrum and breast
milk, including multiple species of lactic acid bacteria as well as
species typically colonizing the oral cavity of infants (Cabrera-
Rubio et al., 2012). While this may contribute to the intestinal
community of breastfed infants, it is still unclear whether the com-
position of species in breast milk is driven by transfer from infant
to mother. The chemical composition of breast milk does influence
the gut microbiome through supplying unique oligosaccharides
that are selectively utilized by Bifidobacterium spp. (Turroni et al.,
2012).
There are conflicting reports regarding differences in the rel-
ative abundance of these bacteria between breast and formula
fed infants. Many studies have reported that formula-fed infants
display dominance of Bifidobacterium spp. similar to what has
been observed in breastfed infants (Harmsen et al., 2000;Fallani
et al., 2010,2011). However, another study reported approxi-
mately double the count of Bifidobacterium in breast fed infants
compared to those fed formula (Bezirtzoglou et al., 2011). For-
mula feeding was also associated with higher levels of Atopo-
bium (Bezirtzoglou et al., 2011); which corroborated reports by
Fallani et al. (2010),although they only noted Atopobium increases
in formula fed infants delivered by Cesearean section or whose
mother’s had been administered antibiotics. Higher numbers
of Bacteroides spp. as well as members of the Enterobacteri-
aceae have also been reported in formula-fed infants (Harmsen
et al., 2000;Fallani et al., 2010). Despite significant evidence
that Bifidobacterium is an important early colonizer in neonates,
Palmer et al. (2007) reported that Bifidobacterium was not present
in significant amounts in the infant gut (Palmer et al., 2007).
However, it is important to highlight that within their cohort,
there was a mixture of breast and formula-feeding, antibiotics
were provided to infants and a small subset required specialized
hospitalization.
The variability reported with regard to Bifidobacterium
abundance could be driven by differences in infant for-
mula composition. Formulas supplemented with the prebiotics
FIGURE 1 |Representation of the infant gut microbiome development from birth to 3 years of age. By 3 years old, toddler’s microbiomes are similar to
that in adults and long-term dietary patterns are beginning to establish.
Frontiers in Microbiology |Evolutionary and Genomic Microbiology September 2014 |Volume 5 |Article 494 |2
Voreades etal. Diet and the gut microbiome
galacto-oligosaccharide (GOS) and fructo-oligosaccharide (FOS)
may account for high levels of Bifidobacterium found in many
formula-fed infants (Marques et al., 2010;Oozeer et al., 2013). A
recent review discusses evidence supporting GOS and FOS sup-
plementation effects on the gut (Oozeer et al., 2013). Infant gut
microbial populations provided with either human breast milk
or prebiotic supplemented infant formula had similar levels of
Bifidobacterium; whereas gut microbial populations of infants
given traditional formula was reported to have about 20% fewer
Bifidobacterium (Knol et al., 2005). Additionally, the species com-
position of Bifidobacterium was similar between infants given
human breast milk and those on prebiotic supplemented for-
mula. However, traditional formula fed infants had markedly
different gut microbial communities and even the specific Bifi-
dobacterium species differed with higher relative abundances of
B. cantenulatum and B. adolescentis, which are typically repre-
sented in adult populations. Another potential explanation for
the variation in studies reporting bacterial abundances, partic-
ularly with regard to breast-feeding could be due to differences
in the maternal-diet (Cabrera-Rubio et al., 2012). Characteriza-
tion of the placental microbiome suggests that it is colonized
by the mother’s oral microbiome (Aagaard et al., 2014). Another
recent study showing that pre and post-natal maternal con-
sumption of a high fat diet, independent of obesity in the
mother, resulted in dysbiosis of the infant gut in a primate
model (Ma et al., 2014). Together, these studies suggest that
maternal diet may play a significant but previously unrecognized
role in determining early colonization and establishment of the
infant microbiome. Conduct of randomized trials in which the
maternal diet is controlled or large-scale cross-sectional studies
of pregnant mothers adhering to different diets (Western, veg-
etarian, gluten-free, etc) are necessary to further develop this
hypothesis.
WEANING AND THE SHIFT TOWARD AN ADULT MICROBIOME
Around the age of 1–2 years old, the infant gut microbiome
undergoes its second shift and the stable adult microbiome
begins to emerge, further supporting the significant role of
the diet in influencing the microbial community (De Fil-
ippo et al., 2010;Bergström et al., 2014). One study reported
that although there were differences in the microbiome pre-
and post-weaning, the impacts of earlier colonization events
(delivery mode, formula or breastfed, etc.) were still appar-
ent (Fallani et al., 2011). Another study comparing Italian vs.
African children’s gut microbiomes showed that after wean-
ing and solid foods were introduced there was a significant
diet-related shift in the gut microbiome profiles. Prior to the
introduction of their respective Western or African diets, the
children across both populations that were still breast-feeding
clustered together and had similar Bifidobacterium species domi-
nance. Only children who were already weaned reliably clustered
together into distinct geographic groupings. This study rein-
forced two important points related to dietary drivers of the
gut microbiome development in children. First, breast-feeding,
regardless of duration supports a specific bacterial state that
is unique and markedly different from that observed in indi-
viduals consuming solid foods. Second, once solid foods are
introduced, its role in shaping long-term gut microbiome pro-
files is so strong that individual’s cluster based on diet type over
other environmental and physiological factors (De Filippo et al.,
2010).
A similarly significant shift was reported by Bergström et al.
(2014) ina3yearDanish study with a cohort of 330 infants.
They reported that between 9 and 18 months, the infant gut bac-
terial abundances changed drastically with the introduction of
solid foods. Specifically, Bacteroidetes-related species increased.
Whereas Bifidobacterium and Lactobacillus species and Enter-
obacteriaceae declined, various species within Firmicutes phylum
were also reported to increase. This bacterial taxa shift is logi-
cal given that breast and/or formula-feeding has ceased, depleting
the primary fuel source for these bacteria. In addition, butyrate
producing bacteria such as Clostridium leptum group, E. halli, and
Roseburia species increased. Typically, butyrate producing bacteria
are responsible for the breakdown of otherwise indigestible com-
plex plant polysaccharides and resistant starches. Anecdotally, this
study found that the longer infants were breast and/or formula-fed,
the lower their levels of butyrate producing bacteria. Additionally,
more and different species begin to appear with introduction of
solid foods (Koenig et al., 2011;Bergström et al., 2014).
EMERGENCE OF A STABLE GUT PROFILE
From 18 to 36 months, the infant gut microbiome undergoes its
final significant shift to a more stable microbial profile composed
primarily of the bacterial phyla Bacteriodetes and Firmicutes.
This shift represents a temporal change that can be attributed
to the continued influence of a varied solid food diet (De Fil-
ippo et al., 2010;Koenig et al., 2011;Bergström et al., 2014).
The earlier that solid food is introduced into the diet, the more
quickly the gut microbiome begins to resemble a stable adult-
like microbiome (Bergström et al., 2014). The specific proportion
of Firmicutes and Bacteroidetes is strongly influenced by diet.
This was best demonstrated in the previously discussed work
by De Filippo et al. (2010) where the distinct microbial signa-
tures of the two groups of children were indicative of their
respective dietary habits. The most compelling evidence for this
was the dominance of Prevotella, capable of digesting complex
plant polysaccharides, in African children and its absence in Ital-
ian children. Similar diet-driven influences were reported in a
detailed temporal study of a single infant. This study demon-
strated that introduction of peas, formula, and other solid foods
led to an emerging co-dominance between Firmicutes and Bac-
teroidetes, with the increase in Bacteroidetes potentially resulting
from requirements for the breakdown of newly introduced plant
polysaccharides (Koenig et al., 2011). The previously mentioned
emergence of a stable gut microbiome can be substantially derailed
if the infant experiences either severe acute malnutrition or mod-
erate acute malnutrition. Emerging research is demonstrating
that either of these malnutrition states has the potential to sig-
nificantly alter the development of a healthy gut microbiome
profile, regardless of diet-based interventions (Subramanian et al.,
2014). These recent findings not only support a link between
diet and the development of a particular gut microbiota and
microbiome, but illustrate that nutrient quantity can impact
development too.
www.frontiersin.org September 2014 |Volume 5 |Article 494 |3
Voreades etal. Diet and the gut microbiome
THE ADULT MICROBIOME
The typical adult intestinal microbiome is primarily comprised
of approximately six or seven different bacterial phyla, of which
Bacteroidetes and Firmicutes dominate (Eckburg et al., 2005).
Less abundant phyla can include Proteobacteria, Verrucomicro-
biota, Actinobacteria, and Euryarchaeota. A recent study followed
changes in the microbiome of 37 adults for up to 5 years and
reported that 60–70% of the bacterial strains present remained
unchanged over the course of the study and that the most sta-
ble members of the microbiome tended to be the most abundant
(Faith et al., 2013). They also observed that at the phyla level,
Bacteroidetes and Actinobacteria populations were less suscepti-
ble to perturbations whereas Firmicutes and Proteobacteria were
significantly less stable. These results are fairly consistent with find-
ings from an earlier study utilizing a microarray-based approach
to determine molecular taxonomy and which followed a smaller
cohort over a longer period of time (Rajilic-Stojanovic et al., 2013).
Both studies reported that the taxa present in an individual remain
fairly consistent over time, although the relative abundances of
these taxa were subject to change. However, data from Rajilic-
Stojanovic et al. (2013) suggests that larger fluctuations occur
between samples taken at longer intervals while Faith et al. (2013)
report the opposite trend, with larger fluctuations occurring in
samples taken over shorter periods of time compared to those
that are temporally farther apart. Despite this resilience, there is
evidence that the diet shapes the relative abundance of dominant
phyla and populations of specific bacterial groups are influenced
by the composition of macronutrients consumed.
DIET-DRIVEN ENTEROTYPES
There have been numerous attempts to identify a “core” micro-
biota, usually defined as bacterial taxa that are shared between
95% of individuals tested (Huse et al., 2012). Identification of a
core microbiome is important for defining a “normal” healthy
state from which major variations may indicate a dysbiotic system
that can result from or contribute to disease development. One
barrier to defining an intestinal core microbiome has been the
vast degree of variation between individuals. The microbial com-
munities identified in samples collected from an individual over
time are more similar to each other than microbial communi-
ties between two individuals, although related persons share more
bacterial strains than unrelated individuals (Palmer et al., 2007;
Yatsunenko et al., 2012;Faith et al., 2013). Although a consensus
for what constitutes a core gut microbiome has been elusive, one
report suggested that an international cohort of 39 individuals
could be assigned to one of three distinct clusters or “enterotypes”
based on metagenomic sequences (Arumugam et al., 2011). They
found that each cluster was dominated by a particular bacterial
genus (Bacteroides,Prevotella, and Ruminococcus) with positive
or negative associations with a number of other genera in the
community. They also reported that each cluster was enriched for
specific gene functions that reflected different microbial trophic
chains. Two of the three original enterotypes, Bacteroides, and Pre-
votella, were later confirmed and long term dietary patterns were
identified as the primary predictor of an individual’s enterotype
(Wu et al., 2011). The Bacteroides enterotype was associated with
a Western-type diet high in proteins and fat, while the Prevotella
enterotype was associated with plant fiber consumption. These
enterotypes appear to be extremely stable, and several studies
utilizing short-term interventions failed to result in a change in
the assigned enterotype of participants (David et al., 2014;Roager
et al., 2014).
The existence of enterotypes provided a convenient way of
classifying individuals based on their fecal microbiota (although
some argue a more appropriate term would be “faecotype”) and
speculation has begun as to whether enterotypes can be used
as a predictor of long term health risks. However, a microbial
survey of several body sites, including stool, conducted with
more than 200 individuals showed only minimal segregation
into the Bacteroides and Prevotella enterotypes rather than the
distinct and well separated clusters previously reported (Huse
et al., 2012). These discrepancies could be due to the fact that
the method for assigning enterotypes is not consistent across
studies. An analysis of archived 16S sequences also showed that
enterotype determination is sensitive to clustering methods and
distance metrics used and that there is a continuum of Bacteroides
abundances across samples rather than a bimodal distribution
(Koren et al., 2013). These studies suggest that the enterotype
concept is not be as clear cut as previously believed, and that
standard methods for defining enterotypes should be developed
and employed before they can be meaningfully tied with clinical
outcomes.
LONG TERM DIETARY PATTERNS AND THE MICROBIOME
Whether enterotypes truly exist or not, it is clear that diet is
an important factor in shaping the microbiome (Figure 2). In
addition to the divergence in microbial composition of Italian
children and those from Burkina Faso shortly after weaning (De
Filippo et al., 2010); other studies have shown microbiota seg-
regation of individuals from Malawi, Venezuela, and the United
States (Yatsunenko et al., 2012); children from Bangladesh and
the United States (Lin et al., 2013), and between rural Africans
and African Americans (Ou et al., 2013) that are at least partially
diet-driven. In the Yatsunenko etal. (2012) study, metagenomic
sequences revealed that enzyme classifications associated with
protein degradation and bile salt metabolism were enriched in
samples from the U.S. population where protein and fat consump-
tion is high. Conversely, glutamate synthase and starch degrading
enzymes were more abundant in the Amerindian and Malawian
samples; consistent with protein poor diets of corn and cassava.
This has been further demonstrated in a recent study of the diver-
sity and metabolism of the microbiome of a Tanzanian hunter
gatherer tribe, the Hadza. This study identified differences in
the microbiome between the sexes which were consistent with
their division of labor with regard to foraging (Schnorr et al.,
2014). They also have many bacterial species associated with fer-
mentation of plant-based fibers and are completely deficient in
Bifidobacterium, which was hypothesized to result from the lack of
meat and dairy in the diet; substrates that allow these bacteria to
continue to colonize Westerners into adulthood. Although com-
parative studies between populations with different diets has been
useful in identifying how dietary patterns shape the microbiome,
these studies have utilized international cohorts that introduce
confounding factors such as extreme differences in culture and
Frontiers in Microbiology |Evolutionary and Genomic Microbiology September 2014 |Volume 5 |Article 494 |4
Voreades etal. Diet and the gut microbiome
FIGURE 2 |The adult gut microbiome is characterized as existing in a steady state that requires a major disturbance to permanently alter that state.
Short-term diet interventions may transiently alter the gut microbiome community structure, but long-term diet changes are required to shift to a new
steady-state.
environment. Relatively few studies have been conducted that
examine the effects of diet on homogenous populations. One
study looked at correlations between specific dietary components
and microbial function and structure in the intestines of a human
cohort known for keeping meticulous diet logs (Muegge et al.,
2011). They found that there were significant correlations between
microbial gene function (Kegg orthologs) and protein intake,con-
firming the difference that was seen across multiple mammalian
species between carnivores and herbivores. They also reported
a correlation between insoluble fiber consumption and bacterial
community membership. A large-scale microbiome sequencing
effort called the American Gut Project is currently underway
and is attempting to address the effects of diet on the adult
microbiome capturing extremes within the American diet (i.e.,
vegan, paleo, etc) where cultural and environmental factors will
be minimized.
DIETARY INTERVENTIONS INTRODUCE TRANSIENT AND SUBTLE
CHANGES IN THE MICROBIOME
Short-term dietary interventions that include introducing novel
food components or altering macronutrient levels have also been
examined for their effects on intestinal microbial populations.
The first of these studies followed obese individuals partitioned
to restricted calorie diet groups that controlled for either fat or
carbohydrate intake (Ley et al., 2006). Regardless of the macronu-
trient composition of the diet, individuals that lost a significant
amount of body weight had a change in their ratio of Bacteroidetes
to Firmicutes, driven by increases in the Bacteroidetes. Weight-
loss driven changes in the microbiome was recently confirmed
in individuals consuming a calorie restricted liquid diet where
it was demonstrated that weight stability of an individual was a
better predictor of fecal microbiome stability than time between
sample collections (Faith et al., 2013). However, this and another
study (Duncan et al., 2008) noted changes in members of the
Firmicutes rather than an increase in Bacteroidetes when corre-
sponding weight loss occurred. Calorie restriction in obese and
overweight individuals has also been shown to increase micro-
bial gene richness, a parameter that was correlated to improved
metabolic parameters (Cotillard et al., 2013;Le Chatelier et al.,
2013).
Several studies have noted rapid but transient changes in fecal
microbial composition immediately following the start of a dietary
intervention study. Wu etal. (2011) conducted a controlled feed-
ing experiment in ten individuals randomized to high fat/low fiber
or high fiber/low fat diets and found that although there was no
increase in community similarity between individuals on the same
diet over a period of 10 days, the first 24 h period was considered an
outlier because transient dramatic shifts occurred in the fecal com-
munities of all individuals. Similarly, switching between animal
and plant-based diets produces similar results (David et al., 2014).
Another interesting finding of the David et al. (2014) study was
that foodborne microbes transiently colonized the gut, introduc-
ing the idea that food may not only select for commensal bacterial
species, but serve as a reservoir for new microbial introductions.
Intentional introduction of food-borne microorganisms (probi-
otics) as well as prebiotic food ingredients and foods high in fiber
can also be a means of subtly changing the relative abundance of
bacterial species in the gut (Preidis and Versalovic, 2009). Thus,
despite the inherent stability of the microbiome over time, changes
related to weight loss and diet composition continue to subtly alter
the composition and relative abundance of our commensal organ-
isms, driving the development of our gut microbiome throughout
adulthood.
THE AGING GUT
As a person ages, the stability and diversity of their gut micro-
biota declines with the state of their health. If health remains
intact however, microbiota composition often retains the stability
www.frontiersin.org September 2014 |Volume 5 |Article 494 |5
Voreades etal. Diet and the gut microbiome
and compositional make-up of a healthy younger adult (Claesson
et al., 2012). The most prevalent age-related factors influenc-
ing the microbial population of the gut are: (1) physiological
changes, (2) dietary choices and malnutrition, (3) living situa-
tion (community-dwelling, hospitalized, or long-term care), and
(4) use of antibiotics (Bartosch et al., 2004;Woodmansey, 2007;
Claesson et al., 2012) and other prescription drugs (Qato et al.,
2008). This section will explore dietary alterations and antibiotic
usage as drivers of change in the elderly gut microbiome and dis-
cuss the use of probiotics and prebiotics as potential solutions for
the restoration of a healthy gut.
Diet is a major influence on the bacterial makeup of the aging
gut. Physiological changes, such as loss of taste and smell, difficulty
chewing or swallowing, impaired digestive function, and lack of
physical mobility can leave elderly individuals consuming a narrow
and nutritionally imbalanced diet, setting the stage for malnutri-
tion (Bartosch et al., 2004;Claesson et al., 2012). Relocation from
an in-home community setting to a long-term care facility can
change dietary intake as well. The move often contributes to a
greater consumption of fat and a decreased intake of fiber, fruits,
vegetables, and meat. These dietary alterations are associated with
a decrease in microbial diversity and increased frailty (Claesson
et al., 2012).
The use of antibiotics in elderly populations is especially preva-
lent in hospital and long-term care facilities. Antibiotics create an
environment of instability by diminishing the population of total
and commensal bacteria and opening the door for pathogenic
bacteria to overpopulate (Claesson et al., 2011). The use of
broad-spectrum antibiotics is associated with the overgrowth of
Clostridium difficile which flourishes in the antibiotic-weakened
gut, often resulting in a life threatening infection (Macfarlane,
2014). As health issues compound and antibiotic use increases,
elderly often see a decline in commensal anaerobes (Bacteroides,
Lactobacillus and Bifidobacterium) accompanied by a rise in pro-
teolytic and pathogenic bacteria (Fusobacteria,Propionibacteria,
Clostridia, and E. coli;Wu et al., 2011). Studies indicate that pro-
biotics may have potential as a therapeutic tool to replenish and
recolonize beneficial bacterial species like Bifidobacterium and Lac-
tobacillus, bringing the elderly gut back into balance (Likotrafiti
et al., 2014).
EFFECTS OF DIET AND MALNUTRITION ON THE ELDERLY MICROBIOME
A number of proposed factors contribute to alterations in the
elderly gut ecosystem and diet is a significant driver of change
(Claesson et al., 2012). Dietary intake can change for a number of
reasons with advanced age. Decline in physical mobility may limit
access to the grocery store or inhibit the ability to cook. Some
elderly lose the desire to eat due to loss of smell and taste or due
to slow digestion and prolonged satiety (Britton and McLaugh-
lin, 2013). Malnutrition is often an unintended consequence of
age-related physiological changes that can lead to changes in the
elderly gut microbiome. Furthermore, studies have shown that
compositional dietary changes can result in almost immediate
alterations in microbial populations. Wu et al. (2011) found that
changes in microbiome composition were detectable within 24 h
of dietary alteration and occurred even faster than transit time
of food through the gut. In an infant population, malnutrition
was shown to delay the maturation of the intestinal microbiota
(Subramanian et al., 2014), and it is likely to have consequences of
a similar magnitude in the elderly gut.
Dietary changes that come with age are also impacted by
living situation. Claesson et al. (2012) found distinct dietary
differences between elderly individuals living in a traditional com-
munity setting compared to those in long-term care facilities.
Community-dwellers may be healthier than their institutional-
ized counterparts for a number of reasons, but they broadly
stated that community-dwellers eat a healthier and more diverse
diet and have a distinct microbiota from those in long-term
care facilities (Claesson et al., 2012). The largest dietary differ-
ences were seen in consumption of fruits, vegetables, and meat.
Community-dwellers correlated 98% with a moderate fat/high
fiber diet and long-term care dwellers correlated 83% with high
fat/low fiber diet (Claesson et al., 2012). The gut microbiota of
community-dwellers was more diverse than long-stay subjects and
grouped more closely with healthy young adults, indicating that
age itself is not the driving factor of microbial change. Similar
to young adults, community-dwellers had a higher proportion
of phylum Firmicutes and unclassified bacteria, and abundant
populations of genera Coprococcus,Roseburia,Ruminococcus, and
Butyricoccus when compared to long-term stay individuals. Long-
stay subjects had a higher incidence of frailty accompanied by
a proportional increase in Bacteroidetes and an increased abun-
dance of Alistipes and Oscillibacter when compared to healthier
community-dwelling elderly (Claesson et al., 2012). Increasingly
frail individuals showed a significant 26-fold reduction in the
number of Lactobacillus and a significant sevenfold increase in
the number of Enterobacteriaceae compared to less frail subjects
(van Tongeren et al., 2005).
ANTIBIOTICS
The compounded effects of poor diet, ailing health, and prolonged
stays in a hospital or long-term care facility reduce the prevalence
of protective gut microbiota and give way to detrimental pop-
ulations (Bartosch et al., 2004;Wu et al., 2011). This leaves the
elderly individual vulnerable to infection and disease and a prime
candidate for antibiotic usage. Unfortunately, antibiotic therapies
only exacerbate the flux and instability of the already fragile gut
microbiome in unhealthy elderly. The use of antibiotics in elderly
populations is especially prevalent in hospital and long-term care
facilities and it is estimated that nearly 20% of elderly patients
in hospitals are receiving antibiotic treatment at any given time
(Bartosch et al., 2004).
Antibiotics cause significant disturbances in gut microbiota
resulting in the suppression of both beneficial and pathogenic
species, allowing the overgrowth of antibiotic-resistant strains. In
young, healthy volunteers administered two separate courses of
the antibiotic ciprofloxacin, a dramatic change in the microbiota
was noted, followed by the return to an alternative stable state of
undetermined consequences (Dethlefsen and Relman, 2011). Use
of broad-spectrum antibiotics is associated with the opportunistic
bacterium Clostridium difficile which flourishes in the antibiotic-
weakened gut and results in severe diarrhea (Macfarlane, 2014).
Elderly hospital patients and others with fragile immune systems
are especially susceptible to this life-threatening infection.
Frontiers in Microbiology |Evolutionary and Genomic Microbiology September 2014 |Volume 5 |Article 494 |6
Voreades etal. Diet and the gut microbiome
Most often, elderly individuals exposed to antibiotics see an
increased relative abundance of Bacteroidetes and a significant
increase in Bacteroidetes:Firmicutes ratio (Claesson et al., 2011).
Beneficial anaerobic species in the colon such as Bifidobacterium,
Lactobacillus, and Bacteroides can be drastically reduced or even
eradicated with the use of antibiotics (Bartosch et al., 2004). Bifi-
dobacterium and Lactobacillus are producers of short chain fatty
acids (SCFA’s), a nutrient vital to the proper function of intesti-
nal cells; the loss of these bacteria can be especially detrimental.
A study examining the differences in bacterial colonies between
healthy elderly, hospitalized patients, and hospitalized patients
receiving antibiotics, found that the hospitalized patients receiving
antibiotics saw a significant reduction in the numbers of Bifidobac-
terium spp. and an increased relative abundance of Enterococcus
faecalis compared to the other two groups. In some patients,
the antibiotic treatment eliminated certain bacterial communities
altogether (Bartosch et al., 2004).
Effects of antibiotic treatment on gut microbiota can differ sig-
nificantly with the type and dose of antibiotic administered. A
study by Bartosch et al. (2004) following elderly patients receiv-
ing antibiotics, found that the same antibiotic, clarithromycin,
had different effects on gut microbiota at different doses. A low
dose of the antibiotic decreased the proportion of Bacteroidetes
(Bacteroides and Parabacteroides) and increased Firmicutes (Alis-
tipes) and a high dose increased the proportion of Bacteroidetes
(Parabacteroides) and decreased the proportion of Firmicutes
(Alistipes;Claesson et al., 2011). Countless variables must be con-
sidered with the use of antibiotics in elderly individuals. What
seems like a lifesaving drug may have detrimental effects on the
aging microbiome and the health of the individual. Additional
research is needed to inform practitioners on the safest ways to
use antibiotics on the elderly while supporting their potentially
fragile gut microbiota.
PROBIOTICS AND PREBIOTICS
Probiotics and prebiotics, when taken together or individually,
may be particularly beneficial in restoring the proper microbial
balance to the elderly gut microbiota, helping to mitigate the detri-
mental effects of antibiotic usage and under nutrition. Probiotics
are live microbes that when administered in sufficient quantities
are beneficial to the host. Prebiotics are non-digestible food ingre-
dients such as inulin or various oligosaccharides, which have been
show to selectively stimulate growth of beneficial bacterial popula-
tions in the large intestine. Probiotic foods and supplements often
contain Bifidobacterium and/or Lactobacillus organisms, both of
which are extremely important to proper function of the intes-
tine (Duncan and Flint, 2013). Bifidobacterium and Lactobacillus
are often depleted in elderly individuals as health deteriorates.
Research shows that consumption of probiotics containing these
strains can result in a notable rise in their abundance along
with a reduction of more pathogenic microorganisms in the gut
(Toward et al., 2012). Prebiotics may support the Bifidobacterium
and Lactobacillus species delivered via probiotic supplementation
by providing a fermentable food source for these bacteria, allow-
ing them to flourish. More specifically, it has been reported that
prebiotics have the ability to exert a bifidogenic effect on human
subjects (O’Connor et al., 2014).
Arecentin vitro study showed promise that the elderly gut
microbiota can in fact be modulated with appropriate probiotics.
Species of Bifidobacterium and Lactobacillus along with two prebi-
otics were added to the fecal batch culture of elderly participants.
The addition of the beneficial bacteria significantly increased
the Bifidobacterium and decreased the Bacteroides count after
fermentation (Likotrafiti et al., 2014). Both probiotic/prebiotic
combinations added to the culture increased the Bifidobacterium
and Lactobacillus count in the vessel representing the distal colon.
These results represented a major shift in the gut microbiota
toward a healthier colon (Likotrafiti et al., 2014). However, pre-
biotics alone have also been shown to improve the health and
alter the gut microbial composition of elderly populations. A
study providing inulin supplementation to an elderly cohort
increased Bifidobacterium levels (Guigoz et al., 2002). Multiple
studies using either fructo or GOSs demonstrated both bifi-
dogenic effects and beneficial immune-related effects. Specific
immune related effects included reduction in pro-inflammatory
cytokines and an increase in the anti-inflammatory cytokine,
IL-10.
While probiotic supplementation has become a widely utilized
tool to positively impact health by assisting with digestion, bol-
stering intestinal barrier function and coordinating with the body
to regulate both the innate and specific immune responses, the
mechanisms by which they exert these beneficial effects is poorly
understood (Siciliano and Mazzeo, 2012). Proteomic-based probi-
otic research is beginning to inform both researchers and industry
that adaptation and adherence properties specific to probiotic
strains influence their ability to colonize the host (Siciliano and
Mazzeo, 2012;van de Guchte etal., 2012). Additionally, these
adaptation and adherence mechanisms have been reported to
potentially be strain specific, making it difficult to globally apply
these mechanisms to all probiotic bacterial strains (Siciliano and
Mazzeo, 2012).
Experimentation on the effects of probiotics and prebiotics of
the elderly gut microbiome is still limited, but results of the avail-
able research lends merit to the notion that beneficial bacteria
in the form of probiotics and the indigestible fibers of prebi-
otics has potential to help restore stability, increase diversity and
beneficially alter the immune system in the aging gut (Vulevic
et al., 2008). However, these beneficial effects must be placed in
perspective given the lack of a mutually agreed upon selection cri-
teria, evaluation methodologies and a clear mechanistic model.
With the reduced cost of sequencing and continued proteomic
research, hopefully researchers will be able to speak with increased
certainty as to the reasons probiotics can be beneficial to human
host.
CONCLUSION
The microbes that reside in our gastrointestinal tract comprise a
dynamic community that changes throughout the lifespan of an
individual. The early years of infancy and childhood are character-
ized by a microbial state that has been described as chaotic because
of the rapid and dramatic fluctuations observed. While the micro-
biota of small children begins to resemble that of adults at a very
early age, there is a paucity of studies examining temporal micro-
bial community shifts in children beyond infancy, so the stability
www.frontiersin.org September 2014 |Volume 5 |Article 494 |7
Voreades etal. Diet and the gut microbiome
of their microbiota is not known. Once stable dietary patterns are
established, the microbiota of adults remains relatively unaltered;
however, significant weight changes have been associated with a
higher amount of microbial instability. Finally, factors related to
aging, including increased use of pharmaceuticals and changes in
diet likely play an important role in shaping the microbial com-
munities residing in the elderly. Changes in physical activity and
hormone levels may also be important determinants of the elderly
microbiome, but they have not yet been investigated with sufficient
depth. Some evidence suggests that the microbial communities of
healthy elderly individuals are similar to that of younger adults,
but whether the health of the individual contributes to microbial
stability or vice versa is not known. Current data suggest that diet
is an important driver in the development of the gut microbiome
and could serve as a means of therapeutic intervention for pre-
vention of diseases. Studies linking the composition and function
of the gut microbiome and disease development certainly high-
light the need for a better understanding of temporal microbiome
dynamics and their predictors.
ACKNOWLEDGMENTS
The authors would like to acknowledge support from NIH
R21CA161472, the Colorado Agricultural Experiment Station,
and Colorado State University Libraries Open Access Research
and Scholarship Fund.
REFERENCES
Aagaard, K., Ma, J., Antony, K. M., Ganu, R., Petrosino, J., and Versalovic, J. (2014).
The placenta harbors a unique microbiome. Sci. Transl. Med. 6, 237ra265. doi:
10.1126/scitranslmed.3008599
Arumugam, M., Raes, J., Pelletier, E., Le Paslier, D., Yamada, T., Mende, D. R., etal.
(2011). Enterotypes of the human gut microbiome. Natu re 473, 174–180. doi:
10.1038/nature09944
Bartosch, S., Fite, A., Macfarlane, G. T., and McMurdo, M. E. (2004). Charac-
terization of bacterial communities in feces from healthy elderly volunteers and
hospitalized elderly patients by using real-time PCR and effects of antibiotic
treatment on the fecal microbiota. Appl. Environ. Microbiol. 70, 3575–3581. doi:
10.1128/AEM.70.6.3575-3581.2004
Bergström, A., Skov, T. H., Bahl, M. I., Roager, H. M., Christensen, L. B., Ejlerskov,
K. T., etal. (2014). Establishment of intestinal microbiota during early life: a
longitudinal, explorative study of a large cohort of Danish infants. Appl. Environ.
Microbiol. 80, 2889–2900. doi: 10.1128/AEM.00342-14
Bezirtzoglou, E., Tsiotsias,A., and Welling, G. W. (2011). Microbiota profile in feces
of breast-and formula-fed newborns by using fluorescence in situ hybridization
(FISH). Anaerobe 17, 478–482. doi: 10.1016/j.anaerobe.2011.03.009
Britton, E., and McLaughlin, J. T. (2013). Ageing and the gut. Proc. Nutr. Soc. 72,
173–177. doi: 10.1017/S0029665112002807
Cabrera-Rubio, R., Collado, M. C., Laitinen, K., Salminen, S., Isolauri, E., and Mira,
A. (2012). The human milk microbiome changes over lactation and is shaped
by maternal weight and mode of delivery. Am. J. Clin. Nutr. 96, 544–551. doi:
10.3945/ajcn.112.037382
Claesson, M. J., Cusack, S., O’Sullivan, O., Greene-Diniz, R., de Weerd, H., Flannery,
E., et al. (2011). Composition, variability, and temporal stability of the intestinal
microbiota of the elderly. Proc. Natl. Acad. Sci. U.S.A. 108(Suppl. 1), 4586–4591.
doi: 10.1073/pnas.1000097107
Claesson, M. J., Jeffery, I. B., Conde, S., Power, S. E., O’Connor, E. M., Cusack, S.,
et al. (2012). Gut microbiota composition correlates with diet and health in the
elderly. Nature 488, 178–184. doi: 10.1038/nature11319
Cotillard, A., Kennedy, S. P., Kong, L. C., Prifti, E., Pons, N., Le Chatelier, E., et al.
(2013). Dietary intervention impact on gut microbial gene richness. Nature 500,
585–588. doi: 10.1038/nature12480
David, L. A., Maurice, C. F., Carmody, R. N., Gootenberg, D. B., But-
ton, J. E., Wolfe, B. E., et al. (2014). Diet rapidly and reproducibly alters
the human gut microbiome. Nature 505, 559–563. doi: 10.1038/nature
12820
De Filippo, C., Cavalieri, D., Di Paola, M., Ramazzotti, M., Poullet, J. B., Massart, S.,
et al. (2010). Impact of diet in shaping gut microbiota revealed by a comparative
study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. U.S.A. 107,
14691–14696. doi: 10.1073/pnas.1005963107
Dethlefsen, L., and Relman, D. A. (2011). Incomplete recovery and individualized
responses of the human distal gut microbiota to repeated antibiotic perturbation.
Proc. Natl. Acad. Sci. U.S.A. 108, 4554–4561. doi: 10.1073/pnas.1000087107
Duncan, S. H., and Flint, H. J. (2013). Probioticsand prebiotics and health in ageing
populations. Maturitas 75, 44–50. doi: 10.1016/j.maturitas.2013.02.004
Duncan, S. H., Lobley, G. E., Holtrop, G., Ince, J., Johnstone, A. M., Louis, P., etal.
(2008). Human colonic microbiota associated with diet, obesity and weight loss.
Int. J. Obes. 32, 1720–1724. doi: 10.1038/ijo.2008.155
Eckburg, P. B., Bik, E. M., Bernstein, C. N., Purdom, E., Dethlefsen, L., Sargent,
M., et al. (2005). Diversity of the human intestinal microbial flora. Science 308,
1635–1638. doi: 10.1126/science.1110591
Faith, J. J., Guruge, J. L., Charbonneau, M., Subramanian, S., Seedorf, H., Goodman,
A. L., et al. (2013). The long-term stability of the human gut microbiota. Science
341, 44–52. doi: 10.1126/science.1237439
Fallani, M., Amarri, S., Uusijarvi, A., Adam, R., Khanna, S., Aguilera, M., etal.
(2011). Determinants of the human infant intestinal microbiota after the intro-
duction of first complementary foods in infant samples from five European
centres. Microbiology 157, 1385–1392. doi: 10.1099/mic.0.042143-0
Fallani, M., Young, D., Scott, J., Norin, E., Amarri, S., Adam, R., etal. (2010).
Intestinal microbiota of 6-week-old infants across Europe: geographic influence
beyond delivery mode, breast-feeding, and antibiotics. J. Pediatr. Gastroenterol.
Nutr. 51, 77–84. doi: 10.1097/MPG.0b013e3181d1b11e
Guigoz, Y., Rochat, F., Perruisseau-Carrier, G., Rochat, I., and Schiffrin, E. J. (2002).
Effects of oligosaccharide on the faecal flora and non-specific immune system in
elderly people. Nutr. Res. 22, 13–25. doi: 10.1016/S0271-5317(01)00354-2
Harmsen, H. J., Wildeboer-Veloo, A. C., Raangs, G. C., Wagendorp, A. A., Klijn, N.,
Bindels, J. G., etal. (2000). Analysis of intestinal flora development in breast-fed
and formula-fed infants by using molecular identification and detection methods.
J. Pediatr. Gastroenterol. Nutr. 30, 61–67. doi: 10.1097/00005176-200001000-
00019
Huse, S. M., Ye, Y., Zhou, Y., and Fodor, A. A. (2012). A core human micro-
biome as viewed through 16S rRNA sequence clusters. PLoS ONE 7:e34242. doi:
10.1371/journal.pone.0034242
Jost, T., Lacroix, C., Braegger, C. P., and Chassard, C. (2012). New insights in gut
microbiota establishment in healthy breast fed neonates. PLoS ONE 7:e44595.
doi: 10.1371/journal.pone.0044595
Kelly, D., King, T., and Aminov, R. (2007). Importance of microbial colonization of
the gut in early life to the development of immunity. Mutat. Res. 622, 58–69. doi:
10.1016/j.mrfmmm.2007.03.011
Knol, J., Boehm, G., Lidestri, M., Negretti, F., Jelinek, J., Agosti, M., et al. (2005).
Increase of faecal bifidobacteria due to dietary oligosaccharides induces a reduc-
tion of clinically relevant pathogen germs in the faeces of formula-fed preterm
infants. Acta Paediatr. Suppl. 94, 31–33. doi: 10.1080/08035320510043529
Koenig, J. E., Spor, A., Scalfone, N., Fricker, A. D., Stombaugh, J., Knight, R.,
et al. (2011). Succession of microbial consortia in the developing infant gut
microbiome. Proc. Natl. Acad. Sci. U.S.A. 108(Suppl. 1), 4578–4585. doi:
10.1073/pnas.1000081107
Koren, O., Knights, D., Gonzalez, A., Waldron, L., Segata, N., Knight, R., et al.
(2013). A guide to enterotypes across the human body: meta-analysis of micro-
bial community structures in human microbiome datasets. PLoS Comput. Biol.
9:e1002863. doi: 10.1371/journal.pcbi.1002863
Le Chatelier, E., Nielsen, T., Qin, J.,Prifti, E., Hildebrand, F., Falony, G., et al. (2013).
Richness of human gut microbiome correlates with metabolic markers. Nature
500, 541–546. doi: 10.1038/nature12506
Ley, R. E., Turnbaugh, P. J., Klein, S., and Gordon, J. I. (2006). Microbial ecol-
ogy: human gut microbes associated with obesity. Nature 444, 1022–1023. doi:
10.1038/4441022a
Likotrafiti, E., Tuohy, K. M., Gibson, G. R., and Rastall, R. A. (2014). An in vitro
study of the effect of probiotics, prebiotics and synbiotics on the elderly faecal
microbiota. Anaerobe 27, 50–55. doi: 10.1016/j.anaerobe.2014.03.009
Lin, A., Bik, E. M., Costello, E. K., Dethlefsen, L., Haque, R., Relman, D. A.,
et al. (2013). Distinct distal gut microbiome diversity and composition in healthy
Frontiers in Microbiology |Evolutionary and Genomic Microbiology September 2014 |Volume 5 |Article 494 |8
Voreades etal. Diet and the gut microbiome
children from Bangladesh and the United States. PLoS ONE 8:e53838. doi:
10.1371/journal.pone.0053838
Ma, J., Prince, A. L., Bader, D., Hu, M., Ganu,R., Baquero, K., et al. (2014). High-fat
maternal diet during pregnancy persistently alters the offspring microbiome in a
primate model. Nat. Commun. 5, 3889. doi: 10.1038/ncomms4889
Macfarlane, S. (2014). Antibiotic treatments and microbes in the gut. Environ.
Microbiol. 16, 919–924. doi: 10.1111/1462-2920.12399
Marques, T. M., Wall, R., Ross, R. P., Fitzgerald, G. F., Ryan, C. A.,
and Stanton, C. (2010). Programming infant gut microbiota: influence of
dietary and environmental factors. Curr. Opin. Biotechnol. 21, 149–156. doi:
10.1016/j.copbio.2010.03.020
Muegge, B. D., Kuczynski, J., Knights, D., Clemente, J. C., González, A.,
Fontana, L., et al. (2011). Diet drives convergence in gut microbiome functions
across Mammalian phylogeny and within humans. Science 332, 970–974. doi:
10.1126/science.1198719
O’Connor, E. M., O’Herlihy, E. A., and O’Toole, P. W. (2014). Gut microbiota in
older subjects: variation, health consequences and dietary intervention prospects.
Proc. Nutr. Soc. 13, 1–11. doi: 10.1017/S0029665114000597
Okada, H., Kuhn, C., Feillet, H., and Bach, J. F. (2010). The “hygiene hypothesis” for
autoimmune and allergic diseases: an update. Clin. Exp. Immunol. 160, 1–9. doi:
10.1111/j.1365-2249.2010.04139.x
Oozeer, R., van Limpt, K., Ludwig, T., Ben Amor, K., Martin, R., Wind, R. D., etal.
(2013). Intestinal microbiology in early life: specific prebiotics can have similar
functionalities as human-milk oligosaccharides. Am. J. Clin. Nutr. 98, 561S–571S.
doi: 10.3945/ajcn.112.038893
Ou, J., Carbonero, F., Zoetendal, E. G., DeLany, J. P., Wang, M., Newton, K.,
et al. (2013). Diet, microbiota, and microbial metabolites in colon cancer risk
in rural Africans and African Americans. Am. J. Clin. Nutr. 98, 111–120. doi:
10.3945/ajcn.112.056689
Palmer, C., Bik, E. M., DiGiulio, D. B., Relman, D. A., and Brown, P. O. (2007).
Development of the human infant intestinal microbiota. PLoS Biol. 5:e177. doi:
10.1371/journal.pbio.0050177
Preidis, G. A., and Versalovic, J. (2009). Targeting the human microbiome with
antibiotics, probiotics, and prebiotics: gastroenterology enters the metagenomics
era. Gastroenterology 136, 2015–2031. doi: 10.1053/j.gastro.2009.01.072
Qato, D. M., Alexander, G., Conti, R. M., Johnson, M., Schumm, P., and Lindau, S.
(2008). Use of prescription and over-the-countermedications and dietar y supple-
ments among older adults in the united states. J. Am. Med. Assoc. 300, 2867–2878.
doi: 10.1001/jama.2008.892
Rajilic-Stojanovic, M., Heilig, H. G., Tims, S., Zoetendal, E. G., and de Vos, W. M.
(2013). Long-term monitoring of the human intestinal microbiota composition.
Environ. Microbiol. 15, 1146–1159. doi: 10.1111/1462-2920.12023
Roager, H. M., Licht, T. R., Poulsen, S. K., Larsen, T. M., and Bahl, M. I. (2014).
Microbial enterotypes, inferred by the prevotella-to-bacteroides ratio, remained
stable during a 6-month randomized controlled diet intervention with the new
nordic diet. Appl. Environ. Microbiol. 80, 1142–1149. doi: 10.1128/AEM.03549-13
Rook, G. W. (2012). Hygiene hypothesis and autoimmune diseases. Clin. Rev. Allergy
Immunol. 42, 5–15. doi: 10.1007/s12016-011-8285-8
Schnorr, S. L., Candela, M., Rampelli, S., Centanni, M., Consolandi, C., Basaglia, G.,
et al. (2014). Gut microbiome of the Hadza hunter-gatherers. Nat. Commun. 5,
3654. doi: 10.1038/ncomms4654
Sekirov,I., Russell, S. L.,Antunes, L. C. M., and Finlay, B. B. (2010). Gut microbiota
in health and disease. Physiol. Rev. 90, 859–904 doi: 10.1152/physrev.00045.2009
Siciliano, R. A., and Mazzeo, M. F. (2012). Molecular mechanisms of probi-
otic action: a proteomic perspective. Curr. Opin. Microbiol. 15, 390–396. doi:
10.1016/j.mib.2012.03.006
Subramanian, S., Huq, S., Yatsunenko, T., Haque, R., Mahfuz, M., Alam, M. A.,
et al. (2014). Persistent gut microbiota immaturity in malnourished Bangladeshi
children. Nature 510, 417–421. doi: 10.1038/nature13421
Tilg, H., and Kaser, A. (2011). Gut microbiome,obesit y, and metabolic dysfunction.
J. Clin. Invest. 121, 2126–2132. doi: 10.1172/JCI58109
Toward, R., Montandon,S., Walton, G., and Gibson, G. R. (2012). Effect of prebiotics
on the human gut microbiota of elderly persons. Gut Microbes 3, 57–60. doi:
10.4161/gmic.19411
Turroni, F., Peano, C., Pass, D. A., Foroni, E., Severgnini, M., Claesson, M. J., etal.
(2012). Diversity of bifidobacteria within the infant gut microbiota. PLoS ONE
7:e36957. doi: 10.1371/journal.pone.0036957
van de Guchte, M., Chaze, T., Jan, G., and Mistou, M.-Y. (2012). Properties of
probiotic bacteria explored by proteomic approaches. Curr. Opin. Microbiol. 15,
381–389. doi: 10.1016/j.mib.2012.04.003
van Tongeren, S. P., Slaets, J. P., Harmsen, H. J., and Welling, G. W. (2005). Fecal
microbiota composition and frailty. Appl. Environ. Microbiol. 71, 6438–6442. doi:
10.1128/AEM.71.10.6438-6442.2005
Vulevic, J., Drakoularakou, A., Yaqoob, P., Tzortzis, G., and Gibson, G. R. (2008).
Modulation of the fecal microflora profile and immune function by a novel trans-
galactooligosaccharide mixture (B-GOS) in healthyelderly volunteers. Am. J. Clin.
Nutr. 88, 1438–1446.
Woodmansey, E. J. (2007). Intestinal bacteria and ageing. J. Appl. Microbiol. 102,
1178–1186. doi: 10.1111/j.1365-2672.2007.03400.x
Wu, G. D., Chen, J., Hoffmann, C., Bittinger, K., Chen, Y. Y., Keilbaugh, S. A.,
et al. (2011). Linking long-term dietary patterns with gut microbial enterotypes.
Science 334, 105–108. doi: 10.1126/science.1208344
Yatsunenko, T., Rey, F. E., Manary, M. J., Trehan, I., Dominguez-Bello, M. G.,
Contreras, M., et al. (2012). Human gut microbiome viewed across age and
geography. Nature 486, 222–227. doi: 10.1038/nature11053
Conflict of Interest Statement: The authors declare that the research was conducted
in the absence of any commercial or financial relationships that could be construed
as a potential conflict of interest.
Received: 02 July 2014; accepted: 02 September 2014; published online: 22 September
2014.
Citation: Voreades N, Kozil A and Weir TL (2014) Diet and the development of the
human intestinal microbiome. Front. Microbiol. 5:494. doi: 10.3389/fmicb.2014.00494
This article was submitted to Evolutionary and Genomic Microbiology, a section of the
journal Frontiers in Microbiology.
Copyright © 2014 Voreades, Kozil and Weir. This is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). The use, dis-
tribution or reproduction in other forums is permitted, provided the original author(s)
or licensor are credited and that the original publication in this journal is cited, in
accordance with accepted academic practice. No use, distribution or reproduction is
permitted which does not comply with these terms.
www.frontiersin.org September 2014 |Volume 5 |Article 494 |9
... This suggests that some amplified genetic material from EVs may originate from ingested, breathed or, more generally, exogenously introduced species with subsequent transport through the bloodstream as part of the EV cargo or the widely reported EV-corona [46]. This finding highlights the potential role of diet and, more generally, the environment, in influencing the microbiome in MS, given the established link between diet and gut microbiota composition [47,48,49]. ...
Preprint
Full-text available
Motivation: The taxonomical characterisation of bacterial species derived from genetic material blood, including reads derived from bacterial extracellular vesicles (bEVs) poses certain challenges, such as the proper discrimination of "true" reads from contaminating reads. This is a common issue in taxa profiling and can lead to the false discovery of species that are present in the sample. To avoid such biases a careful approximation to taxa profiling is necessary. Results: In this work we propose a workflow to analyze the presence of bacterial transcripts as indicative of putative bEVs circulating in the blood of people with MS (pwMS). The workflow includes several reference mapping steps against the host genome and a consensus selection of genera based on different taxa profilers. The consensus selection is performed with a flagging system that removes species with low abundance or with high variation across profilers. Additionally, the inclusion of biological samples from known cultured species as well as the generation of artificial reads constitute two key aspects of this workflow. Availability: The workflow is available at the following repository: https://github.com/NanoNeuro/EV_taxprofiling
... Additionally, cases and controls were not formally matched on possible confounders such as age, sex, and BMI, though these factors did not significantly differ between the groups at baseline, so possibly have not influenced the outcome. Moreover, we have not accounted for the potential influence of diet and other environmental factors other than medication use on faecal VOC composition, possibly affecting the overall outcome [36,37]. Since subjects from both groups derive from the same regions in The Netherlands, it can be speculated that there are no differences in dietary habits and other environmental factors, limiting the risk of type I error. ...
Article
Full-text available
The gut microbiota and its related metabolites differ between inflammatory bowel disease (IBD) patients and healthy controls. In this study, we compared faecal volatile organic compound (VOC) patterns of paediatric IBD patients and controls with gastrointestinal symptoms (CGIs). Additionally, we aimed to assess if baseline VOC profiles could predict treatment response in paediatric IBD patients. We collected faecal samples from a cohort of de novo therapy-naïve paediatric IBD patients and CGIs. VOCs were analysed using gas chromatography–ion mobility spectrometry (GC-IMS). Response was defined as a combination of clinical response based on disease activity scores, without requiring treatment escalation. We included 109 paediatric IBD patients and 75 CGIs, aged 4 to 17 years. Faecal VOC profiles of paediatric IBD patients were distinguishable from those of CGIs (AUC ± 95% CI, p-values: 0.71 (0.64–0.79), <0.001). This discrimination was observed in both Crohn’s disease (CD) (0.75 (0.67–0.84), <0.001) and ulcerative colitis (UC) (0.67 (0.56–0.78), 0.01) patients. VOC profiles between CD and UC patients were not distinguishable (0.57 (0.45–0.69), 0.87). Baseline VOC profiles of responders did not differ from non-responders (0.70 (0.58–0.83), 0.1). In conclusion, faecal VOC profiles of paediatric IBD patients differ significantly from those of CGIs.
... Just as each of us are indelibly linked to the trillions of bacteria that live out their days in our intestines and on our bodies, these communities link us to the environments outside of our skins; a significant amount of the microbiota is acquired from the environment during and after birth, and remains in dynamic contact with environmental biota throughout life. Our microbiota is dynamic, and is affected by the places we live and the food we eat (David et al., 2014;Voreades et al., 2014) forming an interface between human physiology and the environment, interpreting and responding to signals from each. ...
Article
Full-text available
Human-microbe relations have undergone a profound shift over the past 100 years. The discovery of antibiotics, increasing levels of pollution, and urban and agricultural intensification have led to the proliferation and diversification of novel resistance genes and microorganisms. This abundance has unfolded against a backdrop of microbial absence that is the other side of the antimicrobial coin; reductions in the quantity and diversity of human-microbe interactions are now registering as epidemics of chronic non-communicable diseases in urban populations. Building from this paradoxical situation of ‘abundance’ and ‘absence’, this article reviews the molecular-genetic, macroscale-infrastructural, and community-ecological aspects of microbial evolution at a time when human actions are a critical force in shaping their directions.
... The intestinal microbiome is crucial for human health and is substantially influenced by diet [46]. Similarly, in fish, intestinal microbiota plays an important role in nutrition, immunity, and resistance to invading pathogens, and diet greatly influences its composition [47]. ...
Article
Full-text available
DL-methionyl–DL-methionine (AQUAVI® Met-Met) (Met-Met) (0.10%, 0.20%, 0.30%, and 0.40%) or DL-methionine (DL-Met) (0.10%, 0.20%, 0.30%, and 0.40%) were added to a low-fishmeal diet in an attempt to reduce fishmeal in the diet of Micropterus salmoides (M. salmoides). The fish were randomly allocated into ten experimental groups (n = 100), each with 4 replicates of 25 fish (16.39 ± 0.01 g) each. Compared to 25% FM, 0.40% of DL-Met and 0.10% of Met-Met promoted growth, and 0.10% of Met-Met decreased FCR. Compared to 25% FM, the supplementation of Met-Met or DL-Met improved the intestinal antioxidant capacity by upregulating the NF-E2-related factor 2-mediated antioxidant factors and enzyme activities and nuclear factor kappa-B-mediated anti-inflammatory factors while downregulating the pro-inflammatory factors, thereby exerting anti-inflammatory effects. Moreover, 0.10% of the Met-Met diet affected the Firmicutes-to-Bacteroidota ratio, increased the levels of Proteobacteria, changed the composition of intestinal flora (Roseburia, Lachnospiraceae_NK4A136_group, and unclassified_Oscillospiraceae), and enhanced intestinal dominant bacteria (Caldicoprobacter, Pseudogracilibacillus, and Parasutterella), leading to improved gut health. In summary, the supplementation of DL-Met or Met-Met alleviated the adverse effect of fishmeal reduction (from 40 to 25%) on the growth performance and intestinal health of M. salmoides.
... These conditions may significantly impact the well-being of both infants and their caregivers. A number of studies have shown that both the composition and diversity of the intestinal microbiota play important roles in the development and function of the gastrointestinal tract, influencing various aspects of health [3][4][5][6][7][8][9][10][11][12][13][14][15][16][17]. ...
Article
Full-text available
Infantile functional gastrointestinal disorders, such as colic, constipation, diarrhea, and gastroesophageal reflux (regurgitation), often occur in early infancy and, representing one of the causes of significant parental anxiety, lead to a significant strain on the healthcare resources. In this study, we aimed to evaluate the effects of Lactobacillus reuteri drops (L. reuteri NCIMB 30351) on the symptoms of infantile colic, constipation, diarrhea, and gastroesophageal reflux, as well as on the levels of intestinal microbiota in full-term newborns during the first months of life. A randomized, placebo-controlled, single-masked (blinded), post-marketing clinical study was conducted in two clinical units—Children’s City Clinical Hospital of Moscow and Medical Center “St. Andrew’s Hospitals-NEBOLIT” from March 2020 to May 2022 in 90 infants aged from 1 to 4 months (mean age (± SD) 12.3 ± 5.09 weeks; 53.3% females, 46.7% males). Patients with colic, regurgitation (single symptom or combination of several symptoms), and constipation or diarrhea were randomly allocated in two parallel arms to receive either 5 drops (2 × 10⁸ colony forming unit) of L. reuteri NCIMB 30351 (n = 60) or masked placebo (n = 30) for 25 consecutive days. Two treatment arms had equal numbers of patients with constipation and diarrhea (n = 30 each). Daily crying times and their duration, evacuations, and regurgitations were recorded in a structured diary. The levels of gut microbiota were analyzed by deep sequencing of bacterial 16S rRNA gene. Infants with colic receiving supplementary L. reuteri NCIMB 30351 for 25 days had significant reduction in the numbers of colic (change from baseline − 6.3 (7.34) vs − 3.0 (7.29) in placebo, P < 0.05) and numbers of crying cases and mean duration of crying (decrease from baseline − 144 (70.7) minutes, lower in the diarrhea subgroup than in constipation infants, compared with − 80 (58.9) in placebo, P < 0.0001), as well as regurgitation numbers (decreased by − 4.8 (2.49) with L. reuteri vs − 3 (7.74) with placebo). We also observed increased numbers of evacuations in infants with constipation (L. reuteri 2.2 (2.4) vs 0.9 (1.06) in placebo, P < 0.05). There was a remarkable reduction of evacuations in infants with diarrhea, while not statistically significant. The analysis of bacterial 16S rRNA gene in the collected samples showed that L. reuteri positively influences the proportions of prevalent species, while it negatively affects both conditionally pathogenic and commensal microbes. Additional in vitro test for formation of Clostridium colonies in the presence of the probiotic demonstrated that L. reuteri effectively inhibits the growth of pathogenic Clostridium species. No adverse events were reported in this study. Conclusion: The uptake of L. reuteri NCIMB 30351 leads to a significant reduction in the number of regurgitations, feeding-induced constipations, and diarrhea as well as mean daily numbers of crying and crying duration in infants during the first months of life. Our results suggest that L. reuteri NCIMB 30351 represents a safe and effective treatment for colic in newborns. Trial registration: ClinicalTrials.gov: NCT04262648. What is Known: • Infantile functional gastrointestinal disorders, such as colic, constipation, diarrhea, and gastroesophageal reflux (regurgitation), often occur in early infancy and, represent one of the causes of significant parental anxiety. • A number of studies have shown that both the composition and diversity of the intestinal microbiota play important roles in the development and function of the gastrointestinal tract. What is New: • The uptake of L. reuteri NCIMB 30351 leads to a significant reduction in the number of regurgitations, feeding-induced constipations, and diarrhea as well as mean daily numbers of crying and crying duration in infants during the first months of life. • L. reuteri positively influences the proportions of prevalent species, while it negatively affects both conditionally pathogenic and commensal microbes in gut microbiota.
... The gut microbiota partially regulates the immune function of L. vannamei (Li et al., 2018). Moreover, nutrient availability influence the diversity of the gut microbiota (Voreades et al., 2014). The gut microbiota of L. vannamei was composed of five main phyla: Proteobacteria, Firmicutes, Bacteroidetes, Acidobacteria, and Actinobacillus. ...
Article
Full-text available
Non-communicable diseases (NCDs) pose a global health challenge, leading to substantial morbidity, mortality, and economic strain. Our review underscores the escalating incidence of NCDs worldwide and highlights the potential of regenerative agriculture (RA) products in mitigating these diseases. We also explore the efficacy of dietary interventions in NCD management and prevention, emphasizing the superiority of plant-based diets over those high in processed foods and red meat. Examining the role of the gut microbiome in various diseases, including liver disorders, allergies, metabolic syndrome, inflammatory bowel disease, and colon cancer, we find compelling evidence implicating its influence on disease development. Notably, dietary modifications can positively affect the gut microbiome, fostering a symbiotic relationship with the host and making this a critical strategy in disease prevention and treatment. Investigating agricultural practices, we identify parallels between soil/plant and human microbiome studies, suggesting a crucial link between soil health, plant- and animal-derived food quality, and human well-being. Conventional/Industrial agriculture (IA) practices, characterized in part by use of chemical inputs, have adverse effects on soil microbiome diversity, food quality, and ecosystems. In contrast, RA prioritizes soil health through natural processes, and includes avoiding synthetic inputs, crop rotation, and integrating livestock. Emerging evidence suggests that food from RA systems surpasses IA-produced food in quality and nutritional value. Recognizing the interconnection between human, plant, and soil microbiomes, promoting RA-produced foods emerges as a strategy to improve human health and environmental sustainability. By mitigating climate change impacts through carbon sequestration and water cycling, RA offers dual benefits for human and planetary health and well-being. Emphasizing the pivotal role of diet and agricultural practices in combating NCDs and addressing environmental concerns, the adoption of regional RA systems becomes imperative. Increasing RA integration into local food systems can enhance food quality, availability, and affordability while safeguarding human health and the planet's future.
Article
The peculiarities of the influence of cytokines and metabolites of the systemic inflammatory reaction, stress-implementing and nutritional factors contributing to the transformation of the phenotype of the resident intestinal microflora with an increase in its virulence are described. From the perspective of gene expression, protein and phospholipids conformations, the influence of temperature as a signaling factor in increasing the virulence of the intestinal microbiome is considered. Evolutionarily formed mechanisms of expression of the maximum pathogenic phenotype of microorganisms and, thus, achieving an increase in their biomass and maximum dissemination through the microorganism compartments increases the probability of the commensals transmission to another biotope, i.e. increases the probability of their survival after the death of the host organism. To prevent bacterial translocation after the relief of critical conditions, early enteral administration of β-glucans in food mixtures, iron excretion, relief of inorganic phosphate deficiency, including by induction of alkaline phosphatase synthesis.
Article
Full-text available
The gut microbiota (GM), as a forgotten organ, refers to the microbial community that resides in the gastrointestinal tract and plays a critical role in a variety of physiological activities in different body organs. The GM affects its targets through neurological, metabolic, immune, and endocrine pathways. The GM is a dynamic system for which exogenous and endogenous factors have negative or positive effects on its density and composition. Since the mid-twentieth century, laboratory animals are known as the major tools for preclinical research; however, each model has its own limitations. So far, two main models have been used to explore the effects of the GM under normal and abnormal conditions: the isolated germ-free and antibiotic-treated models. Both methods have strengths and weaknesses. In many fields of host-microbe interactions, research on these animal models are known as appropriate experimental subjects that enable investigators to directly assess the role of the microbiota on all features of physiology. These animal models present biological model systems to either study outcomes of the absence of microbes, or to verify the effects of colonization with specific and known microbial species. This paper reviews these current approaches and gives advantages and disadvantages of both models.
Article
Full-text available
Complex gene-environment interactions are considered important in the development of obesity(1). The composition of the gut microbiota can determine the efficacy of energy harvest from food(2-4) and changes in dietary composition have been associated with changes in the composition of gut microbial populations(5,6). The capacity to explore microbiota composition was markedly improved by the development of metagenomic approaches(7,8), which have already allowed production of the first human gut microbial gene catalogue(9) and stratifying individuals by their gut genomic profile into different enterotypes(10), but the analyses were carried out mainly in nonintervention settings. To investigate the temporal relationships between food intake, gut microbiota and metabolic and inflammatory phenotypes, we conducted diet-induced weight-loss and weight-stabilization interventions in a study sample of 38 obese and 11 overweight individuals. Here we report that individuals with reduced microbial gene richness (40%) present more pronounced dys-metabolism and low-grade inflammation, as observed concomitantly in the accompanying paper(11). Dietary intervention improves low gene richness and clinical phenotypes, but seems to be less efficient for inflammation variables in individuals with lower gene richness. Low gene richness may therefore have predictive potential for the efficacy of intervention.
Article
Full-text available
Therapeutic food interventions have reduced mortality in children with severe acute malnutrition (SAM) but incomplete restoration of healthy growth remains a major problem1,2. The relationships between the type of nutritional intervention, the gut microbiota, and therapeutic responses are unclear. In the current study, bacterial species whose proportional representation define a healthy gut microbiota as it assembles during the first two postnatal years were identified by applying a machine-learning-based approach to 16S rRNA datasets generated from monthly fecal samples obtained from a birth-cohort of children, living in an urban slum of Dhaka, Bangladesh, who exhibited consistently healthy growth. These age-discriminatory bacterial species were incorporated into a model that computes a ‘relative microbiota maturity index’ and ‘microbiota-for-age Z-score’ that compare development (defined here as maturation) of a child’s fecal microbiota relative to healthy children of similar chronologic age. The model was applied to twins and triplets (to test for associations of these indices with genetic and environmental factors including diarrhea), children with SAM enrolled in a randomized trial of two food interventions, and children with moderate acute malnutrition. Our results indicate that SAM is associated with significant relative microbiota immaturity that is only partially ameliorated following two widely used nutritional interventions. Immaturity is also evident in less severe forms of malnutrition and correlates with anthropometric measurements. Microbiota maturity indices provide a microbial measure of human postnatal development, a way of classifying malnourished states, and a parameter for judging therapeutic efficacy. More prolonged interventions with existing or new therapeutic foods and/or addition of gut microbes may be needed to achieve enduring repair of gut microbiota immaturity in childhood malnutrition and improve clinical outcomes.
Article
Full-text available
Humans and their microbiomes have coevolved as a physiologic community composed of distinct body site niches with metabolic and antigenic diversity. The placental microbiome has not been robustly interrogated, despite recent demonstrations of intracellular bacteria with diverse metabolic and immune regulatory functions. A population-based cohort of placental specimens collected under sterile conditions from 320 subjects with extensive clinical data was established for comparative 16S ribosomal DNA-based and whole-genome shotgun (WGS) metagenomic studies. Identified taxa and their gene carriage patterns were compared to other human body site niches, including the oral, skin, airway (nasal), vaginal, and gut microbiomes from nonpregnant controls. We characterized a unique placental microbiome niche, composed of nonpathogenic commensal microbiota from the Firmicutes, Tenericutes, Proteobacteria, Bacteroidetes, and Fusobacteria phyla. In aggregate, the placental microbiome profiles were most akin (Bray-Curtis dissimilarity <0.3) to the human oral microbiome. 16S-based operational taxonomic unit analyses revealed associations of the placental microbiome with a remote history of antenatal infection (permutational multivariate analysis of variance, P = 0.006), such as urinary tract infection in the first trimester, as well as with preterm birth <37 weeks (P = 0.001).
Article
Full-text available
The intestinal microbiome is a unique ecosystem and an essential mediator of metabolism and obesity in mammals. However, studies investigating the impact of the diet on the establishment of the gut microbiome early in life are generally lacking, and most notably so in primate models. Here we report that a high-fat maternal or postnatal diet, but not obesity per se, structures the offspring's intestinal microbiome in Macaca fuscata (Japanese macaque). The resultant microbial dysbiosis is only partially corrected by a low-fat, control diet after weaning. Unexpectedly, early exposure to a high-fat diet diminished the abundance of non-pathogenic Campylobacter in the juvenile gut, suggesting a potential role for dietary fat in shaping commensal microbial communities in primates. Our data challenge the concept of an obesity-causing gut microbiome and rather provide evidence for a contribution of the maternal diet in establishing the microbiota, which in turn affects intestinal maintenance of metabolic health.
Article
Full-text available
Alterations in intestinal microbiota composition and function have been linked to conditions including functional gastrointestinal disorders, obesity and diabetes. The gut microbiome encodes metabolic capability in excess of that encoded by the human genome, and bacterially produced enzymes are important for releasing nutrients from complex dietary ingredients. Previous culture-based studies had indicated that the gut microbiota of older people was different from that of younger adults, but the detailed findings were contradictory. Small-scale studies had also shown that the microbiota composition could be altered by dietary intervention or supplementation. We showed that the core microbiota and aggregate composition in 161 seniors was distinct from that of younger persons. To further investigate the reasons for this variation, we analysed the microbiota composition of 178 elderly subjects for whom the dietary intake data were available. The data revealed distinct microbiota composition groups, which overlapped with distinct dietary patterns that were governed by where people lived: at home, in rehabilitation or in long-term residential care. These diet-microbiota separations correlated with cluster analysis of NMR-derived faecal metabolites and shotgun metagenomic data. Major separations in the microbiota correlated with selected clinical measurements. It should thus be possible to programme the microbiota to enrich bacterial species and activities that promote healthier ageing. A number of other studies have investigated the effect of certain dietary components and their ability to modulate the microbiota composition to promote health. This review will discuss dietary interventions conducted thus far, especially those in elderly populations and highlight their impact on the intestinal microbiota.