Hostname: page-component-8448b6f56d-c47g7 Total loading time: 0 Render date: 2024-04-19T20:50:35.574Z Has data issue: false hasContentIssue false

Current and emerging concepts on the role of peripheral signals in the control of food intake and development of obesity

Published online by Cambridge University Press:  13 March 2012

F. A. Duca
Affiliation:
INRA and AgroParis Tech, UMR 1319, MICALIS, Neurobiology of Ingestive Behavior, 78350 Jouy-en-Josas, Cedex, France University Pierre and Marie Curie, Paris75005, France
M. Covasa*
Affiliation:
INRA and AgroParis Tech, UMR 1319, MICALIS, Neurobiology of Ingestive Behavior, 78350 Jouy-en-Josas, Cedex, France Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA91766, USA
*
*Corresponding author: M. Covasa, fax +33 1 34 65 24 92, email mcovasa@jouy.inra.fr
Rights & Permissions [Opens in a new window]

Abstract

The gastrointestinal peptides are classically known as short-term signals, primarily inducing satiation and/or satiety. However, accumulating evidence has broadened this view, and their role in long-term energy homeostasis and the development of obesity has been increasingly recognised. In the present review, the recent research involving the role of satiation signals, especially ghrelin, cholecystokinin, glucagon-like peptide 1 and peptide YY, in the development and treatment of obesity will be discussed. Their activity, interactions and release profile vary constantly with changes in dietary and energy influences, intestinal luminal environment, body weight and metabolic status. Manipulation of gut peptides and nutrient sensors in the oral and postoral compartments through diet and/or changes in gut microflora or using multi-hormone ‘cocktail’ therapy are among promising approaches aimed at reducing excess food consumption and body-weight gain.

Type
Review Article
Copyright
Copyright © The Authors 2012

Obesity rates continue to rise worldwide, with no immediate cure in sight. While increased food intake coupled with decreased energy expenditure generally accounts for rising obesity rates, this equation is influenced by a multitude of factors including genetic, physiological, neural, metabolic, social and environmental factors (for a review, see Berthoud(Reference Berthoud1)). With large-scale attempts at increasing energy expenditure mostly unsuccessful, the necessity for therapy in combating obesity has led to important advances in understanding the mechanisms controlling meal size and energy regulation. Throughout a meal, ingested nutrients interact at multiple sites generating signals regarding energy load, meal composition and size. Signals from the oral cavity, gastrointestinal (GI) tract, adjacent alimentary organs, and muscle and adipose tissue all converge in the brain to control short-term food intake and achieve long-term energy balance. The present study reviews new emerging evidence for the role of GI signals in controlling appetite and energy balance, their adaptive functions in the face of constant environmental changes and their potential therapeutic role in the prevention, perpetuation and treatment of obesity. Since peripheral signals are sensed by the brain and the ongoing bidirectional dialogue between the gut and the brain is pivotal to the control of energy intake, the key brain areas integrating this complex information, the sensing neurons and central peptides involved in the regulation of energy homeostasis will also be briefly mentioned.

In today's modern society, where there is an abundance of food, most meals are not initiated by physiological need (‘hunger’); therefore, the main action of most peripheral signals is not to initiate feeding, but to control the size of the meal once eating begins. The GI tract is host to a vast array of chemical and neural signals controlling food intake. These signals arising from the periphery are classically divided into short-term ‘episodic’ signals, which are rhythmically released in response to eating such as GI peptide hormones, and long-term ‘tonic’ signals, such as insulin, leptin and adipokines, that are released in proportion to the amount of fat stores, reflecting the metabolic state(Reference Woods2, Reference Blundell3). Some of the GI peptides such as cholecystokinin (CCK) generate signals leading to meal termination (satiation), while others such as peptide YY (PYY)(3–36) also play a role in controlling eating during the postprandial period (satiety). Most short-term, episodic peptide signals are secreted from the gut in response to specific nutrients, and act on local sensory nerves, relaying messages to the hindbrain that contribute to satiation and/or satiety(Reference Woods4). Although known for their short-term effects, new accumulating evidence suggests a broader role in the long-term regulation of appetite and energy balance(Reference Neary and Batterham5). Tonic signals reflect energy storage levels and, via an endocrine mode of action, regulate body weight and stored energy by acting on hypothalamic neurons(Reference Woods2). A constant reciprocal relationship exists between episodic and tonic signals, with episodic signals overcoming tonic influences, thus driving eating even in an energy-repleted condition such as obesity, while, on the other hand, tonic signals can modulate the strength of episodic signalling, therefore contributing to the short-term control of food intake.

Gut–brain integration

Satiation, adiposity and other neural signals, such as gastric distension, are integrated in the caudal brainstem and/or hypothalamus where an appropriate response is generated, ultimately affecting meal size and energy homeostasis, as depicted in Fig. 1. The caudal brainstem is a key recipient integrating not only sensory information from neural gustatory and gut vagal afferents that synapse in the nucleus of the solitary tract(Reference Berthoud1), but also humoral information from endocrine signals, via area postrema such as leptin, ghrelin and amylin which all contain receptors in the caudal brainstem(Reference Faulconbridge, Cummings and Kaplan6Reference Sexton, Paxinos and Kenney8). While decereberate animals can effectively control meal size through caudal brainstem integration, they lack the ability to seek food and compensate total energy intake when fasted, demonstrating the role of higher-order hypothalamic input in the regulation of weight gain(Reference Grill9).

Fig. 1 Food intake is controlled by complex neural, hormonal and metabolic signals. In the oral cavity, nutrient and non-nutrient tastants activate several taste proteins, such as type 1 taste receptor (T1R) 2/3 and fatty acid translocase CD-36 (CD36). This sensory input is processed by the hindbrain, further stimulating ingestion. The presence of nutrients triggers the release of gastrointestinal (GI) peptides from the stomach and intestine that either act locally on specific receptors distributed along vagal afferents which synapse with the first-order neurons in the hindbrain, or enter the bloodstream (along with signals from the adipose tissue such as leptin) and activate receptors located on hypothalamic neurons. The release of these GI signals, especially in the distal intestine, is also affected by the microflora within the gut. Gut microbiota dispersed primarily throughout the distal intestine (represented by oval and round shapes in the intestinal lumen) influences peptide secretion possibly through nutrient receptors, such as T1R2/R3 and G-protein-coupled receptor (GPCR) or bacterial by-products such as SCFA, that serve as ligands for GPCR. These signals, along with other sensory inputs, are integrated with circuits from higher-order brain areas which in turn alter food intake, energy expenditure and body adiposity. With the exception of ghrelin that has a stimulatory effect on food intake, the final actions of these peptides are inhibition of food intake. LCFA, long-chain fatty acid; GLUC, glucose; CCK, cholecystokinin; GLP-1, glucagon-like peptide-1; PYY, peptide YY; CCK-1R, cholecystokinin-1 receptor; GHSR, growth hormone secretagogue receptor; LepR, leptin receptor; GLP-1R, glucagon-like peptide-1 receptor; NPY2R, neuropeptide Y 2-receptor.

The hypothalamus, specifically the arcuate nucleus (ARC), is the main relay station that receives and integrates nutritional information via circulating hormones and metabolites through a saturable carrier across the blood–brain barrier(Reference Banks, Kastin and Huang10Reference Moran12), and/or from direct access through the incomplete portion of the blood–brain barrier(Reference Munzberg13). In addition, hypothalamic nuclei receive indirect information from peripheral signals via brainstem neural pathways(Reference Blevins and Baskin14). Finally, dopaminergic and endocannabinoid systems that are involved in food reward, as well as higher-order brain inputs related to emotions, motivation and learned behaviour, also converge in the hypothalamus(Reference Langhans and Geary15). Taken together, the hypothalamus coordinates multiple levels of information and provides subsequent efferent signals to regulate overall energy homeostasis. A major site for this regulation within the ARC is through two populations of neurons with opposing effects on food intake. There are an anorexigenic group containing pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript, and an orexigenic group containing neuropeptide Y (NPY) and agouti-related peptide (AgRP), that branch and relay information to other areas involved in food intake and energy homeostasis, such as the lateral and ventromedial hypothalamus, the paraventricular nucleus and the nucleus of the solitary tract(Reference Lenard and Berthoud16). Therefore, the overall control of food intake and regulation of body weight occurs predominantly in the central nervous system. However, peripheral inputs have a major impact on the subsequent actions of the central nervous system, since they relay first-hand primary information regarding the current meal and the metabolic status of the body.

Oral nutrient sensing

In obesity, an increased motivation to eat overrides homeostatic regulation, resulting in sustained and escalating overeating despite normal or excessive energy storage. Sugars and fats are palatable to both humans and animals, and are preferred and consumed in large quantities(Reference Rolls17). Thus, identification of mechanisms involved in the detection of chemical compounds such as sugars and fats has major nutritional and clinical significance. In the past decade, the field has made significant progress with the discovery and characterisation of sweet taste receptors of the type 1 taste receptor (T1R) family, their expression, distribution, functional role and the molecular components of taste transduction signalling pathways (for a review, see Bachmanov & Beauchamp(Reference Bachmanov and Beauchamp18)). Chemosensing of nutrients begins in the oral cavity where taste signalling molecules are contained in epithelial cells of specialised taste buds that undergo a cascade of intracellular events leading to neurotransmitter release and activation of gustatory afferent nerve fibres(Reference Berthoud19). On the tongue, the G-protein-coupled receptors (GPCR) T1R2 and T1R3 form a heterodimeric combination to detect sweet tastants while the T1R1/T1R3 combination and the type 2 taste receptor family are responsible for amino acid (umami) and bitter tastes, respectively(Reference Chandrashekar, Mueller and Hoon20Reference Nelson, Hoon and Chandrashekar22). The vast overconsumption of sugar in the Western diet provides a possible role for sweet taste in the development of obesity. Knockout mice of either T1R2 or T1R3 have a dramatic loss of sweet taste perception, while abolishing both T1R2 and T1R3 receptors leads to a complete loss of sweet taste, demonstrating the importance of these proteins in sweet detection(Reference Zhao, Zhang and Hoon23). Furthermore, genetic variations in the genes encoding these receptors are associated with differences in sensitivity to sweet taste in both rodents and human subjects(Reference Inoue, Glendinning and Theodorides24, Reference Eny, Wolever and Corey25). An association between sugar consumption and variation in the TAS1R2 gene has recently been reported in two obese populations(Reference Eny, Wolever and Corey25). Although genetic association studies lack functional links, nevertheless, this finding suggests that increased sugar consumption may be a result of genetic variations and subsequent change in sweet taste sensitivity. Whether genetic variations in sweet tasting, either induced or spontaneous, are linked to increased obesity, or whether individuals with defects in sweet taste perception are less obese is not clear. Interestingly, obese individuals do have altered sweet taste perception(Reference Donaldson, Bennett and Baic26) and ‘liking’ for sweetness in the obese increases as a function of sweetness and BMI(Reference Bartoshuk, Duffy and Hayes27). However, Grinker and co-workers(Reference Grinker28, Reference Grinker and Hirsch29) reported no difference in the sensitivity for sweets between obese and normal-weight individuals. In rodents, obese-prone as well as obese rats lacking a functional CCK-1 receptor show an increased avidity for high concentrations of sucrose solutions compared with their lean counterparts(Reference De Jonghe, Hajnal and Covasa30, Reference Shin and Berthoud31). These obesity-induced changes in sweet taste responses were completely reversed after weight loss, suggesting that they were secondary to the obese state(Reference Shin and Berthoud31). Together, these findings underscore the importance of both the effects of genotype on sweet preferences as well as the prevailing dietary environment and pathological conditions such as obesity that can modify and account for individual differences in food preferences.

Dietary fats are also detected in the oral cavity mainly through tactile (texture) and olfactory cues, although gustatory cues have also been suggested(Reference Mattes32). Several detection mechanisms for NEFA have been reported in rodents. They act through the Kv1·5 delayed rectifying potassium channel and the fatty acid translocase CD36 (CD36), coined the putative NEFA receptor(Reference Fukuwatari, Kawada and Tsuruta33, Reference Gilbertson, Fontenot and Liu34). Localised in circumvallate and foliate taste buds(Reference Laugerette, Passilly-Degrace and Patris35), CD36 mediates preference for both long-chain fatty acids (LCFA) and TAG in rodents and deletion of its gene greatly reduces fat preference and intake in mice(Reference Laugerette, Passilly-Degrace and Patris35, Reference Sclafani, Ackroff and Abumrad36). Additionally, deletion of the gene abolishes digestive secretions initiated by orally deposited LCFA, further demonstrating its role as a lipid sensor. However, development of fat preference can be acquired independent of CD36 through other modalities such as learned associations or post-oral reinforcing actions of fat(Reference Sclafani, Ackroff and Abumrad36). Recent evidence shows that despite the reported role of CD36 in glucose and lipid metabolic abnormalities, there was no association between CD36 gene variants and obesity risks(Reference Choquet, Labrune and De Graeve37). However, diet-induced obese rodents exhibited a decreased expression of CD36(Reference Zhang, Zhou and Ban38). Thus, the degree of the direct involvement of CD36 in modulating fat intake and preference and associated metabolic disorders still requires greater evaluation.

In addition to CD36, several GPCR identified on the lingual epithelium have been shown to bind to short- (GPCR41 (NEFA2), GPCR43 (NEFA3)), medium- and long-chain (GPCR40 (NEFA1) and GPCR120) NEFA(Reference Mattes39). As with CD36, GPCR120 and GPCR40 knockout mice displayed decreased preference to LCFA(Reference Cartoni, Yasumatsu and Ohkuri40). Their presence and functions in human lingual tissue, however, are not known. Furthermore, the picture of fat taste detection in humans is less clear with no identified receptors for TAG, the main form of dietary lipids, and a lack of knowledge of possible transduction mechanisms. Further, we still do not fully understand whether lipids are processed by the somatosensory or the gustatory system. Although CD36 is expressed in human taste cells, and may be involved in dietary LCFA detection(Reference Laugerette, Passilly-Degrace and Patris35), the role of the gustatory apparatus in fat detection and preference in humans remains largely unresolved. However, recent findings have shown that hypersensitivity to lipids in human subjects was associated with a lower BMI, as well as a decreased consumption of lipids and total energy(Reference Stewart, Feinle-Bisset and Golding41). Thus, this represents a prolific and promising area for research aiming at developing strategies for modulating taste receptor functions to curb appetite given that taste is a major factor accounting for increased preference for palatable foods resulting in excess weight gain.

Gut chemosensation

The finding that the molecular sensing elements and pathways that mediate oral taste signalling are also present and operate in the GI tract(Reference Tanaka, Katsuma and Adachi42Reference Liou, Lu and Sei44) has added a new dimension to the role of the gut in controlling appetite. For example, T1R, type 2 taste receptors, α-gustducin and transient receptor potential member 5 lingual taste molecules are all expressed in the upper GI mucosa(Reference Hass, Schwarzenbacher and Breer45, Reference Bezencon, le Coutre and Damak46) and are subject to dynamic metabolic control of the intestinal luminal environment. In addition, they co-localise with gut peptide producing cells(Reference Sutherland, Young and Cooper47, Reference Rozengurt, Wu and Chen48) in the intestinal epithelium and mediate peptide release(Reference Margolskee, Dyer and Kokrashvili43, Reference Jang, Kokrashvili and Theodorakis49). Specifically, the T1R2/R3 sweet taste receptor found on the tongue is also present in enteroendocrine L-cells of mice and humans(Reference Margolskee, Dyer and Kokrashvili43, Reference Bezencon, le Coutre and Damak46, Reference Young, Sutherland and Pezos50, Reference Dyer, Salmon and Zibrik51), and is required for glucagon-like peptide-1 (GLP-1) release after a glucose load(Reference Margolskee, Dyer and Kokrashvili43). Similarly, α-gustducin co-localises with GLP-1 in the intestinal epithelium(Reference Rozengurt, Wu and Chen48), and T1R3 and α-gustducin knockout mice have impaired GLP-1 release(Reference Margolskee, Dyer and Kokrashvili43, Reference Jang, Kokrashvili and Theodorakis49). In human subjects, administration of lactisole, a T1R3 antagonist, dose-dependently decreased the glucose-stimulating release of both GLP-1 and PYY(Reference Jang, Kokrashvili and Theodorakis49). Also, specific GPCR have been identified in enteroendocrine cells, providing a mechanism for lipid-induced secretion of GI peptides. For example, GPCR120 and GPCR40 have been implicated in CCK and GLP-1 release via LCFA, while GPCR41 knockout mice have a blunted release of PYY(Reference Tanaka, Katsuma and Adachi42, Reference Liou, Lu and Sei44, Reference Hirasawa, Tsumaya and Awaji52, Reference Tanaka, Yano and Adachi53). As shown in Fig. 1, these data demonstrate that taste and nutrient receptors in the gut provide feedback information in response to luminal nutrients through mediating hormone secretion. However, the ability to manipulate these receptors and with them peptide secretion so far has proved elusive. Several groups have shown that artificial sweeteners known to bind to T1R3 were unable to stimulate GLP-1 release, while glucose is more effective than fructose in inducing release(Reference Steinert, Frey and Topfer54). Therefore, while the ability to enhance satiation signalling could be beneficial for the treatment of metabolic disorders, non-taste mechanisms are major factors influencing gut peptide release.

Gut peptides

Recent successes from bariatric surgery in achieving massive weight loss in morbidly obese patients, which are associated with enhanced secretion of anorexigenic peptides, have propelled gut hormones to the forefront of research seeking an alternative, non-surgical, treatment for obesity. A growing number of neural and humoral factors are released from the gut during feeding, and they play a prominent role in the cascade of events controlling appetite.

Ghrelin

In addition to gastric mechanoreceptors that respond vagally to stretch and tension(Reference Phillips and Powley55), the stomach is the site of release for ghrelin, the only known peripheral orexigenic hormone(Reference Tschop, Smiley and Heiman56Reference Wren, Small and Abbott58). Produced mainly by the X/A-type cells in the gastric mucosa(Reference Date, Kojima and Hosoda59, Reference Kojima, Hosoda and Date60), plasma ghrelin levels are high during fasting, greatly decreased during re-feeding and rise before the onset of a meal, defining the peptide as a possible meal initiator(Reference Cummings, Frayo and Marmonier61, Reference Cummings, Purnell and Frayo62). However, the ability of ghrelin to initiate intake has recently been contested by the fact that ghrelin levels actually peak in response to habitual meal patterns, thus rising in anticipation of meal, and perhaps better preparing the GI tract for an upcoming meal(Reference Frecka and Mattes63). Des-acyl ghrelin is the prominent form in the plasma(Reference Hosoda, Kojima and Mizushima64), but the biologically active form requires acylation by the gastric O-acyl transferase (GOAT) enzyme(Reference Yang, Brown and Liang65). Ghrelin activates NPY/AgRP neurons within the ARC(Reference Tamura, Kamegai and Shimizu66, Reference Nakazato, Murakami and Date67) through growth hormone secretagogue receptor-1a. Thus, the orexigenic effect of ghrelin is dependent on the release of NPY and AgRP and their subsequent inhibitory action on POMC neurons(Reference Chen, Trumbauer and Chen68). In addition to a central action, vagal afferents innervating the stomach express the ghrelin receptor, indicating a possible peripheral mechanism(Reference le Roux, Neary and Halsey69, Reference Date, Murakami and Toshinai70). Furthermore, ghrelin down-regulates anorexigenic peptide receptors for PYY, GLP-1 and CCK(Reference de Lartigue, Dimaline and Varro71, Reference Burdyga, Varro and Dimaline72), thus strengthening its orexigenic effects.

Although initial pharmacological data provided strong evidence on the role of ghrelin in the control of food intake, more recent studies using targeted deletion of ghrelin and its receptor(Reference Sun, Asnicar and Saha73, Reference Wortley, del Rincon and Murray74), ghrelin overexpression(Reference Gardiner, Campbell and Patterson75) or manipulation of ghrelin activation pathways(Reference Kirchner, Gutierrez and Solenberg76) have raised new and intriguing questions on the functional role of ghrelin. For example, abolishing ghrelin or ghrelin receptor activity in obese mice results in decreased food intake, body weight and adiposity and improvement in metabolic parameters(Reference Sun, Asnicar and Saha73, Reference Asakawa, Inui and Kaga77, Reference Shearman, Wang and Helmling78). Furthermore, transgenic mice overexpressing ghrelin are resistant to high-fat (HF) diet-induced obesity(Reference Gardiner, Campbell and Patterson75). This differential effect of ghrelin on feeding and obesity when animals are on a HF diet suggests a crucial role of ghrelin as a key homeostatic signal modulating energy balance and lipid metabolism. Indeed, GOAT, the enzyme responsible for ghrelin acylation, is regulated by dietary lipids, such as medium-chain fatty acids acting as substrates(Reference Kirchner, Gutierrez and Solenberg76). This suggests that the GOAT–ghrelin system acts as a lipid ‘sensor’ to inform the hypothalamus of available energy for distribution. The role of ghrelin on hypothalamic and peripheral lipid metabolism has been shown in several papers and recently reviewed(Reference Varela, Vazquez and Cordido79). Although much remains to be done in identifying the physiological and neuronal pathways of ghrelin's role under various feeding and metabolic conditions, it is clear that ghrelin has an important physiological and pathophysiological role in appetite as an anticipatory meal signal and as a signal for energy deficits. Consistent with the latter, ghrelin is a promising candidate for obesity management.

A role of ghrelin in long-term weight regulation has been suggested. For example, ghrelin increases the production of fat storage proteins, resulting in intracytoplasmic lipid accumulation(Reference Rodriguez, Gomez-Ambrosi and Catalan80), reduces fat utilisation, increases adipose tissue and promotes weight gain(Reference Tschop, Smiley and Heiman56). While individuals with the Prader–Willi syndrome have elevated levels of ghrelin even before the onset of obesity(Reference Cummings, Clement and Purnell81, Reference Feigerlova, Diene and Conte-Auriol82), an inverse correlation exists between ghrelin levels and obesity-related parameters such as BMI, visceral adiposity, hyperleptinaemia, abnormal glucose homeostasis and insulin resistance(Reference Goldstone, Thomas and Brynes83, Reference Tschop, Weyer and Tataranni84). Plasma ghrelin levels are reduced in obese individuals compared with normal-weight individuals, an effect that may result in limiting intake(Reference le Roux, Patterson and Vincent85). However, unlike lean individuals, the obese fail to significantly decrease ghrelin levels after a meal, suggesting a role for ghrelin in overconsumption due to a blunted postprandial response(Reference le Roux, Patterson and Vincent85). Adding to this, the beneficial weight loss from bariatric surgery may be due in part to changes in ghrelin release(Reference Beckman, Beckman and Earthman86). Most evidence shows that ghrelin fasting and postprandial levels are decreased significantly after gastric bypass but others have reported unchanged or even increased levels of plasma ghrelin after surgery which had been attributed to differences in pre- and post-operative conditions and surgical methods(Reference Beckman, Beckman and Earthman86). The mechanisms for reduced plasma ghrelin levels after gastric bypass are not clearly known, although the loss of ghrelin-producing cells and the absence of gastric mucosal contact with nutrients have been suggested as possible causes(Reference Kojima and Kangawa87). Additionally, gastric bypass surgery in rodents lowered growth hormone secretagogue receptor-1a protein expression in the hypothalamus, providing another mechanism for weight loss after surgery(Reference Wang and Liu88).

Approaches aimed at blocking the activity of ghrelin (e.g. anti-ghrelin vaccines)(Reference Vizcarra, Kirby and Kim89) and its receptor(Reference Asakawa, Inui and Kaga77) or inactivating the acylation process using GOAT enzyme inhibitors have all been proposed as a potential target for obesity treatment(Reference Gualillo, Lago and Dieguez90). However, ghrelin receptor antagonists have had mixed results, and although the anti-ghrelin vaccine proved effective in animal models, it failed to reduce weight in obese human subjects(Reference Zorrilla, Iwasaki and Moss91, 92). On the other hand, GOAT antagonists that reduce acyl ghrelin prove to be promising, with recent results showing decreased hunger, body weight and fat mass in HF-fed mice(Reference Barnett, Hwang and Taylor93). Furthermore, rodents treated with a ghrelin-specific RNA spiegelmer, an l-isomer oligonucleotide, which binds and blocks acylated ghrelin, have decreased food intake and body weight; however, it has yet to be tested in human subjects(Reference Shearman, Wang and Helmling78, Reference Kobelt, Helmling and Stengel94). Finally, some, but not all, linkage and genomic studies showed associations between several ghrelin variants and the obese phenotype, further implicating ghrelin as a major candidate involved in long-term energy balance (for a review, see Barnett et al. (Reference Barnett, Hwang and Taylor93)).

Cholecystokinin

In the proximal intestine, CCK released from mucosal enteroendocrine I-cells, mainly in response to fats and proteins, stimulates pancreatic secretion, bile release, gallbladder contraction, slowing of gastric emptying and inhibition of food intake, thus controlling the passage of the ingesta(Reference Moran and Kinzig95). Most of CCK's actions, including control of food intake, are mediated through CCK-1R acting through a paracrine mode of action on vagal afferent neurons(Reference Ritter, Covasa and Matson96). CCK interacts with other signals such as gastric distention(Reference Kissileff, Carretta and Geliebter97), oestradiol(Reference Asarian and Geary98), 5-hydroxytryptamine(Reference Hayes and Covasa99, Reference Hayes, Savastano and Covasa100) and leptin(Reference Peters, Simasko and Ritter101) to enhance its anorexigenic effects while also mediating the effect of other signals such as ghrelin(Reference Degen, Drewe and Piccoli102), PYY(Reference Degen, Drewe and Piccoli102, Reference Brennan, Little and Feltrin103) and apoA-IV(Reference Lo, Zhang and Pearson104).

Although predominantly viewed as a short-term satiation signal, there is also evidence that CCK plays a role in the pathogenesis of obesity in human subjects. For example, CCK-1R gene promoter polymorphism is associated with body fat(Reference Funakoshi, Miyasaka and Matsumoto105) and obese carriers of variants in the CCK gene have an increased risk of eating large portion sizes, with a 60 % increased risk for carriers of CCK_H3(Reference de Krom, van der Schouw and Hendriks106). Further, obese women have lower fasting plasma CCK concentrations and exhibit a blunt postprandial CCK response, possibly indicating a dysfunctional secretion and thus a decrease in the signalling pathway(Reference Zwirska-Korczala, Konturek and Sodowski107). However, plasma CCK concentration in obese subjects remains elevated following consumption of a fatty meal, which could lead to CCK-1R desensitisation(Reference French, Murray and Rumsey108). Manipulation of endogenous CCK levels either through diet (e.g. addition of LCFA), by inhibiting CCK degradation, or through chronic exogenous administration of CCK have all been shown to decrease energy intake and/or body weight(Reference Ritter, Covasa and Matson96). Whether these approaches can lead to sustained changes in CCK responses without the development of tolerance effects, resulting in a consistent reduction in appetite, in obese subjects requires further investigation. However, recent findings showing that CCK-58 is more potent than CCK-8 in reducing food intake(Reference Glatzle, Raybould and Kueper109) and that morbidly obese subjects have elevated CCK levels even 20 years after jejunoileal bypass(Reference Naslund, Gryback and Hellstrom110) strengthen the role of CCK as a therapeutical candidate in obesity management. Furthermore, pharmacological studies employing CCK-1R agonists have been promising, with, at least, initial studies showing a significant weight loss(Reference Roses111). Studies using longer forms of CCK, such as CCK-58 that prolong the intermeal interval(Reference Lateef, Washington and Sayegh112), may prove more beneficial in curbing intake. Lastly, CCK may have a role in long-term energy balance by interacting with leptin(Reference Matson, Reid and Cannon113Reference Emond, Schwartz and Ladenheim115) and amylin(Reference Bhavsar, Watkins and Young116). Indeed, administration of CCK with either leptin or amylin increases the magnitude of feeding suppression while CCK/leptin enhances body-weight suppression(Reference Matson, Reid and Cannon113, Reference Matson, Reid and Ritter114, Reference Bhavsar, Watkins and Young116, Reference Matson and Ritter117). Uncovering the most effective strategy of manipulating the CCK system leading to enhancement of the effects on intake, adiposity and other metabolic improvements remains a promising area of intense investigation.

Glucagon-like peptide-1

Another major hormone that exerts a profound effect on eating behaviour, GI functions, nutrient utilisation and energy homeostasis is GLP-1. GLP-1 is a post-translational product of the proglucagon gene expressed in the α-cells of the pancreas, L-cells of the small intestine and colon, and neurons in the central nervous system(Reference Holst118). Secretion of GLP-1 is governed by a neural–humoral reflex, the presence of nutrients and other endocrine factors in the intestinal tract(Reference Dube and Brubaker119). GLP-1 is present in two forms, GLP-1(1–36) and GLP-1(1–37), which undergo enzymatic cleavage to yield the bioactive forms of the peptide: GLP-1(7–36) and GLP-1(7–37)(Reference Holst, Orskov and Nielsen120, Reference Mojsov, Weir and Habener121), which enter the circulation via the lymphatic system(Reference D'Alessio, Lu and Sun122). Most of the peptide is rapidly degraded by dipeptidyl peptidase IV(Reference Mentlein, Gallwitz and Schmidt123), which results in a relatively short half-life in the circulation(Reference Deacon, Pridal and Klarskov124) and limits its effects on weight loss. GLP-1 has potent effects on (1) regulating GI functions such as gastric emptying, motility and pancreatic secretions(Reference Imeryuz, Yegen and Bozkurt125Reference Tolessa, Gutniak and Holst128), (2) suppression of food intake(Reference Donahey, van Dijk and Woods129Reference Turton, O'Shea and Gunn131) and (3) regulation of blood glucose levels by stimulating insulin secretion(Reference Larsen and Holst132). Both systemic and central exogenous GLP-1 are effective in decreasing food intake(Reference Tang-Christensen, Larsen and Goke130, Reference Turton, O'Shea and Gunn131, Reference Rodriquez de Fonseca, Navarro and Alvarez133) by acting either locally through vagal afferents or centrally through brain neurons arising from the hindbrain that maintain synaptic connections with hypothalamic areas(Reference Turton, O'Shea and Gunn131, Reference Abbott, Monteiro and Small134Reference Hayes, Leichner and Zhao136). Indeed, recent work by Kanoski et al. (Reference Kanoski, Fortin and Arnold137) shows that the suppressive effects of long-lasting GLP-1 agonists are not exclusively mediated by a peripheral action, but also require some central activation, while others have shown that blood-borne GLP-1 does not require peripheral participation(Reference Ruttimann, Arnold and Hillebrand138, Reference Zhang and Ritter139). However, the role of endogenous GLP-1 as a true satiation peptide is still contentious, and it may only have a role in decreasing appetite during times of low intake, not during large meals. This is evidenced by the fact that GLP-1R antagonism only increases intake during the light, but not the dark, cycle, which is the largest meal for a rodent(Reference Williams, Baskin and Schwartz135, Reference Ruttimann, Arnold and Geary140). Furthermore, although circulating GLP-1 remains elevated for several hours(Reference Orskov, Wettergren and Holst141), it does not appear to promote satiety, as it is unable to increase the intermeal interval(Reference Ruttimann, Arnold and Hillebrand138). GLP-1 also interacts with both central(Reference Ma, Bruning and Ashcroft142, Reference Seo, Ju and Chung143) and peripheral(Reference Asarian144, Reference Neary, Small and Druce145) peptides that control food intake, as well as with long-term energy-regulating hormones such as leptin(Reference Bojanowska and Nowak146, Reference Nowak and Bojanowska147).

In pathological states characterised by imbalanced energy homeostasis, studies have shown that while fasting levels are maintained, GLP-1 secretion is markedly decreased(Reference Naslund, Gryback and Backman148Reference Williams, Hyvarinen and Lilly150). Interestingly, though, is the fact that potency of GLP-1 appears to be maintained in obese models, although a HF diet can abate the anorexic response(Reference Williams, Hyvarinen and Lilly150, Reference Hayes, Kanoski and Alhadeff151) (F. A. Duca and M. Covasa, unpublished results). Further proof for GLP-1’s role in weight-loss treatment comes from data showing a dramatic fasting and postprandial increase in plasma levels of GLP-1 after gastric bypass surgery(Reference Beckman, Beckman and Earthman86). Whether these changes have a direct bearing on weight loss is not known, particularly given the rapid degradation of GLP-1 in the blood. However, other sources of GLP-1 signalling that avoid peptide degradation via activation of vagal afferents(Reference Abbott, Monteiro and Small134, Reference Ruttimann, Arnold and Hillebrand138) or the caudal brainstem(Reference Baggio, Huang and Brown152) or entrance into the lymphatic system(Reference D'Alessio, Lu and Sun122) may be partly responsible for its delayed effects on weight loss(Reference Berthoud, Shin and Zheng153). Given all this, several GLP-1R agonists (exenatide and liraglutide) or dipeptidyl peptidase IV inhibitors have been developed to prolong its anorexigenic effects. Although originally developed for treating diabetes, both liraglutide and exenatide produced significant weight loss(Reference Astrup, Rossner and Van Gaal154, Reference Nayak, Govindan and Baskar155). In a smaller study, dual treatment of extenatide and insulin to type 2 diabetic obese patients yielded a 12·8 % reduction in body weight after 1 year(Reference Nayak, Govindan and Baskar155). Although more studies on obese patients are needed, the initial success of GLP-1 treatment coupled with the fact that sensitivity seems to be maintained in obesity establishes GLP-1 as a promising therapeutic target.

Peptide YY

Co-secreted with GLP-1 from enteroendocrine L-cells in response to a meal(Reference Adrian, Ferri and Bacarese-Hamilton156), PYY mediates several GI functions such as inhibition of gastric emptying and secretion, GI motility, gall bladder emptying, and pancreatic and intestinal secretion. PYY(3–36) is the active and major circulating form, resulting from the cleavage of PYY(1–36) by dipeptidyl peptidase IV(Reference Eberlein, Eysselein and Schaeffer157). PYY levels are increased within 15 min following a meal and remain elevated for up to 6 h(Reference Adrian, Ferri and Bacarese-Hamilton156). The anorectic effect of centrally and peripherally administered PYY(3–36) is mediated by NPY2 receptors in the ARC, down-regulating orexigenic NPY mRNA(Reference Acuna-Goycolea and van den Pol158, Reference Batterham, Cowley and Small159) while possibly up-regulating POMC mRNA(Reference Batterham, Cowley and Small159, Reference Challis, Pinnock and Coll160), although the effect on POMC has been challenged(Reference Acuna-Goycolea and van den Pol158, Reference Challis, Coll and Yeo161, Reference Halatchev, Ellacott and Fan162). However, PYY may also act on vagal afferents that express Y2 receptors(Reference Koda, Date and Murakami163), since vagotomy abolishes the suppressive effect of exogenous PYY(Reference Abbott, Monteiro and Small134, Reference Koda, Date and Murakami163).

The fact that PYY plasma levels stay elevated long after a meal suggests a role for PYY in satiety(Reference Moran and Dailey164). Additionally, several studies have shown a positive correlation between postprandial PYY levels and ratings of satiety in human subjects(Reference Guo, Ma and Enriori165, Reference le Roux, Batterham and Aylwin166). Therefore, treatment strategies involving PYY may target lowering body weight via enhanced satiety. PYY may indeed have a role in the pathogenesis of obesity as diet-induced obese rats display reduced levels of PYY(Reference le Roux, Batterham and Aylwin166, Reference Yang, Wang and Xu167), and human studies have shown a negative correlation between fasting PYY and BMI in adults(Reference Batterham, Cohen and Ellis168), as well as a decreased postprandial response(Reference le Roux, Batterham and Aylwin166). Furthermore, PYY levels are greatly increased in both fasted and fed animals following gastric bypass surgery(Reference Chandarana, Gelegen and Karra169, Reference Shin, Zheng and Townsend170), suggesting an important role of PYY in weight loss. The increased PYY levels in gastric bypass patients can last for years(Reference Naslund, Gryback and Hellstrom110), a phenomenon attributed to alterations in L-cell functions(Reference Chandarana, Gelegen and Karra169). Finally, chronic administration of PYY reduces body weight in animal models, while PYY-null mice developed hyperphagia and increased adiposity that was subsequently reversed by PYY(3–36) treatment(Reference Batterham, Heffron and Kapoor171). However, to date, therapeutic treatments with PYY(3–36), its analogues or combination therapy have had only modest results(Reference Gantz, Erondu and Mallick172, Reference Reidelberger, Haver and Apenteng173). Nevertheless, because PYY increases the intermeal interval, it makes it an interesting peptide with potential therapeutic effects particularly when combined with other satiation peptides such as GLP-1 or oxyntomodulin to reduce long-term energy intake(Reference Neary, Small and Druce145, Reference Field, Wren and Peters174).

Other gut anorexigenic peptides

Several other peptides are released from the gut in response to nutrients which include gastrin-releasing peptide, apoA-IV, enterostatin, pancreatic polypeptide, amylin, glucagon and oxyntomodulin (OXM), to name a few. While a role for most of these peptides in obesity treatment remains uncertain and is still under consideration, the effects of OXM, which has potent anorectic, incretin and energy expenditure properties(Reference Wynne and Bloom175, Reference Wynne, Park and Small176), look more promising with recent data showing decreased food intake and sustained weight loss in diet-induced obese mice following infusion with an OXM analogue(Reference Liu, Ford and Druce177).

Gut peptide interactions

Control of food intake is orchestrated, in part, by highly complex interactions between gut peptides. Since single hormone therapy poses several challenges, including rapid peptide degradation, tolerance, redundancy and compensatory mechanisms, the use of multi-hormone therapy has proved more effective. Roth et al. showed that treatment of PYY, a GLP-1 analogue, or amylin with co-administration of leptin all decreased weight in obese rats, but only amylin and leptin had a synergistic effect. This treatment of pramlinitide (an amylin analogue) and metreleptin (recombinant leptin) elicited a weight loss of 12·7 % in obese human subjects(Reference Roth, Roland and Cole178, Reference Trevaskis, Coffey and Cole179). The addition of CCK with leptin and pramlintide may prove to be even more effective than either the two treatments alone, since CCK and leptin co-treatment in rodents synergistically reduces meal size and reduces body weight(Reference Matson, Reid and Cannon113, Reference Matson, Reid and Ritter114). Treatment with all three peptides increased weight loss by 40 % compared with just the leptin and pramlinitide combination in obese rats(Reference Trevaskis, Turek and Griffin180). Furthermore, combination of low doses of PYY(3–36) with OXM or GLP-1 in human subjects resulted in a 42·7 and 27 % reduction, respectively, in energy intake compared with controls and was significantly greater than that produced by either hormone independently(Reference Neary, Small and Druce145, Reference Field, Wren and Peters174). Thus, ‘cocktail’ treatments appear to be more effective in suppressing energy intake and sustaining weight loss. Attempts have been made at replicating complex neurohormonal responses following a meal by designing treatment combination targeting both episodic and tonic signals, such as CCK, amylin and leptin(Reference Trevaskis, Turek and Griffin180). The ability of a multi-faceted treatment to decrease meal size while simultaneously increasing the intermeal interval and background tonic signalling may result in significant energy reductions and long-term weight loss. Additionally, the synergistic property of ‘cocktail’ therapy allows for lower doses of peptides within the treatment, thus limiting potential tolerance effects normally observed with long-term single drug treatment. In summary, manipulating gut hormones through dietary or combination therapy to mimic a more complete post-ingestive response could prove an effective treatment approach to curb appetite and weight gain. However, to date, their effects in humans are largely unknown but the initial success in animals is promising.

Modulation of gut peptides by the gut luminal environment

Dietary influences

Responses to GI appetite-related signals are not fixed, and they change considerably in response to the dietary and endocrine milieu. Some changes can lead to defects in release, or functionality, of the peptides resulting in increases in food intake and ultimately obesity. For example, rats adapted to a HF diet become less sensitive to both exogenous and endogenous CCK as well as to gastric and intra-intestinal lipid loads, and exhibit hyperphagia and weight gain (for a review, see Covasa(Reference Covasa181)). The reduced sensitivity to CCK after HF exposure occurs both in rat pups(Reference Swartz, Savastano and Covasa182) and adults(Reference Covasa and Ritter183, Reference Savastano and Covasa184) and can be reversed when switched to a low-fat diet(Reference Swartz, Savastano and Covasa182). These behavioural responses have been associated with rapid physiological, enzymatic and molecular changes in CCK and CCK-dependent physiological functions and signalling pathways, as well as with reduced neuronal activation in enteric, vagal and the nucleus of the solitary tract neurons, areas densely populated with CCK-1 receptors(Reference Covasa181). Similarly, human subjects adapted to a HF diet reported greater hunger during a duodenal lipid infusion(Reference Boyd, O'Donovan and Doran185), and had increased daily food consumption and body weights(Reference Bray, Paeratakul and Popkin186). The effects of HF feeding are not limited to CCK. Similarly, long-term exposure to a HF diet resulted in both decreased sensitivity to an exogenous analogue of GLP-1 and decreased plasma GLP-1(Reference Williams, Hyvarinen and Lilly150) (F. A. Duca and M. Covasa, unpublished results). Furthermore, Chandarana et al. (Reference Chandarana, Gelegen and Karra169) have recently shown that while short-term HF diet exposure did not alter fasting circulating acyl-ghrelin, total PYY or active GLP-1 concentrations, prolonged exposure with the development of obesity significantly diminished the levels of these peptides. These lower levels of circulating peptides such as PYY may result in increased intake in obese human subjects(Reference le Roux, Batterham and Aylwin166). Obesity is often associated not only with changes in responsiveness to peripheral peptides and nutrients but also with central peptides, in several obese models. For example, HF feeding significantly increases the expression of centrally acting peptides such as orexin(Reference Wortley, Chang and Davydova187), galanin(Reference Leibowitz, Akabayashi and Wang188), AgRP(Reference Wang, Storlien and Huang189) and NPY(Reference Huang, Xin and McLennan190) in the hypothalamus. Thus, potentiating positive feedback involving orexigenic signals, coupled with decreased negative feedback from anorexigenic signals, following HF feeding may be responsible for the overconsumption on a HF diet. It is clear that the obese state is associated with changes in hormone release induced by food intake, but it remains uncertain how obesity influences hormone concentrations or changes in sensitivity to the hormones that might exacerbate the obese condition.

Microbiota influences

In addition to the presence of nutrients, the intestinal epithelium comes in direct contact with trillions of diverse, complex bacteria and other micro-organisms collectively termed the microbiota. Growing evidence demonstrates that the gut microbiota contributes to the development of diet-induced obesity(Reference Ley, Turnbaugh and Klein191Reference Turnbaugh, Ridaura and Faith193). For example, colonisation of adult germ-free mice with a distal gut microbial community harvested from conventionally raised mice leads to a dramatic increase in body fat within 10–14 d, despite an associated decrease in food consumption and increased energy expenditure(Reference Backhed, Ding and Wang194). Additionally, germ-free mice are resistant to diet-induced obesity when fed a HF/high-sugar ‘Western’ diet(Reference Backhed, Manchester and Semenkovich195, Reference Rabot, Membrez and Bruneau196). Interestingly, the obese phenotype is transmissible: germ-free mice that receive an ‘obese microbiota’ display significantly greater fat mass than those that received a ‘lean microbiota’(Reference Turnbaugh, Ley and Mahowald197). Finally, the diet can profoundly alter the composition of the gut microbial population(Reference de La Serre, Ellis and Lee198Reference Mozes, Bujnakova and Sefcikova200), possibly contributing to weight gain.

In addition to its profound effect on modulating host energy homeostasis and metabolism(Reference Musso, Gambino and Cassader201), as shown in Fig. 1, there is evidence that microbiota-generated by-products affect the functional expression of intestinal nutrient-responsive GPCR(Reference Samuel, Shaito and Motoike202, Reference Dewulf, Cani and Neyrinck203), GI hormones(Reference Cani and Delzenne204), and nutrient transport and taste(Reference Swartz, Duca and de Wouters205). Studies examining a direct role of the microbiota in the control of food intake and body adiposity are in infancy; however, indirect evidence suggests a role of the gut microbiota in the secretion and function of GI peptides, such as 5-hydroxytryptamine, GLP-1, GLP-2, PYY and ghrelin(Reference Cani, Montoya and Neyrinck206Reference Wikoff, Anfora and Liu209). Additionally, obesity has been associated with diet-induced, low-grade gut inflammation or the ‘metabolic endotoxaemia’ condition resulting from a substantial increase in bacterially derived lipopolysaccharide and increased gut permeability, a condition improved by altering the gut microbiota(Reference Cani and Delzenne210), involving GLP-2(Reference Cani, Possemiers and Van de Wiele207)- and endocannabinoid(Reference Muccioli, Naslain and Backhed211)-dependent mechanisms. This implicates the gut microbes as targets in metabolic disorders such as obesity and diabetes. As such, decreasing inflammation by increasing microbial fermentation, either through the diet or the aid of prebiotics, results in lowered appetite, elevated plasma levels of GLP-1(Reference Cani, Montoya and Neyrinck206, Reference Delzenne, Cani and Daubioul208), GLP-2(Reference Cani, Possemiers and Van de Wiele207) and PYY(Reference Delzenne, Cani and Daubioul208, Reference Gee and Johnson212), and decreased levels of ghrelin(Reference Cani, Montoya and Neyrinck206). Thus, the microbiota profile can be modified in the interest of improving metabolic parameters such as glucose homeostasis and leptin sensitivity, and to control the activity of gut hormones through its effects on enteroendocrine cell number and increased cell differentiation(Reference Everard, Lazarevic and Derrien213). Furthermore, SCFA, by-products of polysaccharide degradation by the gut microbiota, are ligands for intestinal GPCR which are candidate mechanisms for peptide release(Reference Le Poul, Loison and Struyf214). They induce enhancement in colonic motility via 5-hydroxytryptamine release(Reference Wikoff, Anfora and Liu209, Reference Uribe, Alam and Johansson215) and stimulate leptin and PYY secretion(Reference Samuel, Shaito and Motoike202). We have recently shown that mice devoid of the gut microbiota exhibit altered expression of lingual and intestinal epithelium GPCR for both sweet and lipid tastants(Reference Swartz, Duca and de Wouters205) (F. A. Duca and M. Covasa, unpublished results). Furthermore, germ-free mice have decreased expression of the intestinal satiety peptides CCK, GLP-1 and PYY and lower levels of circulating leptin, PYY and ghrelin. These changes were associated with altered preference for, and intake of, sugars and oils(Reference Swartz, Duca and de Wouters205) (F. A. Duca and M. Covasa, unpublished results). These data show that the microbiota has a potent modulatory role for the signalling elements known to be involved in the control of food intake and regulation of energy balance, resulting in behavioural changes. Thus, microbial components target molecular regulatory systems with a major role in metabolism, nutrient sensing and absorption, gut barrier integrity, gut hormones, systemic inflammation and fat tissue metabolism. The precise mechanisms responsible for these changes including their overall significance as it relates to weight gain are largely unknown, although several mechanisms have been put forward mainly involving the suppression of the intestinal lipoprotein lipase inhibitor Fiaf, inactivation of AMP-activated protein kinase pathways and efficient energy extraction from complex carbohydrates(Reference Musso, Gambino and Cassader201). Nevertheless, it is evident that changes in the gut microbiome with a shift towards improving efficiency of energy extraction and excess energy availability are neither sufficient nor can they explain the dramatic rise in the obesity epidemic within the past years. Despite major advances at the host–microbial interface and the intriguing link with the host metabolic phenotypes such as obesity and diabetes, significant work still lies ahead in deciphering the mechanisms by which the microbiota affects the regulatory systems governing energy homeostasis. Studies so far have generated more questions than answers. A major challenge is the inherent difficulty of teasing apart the complex interactions between the microbiota and the host at multiple levels that expand through several physiological and neural systems from the periphery to the brain. Because of this, some previous results are inconclusive, even controversial, with multiple confounding variables making it difficult to distinguish and separate the effect from the cause or contributing factors. Thus, it is imperative that future studies (1) uncover the identity of specific species of bacteria that are associated with obesity, (2) identify the molecular targets and understand the mechanisms of action, and (3) develop the delivery tools, including ‘targeted’ prebiotic and probiotic treatment to manipulate the microbiota profile acting on specific pathways to sustain desired intake and weight, and alleviate metabolic parameters associated with obesity. This may be especially important, since current exogenous peptide administration treatments can induce side effects and have short-lived success rates from developed tolerance, while the option of altering endogenous GI peptide levels through microbiota manipulations could be safer and long-lasting. Nevertheless, based on the existing body of evidence, the gut microbiota qualifies as an important additional factor to an already complex and redundant homeostatic and appetite-controlling system, which, undoubtedly, adds a new and critical dimension to our understanding of the role of the gut in the control of food intake, obesity and associated metabolic disorders.

Hedonic influences and interactions with homeostatic controls

Despite the strong regulation of the homeostatic system, it has become apparent that the non-homeostatic (hedonic or food reward) system plays an integral role in feeding behaviour. In obesity, an increased motivation to eat overrides homeostatic regulation resulting in sustained and escalating overeating despite normal or excessive energy storage. Palatable foods can be overconsumed for their pleasurable effects, with hedonic responses generated in the cortico-limbic structures overriding physiological control of appetite (for reviews on the hedonic system, see reports by several researchers(Reference Berthoud, Lenard and Shin216Reference Kenny218)). Interestingly, obesity is associated with alterations in the reward system, specifically the mesolimbic dopamine (DA) pathway, possibly resulting in overeating of palatable foods(Reference Shin and Berthoud31). For instance, obese individuals have blunted striatal activation in response to highly palatable foods, and individuals with a point mutation in the DA D2 receptor (D2R) are predisposed to obesity and substance abuse(Reference Stice, Spoor and Bohon219, Reference Stice, Spoor and Bohon220). In line with this, D2R levels are decreased in several models of obesity, and obesity-prone rats have lower basal and stimulated DA levels(Reference Geiger, Behr and Frank221). Furthermore, sites in the lateral hypothalamus which receive input from hedonic striatal projections are less responsive during obesity and overeating, and striatal D2R knockdown rats have blunted rewarding lateral hypothalamus stimulation(Reference Johnson and Kenny222). Thus, obesity is associated with hyporesponsivity of the mesolimbic dopamine pathway possibly through decreased D2R signalling, leading to overconsumption in order to alleviate deficits in reward signalling. However, it is still debated as to whether these alterations are a cause or consequence of the obese state(Reference Kenny218). Recent work comparing individuals with a low or high risk for obesity development shows that normal-weight individuals predisposed to obesity exhibit elevated dorsal striatum responses to food reward(Reference Stice, Yokum and Burger223, Reference Stice, Yokum and Bohon224). This led to the suggestion that reduced dorsal striatum responses to food and decreased D2R availability reported in the obese are indicative of a consequence rather than the cause of hyperphagia(Reference Stice, Yokum and Blum225). Thus, the current ‘feed-forward’ model postulates that increased initial responses of the DA system reflect enhanced responses to palatable foods and subsequent overeating. This, in turn, leads to the down-regulation of DA signalling, abrogating striatal responses to food intake and a return to prior reward experience from increased consumption.

While the reward system can override homeostatic signals, we also know there is a high degree of interaction between the two systems that influence appetite behaviour. For example, activation of the leptin receptor in the ventral tegmental area inhibits DA neurons and subsequent food intake, while leptin decreases both basal and food-evoked levels of DA(Reference Krugel, Schraft and Kittner226, Reference Hommel, Trinko and Sears227). However, leptin appears to be necessary for normal mesolimbic DA signalling, as ob/ob mice have reduced DA production and decreased DA release in the nucleus accumbens, which are corrected with leptin treatment(Reference Fulton, Pissios and Manchon228). More importantly, leptin resistance also results in altered DA signalling, indicating that while leptin is necessary for normal DA signalling, exogenously administered and obesity-induced leptin resistance causes hyposensitivity of the mesolimbic DA system(Reference Geiger, Behr and Frank221, Reference Pfaffly, Michaelides and Wang229). On the other hand, leptin is also regulated by D2R activation, as the injection of a D2R agonist reduces leptin levels, and D2R knockout mice have increased leptin signalling and sensitivity(Reference Kim, Yoon and Lee230). Furthermore, studies show that ghrelin stimulates DA release and increases activity in reward areas of the brain in human subjects(Reference Naleid, Grace and Cummings231, Reference Malik, McGlone and Bedrossian232). Thus, in addition to increased intake through alterations in homeostatic signalling, leptin resistance can cause an inopportune hyposensitivity of the hedonic system possibly exacerbating energy excess. Taken together, it is clear that a significant interaction between the homeostatic and hedonic systems occurs to affect food intake, yet potent hedonic signalling can overcome physiological appetite signals resulting in overeating. On the other hand, DA circuitries, for example, are a major site of receipt and convergence of post-oral, metabolic, hormonal and visceral cues that interact with and modulate cognitive and reward functions that drive consumption. A clear understanding of controlling this delicate reciprocal balance between the hedonic and homeostatic processes to meet energy needs, as well its dynamic and complex neurocircuitries in altered food intake and obesity, is still lacking and under intense investigation.

Perspectives and conclusions

The knowledge on the role of the GI tract in the control of food intake and the regulation of body weight is rapidly evolving. Our constant discoveries of the complexity of systems and pathways involved in this process have moved the field beyond the rather simplistic view that obesity is a simple result of excess energy balance. Clearly, food consumption is controlled by a multitude of complex factors involving metabolic and hedonic components that converge and interact at various levels of the gut–brain axis. Disruptions in both the homeostatic and hedonic systems can result in chronic positive energy balance and ensuing obesity. Although significant progress has been made recently in identifying the neural substrates and brain circuitries involved in responses to overconsumption of palatable food and weight gain, it is not clear how precisely the hedonic system influences food intake or yet how it overcomes the homeostatic system. Similarly, the influence of intrinsic or diet-induced alterations on the responsiveness of the reward and metabolic systems, and how these effects contribute to overeating and obesity, remains unclear. The evidence that oral and post-oral signals are disrupted in obesity has generated interest in developing therapeutic strategies against obesity. It has become increasingly obvious that monotherapies, or by targeting single homeostatic or hedonic components, are largely insufficient and ultimately ineffectual in producing the desired weight-loss effects. Thus, taking advantage of the interactions that normally occur between various gut hormones involved in appetite and energy regulation proves a more promising strategy in controlling meal size and subsequent weight gain. Using combination therapies, it may be possible to simulate physiological levels of key GI signals, such as those observed in bariatric surgery patients with severe weight loss. The therapeutic success of combining short- and long-term signals in rodents warrants further investigation, and long-term human clinical studies should be conducted as these ‘cocktail’ therapies may prove to be the most effective treatment for sustained weight loss. Furthermore, diet has a major role in modulating the release and action of gut hormones and dietary manipulations can result in changes in sensitivity to both hedonic and metabolic appetite-related signals that affect the behavioural control of intake. More work should be directed towards understanding how diet-induced adaptational changes can lead to deficits in the sensitivity of the reward as well as post-absorptive metabolical signalling, and how these effects contribute to overeating and obesity. At the same time, studies examining factors controlling food intake need to consider the profound impact that alterations in energy balance exert on hormone and nutrient levels which in turn influence brain regions involved in ingestive behaviour, thereby perpetuating obesity. In line with this, critical areas for future research should also involve investigating potential mechanisms that predispose individuals to obesity. This will provide useful insights into inter-individual variability in the aetiology of obesity.

Finally, the microbiota residing in the GI tract has a major impact on enteroendocrine gut functions and molecular chemosensory machinery that influence host physiology and metabolism, and affect adiposity and obesity. Future work should focus on understanding the intricate mechanisms by which nutrients and nutrient-sensing molecules, non-nutrient tastants, as well as microflora all affect GI peptide secretion, and how this can be modulated in the face of constant changes of the gut environment to control ingestion. For instance, recent work in rodents with prebiotic treatment shows significant decreases in inflammation and weight; however, studies involving long-term prebiotic treatment in human subjects are scarce and needed. Thus, developing effective treatments for obesity will ultimately require a comprehensive understanding of the complexity of systems that regulate body weight and their interactions. Although much remains to be done, the novel discoveries and experimental approaches captured in the present review will undoubtedly continue to raise more questions and pose new challenges in our quest to finding a cure to curb obesity.

Acknowledgements

The review was supported by INRA through a scientific package awarded to M. C. and a graduate fellowship to F. A. D. F. A. D. and M. C. wrote the manuscript. The authors declare that they have no conflict of interest.

References

1Berthoud, HR (2005) Brain, appetite and obesity. Physiol Behav 85, 12.CrossRefGoogle ScholarPubMed
2Woods, SC (2005) Signals that influence food intake and body weight. Physiol Behav 86, 709716.CrossRefGoogle ScholarPubMed
3Blundell, JE (2006) Perspective on the central control of appetite. Obesity (Silver Spring) 14, Suppl. 4, 160S163S.CrossRefGoogle ScholarPubMed
4Woods, SC (2009) The control of food intake: behavioral versus molecular perspectives. Cell Metab 9, 489498.CrossRefGoogle ScholarPubMed
5Neary, MT & Batterham, RL (2009) Gut hormones: implications for the treatment of obesity. Pharmacol Ther 124, 4456.CrossRefGoogle ScholarPubMed
6Faulconbridge, LF, Cummings, DE, Kaplan, JM, et al. (2003) Hyperphagic effects of brainstem ghrelin administration. Diabetes 52, 22602265.CrossRefGoogle ScholarPubMed
7Grill, HJ (2010) Leptin and the systems neuroscience of meal size control. Front Neuroendocrinol 31, 6178.CrossRefGoogle ScholarPubMed
8Sexton, PM, Paxinos, G, Kenney, MA, et al. (1994) In vitro autoradiographic localization of amylin binding sites in rat brain. Neuroscience 62, 553567.CrossRefGoogle ScholarPubMed
9Grill, HJ (2006) Distributed neural control of energy balance: contributions from hindbrain and hypothalamus. Obesity (Silver Spring) 14, Suppl. 5, 216S221S.CrossRefGoogle ScholarPubMed
10Banks, WA, Kastin, AJ, Huang, W, et al. (1996) Leptin enters the brain by a saturable system independent of insulin. Peptides 17, 305311.CrossRefGoogle ScholarPubMed
11Burguera, B & Couce, ME (2001) Leptin access into the brain: a saturated transport mechanism in obesity. Physiol Behav 74, 717720.CrossRefGoogle Scholar
12Moran, TH (2009) Hypothalamic nutrient sensing and energy balance. Forum Nutr 63, 94101.CrossRefGoogle ScholarPubMed
13Munzberg, H (2008) Differential leptin access into the brain – a hierarchical organization of hypothalamic leptin target sites? Physiol Behav 94, 664669.CrossRefGoogle Scholar
14Blevins, JE & Baskin, DG (2010) Hypothalamic-brainstem circuits controlling eating. Forum Nutr 63, 133140.CrossRefGoogle ScholarPubMed
15Langhans, W & Geary, N (2010) Overview of the physiological control of eating. Forum Nutr 63, 953.CrossRefGoogle ScholarPubMed
16Lenard, NR & Berthoud, HR (2008) Central and peripheral regulation of food intake and physical activity: pathways and genes. Obesity (Silver Spring) 16, Suppl. 3, S11S22.CrossRefGoogle ScholarPubMed
17Rolls, BJ (2003) The supersizing of America: portion size and the obesity epidemic. Nutr Today 38, 4253.CrossRefGoogle ScholarPubMed
18Bachmanov, AA & Beauchamp, GK (2007) Taste receptor genes. Annu Rev Nutr 27, 389414.CrossRefGoogle ScholarPubMed
19Berthoud, H-R (2002) Multiple neural systems controlling food intake and body weight. Neurosci Biobehav Rev 26, 393428.CrossRefGoogle ScholarPubMed
20Chandrashekar, J, Mueller, KL, Hoon, MA, et al. (2000) T2Rs function as bitter taste receptors. Cell 100, 703711.CrossRefGoogle ScholarPubMed
21Li, X, Staszewski, L, Xu, H, et al. (2002) Human receptors for sweet and umami taste. Proc Natl Acad Sci U S A 99, 46924696.CrossRefGoogle ScholarPubMed
22Nelson, G, Hoon, MA, Chandrashekar, J, et al. (2001) Mammalian sweet taste receptors. Cell 106, 381390.CrossRefGoogle ScholarPubMed
23Zhao, GQ, Zhang, Y, Hoon, MA, et al. (2003) The receptors for mammalian sweet and umami taste. Cell 115, 255266.CrossRefGoogle ScholarPubMed
24Inoue, M, Glendinning, JI, Theodorides, ML, et al. (2007) Allelic variation of the Tas1r3 taste receptor gene selectively affects taste responses to sweeteners: evidence from 129.B6-Tas1r3 congenic mice. Physiol Genomics 32, 8294.CrossRefGoogle ScholarPubMed
25Eny, KM, Wolever, TM, Corey, PN, et al. (2010) Genetic variation in TAS1R2 (Ile191Val) is associated with consumption of sugars in overweight and obese individuals in 2 distinct populations. Am J Clin Nutr 92, 15011510.CrossRefGoogle ScholarPubMed
26Donaldson, LF, Bennett, L, Baic, S, et al. (2009) Taste and weight: is there a link? Am J Clin Nutr 90, 800S803S.CrossRefGoogle ScholarPubMed
27Bartoshuk, LM, Duffy, VB, Hayes, JE, et al. (2006) Psychophysics of sweet and fat perception in obesity: problems, solutions and new perspectives. Philos Trans R Soc Lond B Biol Sci 361, 11371148.CrossRefGoogle ScholarPubMed
28Grinker, J (1978) Obesity and sweet taste. Am J Clin Nutr 31, 10781087.CrossRefGoogle ScholarPubMed
29Grinker, J & Hirsch, J (1972) Metabolic and behavioural correlates of obesity. Ciba Found Symp 8, 349369.Google ScholarPubMed
30De Jonghe, BC, Hajnal, A & Covasa, M (2005) Increased oral and decreased intestinal sensitivity to sucrose in obese, prediabetic CCK-A receptor-deficient OLETF rats. Am J Physiol Regul Integr Comp Physiol 288, R292R300.CrossRefGoogle ScholarPubMed
31Shin, AC & Berthoud, HR (2011) Food reward functions as affected by obesity and bariatric surgery. Int J Obes (Lond) 35, Suppl. 3, S40S44.CrossRefGoogle ScholarPubMed
32Mattes, RD (2011) Oral fatty acid signaling and intestinal lipid processing: support and supposition. Physiol Behav 105, 2735.CrossRefGoogle ScholarPubMed
33Fukuwatari, T, Kawada, T, Tsuruta, M, et al. (1997) Expression of the putative membrane fatty acid transporter (FAT) in taste buds of the circumvallate papillae in rats. FEBS Lett 414, 461464.Google ScholarPubMed
34Gilbertson, TA, Fontenot, DT, Liu, L, et al. (1997) Fatty acid modulation of K+ channels in taste receptor cells: gustatory cues for dietary fat. Am J Physiol 272, C1203C1210.CrossRefGoogle ScholarPubMed
35Laugerette, F, Passilly-Degrace, P, Patris, B, et al. (2005) CD36 involvement in orosensory detection of dietary lipids, spontaneous fat preference, and digestive secretions. J Clin Invest 115, 31773184.CrossRefGoogle ScholarPubMed
36Sclafani, A, Ackroff, K & Abumrad, NA (2007) CD36 gene deletion reduces fat preference and intake but not post-oral fat conditioning in mice. Am J Physiol Regul Integr Comp Physiol 293, R1823R1832.CrossRefGoogle Scholar
37Choquet, H, Labrune, Y, De Graeve, F, et al. (2010) Lack of association of CD36 SNPs with early onset obesity: a meta-analysis in 9,973 European subjects. Obesity 19, 833839.CrossRefGoogle Scholar
38Zhang, XJ, Zhou, LH, Ban, X, et al. (2011) Decreased expression of CD36 in circumvallate taste buds of high-fat diet induced obese rats. Acta Histochem 113, 663667.CrossRefGoogle ScholarPubMed
39Mattes, RD (2009) Is there a fatty acid taste? Annu Rev Nutr 29, 305327.CrossRefGoogle Scholar
40Cartoni, C, Yasumatsu, K, Ohkuri, T, et al. (2010) Taste preference for fatty acids is mediated by GPR40 and GPR120. J Neurosci 30, 83768382.CrossRefGoogle ScholarPubMed
41Stewart, JE, Feinle-Bisset, C, Golding, M, et al. (2010) Oral sensitivity to fatty acids, food consumption and BMI in human subjects. Br J Nutr 104, 145152.CrossRefGoogle ScholarPubMed
42Tanaka, T, Katsuma, S, Adachi, T, et al. (2008) Free fatty acids induce cholecystokinin secretion through GPR120. Naunyn Schmiedebergs Arch Pharmacol 377, 523527.CrossRefGoogle ScholarPubMed
43Margolskee, RF, Dyer, J, Kokrashvili, Z, et al. (2007) T1R3 and gustducin in gut sense sugars to regulate expression of Na+-glucose cotransporter 1. Proc Natl Acad Sci U S A 104, 1507515080.CrossRefGoogle ScholarPubMed
44Liou, AP, Lu, X, Sei, Y, et al. (2010) The G-protein-coupled receptor GPR40 directly mediates long-chain fatty acid-induced secretion of cholecystokinin. Gastroenterology 140, 903912.CrossRefGoogle ScholarPubMed
45Hass, N, Schwarzenbacher, K & Breer, H (2010) T1R3 is expressed in brush cells and ghrelin-producing cells of murine stomach. Cell Tissue Res 339, 493504.CrossRefGoogle ScholarPubMed
46Bezencon, C, le Coutre, J & Damak, S (2007) Taste-signaling proteins are coexpressed in solitary intestinal epithelial cells. Chem Senses 32, 4149.CrossRefGoogle ScholarPubMed
47Sutherland, K, Young, RL, Cooper, NJ, et al. (2007) Phenotypic characterization of taste cells of the mouse small intestine. Am J Physiol Gastrointest Liver Physiol 292, G1420G1428.CrossRefGoogle ScholarPubMed
48Rozengurt, N, Wu, SV, Chen, MC, et al. (2006) Colocalization of the alpha-subunit of gustducin with PYY and GLP-1 in L cells of human colon. Am J Physiol Gastrointest Liver Physiol 291, G792G802.CrossRefGoogle Scholar
49Jang, HJ, Kokrashvili, Z, Theodorakis, MJ, et al. (2007) Gut-expressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proc Natl Acad Sci U S A 104, 1506915074.CrossRefGoogle ScholarPubMed
50Young, RL, Sutherland, K, Pezos, N, et al. (2009) Expression of taste molecules in the upper gastrointestinal tract in humans with and without type 2 diabetes. Gut 58, 337346.CrossRefGoogle ScholarPubMed
51Dyer, J, Salmon, KS, Zibrik, L, et al. (2005) Expression of sweet taste receptors of the T1R family in the intestinal tract and enteroendocrine cells. Biochem Soc Trans 33, 302305.CrossRefGoogle ScholarPubMed
52Hirasawa, A, Tsumaya, K, Awaji, T, et al. (2005) Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat Med 11, 9094.CrossRefGoogle ScholarPubMed
53Tanaka, T, Yano, T, Adachi, T, et al. (2008) Cloning and characterization of the rat free fatty acid receptor GPR120: in vivo effect of the natural ligand on GLP-1 secretion and proliferation of pancreatic beta cells. Naunyn Schmiedebergs Arch Pharmacol 377, 515522.CrossRefGoogle ScholarPubMed
54Steinert, RE, Frey, F, Topfer, A, et al. (2011) Effects of carbohydrate sugars and artificial sweeteners on appetite and the secretion of gastrointestinal satiety peptides. Br J Nutr 105, 13201328.CrossRefGoogle ScholarPubMed
55Phillips, RJ & Powley, TL (2000) Tension and stretch receptors in gastrointestinal smooth muscle: re-evaluating vagal mechanoreceptor electrophysiology. Brain Res Brain Res Rev 34, 126.CrossRefGoogle ScholarPubMed
56Tschop, M, Smiley, DL & Heiman, ML (2000) Ghrelin induces adiposity in rodents. Nature 407, 908913.CrossRefGoogle ScholarPubMed
57Wren, AM, Seal, LJ, Cohen, MA, et al. (2001) Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab 86, 5992.CrossRefGoogle ScholarPubMed
58Wren, AM, Small, CJ, Abbott, CR, et al. (2001) Ghrelin causes hyperphagia and obesity in rats. Diabetes 50, 25402547.CrossRefGoogle ScholarPubMed
59Date, Y, Kojima, M, Hosoda, H, et al. (2000) Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology 141, 42554261.CrossRefGoogle Scholar
60Kojima, M, Hosoda, H, Date, Y, et al. (1999) Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402, 656660.CrossRefGoogle ScholarPubMed
61Cummings, DE, Frayo, RS, Marmonier, C, et al. (2004) Plasma ghrelin levels and hunger scores in humans initiating meals voluntarily without time- and food-related cues. Am J Physiol Endocrinol Metab 287, E297E304.CrossRefGoogle ScholarPubMed
62Cummings, DE, Purnell, JQ, Frayo, RS, et al. (2001) A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 50, 17141719.CrossRefGoogle ScholarPubMed
63Frecka, JM & Mattes, RD (2008) Possible entrainment of ghrelin to habitual meal patterns in humans. Am J Physiol Gastrointest Liver Physiol 294, G699G707.CrossRefGoogle ScholarPubMed
64Hosoda, H, Kojima, M, Mizushima, T, et al. (2003) Structural divergence of human ghrelin. Identification of multiple ghrelin-derived molecules produced by post-translational processing. J Biol Chem 278, 6470.CrossRefGoogle ScholarPubMed
65Yang, J, Brown, MS, Liang, G, et al. (2008) Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone. Cell 132, 387396.CrossRefGoogle ScholarPubMed
66Tamura, H, Kamegai, J, Shimizu, T, et al. (2002) Ghrelin stimulates GH but not food intake in arcuate nucleus ablated rats. Endocrinology 143, 32683275.CrossRefGoogle Scholar
67Nakazato, M, Murakami, N, Date, Y, et al. (2001) A role for ghrelin in the central regulation of feeding. Nature 409, 194198.CrossRefGoogle ScholarPubMed
68Chen, HY, Trumbauer, ME, Chen, AS, et al. (2004) Orexigenic action of peripheral ghrelin is mediated by neuropeptide Y and agouti-related protein. Endocrinology 145, 26072612.CrossRefGoogle ScholarPubMed
69le Roux, CW, Neary, NM, Halsey, TJ, et al. (2005) Ghrelin does not stimulate food intake in patients with surgical procedures involving vagotomy. J Clin Endocrinol Metab 90, 45214524.CrossRefGoogle ScholarPubMed
70Date, Y, Murakami, N, Toshinai, K, et al. (2002) The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats. Gastroenterology 123, 11201128.CrossRefGoogle ScholarPubMed
71de Lartigue, G, Dimaline, R, Varro, A, et al. (2007) Cocaine- and amphetamine-regulated transcript: stimulation of expression in rat vagal afferent neurons by cholecystokinin and suppression by ghrelin. J Neurosci 27, 28762882.CrossRefGoogle ScholarPubMed
72Burdyga, G, Varro, A, Dimaline, R, et al. (2006) Ghrelin receptors in rat and human nodose ganglia: putative role in regulating CB-1 and MCH receptor abundance. Am J Physiol Gastrointest Liver Physiol 290, G1289G1297.CrossRefGoogle ScholarPubMed
73Sun, Y, Asnicar, M, Saha, PK, et al. (2006) Ablation of ghrelin improves the diabetic but not obese phenotype of ob/ob mice. Cell Metab 3, 379386.CrossRefGoogle Scholar
74Wortley, KE, del Rincon, JP, Murray, JD, et al. (2005) Absence of ghrelin protects against early-onset obesity. J Clin Invest 115, 35733578.CrossRefGoogle ScholarPubMed
75Gardiner, JV, Campbell, D, Patterson, M, et al. (2010) The hyperphagic effect of ghrelin is inhibited in mice by a diet high in fat. Gastroenterology 138, 24682476, 2476 e1.CrossRefGoogle ScholarPubMed
76Kirchner, H, Gutierrez, JA, Solenberg, PJ, et al. (2009) GOAT links dietary lipids with the endocrine control of energy balance. Nat Med 15, 741745.CrossRefGoogle ScholarPubMed
77Asakawa, A, Inui, A, Kaga, T, et al. (2003) Antagonism of ghrelin receptor reduces food intake and body weight gain in mice. Gut 52, 947952.CrossRefGoogle ScholarPubMed
78Shearman, LP, Wang, SP, Helmling, S, et al. (2006) Ghrelin neutralization by a ribonucleic acid-SPM ameliorates obesity in diet-induced obese mice. Endocrinology 147, 15171526.CrossRefGoogle ScholarPubMed
79Varela, L, Vazquez, MJ, Cordido, F, et al. (2011) Ghrelin and lipid metabolism: key partners in energy balance. J Mol Endocrinol 46, R43R63.Google ScholarPubMed
80Rodriguez, A, Gomez-Ambrosi, J, Catalan, V, et al. (2009) Acylated and desacyl ghrelin stimulate lipid accumulation in human visceral adipocytes. Int J Obes (Lond) 33, 541552.CrossRefGoogle ScholarPubMed
81Cummings, DE, Clement, K, Purnell, JQ, et al. (2002) Elevated plasma ghrelin levels in Prader Willi syndrome. Nat Med 8, 643644.CrossRefGoogle ScholarPubMed
82Feigerlova, E, Diene, G, Conte-Auriol, F, et al. (2008) Hyperghrelinemia precedes obesity in Prader-Willi syndrome. J Clin Endocrinol Metab 93, 28002805.CrossRefGoogle ScholarPubMed
83Goldstone, AP, Thomas, EL, Brynes, AE, et al. (2004) Elevated fasting plasma ghrelin in Prader-Willi syndrome adults is not solely explained by their reduced visceral adiposity and insulin resistance. J Clin Endocrinol Metab 89, 17181726.CrossRefGoogle Scholar
84Tschop, M, Weyer, C, Tataranni, PA, et al. (2001) Circulating ghrelin levels are decreased in human obesity. Diabetes 50, 707709.CrossRefGoogle ScholarPubMed
85le Roux, CW, Patterson, M, Vincent, RP, et al. (2005) Postprandial plasma ghrelin is suppressed proportional to meal calorie content in normal-weight but not obese subjects. J Clin Endocrinol Metab 90, 10681071.CrossRefGoogle Scholar
86Beckman, LM, Beckman, TR & Earthman, CP (2010) Changes in gastrointestinal hormones and leptin after Roux-en-Y gastric bypass procedure: a review. J Am Diet Assoc 110, 571584.CrossRefGoogle ScholarPubMed
87Kojima, M & Kangawa, K (2005) Ghrelin: structure and function. Physiol Rev 85, 495522.CrossRefGoogle ScholarPubMed
88Wang, Y & Liu, J (2010) Combination of bypassing stomach and vagus dissection in high-fat diet-induced obese rats – a long-term investigation. Obes Surg 20, 375379.CrossRefGoogle ScholarPubMed
89Vizcarra, JA, Kirby, JD, Kim, SK, et al. (2007) Active immunization against ghrelin decreases weight gain and alters plasma concentrations of growth hormone in growing pigs. Domest Anim Endocrinol 33, 176189.CrossRefGoogle ScholarPubMed
90Gualillo, O, Lago, F & Dieguez, C (2008) Introducing GOAT: a target for obesity and anti-diabetic drugs? Trends Pharmacol Sci 29, 398401.CrossRefGoogle ScholarPubMed
91Zorrilla, EP, Iwasaki, S, Moss, JA, et al. (2006) Vaccination against weight gain. Proc Natl Acad Sci U S A 103, 1322613231.CrossRefGoogle ScholarPubMed
92 Cytos Biotechnology. (2006) Phase I/IIa clinical trial with obese individuals shows no effect of CYT009-GhrQb on weight loss Cytos Biotechnology: Media Release [serial on the Internet]. http://www.cytos.com/doc/Cytos_Press_E_061107.pdf.Google Scholar
93Barnett, BP, Hwang, Y, Taylor, MS, et al. (2010) Glucose and weight control in mice with a designed ghrelin O-acyltransferase inhibitor. Science 330, 16891692.CrossRefGoogle ScholarPubMed
94Kobelt, P, Helmling, S, Stengel, A, et al. (2006) Anti-ghrelin Spiegelmer NOX-B11 inhibits neurostimulatory and orexigenic effects of peripheral ghrelin in rats. Gut 55, 788792.CrossRefGoogle ScholarPubMed
95Moran, TH & Kinzig, KP (2004) Gastrointestinal satiety signals II. Cholecystokinin. Am J Physiol Gastrointest Liver Physiol 286, G183G188.CrossRefGoogle ScholarPubMed
96Ritter, RC, Covasa, M & Matson, CA (1999) Cholecystokinin: proofs and prospects for involvement in control of food intake and body weight. Neuropeptides 33, 387399.CrossRefGoogle ScholarPubMed
97Kissileff, HR, Carretta, JC, Geliebter, A, et al. (2003) Cholecystokinin and stomach distension combine to reduce food intake in humans. Am J Physiol Regul Integr Comp Physiol 285, R992R998.CrossRefGoogle ScholarPubMed
98Asarian, L & Geary, N (2007) Estradiol enhances cholecystokinin-dependent lipid-induced satiation and activates estrogen receptor-{alpha}-expressing cells in the nucleus tractus solitarius of ovariectomized rats. Endocrinology 148, 56565666.CrossRefGoogle ScholarPubMed
99Hayes, MR & Covasa, M (2005) CCK and 5-HT act synergistically to suppress food intake through simultaneous activation of CCK-1 and 5-HT3 receptors. Peptides 26, 23222330.CrossRefGoogle Scholar
100Hayes, MR, Savastano, DM & Covasa, M (2004) Cholecystokinin-induced satiety is mediated through interdependent cooperation of CCK-A and 5-HT3 receptors. Physiol Behav 82, 663669.CrossRefGoogle Scholar
101Peters, JH, Simasko, SM & Ritter, RC (2006) Modulation of vagal afferent excitation and reduction of food intake by leptin and cholecystokinin. Physiol Behav 89, 477485.CrossRefGoogle ScholarPubMed
102Degen, L, Drewe, J, Piccoli, F, et al. (2007) Effect of CCK-1 receptor blockade on ghrelin and PYY secretion in men. Am J Physiol Regul Integr Comp Physiol 292, R1391R1399.CrossRefGoogle ScholarPubMed
103Brennan, IM, Little, TJ, Feltrin, KL, et al. (2008) Dose-dependent effects of cholecystokinin-8 on antropyloroduodenal motility, gastrointestinal hormones, appetite, and energy intake in healthy men. Am J Physiol Endocrinol Metab 295, E1487E1494.CrossRefGoogle ScholarPubMed
104Lo, CM, Zhang, DM, Pearson, K, et al. (2007) Interaction of apolipoprotein AIV with cholecystokinin on the control of food intake. Am J Physiol Regul Integr Comp Physiol 293, R1490R14R4.CrossRefGoogle ScholarPubMed
105Funakoshi, A, Miyasaka, K, Matsumoto, H, et al. (2000) Gene structure of human cholecystokinin (CCK) type-A receptor: body fat content is related to CCK type-A receptor gene promoter polymorphism. FEBS Lett 466, 264266.CrossRefGoogle Scholar
106de Krom, M, van der Schouw, YT, Hendriks, J, et al. (2007) Common genetic variations in CCK, leptin, and leptin receptor genes are associated with specific human eating patterns. Diabetes 56, 276280.CrossRefGoogle ScholarPubMed
107Zwirska-Korczala, K, Konturek, SJ, Sodowski, M, et al. (2007) Basal and postprandial plasma levels of PYY, ghrelin, cholecystokinin, gastrin and insulin in women with moderate and morbid obesity and metabolic syndrome. J Physiol Pharmacol 58, Suppl. 1, 1335.Google ScholarPubMed
108French, SJ, Murray, B, Rumsey, RD, et al. (1993) Preliminary studies on the gastrointestinal responses to fatty meals in obese people. Int J Obes Relat Metab Disord 17, 295300.Google ScholarPubMed
109Glatzle, J, Raybould, HE, Kueper, MA, et al. (2008) Cholecystokinin-58 is more potent in inhibiting food intake than cholecystokinin-8 in rats. Nutr Neurosci 11, 6974.CrossRefGoogle ScholarPubMed
110Naslund, E, Gryback, P, Hellstrom, PM, et al. (1997) Gastrointestinal hormones and gastric emptying 20 years after jejunoileal bypass for massive obesity. Int J Obes Relat Metab Disord 21, 387392.CrossRefGoogle ScholarPubMed
111Roses, AD (2004) Pharmacogenetics and drug development: the path to safer and more effective drugs. Nat Rev Genet 5, 645656.CrossRefGoogle ScholarPubMed
112Lateef, DM, Washington, MC & Sayegh, AI (2011) The short term satiety peptide cholecystokinin reduces meal size and prolongs intermeal interval. Peptides 32, 12891295.CrossRefGoogle ScholarPubMed
113Matson, CA, Reid, DF, Cannon, TA, et al. (2000) Cholecystokinin and leptin act synergistically to reduce body weight. Am J Physiol Regul Integr Comp Physiol 278, R882R890.CrossRefGoogle ScholarPubMed
114Matson, CA, Reid, DF & Ritter, RC (2002) Daily CCK injection enhances reduction of body weight by chronic intracerebroventricular leptin infusion. Am J Physiol Regul Integr Comp Physiol 282, R1368R1373.CrossRefGoogle ScholarPubMed
115Emond, M, Schwartz, GJ, Ladenheim, EE, et al. (1999) Central leptin modulates behavioral and neural responsivity to CCK. Am J Physiol 276, R1545R1549.Google ScholarPubMed
116Bhavsar, S, Watkins, J & Young, A (1998) Synergy between amylin and cholecystokinin for inhibition of food intake in mice. Physiol Behav 64, 557561.CrossRefGoogle ScholarPubMed
117Matson, CA & Ritter, RC (1999) Long-term CCK-leptin synergy suggests a role for CCK in the regulation of body weight. Am J Physiol Regul Integr Comp Physiol 276, R1038R1045.CrossRefGoogle ScholarPubMed
118Holst, JJ (2010) Glucagon and glucagon-like peptides 1 and 2. Results Probl Cell Differ 50, 121135.Google Scholar
119Dube, PE & Brubaker, PL (2004) Nutrient, neural and endocrine control of glucagon-like peptide secretion. Horm Metab Res 36, 755760.CrossRefGoogle ScholarPubMed
120Holst, JJ, Orskov, C, Nielsen, OV, et al. (1987) Truncated glucagon-like peptide I, an insulin-releasing hormone from the distal gut. FEBS Lett 211, 169174.CrossRefGoogle ScholarPubMed
121Mojsov, S, Weir, GC & Habener, JF (1987) Insulinotropin: glucagon-like peptide I (7–37) co-encoded in the glucagon gene is a potent stimulator of insulin release in the perfused rat pancreas. J Clin Invest 79, 616619.CrossRefGoogle ScholarPubMed
122D'Alessio, D, Lu, W, Sun, W, et al. (2007) Fasting and postprandial concentrations of GLP-1 in intestinal lymph and portal plasma: evidence for selective release of GLP-1 in the lymph system. Am J Physiol Regul Integr Comp Physiol 293, R2163R2169.CrossRefGoogle ScholarPubMed
123Mentlein, R, Gallwitz, B & Schmidt, WE (1993) Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1(7–36)amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur J Biochem 214, 829835.CrossRefGoogle ScholarPubMed
124Deacon, CF, Pridal, L, Klarskov, L, et al. (1996) Glucagon-like peptide 1 undergoes differential tissue-specific metabolism in the anesthetized pig. Am J Physiol 271, E458E464.Google ScholarPubMed
125Imeryuz, N, Yegen, BC, Bozkurt, A, et al. (1997) Glucagon-like peptide-1 inhibits gastric emptying via vagal afferent-mediated central mechanisms. Am J Physiol 273, G920G927.Google ScholarPubMed
126Little, TJ, Pilichiewicz, AN, Russo, A, et al. (2006) Effects of intravenous glucagon-like peptide-1 on gastric emptying and intragastric distribution in healthy subjects: relationships with postprandial glycemic and insulinemic responses. J Clin Endocrinol Metab 91, 19161923.CrossRefGoogle ScholarPubMed
127Shiiya, T, Nakazato, M, Mizuta, M, et al. (2002) Plasma ghrelin levels in lean and obese humans and the effect of glucose on ghrelin secretion. J Clin Endocrinol Metab 87, 240244.CrossRefGoogle ScholarPubMed
128Tolessa, T, Gutniak, M, Holst, JJ, et al. (1998) Glucagon-like peptide-1 retards gastric emptying and small bowel transit in the rat: effect mediated through central or enteric nervous mechanisms. Dig Dis Sci 43, 22842290.CrossRefGoogle ScholarPubMed
129Donahey, JC, van Dijk, G, Woods, SC, et al. (1998) Intraventricular GLP-1 reduces short- but not long-term food intake or body weight in lean and obese rats. Brain Res 779, 7583.CrossRefGoogle ScholarPubMed
130Tang-Christensen, M, Larsen, PJ, Goke, R, et al. (1996) Central administration of GLP-1-(7–36) amide inhibits food and water intake in rats. Am J Physiol 271, R848R856.Google ScholarPubMed
131Turton, MD, O'Shea, D, Gunn, I, et al. (1996) A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379, 6972.CrossRefGoogle ScholarPubMed
132Larsen, PJ & Holst, JJ (2005) Glucagon-related peptide 1 (GLP-1): hormone and neurotransmitter. Regul Pept 128, 97107.CrossRefGoogle ScholarPubMed
133Rodriquez de Fonseca, F, Navarro, M, Alvarez, E, et al. (2000) Peripheral versus central effects of glucagon-like peptide-1 receptor agonists on satiety and body weight loss in Zucker obese rats. Metabolism 49, 709717.CrossRefGoogle ScholarPubMed
134Abbott, CR, Monteiro, M, Small, CJ, et al. (2005) The inhibitory effects of peripheral administration of peptide YY(3–36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Res 1044, 127131.CrossRefGoogle ScholarPubMed
135Williams, DL, Baskin, DG & Schwartz, MW (2009) Evidence that intestinal glucagon-like peptide-1 plays a physiological role in satiety. Endocrinology 150, 16801687.CrossRefGoogle Scholar
136Hayes, MR, Leichner, TM, Zhao, S, et al. (2011) Intracellular signals mediating the food intake-suppressive effects of hindbrain glucagon-like peptide-1 receptor activation. Cell Metab 13, 320330.CrossRefGoogle ScholarPubMed
137Kanoski, SE, Fortin, SM, Arnold, M, et al. (2011) Peripheral and central GLP-1 receptor populations mediate the anorectic effects of peripherally administered GLP-1 receptor agonists, liraglutide and exendin-4. Endocrinology 152, 31033112.CrossRefGoogle ScholarPubMed
138Ruttimann, EB, Arnold, M, Hillebrand, JJ, et al. (2009) Intrameal hepatic portal and intraperitoneal infusions of glucagon-like peptide-1 reduce spontaneous meal size in the rat via different mechanisms. Endocrinology 150, 11741181.CrossRefGoogle ScholarPubMed
139Zhang, J & Ritter, RC (2011) Circulating GLP-1 and CCK-8 reduce food intake by capsaicin-insensitive, non-vagal mechanisms. Am J Physiol Regul Integr Comp Physiol 302, R264R273.CrossRefGoogle Scholar
140Ruttimann, EB, Arnold, M, Geary, N, et al. (2010) GLP-1 antagonism with exendin (9–39) fails to increase spontaneous meal size in rats. Physiol Behav 100, 291296.CrossRefGoogle ScholarPubMed
141Orskov, C, Wettergren, A & Holst, JJ (1996) Secretion of the incretin hormones glucagon-like peptide-1 and gastric inhibitory polypeptide correlates with insulin secretion in normal man throughout the day. Scand J Gastroenterol 31, 665670.CrossRefGoogle ScholarPubMed
142Ma, X, Bruning, J & Ashcroft, FM (2007) Glucagon-like peptide 1 stimulates hypothalamic proopiomelanocortin neurons. J Neurosci 27, 71257129.CrossRefGoogle ScholarPubMed
143Seo, S, Ju, S, Chung, H, et al. (2008) Acute effects of glucagon-like peptide-1 on hypothalamic neuropeptide and AMP activated kinase expression in fasted rats. Endocr J 55, 867874.CrossRefGoogle ScholarPubMed
144Asarian, L (2009) Loss of cholecystokinin and glucagon-like peptide-1-induced satiation in mice lacking serotonin 2C receptors. Am J Physiol Regul Integr Comp Physiol 296, R51R56.CrossRefGoogle ScholarPubMed
145Neary, NM, Small, CJ, Druce, MR, et al. (2005) Peptide YY3–36 and glucagon-like peptide-17–36 inhibit food intake additively. Endocrinology 146, 51205127.CrossRefGoogle ScholarPubMed
146Bojanowska, E & Nowak, A (2007) Interactions between leptin and exendin-4, a glucagon-like peptide-1 agonist, in the regulation of food intake in the rat. J Physiol Pharmacol 58, 349360.Google ScholarPubMed
147Nowak, A & Bojanowska, E (2008) Effects of peripheral or central GLP-1 receptor blockade on leptin-induced suppression of appetite. J Physiol Pharmacol 59, 501510.Google ScholarPubMed
148Naslund, E, Gryback, P, Backman, L, et al. (1998) Distal small bowel hormones: correlation with fasting antroduodenal motility and gastric emptying. Dig Dis Sci 43, 945952.CrossRefGoogle ScholarPubMed
149Rask, E, Olsson, T, Soderberg, S, et al. (2001) Impaired incretin response after a mixed meal is associated with insulin resistance in nondiabetic men. Diabetes Care 24, 16401645.CrossRefGoogle ScholarPubMed
150Williams, DL, Hyvarinen, N, Lilly, N, et al. (2011) Maintenance on a high-fat diet impairs the anorexic response to glucagon-like-peptide-1 receptor activation. Physiol Behav 103, 557564.CrossRefGoogle ScholarPubMed
151Hayes, MR, Kanoski, SE, Alhadeff, AL, et al. (2011) Comparative effects of the long-acting GLP-1 receptor ligands, liraglutide and exendin-4, on food intake and body weight suppression in rats. Obesity (Silver Spring) 19, 13421349.CrossRefGoogle ScholarPubMed
152Baggio, LL, Huang, Q, Brown, TJ, et al. (2004) Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology 127, 546558.CrossRefGoogle ScholarPubMed
153Berthoud, HR, Shin, AC & Zheng, H (2011) Obesity surgery and gut-brain communication. Physiol Behav 105, 106119.CrossRefGoogle ScholarPubMed
154Astrup, A, Rossner, S, Van Gaal, L, et al. (2009) Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet 374, 16061616.CrossRefGoogle ScholarPubMed
155Nayak, UA, Govindan, J, Baskar, V, et al. (2010) Exenatide therapy in insulin-treated type 2 diabetes and obesity. QJM 103, 687694.CrossRefGoogle ScholarPubMed
156Adrian, TE, Ferri, GL, Bacarese-Hamilton, AJ, et al. (1985) Human distribution and release of a putative new gut hormone, peptide YY. Gastroenterology 89, 10701077.CrossRefGoogle ScholarPubMed
157Eberlein, GA, Eysselein, VE, Schaeffer, M, et al. (1989) A new molecular form of PYY: structural characterization of human PYY(3–36) and PYY(1–36). Peptides 10, 797803.CrossRefGoogle Scholar
158Acuna-Goycolea, C & van den Pol, AN (2005) Peptide YY(3–36) inhibits both anorexigenic proopiomelanocortin and orexigenic neuropeptide Y neurons: implications for hypothalamic regulation of energy homeostasis. J Neurosci 25, 1051010519.CrossRefGoogle ScholarPubMed
159Batterham, RL, Cowley, MA, Small, CJ, et al. (2002) Gut hormone PYY(3–36) physiologically inhibits food intake. Nature 418, 650654.CrossRefGoogle ScholarPubMed
160Challis, BG, Pinnock, SB, Coll, AP, et al. (2003) Acute effects of PYY3–36 on food intake and hypothalamic neuropeptide expression in the mouse. Biochem Biophys Res Commun 311, 915919.CrossRefGoogle ScholarPubMed
161Challis, BG, Coll, AP, Yeo, GS, et al. (2004) Mice lacking pro-opiomelanocortin are sensitive to high-fat feeding but respond normally to the acute anorectic effects of peptide-YY(3–36). Proc Natl Acad Sci U S A 101, 46954700.CrossRefGoogle Scholar
162Halatchev, IG, Ellacott, KL, Fan, W, et al. (2004) Peptide YY3–36 inhibits food intake in mice through a melanocortin-4 receptor-independent mechanism. Endocrinology 145, 25852590.CrossRefGoogle ScholarPubMed
163Koda, S, Date, Y, Murakami, N, et al. (2005) The role of the vagal nerve in peripheral PYY3–36-induced feeding reduction in rats. Endocrinology 146, 23692375.CrossRefGoogle ScholarPubMed
164Moran, TH & Dailey, MJ (2011) Intestinal feedback signaling and satiety. Physiol Behav 105, 7781.CrossRefGoogle ScholarPubMed
165Guo, Y, Ma, L, Enriori, PJ, et al. (2006) Physiological evidence for the involvement of peptide YY in the regulation of energy homeostasis in humans. Obesity (Silver Spring) 14, 15621570.CrossRefGoogle ScholarPubMed
166le Roux, CW, Batterham, RL, Aylwin, SJ, et al. (2006) Attenuated peptide YY release in obese subjects is associated with reduced satiety. Endocrinology 147, 38.CrossRefGoogle ScholarPubMed
167Yang, N, Wang, C, Xu, M, et al. (2005) Interaction of dietary composition and PYY gene expression in diet-induced obesity in rats. J Huazhong Univ Sci Technolog Med Sci 25, 243246.Google ScholarPubMed
168Batterham, RL, Cohen, MA, Ellis, SM, et al. (2003) Inhibition of food intake in obese subjects by peptide YY3–36. N Engl J Med 349, 941948.CrossRefGoogle ScholarPubMed
169Chandarana, K, Gelegen, C, Karra, E, et al. (2011) Diet and gastrointestinal bypass-induced weight loss: the roles of ghrelin and peptide YY. Diabetes 60, 810818.CrossRefGoogle ScholarPubMed
170Shin, AC, Zheng, H, Townsend, RL, et al. (2010) Meal-induced hormone responses in a rat model of Roux-en-Y gastric bypass surgery. Endocrinology 151, 15881597.CrossRefGoogle Scholar
171Batterham, RL, Heffron, H, Kapoor, S, et al. (2006) Critical role for peptide YY in protein-mediated satiation and body-weight regulation. Cell Metab 4, 223233.CrossRefGoogle ScholarPubMed
172Gantz, I, Erondu, N, Mallick, M, et al. (2007) Efficacy and safety of intranasal peptide YY3–36 for weight reduction in obese adults. J Clin Endocrinol Metab 92, 17541757.CrossRefGoogle ScholarPubMed
173Reidelberger, RD, Haver, AC, Apenteng, BA, et al. (2011) Effects of exendin-4 alone and with peptide YY(3–36) on food intake and body weight in diet-induced obese rats. Obesity (Silver Spring) 19, 121127.CrossRefGoogle ScholarPubMed
174Field, BCT, Wren, AM, Peters, V, et al. (2010) PYY3–36 and oxyntomodulin can be additive in their effect on food intake in overweight and obese humans. Diabetes 59, 16351639.CrossRefGoogle ScholarPubMed
175Wynne, K & Bloom, SR (2006) The role of oxyntomodulin and peptide tyrosine-tyrosine (PYY) in appetite control. Nat Clin Pract Endocrinol Metab 2, 612620.CrossRefGoogle ScholarPubMed
176Wynne, K, Park, AJ, Small, CJ, et al. (2006) Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial. Int J Obes (Lond) 30, 17291736.CrossRefGoogle ScholarPubMed
177Liu, YL, Ford, HE, Druce, MR, et al. (2010) Subcutaneous oxyntomodulin analogue administration reduces body weight in lean and obese rodents. Int J Obes (Lond) 34, 17151725.CrossRefGoogle ScholarPubMed
178Roth, JD, Roland, BL, Cole, RL, et al. (2008) Leptin responsiveness restored by amylin agonism in diet-induced obesity: evidence from nonclinical and clinical studies. Proc Natl Acad Sci U S A 105, 72577262.CrossRefGoogle ScholarPubMed
179Trevaskis, JL, Coffey, T, Cole, R, et al. (2008) Amylin-mediated restoration of leptin responsiveness in diet-induced obesity: magnitude and mechanisms. Endocrinology 149, 56795687.CrossRefGoogle ScholarPubMed
180Trevaskis, JL, Turek, VF, Griffin, PS, et al. (2010) Multi-hormonal weight loss combinations in diet-induced obese rats: therapeutic potential of cholecystokinin? Physiol Behav 100, 187195.CrossRefGoogle ScholarPubMed
181Covasa, M (2010) Deficits in gastrointestinal responses controlling food intake and body weight. Am J Physiol Regul Integr Comp Physiol 299, R1423R1439.CrossRefGoogle ScholarPubMed
182Swartz, TD, Savastano, DM & Covasa, M (2010) Reduced sensitivity to cholecystokinin in male rats fed a high-fat diet is reversible. J Nutr 140, 16981703.CrossRefGoogle ScholarPubMed
183Covasa, M & Ritter, RC (1998) Rats maintained on high-fat diets exhibit reduced satiety in response to CCK and bombesin. Peptides 19, 14071415.CrossRefGoogle Scholar
184Savastano, DM & Covasa, M (2005) Adaptation to a high-fat diet leads to hyperphagia and diminished sensitivity to cholecystokinin in rats. J Nutr 135, 19531959.CrossRefGoogle ScholarPubMed
185Boyd, KA, O'Donovan, DG, Doran, S, et al. (2003) High-fat diet effects on gut motility, hormone, and appetite responses to duodenal lipid in healthy men. Am J Physiol Gastrointest Liver Physiol 284, G188G196.CrossRefGoogle ScholarPubMed
186Bray, GA, Paeratakul, S & Popkin, BM (2004) Dietary fat and obesity: a review of animal, clinical and epidemiological studies. Physiol Behav 83, 549555.CrossRefGoogle ScholarPubMed
187Wortley, KE, Chang, G-Q, Davydova, Z, et al. (2003) Orexin gene expression is increased during states of hypertriglyceridemia. Am J Physiol Regul Integr Comp Physiol 284, R1454R1465.CrossRefGoogle ScholarPubMed
188Leibowitz, SF, Akabayashi, A & Wang, J (1998) Obesity on a high-fat diet: role of hypothalamic galanin in neurons of the anterior paraventricular nucleus projecting to the median eminence. J Neurosci 18, 27092719.CrossRefGoogle Scholar
189Wang, H, Storlien, LH & Huang, X-F (2002) Effects of dietary fat types on body fatness, leptin, and ARC leptin receptor, NPY, and AgRP mRNA expression. Am J Physiol Endocrinol Metab 282, E1352E1359.CrossRefGoogle ScholarPubMed
190Huang, XF, Xin, X, McLennan, P, et al. (2004) Role of fat amount and type in ameliorating diet-induced obesity: insights at the level of hypothalamic arcuate nucleus leptin receptor, neuropeptide Y and pro-opiomelanocortin mRNA expression. Diabetes Obes Metab 6, 3544.CrossRefGoogle ScholarPubMed
191Ley, RE, Turnbaugh, PJ, Klein, S, et al. (2006) Microbial ecology: human gut microbes associated with obesity. Nature 444, 10221023.CrossRefGoogle ScholarPubMed
192Turnbaugh, PJ, Backhed, F, Fulton, L, et al. (2008) Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 3, 213223.CrossRefGoogle ScholarPubMed
193Turnbaugh, PJ, Ridaura, VK, Faith, JJ, et al. (2009) The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med 1, 6ra14.CrossRefGoogle ScholarPubMed
194Backhed, F, Ding, H, Wang, T, et al. (2004) The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 101, 1571815723.CrossRefGoogle ScholarPubMed
195Backhed, F, Manchester, JK, Semenkovich, CF, et al. (2007) Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A 104, 979984.CrossRefGoogle ScholarPubMed
196Rabot, S, Membrez, M, Bruneau, A, et al. (2010) Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J 24, 49484959.Google ScholarPubMed
197Turnbaugh, PJ, Ley, RE, Mahowald, MA, et al. (2006) An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 10271031.CrossRefGoogle ScholarPubMed
198de La Serre, CB, Ellis, CL, Lee, J, et al. (2010) Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am J Physiol Gastrointest Liver Physiol 299, G440G448.CrossRefGoogle ScholarPubMed
199Hildebrandt, MA, Hoffmann, C, Sherrill-Mix, SA, et al. (2009) High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology 137, 17161724, e1–2.CrossRefGoogle ScholarPubMed
200Mozes, S, Bujnakova, D, Sefcikova, Z, et al. (2008) Developmental changes of gut microflora and enzyme activity in rat pups exposed to fat-rich diet. Obesity (Silver Spring) 16, 26102615.CrossRefGoogle ScholarPubMed
201Musso, G, Gambino, R & Cassader, M (2010) Gut microbiota as a regulator of energy homeostasis and ectopic fat deposition: mechanisms and implications for metabolic disorders. Curr Opin Lipidol 21, 7683.CrossRefGoogle ScholarPubMed
202Samuel, BS, Shaito, A, Motoike, T, et al. (2008) Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci U S A 105, 1676716772.CrossRefGoogle ScholarPubMed
203Dewulf, EM, Cani, PD, Neyrinck, AM, et al. (2011) Inulin-type fructans with prebiotic properties counteract GPR43 overexpression and PPAR-γ-related adipogenesis in the white adipose tissue of high-fat diet-fed mice. J Nutr Biochem 22, 712722.CrossRefGoogle Scholar
204Cani, PD & Delzenne, NM (2009) The role of the gut microbiota in energy metabolism and metabolic disease. Curr Pharm Des 15, 15461558.CrossRefGoogle ScholarPubMed
205Swartz, TD, Duca, FA, de Wouters, T, et al. (2011) Up-regulation of intestinal type 1 taste receptor 3 and sodium glucose luminal transporter-1 expression and increased sucrose intake in mice lacking gut microbiota. Br J Nutr 107, 621630.CrossRefGoogle ScholarPubMed
206Cani, PD, Montoya, ML, Neyrinck, AM, et al. (2004) Potential modulation of plasma ghrelin and glucagon-like peptide-1 by anorexigenic cannabinoid compounds, SR141716A (rimonabant) and oleoylethanolamide. Br J Nutr 92, 757761.CrossRefGoogle ScholarPubMed
207Cani, PD, Possemiers, S, Van de Wiele, T, et al. (2009) Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 58, 10911103.CrossRefGoogle ScholarPubMed
208Delzenne, NM, Cani, PD, Daubioul, C, et al. (2005) Impact of inulin and oligofructose on gastrointestinal peptides. Br J Nutr 93, Suppl. 1, S157S161.CrossRefGoogle ScholarPubMed
209Wikoff, WR, Anfora, AT, Liu, J, et al. (2009) Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A 106, 36983703.CrossRefGoogle ScholarPubMed
210Cani, PD & Delzenne, NM (2009) Interplay between obesity and associated metabolic disorders: new insights into the gut microbiota. Curr Opin Pharmacol 9, 737743.CrossRefGoogle ScholarPubMed
211Muccioli, GG, Naslain, D, Backhed, F, et al. (2010) The endocannabinoid system links gut microbiota to adipogenesis. Mol Syst Biol 6, 392.CrossRefGoogle ScholarPubMed
212Gee, JM & Johnson, IT (2005) Dietary lactitol fermentation increases circulating peptide YY and glucagon-like peptide-1 in rats and humans. Nutrition 21, 10361043.CrossRefGoogle ScholarPubMed
213Everard, A, Lazarevic, V, Derrien, M, et al. (2011) Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 60, 27752786.CrossRefGoogle ScholarPubMed
214Le Poul, E, Loison, C, Struyf, S, et al. (2003) Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem 278, 2548125489.CrossRefGoogle ScholarPubMed
215Uribe, A, Alam, M, Johansson, O, et al. (1994) Microflora modulates endocrine cells in the gastrointestinal mucosa of the rat. Gastroenterology 107, 12591269.CrossRefGoogle ScholarPubMed
216Berthoud, HR, Lenard, NR & Shin, AC (2011) Food reward, hyperphagia, and obesity. Am J Physiol Regul Integr Comp Physiol 300, R1266R1277.CrossRefGoogle ScholarPubMed
217Finlayson, G, King, N & Blundell, JE (2007) Liking vs. wanting food: importance for human appetite control and weight regulation. Neurosci Biobehav Rev 31, 9871002.CrossRefGoogle ScholarPubMed
218Kenny, PJ (2010) Reward mechanisms in obesity: new insights and future directions. Neuron 69, 664679.CrossRefGoogle Scholar
219Stice, E, Spoor, S, Bohon, C, et al. (2008) Relation between obesity and blunted striatal response to food is moderated by TaqIA A1 allele. Science 322, 449452.CrossRefGoogle ScholarPubMed
220Stice, E, Spoor, S, Bohon, C, et al. (2008) Relation of reward from food intake and anticipated food intake to obesity: a functional magnetic resonance imaging study. J Abnorm Psychol 117, 924935.CrossRefGoogle ScholarPubMed
221Geiger, BM, Behr, GG, Frank, LE, et al. (2008) Evidence for defective mesolimbic dopamine exocytosis in obesity-prone rats. FASEB J 22, 27402746.CrossRefGoogle ScholarPubMed
222Johnson, PM & Kenny, PJ (2010) Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. Nat Neurosci 13, 635641.CrossRefGoogle ScholarPubMed
223Stice, E, Yokum, S, Burger, KS, et al. (2011) Youth at risk for obesity show greater activation of striatal and somatosensory regions to food. J Neurosci 31, 43604366.CrossRefGoogle ScholarPubMed
224Stice, E, Yokum, S, Bohon, C, et al. (2010) Reward circuitry responsivity to food predicts future increases in body mass: moderating effects of DRD2 and DRD4. Neuroimage 50, 16181625.CrossRefGoogle ScholarPubMed
225Stice, E, Yokum, S, Blum, K, et al. (2010) Weight gain is associated with reduced striatal response to palatable food. J Neurosci 30, 1310513109.CrossRefGoogle ScholarPubMed
226Krugel, U, Schraft, T, Kittner, H, et al. (2003) Basal and feeding-evoked dopamine release in the rat nucleus accumbens is depressed by leptin. Eur J Pharmacol 482, 185187.CrossRefGoogle ScholarPubMed
227Hommel, JD, Trinko, R, Sears, RM, et al. (2006) Leptin receptor signaling in midbrain dopamine neurons regulates feeding. Neuron 51, 801810.CrossRefGoogle ScholarPubMed
228Fulton, S, Pissios, P, Manchon, RP, et al. (2006) Leptin regulation of the mesoaccumbens dopamine pathway. Neuron 51, 811822.CrossRefGoogle ScholarPubMed
229Pfaffly, J, Michaelides, M, Wang, GJ, et al. (2010) Leptin increases striatal dopamine D2 receptor binding in leptin-deficient obese (ob/ob) mice. Synapse 64, 503510.CrossRefGoogle ScholarPubMed
230Kim, KS, Yoon, YR, Lee, HJ, et al. (2010) Enhanced hypothalamic leptin signaling in mice lacking dopamine D2 receptors. J Biol Chem 285, 89058917.CrossRefGoogle ScholarPubMed
231Naleid, AM, Grace, MK, Cummings, DE, et al. (2005) Ghrelin induces feeding in the mesolimbic reward pathway between the ventral tegmental area and the nucleus accumbens. Peptides 26, 22742279.CrossRefGoogle ScholarPubMed
232Malik, S, McGlone, F, Bedrossian, D, et al. (2008) Ghrelin modulates brain activity in areas that control appetitive behavior. Cell Metab 7, 400409.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Food intake is controlled by complex neural, hormonal and metabolic signals. In the oral cavity, nutrient and non-nutrient tastants activate several taste proteins, such as type 1 taste receptor (T1R) 2/3 and fatty acid translocase CD-36 (CD36). This sensory input is processed by the hindbrain, further stimulating ingestion. The presence of nutrients triggers the release of gastrointestinal (GI) peptides from the stomach and intestine that either act locally on specific receptors distributed along vagal afferents which synapse with the first-order neurons in the hindbrain, or enter the bloodstream (along with signals from the adipose tissue such as leptin) and activate receptors located on hypothalamic neurons. The release of these GI signals, especially in the distal intestine, is also affected by the microflora within the gut. Gut microbiota dispersed primarily throughout the distal intestine (represented by oval and round shapes in the intestinal lumen) influences peptide secretion possibly through nutrient receptors, such as T1R2/R3 and G-protein-coupled receptor (GPCR) or bacterial by-products such as SCFA, that serve as ligands for GPCR. These signals, along with other sensory inputs, are integrated with circuits from higher-order brain areas which in turn alter food intake, energy expenditure and body adiposity. With the exception of ghrelin that has a stimulatory effect on food intake, the final actions of these peptides are inhibition of food intake. LCFA, long-chain fatty acid; GLUC, glucose; CCK, cholecystokinin; GLP-1, glucagon-like peptide-1; PYY, peptide YY; CCK-1R, cholecystokinin-1 receptor; GHSR, growth hormone secretagogue receptor; LepR, leptin receptor; GLP-1R, glucagon-like peptide-1 receptor; NPY2R, neuropeptide Y 2-receptor.