gut microbiota, enteroendocrine functions and metabolism

6
COPHAR-1227; NO. OF PAGES 6 Please cite this article in press as: Cani PD, et al.: Gut microbiota, enteroendocrine functions and metabolism, Curr Opin Pharmacol (2013), http://dx.doi.org/10.1016/j.coph.2013.09.008 Gut microbiota, enteroendocrine functions and metabolism Patrice D Cani, Amandine Everard and Thibaut Duparc The gut microbiota affects host metabolism through a number of physiological processes. Emerging evidence suggests that gut microbes interact with the host through several pathways involving enteroendocrine cells (e.g. L cells). The activation of specific G protein coupled receptors expressed on L cells (e.g. GPR41, GPR43, GPR119 and TGR5) triggers the secretion of glucagon-like peptides (GLP-1 and GLP-2) and PYY. These gut peptides are known to control energy homeostasis, glucose metabolism, gut barrier function and metabolic inflammation. Here, we explore how crosstalk between the ligands produced by the gut microbiota (short chain fatty acids, or SCFAs), or produced by the host but influenced by gut microbes (endocannabinoids and bile acids), impact host physiology. Addresses Universite ´ catholique de Louvain, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Metabolism and Nutrition Research Group, Av. E. Mounier, 73 Box B1.73.11, B-1200 Brussels, Belgium Corresponding author: Cani, Patrice D ([email protected]) Current Opinion in Pharmacology 2013, 13:xxyy This review comes from a themed issue on Endocrine and metabolic diseases Edited by Frank Reimann and Fiona M Gribble S1471-4892/$ see front matter, # 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.coph.2013.09.008 Introduction Obesity is associated with numerous metabolic comor- bidities, such as insulin resistance, diabetes, cardiovas- cular disease and non-alcoholic fatty liver disease. The reduction of excessive body weight is effective for alle- viating many of these metabolic abnormalities; however, the prevention of positive energy balance remains a widely practised cornerstone of obesity-prevention strat- egies [1]. Therefore, more effective weight loss induction strategies are needed to decrease the incidence and severity of metabolic abnormalities. Alternatively, approaches that can treat obesity-related metabolic abnormalities independent of weight loss are extremely attractive. Recent data indicate that certain microbes may provide templates for the development of such strategies (reviewed in [2,3]). Over the years, the understanding of the role of these cells (i.e. gut bacteria) has changed, leading to novel and interesting findings. The gut micro- biota has profound impacts on host physiology, including in the control of energy homeostasis, the immune system, vitamin synthesis and digestion [4,5,6 ]. Owing to its huge surface area and multiple functions, the intestine represents one of the most important organs, and it permits vital interactions with the external world, in- cluding the gut microbiota. The intestinal epithelium allows the absorption of nutrients and fluids while acting as an efficient barrier against toxins and microorganisms. The gut epithelium is comprised of different cell types, including epithelial absorptive cells, Goblet cells, Paneth cells and enteroendocrine cells. Enteroendocrine cells represent approximately 1% of all epithelial cells in the intestine and are subdivided into more than 10 different cell types based on their major secretory products and their localisation along the gastrointestinal tract. Given that multiple biological functions are physiologically regulated by the gut hormones produced by enteroendocrine cells (e.g. food intake, gastric emptying, gut motility, gut barrier formation, glucose metabolism) (Figure 1), these cells have been positioned at the forefront of research to find novel therapies. The enteroendocrine cells play key roles in the maintenance of gut homeostasis both by enabling nutrient absorption and by preserving the essential function of the gut as a barrier between the external environment (gut lumen) and the host tissues. Because enteroendocrine cells are distributed all along the gastrointestinal tract and are in close proximity to gut bacteria and/or metabolites pro- duced by the metabolic activity of the gut microbiota, it is of utmost interest to unravel the crosstalk between the gut microbiota and these cells and the impact of this crosstalk on host physiology (Figure 1). However, the study of the gut microbiota and its relationship with the host represents a real challenge. Among the enteroendocrine cells, L cells have attracted particular interest because of the pleiotropic effects of their secreted peptides [79]. Although these cells are known to respond to nutrients present in the intestinal lumen, they are also able to detect many other luminal compounds. This is especially true in the colon, where the gut microbiota and most of its metab- olites are largely present. In this review, we discuss the body of evidence linking gut microbes (and specific metabolites produced by these bacteria) with enteroendocrine function and thereby host physiology. Gut microbiota and enteroendocrine functions: effects on glucose and energy homeostasis We have previously shown that the modulation of the gut microbiota using prebiotics in mice and humans Available online at www.sciencedirect.com ScienceDirect www.sciencedirect.com Current Opinion in Pharmacology 2013, 13:16

Upload: thibaut

Post on 15-Dec-2016

216 views

Category:

Documents


1 download

TRANSCRIPT

COPHAR-1227; NO. OF PAGES 6

Gut microbiota, enteroendocrine functions and metabolismPatrice D Cani, Amandine Everard and Thibaut Duparc

Available online at www.sciencedirect.com

ScienceDirect

The gut microbiota affects host metabolism through a number

of physiological processes. Emerging evidence suggests that

gut microbes interact with the host through several pathways

involving enteroendocrine cells (e.g. L cells). The activation of

specific G protein coupled receptors expressed on L cells (e.g.

GPR41, GPR43, GPR119 and TGR5) triggers the secretion of

glucagon-like peptides (GLP-1 and GLP-2) and PYY. These gut

peptides are known to control energy homeostasis, glucose

metabolism, gut barrier function and metabolic inflammation.

Here, we explore how crosstalk between the ligands produced

by the gut microbiota (short chain fatty acids, or SCFAs), or

produced by the host but influenced by gut microbes

(endocannabinoids and bile acids), impact host physiology.

Addresses

Universite catholique de Louvain, Louvain Drug Research Institute,

WELBIO (Walloon Excellence in Life sciences and BIOtechnology),

Metabolism and Nutrition Research Group, Av. E. Mounier, 73 Box

B1.73.11, B-1200 Brussels, Belgium

Corresponding author: Cani, Patrice D ([email protected])

Current Opinion in Pharmacology 2013, 13:xx–yy

This review comes from a themed issue on Endocrine and metabolic

diseases

Edited by Frank Reimann and Fiona M Gribble

S1471-4892/$ – see front matter, # 2013 Elsevier Ltd. All rights

reserved.

http://dx.doi.org/10.1016/j.coph.2013.09.008

IntroductionObesity is associated with numerous metabolic comor-

bidities, such as insulin resistance, diabetes, cardiovas-

cular disease and non-alcoholic fatty liver disease. The

reduction of excessive body weight is effective for alle-

viating many of these metabolic abnormalities; however,

the prevention of positive energy balance remains a

widely practised cornerstone of obesity-prevention strat-

egies [1]. Therefore, more effective weight loss induction

strategies are needed to decrease the incidence and

severity of metabolic abnormalities. Alternatively,

approaches that can treat obesity-related metabolic

abnormalities independent of weight loss are extremely

attractive. Recent data indicate that certain microbes may

provide templates for the development of such strategies

(reviewed in [2,3]). Over the years, the understanding of

the role of these cells (i.e. gut bacteria) has changed,

leading to novel and interesting findings. The gut micro-

biota has profound impacts on host physiology, including

Please cite this article in press as: Cani PD, et al.: Gut microbiota, enteroendocrine functions an

www.sciencedirect.com

in the control of energy homeostasis, the immune system,

vitamin synthesis and digestion [4,5,6�].

Owing to its huge surface area and multiple functions, the

intestine represents one of the most important organs, and

it permits vital interactions with the external world, in-

cluding the gut microbiota. The intestinal epithelium

allows the absorption of nutrients and fluids while acting

as an efficient barrier against toxins and microorganisms.

The gut epithelium is comprised of different cell types,

including epithelial absorptive cells, Goblet cells, Paneth

cells and enteroendocrine cells. Enteroendocrine cells

represent approximately 1% of all epithelial cells in the

intestine and are subdivided into more than 10 different

cell types based on their major secretory products and their

localisation along the gastrointestinal tract. Given that

multiple biological functions are physiologically regulated

by the gut hormones produced by enteroendocrine cells

(e.g. food intake, gastric emptying, gut motility, gut barrier

formation, glucose metabolism) (Figure 1), these cells have

been positioned at the forefront of research to find novel

therapies. The enteroendocrine cells play key roles in the

maintenance of gut homeostasis both by enabling nutrient

absorption and by preserving the essential function of the

gut as a barrier between the external environment (gut

lumen) and the host tissues. Because enteroendocrine cells

are distributed all along the gastrointestinal tract and are in

close proximity to gut bacteria and/or metabolites pro-

duced by the metabolic activity of the gut microbiota, it

is of utmost interest to unravel the crosstalk between the

gut microbiota and these cells and the impact of this

crosstalk on host physiology (Figure 1). However, the study

of the gut microbiota and its relationship with the host

represents a real challenge. Among the enteroendocrine

cells, L cells have attracted particular interest because of

the pleiotropic effects of their secreted peptides [7–9].

Although these cells are known to respond to nutrients

present in the intestinal lumen, they are also able to detect

many other luminal compounds. This is especially true in

the colon, where the gut microbiota and most of its metab-

olites are largely present.

In this review, we discuss the body of evidence linking

gut microbes (and specific metabolites produced by these

bacteria) with enteroendocrine function and thereby host

physiology.

Gut microbiota and enteroendocrinefunctions: effects on glucose and energyhomeostasisWe have previously shown that the modulation of the

gut microbiota using prebiotics in mice and humans

d metabolism, Curr Opin Pharmacol (2013), http://dx.doi.org/10.1016/j.coph.2013.09.008

Current Opinion in Pharmacology 2013, 13:1–6

2 Endocrine and metabolic diseases

COPHAR-1227; NO. OF PAGES 6

Figure 1

OEA

2-OG

GLP-1

PYY

GLP-2

SCFA Bile acids

Brain Gl tract

LiverPancreas

Adiposetissue

Muscles

GPR43 GPR119

L-cell

TGR5GPR41

Current Opinion in Pharmacology

Schematic overview of the interactions between the gut microbiota, specific metabolites, enteroendocrine L cell functions and targeted organs.

Enteroendocrine L cells express several G protein coupled receptors (GPCRs) (e.g. GPR43, GPR41, GPR119 and TGR5). The endogenous and

physiological ligands of the different receptors are indicated above each receptor and connected with an arrow. The gut microbiota produces several

metabolites, including short-chain fatty acids (SCFAs) that are able to trigger gut peptide secretion by acting directly on specific receptors such as

GPR41 and 43. Specific changes in the gut microbiota have also been associated with changes in N-oleoylethanolamide (OEA) and 2-oleoylglycerol (2-

OG). These bioactive lipids belong to the endocannabinoid family and serve as ligands of the GPR119 receptor. TGR5 (also known as M-BAR, GPBAR-

1 or GPR131) is targeted by bile acids. The stimulation of these receptors by their putative ligands promotes the secretion of gut peptides (GLP-1,

GLP-2 and PYY) by the L cells. These peptides target several organs, including the gastrointestinal tract, brain, adipose tissue and liver, thereby

contributing to improvements in gut barrier function and glucose and energy homeostasis.

significantly affects energy and glucose homeostasis (for

review [10]). In rodents, we found that this was consist-

ently associated with and improved insulin sensitivity

[11] and leptin sensitivity [12], two major features of

obesity and type 2 diabetes. The fermentation of non-

digestible carbohydrates not only changes gut microbiota

composition and activity but also contributes to the

modulation of bioactive metabolites that can reach the

circulation [13��,14]. Among these metabolites, the short-

chain fatty acids (SCFAs, butyrate, propionate and

acetate) are the best studied (Figure 1). Given that

prebiotic fermentation promotes SCFA production, it is

likely that some of the physiological effects associated

with prebiotics use are directly mediated by such metab-

olites [15,16,17�]. In addition to their production by the

fermentation of prebiotics, SCFAs can also be formed

from other dietary sources of complex carbohydrates

[17�,18]. These metabolites have been shown to influ-

ence insulin sensitivity and energy metabolism through

various physiological pathways, including components of

the central nervous system [19–21]. Indeed, these metab-

olites can modify the levels of several gut hormones

Please cite this article in press as: Cani PD, et al.: Gut microbiota, enteroendocrine functions an

Current Opinion in Pharmacology 2013, 13:1–6

involved in glucose and energy homeostasis [22��,23–25]. For example, butyrate and acetate were shown to

suppress weight gain in mice with high-fat diet-induced

obesity, and propionate was shown to reduce food intake

[26]. Lin et al. observed that the oral administration of

butyrate and propionate in mice significantly increased

their plasma levels of glucagon-like peptide-1 (GLP-1)

and glucose-dependent insulinotropic polypeptide (GIP),

accompanied by a moderate increase in peptide YY (PYY)

levels, leading to improved oral glucose tolerance and

reduced fasting insulin and leptin levels; these phenom-

ena are consistent with the observed improvements in

insulin sensitivity [26]. Moreover, butyrate was also

shown to improve insulin sensitivity and increase energy

expenditure in obese mice [27]. Unfortunately, the mol-

ecular pathways underlying these beneficial effects of

SCFAs remain largely unknown. However, the recent

identification of a family of G protein coupled receptors

(GPCRs) that bind these gut microbiome products has

highlighted new potential mechanisms of action for

SCFAs in glucose homeostasis [28]. Interestingly, the

expression of these receptors seems to be regulated by

d metabolism, Curr Opin Pharmacol (2013), http://dx.doi.org/10.1016/j.coph.2013.09.008

www.sciencedirect.com

Gut microbiota and endocrine cells Cani, Everard and Duparc 3

COPHAR-1227; NO. OF PAGES 6

the gut microbiota itself [16,29,30]. Moreover, several

studies have reported that SCFA receptors colocalise in

the epithelium with the enteroendocrine cells that

secrete key modulators of glucose homeostasis such as

PYY, GLP-1 and GIP [31]. Among these receptors,

GPR41 is expressed in the intestinal mucosa and is

particularly abundant in an immortalised murine L cell

line [32], which is consistent with a potential role in

peptide secretion from L, K, and other enteroendocrine

cells, but the role of GPR41 in the regulation of energy

balance and glucose metabolism is poorly understood

(Figure 1). To address this question, a GPR41-deficient

mouse line was generated. These mice have body weights

similar to those of control mice on both normal and high-fat

diets and exhibit normal fasting GLP-1 levels but attenu-

ated GLP-1 secretion in response to butyrate. The GIP and

PYY levels of GPR41-deficient mice were similar to those

of their littermates. These data suggest that only butyrate

stimulation of GLP-1 secretion from L cells is partially

mediated by GPR41 [26]. Tolhurst et al. generated knock-

out mice deficient in GPR43 [25], another SCFA receptor

that is normally expressed in the rat and human colon wall,

mucosa and PYY-expressing enteroendocrine cells [33].

They found that GPR43-deficient mice have reduced

colonic GLP-1 protein content, reduced basal and glu-

cose-stimulated levels of GLP-1 and impaired glucose

tolerance (Figure 1). Other receptors can also stimulate

GLP-1 secretion by L cells. For instance, bile acids can

bind to both nuclear and cell surface receptors. Yet, the gut

microbiota regulates both bile acid synthesis and the

production of secondary bile acids [34]. Two bile acid

receptors are involved in glucose homeostasis. TGR5 is

a GPCR, and its activation improves liver function and

glucose tolerance in obese mice by regulating intestinal

GLP-1 production (Figure 1) [35]. FXR is a nuclear re-

ceptor that is essential for maintaining glucose tolerance

and insulin sensitivity but acts via mechanisms other than

enteroendocrine regulation [36].

Although convincing studies have supported a link be-

tween GPR43/41, enteroendocrine functions and gut

microbiota metabolites (i.e. SCFAs), compelling evi-

dence suggests that other bioactive compounds and

receptors contribute to the secretion of GLP-1/2 and

PYY by the L cells [7]. For instance, bioactive lipids

belonging to the N-acylethanolamine and acylglycerol

families are also able to activate the L cell-specific re-

ceptor GPR119 (Figure 1) [37,38,39��].

To date, few studies have characterised the roles of

specific bacterial species that are able to produce metab-

olites with bioactive effects on these gut hormones.

Interestingly, Akkermansia muciniphila, a mucin-degrading

bacterium that resides in the mucus layer, is able to

produce acetate and propionate [40], which can act on

GPR43. We recently showed that this bacterium was

present at lower levels in obese and type 2 diabetic mice

Please cite this article in press as: Cani PD, et al.: Gut microbiota, enteroendocrine functions an

www.sciencedirect.com

than in healthy mice [41�] and that the daily adminis-

tration of A. muciniphila for 4 weeks reversed high-fat diet-

induced obesity and type 2 diabetes [41�]. Strikingly, we

also showed that the abundance of A. muciniphila was

associated with L cell number and secretory capacity [12].

Therefore, it appears clear that the gut microbiota and

specific associated metabolites are key partners in the

control of glucose and energy homeostasis. Although the

mechanisms underlying this regulation are still unclear,

the gut microbiota constitutes an important potential

target for treatment of metabolic disorders such as obesity

and type 2 diabetes.

Gut microbiota and enteroendocrinefunctions: effects on gut barrier functionGut barrier disruption has been observed in several

pathological situations and is involved in the pathophy-

siologic process of multiple diseases. Obesity is associated

with an increase in gut permeability that leads to the

abnormal translocation of gut bacteria or gut bacteria

components from the intestinal lumen through the host

blood circulation and tissues [42–45]. We have shown that

these changes in gut barrier activity are associated with

increased plasma levels of gram-negative bacterial cell

wall components, namely lipopolysaccharides (LPS).

Importantly, the increase in plasma LPS levels, which

is defined as metabolic endotoxemia, has been shown to

trigger low grade inflammation and metabolic disorders

associated with obesity and type 2 diabetes [42]. Given

the implications of gut barrier alterations on the onset of

metabolic endotoxemia, inflammation and metabolic dis-

orders associated with obesity and type 2 diabetes, it is of

the utmost importance to determine the underlying

mechanisms and the potential opportunities to reverse

the increase in gut permeability related to this pathology.

One of the principal players involved in the changes in

gut barrier function in obesity and type 2 diabetes is the

gut microbiota. Indeed, it is now well established that

obesity is associated with changes in the diversity and

composition of the gut microbiome [46–49]. Moreover,

we have demonstrated that this link is causal because

modulations of the gut microbiota using prebiotics can

improve gut barrier function and ameliorate metabolic

endotoxemia and inflammation in obesity and type 2

diabetes through mechanisms associated with enteroen-

docrine cell function [12,41�,50,51]. To date, the main

enteroendocrine peptide known to be involved in gut

barrier functions and produced by enteroendocrine cells

is the glucagon-like peptide-2 (GLP-2) (Figure 1). This

peptide is produced by L cells; its production is regulated

by the nutritional status of the host, and it increases the

nutrient absorption capacity of the host. Importantly,

GLP-2 maintains gut barrier functions through different

mechanisms. First, GLP-2 has been demonstrated to

induce intestinal epithelial cell proliferation and thereby

d metabolism, Curr Opin Pharmacol (2013), http://dx.doi.org/10.1016/j.coph.2013.09.008

Current Opinion in Pharmacology 2013, 13:1–6

4 Endocrine and metabolic diseases

COPHAR-1227; NO. OF PAGES 6

gut trophic effects [52]. Second, GLP-2 increases the

expression of proteins that link epithelial cells together,

that is, intestinal tight junction proteins [53,54]. Third,

GLP-2 has recently been identified to be involved in

regulating the innate immune system by controlling the

expression of antimicrobial peptides produced by Paneth

cells. Antimicrobial peptides are implicated in host–microbiota segregation and consequently the mainten-

ance of gut barrier functions [55].

Importantly, we have shown that the beneficial effects of

gut microbiota modulation on gut barrier function are

associated with an increase in GLP-2 production related

to an increase in the number of L cells in the intestine

[12,53]. It is worth noting that treatment with a GLP-2

antagonist completely abolished the improvements in gut

barrier function following prebiotic-induced changes in

the gut microbiota. These data suggest that specific

interactions between the gut microbiota and enteroendo-

crine L cells control gut permeability and inflammation in

obesity and type 2 diabetes.

As discussed above, we have recently identified A. muci-niphila as one species that is involved in the crosstalk

between the gut microbiota and intestinal epithelium in

the context of obesity and type 2 diabetes. Compelling

evidence suggests that A. muciniphila seems to play a

special role in gut barrier function. Indeed, several path-

ologies associated with increased gut permeability are

also associated with a decrease in the abundance of A.muciniphila in the gut [56,57]. Importantly, the abundance

of this bacterium negatively correlates with the levels of

gut permeability markers and inflammatory markers

[41�]. In a recent study, we found that A. muciniphilatreatment decreases metabolic endotoxemia and inflam-

mation by improving intestinal mucosal barrier function

in high-fat fed mice [41�]. Our data suggest that the

effects of A. muciniphila on gut barrier function could

be mediated through enteroendocrine L cells.

Indeed, we demonstrated that A. muciniphila adminis-

tration significantly increases the intestinal levels of 2-

oleoylglycerol (2-OG), a bioactive lipid belonging to the

endocannabinoid system [41�]. Interestingly, 2-OG

stimulates the secretion of glucagon-like peptides

through the activation of GPR119 localised on intestinal

L cells (Figure 1) [39��].

It is worth noting that L cells and their secretory products

are not the only enteroendocrine peptides and cells

involved in the relationship between the gut microbiota

and gut barrier function. For instance, intestinal signalling

by serotonin (another enteroendocrine product) is involved

in gut barrier function during obesity. Levels of serotonin

and the type 3 serotonin receptor (5-HT3R) are increased

in mice with genetic or high-fat diet-induced obesity, and

5-HT3R antagonists increase the expression of intestinal

Please cite this article in press as: Cani PD, et al.: Gut microbiota, enteroendocrine functions an

Current Opinion in Pharmacology 2013, 13:1–6

tight junction proteins such as occludin and claudin-1, an

effect that is associated with decreases in metabolic endo-

toxemia and fatty acid liver diseases associated with obesity

[58,59]. A link between specific gut microbes and the

modulation of serotonin production has been proposed

[60,61]; however, the connections between serotonin,

the gut microbiota and the secretory capacity of other

enteroendocrine cells remain to be elucidated.

ConclusionsSeveral studies have shown that modification of the gut

microbiota affects various physiological processes. However,

it has so far proven difficult to determine the precise

mechanisms linking gut microbiota composition and/or

activity with specific metabolic pathways involved in energy

and glucose homeostasis. The gut microbiota represents an

extremely diverse group of microbes and is not a static

ecosystem but is subject to rapid modifications that are

directly associated with nutrient intake. Several studies have

suggested that targeting the gut microbiota and thereby the

production of specific metabolites may be one interesting

approach toward improving metabolic features associated

with obesity and type 2 diabetes. However, it remains to be

determined whether these modifications are a cause or a

consequence of the metabolic changes observed.

Emerging evidence suggests that the gut microbiota

interacts with their hosts through several pathways invol-

ving the enteroendocrine cells. Thus, specific GPCRs

expressed on enteroendocrine L cells may constitute a

putative therapeutic target.

To date, it is not known whether the modifications of

enteroendocrine cell activities by the microbiota are strain

or metabolite dependent. The next challenge is to develop

approaches to decipher the mechanisms involved in mol-

ecular signalling modulated by gut microbes to ultimately

identify new therapeutic targets for the management of

various metabolic conditions. Thus, additional research is

needed to further isolate and identify the key mechanisms

involved in gut microbe–host interactions.

AcknowledgementsPDC is a research associate from the FRS-FNRS (Fonds de la RechercheScientifique) Belgium. AE is a doctoral fellow from the FRS-FNRS.TD is postdoctoral fellow from the FRM (Fondation pour la RechercheMedicale, France). PDC is recipients of an ERC Starting Grant 2013(336452-ENIGMO), FSR and FRSM subsidies (Fonds speciaux derecherches, UCL, Belgium; Fonds de la recherche scientifique medicale,Belgium) and ARC (Action de Recherche Concertee).

References and recommended readingPapers of particular interest, published within the period of review,have been highlighted as:

� of special interest

�� of outstanding interest

1. Dhurandhar NV: When commonsense does not make sense. IntJ Obes (Lond) 2012, 36:1332-1333.

d metabolism, Curr Opin Pharmacol (2013), http://dx.doi.org/10.1016/j.coph.2013.09.008

www.sciencedirect.com

Gut microbiota and endocrine cells Cani, Everard and Duparc 5

COPHAR-1227; NO. OF PAGES 6

2. Dhurandhar NV, Geurts L, Atkinson RL, Casteilla L, Clement K,Gerard P, Vijay-Kumar M, Nam JH, Nieuwdorp M, Trovato G et al.:Harnessing the beneficial properties of adipogenic microbesfor improving human health. Obes Rev 2013, 14:721-735.

3. Dhurandhar NV: A framework for identification of infectionsthat contribute to human obesity. Lancet Infect Dis 2011,11:963-969.

4. Hooper LV, Macpherson AJ: Immune adaptations that maintainhomeostasis with the intestinal microbiota. Nat Rev Immunol2010, 10:159-169.

5. Round JL, Mazmanian SK: The gut microbiota shapes intestinalimmune responses during health and disease. Nat RevImmunol 2009, 9:313-323.

6.�

Tremaroli V, Backhed F: Functional interactions betweenthe gut microbiota and host metabolism. Nature 2012,489:242-249.

This review outlines different mechanisms involving the gut microbiotainteractions and host metabolims. This review is elegantly discussed andillustrated.

7. des Rieux A, Pourcelle V, Cani PD, Marchand-Brynaert J, Preat V:Targeted nanoparticles with novel non-peptidic ligands fororal delivery. Adv Drug Deliv Rev 2013, 65:833-844.

8. Parker HE, Wallis K, Le Roux CW, Wong KY, Reimann F,Gribble FM: Molecular mechanisms underlying bile acid-stimulated glucagon-like peptide-1 secretion. Br J Pharmacol2012, 165:414-423.

9. Reimann F, Habib AM, Tolhurst G, Parker HE, Rogers GJ,Gribble FM: Glucose sensing in L cells: a primary cell study. CellMetab 2008, 8:532-539.

10. Delzenne NM, Neyrinck AM, Backhed F, Cani PD: Targeting gutmicrobiota in obesity: effects of prebiotics and probiotics. NatRev Endocrinol 2011, 7:639-646.

11. Cani PD, Knauf C, Iglesias MA, Drucker DJ, Delzenne NM,Burcelin R: Improvement of glucose tolerance and hepaticinsulin sensitivity by oligofructose requires a functionalglucagon-like peptide 1 receptor. Diabetes 2006,55:1484-1490.

12. Everard A, Lazarevic V, Derrien M, Girard M, Muccioli GM,Neyrinck AM, Possemiers S, Van HA, Francois P, de Vos WM et al.:Responses of gut microbiota and glucose and lipidmetabolism to prebiotics in genetic obese and diet-inducedleptin-resistant mice. Diabetes 2011, 60:2775-2786.

13.��

Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI: Humannutrition, the gut microbiome and the immune system. Nature2011, 474:327-336.

A dense review discussing data supporting the link between gut micro-biota and immune system, in both physiological and pathological situa-tions.

14. Muccioli GG, Naslain D, Backhed F, Reigstad CS, Lambert DM,Delzenne NM, Cani PD: The endocannabinoid system links gutmicrobiota to adipogenesis. Mol Syst Biol 2010, 6:392.

15. Kimura I, Ozawa K, Inoue D, Imamura T, Kimura K, Maeda T,Terasawa K, Kashihara D, Hirano K, Tani T et al.: The gutmicrobiota suppresses insulin-mediated fat accumulation viathe short-chain fatty acid receptor GPR43. Nat Commun 2013,4:1829.

16. Dewulf EM, Cani PD, Neyrinck AM, Possemiers S, Holle AV,Muccioli GG, Deldicque L, Bindels LB, Pachikian BD, Sohet FMet al.: Inulin-type fructans with prebiotic properties counteractGPR43 overexpression and PPARgamma-relatedadipogenesis in the white adipose tissue of high-fat diet-fedmice. J Nutr Biochem 2011, 22:712-722.

17.�

Scott KP, Gratz SW, Sheridan PO, Flint HJ, Duncan SH: Theinfluence of diet on the gut microbiota. Pharmacol Res 2013,69:52-60.

This is an excellent review of the relationship between different nutrients(fat, protein, carbohydrates) and the gut microbiota.

18. Charrier JA, Martin RJ, McCutcheon KL, Raggio AM, Goldsmith F,Goita M, Senevirathne R, Brown IL, Pelkman C, Zhou J et al.: Highfat diet partially attenuates fermentation responses in rats fed

Please cite this article in press as: Cani PD, et al.: Gut microbiota, enteroendocrine functions an

www.sciencedirect.com

resistant starch from high-amylose maize. Obesity (SilverSpring) 2013 http://dx.doi.org/10.1002/oby.20362. [Epub ahead ofprint].

19. Donohoe DR, Wali A, Brylawski BP, Bultman SJ: Microbialregulation of glucose metabolism and cell-cycle progressionin mammalian colonocytes. PLOS ONE 2012, 7:e46589.

20. Kruger C, Kumar KG, Mynatt RL, Volaufova J, Richards BK: Braintranscriptional responses to high-fat diet in Acads-deficientmice reveal energy sensing pathways. PLOS ONE 2012,7:e41709.

21. Anastasovska J, Arora T, Sanchez Canon GJ, Parkinson JR,Touhy K, Gibson GR, Nadkarni NA, So PW, Goldstone AP,Thomas EL et al.: Fermentable carbohydrate altershypothalamic neuronal activity and protects against theobesogenic environment. Obesity (Silver Spring) 2012,20:1016-1023.

22.��

Reimann F, Tolhurst G, Gribble FM: G-protein-coupledreceptors in intestinal chemosensation. Cell Metab 2012,15:421-431.

An outstanding review examining the role of G protein-coupled receptorsas signaling molecules in the context of energy and glucose metabolism.

23. Plaisancie P, Dumoulin V, Chayvialle JA, Cuber JC: Luminalglucagon-like peptide-1(7-36) amide-releasing factors in theisolated vascularly perfused rat colon. J Endocrinol 1995,145:521-526.

24. Tarini J, Wolever TM: The fermentable fibre inulin increasespostprandial serum short-chain fatty acids and reduces free-fatty acids and ghrelin in healthy subjects. Appl Physiol NutrMetab 2010, 35:9-16.

25. Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM,Diakogiannaki E, Cameron J, Grosse J, Reimann F, Gribble FM:Short-chain fatty acids stimulate glucagon-like peptide-1secretion via the G protein-coupled receptor FFAR2. Diabetes2012, 61:364-371.

26. Lin HV, Frassetto A, Kowalik EJ Jr, Nawrocki AR, Lu MM,Kosinski JR, Hubert JA, Szeto D, Yao X, Forrest G et al.: Butyrateand propionate protect against diet-induced obesity andregulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLOS ONE 2012, 7:e35240.

27. Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, Lefevre M, Cefalu WT,Ye J: Butyrate improves insulin sensitivity and increasesenergy expenditure in mice. Diabetes 2009, 58:1509-1517.

28. Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L,Daniels D, Muir AI, Wigglesworth MJ, Kinghorn I, Fraser NJ et al.:The Orphan G protein-coupled receptors GPR41 and GPR43are activated by propionate and other short chain carboxylicacids. J Biol Chem 2003, 278:11312-11319.

29. Duca FA, Swartz TD, Sakar Y, Covasa M: Increased oraldetection, but decreased intestinal signaling for fats in micelacking gut microbiota. PLOS ONE 2012, 7:e39748.

30. Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F,Manchester JK, Hammer RE, Williams SC, Crowley J,Yanagisawa M et al.: Effects of the gut microbiota on hostadiposity are modulated by the short-chain fatty-acid bindingG protein-coupled receptor, Gpr41. Proc Natl Acad Sci U S A2008, 105:16767-16772.

31. Kaji I, Karaki S, Tanaka R, Kuwahara A: Density distribution offree fatty acid receptor 2 (FFA2)-expressing and GLP-1-producing enteroendocrine L cells in human and rat lowerintestine, and increased cell numbers after ingestion of fructo-oligosaccharide. J Mol Histol 2011, 42:27-38.

32. Tazoe H, Otomo Y, Karaki S, Kato I, Fukami Y, Terasaki M,Kuwahara A: Expression of short-chain fatty acid receptorGPR41 in the human colon. Biomed Res 2009, 30:149-156.

33. Karaki S, Mitsui R, Hayashi H, Kato I, Sugiya H, Iwanaga T,Furness JB, Kuwahara A: Short-chain fatty acid receptor,GPR43, is expressed by enteroendocrine cells and mucosalmast cells in rat intestine. Cell Tissue Res 2006, 324:353-360.

34. Claus SP, Tsang TM, Wang Y, Cloarec O, Skordi E, Martin FP,Rezzi S, Ross A, Kochhar S, Holmes E et al.: Systemic

d metabolism, Curr Opin Pharmacol (2013), http://dx.doi.org/10.1016/j.coph.2013.09.008

Current Opinion in Pharmacology 2013, 13:1–6

6 Endocrine and metabolic diseases

COPHAR-1227; NO. OF PAGES 6

multicompartmental effects of the gut microbiome on mousemetabolic phenotypes. Mol Syst Biol 2008, 4:219.

35. Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G,Macchiarulo A, Yamamoto H, Mataki C, Pruzanski M et al.: TGR5-mediated bile acid sensing controls glucose homeostasis. CellMetab 2009, 10:167-177.

36. Ma K, Saha PK, Chan L, Moore DD: Farnesoid X receptor isessential for normal glucose homeostasis. J Clin Invest 2006,116:1102-1109.

37. Syed SK, Bui HH, Beavers LS, Farb TB, Ficorilli J, Chesterfield AK,Kuo MS, Bokvist K, Barrett DG, Efanov AM: Regulation ofGPR119 receptor activity with endocannabinoid-like lipids. AmJ Physiol Endocrinol Metab 2012, 303:E1469-E1478.

38. Lauffer LM, Iakoubov R, Brubaker PL: GPR119 is essential foroleoylethanolamide-induced glucagon-like peptide-1secretion from the intestinal enteroendocrine L-cell. Diabetes2009, 58:1058-1066.

39.��

Hansen KB, Rosenkilde MM, Knop FK, Wellner N, Diep TA,Rehfeld JF, Andersen UB, Holst JJ, Hansen HS: 2-Oleoyl glycerolis a GPR119 agonist and signals GLP-1 release in humans. JClin Endocrinol Metab 2011, 96:E1409-E1417.

A very interesting study showing that 2OG and other 2-monoacylglycerolsthat belong to the endocannabinoid system and formed during fatdigestion can activate GPR119 and cause incretin release.

40. Derrien M, Vaughan EE, Plugge CM, de Vos WM: Akkermansiamuciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol 2004,54:1469-1476.

41.�

Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB,Guiot Y, Derrien M, Muccioli GG, Delzenne NM et al.: Cross-talkbetween Akkermansia muciniphila and intestinal epitheliumcontrols diet-induced obesity. Proc Natl Acad Sci U S A 2013,110:9066-9071.

This is the first study showing the role of Akkermansia muciniphila onglucose, energy metabolism and gut barrier function.

42. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D,Neyrinck AM, Fava F, Tuohy KM, Chabo C et al.: Metabolicendotoxemia initiates obesity and insulin resistance. Diabetes2007, 56:1761-1772.

43. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM,Delzenne NM, Burcelin R: Changes in gut microbiota controlmetabolic endotoxemia-induced inflammation in high-fatdiet-induced obesity and diabetes in mice. Diabetes 2008,57:1470-1481.

44. Brun P, Castagliuolo I, Leo VD, Buda A, Pinzani M, Palu G,Martines D: Increased intestinal permeability in obese mice:new evidence in the pathogenesis of nonalcoholicsteatohepatitis. Am J Physiol Gastrointest Liver Physiol 2007,292:G518-G525.

45. Amar J, Serino M, Lange C, Chabo C, Iacovoni J, Mondot S,Lepage P, Klopp C, Mariette J, Bouchez O et al.: Involvement oftissue bacteria in the onset of diabetes in humans: evidencefor a concept. Diabetologia 2011, 54:3055-3061.

46. Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD,Gordon JI: Obesity alters gut microbial ecology. Proc Natl AcadSci U S A 2005, 102:11070-11075.

47. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y,Shen D et al.: A metagenome-wide association study ofgut microbiota in type 2 diabetes. Nature 2012,490:55-60.

Please cite this article in press as: Cani PD, et al.: Gut microbiota, enteroendocrine functions an

Current Opinion in Pharmacology 2013, 13:1–6

48. Karlsson FH, Tremaroli V, Nookaew I, Bergstrom G, Behre CJ,Fagerberg B, Nielsen J, Backhed F: Gut metagenome inEuropean women with normal, impaired and diabetic glucosecontrol. Nature 2013, 498:99-103.

49. Everard A, Cani PD: Diabetes, obesity and gut microbiota. BestPract Res Clin Gastroenterol 2013, 27:73-83.

50. Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM,Gibson GR, Delzenne NM: Selective increases of bifidobacteriain gut microflora improve high-fat-diet-induced diabetes inmice through a mechanism associated with endotoxaemia.Diabetologia 2007, 50:2374-2383.

51. Neyrinck AM, Van Hee VF, Piront N, De Backer F, Toussaint O,Cani PD, Delzenne NM: Wheat-derived arabinoxylanoligosaccharides with prebiotic effect increase satietogenicgut peptides and reduce metabolic endotoxemia in diet-induced obese mice. Nutr Diabetes 2012, 2:e28.

52. Drucker DJ, Erlich P, Asa SL, Brubaker PL: Induction of intestinalepithelial proliferation by glucagon-like peptide 2. Proc NatlAcad Sci U S A 1996, 93:7911-7916.

53. Cani PD, Possemiers S, Van de WT, Guiot Y, Everard A, Rottier O,Geurts L, Naslain D, Neyrinck AM, Lambert DM et al.: Changes ingut microbiota control inflammation in obese mice through amechanism involving GLP-2-driven improvement of gutpermeability. Gut 2009, 58:1091-1103.

54. Moran GW, O’Neill C, McLaughlin JT: GLP-2 enhances barrierformation and attenuates TNFalpha-induced changes in aCaco-2 cell model of the intestinal barrier. Regul Pept 2012,178:95-101.

55. Lee SJ, Lee J, Li KK, Holland D, Maughan H, Guttman DS, Yusta B,Drucker DJ: Disruption of the murine Glp2r impairs Paneth cellfunction and increases susceptibility to small bowel enteritis.Endocrinology 2012, 153:1141-1151.

56. Png CW, Linden SK, Gilshenan KS, Zoetendal EG,McSweeney CS, Sly LI, McGuckin MA, Florin TH: Mucolyticbacteria with increased prevalence in IBD mucosa augment invitro utilization of mucin by other bacteria. Am J Gastroenterol2010, 105:2420-2428.

57. Santacruz A, Collado MC, Garcia-Valdes L, Segura MT, Martin-Lagos JA, Anjos T, Marti-Romero M, Lopez RM, Florido J,Campoy C et al.: Gut microbiota composition is associatedwith body weight, weight gain and biochemical parameters inpregnant women. Br J Nutr 2010, 104:83-92.

58. Haub S, Ritze Y, Ladel I, Saum K, Hubert A, Spruss A, Trautwein C,Bischoff SC: Serotonin receptor type 3 antagonists improveobesity-associated fatty liver disease in mice. J Pharmacol ExpTher 2011, 339:790-798.

59. Bertrand RL, Senadheera S, Markus I, Liu L, Howitt L, Chen H,Murphy TV, Sandow SL, Bertrand PP: A Western diet increasesserotonin availability in rat small intestine. Endocrinology 2011,152:36-47.

60. Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, Deng Y,Blennerhassett P, Macri J, McCoy KD et al.: The intestinalmicrobiota affect central levels of brain-derived neurotropicfactor and behavior in mice. Gastroenterology 2011, 141:599-609 e591–e593.

61. Nishino R, Mikami K, Takahashi H, Tomonaga S, Furuse M,Hiramoto T, Aiba Y, Koga Y, Sudo N: Commensal microbiotamodulate murine behaviors in a strictly contamination-freeenvironment confirmed by culture-based methods.Neurogastroenterol Motil 2013, 25:521-528.

d metabolism, Curr Opin Pharmacol (2013), http://dx.doi.org/10.1016/j.coph.2013.09.008

www.sciencedirect.com