14
Central Control of Body Weight and Appetite Stephen C. Woods and David A. D’Alessio Departments of Psychiatry (S.C.W.) and Medicine (D.A.D.), University of Cincinnati, Cincinnati, Ohio 45237 Context: Energy balance is critical for survival and health, and control of food intake is an integral part of this process. This report reviews hormonal signals that influence food intake and their clinical applications. Evidence Acquisition: A relatively novel insight is that satiation signals that control meal size and adiposity signals that signify the amount of body fat are distinct and interact in the hypothalamus and elsewhere to control energy homeostasis. This review focuses upon recent literature address- ing the integration of satiation and adiposity signals and therapeutic implications for treatment of obesity. Evidence Synthesis: During meals, signals such as cholecystokinin arise primarily from the GI tract to cause satiation and meal termination; signals secreted in proportion to body fat such as insulin and leptin interact with satiation signals and provide effective regulation by dictating meal size to amounts that are appropriate for body fatness, or stored energy. Although satiation and adiposity signals are myriad and redundant and reduce food intake, there are few known orexigenic signals; thus, initiation of meals is not subject to the degree of homeostatic regulation that cessation of eating is. There are now drugs available that act through receptors for satiation factors and which cause weight loss, demonstrating that this system is amenable to manipulation for therapeutic goals. Conclusions: Although progress on effective medical therapies for obesity has been relatively slow in coming, advances in understanding the central regulation of food intake may ultimately be turned into useful treatment options. (J Clin Endocrinol Metab 93: S37–S50, 2008) T he past decade has seen an increasing recognition that a com- plex interplay exists between the central nervous system (CNS) and the activity of numerous organs involved in energy homeostasis. This requires the transmission of key information to the brain, and control of food intake is one component of energy balance where endocrine signaling from the periphery to the CNS has a particularly important role. Considered broadly, energy homeostasis consists of the interrelated processes inte- grated by the brain to maintain energy stores at appropriate levels for given environmental conditions. Energy homeostasis thus includes the regulation of nutrient levels in key storage or- gans (e.g. fat in adipose tissue and glycogen in the liver and elsewhere) as well as in the blood (e.g. blood glucose). To ac- complish this, the brain receives continuous information about energy stores and fluxes in critical organs, about food that is being eaten and absorbed, and about basal and situational en- ergy needs by tissues. The brain in turn controls tissues that have important roles in energy homeostasis, like the liver and mus- culoskeletal system, as well as the secretion of key metabolically active hormones, primarily through the autonomic nervous sys- tem. The brain is thus able to respond to ongoing as well as unanticipated demands via well-coordinated responses to pre- vent shortfalls in energy stores while maintaining biochemical homeostasis. This review focuses on hormonal and related sig- nals that inform the brain of energy levels, thereby influencing energy intake and ultimately body weight. As a general rule, signals arising in the periphery that influ- ence food intake and energy expenditure can be partitioned into two broad categories (Fig. 1) (1–3). One comprises the signals generated during meals that cause satiation (i.e. feelings of full- 0021-972X/08/$15.00/0 Printed in U.S.A. Copyright © 2008 by The Endocrine Society doi: 10.1210/jc.2008-1630 Received July 28, 2008. Accepted September 8, 2008. Abbreviations: AgRP, Agouti-related peptide; apo A-IV, Apolipoprotein A-IV; ARC, arcuate nucleus; CCK, cholecystokinin; DPP-IV, dipeptidyl peptidase IV; GI, gastrointestinal; GLP-1, glucagon-like peptide-1; GLP-1r, GLP-1 receptor; GRP, gastrin-releasing peptide; LHA, lateral hypothalamic area; MC3R, melanocortin 3 receptor; MC4R, melanocortin 4 recep- tor; MSH, -melanocyte-stimulating hormone; NMB, neuromedin B; NPY, neuropep- tide-Y; POMC, proopiomelanocortin; PVN, paraventricular nuclei; PYY, peptide tyrosine-tyrosine. S U P P L E M E N T Review J Clin Endocrinol Metab, November 2008, 93(11):S37–S50 jcem.endojournals.org S37

s u p p l e m e n t

Embed Size (px)

Citation preview

Page 1: s u p p l e m e n t

Central Control of Body Weight and Appetite

Stephen C. Woods and David A. D’Alessio

Departments of Psychiatry (S.C.W.) and Medicine (D.A.D.), University of Cincinnati, Cincinnati, Ohio 45237

Context: Energy balance is critical for survival and health, and control of food intake is an integralpart of this process. This report reviews hormonal signals that influence food intake and theirclinical applications.

Evidence Acquisition: A relatively novel insight is that satiation signals that control meal size andadiposity signals that signify the amount of body fat are distinct and interact in the hypothalamusand elsewhere to control energy homeostasis. This review focuses upon recent literature address-ing the integration of satiation and adiposity signals and therapeutic implications for treatmentof obesity.

Evidence Synthesis: During meals, signals such as cholecystokinin arise primarily from the GI tractto cause satiation and meal termination; signals secreted in proportion to body fat such as insulinand leptin interact with satiation signals and provide effective regulation by dictating meal size toamounts that are appropriate for body fatness, or stored energy. Although satiation and adipositysignals are myriad and redundant and reduce food intake, there are few known orexigenic signals;thus, initiation of meals is not subject to the degree of homeostatic regulation that cessation of eatingis. There are now drugs available that act through receptors for satiation factors and which causeweight loss, demonstrating that this system is amenable to manipulation for therapeutic goals.

Conclusions: Although progress on effective medical therapies for obesity has been relatively slowin coming, advances in understanding the central regulation of food intake may ultimately beturned into useful treatment options. (J Clin Endocrinol Metab 93: S37–S50, 2008)

The past decade has seen an increasing recognition that a com-plex interplay exists between the central nervous system

(CNS) and the activity of numerous organs involved in energyhomeostasis. This requires the transmission of key informationto the brain, and control of food intake is one component ofenergy balance where endocrine signaling from the periphery tothe CNS has a particularly important role. Considered broadly,energy homeostasis consists of the interrelated processes inte-grated by the brain to maintain energy stores at appropriatelevels for given environmental conditions. Energy homeostasisthus includes the regulation of nutrient levels in key storage or-gans (e.g. fat in adipose tissue and glycogen in the liver andelsewhere) as well as in the blood (e.g. blood glucose). To ac-complish this, the brain receives continuous information aboutenergy stores and fluxes in critical organs, about food that is

being eaten and absorbed, and about basal and situational en-ergy needs by tissues. The brain in turn controls tissues that haveimportant roles in energy homeostasis, like the liver and mus-culoskeletal system, as well as the secretion of key metabolicallyactive hormones, primarily through the autonomic nervous sys-tem. The brain is thus able to respond to ongoing as well asunanticipated demands via well-coordinated responses to pre-vent shortfalls in energy stores while maintaining biochemicalhomeostasis. This review focuses on hormonal and related sig-nals that inform the brain of energy levels, thereby influencingenergy intake and ultimately body weight.

As a general rule, signals arising in the periphery that influ-ence food intake and energy expenditure can be partitioned intotwo broad categories (Fig. 1) (1–3). One comprises the signalsgenerated during meals that cause satiation (i.e. feelings of full-

0021-972X/08/$15.00/0

Printed in U.S.A.

Copyright © 2008 by The Endocrine Society

doi: 10.1210/jc.2008-1630 Received July 28, 2008. Accepted September 8, 2008.

Abbreviations: AgRP, Agouti-related peptide; apo A-IV, Apolipoprotein A-IV; ARC, arcuatenucleus; CCK, cholecystokinin; DPP-IV, dipeptidyl peptidase IV; GI, gastrointestinal; GLP-1,glucagon-like peptide-1; GLP-1r, GLP-1 receptor; GRP, gastrin-releasing peptide; LHA,lateral hypothalamic area; MC3R, melanocortin 3 receptor; MC4R, melanocortin 4 recep-tor; �MSH, �-melanocyte-stimulating hormone; NMB, neuromedin B; NPY, neuropep-tide-Y; POMC, proopiomelanocortin; PVN, paraventricular nuclei; PYY, peptidetyrosine-tyrosine.

S U P P L E M E N T

R e v i e w

J Clin Endocrinol Metab, November 2008, 93(11):S37–S50 jcem.endojournals.org S37

Page 2: s u p p l e m e n t

ness that contribute to the decision to stop eating) and/or satiety(i.e. prolongation of the interval until hunger or a drive to eatreappears). The prototypical satiation signal is the duodenal pep-tide cholecystokinin (CCK), which is secreted in response to di-etary lipid or protein and which activates receptors on local sen-sory nerves in the duodenum, sending a message to the brain viathe vagus nerve that contributes to satiation. The second cate-gory includes hormones such as insulin and leptin that are se-creted in proportion to the amount of fat in the body. These“adiposity” hormones enter the brain by transport through theblood-brain barrier and interact with specific neuronal receptorsprimarily in the hypothalamus to affect energy balance. Satiationand adiposity signals interact with other factors in the hypothal-amus and elsewhere in the brain to control appetite and bodyweight, and they are the topic of this review.

Body weight (adiposity) is a homeostatically regulated vari-able, and its long-term maintenance can only occur via a closelinkage of energy intake to energy expenditure. This means thatover long intervals, the amount of food consumed must provideenergy equivalent to the amount of energy expended. Humansand most mammals acquire energy in discrete episodes or meals.For many modern-day humans, the impetus to begin a meal israrely if ever based on a biological deficit or need such as insuf-ficient glucose. Rather, “appetite” and “hunger” occur, andmeals are initiated based on habit, time of day, specific socialsituations, convenience, or stress, factors that are not linked toenergy needs and so are nonhomeostatic (4–6). Arguably, this

has been the case throughout human evolution as well, with theinitiation of feeding governed by nonhomeostatic factors such asfood availability or having a safe haven to eat. Thus, the homeo-static influence over food intake is often left to the control overhow many calories are consumed once a meal begins; i.e. on mealsize. Consistent with this, many of the secretions of the gastro-intestinal (GI) tract during a meal, such as CCK, are proportionalto the number of calories consumed, and some of these secretionsfunction as satiation signals to the CNS to help limit meal size (seereviews in Refs. 7–9).

In contrast to satiation signals that are phasically secretedduring meals, adiposity signals are more tonically active, pro-viding an ongoing message to the brain proportional to totalbody fat. Insulin is tonically secreted in basal amounts, withphasic increments occurring during meals, and both componentsof total insulin secretion (i.e. basal and meal-stimulated) are di-rectly proportional to body fat (10). Leptin is secreted in directproportion to body adiposity, following a diurnal pattern withless direct connection to meals than insulin (11). As an individualchanges body weight through caloric restriction or overeating,the amounts of insulin and leptin secreted into the blood changein parallel, and this in turn is reflected as an altered signal of bodyfatness, tantamount to body energy stores, reaching the brain(1–3). These adiposity signals interact with anabolic and cata-bolic neural circuits, causing a change in sensitivity of the brainto satiation signals. For example, during food deprivation ordieting, reduced brain insulin/leptin signaling renders neural cir-

FIG. 1. Model summarizing different levels of control over energy homeostasis. During meals, signals such as CCK, GLP-1, and distension of the stomach that arisefrom the gut (stomach and intestine) trigger nerve impulses in sensory nerves traveling to the hindbrain. These satiation signals synapse with neurons in the nucleus ofthe solitary tract (NTS) where they influence meal size. Ghrelin from the stomach both acts on the vagus nerve and stimulates neurons in the ARC directly. Signalsrelated to body fat content such as leptin and insulin, collectively called adiposity signals, circulate in the blood to the brain. They pass through the blood-brain barrierin the region of the ARC and interact with neurons that synthesize POMC or NPY and AgRP. ARC neurons in turn project to other hypothalamic areas including thePVN and the LHA. The net output of the PVN is catabolic and enhances the potency of satiation signals in the hindbrain. The net output of the LHA, on the other hand,is anabolic, suppressing the activity of the satiation signals. In this way body fat content tends to remain relatively constant over long intervals by means of changes ofmeal size.

S38 Woods and D’Alessio Central Control of Body Weight and Appetite J Clin Endocrinol Metab, November 2008, 93(11):S37–S50

Page 3: s u p p l e m e n t

cuits controlling meal size less sensitive to satiation signals suchas CCK. As a consequence, the homeostatic setting is geared forthe intake of larger meals because more food must be consumedbefore a sufficient satiation signal is generated to stop eating.This situation of extra-large meals persists until body weight(and hence the insulin/leptin signal) returns to normal. Con-versely, after excessive weight gain, the increased insulin/leptinbrain signal results in increased sensitivity to satiation signals,and smaller meals are consumed until the excess weight is lost. Amajor dilemma facing clinicians and others involved in publichealth is the slippage in the latter limb of this homeostatic systemthat permits excessive energy storage, obesity, and the associateddiseases of nutritional excess; e.g. diabetes, cardiovascular dis-ease, and some cancers.

Satiation Signals

By definition, satiation factors, when administered to humans oranimals at the start of a meal, result in a smaller-than-normalmeal being consumed. Exogenously administered satiation fac-tors, or endogenous secretion of these compounds, activates spe-cific receptors that cause premature cessation of eating. There areseveral excellent reviews of satiation signals such that only thesalient points need to be reviewed here (7–9, 12–14). It is gen-erally accepted that for an endogenous compound to be consid-ered a satiation signal, it is secreted in response to food ingestion,acts within the time frame of a single meal, reduces meal sizewithout creating malaise or incapacitation, and is effective atphysiological doses, and removing or antagonizing its endoge-nous activity increases meal size (7, 15).

The best-established satiation signals are secreted from spe-cialized enteroendocrine cells in the wall of the GI tract in re-sponse to the digestion and absorption of meals. In the classicmodel of satiation, local sensory nerves express receptors forthese gut peptides as they are secreted such that the brain isimmediately informed about the nutritional content of the mealby monitoring the level of hormones secreted to cope with it. Asa complex meal is consumed, the mix of macronutrients (carbo-hydrates, fats, and proteins) stimulates a proportional blend ofsatiation peptides, and an overall message indicating meal con-tent is integrated in the hindbrain where it activates appropriateresponses, including ultimately cessation of the meal. In humans,this is associated with a sensation of fullness. Although not allintestinal hormones double as satiation signals, they all presum-ably contribute to the assimilation of nutrients; i.e. by stimulat-ing the secretion of appropriate enzymes, water, and other com-pounds into the lumen of the gut and regulating GI motility.Table 1 provides a list of GI and other meal-related hormones/peptides that are generally considered to be satiation signals.

CholecystokininCCK was the compound first identified to fit the criteria for

a satiation factor, so that much is known about its actions. Con-sequently, CCK has become the model for the larger class ofsatiation factors. When food containing fat or protein is con-sumed and enters the duodenum, CCK is secreted from I cells.

CCK enters the blood and has hormonal influences on gut mo-tility, contraction of the gallbladder, pancreatic enzyme secre-tion, gastric emptying, and gastric acid secretion (16–19). How-ever, CCK also diffuses locally to provide a paracrine stimulus toCCK-1 receptors on nearby branches of vagal sensory nerves(20–23). Through this mechanism a message, generally that in-gested fat/protein is being processed and will soon be absorbed,is conveyed to the hindbrain and relayed to the hypothalamuswhere it is integrated into the composite information on energyhomeostasis (20, 24–28) (Fig. 2).

The decrease of meal size elicited by exogenous CCK is dose-dependent, with higher doses causing greater reductions of mealsize. However, CCK does not prevent meals from occurring;rather, it decreases the size of the meal once it has begun, reducinghunger and increasing fullness without concomitant sensationsof illness or malaise. When a CCK-1 receptor antagonist is ad-ministered before the presentation of food to animals or humans,larger-than-normal meals are consumed (29–31), providingcompelling evidence that endogenous CCK normally acts to sup-

TABLE 1. GI hormones that affect satiation

Peptide Effect on food intake

CCK DecreaseGLP-1 DecreasePYY DecreaseApo A-IV DecreaseEnterostatin DecreaseBombesin/GRP/NMB DecreaseOxyntomodulin DecreaseAmylin DecreaseGhrelin Increase

FIG. 2. Satiation signals arising in the GI system converge on the dorsalhindbrain (D) where they are integrated with taste and other inputs. The dorsalhindbrain makes direct connections with the ventral hindbrain (V) where neuralcircuits direct the autonomic nervous system to influence blood glucose andwhere the motor control over eating behavior is located. The dorsal hindbrainalso conveys information on satiation and other factors anteriorly to thehypothalamus and other brain areas. These areas in turn integrate satiation andadiposity signals as well as available nutrients with experience, the social situationand stressors, and with time of day and other factors. The integrated informationis then conveyed posteriorly back to the ventral hindbrain as well as to thepituitary to influence all aspects of energy homeostasis. Animals lacking neuralconnections between the hindbrain and the hypothalamus reduce the intake ofindividual bouts of eating when the stomach is distended or they areadministered CCK. However, those animals cannot regulate their body weightand are not sensitive to past experience, time of day, or social factors (243).

J Clin Endocrinol Metab, November 2008, 93(11):S37–S50 jcem.endojournals.org S39

Page 4: s u p p l e m e n t

press intake during meals. However, the effect of exogenousCCK is short-lived; the meal-reducing signal does not carry overto a second meal. Moreover, the effect of repeated or chronicadministration of CCK (32, 33) has no effect on body weightbecause the short-term regulation of meal size is overridden byoverall energy balance. That is, in the context of lower adiposity,leptin and insulin signals to the brain are reduced and satiationfactors like CCK have a reduced effect to restrict meal size.

There is strong experimental evidence supporting a role forCCK as a satiation factor in humans. CCK levels increase aftermeals, and infusion of an exogenous CCK-1 receptor agonist,CCK-33, to postprandial levels suppresses food intake (34, 35).Furthermore, infusion of a CCK-1 receptor antagonist to healthyhumans causes an increase in caloric intake, strongly implicatingendogenous CCK as a brake on meal size (29, 36). The effects ofCCK-1 agonism and antagonism are similar in lean and obesehumans and are associated with appropriate changes in hungerand fullness. Interestingly, blockade of CCK-1 receptors affectsthe postprandial responses of other GI hormones, attenuatingthe usual rise in peptide YY (PYY) and abolishing the suppres-sion of ghrelin (37). This latter finding suggests that beyonddirectly inhibiting food intake, CCK acts as a proximal mediatorof the broader satiation process. Two minor single nucleotidepolymorphisms in the CCK-1 receptor gene promoter have beenassociated with increased body fatness, but the mechanism ofthis association has not been explained (38).

Glucagon-like peptide-1Glucagon-like peptide-1 (GLP-1) is derived from progluca-

gon in intestinal L cells that are most prevalent in the ileum andcolon (39). GLP-1 secretion is elicited by nutrients, but the mech-anism whereby the distal L cells are stimulated early within mealsmay require neurohumoral signals initiated in the proximal re-gions of the small intestine (40). GLP-1 has a broad range ofactions on glucose metabolism (41), most prominently stimula-tion of insulin secretion, but also inhibition of glucagon release.Because GLP-1 inhibits GI motility and secretions, it has beenimplicated as a major component of the “ileal brake,” an inhib-itory feedback mechanism that regulates transit of nutrientsthrough the course of the GI tract (42, 43). It has generally beenassumed that GLP-1 mediates these various actions through anendocrine mechanism, by binding directly to key target tissueslike pancreatic islet cells. However, this mechanism has recentlybeen called into question, in large part because GLP-1 is rapidlymetabolized in the circulation by the protease dipeptidyl pepti-dase IV (DPP-IV) (44). Indeed, the half-life of GLP-1 in humanplasma is only 1–2 min, and the product of DPP-IV action, atruncated GLP-1, is inactive with regard to glucose metabolism(45). Because the GLP-1 receptor (GLP-1r) is expressed by pe-ripheral and CNS neurons as well as by cells in the pancreaticislets and the GI tract, recent attention has focused on neuralmechanisms of GLP-1 action (46–48).

GLP-1 administration reduces food intake in animals andhumans (49–54), and these anorectic actions are thought to bemediated through both peripheral and central mechanisms. Apopulation of neurons that synthesize GLP-1 is located in thebrain stem and projects to hypothalamic and brain stem areas

important in the control of energy homeostasis (55, 56). Cen-trally administered GLP-1 reduces food intake through at leasttwo mechanisms. GLP-1r in the hypothalamus appear to reduceintake by acting on caloric homeostatic circuits (57–60),whereas GLP-1r in the amygdala reduce food intake by elicitingsymptoms of stress or malaise (61, 62).

Systemically administered GLP-1 elicits satiation in healthy(53), obese (63), and diabetic (64, 65) humans. Because the half-life of active GLP-1 is less than 2 min, any direct effects are likelytransient, and the reduction of food intake may result from in-hibitory effects of GLP-1 on GI transit and reduced gastric emp-tying (66). However, peripherally administered GLP-1 doescross the blood-brain barrier (67), perhaps enabling circulatingGLP-1 to interact with the brain GLP-1r. Because it both reducesfood intake and stimulates insulin secretion, the GLP-1 systemhas been adapted to the treatment of type 2 diabetes (68, 69).DPP-IV-resistant, long-acting GLP-1r agonists are effective atreducing blood glucose in persons with type 2 diabetes and alsocause weight loss (70). In addition, inhibitors of DPP-IV, whichelevate endogenous GLP-1 levels, are also effective at improvingglycemic control in diabetic patients (71, 72).

The effect of chronic GLP-1r agonists to cause weight loss isnot consistent with the general principle that satiation peptidesare subservient to long-term regulators of energy balance, suchas leptin (3). One potential explanation is that because GLP-1can activate nonhomeostatic pathways that suppress food intakeand otherwise reduce body weight, the effects of chronic admin-istration of GLP-1r agonists work around the homeostatic sys-tems controlling body weight. Indeed, nausea, a hallmark featureof nonhomeostatic anorexia, is very common with GLP-1r ag-onist treatment (73), although this response wanes with contin-ued treatment and is not present in all persons who lose weight.Another possible explanation for why patients treated with in-dictable GLP-1r agonists lose weight is that repeated pharma-cological doses of a satiation signal can overcome homeostaticrestraints. Consistent with this hypothesis is the failure ofDPP-IV inhibitor treatment, which has a smaller effect to raisecirculating concentrations of GLP-1r agonist activity than doinjectable GLP-1 mimetics to cause weight loss. Although it is notyet clear how GLP-1r agonists cause weight loss, the fact thatthey do challenges the current model of the interaction of sati-ation factors with overall energy homeostasis.

Glicentin, GLP-2, oxyntomodulin, and glucagonOther peptides derived from the processing of proglucagon

include glicentin, GLP-2, and oxyntomodulin, as well as gluca-gon itself (39). Glicentin inhibits gastric acid secretion (74), butat least in rats, does not affect food intake (75). In contrast,oxyntomodulin, a C-terminally extended congener of glucagon,does reduce food intake in animals when given centrally or pe-ripherally (75, 76). Although a unique receptor for oxyntomodu-lin has yet to be identified, oxyntomodulin may exert its ano-rectic effect through the GLP-1r because subthreshold doses ofthe GLP-1r antagonist, exendin (9–39), block both GLP-1 andoxyntomodulin-induced reductions in food intake (75). Oxyn-tomodulin is thought to cross the blood-brain barrier and stim-ulate neurons in the arcuate nucleus (ARC) that express GLP-1r

S40 Woods and D’Alessio Central Control of Body Weight and Appetite J Clin Endocrinol Metab, November 2008, 93(11):S37–S50

Page 5: s u p p l e m e n t

and control energy homeostasis (77, 78). Long-term treatmentwith oxyntomodulin causes a persistent decrease in food intakeand attenuated weight gain in rats (79). Interestingly, weight lossin animals given oxyntomodulin chronically is greater than whatwould be anticipated from reduced caloric intake (78), suggest-ing additional effects on energy expenditure.

Short-term treatment with iv oxyntomodulin decreases hun-ger, reduces consumption of an ad libitum meal, and has ananorectic action that persists for 12 h after cessation of treatmentin lean humans (80). Moreover, in obese subjects randomized toinjections of oxyntomodulin or placebo before each meal for 4wk, there was a significant loss of weight associated with activetreatment. Oxyntomodulin caused an approximately 0.5 kg/wkreduction in body weight and was generally well tolerated. Thesefindings suggest that at least two proglucagon products may havea role in the regulation of food intake. Distinguishing betweenthe effects of GLP-1 and oxyntomodulin, in particular the rela-tive in vivo actions on the GLP-1r, is an important next step inapplying these compounds to clinical medicine.

GLP-2 acts through a specific GLP-2 receptor to stimulateintestinal mucosal growth and has become the focus of researchon short-bowel syndrome (81, 82). GLP-2 and GLP-1 havenearly 50% sequence homology and are secreted in parallel fromperfused ileal preparations (83). Intracranial administration ofGLP-2 reduces food intake in rats, and the effect can be blockedwith a specific GLP-1r antagonist (84). More recent studies inhumans found no effect of iv GLP-2 on food intake (85).

Glucagon is the most widely studied hormone cleaved frompreproglucagon (39). It is secreted from both pancreatic A cellsand probably also in small amounts from the distal intestine. Thebest known action of glucagon is to increase hepatic glucoseproduction by stimulating glycogenolysis and gluconeogenesis.Glucagon also reduces meal size when administered systemically(86, 87), but not centrally (88), the signal being detected in theliver and relayed to the brain (89). A role for glucagon in thenormal control of meal size was demonstrated by the observationthat blocking endogenous glucagon action in rats increases foodintake (90, 91). There is little convincing evidence that glucagonplays a role in food intake in healthy humans.

Peptide tyrosine-tyrosine (PYY)PYY is a member of a family of homologous peptides that also

includes pancreatic polypeptide and neuropeptide-Y (NPY).Like proglucagon-derived peptides, PYY is synthesized and se-creted by L cells in the distal ileum and colon (92). PYY is secretedas PYY (1–36) and is metabolized to PYY (3–36) by DPP-IV (93,94). Receptors that mediate the effects of PYY, including reduc-tion of food intake, belong to the NPY receptor family and in-clude Y1, Y2, Y4, and Y5 (95). However, PYY (3–36) is a highlyselective agonist activity for the Y2 receptor, and it also reducesfood intake in humans and animals (96, 97). Like GLP-1, PYYhas been implicated in GI motility and is considered a majorcomponent of the ileal brake (98, 99). Secretion of PYY is stim-ulated by food intake (100) and also by the presence of nutrientswithin the ileum itself (101); lipid seems to be a particularlyeffective stimulus. Similar to the case with GLP-1, it is not clearwhether PYY release requires direct nutrient contact with L cells,

or if neurohumoral signals originating from the more proximalGI tract mediate the response. There is evidence that PYY influ-ences food intake through its interaction with Y2 receptors in theARC because it freely crosses the blood-brain barrier (102) andbecause systemic PYY (3–36) is ineffective in reducing food in-take in the Y2-deficient mouse (103).

Studies in humans have demonstrated that PYY signaling re-duces food intake and that abnormalities in this system arepresent in obese subjects. PYY secretion is proportional to thecaloric content of meals, with larger meals eliciting a significantlylarger response (104, 105). Fasting and postprandial levels ofPYY are lower in obese adults compared with lean controls (105,106). The attenuated rise in PYY after eating has also been ob-served in obese adolescents (107). Infusion of PYY (3–36) intolean and obese humans reduced food consumption measuredduring test meals (104–106), although there remains some ques-tion as to whether this effect of PYY (3–36) is pharmacologicalor occurs at circulating levels seen after eating. Obese subjects didnot differ in their sensitivity to PYY, and there was no differencein the relative PYY (1–36) and PYY (3–36) levels after exogenousadministration. Taken together, these findings suggest that ab-normal PYY production, rather than anorectic action of metab-olism, could contribute to obesity.

Interestingly, there is evidence that genetic variations in thePYY and Y2 sequences are associated with body weight. A com-mon gene polymorphism in the Y2 receptor has been associatedwith a reduced likelihood of obesity in a cohort study of men witha broad range of BMI (108). Additionally, in a survey of lean andvery obese men, a polymorphism in the PYY allele was found tosegregate with increased body weight; this genetic variant of PYYwas also found to have reduced binding to its receptor and anattenuated anorectic effect in mice (109). In total, the data col-lected in human studies support a role for PYY in the regulationof food intake and build the strongest case for any of the satiationfactors in the pathogenesis of obesity.

Apolipoprotein A-IVApolipoprotein A-IV (apo A-IV) is synthesized by intestinal

mucosal cells during the packaging of digested lipids into chy-lomicrons that subsequently enter the blood via the lymphaticsystem (110). Apo A-IV is also synthesized in the ARC (111).Systemic or central administration of apo A-IV reduces foodintake and body weight of rats (112), and administration of apoA-IV antibodies increases food intake (113). Apo A-IV appearsto work by interacting with the CCK signal (114). Because bothintestinal and hypothalamic apo A-IV are regulated by absorp-tion of lipid but not carbohydrate (115), this peptide may be animportant link between short- and long-term regulators of bodyfat (see review by Tso and Liu in Ref. 116).

EnterostatinA second digestion-related peptide, enterostatin, is also

closely tied to intestinal processing of lipid. The exocrine pan-creas secretes lipase and colipase to aid in the digestion of fat, andenterostatin, a pentapeptide, is cleaved from colipase in the in-testinal lumen and enters the circulation. Administration of ex-ogenous enterostatin either systemically (117, 118) or directly

J Clin Endocrinol Metab, November 2008, 93(11):S37–S50 jcem.endojournals.org S41

Page 6: s u p p l e m e n t

into the brain (119) reduces food intake, and, when rats are givena choice of foods, the reduction is specific for fats; that is, en-terostatin does not decrease the intake of carbohydrate or pro-tein (120). Therefore, two peptides that are secreted from the gutduring the digestion and absorption of lipids, apo A-IV and en-terostatin, act as signals that decrease food intake, and at leastone of them selectively reduces the intake of fat. Macronutrientspecificity has not been assessed with apo A-IV. There are no datafrom human studies to confirm the findings with apo A-IV andenterostatin on food intake.

Bombesin-family peptidesMembers of the bombesin family of peptides including bomb-

esin itself (an amphibian peptide) and its mammalian analogs,gastrin-releasing peptide (GRP) and neuromedin B (NMB), re-duce food intake when administered systemically in humans andanimals or into the CNS of animals (121–123). Consistent withthe possibility that these peptides act endogenously to reducefood intake, mice deficient for the GRP receptor eat significantlylarger meals and develop late-onset obesity (124). Whereas mostsatiation factors act by reducing the size of an ongoing meal (4),bombesin peptides are an interesting exception in that when theyare administered between meals, they increase the amount oftime until the subsequent meal begins; i.e. they increase satiety aswell as satiation (125, 126).

Both bombesin and GRP reduce food intake when infusedinto human subjects (127, 128). Antagonism of endogenousbombesin receptors has demonstrable effects on gastric, intesti-nal, and gallbladder function but has not been studied with re-gard to food intake (129, 130). Therefore the case for GRP as aphysiological mediator of satiation in humans is not as strong asfor CCK.

AmylinAmylin (also called islet amyloid polypeptide) is a peptide

hormone secreted by pancreatic B cells in tandem with insulinsecretion, which inhibits gastric emptying and gastric acid se-cretion, lowers glucagon concentrations, and reduces food in-take (131). Amylin causes a dose-dependent reduction of mealsize when administered systemically or directly into the brain(132–136), and antagonism of amylin action in the CNS causesincreased food intake and body weight (137). Consistent withthis, targeted deletion of the amylin gene causes increased bodyweight in mice.

Amylin signals through the calcitonin receptor when it hasbeen modified by receptor activity modifying proteins (138,139), and in contrast to many satiation peptides that reduce foodintake by stimulating the visceral afferent nerves, amylin seemsto act as a hormone, directly stimulating neurons in the areapostrema in the hindbrain (133, 140). In fact, the anorectic ac-tion of amylin shares features with both satiation signals (phasic,meal-induced secretion) and adiposity signals (chronic interrup-tion of action in rodents causes increased body fatness). Amylinseems to interact with both types of regulation in that the abilityof amylin to reduce meal size is augmented when brain insulinaction is elevated (141) and the effects of CCK and bombesin aremuted in the absence of amylin signaling (142).

Amylin has been developed as a therapeutic, with the syn-thetic analog pramlintide now available for the treatment of type1 and type 2 diabetes and clinical trials under way to determinethe efficacy in treating obesity. In diabetic patients treated withpramlintide for 1 yr, weight loss averaged approximately 2 kgrelative to placebo-treated controls (143, 144). Similar to treat-ment with the GLP-1r agonist exendin-4, pramlintide presentssupraphysiological levels of amylin-like activity to patients, andthe primary side effect is nausea. Nonetheless, the clinical expe-rience with pramlintide supports the idea that chronic activity ofa satiating compound can cause weight loss.

GhrelinGhrelin, a product of specific endocrine cells in the stomach

and duodenum, actually stimulates food intake and is the mostpotent known circulating orexigen (145). Ghrelin is secretedfrom the fundic region of the stomach and has been identified asthe endogenous ligand for the GH secretagogue receptor. Fastingincreases plasma ghrelin (146), and exogenous ghrelin increasesfood intake when administered peripherally or centrally (147–149). Ghrelin has also been linked to the anticipatory aspects ofmeal ingestion because levels peak shortly before scheduledmeals in humans and rats (150) and fall shortly after meals end.Moreover, elevated ghrelin has been linked to the hyperphagiaand obesity of individuals with the Prader-Willi syndrome (151).Ghrelin is also unique among the GI signals in that its messageappears to be conveyed directly to receptors in the hypothalamus(152–155), although, as is the case for CCK, GLP-1, and otherGI signals, there are ghrelin receptors on vagal sensory nerves(156) but they do not appear to signal satiation (157).

Satiation signals: summary of general principlesSatiation appears to be a complex phenomenon, mediated by

a number of GI peptides. Although it is clear that the differentsatiation factors respond to specific nutrient stimuli (e.g. CCK toprotein and fat, GLP-1 to carbohydrate and fat, PYY primarilyto fat, and so on), it has not been proven that mixed meals ofdiffering macronutrient content elicit the release of distinct cock-tails of GI hormones. However, given the wide range of specificfactors that seem to mediate satiation, it is logical to presume thatthis process is subject to highly refined regulation. Especiallyimportant is the modulation of the action of satiation by factorssuch as leptin and insulin that are responsive to body adiposity.This interaction is the critical site of endocrine regulation ofeating and energy homeostasis.

A key feature of the system regulating food intake is that mostif not all of the peptides that are made in the GI tract and influencesatiation are also synthesized in the brain. This includes CCK,GLP-1, GLP-2, oxyntomodulin, apo A-IV, GRP, NMB, PYY,and ghrelin. Exceptions are the pancreatic hormones that influ-ence energy homeostasis (i.e. insulin, glucagon, and amylin) andthe adipose tissue/stomach hormone leptin; and each of theselatter hormones has long-term effects on body fat as well. Thefact that so many peripheral signals that influence food intake arealso synthesized locally in the brain raises the question ofwhether and how the same signals secreted from different placesin the body interact. A simple generalization is that, if a peptide

S42 Woods and D’Alessio Central Control of Body Weight and Appetite J Clin Endocrinol Metab, November 2008, 93(11):S37–S50

Page 7: s u p p l e m e n t

reduces (or increases) food intake when administered systemi-cally, it probably has the same action when administered cen-trally. With regard to changes of food intake, this is true of CCK,GLP-1, apo A-IV, GRP, NMB, PYY, and ghrelin. Interestingly,it is also generally true that peptide signals that are not synthe-sized in the brain nonetheless have the same effect on food intakewhen administered directly into the brain. This is true of leptin,insulin, and amylin, but not glucagon.

It is worth contemplating why there is such a large imbalanceamong GI hormones that suppress and stimulate food intake; i.e.whereas numerous peptides secreted from the stomach and in-testines decrease food intake, only one known factor, ghrelin,increases it. One possibility relates to the phenomenon of sati-ation itself and the benefits of meal termination. Meals end longbefore any physical limit of the stomach is reached. This is easilydemonstrated when food is diluted with noncaloric bulk andanimals increase the volume of food consumed to attain theirnormal caloric load (158, 159). It has therefore been argued thata primary function of satiation signals is to prevent the consump-tion of too many calories at one time, lest the influx of nutrientsand substrates overwhelm the capacity of the animal to maintainhomeostasis (5, 160). That is, an important role of the GI tractis to analyze and respond to the incoming macronutrients whilea meal is being eaten, helping to preempt excessive challenges tobiochemical homeostasis. A corollary to this role for satiationsignals is that adequate food intake does not require much spe-cific stimulation, only the absence of inhibitory signals for foodintake combined with the presence of food. Until recently therewas some question as to whether manipulation of meal size byfactors activating the satiation system could have therapeuticefficacy for weight reduction. One possibility was that becausethe effects of satiation peptides are dependent on body adiposity,their action would become muted as food intake decreaseddue to homeostatic regulation of body energy stores. Evidencefor this was demonstrated for the satiating action of CCK ingenetically obese Zucker rats (161, 162), but not for rats ren-dered obese by lesions of the ventromedial hypothalamus (163).Furthermore, rats genetically prone to becoming obese when feda high-fat diet (diet-induced obesity rats) are actually more sen-sitive to the satiating action of CCK both before and after be-coming obese (164). Some strains of genetically obese mice arecomparably as sensitive as lean controls (165), and CCK workswell in obese humans when administered iv (166). Hence, thereis no general principle with regard to sensitivity in obesity, atleast with regard to CCK. There is evidence that the satiatingaction of GLP-1 is leptin-dependent (167). All of these observa-tions were made with animals having relatively free access totheir diets so that they could vary their meal patterns in theservice of maintaining body fat; i.e. when individuals are con-strained to eating only small meals, they compensate by eatingmore often, thereby maintaining daily caloric intake and bodyweight. This has been observed when CCK is administered be-fore every meal in experimental animals; animals receiving thistreatment eat smaller and more frequent meals while keepingbody weight constant (33, 168). In contrast, if animals are con-strained to eating only three meals a day and receive a satiatingpeptide at each mealtime, they cannot compensate and conse-

quently lose weight (169). The advent of long-acting formula-tions of GLP-1 and amylin, peptides that seem to have a role insatiation (170, 171), has resulted in weight loss (172, 173). How-ever, at present it is not clear that the chronic effects of GLP-1 andamylin receptor agonists are entirely due to continued hypopha-gia as opposed to other, nonbehavioral actions of the com-pounds.Understandinghowachronicpharmacological stimulusto the satiation system lowers body weight is important for re-fining the models for normal energy homeostasis.

Adiposity Signals

Insulin from the pancreatic B cells and leptin from white adipo-cytes (as well as the stomach and other tissues) are each secretedin direct proportion to body fat. Both hormones are transportedthrough the blood-brain barrier (174, 175) and gain access toneurons in the hypothalamus and elsewhere in the brain to in-fluence energy homeostasis. Hence, neurons sensitive to insulinand/or leptin receive a signal directly proportional to the amountof fat in the body. Consistent with this, if exogenous insulin orleptin is added locally into the brain, the individual responds asif excess fat exists in the body; i.e. food intake is reduced andbody weight is lost. Analogously, if either the leptin or the insulinsignal is reduced locally in the brain, the individual responds asif insufficient fat is present in the body, more food is eaten, andthe individual gains weight. There are many reviews of thesephenomena (1, 2, 87, 176–178).

An important distinction is that whereas satiation signals pri-marily influence how many calories are eaten during individualmeals, adiposity signals are more directly related to how much fatthe body carries and maintains. Developing novel compoundsthat interact directly with the normal detection of and responseto adiposity signals would therefore seem a more promising ther-apeutic approach for obesity. A key question therefore is whetheragonists for the leptin and/or insulin receptor would be viabletargets for the pharmaceutical industry. One obstacle is that tobe effective, any compound would have to gain access to keyreceptors in the brain, yet any such compound would most likelyhave to be administered systemically. Administering insulin sys-temically elicits hypoglycemia and other side effects, and hypo-glycemia per se increases food intake (179–181), thus workingagainst the therapeutic intent. Systemically administered insulindoes result in reduced food intake when plasma glucose is pre-vented from decreasing in animal models (182, 183), but thiswould be difficult to achieve therapeutically. Alternatively, thereare reports that some formulations of nasally administered in-sulin elicit reduced food intake and body weight in humans with-out altering plasma glucose (184, 185).

Leptin does not have the same counterproductive systemiceffects as insulin, and in fact improves insulin sensitivity andcirculating lipoprotein concentrations in subjects with metabolicabnormalities associated with anti-HIV treatment (186). More-over, chronic leptin treatment of patients with generalized lipo-dystrophy causes significant improvements of insulin resistance,hypertriglyceridemia, hepatic steatosis, and glucose metabolism(187), responses found across the spectrum of lipodystrophies.

J Clin Endocrinol Metab, November 2008, 93(11):S37–S50 jcem.endojournals.org S43

Page 8: s u p p l e m e n t

However, the results of clinical trials using leptin in healthy obesesubjects have been variable, with significant weight loss, but notof a remarkable magnitude, and some bothersome side effectsrelated to peptide administration.

Insulin and leptin resistance characterize the obese state,meaning that more of each hormone is required to achieve aparticular physiological effect than occurs in lean individuals.Individuals with diabetes cannot achieve a maximum insulinresponse because of defects in insulin secretion and insulin ac-tion. The insulin and leptin resistance that characterizes periph-eral tissues in obesity is also manifested in the brain. For onething, the transport of both hormones from the blood to the brainis compromised in obesity such that less signal reaches criticalneurons (188–190), and the ability of those neurons to respondis also compromised. When insulin is administered locally intothe brain near the hypothalamus, both genetically obese (191)and dietary obese individuals (192) have a reduced or absentreduction of food intake, and this is the case for leptin as well(193). Hence, an inability on the part of the brain to respond tosignals indicating that there is excess fat in the body may be acontributing and/or confounding factor in obesity. An insulinmimetic that interacts with the insulin receptor and is efficaciouswhen given orally or directly into the brain has been reported toreduce food intake and body weight in obese rodents (194, 195),but it evidently has problematic side effects.

Central Integrating Circuits

Although receptors for insulin and leptin are found in severaldiscrete areas throughout the brain, many that are especiallyimportant for controlling energy homeostasis are localized in theARC of the hypothalamus (Fig. 1). The ARC is ideally suited asa receptor site for body adiposity as well as for integration ofdiverse hormonal and neural signals because there is evidencethat blood-borne molecules have relatively greater access to re-ceptors there than to other brain areas, and this is thought to bedue in part to a relatively leaky blood-brain barrier in the ARC(196, 197). Two categories of ARC neurons are particularly im-portant. One synthesizes the prepropeptide, proopiomelanocor-tin (POMC), and in the ARC POMC is cleaved to �-melanocyte-stimulating hormone (�MSH) as a neurotransmitter. �MSH actsat melanocortin 3 and melanocortin 4 receptors (MC3R andMC4R) on neurons in other hypothalamic areas and elsewherein the brain to reduce food intake, and synthetic agonists forMC3R/MC4R are available that cause hypophagia andweight loss in experimental animals (see reviews in Refs. 1, 2,196, and 198 –202). The catabolic action of both leptin andinsulin relies upon �MSH signaling because administration ofantagonists to MC3R/MC4R blocks each of their actions inthe brain (203, 204).

The second group of ARC neurons synthesizes and secretestwo neuropeptides important in energy homeostasis, and theiraxons project to many of the same brain areas as POMC neurons.Agouti-related peptide (AgRP) is an antagonist at MC3R andMC4R (199) such that one action of these neurons is to counterthe activity of POMC neurons. NPY acts at Y receptors to stim-

ulate food intake (205–207). When either AgRP or NPY is ad-ministered chronically into the brain, body weight increases(202, 208–211). In fact, when a single dose of AgRP is admin-istered into the brain near the ARC, food intake is increased for1 wk or more (212, 213). Insulin and leptin each have a net effectto suppress the activity of NPY/AgRP neurons in the ARC.

The POMC and NPY/AgRP neurons in the ARC share manyimportant features. Each is the origin of tracts projecting to otherhypothalamic and brain areas, the two tracts often occurring inparallel. The POMC-originating tract has an overall cataboliceffect such that when it is more active food intake is reduced,energy expenditure is increased, and if prolonged,body fat is lost.Conversely, the NPY/AgRP-originating tract is anabolic, withheightened activity causing more food to be ingested and body fatto increase. Under normal conditions, both circuits are activesuch that a change of input that is either stimulatory or inhibitoryto either type of neuron elicits rapid changes of many energeticparameters. In the acute situation, both food intake and plasmaglucose are altered because the ARC influences circuits project-ing to behavioral sites as well as autonomic circuits influencinghepatic glucose secretion and pancreatic insulin secretion(214–216).

Another important aspect of the area including the ARC andnearby hypothalamic nuclei is that receptors for many of thesatiation signals discussed above are expressed there; i.e. circu-lating ghrelin is thought to interact directly with ARC neurons(152), which are also directly or indirectly sensitive to changes ofCCK, GLP-1, NMB, and apo A-IV. Because most of these pep-tides are made within the brain, the origin of molecules alteringARC activity may not be directly from the plasma as occurs withinsulin, leptin, and ghrelin. Numerous circuits from the hind-brain satiation area and elsewhere in the brain project to theregion of the ARC (25, 27, 217). Finally, ARC neurons are alsosensitive to local levels of energy-rich nutrients, including glucose(218), some long-chain fatty acids �e.g. oleic acid (219, 220)�,and some amino acids �e.g. leucine (221)�.

Thus, the ARC is situated to be sensitive to a wide array ofsignals important in energy regulation (216, 222–224). It is di-rectly sensitive to hormones whose secretion is proportional tobody fat (insulin and leptin); it receives information on ongoingmeals either directly or indirectly; and it is sensitive to local levelsof nutrients. Importantly, numerous neuronal circuits intercon-nect the ARC and nearby hypothalamic areas with the nucleus ofthe solitary tract, enabling the hypothalamic homeostatic net-work to be constantly aware of ongoing GI activity while at thesame time influencing brain stem autonomic areas projecting tothe GI tract, liver, pancreas, and other tissues (25, 27, 225–227).The ARC can therefore be considered as a key afferent as well asefferent area for the regulation of energy homeostasis.

Although ARC neurons project throughout the brain, twonearby target areas are thought to be especially important. Theparaventricular nuclei (PVN) express both MC3R/MC4R andvarious Y receptors, and PVN neurons in turn synthesize andsecrete neuropeptides that have a net catabolic action, includingCRH and oxytocin. Administration of exogenous CRH (228) oroxytocin (229) into the brain reduces food intake. Hence, a cat-abolic circuit exists in which an increase of body fat is associated

S44 Woods and D’Alessio Central Control of Body Weight and Appetite J Clin Endocrinol Metab, November 2008, 93(11):S37–S50

Page 9: s u p p l e m e n t

with increased insulin and leptin, increased �MSH, and de-creased NPY and AgRP activity, and consequently increased ac-tivity of CRH, oxytocin, and other catabolic signals; all of theselead in turn to reduced food intake and increased energyexpenditure.

The lateral hypothalamic area (LHA) has a contrasting profilefrom the PVN (230). It also receives direct inputs from the ARC,and it contains neurons that synthesize and secrete anabolic pep-tides, including melanin-concentrating hormone and the orex-ins. Administration of melanin-concentrating hormone (231,232) or orexin agonists (233) increases food intake and bodyweight gain. The architecture and functioning of these opposinghypothalamic circuits therefore enables rapid and fine-tunedcontrol over energy homeostasis because the brain can simulta-neously turn up one system (e.g. catabolic or anabolic) whileturning down the other.

Integration of Satiation and Adiposity Signals

Total food intake each day is the sum of the intake in individualmeals (including snacks). As discussed above, the time that mealsare initiated is often under the control of nonhomeostatic influ-ences (4, 5, 160, 234). Hence, whatever regulatory control existsfor body weight must be exerted on how much is eaten in indi-vidual meals, and meal termination is consequently under theinfluence of satiation signals. The efficacy of satiation signals toterminate a meal varies with the amount of fat in the body assignaled to the brain by leptin and insulin. When an individualhas been food restricted (or been on a diet) and loses weight,leptin and insulin secretion both decline, and a reduced adipositysignal reaches the ARC. This in turn lowers sensitivity to satia-tion signals such as CCK, and the consequence is that more foodis eaten during meals before satiation or feeling full occurs. Con-versely, individuals who have overeaten and gained weight haveelevated levels of adiposity signals and enhanced sensitivity tosatiation signals, reducing the trajectory of weight gain or evenpromoting weight loss. When low doses of leptin or insulin areinfused directly into the brain near the ARC, they greatly enhancethe ability of satiation signals to reduce food intake �e.g. muchless CCK or other satiation signals is required to terminate a meal(235–241)�, andwhen theadiposity signal in thebrain is reduced,satiation signals are less efficacious (242).

Conclusion

The brain receives and integrates diverse information pertinentto the maintenance of energy homeostasis. Adiposity signals suchas insulin and leptin act in the arcuate nuclei to provide a back-ground tone, and this tone in turn determines the sensitivity ofthe brain to satiation signals influencing how much food is eatenat any one time. It is important to recognize that this “homeo-static” mechanism provides at most a background influence, andthat it only subtly influences intake during any given meal. Thisis because social factors, palatability, habits, the presence ofpredators, stress, and many other factors are also always at work,

influencing not only when meals occur but how much food isconsumed as well. Only when extraneous factors are tightly con-trolled in laboratory animal experiments, or else when ingestionis precisely monitored and quantified over periods of days orweeks in free-feeding humans, do the effects of these homeostaticsignals become apparent.

Acknowledgments

Address all correspondence and requests for reprints to: Stephen C.Woods, Department of Psychiatry, University of Cincinnati, 2170East Galbraith Road, Cincinnati, Ohio 45237. E-mail: [email protected].

This work was supported by National Institutes of Health AwardsDK 17844 and DK 57900.

Disclosure Summary: S.C.W. received lecture fees from Sanofi-Aven-tis and from Merck. D.A.D. received lecture fees from Merck, Takeda,and MannKind.

References

1. Schwartz MW, Woods SC, Porte Jr D, Seeley RJ, Baskin DG 2000 Centralnervous system control of food intake. Nature 404:661–671

2. Woods SC, Seeley RJ, Porte Jr D, Schwartz MW 1998 Signals that regulatefood intake and energy homeostasis. Science 280:1378–1383

3. Woods SC 2005 Signals that influence food intake and body weight. PhysiolBehav 86:709–716

4. Strubbe JH, Woods SC 2004 The timing of meals. Psychol Rev 111:128–1415. Woods SC 1991 The eating paradox: how we tolerate food. Psychol Rev

98:488–5056. Woods SC, Ramsay DS 2007 Homeostasis: beyond Curt Richter. Appetite

49:388–3987. Moran TH, Kinzig KP 2004 Gastrointestinal satiety signals II. Cholecysto-

kinin. Am J Physiol 286:G183–G1888. Moran TH 2004 Gut peptides in the control of food intake: 30 years of ideas.

Physiol Behav 82:175–1809. Strader AD, Woods SC 2005 Gastrointestinal hormones and food intake.

Gastroenterology 128:175–19110. Polonsky KS, Given E, Carter V 1988 Twenty-four-hour profiles and pulsatile

patterns of insulin secretion in normal and obese subjects. J Clin Invest 81:442–448

11. Teff KL, Townsend RR 2004 Prolonged mild hyperglycemia induces vagallymediated compensatory increase in C-peptide secretion in humans. J ClinEndocrinol Metab 89:5606–5613

12. Kaplan JM, Moran TH 2004 Gastrointestinal signaling in the control of foodintake. In: Stricker EM, Woods SC, eds. Handbook of behavioral neurobi-ology. Neurobiology of food and fluid intake. Vol. 4, no. 2. New York:Kluwer Academic/Plenum Publishing; 273–303

13. Wren AM, Bloom SR 2007 Gut hormones and appetite control. Gastroen-terology 132:2116–2130

14. Wren AM 2008 Gut and hormones and obesity. Front Horm Res 36:165–18115. Smith GP, Gibbs J 1992 The development and proof of the cholecystokinin

hypothesis of satiety. In: Dourish CT, Cooper SJ, Iversen SD, Iversen LL, eds.Multiple cholecystokinin receptors in the CNS. Oxford, UK: Oxford Uni-versity Press; 166–182

16. Chandra R, Liddle RA 2007 Cholecystokinin. Curr Opin Endocrinol Dia-betes Obes 14:63–67

17. Grider JR 1994 Role of cholecystokinin in the regulation of gastrointestinalmotility. J Nutr 124:1334S–1339S

18. Raybould HE 2007 Mechanisms of CCK signaling from gut to brain. CurrOpin Pharmacol 7:570–574

19. Schwartz GJ, Moran TH, White WO, Ladenheim EE 1997 Relationshipsbetween gastric motility and gastric vagal afferent responses to CCK and GRPin rats differ. Am J Physiol 272:R1726—R1733

20. Edwards GL, Ladenheim EE, Ritter RC 1986 Dorsomedial hindbrain par-ticipation in cholecystokinin-induced satiety. Am J Physiol 251:R971–R977

21. Lorenz DN, Goldman SA 1982 Vagal mediation of the cholecystokinin sa-tiety effect in rats. Physiol Behav 29:599–604

J Clin Endocrinol Metab, November 2008, 93(11):S37–S50 jcem.endojournals.org S45

Page 10: s u p p l e m e n t

22. Moran TH, Shnayder L, Hostetler AM, McHugh PR 1988 Pylorectomy re-duces the satiety action of cholecystokinin. Am J Physiol 255:R1059–R1063

23. Moran TH, Baldessarini AR, Salorio CF, Lowery T, Schwartz GJ 1997 Vagalafferent and efferent contributions to the inhibition of food intake by cho-lecystokinin. Am J Physiol 272:R1245—R1251

24. Berthoud HR, Earle T, Zheng H, Patterson LM, Phifer C 2001 Food-relatedgastrointestinal signals activate caudal brainstem neurons expressing bothNMDA and AMPA receptors. Brain Res 915:143–154

25. Berthoud HR 2007 Interactions between the “cognitive” and “metabolic”brain in the control of food intake. Physiol Behav 91:486–498

26. Rinaman L 2004 Hindbrain contributions to anorexia. Am J Physiol RegulIntegr Comp Physiol 287:R1035—R1036

27. Rinaman L 2007 Visceral sensory inputs to the endocrine hypothalamus.Front Neuroendocrinol 28:50–60

28. Rinaman L, Hoffman GE, Dohanics J, Le WW, Stricker EM, Verbalis JG1995 Cholecystokinin activates catecholaminergic neurons in the caudal me-dulla that innervate the paraventricular nucleus of the hypothalamus in rats.J Comp Neurol 360:246–256

29. Beglinger C, Degen L, Matzinger D, D’Amato M, Drewe J 2001 Loxiglumide,a CCK-A receptor antagonist, stimulates calorie intake and hunger feelings inhumans. Am J Physiol 280:R1149–R1154

30. Hewson G, Leighton GE, Hill RG, Hughes J 1988 The cholecystokinin re-ceptor antagonist L364,718 increases food intake in the rat by attenuation ofendogenous cholecystokinin. Br J Pharmacol 93:79–84

31. Reidelberger RD, O’Rourke MF 1989 Potent cholecystokinin antagonistL-364,718 stimulates food intake in rats. Am J Physiol 257:R1512–R1518

32. Crawley JN, Beinfeld MC 1983 Rapid development of tolerance to the be-havioural actions of cholecystokinin. Nature 302:703–706

33. West DB, Fey D, Woods SC 1984 Cholecystokinin persistently suppressesmeal size but not food intake in free-feeding rats. Am J Physiol 246:R776 —R787

34. Lieverse RJ, Jansen JB, Masclee AA, Lamers CB 1995 Satiety effects of aphysiological dose of cholecystokinin in humans. Gut 36:176–179

35. Gutzwiller JP, Degen L, Matzinger D, Prestin S, Beglinger C 2004 Interactionbetween GLP-1 and CCK-33 in inhibiting food intake and appetite in men.Am J Physiol Regul Integr Comp Physiol 287:R562—R567

36. Lieverse RJ, Jansen JB, Masclee AA, Rovati LC, Lamers CB 1994 Effect of alow dose of intraduodenal fat on satiety in humans: studies using the type Acholecystokinin receptor antagonist loxiglumide. Gut 35:501–505

37. Degen L, Drewe J, Piccoli F, Grani K, Oesch S, Bunea R, D’Amato M, Beg-linger C 2007 Effect of CCK-1 receptor blockade on ghrelin and PYY secre-tion in men. Am J Physiol Regul Integr Comp Physiol 292:R1391–R1399

38. Funakoshi A, Miyasaka K, Matsumoto H, Yamamori S, Takiguchi S,Kataoka K, Takata Y, Matsusue K, Kono A, Shimokata H 2000 Gene struc-ture of human cholecystokinin (CCK) type-A receptor: body fat content isrelated to CCK type-A receptor gene promoter polymorphism. FEBS Lett466:264–266

39. Holst JJ 1997 Enteroglucagon. Annu Rev Physiol 59:257–27140. Dube PE, Brubaker PL 2004 Nutrient, neural and endocrine control of glu-

cagon-like peptide secretion. Horm Metab Res 36:755–76041. Drucker DJ, Nauck MA 2006 The incretin system: glucagon-like peptide-1

receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes.Lancet 368:1696–1705

42. Giralt M, Vergara P 1999 Glucagonlike peptide-1 (GLP-1) participation inileal brake induced by intraluminal peptones in rat. Dig Dis Sci 44:322–329

43. Nauck MA, Niedereichholz U, Ettler R, Holst JJ, Orskov C, Ritzel R, SchmiegelWH 1997 Glucagon-like peptide-1 inhibition of gastric emptying outweighs itsinsulinotropic effects in healthy humans. Am J Physiol 273:E981–E988

44. Mentlein R 1999 Dipeptidyl-peptidase IV (CD26)–role in the inactivation ofregulatory peptides. Regul Pept 85:9–24

45. Vahl TP, Paty BW, Fuller BD, Prigeon RL, D’Alessio DA 2003 Effects ofGLP-1-(7–36)NH2, GLP-1-(7–37), and GLP-1- (9–36)NH2 on intravenousglucose tolerance and glucose-induced insulin secretion in healthy humans.J Clin Endocrinol Metab 88:1772–1779

46. Knauf C, Cani PD, Perrin C, Iglesias MA, Maury JF, Bernard E, BenhamedF, Gremeaux T, Drucker DJ, Kahn CR, Girard J, Tanti JF, Delzenne NM,Postic C, Burcelin R 2005 Brain glucagon-like peptide-1 increases insulinsecretion and muscle insulin resistance to favor hepatic glycogen storage.J Clin Invest 115:3554–3563

47. Knauf C, Cani PD, Kim DH, Iglesias MA, Chabo C, Waget A, Colom A,Rastrelli S, Delzenne NM, Drucker DJ, Seeley RJ, Burcelin R 2008 Role ofcentral nervous system glucagon-like peptide-1 receptors in enteric glucose-sensing. Diabetes, 57:2603–2612

48. Vahl TP, Tauchi M, Durler TS, Elfers EE, Fernandes TM, Bitner RD, Ellis KS,Woods SC, Seeley RJ, Herman JP, D’Alessio DA 2007 Glucagon-like pep-

tide-1 (GLP-1) receptors expressed on nerve terminals in the portal vein me-diate the effects of endogenous GLP-1 on glucose tolerance in rats. Endocri-nology 148:4965–4973

49. Tang-Christensen M, Larsen PJ, Goke R, Fink-Jensen A, Jessop DS, MollerM, Sheikh SP 1996 Central administration of GLP-1-(7–36) amide inhibitsfood and water intake in rats. Am J Physiol 271:R848–R856

50. Turton MD, O’Shea D, Gunn I, Beak SA, Edwards CM, Meeran K, Choi SJ,Taylor GM, Heath MM, Lambert PD, Wilding JP, Smith DM, Ghatei MA,Herbert J, Bloom SR 1996 A role for glucagon-like peptide-1 in the centralregulation of feeding �see comments�. Nature 379:69–72

51. Donahey JC, van Dijk G, Woods SC, Seeley RJ 1998 Intraventricular GLP-1reduces short- but not long-term food intake or body weight in lean and obeserats. Brain Res 779:75–83

52. Naslund E, Gutniak M, Skogar S, Rossner S, Hellstrom PM 1998 Glucagon-like peptide 1 increases the period of postprandial satiety and slows gastricemptying in obese men. Am J Clin Nutr 68:525–530

53. Gutzwiller JP, Goke B, Drewe J, Hildebrand P, Ketterer S, Handschin D,Winterhalder R, Conen D, Beglinger C 1999 Glucagon-like peptide-1: a po-tent regulator of food intake in humans. Gut 44:81–86

54. van Dijk G, Thiele TE, Donahey JCK, Campfield LA, Smith FJ, Burn P,Bernstein IL, Woods SC, Seeley RJ 1996 Central infusion of leptin and GLP-1(7–36) amide differentially stimulate c-Fos-like immunoreactivity in the ratbrain. Am J Physiol 271:R1096–R1100

55. Shughrue PJ, Lane MV, Merchenthaler I 1996 Glucagon-like peptide-1receptor (GLP1-R) mRNA in the rat hypothalamus. Endocrinology 137:5159 –5162

56. Navarro M, Rodriquez de Fonseca F, Alvarez E, Chowen JA, Zueco JA,Gomez R, Eng J, Blazquez E 1996 Colocalization of glucagon-like peptide-1(GLP-1) receptors, glucose transporter GLUT-2, and glucokinase mRNAs inrat hypothalamic cells: evidence for a role of GLP-1 receptor agonists as aninhibitory signal for food and water intake. J Neurochem 67:1982–1991

57. Kinzig KP, D’Alessio DA, Seeley RJ 2002 The diverse roles of specific GLP-1receptors in the control of food intake and the response to visceral illness.J Neurosci 22:10470–10476

58. Larsen PJ, Tang-Christensen M, Jessop DS 1997 Central administration ofglucagon-like peptide-1 activates hypothalamic neuroendocrine neurons inthe rat. Endocrinology 138:4445–4455

59. McMahon LR, Wellman PJ 1997 Decreased intake of a liquid diet in non-food-deprived rats following intra-PVN injections of GLP-1 (7–36) amide.Pharmacol Biochem Behav 58:673–677

60. McMahon LR, Wellman PJ 1997 PVN infusion of GLP-1(7–36) amide sup-presses feeding and drinking but does not induce conditioned taste aversionsor alter locomotion in rats. Am J Physiol 274:R23–R29

61. Seeley RJ, Blake K, Rushing PA, Benoit SC, Eng J, Woods SC, D’Alessio D2000 The role of CNS GLP-1-(7–36) amide receptors in mediating the visceralillness effects of lithium chloride. J Neurosci 20:1616–1621

62. Kinzig KP, D’Alessio DA, Herman JP, Sakai RR, Vahl TP, Figueredo HF,Murphy EK, Seeley RJ 2003 CNS glucagon-like peptide-1 receptors mediateendocrine and anxiety responses to interoceptive and psychogenic stressors.J Neurosci 23:6163–6170

63. Naslund E, Barkeling B, King N, Gutniak M, Blundell JE, Holst JJ, RossnerS, Hellstrom PM 1999 Energy intake and appetite are suppressed by gluca-gon-like peptide-1 (GLP-1) in obese men. Int J Obes Relat Metab Disord23:304–311

64. Toft-Nielsen MB, Madsbad S, Holst JJ 1999 Continuous subcutaneous in-fusion of glucagon-like peptide 1 lowers plasma glucose and reduces appetitein type 2 diabetic patients. Diabetes Care 22:1137–1143

65. Gutzwiller JP, Drewe J, Goke B, Schmidt H, Rohrer B, Lareida J, BeglingerC 1999 Glucagon-like peptide-1 promotes satiety and reduces food intake inpatients with diabetes mellitus type 2. Am J Physiol 276:R1541—R1544

66. Delgado-ArosS,KimDY,BurtonDD,ThomfordeGM,StephensD,BrinkmannBH, Vella A, Camilleri M 2002 Effect of GLP-1 on gastric volume, emptying,maximum volume ingested, and postprandial symptoms in humans. Am JPhysiol 282:G424–G431

67. Kastin AJ, Akerstrom V, Pan W 2002 Interactions of glucagon-like peptide-1(GLP-1) with the blood-brain barrier. J Mol Neurosci 18:7–14

68. Ahren B 2007 GLP-1-based therapy of type 2 diabetes: GLP-1 mimetics andDPP-IV inhibitors. Curr Diab Rep 7:340–347

69. Madsbad S, Krarup T, Deacon CF, Holst JJ 2008 Glucagon-like peptidereceptor agonists and dipeptidyl peptidase-4 inhibitors in the treatment ofdiabetes: a review of clinical trials. Curr Opin Clin Nutr Metab Care 11:491–499

70. Mafong DD, Henry RR 2008 Exenatide as a treatment for diabetes andobesity: implications for cardiovascular risk reduction. Curr Atheroscler Rep10:55–60

S46 Woods and D’Alessio Central Control of Body Weight and Appetite J Clin Endocrinol Metab, November 2008, 93(11):S37–S50

Page 11: s u p p l e m e n t

71. Flatt PR, Bailey CJ, Green BD 2008 Dipeptidyl peptidase IV (DPP IV) andrelated molecules in type 2 diabetes. Front Biosci 13:3648–3660

72. Pei Z 2008 From the bench to the bedside: dipeptidyl peptidase IV inhibitors,a new class of oral antihyperglycemic agents. Curr Opin Drug Discov Devel11:512–532

73. Kim D, MacConell L, Zhuang D, Kothare PA, Trautmann M, Fineman M,Taylor K 2007 Effects of once-weekly dosing of a long-acting release formu-lation of exenatide on glucose control and body weight in subjects with type2 diabetes. Diabetes Care 30:1487–1493

74. Kirkegaard P, Moody AJ, Holst JJ, Loud FB, Olsen PS, Christiansen J 1982Glicentin inhibits gastric acid secretion in the rat. Nature 297:156–157

75. Dakin CL, Gunn I, Small CJ, Edwards CM, Hay DL, Smith DM, Ghatei MA,Bloom SR 2001 Oxyntomodulin inhibits food intake in the rat. Endocrinol-ogy 142:4244–4250

76. Dakin CL, Small CJ, Batterham RL, Neary NM, Cohen MA, Patterson M,Ghatei MA, Bloom SR 2004 Peripheral oxyntomodulin reduces food intakeand body weight gain in rats. Endocrinology 145:2687–2695

77. Wynne K, Park AJ, Small CJ, Meeran K, Ghatei MA, Frost GS, Bloom SR2006 Oxyntomodulin increases energy expenditure in addition to decreasingenergy intake in overweight and obese humans: a randomised controlled trial.Int J Obes (Lond) 30:1729–1736

78. Wynne K, Bloom SR 2006 The role of oxyntomodulin and peptide ty-rosine-tyrosine (PYY) in appetite control. Nat Clin Pract EndocrinolMetab 2:612– 620

79. Dakin CL, Small CJ, Park AJ, Seth A, Ghatei MA, Bloom SR 2002 RepeatedICV administration of oxyntomodulin causes a greater reduction in bodyweight gain than in pair-fed rats. Am J Physiol Endocrinol Metab 283:E1173—E1177

80. Cohen MA, Ellis SM, Le Roux CW, Batterham RL, Park A, Patterson M,Frost GS, Ghatei MA, Bloom SR 2003 Oxyntomodulin suppresses appetiteand reduces food intake in humans. J Clin Endocrinol Metab 88:4696–4701

81. Drucker DJ 1999 Glucagon-like peptide 2. Trends Endocrinol Metab 10:153–156

82. Jeppesen PB 2003 Clinical significance of GLP-2 in short-bowel syndrome. JNutr 133:3721–3724

83. Orskov C, Holst JJ, Knuhtsen S, Baldissera FG, Poulsen SS, Nielsen OV 1986Glucagon-like peptides GLP-1 and GLP-2, predicted products of the glucagongene, are secreted separately from pig small intestine but not pancreas. En-docrinology 119:1467–1475

84. Tang-Christensen M, Larsen PJ, Thulesen J, Romer J, Vrang N 2000 Theproglucagon-derived peptide, glucagon-like peptide-2, is a neurotransmitterinvolved in the regulation of food intake. Nat Med 6:802–807

85. Schmidt PT, Naslund E, Gryback P, Jacobsson H, Hartmann B, Holst JJ,Hellstrom PM 2003 Peripheral administration of GLP-2 to humans has noeffect on gastric emptying or satiety. Regul Pept 116:21–25

86. Geary N 1998 Glucagon and the control of meal size. In: Smith GP, ed.Satiation. From gut to brain. New York: Oxford University Press; 164–197

87. Woods SC, Lutz TA, Geary N, Langhans W 2006 Pancreatic signals con-trolling food intake; insulin, glucagon, and amylin. Philos Trans R Soc LondB Biol Sci 361:1219–1235

88. Woods SC, Lotter EC, McKay LD, Porte Jr D 1979 Chronic intracerebro-ventricular infusion of insulin reduces food intake and body weight of ba-boons. Nature 282:503–505

89. Geary N, Le Sauter J, Noh U 1993 Glucagon acts in the liver to controlspontaneous meal size in rats. Am J Physiol 264:R116–R122

90. Langhans W, Zieger U, Scharrer E, Geary N 1982 Stimulation of feedingin rats by intraperitoneal injection of antibodies to glucagon. Science218:894 – 896

91. LeSauterJ,NohU,GearyN1991Hepaticportal infusionofglucagonantibodiesincreases spontaneous meal size in rats. Am J Physiol 261:R162–R165

92. Adrian TE, Bacarese-Hamilton AJ, Smith HA, Chohan P, Manolas KJ,Bloom SR 1987 Distribution and postprandial release of porcine peptide YY.J Endocrinol 113:11–14

93. Grandt D, Schimiczek M, Beglinger C, Layer P, Goebell H, Eysselein VE,Reeve Jr JR 1994 Two molecular forms of peptide YY (PYY) are abundantin human blood: characterization of a radioimmunoassay recognizing PYY1–36 and PYY 3–36. Regul Pept 51:151–159

94. Mentlein R, Dahms P, Grandt D, Kruger R 1993 Proteolytic processing ofneuropeptide Y and peptide YY by dipeptidyl peptidase IV. Regul Pept 49:133–144

95. Larhammar D 1996 Structural diversity of receptors for neuropeptide Y,peptide YY and pancreatic polypeptide. Regul Pept 65:165–174

96. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL,Wren AM, Brynes AE, Low MJ, Ghatei MA, Cone RD, Bloom SR 2002

Gut hormone PYY(3–36) physiologically inhibits food intake. Nature418:650 – 654

97. Vincent RP, le Roux CW 2008 The satiety hormone peptide YY as a regulatorof appetite. J Clin Pathol 61:548–552

98. Lin HC, Zhao XT, Wang L, Wong H 1996 Fat-induced ileal brake in the dogdepends on peptide YY. Gastroenterology 110:1491–1495

99. Pironi L, Stanghellini V, Miglioli M, Corinaldesi R, De Giorgio R, Ruggeri E,Tosetti C, Poggioli G, Morselli Labate AM, Monetti N, Gozzetti G, Barbara L,Go VLW 1993 Fat-induced ileal brake in humans: a dose-dependent phe-nomenon correlated to the plasma levels of peptide YY. Gastroenterology105:733–739

100. Pfluger PT, Kampe J, Castaneda TR, Vahl T, D’Alessio DA, Kruthaupt T,Benoit SC, Cuntz U, Rochlitz HJ, Moehlig M, Pfeiffer AF, Koebnick C,Weickert MO, Otto B, Spranger J, Tschop MH 2007 Effect of human bodyweight changes on circulating levels of peptide YY and peptide YY3–36.J Clin Endocrinol Metab 92:583–588

101. Spiller RC, Trotman IF, Higgins BE, Ghatei MA, Grimble GK, Lee YC,Bloom SR, Misiewicz JJ, Silk DB 1984 The ileal brake—inhibition of jejunalmotility after ileal fat perfusion in man. Gut 25:365–374

102. Nonaka N, Shioda S, Niehoff ML, Banks WA 2003 Characterization ofblood-brain barrier permeability to PYY3–36 in mouse. J Pharmacol ExpTher 306:948–953

103. Batterham RL, Bloom SR 2003 The gut hormone peptide YY regulates ap-petite. Ann NY Acad Sci 994:162–168

104. Degen L, Oesch S, Casanova M, Graf S, Ketterer S, Drewe J, Beglinger C 2005Effect of peptide YY3–36 on food intake in humans. Gastroenterology 129:1430–1436

105. le Roux CW, Batterham RL, Aylwin SJ, Patterson M, Borg CM, Wynne KJ,Kent A, Vincent RP, Gardiner J, Ghatei MA, Bloom SR 2006 Attenuatedpeptide YY release in obese subjects is associated with reduced satiety. En-docrinology 147:3–8

106. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS,Ghatei MA, Bloom SR 2003 Inhibition of food intake in obese subjects bypeptide YY3–36. N Engl J Med 349:941–948

107. Stock S, Leichner P, Wong AC, Ghatei MA, Kieffer TJ, Bloom SR, ChanoineJP 2005 Ghrelin, peptide YY, glucose-dependent insulinotropic polypeptide,and hunger responses to a mixed meal in anorexic, obese, and control femaleadolescents. J Clin Endocrinol Metab 90:2161–2168

108. Lavebratt C, Alpman A, Persson B, Arner P, Hoffstedt J 2006 Commonneuropeptide Y2 receptor gene variant is protective against obesity amongSwedish men. Int J Obes (Lond) 30:453–459

109. Ahituv N, Kavaslar N, Schackwitz W, Ustaszewska A, Collier JM, Hebert S,Doelle H, Dent R, Pennacchio LA, McPherson R 2006 A PYY Q62P variantlinked to human obesity. Hum Mol Genet 15:387–391

110. Tso P, Liu M, Kalogeris TJ, Thomson AB 2001 The role of apolipoproteinA-IV in the regulation of food intake. Annu Rev Nutr 21:231–254

111. Liu M, Doi T, Shen L, Woods SC, Seeley RJ, Zheng S, Jackman A, Tso P 2001Intestinal satiety protein apolipoprotein AIV is synthesized and regulated inrat hypothalamus. Am J Physiol 280:R1382–R1387

112. Fujimoto K, Fukagawa K, Sakata T, Tso P 1993 Suppression of food intakeby apolipoprotein A-IV is mediated through the central nervous system inrats. J Clin Invest 91:1830–1833

113. Fujimoto K, Machidori H, Iwakiri R, Yamamoto K, Fujisaki J, Sakata T, TsoP 1993 Effect of intravenous administration of apolipoprotein A-IV on pat-terns of feeding, drinking and ambulatory activity in rats. Brain Res 608:233–237

114. Lo CM, Zhang DM, Pearson K, Ma L, Sun W, Sakai RR, Davidson WS, LiuM, Raybould HE, Woods SC, Tso P 2007 Interaction of apolipoprotein AIVwith cholecystokinin on the control of food intake. Am J Physiol Regul IntegrComp Physiol 293:R1490—R1494

115. Liu M, Shen L, Doi T, Woods SC, Seeley RJ, Tso P 2003 Neuropeptide Y andlipid increase apolipoprotein AIV gene expression in rat hypothalamus. BrainRes 971:232–238

116. Tso P, Liu M 2004 Apolipoprotein A-IV, food intake, and obesity. PhysiolBehav 83:631–643

117. Okada S, York DA, Bray GA, Erlanson-Albertsson C 1991 Enterostatin(Val-Pro-Asp-Pro-Arg), the activation peptide of procolipase, selectively re-duces fat intake. Physiol Behav 49:1185–1189

118. Shargill NS, Tsuji S, Bray GA, Erlanson-Albertsson C 1991 Enterostatinsuppresses food intake following injection into the third ventricle of rats.Brain Res 544:137–140

119. Mei J, Erlanson-Albertsson C 1992 Effect of enterostatin given intrave-nously and intracerebroventricularly on high-fat feeding in rats. RegulPept 41:209 –218

120. Okada S, York DA, Bray GA, Mei J, Erlanson-Albertsson C 1992 Differential

J Clin Endocrinol Metab, November 2008, 93(11):S37–S50 jcem.endojournals.org S47

Page 12: s u p p l e m e n t

inhibition of fat intake in two strains of rat by the peptide enterostatin. Am JPhysiol 262:R1111–R1116

121. Muurahainen NE, Kissileff HR, Pi-Sunyer FX 1993 Intravenous infusion ofbombesin reduces food intake in humans. Am J Physiol 264:R350–R354

122. Ladenheim EE, Wirth KE, Moran TH 1996 Receptor subtype mediation offeeding suppression by bombesin-like peptides. Pharmacol Biochem Behav54:705–711

123. Stein LJ, Woods SC 1982 Gastrin releasing peptide reduces meal size in rats.Peptides 3:833–835

124. Ladenheim EE, Hampton LL, Whitney AC, White WO, Battey JF, Moran TH2002 Disruptions in feeding and body weight control in gastrin-releasingpeptide receptor deficient mice. J Endocrinol 174:273–281

125. Stuckey JA, Gibbs J, Smith GP 1985 Neural disconnection of gut from brainblocks bombesin-induced satiety. Peptides 6:1249–1252

126. Rushing PA, Henderson RP, Gibbs J 1998 Prolongation of the postprandialintermeal interval by gastrin-releasing peptide 1–27 in spontaneously feedingrats. Peptides 19:175–177

127. Lieverse RJ, Jansen JB, van de Zwan A, Samson L, Masclee AA, Rovati LC,Lamers CB 1993 Bombesin reduces food intake in lean man by a cholecys-tokinin-independent mechanism. J Clin Endocrinol Metab 76:1495–1498

128. Gutzwiller JP, Drewe J, Hildebrand P, Rossi L, Lauper JZ, Beglinger C 1994Effect of intravenous human gastrin-releasing peptide on food intake in hu-mans. Gastroenterology 106:1168–1173

129. Hildebrand P, Lehmann FS, Ketterer S, Christ AD, Stingelin T, Beltinger J,Gibbons AH, Coy DH, Calam J, Larsen F, Beglinger C 2001 Regulation ofgastric function by endogenous gastrin releasing peptide in humans: studieswith a specific gastrin releasing peptide receptor antagonist. Gut 49:23–28

130. Degen LP, Peng F, Collet A, Rossi L, Ketterer S, Serrano Y, Larsen F, BeglingerC, Hildebrand P 2001 Blockade of GRP receptors inhibits gastric emptyingand gallbladder contraction but accelerates small intestinal transit. Gastro-enterology 120:361–368

131. Ludvik B, Kautzky-Willer A, Prager R, Thomaseth K, Pacini G 1997 Amylin:history and overview. Diabet Med 14(Suppl 2):S9–S13

132. Chance WT, Balasubramaniam A, Zhang FS, Wimalawansa SJ, Fischer JE1991 Anorexia following the intrahypothalamic administration of amylin.Brain Res 539:352–354

133. Lutz TA 2006 Amylinergic control of food intake. Physiol Behav 89:465–471134. Lutz TA, Del Prete E, Scharrer E 1994 Reduction of food intake in rats by

intraperitoneal injection of low doses of amylin. Physiol Behav 55:891–895135. Lutz TA, Geary N, Szabady MM, Del Prete E, Scharrer E 1995 Amylin

decreases meal size in rats. Physiol Behav 58:1197–1202136. Rushing PA, Hagan MM, Seeley RJ, Lutz TA, Woods SC 2000 Amylin: a

novel action in the brain to reduce body weight. Endocrinology 141:850–853137. Rushing PA, Hagan MM, Seeley RJ, Lutz TA, D’Alessio DA, Air EL, Woods

SC 2001 Inhibition of central amylin signaling increases food intake and bodyadiposity in rats. Endocrinology 142:5035

138. Martinez A, Kapas S, Miller MJ, Ward Y, Cuttitta F 2000 Coexpression ofreceptors for adrenomedullin, calcitonin gene-related peptide, and amylin inpancreatic �-cells. Endocrinology 141:406–411

139. Christopoulos A, Christopoulos G, Morfis M, Udawela M, Laburthe M,Couvineau A, Kuwasako K, Tilakaratne N, Sexton PM 2003 Novel receptorpartners and function of receptor activity-modifying proteins. J Biol Chem278:3293–3297

140. Lutz TA, Senn M, Althaus J, Del Prete E, Ehrensperger F, Scharrer E 1998Lesion of the area postrema/nucleus of the solitary tract (AP/NTS) attenuatesthe anorectic effects of amylin and calcitonin gene-related peptide (CGRP) inrats. Peptides 19:309–317

141. Rushing PA, Lutz TA, Seeley RJ, Woods SC 2000 Amylin and insulin interactto reduce food intake in rats. Horm Metab Res 32:62–65

142. Mollet A, Meier S, Grabler V, Gilg S, Scharrer E, Lutz TA 2003 Endogenousamylin contributes to the anorectic effects of cholecystokinin and bombesin.Peptides 24:91–98

143. Whitehouse F, Kruger DF, Fineman M, Shen L, Ruggles JA, Maggs DG,Weyer C, Kolterman OG 2002 A randomized study and open-label extensionevaluating the long-term efficacy of pramlintide as an adjunct to insulintherapy in type 1 diabetes. Diabetes Care 25:724–730

144. Hollander PA, Levy P, Fineman MS, Maggs DG, Shen LZ, Strobel SA, WeyerC, Kolterman OG 2003 Pramlintide as an adjunct to insulin therapy improveslong-term glycemic and weight control in patients with type 2 diabetes: a1-year randomized controlled trial. Diabetes Care 26:784–790

145. Wiedmer P, Nogueiras R, Broglio F, D’Alessio D, Tschop MH 2007 Ghrelin,obesity and diabetes. Nat Clin Pract Endocrinol Metab 3:705–712

146. Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, Weigle DS2001 A preprandial rise in plasma ghrelin levels suggests a role in meal ini-tiation in humans. Diabetes 50:1714–1719

147. Asakawa A, Inui A, Kaga T, Yuzuriha H, Nagata T, Ueno N, Makino S,Fujimiya M, Niijima A, Fujino MA, Kasuga M 2001 Ghrelin is an appetite-stimulatory signal from stomach with structural resemblance to motilin. Gas-troenterology 120:337–345

148. Wren AM, Small CJ, Abbott CR, Dhillo WS, Seal LJ, Cohen MA, BatterhamRL, Taheri S, Stanley SA, Ghatei MA, Bloom SR 2001 Ghrelin causes hy-perphagia and obesity in rats. Diabetes 50:2540–2547

149. Tschop M, Smiley DL, Heiman ML 2000 Ghrelin induces adiposity in ro-dents. Nature 407:908–913

150. Drazen DL, Vahl TP, D’Alessio DA, Seeley RJ, Woods SC 2006 Effects of afixed meal pattern on ghrelin secretion: evidence for a learned response in-dependent of nutrient status. Endocrinology 147:23–30

151. Cummings DE, Clement K, Purnell JQ, Vaisse C, Foster KE, Frayo RS,Schwartz MW, Basdevant A, Weigle DS 2002 Elevated plasma ghrelin levelsin Prader Willi syndrome. Nat Med 8:643–644

152. Cowley MA, Smith RG, Diano S, Tschop M, Pronchuk N, Grove KL,Strasburger CJ, Bidlingmaier M, Esterman M, Heiman ML, Garcia-SeguraLM, Nillni EA, Mendez P, Low MJ, Sotonyi P, Friedman JM, Liu H, Pinto S,Colmers WF, Cone RD, Horvath TL 2003 The distribution and mechanismof action of ghrelin in the CNS demonstrates a novel hypothalamic circuitregulating energy homeostasis. Neuron 37:649–661

153. Lawrence CB, Snape AC, Baudoin FM, Luckman SM 2002 Acute centralghrelin and GH secretagogues induce feeding and activate brain appetitecenters. Endocrinology 143:155–162

154. Cummings DE 2006 Ghrelin and the short- and long-term regulation ofappetite and body weight. Physiol Behav 89:71–84

155. Traebert M, Riediger T, Whitebread S, Scharrer E, Schmid HA 2002 Ghrelinacts on leptin-responsive neurones in the rat arcuate nucleus. J Neuroendo-crinol 14:580–586

156. Date Y, Murakami N, Toshinai K, Matsukura S, Niijima A, Matsuo H,Kangawa K, Nakazato M 2002 The role of the gastric afferent vagal nerve inghrelin-induced feeding and growth hormone secretion in rats. Gastroenter-ology 123:1120–1128

157. Arnold M, Mura A, Langhans W, Geary N 2006 Gut vagal afferents are notnecessary for the eating-stimulatory effect of intraperitoneally injected gh-relin in the rat. J Neurosci 26:11052–11060

158. Adolph EF 1947 Urges to eat and drink in rats. Am J Physiol 151:110–125159. Janowitz HD, Grossman MI 1949 Effect of variations in nutritive density of

food in dogs and rats. Am J Physiol 158:184–193160. Woods SC, Strubbe JH 1994 The psychobiology of meals. Psychon Bull Rev

1:141–155161. McLaughlin CL, Baile CA 1979 Cholecystokinin, amphetamine and diaze-

pam and feeding in lean and obese Zucker rats. Pharmacol Biochem Behav10:87–93

162. Niederau C, Meereis-Schwanke K, Klonowski-Stumpe H, Herberg L 1997CCK resistance in Zucker obese versus lean rats. Regul Pept 70:97–104

163. Kulkosky PJ, Breckenridge C, Krinsky R, Woods SC 1976 Satiety elicited bythe C-terminal octapeptide of cholecystokinin-pancreozymin in normal andVMH-lesioned rats. Behav Biol 18:227–234

164. Chandler PC, Wauford PK, Oswald KD, Maldonado CR, Hagan MM 2004Change in CCK-8 response after diet-induced obesity and MC3/4-receptorblockade. Peptides 25:299–306

165. Strohmayer AJ, Smith GP 1981 Cholecystokinin inhibits food intake in ge-netically obese (C57BL/6j-ob) mice. Peptides 2:39–43

166. Pi-Sunyer X, Kissileff HR, Thornton J, Smith GP 1982 C-terminal octapep-tide of cholecystokinin decreases food intake in obese men. Physiol Behav29:627–630

167. Williams DL, Baskin DG, Schwartz MW 2006 Leptin regulation of the an-orexic response to glucagon-like peptide-1 receptor stimulation. Diabetes55:3387–3393

168. West DB, Greenwood MRC, Marshall KA, Woods SC 1987 Lithium chlo-ride, cholecystokinin and meal patterns: evidence the cholecystokinin sup-presses meal size in rats without causing malaise. Appetite 8:221–227

169. West DB, Williams RH, Braget DJ, Woods SC 1982 Bombesin reduces foodintake of normal and hypothalamically obese rats and lowers body weightwhen given chronically. Peptides 3:61–67

170. D’Alessio DA, Vahl TP 2004 Glucagon-like peptide 1: evolution of an in-cretin into a treatment for diabetes. Am J Physiol Endocrinol Metab 286:E882—E890

171. D’Alessio DA, Vahl TP 2005 Utilizing the GLP-1 signaling system to treatdiabetes: sorting through the pharmacologic approaches. Curr Diab Rep5:346–352

172. Buse JB, Klonoff DC, Nielsen LL, Guan X, Bowlus CL, Holcombe JH, MaggsDG, Wintle ME 2007 Metabolic effects of two years of exenatide treatmenton diabetes, obesity, and hepatic biomarkers in patients with type 2 diabetes:

S48 Woods and D’Alessio Central Control of Body Weight and Appetite J Clin Endocrinol Metab, November 2008, 93(11):S37–S50

Page 13: s u p p l e m e n t

an interim analysis of data from the open-label, uncontrolled extension ofthree double-blind, placebo-controlled trials. Clin Ther 29:139–153

173. Poon T, Nelson P, Shen L, Mihm M, Taylor K, Fineman M, Kim D 2005Exenatide improves glycemic control and reduces body weight in subjectswith type 2 diabetes: a dose-ranging study. Diabetes Technol Ther 7:467–477

174. Banks WA 2006 The blood-brain barrier as a regulatory interface in thegut-brain axes. Physiol Behav 89:472–476

175. Woods SC, Seeley RJ, Baskin DG, Schwartz MW 2003 Insulin and the blood-brain barrier. Curr Pharm Des 9:795–800

176. Benoit SC, Clegg DJ, Seeley RJ, Woods SC 2004 Insulin and leptin as adi-posity signals. Recent Prog Horm Res 59:267–285

177. Friedman JM 2002 The function of leptin in nutrition, weight, and physiol-ogy. Nutr Rev 60:S1–S14; discussion S68- 84:85–87

178. Seeley RJ, Woods SC 2003 Monitoring of stored and available fuel by theCNS: implications for obesity. Nat Rev Neurosci 4:901–909

179. Grossman SP 1986 The role of glucose, insulin and glucagon in the regulationof food intake and body weight. Neurosci Biobehav Rev 10:295–315

180. Langhans W 1996 Metabolic and glucostatic control of feeding. Proc NutrSoc 55:497–515

181. Lotter EC, Woods SC 1977 Injections of insulin and changes of body weight.Physiol Behav 18:293–297

182. Nicolaidis S, Rowland N 1976 Metering of intravenous versus oral nutrientsand regulation of energy balance. Am J Physiol 231:661–668

183. Vanderweele DA, Haraczkiewicz E, Van Itallie TB 1982 Elevated insulin andsatiety in obese and normal weight rats. Appetite 3:99–109

184. Hallschmid M, Benedict C, Schultes B, Fehm HL, Born J, Kern W 2004Intranasal insulin reduces body fat in men but not in women. Diabetes 53:3024–3029

185. Hallschmid M, Benedict C, Born J, Fehm HL, Kern W 2004 Manipulatingcentral nervous mechanisms of food intake and body weight regulation byintranasal administration of neuropeptides in man. Physiol Behav 83:55–64

186. Lee JH, Chan JL, Sourlas E, Raptopoulos V, Mantzoros CS 2006 Recombi-nant methionyl human leptin therapy in replacement doses improves insulinresistance and metabolic profile in patients with lipoatrophy and metabolicsyndrome induced by the highly active antiretroviral therapy. J Clin Endo-crinol Metab 91:2605–2611

187. Javor ED, Cochran EK, Musso C, Young JR, Depaoli AM, Gorden P 2005Long-term efficacy of leptin replacement in patients with generalized lipo-dystrophy. Diabetes 54:1994–2002

188. Israel PA, Park CR, Schwartz MW, Green PK, Sipols AJ, Woods SC, Porte JrD, Figewicz DP 1993 Effect of diet-induced obesity and experimental hyper-insulinemia on insulin uptake into CSF of the rat. Brain Res Bull 30:571–575

189. Owen OE, Reichard GAJ, Boden G, Shuman C 1974 Comparative measure-ments of glucose, �-hydroxybutyrate, acetoacetate, and insulin in blood andcerebrospinal fluid during starvation. Metabolism 23:7–14

190. Stein LJ, Dorsa DM, Baskin DG, Figlewicz DP, Ikeda H, Frankmann SP,Greenwood MR, Porte Jr D, Woods SC 1983 Immunoreactive insulin levelsare elevated in the cerebrospinal fluid of genetically obese Zucker rats. En-docrinology 113:2299–2301

191. Ikeda H, West DB, Pustek JJ, Figlewicz DP, Greenwood MRC, Porte Jr D,Woods SC 1986 Intraventricular insulin reduces food intake and body weightof lean but not obese Zucker rats. Appetite 7:381–386

192. Woods SC, D’Alessio DA, Tso P, Rushing PA, Clegg DJ, Benoit SC, GotohK, Liu M, Seeley RJ 2004 Consumption of a high-fat diet alters the homeo-static regulation of energy balance. Physiol Behav 83:573–578

193. Cota D, Matter EK, Woods SC, Seeley RJ 2008 The role of hypothalamicmTORC1 signaling in diet-induced obesity. J Neurosci 28:7202–7208

194. Air EL, Strowski MZ, Benoit SC, Conarello SL, Salituro GM, Guan XM, Liu K,Woods SC, Zhang BB 2002 Small molecule insulin mimetics reduce food intakeand body weight and prevent development of obesity. Nat Med 8:179–183

195. Zhang B, Salituro G, Szalkowski D, Li Z, Zhang Y, Royo I, Vilella D, DiezMT, Pelaez F, Ruby C, Kendall RL, Mao X, Griffin P, Calaycay J, Zierath JR,Heck JV, Smith RG, Moller DE 1999 Discovery of a small molecule insulinmimetic with antidiabetic activity in mice. Science 284:974–977

196. Cone RD, Cowley MA, Butler AA, Fan W, Marks DL, Low MJ 2001 Thearcuate nucleus as a conduit for diverse signals relevant to energy homeosta-sis. Int J Obes Relat Metab Disord 25(Suppl 5):S63—S67

197. Peruzzo B, Pastor FE, Blazquez JL, Schobitz K, Pelaez B, Amat P, RodriguezEM 2000 A second look at the barriers of the medial basal hypothalamus. ExpBrain Res 132:10–26

198. Ahima RS, Saper CB, Flier JS, Elmquist JK 2000 Leptin regulation of neu-roendocrine systems. Front Neuroendocrinol 21:263–307

199. Cone RD 2005 Anatomy and regulation of the central melanocortin system.Nat Neurosci 8:571–578

200. Elmquist JK, Coppari R, Balthasar N, Ichinose M, Lowell BB 2005 Identi-

fying hypothalamic pathways controlling food intake, body weight, and glu-cose homeostasis. J Comp Neurol 493:63–71

201. Schwartz MW, Porte Jr D 2005 Diabetes, obesity, and the brain. Science307:375–379

202. Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW 2006Central nervous system control of food intake and body weight. Nature443:289–295

203. Benoit SC, Air EL, Coolen LM, Strauss R, Jackman A, Clegg DJ, Seeley RJ,Woods SC 2002 The catabolic action of insulin in the brain is mediated bymelanocortins. J Neurosci 22:9048–9052

204. Seeley R, Yagaloff K, Fisher S, Burn P, Thiele T, van DG, Baskin D, SchwartzM 1997 Melanocortin receptors in leptin effects. Nature 390:349

205. Stanley BG, Leibowitz SF 1984 Neuropeptide Y: stimulation of feedingand drinking by injection into the paraventricular nucleus. Life Sciences35:2635–2642

206. Clark JT, Kalra PS, Crowley WR, Kalra SP 1984 Neuropeptide Y and humanpancreatic polypeptide stimulate feeding behavior in rats. Endocrinology115:427–429

207. Corp ES, Melville LD, Greenberg D, Gibbs J, Smith GP 1990 Effect of fourthventricular neuropeptide Y and peptide YY on ingestive and other behaviors.Am J Physiol 259:R317–R323

208. Zarjevski N, Cusin I, Vetter R, Rohner-Jeanrenaud F, Jeanrenaud B 1993Chronic intracerebroventricular neuropeptide-Y administration to normalrats mimics hormonal and metabolic changes of obesity. Endocrinology 133:1753–1758

209. Wilson BD, Ollmann MM, Barsh GS 1999 The role of agouti-related proteinin regulating body weight. Mol Med Today 5:250–256

210. Vettor R, Zarjevski N, Cusin I, Johner-Jeanrenaud F, Jeanrenaud B 1994Induction and reversibility of an obesity syndrome by intracerebroventricularneuropeptide Y administration to normal rats. Diabetologia 37:1202–1208

211. Ollmann M, Wilson B, Yang Y, Kerns J, Chen Y, Gantz I, Barsh G 1997Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 278:135–138

212. Hagan MM, Rushing PA, Pritchard LM, Schwartz MW, Strack AM, Van derPloeg HT, Woods SC, Seeley RJ 2000 Long-term orexigenic effects of AgRP-(83–132) involve mechanisms other than melanocortin receptor blockade.Am J Physiol 279:R47–R52

213. Hagan MM, Benoit SC, Rushing PA, Pritchard LM, Woods SC, Seeley RJ2001 Immediate and prolonged patterns of agouti-related peptide-(83–132)-induced c-Fos activation in hypothalamic and extrahypothalamic sites. En-docrinology 142:1050–1056

214. Porte Jr D, Baskin DG, Schwartz MW 2005 Insulin signaling in the centralnervous system: a critical role in metabolic homeostasis and disease from C.elegans to humans. Diabetes 54:1264–1276

215. Schwartz MW 2006 Central nervous system regulation of food intake. Obe-sity (Silver Spring) 14(Suppl 1):1S–8S

216. Woods SC, Seeley RJ, Cota D 2008 Regulation of food intake through hypo-thalamic signaling networks involving mTOR. Annu Rev Nutr 28:295–311

217. Berthoud HR 2002 Multiple neural systems controlling food intake and bodyweight. Neurosci Biobehav Rev 26:393–428

218. Levin BE 2006 Metabolic sensing neurons and the control of energy ho-meostasis. Physiol Behav 89:486–489

219. Lam TK, Pocai A, Gutierrez-Juarez R, Obici S, Bryan J, Aguilar-Bryan L,Schwartz GJ, Rossetti L 2005 Hypothalamic sensing of circulating fatty acidsis required for glucose homeostasis. Nat Med 11:320–327

220. Obici S, Feng Z, Morgan K, Stein D, Karkanias G, Rossetti L 2002 Centraladministration of oleic acid inhibits glucose production and food intake.Diabetes 51:271–275

221. Cota D, Proulx K, Woods SC, Seeley RJ 2006 Role of leucine in regulatingfood intake. Science 313:1236–1238

222. Cota D, Proulx K, Seeley RJ 2007 The role of CNS fuel sensing in energy andglucose regulation. Gastroenterology 132:2158–2168

223. Seeley RJ, York DA 2005 Fuel sensing and the central nervous system (CNS):implications for the regulation of energy balance and the treatment for obe-sity. Obes Rev 6:259–265

224. Fehm HL, Kern W, Peters A 2006 The selfish brain: competition for energyresources. Prog Brain Res 153:129–140

225. Berthoud HR 2006 Homeostatic and non-homeostatic pathways involved inthe control of food intake and energy balance. Obesity (Silver Spring)14(Suppl 5):197S–200S

226. Grill HJ, Kaplan JM 2002 The neuroanatomical axis for control of energybalance. Front Neuroendocrinol 23:2–40

227. Schwartz GJ 2006 Integrative capacity of the caudal brainstem in the controlof food intake. Philos Trans R Soc Lond B Biol Sci 361:1275–1280

228. Richard D, Huang Q, Timofeeva E 2000 The corticotropin-releasing hor-

J Clin Endocrinol Metab, November 2008, 93(11):S37–S50 jcem.endojournals.org S49

Page 14: s u p p l e m e n t

mone system in the regulation of energy balance in obesity. Int J Obes RelatMetab Disord 24:S36—S39

229. Verbalis JG, Blackburn RE, Olson BR, Stricker EM 1993 Central oxytocininhibition of food and salt ingestion: a mechanism for intake regulation ofsolute homeostasis. Regul Pept 45:149–154

230. Broberger C, De Lecea L, Sutcliffe JG, Hokfelt T 1998 Hypocretin/orexin-and melanin-concentrating hormone-expressing cells form distinct popula-tions in the rodent lateral hypothalamus: relationship to the neuropeptide Yand agouti gene-related protein systems. J Comp Neurol 402:460–474

231. Qu D, Ludwig DS, Gammeltoft S, Piper M, Pelleymounter MA, Cullen MJ,Mathes WF, Przypek R, Kanarek R, Maratos-Flier E 1996 A role for melanin-concentrating hormone in the central regulation of feeding behaviour. Nature380:243–247

232. Nahon JL 2006 The melanocortins and melanin-concentrating hormone inthe central regulation of feeding behavior and energy homeostasis. C R Biol329:623–638; discussion 653–655

233. Sweet DC, Levine AS, Billington CJ, Kotz CM 1999 Feeding response tocentral orexins. Brain Res 821:535–538

234. Strubbe JH, van Dijk G 2002 The temporal organization of ingestive behav-iour and its interaction with regulation of energy balance. Neurosci BiobehavRev 26:485–498

235. Matson CA, Wiater MF, Kuijper JL, Weigle DS 1997 Synergy between

leptin and cholecystokinin (CCK) to control daily caloric intake. Peptides18:1275–1278

236. Matson CA, Ritter RC 1999 Long-term CCK-leptin synergy suggests a rolefor CCK in the regulation of body weight. Am J Physiol 276:R1038–R1045

237. Riedy CA, Chavez M, Figlewicz DP, Woods SC 1995 Central insulin en-hances sensitivity to cholecystokinin. Physiol Behav 58:755–760

238. Emond M, Schwartz GJ, Ladenheim EE, Moran TH 1999 Central leptinmodulates behavioral and neural responsivity to CCK. Am J Physiol 276:R1545–R1549

239. Figlewicz DP, Stein LJ, West D, Porte Jr D, Woods SC 1986 Intracisternalinsulin alters sensitivity to CCK-induced meal suppression in baboons. Am JPhysiol 250:R856–R860

240. Ladenheim EE, Emond M, Moran TH 2005 Leptin enhances feeding sup-pression and neural activation produced by systemically administered bomb-esin. Am J Physiol Regul Integr Comp Physiol 289:R473–R477

241. Morton GJ, Blevins JE, Williams DL, Niswender KD, Gelling RW, RhodesCJ, Baskin DG, Schwartz MW 2005 Leptin action in the forebrain regulatesthe hindbrain response to satiety signals. J Clin Invest 115:703–710

242. McMinn JE, Sindelar DK, Havel PJ, Schwartz MW 2000 Leptin deficiencyinduced by fasting impairs the satiety response to cholecystokinin. Endocri-nology 141:4442–4448

243. Grill HJ, Smith GP 1988 Cholecystokinin decreases sucrose intake in chronicdecerebrate rats. Am J Physiol 254:R853–R856

S50 Woods and D’Alessio Central Control of Body Weight and Appetite J Clin Endocrinol Metab, November 2008, 93(11):S37–S50