You are on page 1of 10

Review

The microbiota–gut–brain axis in obesity


Cristina Torres-Fuentes*, Harriët Schellekens*, Timothy G Dinan, John F Cryan

Changes in microbial diversity and composition are increasingly associated with several disease states including Lancet Gastroenterol Hepatol
obesity and behavioural disorders. Obesity-associated microbiota alter host energy harvesting, insulin resistance, 2017
inflammation, and fat deposition. Additionally, intestinal microbiota can regulate metabolism, adiposity, Published Online
August 24, 2017
homoeostasis, and energy balance as well as central appetite and food reward signalling, which together have
http://dx.doi.org/10.1016/
crucial roles in obesity. Moreover, some strains of bacteria and their metabolites might target the brain directly via S2468-1253(17)30147-4
vagal stimulation or indirectly through immune-neuroendocrine mechanisms. Therefore, the gut microbiota is *Joint first authors
becoming a target for new anti-obesity therapies. Further investigations are needed to elucidate the intricate APC Microbiome Institute,
gut-microbiota–host relationship and the potential of gut-microbiota-targeted strategies, such as dietary (C Torres-Fuentes PhD,
interventions and faecal microbiota transplantation, as promising metabolic therapies that help patients to maintain H Schellekens PhD,
a healthy weight throughout life. Prof T G Dinan MD,
Prof J F Cryan PhD), Department
of Psychiatry and
Introduction homoeostasis and the central appetite mechanism.10 Neurobehavioural Sciences
Hippocrates, the father of modern medicine, famously Thus, modulation of intestinal microbiota by dietary (Prof T G Dinan), and
said “all disease starts in the gut”. The past two decades interventions, including prebiotics, probiotics, or faecal Department of Anatomy and
Neuroscience (H Schellekens,
have seen an abundance of research supporting this microbiota transplantation (FMT), might provide Prof J F Cryan), University
concept. The human intestine hosts tens of trillions of potential novel anti-obesity strategies. Growing evidence College Cork, Cork, Ireland
microorganisms, including archaea, bacteria, viruses, suggests that the success of bariatric surgery is due to Correspondence to:
phages, fungi, protists, and nematodes, but is dominated its effects on the microbiota (panel 1; figure 1).11 These Prof John F Cryan, Department of
by bacteria from the phyla Firmicutes and Bacteriodetes. findings have important implications for society since Anatomy and Neuroscience,
University College Cork,
This gut microbiota ecosystem is established after birth obesity and its comorbidities (ie, type 2 diabetes, Cork T12 XF62, Ireland
following the transfer of maternal and environmental cardiovascular disease, and increased pro-inflammatory j.cryan@ucc.ie
bacteria, and continues to develop until adulthood.1 status), are serious and widespread health concerns
Despite being overlooked for years, the gut microbiota worldwide and urgently need to be addressed.14,15
is one of the largest components of our body, weighing
approximately 1–2 kg, and contains over 100 times more
genes than does the human genome.2 Commensal gut Panel 1: Bariatric surgery and the microbiome
bacteria have a crucial symbiotic relationship with the Bariatric surgery is an intervention indicated for severe obesity
human body throughout its evolution, protecting and and is the only treatment that leads to substantial and
supporting the structure of intestinal mucosa. Gut sustained weight loss. Different bariatric surgeries are
bacteria are therefore becoming increasingly recognised available, such as adjustable gastric banding, sleeve
as key regulators of host physiology and pathophysiology, gastrectomy, Roux-en-Y gastric bypass (RYGB), and
and undeniably have a role in health and disease.3 biliopancreatic diversion.11 Results from human and animal
Alterations in the composition of the human gut studies show that RYGB reduces adiposity, improves
microbiota occur in metabolic disorders such as obesity, hormonal (increased glucagon-like peptide-1 and peptide YY)
diabetes,3 and eating disorders, as well as stress-related and inflammatory status, and increases the pool of bile acids,
neuropsychiatric disorders including depression4 and as well as improving insulin sensitivity (figure 1).11 RYGB also
anxiety,5 which are also characterised by changes in affects the composition of the gut microbiota, leading to
eating behaviour. Moreover, gut microbiota regulate fat increased diversity.1 For example, although with variation
storage6 and can harvest energy from the diet.7 Several across studies, after bariatric surgery, patients and mice have
studies have found that intestinal microbiota also affect an increased abundance of Gammaproteobacteria and
inflammation, insulin, and glucose metabolism, as well Verrucomicrobia (Akkermansia) and a decreased abundance of
as hepatic lipid metabolism.8 Furthermore, gut bacteria Firmicutes.11,12 Moreover, faecal microbiota transplantation
can directly affect the CNS via modulation of endocrine from mice that had RYGB into germ-free mice resulted in
signalling pathways of the microbiota–gut–brain axis weight loss and decreased fat mass in the recipient animals,
such as, glucagon-like peptide-1 (GLP-1) and peptide YY potentially due to altered microbial production of short-chain
signalling, or activation of reward pathways.9 fatty acids, increased propionate levels, and decreased acetate
A healthy gut microbiota is crucial for proper levels.13 Therefore, bariatric surgery leads to specific changes in
metabolic function and homoeostasis, which sub­ the gut microbiota, causing changes in the composition of
stantially benefits the host in exchange for living and short-chain fatty acids and thus influencing host metabolism,
proliferating in the intestinal habitat. Alterations in the including gut hormone secretion and insulin sensitivity.
composition of the microbiota, especially early in These effects might be central to the success of RYGB in
life, might cause obesity and diabetes by substantially treating obesity, metabolic syndrome, and diabetes.11,12
modifying the host’s metabolism and affecting

www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4 1


Review

Nutrients
Roux-en-Y gastric bypass

↓Ghrelin
↓Appetite

↑Insulin sensitivity GPR43


Bile acids pool
↓Adiposity
↓Inflammation
↓Leptin
↑Proteobacteria
↑Verrucomicrobia (Akkermansia)
↑Diversity

↓Firmicutes/Bacteroidetes GPR41 GPR43


SCFAs
Butyrate
↓Acetate
↑Propionate

↓GLP-1/PYY/CCK

↑Satiety

GLP-1

Figure 1: Effects of Roux-en-Y gastric bypass surgery on the gut microbiota and its metabolic outcomes
SCFAs=short-chain fatty acids. GLP-1=glucagon-like peptide 1. PYY=peptide YY. CCK=cholecystokinin. GPR41=G-protein coupled receptor 41. GPR43=G-protein
coupled receptor 43.

In addition to metabolic changes, obesity is a disorder of which are indicative of an energy deficit.17 Conversely,
the brain with altered food intake behaviour. Research is specific alterations in gut microbial composition have
focusing on whether the microbiome can affect brain been linked with obesity; a wider variety of Bacteroidetes,
processes and modify the homoeostatic and hedonic which break down plant starches and fibres for energy,
control of food intake.16 We review the gut microbiota as a was found in individuals with normal body-mass index
key regulator of host metabolism, central appetite, and than in obese individuals, in whom an increase in the
food reward, and its role in metabolic disorders such as abundance of Firmicutes was found.18 Despite large
obesity, summarise the literature on potential mechanisms variations in composition among individuals, changes in
of gut–brain axis signalling, review how some bacterial an overall core human microbiome were associated
strains might contribute to, or protect against, metabolic with obese individuals.19 The obese phenotype was
disease, and address how FMT, bariatric surgery, and transmittable via intestinal microbiota alone in germ-free
dietary interventions might be novel metabolic therapies mice20 and human beings21 and was reversed in germ-free
in clinical practice in obesity management. mice following co-housing with mice transplanted
with the lean microbiota.21 These findings show the
Obesity-associated microbiota transmissible, rapid, and modifiable nature of inter­
One of the earliest key findings implicating a role for actions between diet and the microbiota in obesity and
intestinal microbiota in energy balance originated from metabolic syndrome.
germ-free mice, which completely lack intestinal More recently the link between the microbiota and
microbiota from birth. These mice were protected against obesity was tested in a meta-analysis in which datasets
obesity and were significantly leaner than were control from ten previous studies were pooled using a random-
mice, despite consuming more calories.6 Furthermore, effect model; no significant associations were observed
these germ-free mice had altered plasma lipid metabolic for the ratio of Bacteroidetes to Firmicutes or their
markers and lower concentrations of ghrelin and leptin, individual relative abundance.22 The researchers showed

2 www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4


Review

Circulation

Hypothalamus
NTS
Satiety ARC
Vagal afferent B cells
NPY/AgRP CART/ Bacterial neuroactives Gut peptides Cytokines
GABA POMC nerve
(GLP-1, PYY, CCK)

Adipose tissue Enteric Enteric nervous


muscles system neuron
↑Leptin GLP-1
↑Insulin sensitivity DC
↓Ghrelin EEC
Pancreas
FXR
SCFAs
FFAR

Taste receptor Toll-like TGR5


GLP-1 LPS receptor
Conjugated
Metabolites bile acids
SCFAs, GABA, 5-HT, NTs
Bile acids
Nutrients Microbiota

Figure 2: Microbiota-driven mechanisms of metabolism and appetite regulation


Intestinal microorganisms convert dietary nutrients into metabolites such as short-chain fatty acids (SCFAs), γ-aminobutyric acid (GABA), serotonin (5-HT),
and other neurotransmitters (NTs), which have different peripheral and central effects modifying host metabolism and central regulation of appetite directly via
vagal stimulation or indirectly through immune–neuroendocrine mechanisms. Enteroendocrine cells (EECs) are activated by these microbial-derived metabolites via
activation of different receptors (eg, fatty acid receptors [FFARs] and taste receptors), leading to the production of gut hormones such as glucagon-like peptide-1
(GLP-1), peptide YY (PYY), and cholecystokinin (CCK). These gut hormones signal from the gut to the nucleus tractus solitarius (NTS) in the brain via the vagus nerve
and direct secretion into the circulatory system. Information from the NTS is distributed to the arcuate nucleus (ARC) in the hypothalamus, where appetite and
energy balance is regulated. The ARC contains neuropeptide Y (NPY), agouti-related protein (AgRP), anorexigenic peptides, cocaine amphetamine-regulated
transcript (CART), and pro-opiomelanocortin (POMC) neurons. Moreover, gut microorganisms might also use bile acids and their conjugates, activating farnesoid X
receptor (FXR) and Takeda G-protein-coupled receptor 5 (TGR5) and increasing GLP-1 secretion by EECs. GLP-1 in the gut is also essential for glucose tolerance and
insulin sensitivity. Moreover, microbial-derived metabolites might also lead to other peripheral effects such as increased leptin production by adipose tissue or
decreased ghrelin production in the stomach. Additionally, gut microbiota has been associated with inflammation via release of lipopolysaccharide (LPS), which leads
to activation of immune cells, such as B cells or dendritic cells (DCs), and cytokine production.

that, after a power analysis, most studies have a large altered the microbiome of recipient mice.24 More recently,
interpersonal variation and insufficient sample sizes, a metagenome-wide association study found a decreased
and do not have sufficient power to detect a 5% difference abundance of a glutamate-fermenting commensal bacteria
in the diversity of microbiota. in obese individuals, which was inversely correlated with
Complex interactions between environmental factors, serum glutamate concentration. This abundance was
host genetics, and the gut microbiota play an increased after bariatric surgery, which highlights further
essential part in the pathophysiology of obesity and unknown links between intestinal microbiota alterations,
diabetes. Alterations in the gut microbiota following circulating amino-acids, and obesity.25
environmental reprogramming ameliorates the develop-
ment of metabolic syndrome in different strains Metabolism and appetite regulation
of mice (obesity/diabetes-prone C57Bl/6J, obesity/ Although the gut microbiota is a contributing and
diabetes-resistant 129S1/SvImJ, and obesity-prone but potential causal factor for obesity and metabolic
diabetes-resistant 129S6/SvEvTac mice).23 Moreover, syndrome, the exact mechanisms underlying this
a twin study24 found that host genetics affect the relationship are unclear. Nevertheless, the intestinal
composition of the gut microbiota as well as host microbiota produce bioactive metabolites in a
metabolism; monozygotic twins had a more similar gut diet-dependent manner, including short-chain fatty
microbiota composition than did dizygotic twins and gut acids and conjugated fatty acids.26 These microbiota-
microbiota were enriched in particular taxa including derived metabolites have peripheral effects but also
Christensenellaceae minuta. Furthermore, an obesity- modulate the brain via direct or indirect mechanisms,
associated microbiome enriched with C minuta and which modifies host metabolism and the central
transplanted to germ-free mice reduced weight gain and regulation of appetite and food intake (figure 2).9,16

www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4 3


Review

Peripheral metabolic signalling The gut microbiota is also associated with inflammation
Intestinal microbiota can affect peripheral metabolic in obesity. Increased plasma levels of lipopolysaccharide,
function. Metagenomics and biochemical analysis in an endotoxin in the cell wall of Gram-negative
genetically obese mice (ob/ob, leptin-deficient mice) bacteria, causes metabolic endotoxaemia, inducing a
revealed that an obesity-associated microbiota increases strong immune system response and contributing to
the efficiency of calorie uptake from ingested foods and obesity-related low-grade inflammation.40 Dietary fat is
affects energy balance by influencing energy use crucial in this process because it inc­reases intestinal
and storage.7 Thus, an obesity-associated microbiota lipopolysaccharide absorption through in­corpor­ation
provides more energy to the host from otherwise into chylomicrons. Moreover, impaired intestinal barrier
indigestible carbohydrates and proteins than does a integrity might also contribute to this metabolic
lean-associated gut microbiota, via increased production endotoxaemia.40 However, some gut bacteria could
of different primary fermentation enzymes and nutrient prevent this endotoxaemia by protecting intestinal
transporters.27 However, the proportions of Firmicutes barrier integrity via a different mechanism, which leads
and Bacteroidetes are unrelated to markers of energy to thickening of the mucus layer or upregulating tight
harvesting, highlighting that this relationship might be junction protein expression.41
more complex than previously considered.28
Furthermore, the gut microbiota can affect other Microbiota and obesity—from gut to brain
obesity-associated factors such as hyperglycaemia and Appetite, food intake, and energy balance are centrally
insulin resistance.29 Germ-free mice have resistance to regulated by a complex network of neuroendocrine
these high-fat-diet-induced metabolic complications,30 factors and their receptors, which mediate the
which is lost after FMT from conventionally raised mice.6 bidirectional communication between the gastro­
The gut microbiota changes the composition and relative intestinal tract and the brain (figure 2).16 The presence
abundance of bile acid species, which might explain of nutrients in the gastrointestinal tract after meal
its effect on glucose and insulin homoeostasis.31 ingestion causes complex neural and hormonal signalling
A reduced bile acid concentration in the gut has been to the brain to inform of the ongoing change in nutritional
associated with bacterial overgrowth and inflammation.31 status. This signalling is mediated by afferent nerve fibres
Additionally, some gut bacteria metabolise bile acids and from the autonomic nervous system, such as the vagus
their conjugates for a source of energy, causing activation nerve, that project information from the gut to the
of bile acid receptors farnesoid X receptor (FXR) and nucleus tractus solitarius (NTS) as well as by effector
Takeda G-protein-coupled receptor 5 (TGR5), which are fibres that project to the smooth muscles of the gut.
essential receptors for maintaining glucose tolerance and Information from the NTS is then distributed to the
insulin sensitivity, in the intestine and liver (figure 2).32,33 hypothalamus, which regulates energy balance, appetite,
Activation of TGR5 improves liver function and glucose and food intake in the neurons of the arcuate nucleus
tolerance in obese mice by regulating intestinal GLP-1 (ARC).42 The ARC contains orexigenic neuropeptide Y,
production,32 whereas FXR deficiency in leptin-deficient agouti-related protein, and anorexigenic peptides, as well
mice protects against obesity and improves insulin as cocaine amphetamine-regulated transcript, and pro-
sensitivity.33 Obese patients and those with type 2 diabetes opiomelanocortin neurons (figure 2).42 This central role of
have altered bile acid metabolism,34 and administration the vagus nerve in appetite signalling is supported by
of bile acids, in both human and animal studies, led to studies that show that vagotomy in animal models causes
improved glycaemic control.35 Furthermore, the effect of a reduction in anorexigenic hormone signalling and
the gut microbiota on serotonin metabolism might also subsequent increases in food intake and weight gain.43,44
influence host glucose homoeostasis;36 pharmacological However, results from human studies are more
stimulation of 5-HT1B or 5-HT4 receptors increased contradictory and the mechanism by which vagal nerve
plasma active GLP-1 concentrations independent of stimulation affects eating behaviour remains unclear.45
feeding and improved glucose tolerance under dipeptidyl Additionally, gut peptide secretion from enteroendocrine
peptidase-4 inhibition in mice.37 cells also contributes to this signalling from the gut to the
The gut microbiota might also affect fat storage and brain via afferent nerve fibres as well as by direct secretion
hepatic lipid metabolism. Bäckhed and colleagues6 into the circulatory system.16 Some bacterial strains can
showed that the fasting-induced adipocyte factor, modify gut hormone secretion, including peptide YY,
a circulating lipoprotein lipase inhibitor, is selectively GLP-1, leptin, and ghrelin, and thus affect appetite and
suppressed by intestinal bacteria, inducing triglyceride satiety via hypothalamic neuroendocrine pathways
deposition in adipocytes.6 Moreover, gut bacteria affect (figure 2).46,47 Microbiota-derived metabolites, such as
the bioavailability of choline, an essential nutrient for short-chain fatty acids, can bind to receptors on
synthesis of very-low-density lipoproteins, which affects enteroendocrine cells, modifying the release of enteric
triglyceride storage in the liver.38 Furthermore, gut hormones into the systemic circulation.48,49 Hence,
microbiota-mediated activation of the bile acid FXR fermentation of non-digestible carbohydrates by the
receptor increases adiposity.39 intestinal microbiota has been shown to increase the

4 www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4


Review

production of short-chain fatty acids and secretion of gut activation, and production of microbial metabolites.4
hormones in animal and human studies.46 Depression-like and anxiety-like behaviours substantially
Acetate, the main short-chain fatty acid secreted by affect food intake, and different associations exist between
intestinal bacteria, directly suppresses appetite via central obesity and affective psychiatric disorders.61 Increases in
hypothalamic mechanisms.50 However, an increase in psychological stress augment the risk of developing
acetate concentration caused by altered microbiota leads anxiety and depression and activate the hedonic signalling
to activation of the parasympathetic nervous system, pathway, stimulating intake of calorie-dense so-called
promoting glucose-stimulated insulin secretion, increased comfort foods.61 Interestingly, FMT from either anxious or
ghrelin secretion, and obesity.51 Moreover, absence of a obese mice, or patients with depression, produces an
microbiota in germ-free mice substantially decreased anxious phenotype in the recipient rodent.63–65 Moreover,
expression of intestinal satiety peptides.17 These germ-free germ-free mice have exaggerated responses to stress66 and
mice also had increased expression of the oral fat taste alterations in neurodevelopment and behaviour.67 Further­
receptor, fatty acid translocase receptor, resulting in an more, modifi­cation of the gut microbiota via prebiotic
increased calorie intake from fats.17 These oral receptors administration has anxiolytic-like and antidepressant-like
transmit information from the taste papillae via nerve effects.68 The vagus nerve links mood to feeding and plays
fibres to the NTS in the brainstem.52 Moreover, in a key part in coordinating feeding behaviours45 and the
enteroendocrine cells, different taste receptors (eg, sweet, behavioural effects modulated by gut bacteria.69 Overall,
fat, bitter, and umami receptors) are expressed and their gut microbiota might clearly affect mood and ultimately
activation leads to secretion of GLP-1, cholecystokinin, affect brain circuits linked to feeding behaviours.
and ghrelin.52 Hence, modulation of taste receptors might
also be involved in the effect of the gut microbiota on the Diet as modifiable factor of microbiota in obesity
host’s appetite control. Another study53 in germ-free mice The composition of the intestinal microbiota and its
showed alteration in the intestinal sweet signalling function are established by the host’s genetic background
protein, T1R3, which led to increased con­sumption of and external factors, including the mode of birth,
nutritive sweet solutions.53 environmental elements, exercise, and nutrition.70 Most
Gut bacteria produce neuroactive metabolites, including notably, the key determinant affecting the composition
serotonin and γ-aminobutyric acid (GABA),54 which affect and activity of the gut microbiota is diet; changes in diet
the central control of appetite.55,56 Serotonin mediates its could explain 57% of structural variations in total gut
appetite-suppressant effects by modulating melanocortin microbiota.71 Different dietary components directly shape
neurons, which control bodyweight homoeostasis.57,58 the gut microbiota,16 and long-term dietary habits have
GABA, the main inhibitory neurotransmitter in the central profound effects on the composition of the intestinal
nervous system and one of the main neurotransmitters microbiota in human beings.72 Western diets, especially
involved in hypothalamic synaptic transmission, stimulates those associated with low levels of microbiota-accessible
feeding, and its synaptic release by agouti-related carbohydrates found in dietary fibre, have possibly
protein-expressing neurons in the hypothalamic ARC is reduced the diversity of microbiota over generations.
required for normal regulation of energy balance.56 By contrast, populations with traditional diets that are
high in fibre and low in sugar and fat, will have increased
Pathways related to mood, reward, and feeding microbiota diversity.73
In addition to the mechanisms mentioned previously, gut Diet-induced obesity in mice following a high fat and
microbiota might also affect host feeding behaviour by high sugar western-style diet was associated with an
modulating reward pathways and altering mood. Some increase in some Firmicutes, possibly caused by their
bacterial species might interact with host metabolism competitive advantage in processing simple sugars, and
through stimulation of systems outside of the a substantially lower abundance of Bacteroidetes.74 These
gastrointestinal tract such as the endocannabinoid changes in microbiota were diminished by subsequent
system, which affects gut barrier function, host dietary manipulations that limit weight gain and
metabolism,59 and the homoeostatic and hedonic control adiposity, reinforcing the interaction between diet and
of appetite and food intake.60 Brain reward signalling is the composition of the distal gut microbiota in metabolic
mediated by the dopaminergic mesolimbic system and function. The composition of the microbiota changes
has been postulated to play a major part in the substantially with age, from early life colonisation, to
development of obesity.61 A human imaging study found ageing-mediated decline in diversity. Altered gut
that increased colonic propionate, one of the main microbiota is particularly relevant for health in ageing
short-chain fatty acids secreted by gut bacteria, reduces because the microbiota might modulate age-related
anticipatory reward responses to high-energy foods via changes in innate immunity, sarcopenia, and the decline
striatal pathways.62 Gut bacteria might also affect this in cognitive function, all of which are elements of frailty.75
reward system by modulating gut hormone secretion.62 Moreover, these factors are modified by diet, with
Gut microbiota affects mood and behaviour via different decreased diversity in microbiota associated with
mechanisms such as vagal nerve stimulation, immune increased frailty and markers of inflammation.76

www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4 5


Review

Thaiss and colleagues77 found that the microbiome obesity84 and metabolic syndrome, including reduction in
contributes to accelerated post-dieting weight regain. body-mass index, waist circumference, blood pressure,
An intestinal microbiome signature that persisted after and blood glucose, as well as improved lipid metabolism.85
successful dieting in obese mice and contributed to However, only 5–10% of dietary polyphenols are absorbed
reduced post-dieting flavonoid concentrations and in the small intestine, whereas the remaining 90–95%
energy expenditure showed that flavonoid-based accumulate in the large intestine and act as energy
post-biotic inter­vention ameliorates excessive secondary substrates for some beneficial bacteria while inhibiting
weight regain. The recognition that diet is a key the growth of pathogenic bacteria.86 Thus, owing to their
determinant in short-term and long-term composition, low bioavailability, this prebiotic effect on the gut
diversity, dynamics of the intestinal microbiota, and sub­ microbiota is believed to be crucial for polyphenol-
sequent microbiota-driven host metabolic functioning, mediated health effects.77,86 Dietary proteins are essential
has generated interest in designing diets that enhance nutrients and are integral for a balanced diet. They are
the growth of specific beneficial anti-obesity gut the major source of nitrogen, which is essential for
microbiota. The table shows the diet’s effects on gut fermentation of carbohydrates and production of
microbiota and host metabolism. Fibre is one of the beneficial products such as short-chain fatty acids.87 The
main dietary components affecting the gut microbiota effect of dietary proteins on the gut microbiota requires
and consists of indigestible carbohydrates, which are investigation; for example, although a high-protein diet
fermented by gut bacteria, causing secretion of beneficial seems to lead to weight loss, it can also cause detrimental
metabolites (eg, short-chain fatty acids).78 High-fibre diets health effects, such as increased risk of colonic diseases,
are associated with different positive metabolic effects especially when combined with low dietary fibre intake
and a diverse, healthy microbiota.16 Dietary fat might also (table). Animal-based diets, which contain more protein
indirectly modulate the gut microbiota through bile acid than do plant-based diets, can increase the abundance of
secretion and composition.79 Bile acids have selective bile-tolerant microorganisms (Alistipes spp, Bilophila spp,
antimicrobial activity and could therefore mediate the and Bacteroides) and decrease that of Firmicutes that
fat-induced effects on the gut microbiota.79 In fact, metabolise dietary plant polysaccharides (Roseburia spp,
reduced concentrations of bile salt are associated Eubacterium rectale, and Ruminococcus bromii).88 Overall,
with bacterial overgrowth in the gut, particularly a high-fibre diet low in saturated fat leads to beneficial
Gram-negative members, including lipopolysaccharide anti-obesity effects (table). However, the precise mech­
pro­ducers and pathogens, leading to increased anisms by which nutritional factors and dietary strategies
inflammation, whereas increased bile salt concentrations modulate the intestinal microbiota and affect host
promote growth of Gram-positive bacteria such as metabolic signalling need further investigation.
Firmicutes.31 Saturated and unsaturated fats have Additionally, the composition of the gut microbiota
different health effects; although saturated fats are not might also be altered by various other factors such as
beneficial for obesity treatment, unsaturated fats have antibiotics, processed food, sweeteners, stress, and even
anti-obesity effects (table).83 Similarly, polyphenols are mode of birth (panel 2). These factors negatively affect
also gaining prominence as positive modulators of the intestinal microbiota, causing dysbiosis and ultimately
gut microbiota because of their positive effects on affecting host physiology and metabolism.

Interaction with the gut microbiota Gut microbiota changes Health effects
Fibre or Principal carbon and energy source for Increase in Bifidobacterium spp, Enhanced intestinal barrier integrity, insulin
carbohydrates16,72,78,80,81 colonic microorganisms; fermented to Bacteroidetes, Akkermansia muciniphila, sensitivity, and intestinal motility; decreased
beneficial metabolites such as Clostridium spp, and Prevotella spp inflammation; improved lipid metabolism;
short-chain fatty acids enhanced intestinal motility; and increased satiety
Proteins16,82 Major source of nitrogen for the gut Increase in Bacteroidetes and a Weight loss and increased risk of atherosclerosis
microbiota, and essential for their decrease in Bifidobacterium spp and colonic diseases
assimilation of carbohydrates, gases,
and putrefactive fermentation products
Saturated fats16,19,74,81–83 Indirect modulation of the gut Increase in Firmicutes, Proteobacteria, Increased endotoxaemia, bodyweight, and
microbiota via bile acid metabolism and Bilophila spp, and a decrease in adiposity, and reduced insulin sensitivity
Bacteroidetes and Bifidobacterium spp
Unsaturated Indirect interaction with the gut Increase in Bifidobacterium spp, Decreased inflammation and adiposity
fats16,19,74,81–83 microbiota via effects on bile acid Lactobacillus spp, and Akkermansia
secretion and composition muciniphla
Polyphenols84–86 Act as an energy substrate for some Increase in Bacteroides spp, Lactobacillus Reduced body-mass index, waist circumference,
beneficial bacteria and inhibit the spp, Bifidobacterium spp, and blood pressure, and blood glucose; and improved
growth of pathogenic bacteria Akkermansia muciniphila, and a decrease lipid metabolism
in Clostridium spp

Table: Effects of the diet on the gut microbiota and host metabolism

6 www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4


Review

Therapeutic strategies
With consideration of the gut microbiota’s key role Panel 2: Negative modulators of gut microbiota
in regulation of the host’s metabolism, energy homo­ Antibiotics
eostasis, and central appetite regulation, it is not surprising Overuse of antibiotics and exposure to antibiotics maternally or via the food chain,
that the microbiota is now a target to combat metabolic especially during early life, can have a large effect on the gut microbiota, with substantial
disorders such as obesity. Gut-microbiota-based treatments disturbances caused to its composition and functionality, which can subsequently disrupt
have subsequent effects on the host’s metabolism. gut barrier function and lead to influx of bacterial fragments into the circulation.
However, advances in the field are needed before gut- The resulting low-grade chronic inflammation and metabolic endotoxaemia can greatly
microbiota-based treatment can be used as a therapeutic affect host metabolism and insulin resistance. This microbiota alteration in early life has
tool to curb appetite and food intake, and restore metabolic long-lasting effects on bodyweight in adulthood. Epidemiological studies show that
imbalances in metabolic disorders such as obesity. exposure to antibiotics early in life is associated with increased risk of obesity and related
metabolic disorders.89
Probiotics
Western diet is driving obesity via different mechanisms in Emulsifiers
which changes in gut microbiota play a key part. Hence, These components of processed foods are used to improve food texture and extend their
western diet, preservatives, and emulsifiers are causing an shelf life. Dietary emulsifiers affect the gut microbiota, promoting colitis and metabolic
increase in the Firmicutes-to-Bacteroidetes ratio, which is syndrome in mice. Emulsifier-induced metabolic syndrome was associated with
characteristic in obese individuals.74,90 Thus, strategies to alterations in the microbiota and increased pro-inflammatory potential. Alterations in the
reverse this obesity-induced change in composition are of composition of gut microbiota included reduction in microbial diversity, a reduced
crucial pharmacological and nutritional interest in the abundance of Bacteroidales, an increased abundance of mucolytic bacteria (including
management of obesity and obesity-related disorders. Ruminococcus gnavus), bloom in Verrumicrobia (especially in Akkermansia muciniphila),
Dietary intervention by probiotic administration might be and enriched Proteobacteria. These changes were sufficient for low-grade inflammation
one of the approaches to modulate and maintain a healthy and metabolic syndrome development, as shown in germ-free mice and using faecal
microbiota. Probiotics are defined as “live microorganisms transplants in mice.90
that, when administrated in adequate amounts, confer Sweeteners
beneficial health effects on the host”.98 Studies highlight Non-caloric artificial sweeteners (NASs) are among the most widely used food additives
the potential role of probiotics in the treatment of worldwide. Although their consumption has been considered safe and beneficial because of
obesity. Hence, different bacterial strains have shown their low-calorie content, NAS consumption can lead to development of glucose intolerance
beneficial anti-obesity effects such as reduction of tissue through compositional and functional alterations in the intestinal microbiota.
inflammation, endotoxaemia, adiposity, bodyweight, leptin These alterations include increased abundance of Bacteroides spp and decreased abundance
levels, and energy intake.47 However, most studies have of Lactobacillus reuteri. NAS-mediated metabolic effects were prevented by antibiotic
used rodent models and, therefore, more studies in human treatment, and were transferrable to germ-free mice upon faecal microbiota transplantation
beings are needed to support the use of these probiotic from NAS-consuming mice, or from microbiota anaerobically incubated in the presence of
strains to treat or prevent obesity. The most common NAS. Similar NAS-induced dysbiosis and glucose intolerance were also seen in healthy
probiotic species that have shown these anti-obesity effects human participants.91
are Bifidobacterium and Lactobacillus spp. More recently,
Akkermansia muciniphila has emerged as one of the main Caesarean section
gut bacteria affecting host metabolism, with the capability Mode of birth is crucial for development of a healthy microbiota. Dominguez-Bello and
to reverse high-fat diet-induced metabolic effects such as colleagues92 showed that delivery mode establishes the acquisition and structure of the
fat-mass gain, metabolic endotoxaemia, adipose tissue initial microbiota in newborn babies. Hence, babies born by caesarean section have a
inflammation, and insulin resistance.99 Beneficial bacteria microbiota composition more similar to that of their mother’s skin, dominated by
interact with different components of the diet, mainly Staphylococcus, Corynebacterium, and Propionibacterium spp, whereas vaginally delivered
insoluble fibre, releasing bioactive metabolites that signal infants acquired bacterial communities more similar to that of their mother’s vaginal
to the host via the gut–brain axis.81 Further studies are microbiota, dominated by Lactobacillus, Prevotella, or Sneathia spp. These alterations in the
poised to elucidate the mechanism by which specific composition of the gut microbiota in early life might affect host physiology and metabolism
probiotics can harvest energy from food and nutrients. later in life. Epidemiological studies93–96 have reported associations between caesarean
section delivery and increased risk of obesity, asthma, allergies, and immune deficiencies.
Prebiotics Stress
In addition to the direct administration of live bacteria in Hypothalamus–pituitary–adrenal axis activity associated with stress affects the composition
probiotics, prebiotics might promote the growth of of the gut microbiota. Thus, early exposure to stress leads to decreased diversity in gut
beneficial bacteria over unfavourable commensal species microbiota and might have long-term effects on its composition. Moreover, chronic stress
in the gut. Prebiotics have been redefined as non-digestible in adulthood affects the composition of the intestinal microbiota, with a decrease in
compounds that, through their meta­ bolism by gut Bacteroides and Clostridium spp, and also leads to increased inflammation, which is
microorganisms, modulate com­position, activity, or both, indicative of immune activation. Furthermore, chronic stress alters the integrity of the
of the gut microbiota, which thus confers a beneficial intestinal barrier, increasing circulating concentrations of immunomodulatory bacterial cell
physiological effect on the host.80 Although dietary wall components such as lipopolysaccharide.97
carbohydrates, such as oligosaccharides (eg, insulin),

www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4 7


Review

Transfer of gut microbiota through a duodenal tube from


Search strategy and selection criteria
healthy donors with a normal body-mass index to obese
The aim of this Review was to identify studies investigating individuals diagnosed with type 2 diabetes increased
the effects of the gut microbiota on host metabolism and insulin sensitivity, faecal microbiota diversity, and butyrate-
central appetite regulation in obesity. Thus, we searched producing intestinal bacteria from the Clostridium
PubMed, Google Scholar, and Web of Science for the terms cluster XIVa, and decreased faecal short-chain fatty acids.103
“obesity”, “microbiota”, “microbiome”, “metabolic syndrome”, Moreover, a case study104 reported that a woman successfully
and “appetite” between Jan 1, 2000, and April 5, 2017. Only treated with FMT for Clostridium difficile developed
papers published in English were reviewed. The final reference new-onset obesity after receiving stool from a healthy but
list was generated on the basis of the relevance to this Review. overweight donor. This finding is controversial, as it is only
a case report; nevertheless, this finding is consistent with
observations in animal models, in which the obesity
fructo-oligosaccharides, and galacto-oligosaccharides, are phenotype was transferred to germ-free mice by FMT
the most commonly used prebiotics, other compounds, from obese donors.21 FMT might also pose some potential
such as polyphenols might also fit this definition.80 risks such as the spread of transmissible disease. However,
Administration of fibre-rich diets increases the abundance although there have been some mild effects such as
of beneficial bacteria in the gut, such as some diarrhoea and fever, no side-effects have been reported.105
Bifidobacterium and Lactobacillus spp, causing anti-obesity
effects, including reduction of endotoxaemia and Conclusions
enhanced intestinal barrier function caused by increased Host–microbe interactions are key for optimal health.
expression of tight junction proteins and decreased Commensal bacteria exert many structural and protective
circulating pro-inflammatory cytokines.47 A lower number effects on intestinal mucosa, but also affect host
of beneficial bacteria have been found in patients with metabolism. Accumulating evidence from both human
obesity and type 2 diabetes than in healthy individuals.47 and rodent studies highlights the central role of the gut
However, most of the studies are based on correlations microbiota in the gut–brain axis and its implication on
and do not establish a causal link between modulation of central appetite modulation. Moreover, the association
the gut microbiota and the observed beneficial metabolic between the composition of the intestinal microbiota and
and physiological effects. Therefore, more studies are metabolic dysfunction or obesity is becoming clear and
needed to elucidate the potential mechanisms involved in has been extensively reported. Therefore, modulation of
beneficial prebiotic-mediated effects.80 the gut microbiota might be a potential therapeutic target
for the treatment of obesity and other metabolic diseases,
FMT such as diabetes. Although use of prebiotics and probiotics
A promising new strategy to alter the composition of is one of the most widely used strategies to modulate the
the gut microbiota is FMT from healthy donors into gut microbiota and hence improve metabolic imbalances,
the patient’s intestinal tract, typically by colonoscopy other strategies, including other dietary components such
or duodenal endoscopy, resulting in the restoration as polyphenols and interventions such as bariatric surgery
of normal gut microbial community structure and and FMT, should also be considered.
functionality.100 Donor microbial strains in human beings Contributors
can colonise the recipient gut microbiota and persist for All authors contributed to the concept, structure, and writing of
at least 3 months after FMT.101 However, donor–recipient the manuscript.
compatibilities are important for successful establish­ Declaration of interests
ment of the donor’s microbial strains in the recipient’s We declare no competing interests.
gut, with a greater chance of prospering if the species Acknowledgments
were already present in the recipient.101 We are supported in part by a grant from Science Foundation Ireland
(Alimentary Pharmabiotic Centre grant number SFI/12/RC/2273) and
FMT has successfully been used to treat Clostridium received funding from the European Community’s Seventh Framework
difficile infections with greater than 90% efficacy.102 Such a Programme “MyNewGut” (grant agreement number FP7/2007–2013).
high efficacy is a hopeful indication of the ability of FMT to The Alimentary Pharmabiotic Centre Microbiome Institute has done
modify the gut microbiota and, therefore, of its potential as studies in collaboration with several companies including Nutritia,
4D Pharma, Cremo, Suntory Wellness, and Mead Johnson.
a therapeutic intervention for other diseases in which
gut microbiota dysbiosis is involved, such as ulcerative References
1 Ottman N, Smidt H, de Vos WM, Belzer C. The function of our
colitis, irritable bowel syndrome, chronic constipation, and microbiota: who is out there and what do they do?
Crohn’s disease. However, no conclusive results yet exist Front Cell Infect Microbiol 2012; 2: 104.
and more studies need to be done to draw conclusions 2 Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human
intestinal microbial flora. Science 2005; 308: 1635–38.
about the efficacy of FMT as a therapeutic intervention for 3 Patterson E, Ryan PM, Cryan JF, et al. Gut microbiota, obesity and
these gastrointestinal disorders.100 Additionally, evidence is diabetes. Postgrad Med J 2016; 92: 286–300.
increasing that FMT might also have potential to treat 4 Dinan TG, Cryan JF. Mood by microbe: towards clinical translation.
Genome Med 2016; 8: 36.
obesity and associated disorders such as type 2 diabetes.

8 www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4


Review

5 Neufeld KA, Kang N, Bienenstock J, Foster JA. Effects of intestinal 30 Bäckhed F, Manchester J, Semenkovich C, Gordon J.
microbiota on anxiety-like behavior. Commun Integr Biol 2011; Mechanisms underlying the resistance to diet-induced obesity in
4: 492–94. germ-free mice. Proc Natl Acad Sci USA 2007; 104: 979–84.
6 Bäckhed F, Ding H, Wang T, et al. The gut microbiota as an 31 Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut
environmental factor that regulates fat storage. microbiome. Curr Opin Gastroenterol 2014; 30: 332–38.
Proc Natl Acad Sci USA 2004; 101: 15718–23. 32 Thomas C, Gioiello A, Noriega L, et al. TGR5-mediated bile acid
7 Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, sensing controls glucose homeostasis. Cell Metab 2009; 10: 167–77.
Gordon JI. An obesity-associated gut microbiome with increased 33 Zhang Y, Ge X, Heemstra LA, et al. Loss of FXR protects against
capacity for energy harvest. Nature 2006; 444: 1027–31. diet-induced obesity and accelerates liver carcinogenesis in ob/ob
8 Khan MJ, Gerasimidis K, Edwards CA, Shaikh MG. Role of gut mice. Mol Endocrinol 2012; 26: 272–80.
microbiota in the aetiology of obesity: proposed mechanisms and 34 Trabelsi MS, Lestavel S, Staels B, Collet X. Intestinal bile acid receptors
review of the literature. J Obes 2016; published online Sept 15. are key regulators of glucose homeostasis. Proc Nutr Soc 2016; 16: 1–11.
DOI:10.1155/2016/7353642. 35 Staels B, Fonseca BA. Bile acids and metabolic regulation:
9 Fetissov SO. Role of the gut microbiota in host appetite control: mechanisms and clinical responses to bile acid sequestration.
bacterial growth to animal feeding behaviour. Nat Rev Endocrinol Diabetes Care 2009; 32: S237–45.
2016; 13: 11–25 36 O’Mahony SM, Clarke G, Borre YE, Dinan TG, Cryan JF.
10 Cox LM, Yamanishi S, Sohn J, et al. Altering the intestinal Serotonin, tryptophan metabolism and the brain-gut-microbiome
microbiota during a critical developmental window has lasting axis. Behav Brain Res 2015; 277: 32–48.
metabolic consequences. Cell 2014; 158: 705–21. 37 Nonogaki K, Kaji T. Pharmacological stimulation of serotonin
11 Aron-Wisnewsky J, Clement K. The effects of gastrointestinal 5-HT1B receptors enhances increases in plasma active glucagon-like
surgery on gut microbiota: potential contribution to improved peptide-1 levels induced by dipeptidyl peptidase-4 inhibition
insulin sensitivity. Curr Atheroscler Rep 2014; 16: 454. independently of feeding in mice. Diabetes Metab 2015; 41: 425–28.
12 Zhang H, DiBaise JK, Zuccolo A, et al. Human gut microbiota in 38 Dumas ME, Barton RH, Toye A, et al. Metabolic profiling reveals a
obesity and after gastric bypass. Proc Natl Acad Sci USA 2009; contribution of gut microbiota to fatty liver phenotype in
106: 2365–70. insulin-resistant mice. Proc Natl Acad Sci USA 2006; 103: 12511–16.
13 Liou AP, Paziuk M, Luevano JM Jr, Machineni S, Turnbaugh PJ, 39 Parseus A, Sommer N, Sommer F, et al. Microbiota-induced obesity
Kaplan LM. Conserved shifts in the gut microbiota due to gastric requires farnesoid X receptor. Gut 2017; 66: 429–37.
bypass reduce host weight and adiposity. Sci Transl Med 2013; 40 Delzenne NM, Neyrinck AM, Bäckhed F, Cani PD. Targeting gut
5: 178ra41. microbiota in obesity: effects of prebiotics and probiotics.
14 Bloom SR, Kuhajda FP, Laher I, et al. The obesity epidemic: Nat Rev Endocrinol 2011; 7: 639–46.
pharmacological challenges. Mol Interv 2008; 8: 82–98. 41 Rooks MG, Garrett WS. Gut microbiota, metabolites and host
15 O’Neill S, O’Driscoll L. Metabolic syndrome: a closer look at the immunity. Nat Rev Immunol 2016; 16: 341–52.
growing epidemic and its associated pathologies. Obes Rev 2015; 42 Schellekens H, Dinan TG, Cryan JF. Lean mean fat reducing
16: 1–12. “ghrelin” machine: hypothalamic ghrelin and ghrelin receptors as
16 Sandhu KV, Sherwin E, Schellekens H, Stanton C, Dinan TG, therapeutic targets in obesity. Neuropharmacology 2010; 58: 2–16.
Cryan JF. Feeding the microbiota-gut-brain axis: diet, microbiome, 43 Abbott CR, Monteiro M, Small CJ, et al. The inhibitory effects of
and neuropsychiatry. Transl Res 2016; 179: 223–44. peripheral administration of peptide YY(3–36) and glucagon-like
17 Duca FA, Swartz TD, Sakar Y, Covasa M. Increased oral detection, peptide-1 on food intake are attenuated by ablation of the
but decreased intestinal signaling for fats in mice lacking gut vagal-brainstem-hypothalamic pathway. Brain Res 2005; 1044: 127–31.
microbiota. PLoS One 2012; 7: e39748. 44 Berthoud HR. The vagus nerve, food intake and obesity. Regul Pept
18 Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, 2008; 149: 15–25.
Gordon JI. Obesity alters gut microbial ecology. 45 Bodenlos JS, Schneider KL, Oleski J, Gordon K, Rothschild AJ,
Proc Natl Acad Sci USA 2005; 102: 11070–75. Pagoto SL. Vagus nerve stimulation and food intake: effect of body
19 Turnbaugh PJ, Gordon JI. The core gut microbiome, energy balance mass index. J Diabetes Sci Technol 2014; 8: 590–95.
and obesity. J Physiol 2009; 587: 4153–58. 46 Everard A, Cani PD. Gut microbiota and GLP-1.
20 Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Rev Endocr Metab Disord 2014. 15: 189–96.
Gordon JI. An obesity-associated gut microbiome with increased 47 Torres-Fuentes C, Schellekens H, Dinan TG, Cryan JF. A natural
capacity for energy harvest. Nature 2006; 444: 1027–31. solution for obesity: bioactives for the prevention and treatment of
21 Ridaura VK, Faith JJ, Rey FE, et al. Gut microbiota from twins weight gain. A review. Nutr Neurosci 2015; 18: 49–65.
discordant for obesity modulate metabolism in mice. Science 2013; 48 Cani PD, Everard A, Duparc T. Gut microbiota, enteroendocrine
341: 1241214. functions and metabolism. Curr Opin Pharmacol 2013; 13: 935–40.
22 Sze MA, Schloss PD. Looking for a signal in the noise: 49 Nohr MK, Pedersen MH, Gille A, et al. GPR41/FFAR3 and
revisiting obesity and the microbiome. MBio 2016; 7: e01018-16. GPR43/FFAR2 as cosensors for short-chain fatty acids in
23 Ussar S, Griffin NW, Bezy O, et al. Interactions between gut enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in
microbiota, host genetics and diet modulate the predisposition to enteric leukocytes. Endocrinology 2013; 154: 3552–64.
obesity and metabolic syndrome. Cell Metab 2015; 22: 516–30. 50 Frost G, Sleeth ML, Sahuri-Arisoylu M, et al. The short-chain fatty
24 Goodrich JK, Waters JL, Poole AC, et al. Human genetics shape the acid acetate reduces appetite via a central homeostatic mechanism.
gut microbiome. Cell 2014. 159: 789–99. Nat Commun 2014; 5: 3611.
25 Liu R, Hong J, Xu X, et al. Gut microbiome and serum metabolome 51 Perry RJ, Peng L, Barry NA, et al. Acetate mediates a
alterations in obesity and after weight-loss intervention. Nat Med microbiome-brain-beta-cell axis to promote metabolic syndrome.
2017; published online June 19. DOI:10.1038/nm.4358. Nature 2016; 534: 213–17.
26 Zhang LS, Davies SS. Microbial metabolism of dietary components 52 Calvo SS, Egan JM. The endocrinology of taste receptors.
to bioactive metabolites: opportunities for new therapeutic Nat Rev Endocrinol 2015; 11: 213–27.
interventions. Genome Med 2016; 8: 46. 53 Swartz TD, Duca FA, de Wouters T, Sakar Y, Covasa M.
27 Krajmalnik-Brown R, Ilhan ZE, Kang DW, DiBaise JK. Effects of gut Up-regulation of intestinal type 1 taste receptor 3 and sodium
microbes on nutrient absorption and energy regulation. glucose luminal transporter-1 expression and increased sucrose
Nutr Clin Pract 2012; 27: 201–14. intake in mice lacking gut microbiota. Br J Nutr 2012; 107: 621–30.
28 Murphy EF, Cotter PD, Healy S, et al. Composition and energy 54 Wall R, Cryan JF, Ross RP, Fitzgerald GF, Dinan TG, Stanton C.
harvesting capacity of the gut microbiota: relationship to diet, Bacterial neuroactive compounds produced by psychobiotics.
obesity and time in mouse models. Gut 2010; 59: 1635–42. Adv Exp Med Biol 2014; 817: 221–39.
29 Boulange CL, Neves AL, Chilloux J, Nicholson JK, Dumas ME. 55 Meng F, Han Y, Srisai D. New inducible genetic method reveals
Impact of the gut microbiota on inflammation, obesity, critical roles of GABA in the control of feeding and metabolism.
and metabolic disease. Genome Med 2016; 8: 42. Proc Natl Acad Sci USA 2016; 113: 3645–50.

www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4 9


Review

56 Delgado TC. Glutamate and GABA in appetite regulation. 81 Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as
Front Endocrinol (Lausanne) 2013; 4: 103. moderators of human metabolism. Nature 2016; 535: 56–64.
57 Heisler LK, Jobst EE, Sutton GM, et al. Serotonin reciprocally 82 Conlon MA, Bird AR. The impact of diet and lifestyle on gut
regulates melanocortin neurons to modulate food intake. Neuron microbiota and human health. Nutrients 2015; 7: 17–44.
2006; 51: 239–49. 83 Caesar R, Tremaroli V, Kovatcheva-Datchary P, Cani PD, Backhed F.
58 Xu Y, Jones JE, Kohno D, et al. 5-HT2CRs expressed by Crosstalk between gut microbiota and dietary lipids aggravates WAT
pro-opiomelanocortin neurons regulate energy homeostasis. inflammation through TLR signaling. Cell Metab 2015; 22: 658–68.
Neuron 2008; 60: 582–89. 84 Cardona F, Andres-Lacueva C, Tulipani S, Tinahones FJ,
59 Cani PD, Plovier H, Van Hul M, et al. Endocannabinoids—at the Queipo-Ortuno MI. Benefits of polyphenols on gut microbiota and
crossroads between the gut microbiota and host metabolism. implications in human health. J Nutr Biochem 2013; 24: 1415–22.
Nat Rev Endocrinol 2016; 12: 133–43. 85 Amiot MJ, Riva C, Vinet A. Effects of dietary polyphenols on
60 Jager G, Witkamp RF. The endocannabinoid system and appetite: metabolic syndrome features in humans: a systematic review.
relevance for food reward. Nutr Res Rev 2014; 27: 172–85. Obes Rev 2016; 17: 573–86.
61 Schellekens H, Finger BC, Dinan TG, Cryan JF. Ghrelin signalling 86 Ozdal T, Sela DA, Xiao J, Boyacioglu D, Chen F, Capanoglu E.
and obesity: at the interface of stress, mood and food reward. The reciprocal interactions between polyphenols and gut microbiota
Pharmacol Ther 2012; 135: 316–26. and effects on bioaccessibility. Nutrients 2016; 8: 78.
62 Byrne CS, Chambers ES, Alhabeeb H, et al. Increased colonic 87 Conlon LE, Wallig MA, Erdman JW Jr. Low-lycopene containing
propionate reduces anticipatory reward responses in the human tomato powder diet does not protect against prostate cancer in
striatum to high-energy foods. Am J Clin Nutr 2016; 104: 5–14. TRAMP mice. Nutr Res 2015; 35: 882–90.
63 Collins SM, Kassam Z, Bercik P. The adoptive transfer of behavioral 88 David LA, Maurice CF, Carmody RN, et al. Diet rapidly and
phenotype via the intestinal microbiota: experimental evidence and reproducibly alters the human gut microbiome. Nature 2014;
clinical implications. Curr Opin Microbiol 2013; 16: 240–45. 505: 559–63.
64 Kelly JR, Borre Y, O’Brien C, et al. Transferring the blues: 89 Cox LM, Blaser MJ. Opinion antibiotics in early life and obesity.
depression-associated gut microbiota induces neurobehavioural Nat Rev Endocrinol 2015; 11: 182–90.
changes in the rat. J Psychiatr Res 2016; 82: 109–18. 90 Chassaing B, Koren O, Goodrich JK, et al. Corrigendum:
65 Bruce-Keller AJ, Salbaum JM, Luo M, et al. Obese-type gut dietary emulsifiers impact the mouse gut microbiota promoting
microbiota induce neurobehavioral changes in the absence of colitis and metabolic syndrome. Nature 2015; 519: 92–96.
obesity. Biol Psychiatry 2015; 77: 607–15. 91 Suez J, Korem T, Zeevi D. Artificial sweeteners induce glucose
66 Sudo N, Chida Y, Aiba Y, et al. Postnatal microbial colonization intolerance by altering the gut microbiota. Nature 2014; 514: 181–86.
programs the hypothalamic-pituitary-adrenal system for stress 92 Dominguez-Bello MG, Costello EK, Contreras M, et al. Delivery mode
response in mice. J Physiol 2004; 558: 263–75. shapes the acquisition and structure of the initial microbiota across
67 Dinan TG, Cryan JF. Gut instincts: microbiota as a key regulator of multiple body habitats in newborns. Proc Natl Acad Sci USA 2010;
brain development, ageing and neurodegeneration. J Physiol 2017; 107: 11971–75.
595: 489–503. 93 Thavagnanam S, Fleming J, Bromley A, Shields MD, Cardwell CR.
68 Burokas A, Arboleya S, Moloney RD, et al. Targeting the A meta-analysis of the association between caesarean section and
microbiota-gut-brain axis: prebiotics have anxiolytic and childhood asthma. Clin Exp Allergy 2008; 38: 629–33.
antidepressant-like effects and reverse the impact of chronic stress 94 Pistiner M, Gold DR, Abdulkerim H, Hoffman E, Celedon JC.
in mice. Biol Psychiatry 2017; published online Feb 24. DOI:10.1016/j. Birth by cesarean section, allergic rhinitis, and allergic sensitization
biopsych.2016.12.031. among children with a parental history of atopy.
69 Bravo JA, Forsythe P, Chew MV, et al. Ingestion of Lactobacillus J Allergy Clin Immunol 2008; 122: 274–79.
strain regulates emotional behavior and central GABA receptor 95 Huh SY, Rifas-Shiman SL, Zera CA, et al. Delivery by caesarean
expression in a mouse via the vagus nerve. Proc Natl Acad Sci USA section and risk of obesity in preschool age children: a prospective
2011; 108: 16050–55. cohort study. Arch Dis Child 2012; 97: 610–16.
70 Borre YE, O’Keeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF. 96 Sevelsted A, Stokholm J, Bonnelykke K, Bisgaard H. Cesarean section
Microbiota and neurodevelopmental windows: implications for and chronic immune disorders. Pediatrics 2015; published online
brain disorders. Trends Mol Med 2014; 20: 509–18. Dec 1. DOI:10.1542/peds.2014-0596.
71 Zhang C, Zhang M, Wang S, et al. Interactions between gut 97 Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of
microbiota, host genetics and diet relevant to development of the gut microbiota on brain and behaviour. Nat Rev Neurosci 2012;
metabolic syndromes in mice. ISME J 2010; 4: 232–41. 13: 701–12.
72 De Filippo C, Cavalieri D, Di Paola M, et al. Impact of diet in shaping 98 WHO, FAO. Probiotics in food. Health and nutritional properties and
gut microbiota revealed by a comparative study in children from guidelines for evaluation. 2006. http://www.fao.org/3/a-a0512e.pdf
Europe and rural Africa. Proc Natl Acad Sci USA 2010; 107: 14691–96. (accessed April 1, 2017).
73 Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, 99 Everard A, Belzer C, Geurts L, et al. Cross-talk between Akkermansia
Wingreen NS, Sonnenburg JL. Diet-induced extinctions in the gut muciniphila and intestinal epithelium controls diet-induced obesity.
microbiota compound over generations. Nature 2016; 529: 212–15. Proc Natl Acad Sci USA 2013; 110: 9066–71.
74 Turnbaugh P, Bäckhed F, Fulton L, Gordon J. Diet-induced obesity 100 Khoruts A, Sadowsky MJ. Understanding the mechanisms of faecal
is linked to marked but reversible alterations in the mouse distal microbiota transplantation. Nat Rev Gastroenterol Hepatol 2016;
gut microbiome. Cell Host Microbe 2008; 3: 213–23. 13: 508–16.
75 O’Toole PW, Jeffery IB. Gut microbiota and aging. Science 2015; 101 Li SS, Zhu A, Benes V. Durable coexistence of donor and recipient
350: 1214–15. strains after fecal microbiota transplantation. Science 2016;
76 Claesson MJ, Jeffery IB, Conde S, et al. Gut microbiota composition 352: 586–89.
correlates with diet and health in the elderly. Nature 2012; 488: 178–84. 102 Li YT, Cai HF, Wang ZH, Xu J, Fang JY. Systematic review with
77 Thaiss CA, Itav S, Rothschild D, et al. Persistent microbiome meta-analysis: long-term outcomes of faecal microbiota
alterations modulate the rate of post-dieting weight regain. Nature transplantation for clostridium difficile infection.
2016; 540: 544–51. Aliment Pharmacol Ther 2016; 43: 445–57.
78 Slavin J. Fiber and prebiotics: mechanisms and health benefits. 103 Vrieze A, Van Nood E, Holleman F. Transfer of intestinal microbiota
Nutrients 2013; 5: 1417–35. from lean donors increases insulin sensitivity in individuals with
79 Graf D, Di Cagno R, Fak F, et al. Contribution of diet to the metabolic syndrome. Gastroenterology 2012; 143: 913–16. e7.
composition of the human gut microbiota. Microb Ecol Health Dis 104 Alang N, Kelly CR. Weight gain after fecal microbiota transplantation.
2015; 26: 26164. Open Forum Infect Dis 2015; 2: ofv004.
80 Bindels LB, Delzenne NM, Cani PD, Walter J. Towards a more 105 Marotz CA, Zarrinpar A. Treating obesity and metabolic syndrome
comprehensive concept for prebiotics. Nat Rev Gastroenterol Hepatol with fecal microbiota transplantation. Yale J Biol Med 2016;
2015; 12: 303–10. 89: 383–88.

10 www.thelancet.com/gastrohep Published online August 24, 2017 http://dx.doi.org/10.1016/S2468-1253(17)30147-4

You might also like