Вы находитесь на странице: 1из 19

The Journal of Nutrition

Critical Review

Microbiota-Gut-Brain Axis: Modulator of Host


Metabolism and Appetite1,2
Marcel van de Wouw,3,4 Harriët Schellekens,3,4 Timothy G Dinan,4,5 and John F Cryan3–5*
3
Department of Anatomy and Neuroscience, 4 APC Microbiome Institute, and 5 Department of Psychiatry and Neurobehavioural
Science, University College Cork, Cork, Ireland

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


Abstract
The gut harbors an enormous diversity of microbes that are essential for the maintenance of homeostasis in health and
disease. A growing body of evidence supports the role of this microbiota in influencing host appetite and food intake.
Individual species within the gut microbiota are under selective pressure arising from nutrients available and other
bacterial species present. Each bacterial species within the gut aims to increase its own fitness, habitat, and survival via
specific fermentation of dietary nutrients and secretion of metabolites, many of which can influence host appetite and
eating behavior by directly affecting nutrient sensing and appetite and satiety-regulating systems. These include
microbiota-produced neuroactives and short-chain fatty acids. In addition, the gut microbiota is able to manipulate
intestinal barrier function, interact with bile acid metabolism, modulate the immune system, and influence host antigen
production, thus indirectly affecting eating behavior. A growing body of evidence indicates that there is a crucial role for
the microbiota in regulating different aspects of eating-related behavior, as well as behavioral comorbidities of eating
and metabolic disorders. The importance of intestinal microbiota composition has now been shown in obesity, anorexia
nervosa, and forms of severe acute malnutrition. Understanding the mechanisms in which the gut microbiota
can influence host appetite and metabolism will provide a better understanding of conditions wherein appetite
is dysregulated, such as obesity and other metabolic or eating disorders, leading to novel biotherapeutic
strategies. J Nutr 2017;147:727–45.

Keywords: nutrition, microbiota, obesity, metabolism, behavior

Introduction
the microbiota-gut-brain axis (7–9). Moreover, the gut micro-
The role of the gut microbiota in the regulation of physiology in biota is now being implicated in various aspects of disease
both health and disease is becoming increasingly recognized (10, 11).
(1, 2). These trillions of intestinal occupants form an integral In particular, the influence of the microbiome on host energy
evolutionary-driven relation with the host in maintaining metabolism and thus metabolic and eating-related disorders is
homeostasis (3). The gut microbiota is comprised of archaea, becoming increasingly established (12, 13). For instance, altered
viruses, protozoa, and mostly bacteria, the quantity of which intestinal microbiota composition has been demonstrated for
exceeds that of the eukaryotic cells in the human body (4, 5). obesity (14–16), anorexia nervosa (17–19), and forms of severe
More startling is the fact that the cumulative amount of unique acute malnutrition such as kwashiorkor (20). In addition, the
genes in the gut microbiota, the microbiome, is 100-fold higher gut microbiota and its metabolites are subsequently altered after
than that of the human genome, further indicating its signifi- interventions that address some of these conditions, such as
cance (6). Accumulating data also show that the gut microbiota bariatric or Roux-en-Y gastric bypass surgery (21–23) and
significantly affects the bidirectional communication between dietary interventions (24). Thus, accumulating evidence indi-
the gastrointestinal tract and the brain, which has been coined cates the potential of targeting the gut microbiota as an effective
therapeutic option for mitigating metabolic and eating disorders
1 (12, 25, 26). It is important to note that the gut microbiota has a
Supported by Science Foundation Ireland (SFI) grant nos. 07/CE/B1368 and
12/RC/2273; the Irish Health Research Board; the Department of Agriculture, Food, key regulatory role in both host metabolism and central appetite,
and the Marine; and Enterprise Ireland (all to JFC). Also supported by SFI grant no. which together can modify host eating behavior in metabolic
SFI/12/RC/2273 and by the Health Research Board through Health Research Awards disorders and eating disorders such as obesity and malnutrition
(grant nos. HRA_POR/2011/23, HRA_POR/2012/32, and HRA_POR/2014/647) and (27–29).
through EU grant 613979 (MYNEWGUT FP7-KBBE-2013-7) (all to TGD).
2
Author disclosures: M van de Wouw, H Schellekens, TG Dinan, and JF Cryan,
The homeostatic control of energy balance is highly regulated
no conflicts of interest. by a complex neuronal network wherein the hypothalamus
*To whom correspondence should be addressed. E-mail: j.cryan@ucc.ie. plays a key role connecting to the brainstem and various
ã 2017 American Society for Nutrition.
Manuscript received December 23, 2016. Initial review completed February 6, 2017. Revision accepted February 21, 2017. 727
First published online March 29, 2017; doi:10.3945/jn.116.240481.
forebrain regions (30). There are various peripheral anorexi- Obesity and metabolic syndrome. Obesity and related met-
genic hormones, such as glucagon-like peptide 1 (GLP-1)6, abolic syndrome have become one of the most prominent health
peptide YY (PYY), insulin, and leptin, that increase satiety, concerns in developed countries worldwide (39), with the WHO
whereas ghrelin is able to induce hunger (31). Within the estimating that nearly 600 million people worldwide are obese
hypothalamus, pro-opiomelanocortin (POMC) and cocaine- (40). Obesity is characterized by excessive adipose tissue, an
and amphetamine-regulated transcript–containing neurons reg- imbalance between energy intake and expenditure favoring a
ulate satiety, whereas neurons that contain neuropeptide Y and positive energy balance, and it is associated with low-grade
those that contain agouti-related peptide are involved in hunger inflammation (41) and insulin resistance (42). As such, obesity is a
signaling (30). These hypothalamic neurons project to other risk factor for numerous medical conditions, such as cardiovascu-
brain circuits such as the mesolimbic rewards circuit, which is lar disease, nonalcoholic fatty liver disease, endocrine and meta-
highly involved in food reward, addiction, and impulsive choice bolic disorders, as well as certain cancers (43). Considerable focus
(32). As such, signaling of the previously mentioned peripheral is being put on the contribution of overeating and a positive energy
hormones has also been implicated in both addiction (33) and balance to body weight gain. In addition, obesity is associated with
impulsivity (34). This additionally highlights the involvement of an increased prevalence of compulsive food behavior (32), as well
physiologic and/or psychological components in eating disorders as food-related impulsivity (44). There is a growing realization that

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


(35, 36). Importantly, the influence of the microbiota-gut-brain there is involvement of the gut microbiota in obesity, especially
axis is becoming increasingly recognized in psychological and considering that gut microbiota features could be used to predict
psychiatric disorders (37, 38), indicating that this axis could glucose responses, enabling personalized dietary interventions (45).
play a role in the physiologic and/or psychological components Notably, the gut microbiota is regarded as an important factor
of metabolic and eating disorders. contributing to nonalcoholic fatty liver disease, which is often
This review focuses on the diverse mechanisms through associated with obesity (46). For #25% of all nonalcoholic fatty
which the gut microbiota influences host metabolism, feeding liver disease patients, this condition progresses into a progressive
behavior, and appetite as a crucial piece of the puzzle in form of liver disease called nonalcoholic steatohepatitis, potentially
conditions wherein food intake and body weight are dysregu- resulting in cirrhosis, hepatocellular carcinomas, and liver dys-
lated. To focus on these, we first provide a brief overview of function (47). The importance of the gut microbiota in obesity is
various food-related disorders in which the gut microbiota is also emphasized by the fact that interventions targeting this
involved. Then we further introduce the role of the gut disorder, including bariatric surgery, and Roux-en-Y gastric bypass
microbiota in metabolic and eating disorders, with particular surgery in particular, induce substantial weight loss and are
emphasis on obesity. After that, we discuss the involvement of associated with alterations in gut microbiota composition (21–23,
gut microbial metabolites in the microbiota-gut-brain axis. 48). This is particularly important in regard to the mechanisms of
Special emphasis is laid on SCFAs and the diverse mechanisms actions, including changes in gut hormones, bile acid, and lipid
by which these are able to regulate host energy metabolism and metabolism, and neuronal signaling (49).
appetite. In addition, we examine the interplay between the gut
microbiota, intestinal barrier permeability, immune system Binge-eating disorder. Binge-eating disorder is characterized
functionality, and gut microbial mimicry. Further, we discuss by excessive, out-of-control, and rapid food intake (50, 51), and is
mechanisms by which the gut microbiota could influence host recognized as a distinct eating disorder by the Diagnostic and
nutrient and taste sensing throughout the gastrointestinal tract. Statistical Manual of Mental Disorders—Fifth Edition (52). It is
Finally, we briefly emphasize the multifaceted nature of eating the most common eating disorder, with an estimated lifetime
disorders in regard to their food-related behavioral components, prevalence of 1.9–2.8% in US adults, yet it often goes unrecog-
other behavioral comorbidities such as anxiety and depression, nized (50, 53). Enhanced food-related impulsivity and compul-
and the role that the gut microbiota plays herein. sivity plays a critical role in the maintenance of this disorder (44).
Binge-eating disorder is not only associated with obesity and
metabolic syndrome, but also with gastrointestinal disorders,
Metabolic and Eating Disorders asthma, and, among women, menstrual dysfunction, pregnancy
complications, intracranial hypertension, and polycystic ovary
The role of the gut microbiota in host metabolism and appetite
syndrome (54, 55). Even though the role of gut microbes in host
has been primarily investigated in regard to obesity and
eating behavior is being elucidated at a rapid rate (28), the link
metabolic syndrome. As such, less work has been done with
between binge-eating disorder and the gut microbiota currently
other eating disorders associated with a positive energy balance,
remains unclear, and more research is warranted.
such as binge-eating disorder and drug-induced obesity, or with
eating disorders associated with a negative energy balance, such
Drug-induced obesity. A change in body weight is often a side-
as anorexia nervosa, cachexia, and infant malnutrition. It is
effect of various medications. Atypical antipsychotics represent
crucial to note that, even though a dysfunctional energy balance
the mainstay of treatment for schizophrenia and bipolar
is common between all mentioned disorders, each disorder has
disorder, and they are known for their various side-effects,
other specific malfunctioning physiologic and psychological
such as weight gain and metabolic dysfunction (56–58). In
factors. As such, each metabolic and eating disorder is discussed
particular, the drug olanzapine is known for its weight gain–
separately in the following section to emphasize their particu-
inducing effects (59, 60). Rodents receiving olanzapine also have
larities and our current knowledge.
an altered gut microbiota composition (61, 62). In addition, rats
that also received an antibiotic cocktail had reduced olanzapine-
6
Abbreviations used: ClpB, caseinolytic protease B; CNS, central nervous induced body weight gain and various markers of metabolic
system; FFA2, FFA receptor 2; FFA3, FFA receptor 3; FXR, farnesoid X receptor; dysfunction, further highlighting the involvement of the gut
GABA, g-aminobutyric acid; GF, germ-free; GIP, gastric inhibitory polypeptide;
GLP-1 glucagon-like peptide 1; GPCR, G protein–coupled receptor; MSH,
microbiota (61). Another medication is the second-generation
melanocyte-stimulating hormone; POMC, pro-opiomelanocortin; PYY, peptide antipsychotic risperidone, often used to treat bipolar disorder
YY. and schizophrenia in children and adolescents (63). Chronic
728 van de Wouw et al.
risperidone treatment results in altered gut microbial composi- mice with an absent microbiota [i.e., germ-free (GF) mice], had
tion, with enriched pathways that have been implicated in dramatically decreased body fat despite having higher food
weight gain (64). Furthermore, mice receiving risperidone intake, both of which were normalized after colonization with
treatment displayed altered gut microbiota composition and a conventional gut microbiota (83). Shortly after this, it was
body weight gain, which was primarily attributed to decreased discovered that there was a different gut microbial ecology
energy expenditure (65). In addition, this trait was transmissible associated with obesity (84, 85), and that the gut microbiota
via fecal matter transplantation and via treatment with phage connected with obesity was associated with an increased capacity
isolated from risperidone-treated microbiota (65). for dietary energy harvest (86). Indeed, colonization of GF mice
with a gut microbiota originating from an obese human resulted
Anorexia nervosa. Anorexia nervosa is characterized by a in a substantial increase in total body fat compared with a lean gut
distorted body image leading to restricted food intake and microbiota (86). Nonetheless, even though a tremendous body of
subsequently severe weight loss, which in addition is associated evidence indicates that the gut microbiota plays a key role in
with hyperactivity and hypothermia (66). Anorexia nervosa has metabolic disorders, it was shown recently that a high-fat diet was
the highest mortality rate among psychiatric illnesses and able to drive obesity in mice, regardless of the gut microbial
significantly affects quality of life (67, 68). There is currently composition (87). In addition, a recent meta-analysis demon-

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


no clear understanding of its etiology, nor is there any medica- strated that the association between the gut microbial composi-
tion present. The role and involvement of the gut microbiota is tion found in human feces and obesity is relatively weak (88).
largely unclear in this disorder, even though current evidence The majority of the composition of the gut microbiota is
indicates that it could represent a potent intervention target (25). composed of the phyla Firmicutes (including Lactobacillus,
Clostridium, and Enterococcus) and Bacteroidetes (including
Cachexia. Cachexia is a metabolic condition caused by severe Bacteroides), with less from the phyla Actinobacteria (Bifi-
illness that is characterized by the loss of skeletal muscle mass, dobacteria), Proteobacteria (Escherichia coli), Fusobacteria,
both dependent of and independent of fat mass loss (69). Fifty Verrucomicrobia, and Cyanobacteria (6, 89). Recently, 2
to eighty percent of all cancer patients are affected by this population-based gut microbiome analysis studies demonstrated
condition, and it ultimately reduces quality of life and survival that the core microbiota within human populations is consid-
(70). Central elements in the pathophysiology of cancer cachexia erably robust (90, 91). Nonetheless, distinct compositional
are a reduced food intake and an abnormal metabolism differences have been demonstrated in individuals with obesity
leading to a negative protein and energy balance (71). These (14, 16), anorexia nervosa (17–19, 92, 93), and kwashiorkor
are primarily driven by the central nervous system (CNS) and (20). In regard to obesity, phylum-level changes, such as
inflammatory pathways (69). Even though the role of the gut increased Actinobacteria (14) and Firmicutes and decreased
microbiota in cancer cachexia is unclear, the former does play a Bacteroidetes, have been reported (84), as well as genus-level
role in both CNS and immune system regulation (72, 73), changes (16). Notably, the changes in Firmicutes and Bacter-
indicating that it may be a potential therapeutic target (26). In oidetes have not consistently been reported (94, 95). Interest-
particular, appetite-related signaling might be of interest here, ingly, anorexia is also associated with decreased Bacteroidetes
with the novel ghrelin-receptor agonist anamorelin recently and increased Actinobacteria and Verrucomicrobia levels (19).
having demonstrated that it increases lean body mass, as well as In addition to compositional differences, studies have
quality of life, in patients with cachexia (74, 75). reported a decrease in gut microbial diversity in obesity (14),
and anorexia nervosa (17–19). Interestingly, a recent study
Infant malnutrition. A mature gut microbiota configuration reported that weight gain in anorexia nervosa did not result in
resembling that of an adult is reached after the first 2–3 y of age the recovery of intestinal microbiota composition perturbations,
(76). Disturbances within this critical time period can result in an but did lead to increased microbial richness (19). This decreased
immature gut microbiota (77). Of note, the consequences of having gut microbial diversity could play a key role in the associated
an immature gut microbiome in later life in will be discussed in the dysregulation of appetite, metabolism, and food-related behaviors.
section Key Determinants of the Adult Gut Microbiota Composi- Alcock et al. (27) hypothesized that decreased microbial diversity,
tion in Metabolic and Eating Disorders. In early life, gut microbiota which tends to be associated with larger populations of the same
maturity is a crucial contributory factor to the prevalence and species, results in individual species having more energy and
severity of kwashiorkor, an enigmatic form of severe acute resources and a higher capacity for host manipulation, because
malnutrition that affects millions of children (20). Notably, this fewer resources are spent on competition. This, combined with the
condition is transferrable by gut microbiota transplantation, fact that many microbes are specialized in the use of specific
highlighting its importance (78, 79). In addition, a recent study nutrients, may result in the possibility that microbes influence the
by Wagner et al. (80) demonstrated that gut microbiota transplan- host to consume their preferred nutrients, or to influence host
tation from undernourished 24-mo-old Bangladeshi children into eating behavior in general (96). This concept is in agreement with
mice resulted in drastic weight loss, which was transmissible from the work of Fetissov (28), who showed that bacterial components
the dams to their offspring. The importance of the gut microbiota is and metabolites are able to influence intestinal satiety pathways,
emphasized by the fact that antibiotics such as amoxicillin and thus controlling host appetite and satiety.
cefdinir reduce mortality in children with kwashiorkor (81, 82).

Key Determinants of the Adult Gut


The Gut Microbiota as a Key Player Microbiota Composition in Metabolic
in Obesity and Metabolic and and Eating Disorders
Eating-Related Disorders
Gut microbial composition, especially in early life, is determined
One of the early and pivotal experiments demonstrating the key by various factors (97, 98). Perturbation of the gut microbiota in
role of the gut microbiota in host energy metabolism showed that the form of severe acute malnutrition during this period is
Microbiota and eating behavior 729
associated with persistent relative microbiota immaturity (77), potentially ketosis and alleviates these cyclical fluctuations in the
and this could also result in alterations in the development of gut microbiota (125). In addition, time-restricted feeding and
organs, tissues, and/or body systems (99), which may subse- fasting reverses numerous features of the metabolic syndrome
quently increase the predisposition to various diseases at an (125, 127–129).
older age (100). Indeed, it is postulated that microbiota Overall, there is a growing body of evidence linking the gut
immaturity could be causally related to the neurologic abnor- microbiota with nutrition, host metabolism, appetite, and
malities associated with child malnutrition (100). It is likely that circadian rhythm. As such, influencing one of these factors will
this could also affect the neuroendocrine systems associated with most likely result in alterations in the others, making the gut
appetite, food intake, and body weight both during this microbiota easily accessible and manipulable for targeting host
developmental phase and later in life. Notably, perturbation of metabolism and appetite control.
the gut microbiota by antibiotic exposure during this period
increases the risk of being overweight or obese later in childhood
(101–104), whereas probiotics have been postulated to have Gut Microbiota and Its Metabolites as a
beneficial effects on both offspring at risk of chronic disease and Therapeutic Target in Host Metabolism
offspring who are healthy (105).
and Appetite Control

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


Furthermore, various other factors, such as host genetics and
day-to-day diet, play a crucial role (106). In regard to diet, it is There are $4 primary means by which the gut microbiota can be
important to note that specific nutrients are correlated with an modulated: 1) the administration of live beneficial bacterial
increase in microbes that have the ability to utilize them (107– strains (probiotics), 2) the administration of host-indigestible
109). For example, the genus Bacteroides is associated with dietary fibers, which undergo bacterial fermentation and subse-
increased consumption of a high-carbohydrate, high-glycemic quently stimulate the growth of certain types of bacteria
index diet, as well as with an increased long-term consumption (prebiotics), 3) the administration of targeted antibiotics, and,
of dietary protein and animal fat (110, 111). Conversely, the finally, 4) fecal microbiota transplantation (Table 1). Both pro-
genus Prevotella is associated with an increased long-term and prebiotics have already been demonstrated to represent a
carbohydrate intake (110). Bifidobacteria have the capacity potent strategy to significantly improve metabolism in overweight
to use host-indigestible complex carbohydrates, which subse- and obese individuals (12, 130). Conversely, the effects of pre- and
quently can stimulate their proliferation and metabolic activity probiotics on undernourished individuals have been relatively
(111–113). A recent study also demonstrated that host- poorly studied, even though current evidence indicates that these
microbiome interactions were particularly influenced by nitro- could improve therapeutic outcomes (26, 140). Rodent studies
gen intake, of which dietary reduction was associated with have demonstrated that probiotic supplementation enhances
health (114). It is also interesting to note that consumption of the recovery from starvation (141–143). Nonetheless, much is still
noncaloric artificial sweetener saccharin alters both gut micro- unknown about the underlying mechanisms under which pre- and
biota composition and functionality, ultimately resulting in probiotics influence host physiology and behavior. The study of
glucose intolerance (115). Nutrient-microbe interactions also gut microbiota–host interactions is therefore crucial for the
include more specific food products and bacterial strains. For development of effective therapeutics targeting the gut micro-
instance, children in a rural African village in Burkina Faso who biota. Since the development of next-generation sequencing-based
are often raised on a polysaccharide-rich diet were found to metagenomics, our understanding of the composition, diversity,
harbor unique microbes associated with an increased capacity to and role of the gut microbiota in human health and disease has
digest cellulose (116). In addition, the gut microbiota of Hadza greatly expanded. However, gut microbial composition provides
hunter-gatherers is more enriched in bacteria associated with the little insight into the interactions between the microbiota and its
breakdown of dietary fibers, resulting in an enhanced ability to host. Research is therefore moving away from just compositional
digest and extract nutrients from fibrous plant foods (117, 118). analysis toward a more functional metabolomic parsing of
Furthermore, marine bacterial-derived enzymes have been microbiota-host interactions (144). In regard to diet and
discovered in the gut microbiota of healthy Japanese individuals, microbiota-gut-brain axis interactions, human brain imaging in
which was linked to increased local seaweed consumption (119, particular is a crucial tool to elucidate microbiota-host interac-
120). It is important to note that specialized taxa can be driven tions (100, 145). This approach will help us understand how the
into low abundance from decreased consumption of the nutri- microbiota influences brain function, and will likely identify
ents they thrive on, potentially resulting in the extinction of such underlying microbiota-gut-brain axis mechanics (145).
taxa (121). Microbial metabolites can affect host metabolism through
The interplay between appetite, metabolism, and gut micro- a variety of pathways, including direct interactions with the
biota is not only restricted to what is being eaten, but is also gastrointestinal tract and peripheral tissues (Figure 1). These
influenced by when food is consumed, and thus host circadian interactions include influencing host gene expression through
rhythm. It is long known that the circadian clock facilitates the epigenetic mechanisms (146), affecting the enteric nervous
anticipation of regularly timed periodic events such as food system and directly inducing vagus nerve signaling (147, 148),
consumption, resulting in optimized energy supply and demand, altering bile acid signaling (149), and affecting central appetite
and thus maximized metabolic fitness (122). The disruption of pathways integrating host energy status (150). It is also worth
behavioral and molecular circadian rhythms in diet-induced noting that individual gut microbes can influence the overall gut
obese mice suggests that the circadian clock is an important part microbiota composition and its metabolites. Interestingly, this
of understanding the mechanisms behind obesity and other particularly plays an important role in SCFA production
metabolic disorders (123). This disruption is not limited to the through a process called bacterial crossfeeding, wherein one
hostÕs endogenous circadian rhythm, but also disrupts the daily species of bacteria provides nutrients for another species (151).
oscillations in gut microbiota composition and function (124– The mechanisms described above are mostly mediated through
126). Interestingly, restricting feeding to a specific period of time gut microbial metabolites, which include, but are not limited to,
(i.e., time-restricted feeding) results in a period of fasting and SCFAs (152), bile acids (153), and various neuroactives.
730 van de Wouw et al.
TABLE 1 Means of gut microbial modulation and their impact in host appetite and energy metabolism

Method of microbial modulation Rationale General findings References

Probiotic administration Live beneficial bacterial strains secrete strain- Particular bacterial strains can affect body weight or 130–132
dependent metabolites and cause shifts in the symptoms associated with being overweight or
microbial composition of the small intestine and, obese, such as metabolic endotoxemia, insulin
to a lesser extent, the colon. Notably, most resistance, and cardiometabolic disorders.
probiotics do not colonize the hostÕs gut, and
continuous consumption often is necessary to
achieve lasting effects.
Prebiotic administration Host-indigestible dietary fibers pass through the These increase satiety and affect postprandial 133, 134
stomach and small intestine and are subsequently glucose and insulin concentrations. They also
fermented, stimulating the growth of certain types have been inconsistently reported to be associ-
of bacteria. ated with decreased energy intake, body weight,

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


insulin secretion, and circulating lipids and
inflammatory markers.
Antibiotic administration Antibiotics have been the cornerstone for treating Prenatal exposure to antibiotics affects birth weight, 103, 104, 135–139
infectious disease. They deplete specific gut obesity, and related metabolic sequelae later in
microbial taxa, depending on the particular life and increases the risk of childhood overweight
antibiotic given, resulting in long-term alterations and obesity when administered at an early age.
in gut microbial composition. Antibiotics interfere with the gut microbiota–
immune interaction, resulting in immune system
perturbations, and are associated with
Clostridium difficile infection when consumed
later in life.
Fecal microbiota transplantation This procedure normally is used after antibiotic Transplantation of an ``obese`` gut microbiota in 20, 86
treatment results in microbiota depletion, or in mice results in a transfer of obesity-associated
studies of germ-free animal models, since fecal symptoms; similar results are obtained in studies
microbiota transplantation provides an elegant way of kwashiorkor.
to demonstrate gut microbial–dependent effects.

One of these gut microbial neuroactives is g-aminobutyric acid production of bile acids is highly regulated through the nuclear
(GABA), which is produced by several Lactobacillus and Bifido- farnesoid X receptor (FXR) by negative feedback inhibition (167),
bacterium strains (154). L. reuteri is able to produce histamine through which bile acids are able to signal as both agonists and
(155, 156), and acetylcholine is produced by both bacteria and antagonists (149). GF and antibiotic studies highlight the impor-
fungi (157, 158). In addition, tryptophan decarboxylase enzymes tance of the gut microbiota in bile acid metabolism and FXR
have been detected in the human gut microbiota, suggesting that signaling (168–171), both of which are crucial for host metab-
the gut microbiota could produce the neurotransmitter trypta- olism and appetite (169, 172). In addition, Takeda G protein–
mine (159). Furthermore, gut microbial b-glucuronidase activity coupled receptor 5 is highly expressed in enteroendocrine L cells,
can increase concentrations of catecholamines as noradrenaline which, upon activation, results in the peripheral release of the
and dopamine (160). Interestingly, close to one-half of the anorexigenic hormones GLP-1 and PYY (173, 174). As such,
dopamine formed in the human body is produced in the enhancing the production of secondary bile acids through the
gastrointestinal tract (161). Finally, indigenous spore-forming increase of bacterial bile salt hydrolase enzymes results in reduced
gut bacteria are able to modulate local and peripheral host weight gain, serum cholesterol, and liver TGs (175). It is also
serotonin production (162). Much research is still needed to interesting to note that bile acids influence gut microbial
identify how local elevations in the gut of these transmitters can composition, indicating a bidirectional interaction (149). This is
affect host metabolism and even brain function, if, indeed, they highlighted in FXR-deficient mice, which have an altered
can. Interestingly, with regard to serotonin-modulating bacteria, microbiota and bile acid composition (169).
they have been demonstrated to affect host physiology, modu- The impact of the gut microbiota is not only restricted to the
lating gastrointestinal motility and platelet function (162). periphery, but can also influence central metabolites. For
As for bile acids, the gut microbiota has a dynamic interplay instance, GF mice have decreased hypothalamic histamine
with bile acid metabolism, conversion, and subsequent signal- concentrations (176). In addition, assessment of the cerebral
ing (149, 163). When food is consumed, primary bile acids stored metabolome of GF mice demonstrated that 38 of the 196
in the gall bladder are secreted in the duodenum, after which these metabolites were significantly altered (177). As such, the
can be deconjugated by gut microbes (i.e., removal of the glycine administration of specific bacterial strains can result in altered
or taurine conjugate), preventing reuptake in the small intestine. CNS metabolites. For instance, Bifidobacteria infantis is able
These deconjugated bile acids subsequently enter the colon and to reduce 5-hydroxyindoleacetic acid concentrations in the
are metabolized into secondary bile acids. At this level, the gut frontal cortex and a decrease in 3,4-dihydroxyphenylacetic
microbiota is able to influence primary bile acid synthesis in the acid in the amygdaloid cortex (178). These findings indicate
liver, both the deconjugation of these bile acids and conversion that specific gut microbes might influence appetite through
into secondary bile acids in the colon (164), and finally the serotonergic signaling (179), or could affect the dopaminergic
reabsorption of primary bile acids in the ileum (165, 166). The mesolimbic rewards circuit, which is highly involved in food
Microbiota and eating behavior 731
Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018
FIGURE 1 Gut microbial metabolite pathways to host appetite and metabolism control. Ingested nutrients are used by gut microbes,
thereby resulting in altered microbiota composition and functionality. These gut microbes influence bile acid metabolism and are able to
manufacture various metabolites, including SCFAs, neuroactives, small protein sequences, and toxins. Several of these gut microbial
metabolites are able to exert their effects through direct interaction with receptors in the gut on enteroendocrine L cells or the vagus nerve, or
by translocating through the intestinal epithelium into the peripheral circulation. Stimulation of L cells by bile acids or SCFAs results in the
release of anorexigenic hormones PYY and GLP-1, and can increase concentrations of peripheral hormones, such as insulin, leptin, and
ghrelin. Signaling mediated by these hormones can be affected by Ig, which in turn can be stimulated or interact with gut microbial–derived
small protein sequences as ClpB. The impact of such protein sequences, as well as gut microbial–derived toxins such as LPS, are highly
influenced by intestinal barrier integrity. LPS is also able to evoke an immune response, possibly leading to a decrease in taste receptor cell
differentiation and decreased taste detection. These pathways cumulatively affect appetite and metabolism, highly regulated by anorexigenic
pro-opiomelanocortin and cocaine- and amphetamine-regulated transcript and orexigenic neuropeptide Y and agouti-regulated peptide–
containing neurons in the hypothalamus, ultimately affecting eating behavior, eating-related behaviors (e.g., cognition and impulsivity), and
behavioral comorbidities associated with disorders in which appetite and metabolism are dysfunctional (e.g., anxiety and depression). Ach,
acetylcholine; ClpB, caseinolytic protease B; DA, dopamine; GABA, g-aminobutyric acid; GLP-1, glucagon-like peptide 1; HI, histamine; NA,
noradrenaline; PYY, peptide YY; TRP, tryptamine; 5-HT, serotonin.

reward, addiction, and impulsive choice (32). In addition, the Gut Microbial SCFAs and Their Impact on
gut microbiota plays a role in the regulation of brain-derived Host Appetite and Metabolism
neurotropic factor (180–182), a key player in the support of the
survival of existing neurons, the growth and differentiation SCFAs are perhaps the most extensively studied molecules with
of new neurons, and the formation of new synapses (183). respect to how the gut microbiota influences host energy
Regulation of central receptor expression has also been metabolism and appetite (Figure 2). Numerous studies have
reported. For instance, Lactobacillus rhamnosus is able to reported that overweight and obese individuals have increased
regulate central GABA receptor expression in a vagus nerve– SCFA concentrations (86, 95, 189, 190), even though this is not
dependent manner (184), and it was recently shown to increase always consistently reported (191). It is also interesting to note
central GABA concentrations (185). Notably, the role of that the previously mentioned second-generation antipsychotic
GABAergic neurotransmission is becoming increasingly recog- risperidone, known to induce weight-gain in children and
nized in host energy metabolism (186). Thus, bacterial strains adolescents, also increases gut microbial–derived SCFA con-
producing or altering these neurochemicals could provide a centrations (64). Conversely, individuals with anorexia nerv-
potent therapeutic for neurological diseases, and could most osa have been demonstrated to have decreased concentrations
likely play a pivotal role in influencing appetite and energy of both acetate and propionate (18), which were not al-
metabolism via modulation of the CNS (187, 188). tered after weight gain (19). SCFAs are generated by the
732 van de Wouw et al.
fermentation of nonhost-digestible dietary fibers by the gut butyrate and propionate are reported to be in the circulation at
microbiota and can provide #10% of total daily caloric intake 5- to 20-mmol/L concentrations, whereas acetate tends to be
(86, 192). As such, the ‘‘obese’’ gut microbiota has an increased within a range of 100–200 mmol/L (205–207). In addition, a
capacity for energy harvest through SCFA production (86). recent study demonstrated that systemic availability of colonic-
Nonetheless, SCFA supplementation tends to result in a reduc- administered acetate, propionate, and butyrate was 36%, 9%,
tion in body weight in both humans (193) and rodents (194– and 2% respectively (208).
196), and ameliorates gut microbial alterations found in mice
with diet-induced obesity to a more lean phenotype (196). In SCFA-induced signaling. SCFAs have many effects at the
addition, GF mice are resistant to diet-induced obesity, and even cellular level, one of which is the regulation of histone
though they have drastically lower SCFA concentrations (197– acetylation and methylation, resulting in altered gene expression
199), this has not been attributed directly to SCFA concentra- (209). This is likely mediated through the facilitation of histone
tions (197). acetyltransferase availability and inhibition of histone deacetylase
Over 95% of gut microbial–derived SCFAs are made up of (210–212). In particular, acetate increases histone acetyltransfer-
acetate, propionate, and butyrate (200). With respect to their ase availability (213), whereas butyrate is the most potent
use as an energy source, butyrate is highly used by colonocytes inhibitor of histone deacetylase classes I and IIa (211, 214).

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


(201–203), and propionate is primarily used by hepatocytes In addition, acetate is converted to acetyl-CoA, after which it is
in the liver (204). As such, only acetate is thought to reach integrated into the citric acid cycle (Krebs cycle), and finally used
the peripheral circulation at relatively high amounts (205), as a mitochondrial energy source (215). Notably, this increase in
resulting in a SCFA concentration gradient. In line with this cellular energy also boosts the mechanistic target of rapamycin,
theory are studies investigating SCFA concentrations in human which is known to be an ATP sensor and thus a homeostatic
subjects during surgery and sudden-death victims, in whom cellular energy sensor (216).

FIGURE 2 Gut microbial–derived SCFAs and their impact on host appetite and metabolism. SCFAs are the product of microbial fermentation in the
colon, in which they are able to induce histone deacetylase inhibition, increase histone acetyltransferase availability, increase serotonin synthesis, and
activate various GPCRs. Activation of these receptors results in the release of anorexigenic hormones, such as GLP-1 and PYY, into the peripheral
circulation. In addition, SCFAs increase concentrations of leptin and insulin. SCFAs are also able to pass through the intestinal epithelium to the portal vein,
in which portal nerves express FFA3, where they are able to induce vagus nerve signaling. In regard to their concentrations in the circulation and brain, only
acetate reaches high concentrations because of both colonic and hepatic clearance. Nonetheless, propionate is able to induce a decreased anticipatory
reward response through currently unknown mechanisms. All these factors cumulatively affect both short- and long-term host energy homeostasis and
appetite. FFA2, FFA receptor 2; FFA3, FFA receptor 3; GLP-1, glucagon-like peptide 1; GPCR, G protein–coupled receptor; GPR109A, niacin receptor 1;
HAT, histone acetyltransferase; HDACi, histone deacetylase inhibition; Olfr78, olfactory receptor 78; PYY, peptide YY; 5-HT, serotonin.

Microbiota and eating behavior 733


Locally in the gut, SCFAs are able to enhance colonic Acetate. The word acetate comes from the Latin word for
serotonin production and secretion (217–219), as well as vinegar, ‘‘acetum,’’ in which it is very abundant. Other dietary
facilitating the secretion of the anorexigenic hormones GLP-1 means of increasing endogenous acetate are alcohol consump-
and PYY from L cells in the gastrointestinal tract into the tion (248), and gut microbial production of acetate. The latter is
circulation (220). These effects are largely mediated through the mediated by the acetyl-CoA or the Wood-Ljungdahl pathway
G protein–coupled receptors (GPCRs) FFA receptor 2 (FFA2/ (249), after which a part is converted into butyrate by bacterial
GPR43) and FFA receptor 3 (FFA3/GPR41) (194, 221, 222). crossfeeding (250, 251). In regard to the effects of acetate on
The SCFAs receptors FFA2 and FFA3 are also expressed in host metabolism, dietary acetate supplementation in obese mice
adipocytes, where they increase the expression and secretion of does not significantly affect cumulative food intake, but does
the anorexigenic hormone leptin in vitro (223–226). Notably, ameliorate body weight gain, TG concentrations, fasting insulin,
FFA3 expression has not been consistently reported in adipo- and leptin (194, 196). This is without affecting fasting glycemia
cytes (223, 224, 227). The importance of these receptors was and oral glucose tolerance, all of which were completely restored
particularly emphasized by a recent study in which it was when either propionate or butyrate were administered (194). In
demonstrated that supplementation with all individual SCFAs, addition, acute oral administration of acetate does not alter
in addition to with a mix of all 3 principal SCFAs, in mice with GLP-1, PYY, insulin, gastric inhibitory polypeptide (GIP), or

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


diet-induced obesity resulted in an attenuation of the obesity- amylin concentrations (194). Acute acetate injection does,
associated decrease in FFA2 and FFA3, which subsequently however, decrease circulating concentrations of FFAs (252),
correlated with various biomarkers of obesity (196). In addition, result in appetite suppression, and suppress hypothalamic
mice overexpressing FFA2 specifically in adipose tissue remained neuronal activation patterning, as well as inducing a change in
lean even on high-fat diets, whereas FFA2-deficient mice were the expression profile of regulatory neuropeptides, favoring
obese while on a normal diet (228). appetite suppression (253). In addition, acetate infusions into
In regard to the nervous system, FFA3 in particular is the distal colon of overweight or obese men increase postpran-
expressed in the enteric nervous system (229) and vagal afferents dial glucose and insulin concentrations and fat oxidation (254).
(230) and throughout the peripheral nervous system (227, 231). Interestingly, a recent study showed that rats with diet-induced
In addition, butyrate is able increase the proportion of cholin- obesity had an increased whole-body acetate turnover, as well as
ergic enteric neurons through epigenetic mechanisms, indicating increased plasma and fecal acetate concentrations (255). It was
that the effects of SCFAs on the nervous system are not limited to subsequently demonstrated that acetate increased glucose-
neuronal activation (232). Importantly, de Vadder et al. (230) stimulated insulin secretion, ghrelin secretion, hyperphagia,
demonstrated that signaling through FFA3 expressed on portal and obesity through the parasympathetic nervous system (255).
nerves by propionate resulted in increased activity in the dorsal These data indicate that acetate may have a causal role in obesity
vagal complex, which receives inputs from the vagus nerve and (256). This is contrary to data demonstrating that acetate could
the hypothalamus, a key brain region in the control of appetite improve metabolic variables associated with obesity (194, 196,
and metabolism. 253, 254). As such, Bindels and Leclercq (257) recently
SCFAs have also been reported to exhibit potent immuno- discussed the current acetate controversy and stressed the
modulatory properties (233, 234). These include the regulation importance of location within the gastrointestinal tract as a
of the chemotaxis and inflammation of neutrophils (235–237) contributing factor for the differential effects of acetate on
and the suppression of inflammatory cytokine production of host metabolism (i.e., proximal or distal colon).
monocytes and macrophages (238, 239), as well as T regulatory,
T helper 1, and T helper 17 cell differentiation and subsequent Propionate. Propionate is widely used in Europe (European
cytokine production (240–243). Moreover, SCFAs alleviate food additive codes: E280–E282) as a food additive because
blood-brain barrier permeability and microglia immaturity in of its antifungal properties (258, 259). Its formation by gut
the CNS in GF mice (244, 245). It is important to note that microbes is mediated via the succinate, acrylate, and propane-
several of these anti-inflammatory effects have also been diol pathways (260, 261). With respect to its effects on host
demonstrated in mice with diet-induced obesity, indicating the metabolism, dietary propionate supplementation ameliorates
multifaceted role of SCFAs in conditions with disordered body weight gain, oral glucose tolerance, TG concentrations,
metabolism and appetite (195, 196). and fasting insulin and leptin in mice (194, 196). In addition,
Even though many of the previously mentioned effects can be acute oral administration increases GIP, insulin, and amylin
induced by all SCFAs, there are distinct differences between the without significantly altering PYY and GLP-1 (194). This could
principal gut microbial–derived SCFAs in the manner in which be due to the fact that oral propionate would most likely not
they affect their host that are important to keep in mind: 1) not reach the colon, where PYY- and GLP-1–containing L cells are
all SCFAs are increased or decreased to the same extent in located. This is in line with the fact that intracolonic adminis-
different pathophysiological conditions; 2) only acetate reaches tration of propionate does result in increased concentrations of
the circulation at high concentrations, also resulting in the PYY and GLP-1 in the circulation (222). Furthermore, acute
suggestion that only gut microbial–derived acetate will induce administration of inulin-propionate ester, which delivers propi-
the GPCR signaling of FFA2 and FFA3, whenever SCFAs are onate directly to the colon, leads to an increase in plasma
required to travel through the peripheral circulation before propionate, PYY, and GLP-1, and a decrease in energy intake
GPCR activation; 3) only acetate directly increases histone and appetite compared with inulin alone in healthy adults (193,
acetyltransferase availability, and butyrate is the most potent 262). This intervention also decreases caudate and nucleus
histone deacetylase inhibitor, indicating that the effects of the accumbens activity, indicating that colonic propionate could
individual SCFAs via epigenetic means might differ; and 4) even attenuate reward-based eating behavior via striatal pathways.
though all SCFAs activate FFA2 and FFA3, various GPCRs are However, this study did not detect any alterations in GLP-1,
specific to 1 or 2 of the principal SCFAs as olfactory receptor 78, PYY, or serum propionate, which were found in the former study
which is only activated by acetate and propionate (246), and (263). This does indicate that the observed effects on caudate
niacin receptor 1, which is activated by butyrate (247). and nucleus accumbens activity were independent of GLP-1 and
734 van de Wouw et al.
PYY and are more likely mediated through currently unknown bacteria, and bacterial products (267, 269). Low-level translo-
mechanisms. Long-term supplementation for 24 wk with this cation of substances or microbes across the tight junctions of
inulin-propionate ester in overweight adults did not significantly these luminal antigens is normal and shape a proper adaptive
affect overall body weight, but did result in decreased body immune system (270). Nonetheless, excessive intestinal perme-
weight gain, without altering basal PYY and GLP-1 concentra- ability, often colloquially referred to as ‘‘leaky gut,’’ is associated
tions (193). As such, these conflicting results call into question with the development of low-grade chronic inflammation and
the therapeutic potential of propionate in the regulation of sepsis, in which inflammatory mediators are thought to exacer-
metabolism and host appetite, and further research is warranted. bate intestinal permeability (271). Interestingly, decreased
intestinal permeability has been reported in individuals suffering
Butyrate. Butyrate, which is infamous for its strong smell of from anorexia nervosa (272), whereas increased intestinal
rancid butter and milk (264), has its etymologic roots in the permeability has been observed in obesity (273, 274) and
Greek word for butter. Butyrate is also widely known for its protein-energy wasting (for in-depth discussion see (275). Of
potent histone deacetylase inhibitory effects, which, combined note, SCFA supplementation has been demonstrated to improve
with its ability to cross the blood-brain barrier, has resulted in deficits in intestinal permeability (276–278). Obesity-associated
butyrate being studied extensively as a neuropharmacologic increases in intestinal permeability can lead to an increased

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


agent (at somewhat supraphysiological doses) to investigate translocation of Gram-negative bacterial-derived LPS into the
epigenetic mechanisms in brain and behavior research (265). circulation, resulting in chronic low-grade systemic inflamma-
Nonetheless, research performed on butyrate from this perspec- tion (273, 279, 280), in a condition called metabolic endotox-
tive has limited relevance to the effect of gut microbial–derived emia (281–283). Interestingly, administration of the antibiotics
butyrate, with many of the studies being focused on the ampicillin and neomycin decreases cecal LPS content and
administration of acute, systemic, and high doses. The produc- metabolic endotoxemia in both mice with diet-induced obesity
tion of butyrate by gut microbes is mediated through the enzyme and ob/ob mice, further emphasizing the role of the gut
butyryl-CoA:acetate CoA transferase (266). Interestingly, both microbiota in metabolic endotoxemia (284).
this enzyme and butyrate have a diurnal variation, which is Increased intestinal permeability can facilitate the transloca-
ablated in mice with diet-induced obesity (126). This also tion of gut microbial metabolites into the peripheral circulation.
appeared to be the case for the less-pronounced diurnal variation Some of these gut microbial metabolites have a structure similar
of propionate, whereas no variation at all was demonstrated for to the hostÕs own molecules that is divergent enough to be
acetate (126). In regard to butyrateÕs effect on host metabolism, recognized as foreign by the hostÕs immune system, in a
dietary butyrate supplementation ameliorates body weight gain, phenomenon known as molecular mimicry (285).
TG concentrations, oral glucose tolerance, and fasting insulin, as Various gut microbes have the capacity to produce protein
well as leptin in mice (194, 196). In addition, acute oral sequences of $5 amino acids that share a sequence that is
administration increases GIP, insulin, amylin, PYY, and GLP-1 identical to various appetite-regulating peptides, and can thus
(194). Oral administration of tributyrin, a TG that contains 3 trigger the production of Ig (286, 287). Ig has the ability to
butyrate moieties, reduces body weight gain in mice with diet- reduce the rate of degradation of gut-derived hormones and
induced obesity, likely because of increased energy expenditure, neuroendocrine factors, which was demonstrated to be the case
which is accompanied by an attenuation in glucose and insulin for the orexigenic hormone ghrelin (288), or can initiate an
tolerance (195). autoimmune response. Coadministration of the hunger hormone
The fact that obesity and metabolic syndrome seem to be ghrelin, together with IgG from obese individuals or ob/ob mice,
associated with increased SCFA concentrations (86, 95, 189, increased food intake in rodents, whereas IgG from anorectic
190), whereas supplementation with SCFAs tends to decrease patients or control animals did not (288). In addition, GF rats
acute food intake and markers associated with these disorders have increased concentrations of IgG directed at the orexigenic
(193–196), highlights the fact that much is still unknown about hormone ghrelin, and decreased concentrations of IgA directed
the role and impact of SCFAs on long-term energy homestasis at the anorexigenic neurotransmitter agouti-related peptide and
and metabolism. In addition, much is still unclear about the the anorexigenic melanocyte-stimulating hormone (MSH)
mechanisms in which gut microbial–derived SCFAs can influ- (286).
ence host appetite and metabolism. In particular, the impact of Moreover, a rat model of intestinal inflammation and
SCFAs in peripheral nervous system signaling remains largely anorexia induced by the chemotherapeutic agent methotrexate
understudied. Nonetheless, current evidence indicates that has been reported to have decreased concentrations of a-MSH
SCFAs represent a potential therapeutic strategy for diseases IgG, which likely plays a role in observed changes in food intake
with alterations in metabolism and appetite. and body weight (289). Acute administration of a-MSH Ig
results in increased food intake (290), even though long-term
effects may actually potentiate a-MSH–mediated signaling
The Interplay between Gut Microbiota and (291). Importantly, E. coli, a prominent inhabitant of the gut
the Immune System on Host Appetite microbiota, is able to produce a small protein sequence and
antigen-mimetic of a-MSH called caseinolytic protease B (ClpB)
and Metabolism
(290, 292, 293). ClpB is elevated in a variety of eating disorders
Gastrointestinal barrier functionality and permeability are associated with inadequate food intake (i.e., anorexia nervosa,
highly affected by the gut microbiota (267). This barrier not bulimia nervosa, and binge-eating disorder), and its concentra-
only regulates the absorption of nutrients, electrolytes, and tions have been correlated with various psychopathologic traits
water, but also prevents toxic substances and pathogens from (294). Interestingly, E. coli produces relatively more ClpB in the
entering the circulation from the lumen, which makes intestinal stationary phase of proliferation than in its exponential growth
barrier functionality a crucial player in the influence of the gut phase, indicating that E. coli induces a more potent anorexigenic
microbiota on its host (268). Barrier function and permeability effect whenever nutrients for E. coli stabilize. In addition,
are affected by several stimuli, including pathogens, commensal systemic administration of E. coli in the stationary phase also
Microbiota and eating behavior 735
increases c-Fos expression in POMC neurons of the arcuate and as increased sucrose preference, has been reported in GF mice
ventromedial nucleus of the hypothalamus generally associated (309). Furthermore, GF mice have an increased oral preference
with decreased food intake (293). Notably, this study harvested for and expression of lingual FA translocase (CD36), but
E. coli proteins by means of centrifugation, meaning that the decreased intestinal expression of the FA receptors GPR40,
observed effects were not necessarily ClpB-mediated (293). FFA3, FFA2, and GPR120, indicating decreased fat signaling
Nonetheless, the administration of ClpB-producing E. coli (310).
results in a short-term reduction in body weight and food intake Current knowledge about the impact of the gut microbiota
compared with the administration of ClpB-deficient E. coli on nutrient and taste sensing and, thus, gustatory function
(290). In addition, ClpB induces neuronal firing in POMC suggests that these effects could be modulated through the
neurons in the arcuate nucleus of the hypothalamus, which is immune system by affecting the continual supply of differenti-
associated with a decrease in food intake (293). It is also worth ated taste-receptor cells. These cells are essential in the detection
noting that female rats exhibit increased concentrations of and of taste compounds and transmit subsequent signals either
affinity for a-MSH IgG in response to acute oral E. coli directly or indirectly via taste bud cells, resulting in oral taste
administration, whereas males had increased a-MSH IgM perception (301, 311). Continual supply of differentiated taste
concentrations indicating sex-specific differences (292). receptor cells is crucial for normal taste function, and disruption

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


of this supply can be detrimental to taste signaling (312).
Activation of the immune system results in decreased cell
The Gut Microbiota in Host Nutrient and renewal and lifespan in both taste receptor and taste bud cells on
the tongue (313–315), which is potentially mediated through
Taste Sensing
mammalian toll-like receptors and type-I and -II IFN receptors,
The ability to sense and taste nutrients throughout the gastro- as these are localized in taste cells (314–316). Several studies
intestinal tract plays a crucial role in the maintenance of nutrient furthermore have pointed to the involvement of immune system
balance, the rewarding aspects of consuming food, and the functionality in taste. For instance, IL-10–knockout mice have a
identification of spoiled food. Interestingly, animal models have reduced number of taste buds and taste receptor cells and have
demonstrated that specific signaling mechanisms affect the an increased inflammatory response to LPS-induced inflamma-
responsiveness of different tastes (i.e., sweet, bitter, salty, sour, tion (317). In addition, TNF-knockout mice have a decreased
and umami). As such, modulation of these signaling mechanisms response to various bitter compounds, but not to other tastes
by gut microbes could influence the host to eat specific nutrients, (318).
therefore increasing their food substrates and survival. One can Interestingly, systemic administration of the bacterial-derived
therefore expect that individuals with eating disorders have an toxin LPS results in an inflammatory response in the tongue,
altered taste responsiveness corresponding with their phenotype. combined with decreased taste cell lifespan and decreased taste
Indeed, obesity is associated with a lowered responsiveness to preference (313, 314, 319–323). Furthermore, prolonged oral
sweet and fatty tastes, resulting in a shift toward liking foods LPS administration in mice results in decreased sweet taste
with higher concentrations of these tastes (295). In addition, receptor expression and a decreased response to sucrose (324).
individuals with anorexia nervosa have impaired taste percep- Notably, LPS administration also results in sickness behavior,
tion (296, 297), which has been shown to improve with weight which includes both anhedonic and anxiogenic components, and
gain (298, 299). it could therefore be a confounding factor when investigating
Signals conveying taste are detected by taste receptor cells behavior in vivo (325, 326). However, LPS administration
on the tongue that are subsequently transmitted through the decreases c-Fos expression in the lingual taste epithelium,
solitary tract into the thalamus, signaling to other brain regions indicating that there is local inhibited cellular activity (314). In
(300, 301). These taste receptor cells also express the anorex- addition, subcutaneous injection of LPS to the ventral surface
igenic hormones GLP-1, PYY, and cholecystokinin, indicating of the tongue results in leukocyte recruitment and inhibits
a peripheral signaling pathway via gastrointestinal hormone sodium-induced taste signaling via the chorda tympani, a
secretion, resulting in decreased appetite (302–305). Interest- primary sensory afferent nerve (327). Cumulatively, these
ingly, PYY knockout mice have a decreased behavioral response results indicate that the gut microbiota could influence host
to both fat- and bitter-tasting compounds (306). Subsequent nutrient and taste signaling, and thus gustatory function,
reconstitution of salivary PYY concentrations in these mice through immune system regulation, of which LPS is a potential
improves their response to fat, but not bitter taste (306). In mediator.
addition, higher concentrations of circulating TNF-a, insulin-
like growth factor 1, and leptin have been detected in individuals
with increased taste-responsiveness, indicating that these par- The Microbiota-Gut Brain Axis in
ticular circulating hormones could play a role in taste perception
Eating-Related Behavior
(307).
To our knowledge, few studies have investigated the role of It is highly likely that other eating-related behaviors besides
the gut microbiota in host nutrient and taste sensing. A recent gustatory function are influenced by the gut microbiota and its
study by Lyte et al. (308) demonstrated with the use of the metabolites. Although direct links with the gut microbiota
selectively bred occidental low- and high-saccharin–consuming have not been investigated extensively yet, it is likely that
rat model that rats innately more prone to saccharin consump- olfactory function, food-related cognitive processing, food-related
tion have a gut microbiota composition that is different from impulsivity and compulsivity, and the social aspects of food intake
those less prone to saccharin consumption. In addition, studies are susceptible to microbiome regulation. Interestingly, the gut
in GF mice suggest that the gut microbiota is able to influence microbiota has recently been hypothesized to play a key role in
taste receptors and, subsequently, taste. For instance, an increase addiction (328), which could have particular relevance to the
in intestinal sweet taste receptors, such as type 1 taste receptor 3, controversial concept of food addiction. In addition, the gut
a-gustducin, and sodium-glucose luminal transporter 1, as well microbiota has already been linked to cocaine use in mice (329)
736 van de Wouw et al.
and alcohol-dependence in humans (330). It is also interesting energy metabolism, will therefore ultimately result in better
to note that the administration of the principal psychoactive therapeutic strategies for obesity and eating disorders.
constituent of cannabis, D9-tetrahydrocannabinol, alleviates the
high-fat diet–induced increases in fat mass and body weight that Acknowledgments
are associated with an altered gut microbiota, although it is We thank Mahendra De Silva and Lilani De Silva for their
unclear whether the altered gut microbiota composition in generous financial assistance to MvdW. MvdW wrote the
conjunction with D9-tetrahydrocannabinol treatment played a manuscript; and HS, TGD, and JFC significantly contributed to
causative role in this reduction in fat mass and body weight (331). the manuscript concept and structure. All authors read and
Overall, much is still unknown about the role of the gut approved the final manuscript.
microbiota in food addiction, and other eating-related behaviors
in particular, and additional research is warranted for a more
comprehensive picture of the role of the gut microbiota in eating-
related behavior. References
1. Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold G,
Quraishi MN, Kinross J, Smidt H, Tuohy KM, et al. The gut microbiota

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


The Microbiota-Gut-Brain Axis in Metabolic and host health: a new clinical frontier. Gut 2016;65:330–9.
and Eating Disorder Comorbidities 2. Goulet O. Potential role of the intestinal microbiota in programming
health and disease. Nutr Rev 2015;73 Suppl 1:32–40.
Eating and metabolic disorders are often associated with other 3. Ley RE, Lozupone CA, Hamady M, Knight R, Gordon JI. Worlds
behavioral comorbidities, such as anxiety and depression (35, within worlds: evolution of the vertebrate gut microbiota. Nat Rev
36), and it is thus conceivable that there is a strong bidirectional Microbiol 2008;6:776–88.
interplay with these behaviors. It is therefore also likely that 4. Whitman WB, Coleman DC, Wiebe WJ. Prokaryotes: the unseen
microbes or microbial-derived metabolites that affect these majority. Proc Natl Acad Sci USA 1998;95:6578–83.
food-related behaviors will invariably alter the hostÕs appetite 5. Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Re-
visiting the ratio of bacterial to host cells in humans. Cell
and metabolism (Figure 1). Importantly, the influence of the
2016;164:337–40.
microbiota-gut-brain axis is also becoming increasingly recog-
6. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L,
nized in behaviors such as anxiety and depression (37, 38, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human
144, 332–334). As such, findings in regard to anxiety, stress, intestinal microbial flora. Science 2005;308:1635–8.
depression, sociability, and other behaviors are pivotal for a 7. Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implica-
comprehensive understanding of eating and metabolic disorders. tions of the brain-gut-enteric microbiota axis. Nat Rev Gastroenterol
Importantly, a recent study indeed demonstrated that the gut Hepatol 2009;6:306–14.
microbiota composition and diversity in individuals with ano- 8. Collins SM, Surette M, Bercik P. The interplay between the intestinal
microbiota and the brain. Nat Rev Microbiol 2012;10:735–42.
rexia nervosa at inpatient admission is correlated with levels of
9. Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the
depression, anxiety, and eating disorder psychopathology (17).
gut microbiota on brain and behaviour. Nat Rev Neurosci
In addition, many food-related behavioral findings in regard to 2012;13:701–12.
the microbiota-gut-brain axis are often mediated through 10. Moloney RD, Desbonnet L, Clarke G, Dinan TG, Cryan JF. The mi-
pathways similar to the ones involved in gut microbiota crobiome: stress, health and disease. Mamm Genome 2014;25(1–
interactions with host metabolism and appetite, including 2):49–74.
modulation of the immune system (37), vagus nerve signaling 11. Doré J, Blottière H. The influence of diet on the gut microbiota and its
(184, 335), and the production of neurochemicals (336). consequences for health. Curr Opin Biotechnol 2015;32:195–9.
12. Ojeda P, Bobe A, Dolan K, Leone V, Martinez K. Nutritional modu-
lation of gut microbiota - the impact on metabolic disease patho-
physiology. J Nutr Biochem 2016;28:191–200.
Conclusion
13. Arora T, Backhed F. The gut microbiota and metabolic disease: current
It is clear that the gut microbiota is a key regulator of host understanding and future perspectives. J Intern Med 2016;280:339–
appetite and metabolism. However, the majority of research has 49.
been focused on obesity and its comorbidities. As such, less is 14. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A,
Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, et al. A core gut
known about other disorders wherein appetite and metabolism microbiome in obese and lean twins. Nature 2009;457:480–4.
are dysregulated, such as binge-eating disorder, drug-induced 15. Turnbaugh PJ, Gordon JI. The core gut microbiome, energy balance
obesity, cachexia, anorexia nervosa, and infant malnutrition. and obesity. J Physiol 2009;587:4153–8.
Nonetheless, mechanisms in which the gut microbiota influences 16. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G,
the hostÕs appetite and metabolism are being elucidated at a Almeida M, Arumugam M, Batto JM, Kennedy S, et al. Richness of
rapid rate, providing a better understanding of its role in human gut microbiome correlates with metabolic markers. Nature
disorders with both a positive and negative energy balance. Such 2013;500:541–6.
information will be crucial for elucidating in which instances 17. Kleiman SC, Watson HJ, Bulik-Sullivan EC, Huh EY, Tarantino LM,
Bulik CM, Carroll IM. The intestinal microbiota in acute anorexia
alterations in gut microbiota composition and function are nervosa and during renourishment: relationship to depression, anxiety,
causative, or merely correlative, because many studies up until and eating disorder psychopathology. Psychosom Med 2015;77:969–
now have only identified associations. In this article, we have 81.
portrayed many of these direct and indirect mechanisms. Gut 18. Morita C, Tsuji H, Hata T, Gondo M, Takakura S, Kawai K,
microbes are able to secrete various substrates and metabolites, Yoshihara K, Ogata K, Nomoto K, Miyazaki K, et al. Gut dysbiosis in
affecting appetite- and metabolism-regulating systems and the patients with anorexia nervosa. PLoS One 2015;10:e0145274.
hostÕs ability to sense and taste nutrients, as well as eating- 19. Mack I, Cuntz U, Gramer C, Niedermaier S, Pohl C, Schwiertz A,
Zimmermann K, Zipfel S, Enck P, Penders J. Weight gain in anorexia
related behaviors and other behavioral comorbidities. This nervosa does not ameliorate the faecal microbiota, branched chain
information, cumulatively contributing to a greater understand- fatty acid profiles, and gastrointestinal complaints. Sci Rep
ing of the gut microbiota and its metabolites in appetite and 2016;6:26752.

Microbiota and eating behavior 737


20. Smith MI, Yatsunenko T, Manary MJ, Trehan I, Mkakosya R, 44. Schag K, Schonleber J, Teufel M, Zipfel S, Giel KE. Food-related
Cheng J, Kau AL, Rich SS, Concannon P, Mychaleckyj JC, et al. Gut impulsivity in obesity and binge eating disorder–a systematic review.
microbiomes of Malawian twin pairs discordant for kwashiorkor. Obesity Rev 2013;14(6):477–95.
Science 2013;339:548–54. 45. Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A,
21. Tremaroli V, Karlsson F, Werling M, Stahlman M, Kovatcheva- Ben-Yacov O, Lador D, Avnit-Sagi T, Lotan-Pompan M, et al. Per-
Datchary P, Olbers T, Fandriks L, le Roux CW, Nielsen J, Backhed F. sonalized nutrition by prediction of glycemic responses. Cell
Roux-en-Y gastric bypass and vertical banded gastroplasty induce 2015;163:1079–94.
long-term changes on the human gut microbiome contributing to fat 46. Bashiardes S, Shapiro H, Rozin S, Shibolet O, Elinav E. Non-alcoholic
mass regulation. Cell Metab 2015;22:228–38. fatty liver and the gut microbiota. Mol Metab 2016;5:782–94.
22. Zhang H, DiBaise JK, Zuccolo A, Kudrna D, Braidotti M, Yu Y, 47. Rinella ME. Nonalcoholic fatty liver disease: a systematic review.
Parameswaran P, Crowell MD, Wing R, Rittmann BE, et al. Human JAMA 2015;313:2263–73.
gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci
48. Palleja A, Kashani A, Allin KH, Nielsen T, Zhang C, Li Y, Brach T,
USA 2009;106:2365–70.
Liang S, Feng Q, Jorgensen NB, et al. Roux-en-Y gastric bypass
23. Gralka E, Luchinat C, Tenori L, Ernst B, Thurnheer M, Schultes B. surgery of morbidly obese patients induces swift and persistent
Metabolomic fingerprint of severe obesity is dynamically affected by changes of the individual gut microbiota. Genome Med 2016;8:67.
bariatric surgery in a procedure-dependent manner. Am J Clin Nutr
49. Chakravartty S, Tassinari D, Salerno A, Giorgakis E, Rubino F. What
2015;102:1313–22.
is the mechanism behind weight loss maintenance with gastric bypass?

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


24. Maukonen J, Saarela M. Human gut microbiota: does diet matter? Curr Obes Rep 2015;4:262–8.
Proc Nutr Soc 2015;74:23–36.
50. Hudson JI, Hiripi E, Pope HG Jr., Kessler RC. The prevalence and
25. Kleiman SC, Carroll IM, Tarantino LM, Bulik CM. Gut feelings: a role correlates of eating disorders in the National comorbidity survey
for the intestinal microbiota in anorexia nervosa? Int J Eat Disord replication. Biol Psychiatry 2007;61:348–58.
2015;48:449–51.
51. Swanson SA, Crow SJ, Le Grange D, Swendsen J, Merikangas KR.
26. Bindels LB, Delzenne NM. Muscle wasting: the gut microbiota as a Prevalence and correlates of eating disorders in adolescents. Results
new therapeutic target? Int J Biochem Cell Biol 2013;45:2186–90. from the national comorbidity survey replication adolescent supple-
27. Alcock J, Maley CC, Aktipis CA. Is eating behavior manipulated by ment. Arch Gen Psychiatry 2011;68:714–23.
the gastrointestinal microbiota? Evolutionary pressures and potential 52. American Psychiatric Association. Diagnostic and statistical manual of
mechanisms. BioEssays 2014;36:940–9. mental disorders. Washington (DC): American Psychiatric Associa-
28. Fetissov SO. Role of the gut microbiota in host appetite control: tion; 2013.
bacterial growth to animal feeding behaviour. Nat Rev Endocrinol 53. Kessler RC, Berglund PA, Chiu WT, Deitz AC, Hudson JI, Shahly V,
2017;13:11–25. Aguilar-Gaxiola S, Alonso J, Angermeyer MC, Benjet C, et al. The
29. Torres-Fuentes C, Schellekens H, Dinan TG, Cryan JF. A natural so- prevalence and correlates of binge eating disorder in the World Health
lution for obesity: bioactives for the prevention and treatment of Organization World Mental Health Surveys. Biol Psychiatry
weight gain. A review. Nutr Neurosci 2015;18:49–65. 2013;73:904–14.
30. Waterson MJ, Horvath TL. Neuronal regulation of energy homeo- 54. Olguin P, Fuentes M, Gabler G, Guerdjikova AI, Keck PE Jr.,
stasis: beyond the hypothalamus and feeding. Cell Metab McElroy SL. Medical comorbidity of binge eating disorder. Eat
2015;22:962–70. Weight Disord 2016 Aug 23 (Epub ahead of print; DOI: 10.1007/
31. Schellekens H, Finger BC, Dinan TG, Cryan JF. Ghrelin signalling and s40519-016-0313-5).
obesity: at the interface of stress, mood and food reward. Pharmacol 55. Mitchell JE. Medical comorbidity and medical complications associ-
Ther 2012;135:316–26. ated with binge-eating disorder. Int J Eat Disord 2016;49:319–23.
32. Murray S, Tulloch A, Gold MS, Avena NM. Hormonal and neural 56. Birkenaes AB, Birkeland KI, Engh JA, Faerden A, Jonsdottir H,
mechanisms of food reward, eating behaviour and obesity. Nat Rev Ringen PA, Friis S, Opjordsmoen S, Andreassen OA. Dyslipidemia
Endocrinol 2014;10:540–52. independent of body mass in antipsychotic-treated patients under real-
33. Engel JA, Jerlhag E. Role of appetite-regulating peptides in the life conditions. J Clin Psychopharmacol 2008;28:132–7.
pathophysiology of addiction: implications for pharmacotherapy. 57. Oriot P, Feys JL, Mertens de Wilmars S, Misson A, Ayache L,
CNS Drugs 2014;28:875–86. Fagnart O, Gruson D, Luts A, Jamart J, Hermans MP, et al. Insulin
34. Anderberg RH, Hansson C, Fenander M, Richard JE, Dickson SL, sensitivity, adjusted beta-cell function and adiponectinaemia among
Nissbrandt H, Bergquist F, Skibicka KP. The stomach-derived lean drug-naive schizophrenic patients treated with atypical antipsy-
hormone ghrelin increases impulsive behavior. Neuropsychopharma- chotic drugs: a nine-month prospective study. Diabetes Metab
cology 2016;41(5):1199–209. 2008;34:490–6.
35. Singh M. Mood, food, and obesity. Front Psychol 2014;5:925. 58. Perez-Iglesias R, Mata I, Pelayo-Teran JM, Amado JA, Garcia-
Unzueta MT, Berja A, Martinez-Garcia O, Vazquez-Barquero JL,
36. Cardi V, Leppanen J, Treasure J. The effects of negative and positive
Crespo-Facorro B. Glucose and lipid disturbances after 1 year of
mood induction on eating behaviour: a meta-analysis of laboratory
antipsychotic treatment in a drug-naive population. Schizophr Res
studies in the healthy population and eating and weight disorders.
2009;107:115–21.
Neurosci Biobehav Rev 2015;57:299–309.
59. Davey KJ, OÕMahony SM, Schellekens H, OÕSullivan O,
37. Dinan TG, Cryan JF. Microbes, immunity, and behavior: psychoneu-
Bienenstock J, Cotter PD, Dinan TG, Cryan JF. Gender-dependent
roimmunology meets the microbiome. Neuropsychopharmacology
consequences of chronic olanzapine in the rat: effects on body weight,
2017;42:178–92.
inflammatory, metabolic and microbiota parameters. Psychopharma-
38. Kelly JR, Clarke G, Cryan JF, Dinan TG. Brain-gut-microbiota axis: cology (Berl) 2012;221:155–69.
challenges for translation in psychiatry. Ann Epidemiol 2016;26:366–72.
60. Citrome L, Holt RI, Walker DJ, Hoffmann VP. Weight gain and
39. Vallis M. Quality of life and psychological well-being in obesity changes in metabolic variables following olanzapine treatment in
management: improving the odds of success by managing distress. Int schizophrenia and bipolar disorder. Clin Drug Investig 2011;31:455–
J Clin Pract 2016;70:196–205. 82.
40. WHO. Obesity and overweight. Factsheet [Internet]. [updated June 2016; 61. Davey KJ, Cotter PD, OÕSullivan O, Crispie F, Dinan TG, Cryan JF,
cited 2016 Dec 5]. Available from: http://www.who.int/mediacentre/ OÕMahony SM. Antipsychotics and the gut microbiome: olanzapine-
factsheets/fs311/en/. induced metabolic dysfunction is attenuated by antibiotic administra-
41. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. tion in the rat. Transl Psychiatry 2013;3:e309.
Annu Rev Immunol 2011;29:415–45. 62. Morgan AP, Crowley JJ, Nonneman RJ, Quackenbush CR, Miller CN,
42. Castro AV, Kolka CM, Kim SP, Bergman RN. Obesity, insulin resis- Ryan AK, Bogue MA, Paredes SH, Yourstone S, Carroll IM, et al. The
tance and comorbidities? Mechanisms of association. Arq Bras En- antipsychotic olanzapine interacts with the gut microbiome to cause
docrinol Metabol 2014;58:600–9. weight gain in mouse. PLoS One 2014;9:e115225.
43. Guh DP, Zhang W, Bansback N, Amarsi Z, Birmingham CL, Anis AH. 63. Bishop JR, Pavuluri MN. Review of risperidone for the treatment of
The incidence of co-morbidities related to obesity and overweight: a pediatric and adolescent bipolar disorder and schizophrenia. Neuro-
systematic review and meta-analysis. BMC Public Health 2009;9:88. psychiatr Dis Treat 2008;4:55–68.

738 van de Wouw et al.


64. Bahr SM, Tyler BC, Wooldridge N, Butcher BD, Burns TL, 85. Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD,
Teesch LM, Oltman CL, Azcarate-Peril MA, Kirby JR, Calarge CA. Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci
Use of the second-generation antipsychotic, risperidone, and second- USA 2005;102:11070–5.
ary weight gain are associated with an altered gut microbiota in 86. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER,
children. Transl Psychiatry 2015;5:e652. Gordon JI. An obesity-associated gut microbiome with increased
65. Bahra SM, Weidemann BJ, Castro AN, Walsh JW, deLeon O, capacity for energy harvest. Nature 2006;444:1027–31.
Burnett CM, Pearson NA, Murry DJ, Grobe JL, Kirby JR. Risperidone- 87. Rabot S, Membrez M, Blancher F, Berger B, Moine D, Krause L,
induced weight gain is mediated through shifts in the gut microbiome and Bibiloni R, Bruneau A, Gerard P, Siddharth J, et al. High fat diet drives
suppression of energy expenditure. EBioMedicine 2015;2:1725–34. obesity regardless the composition of gut microbiota in mice. Sci Rep
66. Hebebrand J, Exner C, Hebebrand K, Holtkamp C, Casper RC, 2016;6:32484.
Remschmidt H, Herpertz-Dahlmann B, Klingenspor M. Hyperactivity 88. Sze MA, Schloss PD. Looking for a signal in the noise: revisiting
in patients with anorexia nervosa and in semistarved rats: evidence obesity and the microbiome. MBio 2016;7:e01018–16.
for a pivotal role of hypoleptinemia. Physiol Behav 2003;79:25–37. 89. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C,
67. Huas C, Caille A, Godart N, Foulon C, Pham-Scottez A, Divac S, Nielsen T, Pons N, Levenez F, Yamada T, et al. A human gut microbial
Dechartres A, Lavoisy G, Guelfi JD, Rouillon F, et al. Factors pre- gene catalogue established by metagenomic sequencing. Nature
dictive of ten-year mortality in severe anorexia nervosa patients. Acta 2010;464:59–65.
Psychiatr Scand 2011;123:62–70. 90. Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M,

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


Vatanen T, Mujagic Z, Vila AV, Falony G, Vieira-Silva S, et al.
68. Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients
Population-based metagenomics analysis reveals markers for gut
with anorexia nervosa and other eating disorders. A meta-analysis of
microbiome composition and diversity. Science 2016;352:565–9.
36 studies. Arch Gen Psychiatry 2011;68:724–31.
91. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K,
69. Fearon K, Arends J, Baracos V. Understanding the mechanisms and
Kurilshikov A, Bonder MJ, Valles-Colomer M, Vandeputte D, et al.
treatment options in cancer cachexia. Nat Rev Clin Oncol 2013;10:90–9.
Population-level analysis of gut microbiome variation. Science
70. Ryan AM, Power DG, Daly L, Cushen SJ, Ni Bhuachalla E, Prado CM. 2016;352:560–4.
Cancer-associated malnutrition, cachexia and sarcopenia: the skeleton in
92. Armougom F, Henry M, Vialettes B, Raccah D, Raoult D. Monitoring
the hospital closet 40 years later. Proc Nutr Soc 2016;75:199–211.
bacterial community of human gut microbiota reveals an increase in
71. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, Lactobacillus in obese patients and Methanogens in anorexic patients.
Jatoi A, Loprinzi C, MacDonald N, Mantovani G, et al. Definition PLoS One 2009;4:e7125.
and classification of cancer cachexia: an international consensus. 93. Million M, Angelakis E, Maraninchi M, Henry M, Giorgi R, Valero R,
Lancet Oncol 2011;12:489–95. Vialettes B, Raoult D. Correlation between body mass index and gut
72. El Aidy S, Dinan TG, Cryan JF. Gut Microbiota: the conductor in concentrations of Lactobacillus reuteri, Bifidobacterium animalis,
the orchestra of immune-neuroendocrine communication. Clin Ther Methanobrevibacter smithii and Escherichia coli. Int J Obes (Lond)
2015;37:954–67. 2013;37:1460–6.
73. Burokas A, Moloney RD, Dinan TG, Cryan JF. Microbiota regulation 94. Duncan SH, Lobley GE, Holtrop G, Ince J, Johnstone AM, Louis P,
of the mammalian gut-brain axis. Adv Appl Microbiol 2015;91:1–62. Flint HJ. Human colonic microbiota associated with diet, obesity and
74. Takayama K, Katakami N, Yokoyama T, Atagi S, Yoshimori K, weight loss. Int J Obes (Lond) 2008;32:1720–4.
Kagamu H, Saito H, Takiguchi Y, Aoe K, Koyama A, et al. Anamor- 95. Schwiertz A, Taras D, Schafer K, Beijer S, Bos NA, Donus C,
elin (ONO-7643) in Japanese patients with non-small cell lung cancer Hardt PD. Microbiota and SCFA in lean and overweight healthy
and cachexia: results of a randomized phase 2 trial. Support Care subjects. Obesity (Silver Spring) 2010;18:190–5.
Cancer 2016;24:3495–505. 96. Norris V, Molina F, Gewirtz AT. Hypothesis: bacteria control host
75. Temel JS, Abernethy AP, Currow DC, Friend J, Duus EM, Yan Y, appetites. J Bacteriol 2013;195:411–6.
Fearon KC. Anamorelin in patients with non-small-cell lung cancer and 97. Borre YE, OÕKeeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF.
cachexia (ROMANA 1 and ROMANA 2): results from two rando- Microbiota and neurodevelopmental windows: implications for brain
mised, double-blind, phase 3 trials. Lancet Oncol 2016;17:519–31. disorders. Trends Mol Med 2014;20:509–18.
76. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, 98. Diaz Heijtz R. Fetal, neonatal, and infant microbiome: Perturbations
Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, and subsequent effects on brain development and behavior. Semin
et al. Human gut microbiome viewed across age and geography. Na- Fetal Neonatal Med 2016;21:410–7.
ture 2012;486:222–7. 99. Barker DJ. The fetal and infant origins of adult disease. BMJ
77. Subramanian S, Huq S, Yatsunenko T, Haque R, Mahfuz M, 1990;301:1111.
Alam MA, Benezra A, DeStefano J, Meier MF, Muegge BD, et al. 100. Goyal MS, Venkatesh S, Milbrandt J, Gordon JI, Raichle ME. Feeding the
Persistent gut microbiota immaturity in malnourished Bangladeshi brain and nurturing the mind: linking nutrition and the gut microbiota to
children. Nature 2014;510:417–21. brain development. Proc Natl Acad Sci USA 2015;112:14105–12.
78. Tilg H, Moschen AR. Malnutrition and microbiota–a new relation- 101. Ajslev TA, Andersen CS, Gamborg M, Sorensen TI, Jess T. Childhood
ship? Nat Rev Gastroenterol Hepatol 2013;10:261–2. overweight after establishment of the gut microbiota: the role of de-
79. Million M, Diallo A, Raoult D. Gut microbiota and malnutrition. livery mode, pre-pregnancy weight and early administration of
Microb Pathog 2016 Feb 4 (Epub ahead of print; DOI: 10.1016/ antibiotics. Int J Obes (Lond) 2011;35:522–9.
j.micpath.2016.02.003). 102. Scott FI, Horton DB, Mamtani R, Haynes K, Goldberg DS, Lee DY,
80. Wagner VE, Dey N, Guruge J, Hsiao A, Ahern PP, Semenkovich NP, Lewis JD. Administration of antibiotics to children before age 2 years in-
Blanton LV, Cheng J, Griffin N, Stappenbeck TS, et al. Effects of a gut creases risk for childhood obesity. Gastroenterology 2016;151(1):120–9 e5.
pathobiont in a gnotobiotic mouse model of childhood undernutrition. 103. Trasande L, Blustein J, Liu M, Corwin E, Cox LM, Blaser MJ. Infant
Sci Transl Med 2016;8:366ra164. antibiotic exposures and early-life body mass. Int J Obes (Lond)
81. Trehan I, Goldbach HS, LaGrone LN, Meuli GJ, Wang RJ, 2013;37:16–23.
Maleta KM, Manary MJ. Antibiotics as part of the management of 104. Azad MB, Bridgman SL, Becker AB, Kozyrskyj AL. Infant antibiotic
severe acute malnutrition. N Engl J Med 2013;368:425–35. exposure and the development of childhood overweight and central
82. Million M, Lagier JC, Raoult D. Meta-analysis on efficacy of amox- adiposity. Int J Obes (Lond) 2014;38:1290–8.
icillin in uncomplicated severe acute malnutrition. Microb Pathog 2016 105. Hashemi A, Villa CR, Comelli EM. Probiotics in early life: a preven-
Jun 29 (Epub ahead of print; DOI: 10.1016/j.micpath.2016.06.025). tative and treatment approach. Food Funct 2016;7:1752–68.
83. Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, 106. Ussar S, Griffin NW, Bezy O, Fujisaka S, Vienberg S, Softic S, Deng L,
Semenkovich CF, Gordon JI. The gut microbiota as an environmental Bry L, Gordon JI, Kahn CR. Interactions between gut microbiota, host
factor that regulates fat storage. Proc Natl Acad Sci USA genetics and diet modulate the predisposition to obesity and metabolic
2004;101:15718–23. syndrome. Cell Metab 2015;22:516–30.
84. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human 107. Conlon MA, Bird AR. The impact of diet and lifestyle on gut micro-
gut microbes associated with obesity. Nature 2006;444:1022–3. biota and human health. Nutrients 2014;7:17–44.

Microbiota and eating behavior 739


108. Sonnenburg JL, Backhed F. Diet-microbiota interactions as modera- 129. Longo VD, Mattson MP. Fasting: molecular mechanisms and clinical
tors of human metabolism. Nature 2016;535:56–64. applications. Cell Metab 2014;19:181–92.
109. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, 130. Le Barz M, Anhe FF, Varin TV, Desjardins Y, Levy E, Roy D,
Griffin NW, Lombard V, Henrissat B, Bain JR, et al. Gut microbiota Urdaci MC, Marette A. Probiotics as complementary treatment for
from twins discordant for obesity modulate metabolism in mice. Sci- metabolic disorders. Diabetes Metab J 2015;39:291–303.
ence 2013;341:1241214. 131. Derrien M, van Hylckama Vlieg JE. Fate, activity, and impact of in-
110. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, gested bacteria within the human gut microbiota. Trends Microbiol
Bewtra M, Knights D, Walters WA, Knight R, et al. Linking long-term 2015;23:354–66.
dietary patterns with gut microbial enterotypes. Science 2011;334:105– 132. Kristensen NB, Bryrup T, Allin KH, Nielsen T, Hansen TH,
8. Pedersen O. Alterations in fecal microbiota composition by probiotic
111. Fava F, Gitau R, Griffin BA, Gibson GR, Tuohy KM, Lovegrove JA. supplementation in healthy adults: a systematic review of randomized
The type and quantity of dietary fat and carbohydrate alter faecal controlled trials. Genome Med 2016;8:52.
microbiome and short-chain fatty acid excretion in a metabolic 133. Kellow NJ, Coughlan MT, Reid CM. Metabolic benefits of dietary
syndrome Ôat-riskÕ population. Int J Obes (Lond) 2013;37:216–23. prebiotics in human subjects: a systematic review of randomised
112. Pokusaeva K, Fitzgerald GF, van Sinderen D. Carbohydrate metabo- controlled trials. Br J Nutr 2014;111:1147–61.
lism in Bifidobacteria. Genes Nutr 2011;6:285–306. 134. Roberfroid M. Prebiotics: the concept revisited. J Nutr 2007;137

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


113. Bondue P, Delcenserie V. Genome of Bifidobacteria and carbohydrate (3 Suppl 2):830S–7S.
metabolism. Korean J Food Sci Anim Resour 2015;35:1–9. 135. Warren CA, Guerrant RL. Pathogenic C difficile is here (and every-
114. Holmes AJ, Chew YV, Colakoglu F, Cliff JB, Klaassens E, Read MN, where) to stay. Lancet 2011;377:8–9.
Solon-Biet SM, McMahon AC, Cogger VC, Ruohonen K, et al. Diet- 136. Macfarlane S. Antibiotic treatments and microbes in the gut. Environ
microbiome interactions in health are controlled by intestinal nitrogen Microbiol 2014;16:919–24.
source constraints. Cell Metab 2017;25:140–151.
137. Jepsen P, Skriver MV, Floyd A, Lipworth L, Schonheyder HC,
115. Suez J, Korem T, Zeevi D, Zilberman-Schapira G, Thaiss CA, Maza O, Sorensen HT. A population-based study of maternal use of amoxicillin
Israeli D, Zmora N, Gilad S, Weinberger A, et al. Artificial sweeteners and pregnancy outcome in Denmark. Br J Clin Pharmacol 2003;55:216–
induce glucose intolerance by altering the gut microbiota. Nature 21.
2014;514:181–6.
138. Vidal AC, Murphy SK, Murtha AP, Schildkraut JM, Soubry A,
116. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Huang Z, Neelon SE, Fuemmeler B, Iversen E, Wang F, et al. Asso-
Massart S, Collini S, Pieraccini G, Lionetti P. Impact of diet in shaping ciations between antibiotic exposure during pregnancy, birth weight
gut microbiota revealed by a comparative study in children from Eu- and aberrant methylation at imprinted genes among offspring. Int J
rope and rural Africa. Proc Natl Acad Sci USA 2010;107:14691–6. Obes (Lond) 2013;37:907–13.
117. Schnorr SL, Candela M, Rampelli S, Centanni M, Consolandi C,
139. Langdon A, Crook N, Dantas G. The effects of antibiotics on the
Basaglia G, Turroni S, Biagi E, Peano C, Severgnini M, et al. Gut mi-
microbiome throughout development and alternative approaches for
crobiome of the Hadza hunter-gatherers. Nat Commun 2014;5:3654.
therapeutic modulation. Genome Med 2016;8:39.
118. Rampelli S, Schnorr SL, Consolandi C, Turroni S, Severgnini M,
140. Sheridan PO, Bindels LB, Saulnier DM, Reid G, Nova E, Holmgren K,
Peano C, Brigidi P, Crittenden AN, Henry AG, Candela M. Meta-
OÕToole PW, Bunn J, Delzenne N, Scott KP. Can prebiotics and pro-
genome sequencing of the Hadza hunter-gatherer gut microbiota. Curr
biotics improve therapeutic outcomes for undernourished individuals?
Biol 2015;25:1682–93.
Gut Microbes 2014;5:74–82.
119. Hehemann JH, Correc G, Barbeyron T, Helbert W, Czjzek M,
141. Dock DB, Latorraca MQ, Aguilar-Nascimento JE, Gomes-da-
Michel G. Transfer of carbohydrate-active enzymes from marine
Silva MH. Probiotics enhance recovery from malnutrition and lessen
bacteria to Japanese gut microbiota. Nature 2010;464:908–12.
colonic mucosal atrophy after short-term fasting in rats. Nutrition
120. Kurokawa K, Itoh T, Kuwahara T, Oshima K, Toh H, Toyoda A, 2004;20:473–6.
Takami H, Morita H, Sharma VK, Srivastava TP, et al. Comparative
142. Gauffin CP, Aguero G, Perdigon G. Adjuvant effects of Lactobacillus
metagenomics revealed commonly enriched gene sets in human gut
casei added to a renutrition diet in a malnourished mouse model.
microbiomes. DNA Res 2007;14(4):169–81.
Biocell 2002;26(1):35–48.
121. Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK,
143. Dock DB, Aguilar-Nascimento JE, Latorraca MQ. Probiotics enhance
Wingreen NS, Sonnenburg JL. Diet-induced extinctions in the gut
the recovery of gut atrophy in experimental malnutrition. Biocell
microbiota compound over generations. Nature 2016;529:212–5.
2004;28(2):143–50.
122. Asher G, Sassone-Corsi P. Time for food: the intimate interplay be-
144. Ji B, Nielsen J. From next-generation sequencing to systematic
tween nutrition, metabolism, and the circadian clock. Cell
modeling of the gut microbiome. Front Genet 2015;6:219.
2015;161:84–92.
145. Foster JA, Lyte M, Meyer E, Cryan JF. Gut microbiota and brain
123. Kohsaka A, Laposky AD, Ramsey KM, Estrada C, Joshu C,
function: an evolving field in neuroscience. Int J Neuropsychopharmacol
Kobayashi Y, Turek FW, Bass J. High-fat diet disrupts behavioral and
2016;19(5)pyv114.
molecular circadian rhythms in mice. Cell Metab 2007;6:414–21.
146. Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour
124. Thaiss CA, Zeevi D, Levy M, Zilberman-Schapira G, Suez J,
–epigenetic regulation of the gut-brain axis. Genes Brain Behav
Tengeler AC, Abramson L, Katz MN, Korem T, Zmora N, et al.
2014;13:69–86.
Transkingdom control of microbiota diurnal oscillations promotes
metabolic homeostasis. Cell 2014;159:514–29. 147. Forsythe P, Bienenstock J, Kunze WA. Vagal pathways for microbiome-
125. Zarrinpar A, Chaix A, Yooseph S, Panda S. Diet and feeding pattern brain-gut axis communication. Adv Exp Med Biol 2014;817:115–33.
affect the diurnal dynamics of the gut microbiome. Cell Metab 148. Obata Y, Pachnis V. The effect of microbiota and the immune system
2014;20:1006–17. on the development and organization of the enteric nervous system.
126. Leone V, Gibbons SM, Martinez K, Hutchison AL, Huang EY, Gastroenterology 2016;151:836–44.
Cham CM, Pierre JF, Heneghan AF, Nadimpalli A, Hubert N, et al. 149. Wahlström A, Sayin SI, Marschall HU, Bäckhed F. Intestinal crosstalk
Effects of diurnal variation of gut microbes and high-fat feeding on between bile acids and microbiota and its impact on host metabolism.
host circadian clock function and metabolism. Cell Host Microbe Cell Metab 2016;24:41–50.
2015;17:681–9. 150. Bauer PV, Hamr SC, Duca FA. Regulation of energy balance by a gut-
127. Chaix A, Zarrinpar A, Miu P, Panda S. Time-restricted feeding is a brain axis and involvement of the gut microbiota. Cellular and
preventative and therapeutic intervention against diverse nutritional molecular life sciences. Cell Mol Life Sci 2016;73:737–55.
challenges. Cell Metab 2014;20:991–1005. 151. Rı́os-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M, de Los
128. Hatori M, Vollmers C, Zarrinpar A, DiTacchio L, Bushong EA, Gill S, Reyes-Gavilán CG, Salazar N. Intestinal short chain fatty acids and
Leblanc M, Chaix A, Joens M, Fitzpatrick JA, et al. Time-restricted their link with diet and human health. Front Microbiol 2016;7:185.
feeding without reducing caloric intake prevents metabolic diseases in 152. Macfarlane S, Macfarlane GT. Regulation of short-chain fatty acid
mice fed a high-fat diet. Cell Metab 2012;15:848–60. production. Proc Nutr Soc 2003;62:67–72.

740 van de Wouw et al.


153. de Aguiar Vallim TQ, Tarling EJ, Edwards PA. Pleiotropic roles of bile 174. Ullmer C, Alvarez Sanchez R, Sprecher U, Raab S, Mattei P,
acids in metabolism. Cell Metab 2013;17:657–69. Dehmlow H, Sewing S, Iglesias A, Beauchamp J, Conde-Knape K.
154. Barrett E, Ross RP, OÕToole PW, Fitzgerald GF, Stanton C. gamma- Systemic bile acid sensing by G protein-coupled bile acid receptor
Aminobutyric acid production by culturable bacteria from the human 1 (GPBAR1) promotes PYY and GLP-1 release. Br J Pharmacol
intestine. J Appl Microbiol 2012;113:411–7. 2013;169:671–84.
155. Thomas CM, Hong T, van Pijkeren JP, Hemarajata P, Trinh DV, Hu W, 175. Joyce SA, MacSharry J, Casey PG, Kinsella M, Murphy EF,
Britton RA, Kalkum M, Versalovic J. Histamine derived from probi- Shanahan F, Hill C, Gahan CG. Regulation of host weight gain and
otic Lactobacillus reuteri suppresses TNF via modulation of PKA and lipid metabolism by bacterial bile acid modification in the gut. Proc
ERK signaling. PLoS One 2012;7:e31951. Natl Acad Sci USA 2014;111:7421–6.
156. Hemarajata P, Spinler JK, Balderas MA, Versalovic J. Identification 176. Hegstrand LR, Hine RJ. Variations of brain histamine levels in germ-
of a proton-chloride antiporter (EriC) by Himar1 transposon muta- free and nephrectomized rats. Neurochem Res 1986;11:185–91.
genesis in Lactobacillus reuteri and its role in histamine production. 177. Matsumoto M, Kibe R, Ooga T, Aiba Y, Sawaki E, Koga Y, Benno Y.
Antonie van Leeuwenhoek 2014;105:579–92. Cerebral low-molecular metabolites influenced by intestinal
157. Girvin GT, Stevenson JW. Cell free choline acetylase from Lactoba- microbiota: a pilot study. Front Syst Neurosci 2013;7:9.
cillus plantarum. Can J Biochem Physiol 1954;32:131–46. 178. Desbonnet L, Garrett L, Clarke G, Bienenstock J, Dinan TG. The
158. Horiuchi Y, Kimura R, Kato N, Fujii T, Seki M, Endo T, Kato T, probiotic Bifidobacteria infantis: an assessment of potential antide-
Kawashima K. Evolutional study on acetylcholine expression. Life Sci pressant properties in the rat. J Psychiatr Res 2008;43:164–74.

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


2003;72:1745–56. 179. Voigt JP, Fink H. Serotonin controlling feeding and satiety. Behav
159. Williams BB, Van Benschoten AH, Cimermancic P, Donia MS, Brain Res 2015;277:14–31.
Zimmermann M, Taketani M, Ishihara A, Kashyap PC, Fraser JS, 180. Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, Deng Y,
Fischbach MA. Discovery and characterization of gut microbiota de- Blennerhassett P, Macri J, McCoy KD, et al. The intestinal microbiota
carboxylases that can produce the neurotransmitter tryptamine. Cell affect central levels of brain-derived neurotropic factor and behavior
Host Microbe 2014;16:495–503. in mice. Gastroenterology 2011;141(2):599–609, e1–3.
160. Asano Y, Hiramoto T, Nishino R, Aiba Y, Kimura T, Yoshihara K, 181. Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety-like
Koga Y, Sudo N. Critical role of gut microbiota in the production of behavior and central neurochemical change in germ-free mice.
biologically active, free catecholamines in the gut lumen of mice. Am J Neurogastroenterol Motil 2011;23(3):255–64, e119.
Physiol Gastrointest Liver Physiol 2012;303:G1288–95. 182. Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, Kubo C,
161. Eisenhofer G, Aneman A, Friberg P, Hooper D, Fandriks L, Lonroth H, Koga Y. Postnatal microbial colonization programs the hypothalamic-
Hunyady B, Mezey E. Substantial production of dopamine in the human pituitary-adrenal system for stress response in mice. J Physiol
gastrointestinal tract. J Clin Endocrinol Metab 1997;82:3864–71. 2004;558:263–75.
162. Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, Nagler CR, 183. Nicodemus KK, Elvevag B, Foltz PW, Rosenstein M, Diaz-Asper C,
Ismagilov RF, Mazmanian SK, Hsiao EY. Indigenous bacteria from the Weinberger DR. Category fluency, latent semantic analysis and
gut microbiota regulate host serotonin biosynthesis. Cell 2015; schizophrenia: a candidate gene approach. Cortex 2014;55:182–91.
161:264–76. 184. Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM,
163. Joyce SA, Gahan CG. Bile acid modifications at the microbe-host Dinan TG, Bienenstock J, Cryan JF. Ingestion of Lactobacillus strain
interface: potential for nutraceutical and pharmaceutical interventions regulates emotional behavior and central GABA receptor expression in a
in host health. Annu Rev Food Sci Technol 2016;7:313–33. mouse via the vagus nerve. Proc Natl Acad Sci USA 2011;108:16050–5.
164. Sayin SI, Wahlstrom A, Felin J, Jantti S, Marschall HU, Bamberg K, 185. Janik R, Thomason LA, Stanisz AM, Forsythe P, Bienenstock J,
Angelin B, Hyotylainen T, Oresic M, Backhed F. Gut microbiota Stanisz GJ. Magnetic resonance spectroscopy reveals oral Lactoba-
regulates bile acid metabolism by reducing the levels of tauro-beta- cillus promotion of increases in brain GABA, N-acetyl aspartate
muricholic acid, a naturally occurring FXR antagonist. Cell Metab and glutamate. Neuroimage 2016;125:988–95.
2013;17:225–35. 186. Xu Y, Tong Q. Expanding neurotransmitters in the hypothalamic neu-
165. Gustafsson BE, Bergstrom S, Lindstedt S, Norman A. Turnover and nature rocircuitry for energy balance regulation. Protein Cell 2011;2:800–13.
of fecal bile acids in germfree and infected rats fed cholic acid-24–14C; 187. Lyte M. Probiotics function mechanistically as delivery vehicles for
bile acids and steroids 41. Proc Soc Exp Biol Med 1957;94:467–71. neuroactive compounds: microbial endocrinology in the design and
166. Riottot M, Sacquet E. Increase in the ileal absorption rate of sodium use of probiotics. BioEssays 2011;33:574–81.
taurocholate in germ-free or conventional rats given an amylomaize- 188. Lyte M. Microbial endocrinology: host-microbiota neuroendocrine
starch diet. Br J Nutr 1985;53:307–10. interactions influencing brain and behavior. Gut Microbes 2014;
167. Sinal CJ, Tohkin M, Miyata M, Ward JM, Lambert G, Gonzalez FJ. 5:381–9.
Targeted disruption of the nuclear receptor FXR/BAR impairs bile 189. Rahat-Rozenbloom S, Fernandes J, Gloor GB, Wolever TM. Evidence
acid and lipid homeostasis. Cell 2000;102:731–44. for greater production of colonic short-chain fatty acids in overweight
168. Li F, Jiang C, Krausz KW, Li Y, Albert I, Hao H, Fabre KM, than lean humans. Int J Obes (Lond) 2014;38:1525–31.
Mitchell JB, Patterson AD, Gonzalez FJ. Microbiome remodelling 190. Fernandes J, Su W, Rahat-Rozenbloom S, Wolever TM, Comelli EM.
leads to inhibition of intestinal farnesoid X receptor signalling and Adiposity, gut microbiota and faecal short chain fatty acids are linked
decreased obesity. Nat Commun 2013;4:2384. in adult humans. Nutr Diabetes 2014;4:e121.
169. Parséus A, Sommer N, Sommer F, Caesar R, Molinaro A, Ståhlman M, 191. Murugesan S, Ulloa-Martinez M, Martinez-Rojano H, Galvan-
Greiner TU, Perkins R, Bäckhed F. Microbiota-induced obesity Rodriguez FM, Miranda-Brito C, Romano MC, Pina-Escobedo A,
requires farnesoid X receptor. Gut 2017;66:429–37. Pizano-Zarate ML, Hoyo-Vadillo C, Garcia-Mena J. Study of the di-
170. Jiang C, Xie C, Li F, Zhang L, Nichols RG, Krausz KW, Cai J, Qi Y, versity and short-chain fatty acids production by the bacterial
Fang ZZ, Takahashi S, et al. Intestinal farnesoid X receptor signaling community in overweight and obese Mexican children. Eur J Clin
promotes nonalcoholic fatty liver disease. J Clin Invest 2015;125:386–402. Microbiol Infect Dis 2015;34:1337–46.
171. Swann JR, Want EJ, Geier FM, Spagou K, Wilson ID, Sidaway JE, 192. McNeil NI. The contribution of the large intestine to energy supplies
Nicholson JK, Holmes E. Systemic gut microbial modulation of bile in man. Am J Clin Nutr 1984;39:338–42.
acid metabolism in host tissue compartments. Proc Natl Acad Sci USA 193. Chambers ES, Viardot A, Psichas A, Morrison DJ, Murphy KG, Zac-
2011;108 Suppl 1:4523–30. Varghese SE, MacDougall K, Preston T, Tedford C, Finlayson GS,
172. Prawitt J, Abdelkarim M, Stroeve JH, Popescu I, Duez H, et al. Effects of targeted delivery of propionate to the human colon on
Velagapudi VR, Dumont J, Bouchaert E, van Dijk TH, Lucas A, et al. appetite regulation, body weight maintenance and adiposity in over-
Farnesoid X receptor deficiency improves glucose homeostasis in weight adults. Gut 2015;64:1744–54.
mouse models of obesity. Diabetes 2011;60:1861–71. 194. Lin HV, Frassetto A, Kowalik EJ Jr., Nawrocki AR, Lu MM,
173. Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, Kosinski JR, Hubert JA, Szeto D, Yao X, Forrest G, et al. Butyrate and
Macchiarulo A, Yamamoto H, Mataki C, Pruzanski M, et al. TGR5- propionate protect against diet-induced obesity and regulate gut
mediated bile acid sensing controls glucose homeostasis. Cell Metab hormones via free fatty acid receptor 3-independent mechanisms.
2009;10:167–77. PLoS One 2012;7:e35240.

Microbiota and eating behavior 741


195. Vinolo MA, Rodrigues HG, Festuccia WT, Crisma AR, Alves VS, 217. Fukumoto S, Tatewaki M, Yamada T, Fujimiya M, Mantyh C,
Martins AR, Amaral CL, Fiamoncini J, Hirabara SM, Sato FT, et al. Voss M, Eubanks S, Harris M, Pappas TN, Takahashi T. Short-chain
Tributyrin attenuates obesity-associated inflammation and insulin fatty acids stimulate colonic transit via intraluminal 5-HT release in
resistance in high-fat-fed mice. Am J Physiol Endocrinol Metab rats. Am J Physiol Regul Integr Comp Physiol 2003;284:R1269–76.
2012;303:E272–82. 218. Grider JR, Piland BE. The peristaltic reflex induced by short-chain
196. Lu Y, Fan C, Li P, Lu Y, Chang X, Qi K. Short chain fatty acids prevent fatty acids is mediated by sequential release of 5-HT and neuronal
high-fat-diet-induced obesity in mice by regulating G protein-coupled CGRP but not BDNF. Am J Physiol Gastrointest Liver Physiol
receptors and gut microbiota. Sci Rep 2016;6:37589. 2007;292:G429–37.
197. Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms 219. Reigstad CS, Salmonson CE, Rainey JF 3rd, Szurszewski JH,
underlying the resistance to diet-induced obesity in germ-free mice. Linden DR, Sonnenburg JL, Farrugia G, Kashyap PC. Gut microbes
Proc Natl Acad Sci USA 2007;104:979–84. promote colonic serotonin production through an effect of short-chain
198. Høverstad T, Midtvedt T. Short-chain fatty acids in germfree mice and fatty acids on enterochromaffin cells. FASEB J 2015;29:1395–403.
rats. J Nutr 1986;116:1772–6. 220. Dumoulin V, Moro F, Barcelo A, Dakka T, Cuber JC. Peptide YY,
199. Rabot S, Membrez M, Bruneau A, Gerard P, Harach T, Moser M, glucagon-like peptide-1, and neurotensin responses to luminal factors in
Raymond F, Mansourian R, Chou CJ. Germ-free C57BL/6J mice are the isolated vascularly perfused rat ileum. Endocrinology 1998;139:3780–
resistant to high-fat-diet-induced insulin resistance and have altered 6.
cholesterol metabolism. FASEB J 2010;24:4948–59. 221. Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM,

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


200. Wong JM, de Souza R, Kendall CW, Emam A, Jenkins DJ. Colonic Diakogiannaki E, Cameron J, Grosse J, Reimann F, Gribble FM.
health: fermentation and short chain fatty acids. J Clin Gastroenterol Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via
2006;40:235–43. the G-protein-coupled receptor FFAR2. Diabetes 2012;61:364–71.
201. Roediger WE. Utilization of nutrients by isolated epithelial cells of the 222. Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA,
rat colon. Gastroenterology 1982;83:424–9. Hanyaloglu AC, Ghatei MA, Bloom SR, Frost G. The short chain fatty
acid propionate stimulates GLP-1 and PYY secretion via free fatty acid
202. Clausen MR, Mortensen PB. Kinetic studies on colonocyte metabo-
receptor 2 in rodents. Int J Obes (Lond) 2015;39:424–9.
lism of short chain fatty acids and glucose in ulcerative colitis. Gut
1995;37:684–9. 223. Zaibi MS, Stocker CJ, OÕDowd J, Davies A, Bellahcene M,
Cawthorne MA, Brown AJ, Smith DM, Arch JR. Roles of GPR41 and
203. Jørgensen JR, Clausen MR, Mortensen PB. Oxidation of short and
GPR43 in leptin secretory responses of murine adipocytes to short
medium chain C2–C8 fatty acids in sprague-dawley rat colonocytes.
chain fatty acids. FEBS Lett 2010;584:2381–6.
Gut 1997;40:400–5.
224. Hong YH, Nishimura Y, Hishikawa D, Tsuzuki H, Miyahara H,
204. Brass EP, Beyerinck RA. Effects of propionate and carnitine on the
Gotoh C, Choi KC, Feng DD, Chen C, Lee HG, et al. Acetate and
hepatic oxidation of short- and medium-chain-length fatty acids.
propionate short chain fatty acids stimulate adipogenesis via GPCR43.
Biochem J 1988;250:819–25.
Endocrinology 2005;146:5092–9.
205. Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT.
225. Xiong Y, Miyamoto N, Shibata K, Valasek MA, Motoike T,
Short chain fatty acids in human large intestine, portal, hepatic and
Kedzierski RM, Yanagisawa M. Short-chain fatty acids stimulate
venous blood. Gut 1987;28:1221–7. leptin production in adipocytes through the G protein-coupled
206. Bloemen JG, Venema K, van de Poll MC, Olde Damink SW, receptor GPR41. Proc Natl Acad Sci USA 2004;101:1045–50.
Buurman WA, Dejong CH. Short chain fatty acids exchange across the 226. Al-Lahham SH, Roelofsen H, Priebe M, Weening D, Dijkstra M,
gut and liver in humans measured at surgery. Clin Nutr 2009;28:657– Hoek A, Rezaee F, Venema K, Vonk RJ. Regulation of adipokine
61. production in human adipose tissue by propionic acid. Eur J Clin
207. Bloemen JG, Olde Damink SW, Venema K, Buurman WA, Jalan R, Invest 2010;40:401–7.
Dejong CH. Short chain fatty acids exchange: is the cirrhotic, 227. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S,
dysfunctional liver still able to clear them? Clin Nutr 2010; Kobayashi M, Hirasawa A, Tsujimoto G. Short-chain fatty acids and
29:365–9. ketones directly regulate sympathetic nervous system via G protein-
208. Boets E, Gomand SV, Deroover L, Preston T, Vermeulen K, De coupled receptor 41 (GPR41). Proc Natl Acad Sci USA 2011;108:8030–5.
Preter V, Hamer HM, Van den Mooter G, De Vuyst L, Courtin CM, 228. Kimura I, Ozawa K, Inoue D, Imamura T, Kimura K, Maeda T,
et al. Systemic availability and metabolism of colonic-derived short- Terasawa K, Kashihara D, Hirano K, Tani T, et al. The gut microbiota
chain fatty acids in healthy subjects: a stable isotope study. J Physiol suppresses insulin-mediated fat accumulation via the short-chain fatty
2017;595:541–55. acid receptor GPR43. Nat Commun 2013;4:1829.
209. Krautkramer KA, Kreznar JH, Romano KA, Vivas EI, Barrett- 229. Kaji I, Akiba Y, Konno K, Watanabe M, Kimura S, Iwanaga T, Kuri A,
Wilt GA, Rabaglia ME, Keller MP, Attie AD, Rey FE, Denu JM. Diet- Iwamoto K, Kuwahara A, Kaunitz JD. Neural FFA3 activation in-
microbiota interactions mediate global epigenetic programming in versely regulates anion secretion evoked by nicotinic ACh receptor
multiple host tissues. Mol Cell 2016;64:982–92. activation in rat proximal colon. J Physiol 2016;594:3339–52.
210. Görisch SM, Wachsmuth M, Tóth KF, Lichter P, Rippe K. Histone 230. De Vadder F, Kovatcheva-Datchary P, Goncalves D, Vinera J,
acetylation increases chromatin accessibility. J Cell Sci 2005;118:5825– Zitoun C, Duchampt A, Backhed F, Mithieux G. Microbiota-
34. generated metabolites promote metabolic benefits via gut-brain neural
211. Waldecker M, Kautenburger T, Daumann H, Busch C, Schrenk D. circuits. Cell 2014;156:84–96.
Inhibition of histone-deacetylase activity by short-chain fatty acids 231. Nøhr MK, Egerod KL, Christiansen SH, Gille A, Offermanns S,
and some polyphenol metabolites formed in the colon. J Nutr Biochem Schwartz TW, Møller M. Expression of the short chain fatty acid
2008;19:587–93. receptor GPR41/FFAR3 in autonomic and somatic sensory ganglia.
212. Soliman ML, Rosenberger TA. Acetate supplementation increases Neuroscience 2015;290:126–37.
brain histone acetylation and inhibits histone deacetylase activity and 232. Soret R, Chevalier J, De Coppet P, Poupeau G, Derkinderen P,
expression. Mol Cell Biochem 2011;352:173–80. Segain JP, Neunlist M. Short-chain fatty acids regulate the enteric
213. Reisenauer CJ, Bhatt DP, Mitteness DJ, Slanczka ER, Gienger HM, neurons and control gastrointestinal motility in rats. Gastroenterology
Watt JA, Rosenberger TA. Acetate supplementation attenuates 2010;138:1772–82.
lipopolysaccharide-induced neuroinflammation. J Neurochem 233. Kim CH, Park J, Kim M. Gut microbiota-derived short-chain fatty
2011;117:264–74. acids, T cells, and inflammation. Immune Netw 2014;14:277–88.
214. Davie JR. Inhibition of histone deacetylase activity by butyrate. J Nutr 234. Corrêa-Oliveira R, Fachi JL, Vieira A, Sato FT, Vinolo MA. Regula-
2003;133(7 Suppl)2485S–93S. tion of immune cell function by short-chain fatty acids. Clin Transl
215. Fujino T, Kondo J, Ishikawa M, Morikawa K, Yamamoto TT. Immunology 2016;5:e73.
Acetyl-CoA synthetase 2, a mitochondrial matrix enzyme involved 235. Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME,
in the oxidation of acetate. J Biol Chem 2001;276:11420–6. Brezillon S, Dupriez V, Vassart G, Van Damme J, et al. Functional
216. Dennis PB, Jaeschke A, Saitoh M, Fowler B, Kozma SC, Thomas G. characterization of human receptors for short chain fatty acids
Mammalian TOR: a homeostatic ATP sensor. Science 2001;294: and their role in polymorphonuclear cell activation. J Biol Chem
1102–5. 2003;278:25481–9.

742 van de Wouw et al.


236. Vinolo MA, Ferguson GJ, Kulkarni S, Damoulakis G, Anderson K, 255. Perry RJ, Peng L, Barry NA, Cline GW, Zhang D, Cardone RL,
Bohlooly YM, Stephens L, Hawkins PT, Curi R. SCFAs induce mouse Petersen KF, Kibbey RG, Goodman AL, Shulman GI. Acetate
neutrophil chemotaxis through the GPR43 receptor. PLoS One mediates a microbiome-brain-beta-cell axis to promote metabolic
2011;6:e21205. syndrome. Nature 2016;534:213–7.
237. Vieira AT, Galvao I, Macia LM, Sernaglia EM, Vinolo MA, Garcia CC, 256. Trent CM, Blaser MJ. Microbially produced acetate: a ‘‘missing link’’
Tavares LP, Amaral FA, Sousa LP, Martins FS, et al. Dietary fiber and the in understanding obesity? Cell Metab 2016;24:9–10.
short-chain fatty acid acetate promote resolution of neutrophilic inflam- 257. Bindels LB, Leclercq I. Colonic acetate in obesity: location matters!
mation in a model of gout in mice. J Leukoc Biol 2017;101:275–84. Clin Sci (Lond) 2016;130:2083–6.
238. Park JS, Lee EJ, Lee JC, Kim WK, Kim HS. Anti-inflammatory effects 258. Scotter MJ, Thorpe SA, Reynolds SL, Wilson LA, Strutt PR. Survey of
of short chain fatty acids in IFN-gamma-stimulated RAW 264.7 baked goods for propionic acid and propionates. Food Addit Contam
murine macrophage cells: involvement of NF-kappaB and ERK 1996;13:133–9.
signaling pathways. Int Immunopharmacol 2007;7:70–7.
259. Brock M, Buckel W. On the mechanism of action of the antifungal
239. Nastasi C, Candela M, Bonefeld CM, Geisler C, Hansen M,
agent propionate. Eur J Biochem 2004;271:3227–41.
Krejsgaard T, Biagi E, Andersen MH, Brigidi P, Odum N, et al. The
effect of short-chain fatty acids on human monocyte-derived dendritic 260. Scott KP, Martin JC, Campbell G, Mayer CD, Flint HJ. Whole-
cells. Sci Rep 2015;5:16148. genome transcription profiling reveals genes up-regulated by growth
on fucose in the human gut bacterium ‘‘Roseburia inulinivorans.’’ J
240. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA,
Bacteriol 2006;188:4340–9.

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


Bohlooly YM, Glickman JN, Garrett WS. The microbial metabolites,
short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 261. Hetzel M, Brock M, Selmer T, Pierik AJ, Golding BT, Buckel W.
2013;341:569–73. Acryloyl-CoA reductase from Clostridium propionicum. An enzyme
complex of propionyl-CoA dehydrogenase and electron-transferring
241. Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson J,
Kim CH. Short-chain fatty acids induce both effector and regulatory flavoprotein. Eur J Biochem 2003;270:902–10.
T cells by suppression of histone deacetylases and regulation of the 262. Polyviou T, MacDougall K, Chambers ES, Viardot A, Psichas A,
mTOR-S6K pathway. Mucosal Immunol 2015;8:80–93. Jawaid S, Harris HC, Edwards CA, Simpson L, Murphy KG, et al.
242. Asarat M, Apostolopoulos V, Vasiljevic T, Donkor O. Short-chain Randomised clinical study: inulin short-chain fatty acid esters for
fatty acids regulate cytokines and Th17/Treg cells in human peripheral targeted delivery of short-chain fatty acids to the human colon.
blood mononuclear cells in vitro. Immunol Invest 2016;45:205–22. Aliment Pharmacol Ther 2016;44:662–72.
243. Tan J, McKenzie C, Vuillermin PJ, Goverse G, Vinuesa CG, 263. Byrne CS, Chambers ES, Alhabeeb H, Chhina N, Morrison DJ,
Mebius RE, Macia L, Mackay CR. Dietary fiber and bacterial SCFA Preston T, Tedford C, Fitzpatrick J, Irani C, Busza A, et al. Increased
enhance oral tolerance and protect against food allergy through di- colonic propionate reduces anticipatory reward responses in the hu-
verse cellular pathways. Cell Reports 2016;15:2809–24. man striatum to high-energy foods. Am J Clin Nutr 2016;104:5–14.
244. Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, 264. Reineccius GH. Flavor chemistry and technology. 2nd ed. Boca Raton
David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, et al. (FL): Taylor & Francis; 2006.
Host microbiota constantly control maturation and function of mi- 265. Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG,
croglia in the CNS. Nat Neurosci 2015;18:965–77. Cryan JF. The neuropharmacology of butyrate: the bread and butter of
245. Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Toth M, the microbiota-gut-brain axis? Neurochem Int 2016;99:110–32.
Korecka A, Bakocevic N, Ng LG, Kundu P, et al. The gut microbiota 266. Louis P, Duncan SH, McCrae SI, Millar J, Jackson MS, Flint HJ. Restricted
influences blood-brain barrier permeability in mice. Sci Transl Med distribution of the butyrate kinase pathway among butyrate-producing
2014;6:263ra158. Erratum in: Sci Transl Med 2014;6:266er7. bacteria from the human colon. J Bacteriol 2004;186:2099–106.
246. Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, 267. Caricilli AM, Castoldi A, Camara NO. Intestinal barrier: a gentle-
Brunet I, Wan LX, Rey F, Wang T, et al. Olfactory receptor responding menÕs agreement between microbiota and immunity. World J Gastro-
to gut microbiota-derived signals plays a role in renin secretion and intest Pathophysiol 2014;5:18–32.
blood pressure regulation. Proc Natl Acad Sci USA 2013;110:4410–5. 268. Farhadi A, Banan A, Fields J, Keshavarzian A. Intestinal barrier: an
247. Thangaraju M, Cresci GA, Liu K, Ananth S, Gnanaprakasam JP, interface between health and disease. J Gastroenterol Hepatol
Browning DD, Mellinger JD, Smith SB, Digby GJ, Lambert NA, et al. 2003;18:479–97.
GPR109A is a G-protein-coupled receptor for the bacterial fermen-
269. Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP.
tation product butyrate and functions as a tumor suppressor in colon.
Breaking down the barriers: the gut microbiome, intestinal permeability
Cancer Res 2009;69:2826–32.
and stress-related psychiatric disorders. Front Cell Neurosci 2015;9:392.
248. Mascord D, Smith J, Starmer GA, Whitfield JB. Effects of increasing
270. Macpherson AJ, Harris NL. Interactions between commensal intestinal
the rate of alcohol metabolism on plasma acetate concentration. Al-
bacteria and the immune system. Nat Rev Immunol 2004;4:478–85.
cohol Alcohol 1992;27:25–8.
271. Andrade ME, Araujo RS, de Barros PA, Soares AD, Abrantes FA,
249. Ragsdale SW, Pierce E. Acetogenesis and the Wood-Ljungdahl pathway
Generoso Sde V, Fernandes SO, Cardoso VN. The role of immuno-
of CO(2) fixation. Biochim Biophys Acta 2008;1784:1873–98.
modulators on intestinal barrier homeostasis in experimental models.
250. De Vuyst L, Leroy F. Cross-feeding between bifidobacteria and Clin Nutr 2015;34:1080–7.
butyrate-producing colon bacteria explains bifdobacterial competi-
tiveness, butyrate production, and gas production. Int J Food 272. Monteleone P, Carratu R, Carteni M, Generoso M, Lamberti M,
Microbiol 2011;149:73–80. Magistris LD, Brambilla F, Colurcio B, Secondulfo M, Maj M. Intestinal
permeability is decreased in anorexia nervosa. Mol Psychiatry 2004;9:76–80.
251. Rivière A, Gagnon M, Weckx S, Roy D, De Vuyst L. Mutual cross-
feeding interactions between Bifidobacterium longum subsp. longum 273. Lam YY, Ha CW, Campbell CR, Mitchell AJ, Dinudom A,
NCC2705 and Eubacterium rectale ATCC 33656 explain the Oscarsson J, Cook DI, Hunt NH, Caterson ID, Holmes AJ, et al. In-
bifidogenic and butyrogenic effects of arabinoxylan oligosaccharides. creased gut permeability and microbiota change associate with mes-
Appl Environ Microbiol 2015;81:7767–81. enteric fat inflammation and metabolic dysfunction in diet-induced
obese mice. PLoS One 2012;7:e34233.
252. Ge H, Li X, Weiszmann J, Wang P, Baribault H, Chen JL, Tian H, Li Y.
Activation of G protein-coupled receptor 43 in adipocytes leads to 274. Brun P, Castagliuolo I, Di Leo V, Buda A, Pinzani M, Palu G,
inhibition of lipolysis and suppression of plasma free fatty acids. Martines D. Increased intestinal permeability in obese mice: new ev-
Endocrinology 2008;149:4519–26. idence in the pathogenesis of nonalcoholic steatohepatitis. Am J
253. Frost G, Sleeth ML, Sahuri-Arisoylu M, Lizarbe B, Cerdan S, Brody L, Physiol Gastrointest Liver Physiol 2007;292:G518–25.
Anastasovska J, Ghourab S, Hankir M, Zhang S, et al. The short-chain 275. Genton L, Cani PD, Schrenzel J. Alterations of gut barrier and gut
fatty acid acetate reduces appetite via a central homeostatic mecha- microbiota in food restriction, food deprivation and protein-energy
nism. Nat Commun 2014;5:3611. wasting. Clin Nutr 2015;34:341–9.
254. van der Beek CM, Canfora EE, Lenaerts K, Troost FJ, Olde Damink SW, 276. Tong LC, Wang Y, Wang ZB, Liu WY, Sun S, Li L, Su DF, Zhang LC.
Holst JJ, Masclee AA, Dejong CH, Blaak EE. Distal, not proximal, colonic Propionate ameliorates dextran sodium sulfate-induced colitis by
acetate infusions promote fat oxidation and improve metabolic markers in improving intestinal barrier function and reducing inflammation and
overweight/obese men. Clin Sci (Lond) 2016;130:2073–82. oxidative stress. Front Pharmacol 2016;7:253.

Microbiota and eating behavior 743


277. Simeoli R, Mattace Raso G, Pirozzi C, Lama A, Santoro A, Russo R, 296. Szalay C, Abraham I, Papp S, Takacs G, Lukats B, Gati A, Karadi Z.
Montero-Melendez T, Berni Canani R, Calignano A, Perretti M, et al. Taste reactivity deficit in anorexia nervosa. Psychiatry Clin Neurosci
An orally administered butyrate-releasing derivative reduces neutro- 2010;64:403–7.
phil recruitment and inflammation in dextran sulphate sodium- 297. Dazzi F, Nitto SD, Zambetti G, Loriedo C, Ciofalo A. Alterations of
induced murine colitis. Br J Pharmacol 2016 Sep 29 (Epub ahead of the olfactory-gustatory functions in patients with eating disorders. Eur
print; DOI: 10.1111/bph.13637). Eat Disord Rev 2013;21:382–5.
278. Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, 298. Aschenbrenner K, Scholze N, Joraschky P, Hummel T. Gustatory and
Wilson KE, Glover LE, Kominsky DJ, Magnuson A, et al. Crosstalk olfactory sensitivity in patients with anorexia and bulimia in the
between microbiota-derived short-chain fatty acids and intestinal course of treatment. J Psychiatr Res 2008;43:129–37.
epithelial HIF augments tissue barrier function. Cell Host Microbe
299. Nozoe S, Masuda A, Naruo T, Soejima Y, Nagai N, Tanaka H.
2015;17:662–71.
Changes in taste responsiveness in patients with anorexia nervosa
279. Fain JN. Release of inflammatory mediators by human adipose tissue during behavior therapy. Physiol Behav 1996;59:549–53.
is enhanced in obesity and primarily by the nonfat cells: a review.
Mediators Inflamm 2010;2010:513948. 300. Rolls ET. Taste, olfactory, and food reward value processing in the
brain. Prog Neurobiol 2015;127–128:64–90.
280. Lassenius MI, Pietilainen KH, Kaartinen K, Pussinen PJ, Syrjanen J,
Forsblom C, Porsti I, Rissanen A, Kaprio J, Mustonen J, et al. Bacterial 301. Breslin PA, Huang L. Human taste: peripheral anatomy, taste trans-
endotoxin activity in human serum is associated with dyslipidemia, duction, and coding. Adv Otorhinolaryngol 2006;63:152–90.
302. Acosta A, Hurtado MD, Gorbatyuk O, La Sala M, Duncan D,

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


insulin resistance, obesity, and chronic inflammation. Diabetes Care
2011;34:1809–15. Aslanidi G, Campbell-Thompson M, Zhang L, Herzog H, Voutetakis A,
281. Hersoug LG, Møller P, Loft S. Gut microbiota-derived lipopolysac- et al. Salivary PYY: a putative bypass to satiety. PLoS One 2011;6:
charide uptake and trafficking to adipose tissue: implications for e26137.
inflammation and obesity. Obes Rev 2016;17:297–312. 303. Feng XH, Liu XM, Zhou LH, Wang J, Liu GD. Expression of
282. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin glucagon-like peptide-1 in the taste buds of rat circumvallate papillae.
Invest 2005;115:1111–9. Acta Histochem 2008;110:151–4.
283. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, 304. Shen T, Kaya N, Zhao FL, Lu SG, Cao Y, Herness S. Co-expression
Neyrinck AM, Fava F, Tuohy KM, Chabo C, et al. Metabolic endo- patterns of the neuropeptides vasoactive intestinal peptide and
toxemia initiates obesity and insulin resistance. Diabetes cholecystokinin with the transduction molecules alpha-gustducin and
2007;56:1761–72. T1R2 in rat taste receptor cells. Neuroscience 2005;130:229–38.
284. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, 305. Shin YK, Martin B, Golden E, Dotson CD, Maudsley S, Kim W,
Delzenne NM, Burcelin R. Changes in gut microbiota control Jang HJ, Mattson MP, Drucker DJ, Egan JM, et al. Modulation of
metabolic endotoxemia-induced inflammation in high-fat diet- taste sensitivity by GLP-1 signaling. J Neurochem 2008;106:455–63.
induced obesity and diabetes in mice. Diabetes 2008;57:1470–81. 306. La Sala MS, Hurtado MD, Brown AR, Bohorquez DV, Liddle RA,
285. Oldstone MB. Molecular mimicry: its evolution from concept to Herzog H, Zolotukhin S, Dotson CD. Modulation of taste responsive-
mechanism as a cause of autoimmune diseases. Monoclon Antib Im- ness by the satiation hormone peptide YY. FASEB J 2013;27:5022–33.
munodiagn Immunother 2014;33:158–65. 307. Wang R, van Keeken NM, Siddiqui S, Dijksman LM, Maudsley S,
286. Fetissov SO, Hamze Sinno M, Coeffier M, Bole-Feysot C, Ducrotte P, Derval D, van Dam PS, Martin B. Higher TNF-alpha, IGF-1, and
Hokfelt T, Dechelotte P. Autoantibodies against appetite-regulating leptin levels are found in tasters than non-tasters. Front Endocrinol
peptide hormones and neuropeptides: putative modulation by gut (Lausanne) 2014;5:125.
microflora. Nutrition 2008;24:348–59. 308. Lyte M, Fodor AA, Chapman CD, Martin GG, Perez-Chanona E,
287. Ericson MD, Schnell SM, Freeman KT, Haskell-Luevano C. A frag- Jobin C, Dess NK. Gut microbiota and a selectively bred taste
ment of the Escherichia coli ClpB heat-shock protein is a micromolar phenotype: a novel model of microbiome-behavior relationships.
melanocortin 1 receptor agonist. Bioorg Med Chem Lett Psychosom Med 2016;78:610–9.
2015;25:5306–8. 309. Swartz TD, Duca FA, de Wouters T, Sakar Y, Covasa M. Up-
288. Takagi K, Legrand R, Asakawa A, Amitani H, Francois M, Tennoune N, regulation of intestinal type 1 taste receptor 3 and sodium glucose
Coeffier M, Claeyssens S, do Rego JC, Dechelotte P, et al. Anti-ghrelin luminal transporter-1 expression and increased sucrose intake in mice
immunoglobulins modulate ghrelin stability and its orexigenic effect in lacking gut microbiota. Br J Nutr 2012;107:621–30.
obese mice and humans. Nat Commun 2013;4:2685. 310. Duca FA, Swartz TD, Sakar Y, Covasa M. Increased oral detection,
289. Coquerel Q, Sinno MH, Boukhettala N, Coeffier M, Terashi M, but decreased intestinal signaling for fats in mice lacking gut micro-
Bole-Feysot C, Breuille D, Dechelotte P, Fetissov SO. Intestinal inflam- biota. PLoS One 2012;7:e39748.
mation influences alpha-MSH reactive autoantibodies: relevance to food
311. Oakley B, Witt M. Building sensory receptors on the tongue. J Neu-
intake and body weight. Psychoneuroendocrinology 2012;37:94–106.
rocytol 2004;33:631–46.
290. Tennoune N, Chan P, Breton J, Legrand R, Chabane YN,
312. Feng P, Huang L, Wang H. Taste bud homeostasis in health, disease,
Akkermann K, Jarv A, Ouelaa W, Takagi K, Ghouzali I, et al. Bacterial
and aging. Chem Senses 2014;39:3–16.
ClpB heat-shock protein, an antigen-mimetic of the anorexigenic
peptide alpha-MSH, at the origin of eating disorders. Transl Psychi- 313. Cohn ZJ, Kim A, Huang L, Brand J, Wang H. Lipopolysaccharide-
atry 2014;4:e458. induced inflammation attenuates taste progenitor cell proliferation and
shortens the life span of taste bud cells. BMC Neurosci 2010;11:72.
291. Lucas N, Legrand R, Ouelaa W, Breton J, Tennoune N, Bole-Feysot C,
Dechelotte P, Fetissov SO. Effects of rabbit anti-alpha-melanocyte- 314. Wang H, Zhou M, Brand J, Huang L. Inflammation activates the inter-
stimulating hormone (alpha-MSH) immunoglobulins on alpha-MSH feron signaling pathways in taste bud cells. J Neurosci 2007;27:10703–
signaling related to food intake control. Neuropeptides 2014;48:21–7. 13.
292. Tennoune N, Legrand R, Ouelaa W, Breton J, Lucas N, Bole-Feysot C, 315. Kim A, Feng P, Ohkuri T, Sauers D, Cohn ZJ, Chai J, Nelson T,
do Rego JC, Dechelotte P, Fetissov SO. Sex-related effects of Bachmanov AA, Huang L, Wang H. Defects in the peripheral taste
nutritional supplementation of Escherichia coli: relevance to eating structure and function in the MRL/lpr mouse model of autoimmune
disorders. Nutrition 2015;31:498–507. disease. PLoS One 2012;7:e35588.
293. Breton J, Tennoune N, Lucas N, Francois M, Legrand R, Jacquemot J, 316. Wang H, Zhou M, Brand J, Huang L. Inflammation and taste disor-
Goichon A, Guerin C, Peltier J, Pestel-Caron M, et al. Gut commensal ders: mechanisms in taste buds. Ann N Y Acad Sci 2009;1170:596–
E. coli proteins activate host satiety pathways following nutrient- 603.
induced bacterial growth. Cell Metab 2016;23:324–34. 317. Feng P, Chai J, Zhou M, Simon N, Huang L, Wang H. Interleukin-10
294. Breton J, Legrand R, Akkermann K, Jarv A, Harro J, Dechelotte P, is produced by a specific subset of taste receptor cells and critical for
Fetissov SO. Elevated plasma concentrations of bacterial ClpB protein maintaining structural integrity of mouse taste buds. J Neurosci
in patients with eating disorders. Int J Eat Disord 2016;49:805–8. 2014;34:2689–701.
295. Berthoud HR, Zheng H. Modulation of taste responsiveness and food 318. Feng P, Jyotaki M, Kim A, Chai J, Simon N, Zhou M, Bachmanov AA,
preference by obesity and weight loss. Physiol Behav 2012;107:527– Huang L, Wang H. Regulation of bitter taste responses by tumor ne-
32. crosis factor. Brain Behav Immun 2015;49:32–42.

744 van de Wouw et al.


319. Aubert A, Dantzer R. The taste of sickness: lipopolysaccharide- 329. Kiraly DD, Walker DM, Calipari ES, Labonte B, Issler O, Pena CJ,
induced finickiness in rats. Physiol Behav 2005;84:437–44. Ribeiro EA, Russo SJ, Nestler EJ. Alterations of the host microbiome
320. Larson SJ. Lipopolysaccharide and interleukin-1beta decrease sucrose affect behavioral responses to cocaine. Sci Rep 2016;6:35455.
intake but do not affect expression of place preference in rats. 330. Leclercq S, Matamoros S, Cani PD, Neyrinck AM, Jamar F, Starkel P,
Pharmacol Biochem Behav 2006;84:429–35. Windey K, Tremaroli V, Backhed F, Verbeke K, et al. Intestinal per-
321. Cross-Mellor SK, Kavaliers M, Ossenkopp KP. The effects of lipo- meability, gut-bacterial dysbiosis, and behavioral markers of alcohol-
polysaccharide and lithium chloride on the ingestion of a bitter-sweet dependence severity. Proc Natl Acad Sci USA 2014;111:E4485–93.
taste: comparing intake and palatability. Brain Behav Immun 331. Cluny NL, Keenan CM, Reimer RA, Le Foll B, Sharkey KA. Preven-
2005;19:564–73.
tion of diet-induced obesity effects on body weight and gut microbiota
322. Feng P, Zhao H, Chai J, Huang L, Wang H. Expression and secretion in mice treated chronically with Delta9-Tetrahydrocannabinol. PLoS
of TNF-alpha in mouse taste buds: a novel function of a specific subset
One 2015;10:e0144270.
of type II taste cells. PLoS One 2012;7:e43140.
332. Dinan TG, Stilling RM, Stanton C, Cryan JF. Collective uncon-
323. Kumarhia D, He L, McCluskey LP. Inflammatory stimuli acutely
scious: how gut microbes shape human behavior. J Psychiatr Res
modulate peripheral taste function. J Neurophysiol 2016;115:2964–75.
2015;63:1–9.
324. Zhu X, He L, McCluskey LP. Ingestion of bacterial lipopolysaccharide
333. Stilling RM, Dinan TG, Cryan JF. The brainÕs Geppetto-microbes as
inhibits peripheral taste responses to sucrose in mice. Neuroscience
2014;258:47–61. puppeteers of neural function and behaviour? J Neurovirol 2016;22:14–

Downloaded from https://academic.oup.com/jn/article-abstract/147/5/727/4584720 by guest on 01 October 2018


21.
325. Anisman H, Merali Z. Anhedonic and anxiogenic effects of cytokine
exposure. Adv Exp Med Biol 1999;461:199–233. 334. Sharon G, Sampson TR, Geschwind DH, Mazmanian SK. The central
326. Konsman JP, Parnet P, Dantzer R. Cytokine-induced sickness behav- nervous system and the gut microbiome. Cell 2016;167:915–32.
iour: mechanisms and implications. Trends Neurosci 2002;25:154–9. 335. Bercik P, Park AJ, Sinclair D, Khoshdel A, Lu J, Huang X, Deng Y,
327. Steen PW, Shi L, He L, McCluskey LP. Neutrophil responses to injury Blennerhassett PA, Fahnestock M, Moine D, et al. The anxiolytic effect
or inflammation impair peripheral gustatory function. Neuroscience of Bifidobacterium longum NCC3001 involves vagal pathways for gut-
2010;167:894–908. brain communication. Neurogastroenterol Motil 2011;23(12):1132–9.
328. Skosnik PD, Cortes-Briones JA. Targeting the ecology within: the role 336. Lyte M. Microbial endocrinology in the microbiome-gut-brain axis:
of the gut-brain axis and human microbiota in drug addiction. Med how bacterial production and utilization of neurochemicals influence
Hypotheses 2016;93:77–80. behavior. PLoS Pathog 2013;9:e1003726.

Microbiota and eating behavior 745

Вам также может понравиться