Hostname: page-component-76fb5796d-5g6vh Total loading time: 0 Render date: 2024-04-26T23:09:13.502Z Has data issue: false hasContentIssue false

Peripheral tissue–brain interactions in the regulation of food intake

Published online by Cambridge University Press:  28 February 2007

Miguel López
Affiliation:
Department of Physiology, School of Medicine, University of Santiago de Compostela, C/San Francisco s/n 15782, Santiago de Compostela, A Coruña, Spain
Sulay Tovar
Affiliation:
Department of Physiology, School of Medicine, University of Santiago de Compostela, C/San Francisco s/n 15782, Santiago de Compostela, A Coruña, Spain
María J. Vázquez
Affiliation:
Department of Physiology, School of Medicine, University of Santiago de Compostela, C/San Francisco s/n 15782, Santiago de Compostela, A Coruña, Spain
Lynda M. Williams
Affiliation:
Energy Balance and Obesity Division, Rowett Research Institute, Aberdeen, UK
Carlos Diéguez*
Affiliation:
Department of Physiology, School of Medicine, University of Santiago de Compostela, C/San Francisco s/n 15782, Santiago de Compostela, A Coruña, Spain
*
*Corresponding author: Professor Carlos Diéguez, fax +34 981574145, fscadigo@usc.es
Rights & Permissions [Opens in a new window]

Abstract

More than 70 years ago the glucostatic, lipostatic and aminostatic hypotheses proposed that the central nervous system sensed circulating levels of different metabolites, changing feeding behaviour in response to the levels of those molecules. In the last 20 years the rapid increase in obesity and associated pathologies in developed countries has involved a substantial increase in the knowledge of the physiological and molecular mechanism regulating body mass. This effort has resulted in the recent discovery of new peripheral signals, such as leptin and ghrelin, as well as new neuropeptides, such as orexins, involved in body-weight homeostasis. The present review summarises research into energy balance, starting from the original classical hypotheses proposing metabolite sensing, through peripheral tissue–brain interactions and coming full circle to the recently-discovered role of hypothalamic fatty acid synthase in feeding regulation. Understanding these molecular mechanisms will provide new pharmacological targets for the treatment of obesity and appetite disorders.

Type
Research Article
Copyright
Copyright © The Authors 2007

Abbreviations:
AgRP

agouti-related peptide

ARC

arcuate nucleus

BBB

blood–brain barrier

CART

cocaine- and amphetamine-regulated transcript

CCK

cholecystokinin

CB

cannabinoids

CNS

central nervous system

DMH

dorsomedial nucleus of the hypothalamus

EC

endocannabinoids

FAS

fatty acid synthase

GHS-R

growth hormone secretagogue receptor

GLP

glucagon-like peptide

NPY

neuropeptide Y

LHA

lateral hypothalamic area

MCH

melanin-concentrating hormone

MCnR

melanocortin receptor (n 1–5)

NTS

nucleus of the solitary tract

OB-Ra–f

isoforms of leptin receptor

OX

orexin

OXM

oxyntomodulin

POMC

pro-opiomelanocortin

PP

pancreatic polypeptide

PVN

paraventricular nucleus

PYY

peptide YY

VMH

ventromedial nucleus of the hypothalamus

The prevalence of overweight and obesity in most developed countries has increased strikingly during the last 30 years (Friedman, Reference Friedman2000, Reference Friedman2003; Flier, Reference Flier2004; Farooqi & O'Rahilly, Reference Farooqi and O'Rahilly2005). Body weight depends on the balance between energy intake and energy consumption. Despite wide daily variation in food intake and energy expenditure, for most individuals body weight remains extremely stable over long periods of time. For this stability to occur, feeding and energy expenditure must be constantly modulated and balanced. Obesity results when the former exceeds the latter and there is an accumulation of an excess of fat in peripheral tissues (e.g. white adipose tissue, which is specifically adapted for this function), liver and muscle, which results in metabolic disease (Friedman, Reference Friedman2000, Reference Friedman2003; Flier, Reference Flier2004; Farooqi & O'Rahilly, Reference Farooqi and O'Rahilly2005). Obesity has a profound impact on human health and lifespan. Being obese correlates not just with associated metabolic dysfunction such as type 2 diabetes and CVD, but is also associated with the occurrence of certain cancers (Calle & Kaaks, Reference Calle and Kaaks2004).

The first hypotheses proposed to explain the periphery–brain interaction in the regulation of food intake were the glucostatic, lipostatic and aminostatic hypotheses. These models proposed that circulating factors, e.g. lipids (lipostatic hypothesis), glucose (glucostatic hypothesis) or protein products (aminostatic hypothesis), that are generated in proportion to body fat stores and/or nutritional status act as signals to the brain, eliciting changes in energy intake and expenditure (Campfield et al. Reference Campfield, Smith and Burn1996). The current ‘obesity epidemic’ has driven forward research efforts in the investigation of body-weight homeostasis. For this reason, in the last decade there has been a major increase in the knowledge of the physiological and molecular mechanism regulating body mass. Animals are now known to regulate body weight by a complex homeostatic mechanism involving interactions between peripheral organs and the central nervous system (CNS). Peripheral organs (such as white adipose tissue), gut, thyroid, muscle and gonads produce signals that inform brain centres of the nutritional, as well as metabolic, status of the animal (Flier, Reference Flier2004; Horvath et al. Reference Horvath, Diano and Tschop2004; Abizaid et al. Reference Abizaid, Gao and Horvath2006; Morton et al. Reference Morton, Cummings, Baskin, Barsh and Schwartz2006). The CNS receives and integrates this entire signalling system, adjusting energy intake (food intake) and energy expenditure, according to the demands of the organism.

The present review summarises the current knowledge about periphery–brain interactions in the regulation of feeding. A full understanding of these mechanisms will allow the establishment of effective therapies to counter eating disorders and obesity.

Gastrointestinal signals regulating food intake

In addition to its evident function in the digestion and absorption of nutrients, the gut and associated organs (liver, pancreas and visceral white adipose tissue depots) play an important role in the control of energy homeostasis, particularly in the short-term regulation of food intake. Both the enteric nervous system and gut hormones are known to control the initiation and termination of individual meals (Halford & Blundell, Reference Halford and Blundell2000a; Badman & Flier, Reference Badman and Flier2005; Perez-Tilve et al. Reference Perez-Tilve, Nogueiras, Mallo, Benoit and Tschoep2006).

Enteric nervous system

The gastrointestinal tract receives a dual extrinsic innervation from the autonomic nervous system via its parasympathetic (cholinergic) division, which includes vagal and pelvic nerves, and its sympathetic (noradrenergic) division, which comprises splanchnic nerves. Parasympathetic innervation is mainly inhibitory, and sympathetic innervation is mainly excitatory. In addition to this autonomic innervation, the gastrointestinal tract also has its own nervous system, i.e. the enteric nervous system (Konturek et al. Reference Konturek, Konturek, Pawlik and Brzozowski2004; Badman & Flier, Reference Badman and Flier2005), which is involved in every aspect of gut function, from mastication to defaecation. Besides these roles, the enteric nervous system is also implicated in gastric and pancreatic exocrine secretion, gut motility, blood supply and hormone release (Konturek et al. Reference Konturek, Konturek, Pawlik and Brzozowski2004; Badman & Flier, Reference Badman and Flier2005).

The enteric nervous system projects to the CNS through vagal and sympathetic (spinal) nerves. These projections transmit a variety of information to several CNS areas, including mechanical stimuli (distension, contraction), chemical stimuli (presence of nutrients in the gut lumen) and neurohumoral stimuli (gut hormones, neurotransmitters and neuromodulators; Langley, Reference Langley1994; Konturek et al. Reference Konturek, Konturek, Pawlik and Brzozowski2004). Most of these afferent vagal fibres terminate in the nucleus of the solitary tract (NTS) in the brainstem, and in laminas I and V of the dorsal horn of spinal cord (Maggi, Reference Maggi1991; Konturek et al. Reference Konturek, Konturek, Pawlik and Brzozowski2004). Some signals from the gut are transmitted from the NTS to higher neural centres, such as the paraventricular (PVN) and arcuate (ARC) nuclei of the hypothalamus (Berthoud et al. Reference Berthoud, Jedrzejewska and Powley1990), the bed nucleus of the stria terminallis and the ventral thalamus. The integration of all these afferent signals related to food presence in the gut regulates the size of individual meals (Flier, Reference Flier2004; Konturek et al. Reference Konturek, Konturek, Pawlik and Brzozowski2004; Badman & Flier, Reference Badman and Flier2005).

Gut hormones

Cholecystokinin

Despite the profuse development of the enteric nervous system, the main route of communication between the brain and the gut in relation to energy homeostasis is via the circulation. One of the hormones first identified in regulating energy homeostasis was the gastrointestinal hormone cholecystokinin (CCK), which is secreted by I cells in the duodenum and the jejunum into the circulation in response to nutrient ingestion (protein and fatty acids; Larsson & Rehfeld, Reference Larsson and Rehfeld1978; Bray, Reference Bray2000; Halford & Blundell, Reference Halford and Blundell2000a; Badman & Flier, Reference Badman and Flier2005; Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005; Perez-Tilve et al. Reference Perez-Tilve, Nogueiras, Mallo, Benoit and Tschoep2006). CKK exists in several molecular forms, the major forms in the plasma being CCK-8, -33 and -39 (Halford & Blundell, Reference Halford and Blundell2000a; Konturek et al. Reference Konturek, Konturek, Pawlik and Brzozowski2004; Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005). Once secreted, CCK reduces meal size and duration in both man and animals (Gibbs et al. Reference Gibbs, Young and Smith1973; Kissileff et al. Reference Kissileff, Pi-Sunyer, Thornton and Smith1981; Smith et al. Reference Smith, Gibbs, Jerome, Pi-Sunyer, Kissileff and Thornton1981a; Pi-Sunyer et al. Reference Pi-Sunyer, Kissileff, Thornton and Smith1982; Muurahainen et al. Reference Muurahainen, Kissileff, DeRogatis and Pi-Sunyer1988) and infusion of a CCK antagonist increases energy intake in human subjects (Beglinger et al. Reference Beglinger, Degen, Matzinger, D'Amato and Drewe2001). However, despite its anorectic actions, repeated administration of CKK does not influence body weight because although meal frequency is increased, there is no overall change in feeding (West et al. Reference West, Fey and Woods1984; Wei & Mojsov, Reference Wei and Mojsov1995). Thus, CCK is mostly involved in the short-term control of food intake, together with distension of the upper gastrointestinal tract (Konturek et al. Reference Konturek, Konturek, Pawlik and Brzozowski2004; Badman & Flier, Reference Badman and Flier2005).

CCK signals via two distinct G-protein-coupled receptors termed CCKA and CCKB (Wank et al. Reference Wank, Harkins, Jensen, Shapira, de Weerth and Slattery1992a; Halford & Blundell, Reference Halford and Blundell2000a). Both receptors are widely expressed in the CNS and in the periphery (Moran et al. Reference Moran, Robinson, Goldrich and McHugh1986; Moran et al. Reference Moran, Norgren, Crosby and McHugh1990; Wank et al. Reference Wank, Harkins, Jensen, Shapira, de Weerth and Slattery1992a,Reference Wank, Pisegna and de Weerthb). The effect of CCK on food intake is mediated via CCKA (Asin et al. Reference Asin, Gore, Bednarz, Holladay and Nadzan1992; Halford & Blundell, Reference Halford and Blundell2000a). CKK crosses the brain–blood barrier (BBB; Reidelberger et al. Reference Reidelberger, Hernandez, Fritzsch and Hulce2004) and acts on neuropeptide Y (NPY) neurons in the dorsomedial nucleus of the hypothalamus (DMH), as well as the NST in the brainstem (Moran et al. Reference Moran, Baldessarini, Salorio, Lowery and Schwartz1997; Bi et al. Reference Bi, Ladenheim, Schwartz and Moran2001). The effects of CKK on feeding are also mediated through paracrine and neuroendocrine activation of vagal fibres (Reidelberger & Solomon, Reference Reidelberger and Solomon1986; Schwartz & Moran, Reference Schwartz and Moran1994; Moran et al. Reference Moran, Baldessarini, Salorio, Lowery and Schwartz1997).

Glucagon-like peptide-1 and oxyntomodulin

The preproglucagon gene product yields two important satiety peptides, glucagon-like peptide (GLP)-1 and oxyntomodulin (OXM; Tang-Christensen et al. Reference Tang-Christensen, Vrang and Larsen2001; Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005). The preproglucagon gene is widely expressed in the gut, the pancreas and the NTS in the brainstem. Tissue-specific processing of preproglucagon by prohormone convertases 1 and 2 produces different products: glucagon is the main product in the pancreas; GLP-1 and -2 and OXM are the major products in CNS and gut (Tang-Christensen et al. Reference Tang-Christensen, Vrang and Larsen2001; Badman & Flier, Reference Badman and Flier2005; Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005).

GLP-1 and OXM are released from L cells in response to NEFA and carbohydrates (Ghatei et al. Reference Ghatei, Uttenthal, Christofides, Bryant and Bloom1983; Le Quellec et al. Reference Le Quellec, Kervran, Blache, Ciurana and Bataille1992; Herrmann et al. Reference Herrmann, Goke, Richter, Fehmann, Arnold and Goke1995; Hirasawa et al. Reference Hirasawa, Tsumaya, Awaji, Katsuma, Adachi, Yamada, Sugimoto, Miyazaki and Tsujimoto2005). Both peptides inhibit feeding when they are centrally or peripherally administrated (Turton et al. Reference Turton, O'Shea, Gunn, Beak, Edwards and Meeran1996; Dakin et al. Reference Dakin, Small, Batterham, Neary, Cohen, Patterson, Ghatei and Bloom2004), and chronic administration of GLP-1 and OXM decreases weight gain and adiposity in rodents (Meeran et al. Reference Meeran, O'Shea, Edwards, Turton, Heath and Gunn1999; Dakin et al. Reference Dakin, Small, Batterham, Neary, Cohen, Patterson, Ghatei and Bloom2004). The actions of both GLP-1 and OXM on feeding may be mediated via the GLP-1 receptor, which is expressed in the hypothalamus, brainstem and periphery (Uttenthal et al. Reference Uttenthal, Toledano and Blazquez1992; Wei & Mojsov, Reference Wei and Mojsov1995; Shughrue et al. Reference Shughrue, Lane and Merchenthaler1996; Bullock et al. Reference Bullock, Heller and Habener1996). The anorectic effect of GLP-1 and OXM is also present in man (Flint et al. Reference Flint, Raben, Astrup and Holst1998, Reference Flint, Raben, Rehfeld, Holst and Astrup2000, Reference Flint, Raben, Ersboll, Holst and Astrup2001; Gutzwiller et al. Reference Gutzwiller, Goke, Drewe, Hildebrand, Ketterer, Handschin, Winterhalder, Conen and Beglinger1999; Naslund et al. Reference Naslund, Barkeling, King, Gutniak, Blundell, Holst, Rossner and Hellstrom1999; Verdich et al. Reference Verdich, Flint, Gutzwiller, Naslund, Beglinger, Hellstrom, Long, Morgan, Holst and Astrup2001; Meier et al. Reference Meier, Gallwitz, Schmidt and Nauck2002). Despite this evidence, it has been reported that some of the anorectic effects of GLP-1 may be related to taste aversion and visceral illness (Shughrue et al. Reference Shughrue, Lane and Merchenthaler1996; Bullock et al. Reference Bullock, Heller and Habener1996; Thiele et al. Reference Thiele, van Dijk, Campfield, Smith, Burn, Woods, Bernstein and Seeley1997; Yamamoto et al. Reference Yamamoto, Lee, Marcus, Williams, Overton, Lopez, Hollenberg, Baggio, Saper, Drucker and Elmquist2002). Regardless of the anorectic actions of GLP-1 and OXM reported in rodents, GLP-1 receptor-knock-out mice have normal feeding behaviour (Scrocchi et al. Reference Scrocchi, Brown, Maclusky, Brubaker, Auerbach, Joyner and Drucker1996, Reference Scrocchi, Brown and Drucker1997, Reference Scrocchi, Hill, Saleh, Perkins and Drucker2000).

Preproglucagon also yields GLP-2. The role of GLP-2 has not been fully established; however, central administration reduces feeding, probably via GLP-1 receptor (Badman & Flier, Reference Badman and Flier2005). No effect of GLP-2 on feeding has been reported in man (Schmidt et al. Reference Schmidt, Naslund, Gryback, Jacobsson, Hartmann, Holst and Hellstrom2003).

Peptide YY

Peptide YY (PYY) is secreted postprandially by the L cells of the gastrointestinal tract, especially in the most distal portions such as the ileum, colon and rectum; PYY secretion is correlated with energy intake (Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005). There are two main forms of PYY in the circulation: PYY1–36; PYY3–36 (Grandt et al. Reference Grandt, Schimiczek, Beglinger, Layer, Goebell, Eysselein and Reeve1994; Batterham et al. Reference Batterham, Cowley, Small, Herzog, Cohen and Dakin2002; Wynne et al. Reference Wynne, Stanley and Bloom2004; Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005). Peripheral administration of PYY has several actions, including delaying gastric emptying and gastric secretion, and increasing ileum absorption. It has also been reported that peripheral administration of PYY3–36 inhibits food intake and reduces weight gain in rodents, primates and man (Batterham et al. Reference Batterham, Cowley, Small, Herzog, Cohen and Dakin2002; Challis et al. Reference Challis, Pinnock, Coll, Carter, Dickson and O'Rahilly2003; Moran et al. Reference Moran, Smedh, Kinzig, Scott, Knipp and Ladenheim2005). PYY crosses the BBB and probably exerts its actions via the presynaptic Y2 receptor of NPY neurons in the ARC, releasing inhibition of pro-opiomelanocortin (POMC) neurons and consequently inhibiting feeding (Broberger et al. Reference Broberger, Landry, Wong, Walsh and Hokfelt1997; Batterham et al. Reference Batterham, Cowley, Small, Herzog, Cohen and Dakin2002; Challis et al. Reference Challis, Pinnock, Coll, Carter, Dickson and O'Rahilly2003).

Despite this evidence, the anorectic effect of PYY3–36 is controversial and not easily duplicated (Tschop et al. Reference Tschop, Castaneda, Joost, Thone-Reineke, Ortmann and Klaus2004; Coll et al. Reference Coll, Challis and O'Rahilly2004a). Indeed, in contrast to peripheral injection central administration of both PYY1–36 and PYY3–36 stimulates feeding in rodents (Stanley et al. Reference Stanley, Daniel, Chin and Leibowitz1985; Clark et al. Reference Clark, Sahu, Kalra, Balasubramaniam and Kalra1987; Hagan et al. Reference Hagan, Castaneda, Sumaya, Fleming, Galloway and Moss1998; Corpa et al. Reference Corpa, McQuade, Krasnicki and Conze2001). It has also been suggested that the anorexic effect of PYY may be partially mediated by an aversive response (Halatchev & Cone, Reference Halatchev and Cone2005).

Bombesin

Bombesin is a peptide that is widely distributed in the mammalian gut. Plasma levels of bombesin increase markedly after food intake (Gibbs et al. Reference Gibbs, Fauser, Rowe, Rolls, Rolls and Maddison1979; Wynne et al. Reference Wynne, Stanley and Bloom2004), and peripheral and central administration of bombesin is anorectic (Gibbs et al. Reference Gibbs, Fauser, Rowe, Rolls, Rolls and Maddison1979; Smith et al. Reference Smith, Jerome and Gibbs1981b). Bombesin is structurally very similar to gastrin-releasing peptide and neuromedin B, and binds to their receptors. Additionally, a bombesin-3 receptor has been cloned (Ladenheim et al. Reference Ladenheim, Moore, Salorio, Mantey, Taylor, Coy, Jensen and Moran1997). Knocking out bombesin-3 receptor induces moderate hyperphagia, obesity and metabolic alterations in mice (Ohki-Hamazaki et al. Reference Ohki-Hamazaki, Watase, Yamamoto, Ogura, Yamano, Yamada, Maeno, Imaki, Kikuyama, Wada and Wada1997).

Gastric inhibitory polypeptide

Gastric inhibitory polypeptide is secreted from the duodenal K cells, predominantly in response to ingested fat. Mice fed a high-fat diet have increased levels of gastric inhibitory polypeptide together with obesity (Wynne et al. Reference Wynne, Stanley and Bloom2004; Badman & Flier, Reference Badman and Flier2005), whereas mice lacking the gastric inhibitory polypeptide receptor are protected against obesity induced by both a high-fat diet and leptin deficiency (ob/ob mice; Miyawaki et al. Reference Miyawaki, Yamada, Ban, Ihara, Tsukiyama and Zhou2002). Thus, gastric inhibitory polypeptide may be involved in the development of obesity in response to high fat intake.

Ghrelin

Ghrelin is a twenty-eight-amino acid acylated hormone mainly synthesised and secreted by the gut in the gastric oxyntic cells (A/X cells) at the fundus of the stomach, as well as the duodenum, ileum, caecum and colon (Kojima et al. Reference Kojima, Hosoda, Date, Nakazato, Matsuo and Kangawa1999; Date et al. Reference Date, Kojima, Hosoda, Sawaguchi, Mondal, Suganuma, Matsukura, Kangawa and Nakazato2000; Gualillo et al. Reference Gualillo, Caminos, Blanco, García-Caballero, Kojima, Kangawa, Diéguez and Casanueva2001; Sakata et al. Reference Sakata, Nakamura, Yamazaki, Matsubara, Hayashi, Kangawa and Sakai2002). Ghrelin expression has also been detected in other tissues, such as the hypothalamus (Kojima et al. Reference Kojima, Hosoda, Date, Nakazato, Matsuo and Kangawa1999; Horvath et al. Reference Horvath, Diano, Sotonyi, Heiman and Tschop2001; Cowley et al. Reference Cowley, Smith, Diano, Tschop, Pronchuk and Grove2003) testis (Barreiro et al. Reference Barreiro, Gaytan, Caminos, Pinilla, Casanueva, Aguilar, Dieguez and Tena-Sempere2002; Tena-Sempere et al. Reference Tena-Sempere, Barreiro, Gonzalez, Gaytan, Zhang, Caminos, Pinilla, Casanueva, Dieguez and Aguilar2002), pituitary (Caminos et al. Reference Caminos, Nogueiras, Blanco, Seoane, Bravo, Alvarez, Garcia-Caballero, Casanueva and Dieguez2003a), ovary (Caminos et al. Reference Caminos, Tena-Sempere, Gaytan, Sanchez-Criado, Barreiro, Nogueiras, Casanueva, Aguilar and Dieguez2003b; Gaytan et al. Reference Gaytan, Barreiro, Chopin, Herington, Morales, Pinilla, Casanueva, Aguilar, Dieguez and Tena-Sempere2003), heart (Iglesias et al. Reference Iglesias, Pineiro, Blanco, Gallego, Dieguez, Gualillo, Gonzalez-Juanatey and Lago2004) and placenta (Gualillo et al. Reference Gualillo, Caminos, Blanco, García-Caballero, Kojima, Kangawa, Diéguez and Casanueva2001).

Ghrelin, which was initially identified as the endogenous ligand of the growth hormone secretagogue receptor (GHS-R), exerts a potent and specific growth hormone-releasing activity in vitro (Kojima et al. Reference Kojima, Hosoda, Date, Nakazato, Matsuo and Kangawa1999) and in vivo (Arvat et al. Reference Arvat, Di Vito, Broglio, Papotti, Muccioli, Dieguez, Casanueva, Deghenghi, Camanni and Ghigo2000; Seoane et al. Reference Seoane, Tovar, Baldelli, Arvat, Ghigo, Casanueva and Diéguez2000), as well as increasing the transcription rate of the Pit-1 gene (García et al. Reference García, Alvarez, Smith and Diéguez2001). Further studies have led to the recognition that ghrelin also plays an important role in energy homeostasis. Ghrelin administration induces positive energy balance in rodents by decreasing fat utilization without markedly changing energy expenditure or locomotor activity (Wren et al. Reference Wren, Small, Ward, Murphy, Dakin, Taheri, Kennedy, Roberts, Morgan, Ghatei and Bloom2000; Nakazato et al. Reference Nakazato, Murakami, Date, Kojima, Matsuo, Kangawa and Matsukura2001). Furthermore, peripheral and central administration of ghrelin to rodents increases feeding, as well as fat mass, and reduces fat utilization (Tschop et al. Reference Tschop, Smiley and Heiman2000; Nakazato et al. Reference Nakazato, Murakami, Date, Kojima, Matsuo, Kangawa and Matsukura2001; Wren et al. Reference Wren, Small, Abbott, Dhillo, Seal, Cohen, Batterham, Taheri, Stanley, Ghatei and Bloom2001b; Seoane et al. Reference Seoane, López, Tovar, Casanueva, Señarís and Diéguez2003). Plasma levels of ghrelin are regulated by food intake, rising during fasting and immediately before meals, and falling after food intake (Ariyasu et al. Reference Ariyasu, Takaya, Tagami, Ogawa, Hosoda and Akamizu2001; Cummings et al. Reference Cummings, Purnell, Frayo, Schmidova, Wisse and Weigle2001; Tschop et al. Reference Tschop, Wawarta, Riepl, Friedrich, Bidlingmaier, Landgraf and Folwaczny2001a). These changes in ghrelin expression are directly modulated by energy intake and nutritional signals such as blood glucose and ingestion of fat or carbohydrate (Tschop et al. Reference Tschop, Smiley and Heiman2000; Sakata et al. Reference Sakata, Nakamura, Yamazaki, Matsubara, Hayashi, Kangawa and Sakai2002). For this reason a physiological role of ghrelin in meal initiation has been proposed (Cummings et al. Reference Cummings, Purnell, Frayo, Schmidova, Wisse and Weigle2001; Cummings & Shannon, Reference Cummings and Shannon2003). This suggestion is supported by experiments (Nakazato et al. Reference Nakazato, Murakami, Date, Kojima, Matsuo, Kangawa and Matsukura2001) that use anti-ghrelin antibodies to block the actions of ghrelin, which results in an attenuation of fasting-induced refeeding.

The effects of ghrelin on feeding and growth hormone secretion are mediated via type 1a GHS-R (Kojima et al. Reference Kojima, Hosoda, Date, Nakazato, Matsuo and Kangawa1999; Tschop et al. Reference Tschop, Smiley and Heiman2000; Chen et al. Reference Chen, Trumbauer, Chen, Weingarth, Adams and Frazier2004; Sun et al. Reference Sun, Wang, Zheng and Smith2004). However, the orexigenic effects of ghrelin are independent of growth hormone-releasing properties (Tschop et al. Reference Tschop, Smiley and Heiman2000; Shintani et al. Reference Shintani, Ogawa, Ebihara, Aizawa-Abe, Miyanaga and Takaya2001; Tamura et al. Reference Tamura, Kamegai, Shimizu, Ishii, Sugihara and Oikawa2002). The expression of GHS-R in the hypothalamus is nutritionally regulated in a nucleus-specific manner, with fasting increasing the mRNA levels of GHS-R in the ARC but not in the ventromedial nucleus of the hypothalamus (VMH). Additionally, the level of GHS-R expression in the ARC, but not in the VMH, is reduced by leptin and increased by ghrelin in a growth hormone-dependent fashion (Fig. 1; Nogueiras et al. Reference Nogueiras, Tovar, Mitchell, Rayner, Archer, Diéguez and Williams2004b).

Fig. 1. Regulation of growth hormone secretogogue receptor (GHS-R) in the rat hypothalamus by leptin and ghrelin. Effects of intracerebroventricular leptin (■; a, b) and ghrelin (; c, d) on GHS-R expression in the arcuate nucleus (a, c) and the ventromedial nucleus of the hypothalamus (b, d) in fed and 48 h-fasted rats. (□), Vehicle. Values are means with their standard errors represented by vertical bars. Mean values were significantly different from the corresponding values for fed rats: *P<0·05. Mean values were significantly different from the corresponding values for vehicle-fed rats: †P<0·05, ††P<0·01, †††P<0·001.

Ghrelin is also important in the regulation of energy homeostasis in man. Intravenous administration of ghrelin to healthy volunteers increases food intake (Wren et al. Reference Wren, Seal, Cohen, Brynes, Frost, Murphy, Dhillo, Ghatei and Bloom2001a). Moreover, the rise in preprandial ghrelin correlates with hunger scores in human subjects eating spontaneously (Cummings et al. Reference Cummings, Frayo, Marmonier, Aubert and Chapelot2004). Interestingly, the levels of ghrelin are correlated with adiposity in man, with an inverse relationship between plasma ghrelin levels and BMI (Tschop et al. Reference Tschop, Weyer, Tataranni, Devanarayan, Ravussin and Heiman2001b). Obese human subjects show reduced levels of plasma ghrelin, which rise to normal after diet-induced weight loss (Hansen et al. Reference Hansen, Dall, Hosoda, Kojima, Kangawa, Christiansen and Jorgensen2002; Cummings et al. Reference Cummings, Weigle, Frayo, Breen, Ma, Dellinger and Purnell2002b). Moreover, in obese individuals the postprandial regulation of ghrelin seems to be altered, which may be related to continuous food intake and/or obesity (English et al. Reference English, Ghatei, Malik, Bloom and Wilding2002). Finally, the severe hyperphagia seen in patients with Prader-Willi syndrome is associated with elevated ghrelin levels, in contrast to other forms of obesity in which ghrelin levels are low (Cummings et al. Reference Cummings, Clement, Purnell, Vaisse, Foster, Frayo, Schwartz, Basdevant and Weigle2002a).

In the CNS the action of ghrelin on feeding is mainly exerted via the ARC. GHS-R mRNA is expressed in neurons in the ARC co-expressing NPY and agoutirelated peptide (AgRP; Guan et al. Reference Guan, Yu, Palyha, McKee, Feighner, Sirinathsinghji, Smith, Van Der Ploeg and Howard1997; Tannenbaum et al. Reference Tannenbaum, Lapointe, Beaudet and Howard1998; Willesen et al. Reference Willesen, Kristensen and Romer1999; Zigman et al. Reference Zigman, Jones, Lee, Saper and Elmquist2006), and the central administration of ghrelin increases the mRNA content of NPY and AgRP in the ARC in fed and fasting conditions (Kamegai et al. Reference Kamegai, Tamura, Shimizu, Ishii, Sugihara and Wakabayashi2001; Nakazato et al. Reference Nakazato, Murakami, Date, Kojima, Matsuo, Kangawa and Matsukura2001; Seoane et al. Reference Seoane, López, Tovar, Casanueva, Señarís and Diéguez2003). There is also some evidence that orexin (also termed hypocretin; OX) neurons in the lateral hypothalamic area (LHA; Lawrence et al. Reference Lawrence, Snape, Baudoin and Luckman2002; Toshinai et al. Reference Toshinai, Date, Murakami, Shimada, Mondal and Shimbara2003) and neurons in the NTS and the area postrema in the brainstem may mediate the orexigenic actions of ghrelin (Nakazato et al. Reference Nakazato, Murakami, Date, Kojima, Matsuo, Kangawa and Matsukura2001; Lawrence et al. Reference Lawrence, Snape, Baudoin and Luckman2002). Recent data also indicate that ghrelin acts in the hypothalamus by altering fatty acid metabolism and AMP-activated protein kinase. It has been demonstrated that ghrelin increases hypothalamic AMP-activated protein kinase phosphorylation levels, activating it. This action may be associated to specific changes in hypothalamic neuropeptides, although the exact molecular mechanisms and anatomical details of this interaction have not been fully identified (Andersson et al. Reference Andersson, Filipsson, Abbott, Woods, Smith, Bloom, Carling and Small2004; Kola et al. Reference Kola, Hubina, Tucci, Kirkham, Garcia, Mitchell, Williams, Hawley, Hardie, Grossman and Korbonits2005).

Despite ghrelin having a potent action in regulating food intake, both ghrelin-knock-out mice and mice lacking GHS-R type 1a have normal feeding patterns and body composition on a standard diet (Sun et al. Reference Sun, Ahmed and Smith2003). However, on a high-fat diet the absence of ghrelin (Wortley et al. Reference Wortley, Del Rincon, Murray, García, Iida, Thorner and Sleeman2005) or the ghrelin receptor (Zigman et al. Reference Zigman, Nakano, Coppari, Balthasar, Marcus and Lee2005) protects against early-onset obesity; in both cases this reduced weight gain is associated with decreased adiposity and increased energy expenditure and locomotor activity. These data suggest that ghrelin, like leptin, may play an important role in the development of hypothalamic systems regulating energy balance (Grove & Cowley, Reference Grove and Cowley2005). Very interestingly, elimination of ghrelin improves the diabetic phenotype but not obese phenotype of ob/ob mice (Sun et al. Reference Sun, Asnicar, Saha, Chan and Smith2006).

Finally, it has recently been reported that obestatin, a new peptide derived from the ghrelin gene, inhibits food intake by acting through the orphan receptor GPR39 (Nogueiras & Tschop, Reference Nogueiras and Tschop2005; Zhang et al. Reference Zhang, Ren, Avsian-Kretchmer, Luo, Rauch, Klein and Hsueh2005). Despite this evidence there are some discrepancies in relation to the anorectic effect of obestatin (Nogueiras et al. Reference Nogueiras, Pfluger, Tovar, Myrtha, Mitchell and Morris2006) as well as its binding to GPR39 (Holst et al. Reference Holst, Egerod, Schild, Vickers, Cheetham, Gerlach, Storjohann, Stidsen, Jones, Beck-Sickinger and Schwartz2006). If the anorectic effect is confirmed, this finding could provide a new drug target for the treatment of obesity.

Adipose tissue hormones

Originally thought of as an inert tissue involved in the storage of energy, it is now clear that adipose tissue is an active endocrine organ (Casanueva & Diéguez, Reference Casanueva and Diéguez1999; Ahima & Flier, Reference Ahima and Flier2000a). Adipocyte hormones regulate appetite, glucose homeostasis, lipid metabolism, endocrine function, cardiovascular physiology, reproduction, immune function and development, amongst other functions (Casanueva & Diéguez, Reference Casanueva and Diéguez1999; Ahima & Flier, Reference Ahima and Flier2000a; Pinto et al. Reference Pinto, Roseberry, Liu, Diano, Shanabrough, Cai, Friedman and Horvath2004; Horvath & Diano, Reference Horvath and Diano2004).

Leptin

Among the adipocyte hormones, the one that has most changed the concept of white adipose tissue as an inert tissue is leptin, the product of the ob (obese) gene (Zhang et al. Reference Zhang, Proenca, Maffei, Barone, Leopold and Friedman1994). Leptin is expressed principally in adipocytes (Zhang et al. Reference Zhang, Proenca, Maffei, Barone, Leopold and Friedman1994), but also at lower levels in gastrointestinal tract (Bado et al. Reference Bado, Levasseur, Attoub, Kermorgant, Laigneau, Bortoluzzi, Moizo, Lehy, Guerre-Millo, Marchand-Brustel and Lewin1998) and placenta (Señarís et al. Reference Señarís, García-Caballero, Casabiell, Gallego, Castro, Considine, Diéguez and Casanueva1997; Masuzaki et al. Reference Masuzaki, Ogawa, Sagawa, Hosoda, Matsumoto, Mise, Nishimura, Yoshimasa, Tanaka, Mori and Nakao1997). Plasma leptin levels reflect both energy stores and acute energy balance. Circulating leptin levels are tightly correlated with adipose tissue mass (Maffei et al. Reference Maffei, Halaas, Ravussin, Pratley, Lee and Zhang1995), and food restriction results in suppression of circulating leptin (Frederich et al. Reference Frederich, Lollmann, Hamann, Napolitano-Rosen, Kahn, Lowell and Flier1995; Maffei et al. Reference Maffei, Halaas, Ravussin, Pratley, Lee and Zhang1995), which can be reversed by refeeding or insulin administration. Peripheral and central leptin administration reduces spontaneous and fasting-induced hyperphagia (Ahima et al. Reference Ahima, Prabakaran, Mantzoros, Qu, Lowell, Maratos-Flier and Flier1996; Ahima, Reference Ahima2000), and chronic peripheral administration reduces feeding, resulting in loss of fat mass and body weight (Halaas et al. Reference Halaas, Gajiwala, Maffei, Cohen, Chait, Rabinowitz, Lallone, Burley and Friedman1995).

The complete lack of leptin seen in the ob/ob mouse has profound consequences on body-weight homeostasis, leading to hyperphagia and obesity, as well as neuroendocrine and immune dysregulation, which is normalised by leptin administration (Campfield et al. Reference Campfield, Smith, Guisez, Devos and Burn1995; Halaas et al. Reference Halaas, Gajiwala, Maffei, Cohen, Chait, Rabinowitz, Lallone, Burley and Friedman1995; Pelleymounter et al. Reference Pelleymounter, Cullen, Baker, Hecht, Winters, Boone and Collins1995). In man leptin deficiency causes morbid obesity and hypogonadism (Montague et al. Reference Montague, Farooqi, Whitehead, Soos, Rau and Wareham1997; Strobel et al. Reference Strobel, Issad, Camoin, Ozata and Strosberg1998), which can be improved by recombinant leptin (Farooqi et al. Reference Farooqi, Jebb, Langmack, Lawrence, Cheetham, Prentice, Hughes, McCamish and O'Rahilly1999; Licinio et al. Reference Licinio, Caglayan, Ozata, Yildiz, de Miranda and O'Kirwan2004). In the same way, defective leptin receptor signalling also has a profound impact on body weight and endocrine function. A point mutation in the intracellular domain of the long isoform of the leptin receptor (OB-Rb) gene, which prevents signalling, results in obesity in db/db mice (Chen et al. Reference Chen, Charlat, Tartaglia, Woolf, Weng and Ellis1996; Lee et al. Reference Lee, Proenca, Montez, Carroll, Darvishzadeh, Lee and Friedman1996). Defects in the human leptin receptor have also been reported; as with leptin deficiency, these subjects have hypogonadism and early-onset morbid obesity (Clement et al. Reference Clement, Vaisse, Lahlou, Cabrol, Pelloux and Cassuto1998, Reference Clement, Vega, Laville, Pelloux, Guy-Grand, Basdevant and Vidal2002).

Leptin binds and activates a receptor of the cytokine receptor family (Tartaglia et al. Reference Tartaglia, Dembski, Weng, Deng, Culpepper and Devos1995). Alternative mRNA splicing and post-translational processing results in several isoforms of the receptor (OB-Ra, OB-Rb, OB-Rc, OB-Re and OB-Rf; Tartaglia, Reference Tartaglia1997; Chua et al. Reference Chua, Koutras, Han, Liu, Kay, Young, Chung and Leibel1997; Ahima & Flier, Reference Ahima and Flier2000b). OB-Rb is the variant implicated in signal transduction (Tartaglia, Reference Tartaglia1997; Ahima & Flier, Reference Ahima and Flier2000b). The other isoforms may act as leptin sequesters and transporters, binding leptin without signal transduction (Friedman & Halaas, Reference Friedman and Halaas1998; Ahima & Flier, Reference Ahima and Flier2000b). Ob-Rb possesses a long intracellular domain that binds to janus kinases (Lee et al. Reference Lee, Proenca, Montez, Carroll, Darvishzadeh, Lee and Friedman1996) and to signal transducers and activators of transcription-3 transcription factors (Vaisse et al. Reference Vaisse, Halaas, Horvath, Darnell, Stoffel and Friedman1996; Hakansson et al. Reference Hakansson, de Lecea, Sutcliffe, Yanagisawa and Meister1999), resulting in signal transduction and mediating the action of leptin on feeding (Lee et al. Reference Lee, Proenca, Montez, Carroll, Darvishzadeh, Lee and Friedman1996). Activation of the janus kinases/signal transducers and activators of transcription pathway induces expression of suppressor of cytokine signalling-3, a cytokine-inducible inhibitor of signalling; suppressor of cytokine signalling-3 expression is up regulated by leptin in hypothalamic nuclei expressing the Ob-Rb receptor (Ahima & Flier, Reference Ahima and Flier2000b; Howard et al. Reference Howard, Cave, Oksanen, Tzameli, Bjorbaek and Flier2004).

Plasma leptin crosses the BBB via a saturable process (Banks et al. Reference Banks, Kastin, Huang, Jaspan and Maness1996; Banks Reference Banks2001a,Reference Banksb), thought to be mediated by OB-Ra and OB-Rc (El Haschimi et al. Reference El Haschimi, Pierroz, Hileman, Bjorbaek and Flier2000; Ahima & Flier, Reference Ahima and Flier2000b). OB-Rb is widely expressed in the hypothalamus (being most abundant in the ARC, the VMH and the DMH) the LHA and the medial preoptic area (Fei et al. Reference Fei, Okano, Li, Lee, Zhao, Darnell and Friedman1997; Elmquist et al. Reference Elmquist, Bjorbaek, Ahima, Flier and Saper1998; Hakansson et al. Reference Hakansson, Brown, Ghilardi, Skoda and Meister1998, Reference Hakansson, de Lecea, Sutcliffe, Yanagisawa and Meister1999). OB-Rb is also expressed in feeding-modulating neurons in the brainstem (Elmquist et al. Reference Elmquist, Ahima, Maratos-Flier, Flier and Saper1997; Mercer et al. Reference Mercer, Moar and Hoggard1998). In the ARC OB-Rb mRNA is expressed by the two major neuronal groups: neurons co-expressing the orexigenic neuropeptides NPY and AgRP (Mercer et al. Reference Mercer, Hoggard, Williams, Lawrence, Hannah, Morgan and Trayhurn1996; Cheung et al. Reference Cheung, Clifton and Steiner1997); a distinct second population of neurons co-expressing the anorexigenic POMC and cocaine- and amphetamine-regulated transcript (CART). Leptin inhibits the activity of orexigenic AgRP/NPY neurons and reduces expression of AgRP and NPY (Stephens et al. Reference Stephens, Basinski, Bristow, Bue-Valleskey, Burgett and Craft1995; Hahn et al. Reference Hahn, Breininger, Baskin and Schwartz1998; Elias et al. Reference Elias, Aschkenasi, Lee, Kelly, Ahima, Bjorbaek, Flier, Saper and Elmquist1999), while activating anorectic CART/POMC neurons (Schwartz et al. Reference Schwartz, Seeley, Woods, Weigle, Campfield, Burn and Baskin1997; Kristensen et al. Reference Kristensen, Judge, Thim, Ribel, Christjansen and Wulff1998; Swart et al. Reference Swart, Jahng, Overton and Houpt2002). In the LHA leptin receptor is expressed in neurons expressing the orexigenic neuropeptides melanin-concentrating hormone (MCH) and the OX, which are inhibited by leptin (Qu et al. Reference Qu, Ludwig, Gammeltoft, Piper, Pelleymounter, Cullen, Mathes, Przypek, Kanarek and Maratos-Flier1996; López et al. Reference López, Seoane, García, Lago, Casanueva, Senarís and Diéguez2000). When leptin levels are low, such as in food restriction and fasting, the expression of orexigenic neuropeptides is increased and orexigenic neurons are activated; in contrast, anorexigenic neuropeptides are decreased and anorexigenic neurons are inhibited. When plasma leptin levels are high, as in the satiated animal, the anorectic pathways are switched on and the orexigenic pathways are switched off (Friedman & Halaas, Reference Friedman and Halaas1998; Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999; Saper et al. Reference Saper, Chou and Elmquist2002; Flier, Reference Flier2004).

The role of leptin in human obesity is intriguing. As described earlier, while there are individuals with defects in leptin synthesis or leptin signalling, these cases are extremely rare. The majority of obese individuals are characterised by high levels of leptin (Maffei et al. Reference Maffei, Halaas, Ravussin, Pratley, Lee and Zhang1995; Considine et al. Reference Considine, Sinha, Heiman, Kriauciunas, Stephens, Nyce, Ohannesian, Marco, McKee and Bauer1996), suggesting leptin insensitivity or resistance; in fact, leptin administration to obese subjects has only a moderate effect on body weight (Heymsfield et al. Reference Heymsfield, Greenberg, Fujioka, Dixon, Kushner, Hunt, Lubina, Patane, Self, Hunt and McCamish1999; Fogteloo et al. Reference Fogteloo, Pijl, Frolich, McCamish and Meinders2003). In rodents diet-induced obesity has also been correlated with the development of leptin resistance (Van Heek et al. Reference Van Heek, Compton, France, Tedesco, Fawzi, Graziano, Sybertz, Strader and Davis1997; Levin & Dunn-Meynell, Reference Levin and Dunn-Meynell2002). Leptin resistance may develop via different mechanisms. Peripheral leptin resistance may be the result of impairment in the function of the saturable leptin transporters in the BBB (Burguera et al. Reference Burguera, Couce, Curran, Jensen, Lloyd, Cleary and Poduslo2000; Furuhata et al. Reference Furuhata, Kagaya, Hirabayashi, Ikeda, Chang, Nishihara and Takahashi2000; Levin et al. Reference Levin, Dunn-Meynell and Banks2004). Central leptin resistance may develop as a result of impaired leptin signalling via OB-Rb in the hypothalamus, which could be related to a decrease in OB-Rb expression (García et al. Reference García, Casanueva, Diéguez and Señarís2000; Seeber et al. Reference Seeber, Smith and Waddell2002; López et al. Reference López, Seoane, Tovar, García, Nogueiras, Diéguez and Señarís2005a), a defect in the intracellular signalling mechanism of the janus kinases/signal transducers and activators of transcription pathway or the over-expression of suppressor of cytokine signalling-3 (El Haschimi et al. Reference El Haschimi, Pierroz, Hileman, Bjorbaek and Flier2000; Howard et al. Reference Howard, Cave, Oksanen, Tzameli, Bjorbaek and Flier2004; Ladyman & Grattan, Reference Ladyman and Grattan2004; Levin et al. Reference Levin, Dunn-Meynell and Banks2004; Munzberg et al. Reference Munzberg, Flier and Bjorbaek2004; Munzberg & Myers, Reference Munzberg and Myers2005).

The role of leptin in the hypothalamus is not only associated with food-intake regulation. Leptin also contributes to the adaptation of the neuroendocrine axis to fasting (Ahima et al. Reference Ahima, Prabakaran, Mantzoros, Qu, Lowell, Maratos-Flier and Flier1996; Casanueva & Diéguez, Reference Casanueva and Diéguez1999). Additionally, leptin is a neurotrophic factor during the development of the hypothalamus, mediating neuronal plasticity (Bouret et al. Reference Bouret, Draper and Simerly2004a,Reference Bouret, Draper and Simerlyb; Bouret & Simerly, Reference Bouret and Simerly2004; Pinto et al. Reference Pinto, Roseberry, Liu, Diano, Shanabrough, Cai, Friedman and Horvath2004). The importance of this function of leptin in the context of obesity is still not clear, but it has been proposed that perturbations in perinatal nutrition that alter leptin levels may have long-term consequences for the formation and function of hypothalamic circuits regulating feeding and body weight in adulthood (López et al. Reference López, Seoane, Tovar, García, Nogueiras, Diéguez and Señarís2005a).

Adiponectin

Adiponectin, also termed adipocyte complement-related protein, apM1 or adipoQ, is a 244-amino acid protein secreted from adipose tissue (Hu et al. Reference Hu, Liang and Spiegelman1996; Berg et al. Reference Berg, Combs and Scherer2002; Tsao et al. Reference Tsao, Lodish and Fruebis2002), the placenta (Caminos et al. Reference Caminos, Nogueiras, Gallego, Bravo, Tovar, García-Caballero, Casanueva and Diéguez2005) and cardiomyocytes (Pineiro et al. Reference Pineiro, Iglesias, Gallego, Raghay, Eiras, Rubio, Dieguez, Gualillo, Gonzalez-Juanatey and Lago2005). Adiponectin has four domains: an amino-terminal signal sequence; a region without homology to other known proteins; a collagen-like region; a carboxy-terminal globular domain. The globular domain forms homotrimers, and additional interactions with collagenous segments cause the formation of higher-molecular-weight complexes (Pajvani et al. Reference Pajvani, Du, Combs, Berg, Rajala, Schulthess, Engel, Brownlee and Scherer2003).

Adiponectin is important in the regulation of energy homeostasis (Scherer et al. Reference Scherer, Williams, Fogliano, Baldini and Lodish1995). Plasma levels of adiponectin are inversely correlated with adiposity in several species, including man (Hu et al. Reference Hu, Liang and Spiegelman1996; Arita et al. Reference Arita, Kihara, Ouchi, Takahashi, Maeda and Miyagawa1999; Hotta et al. Reference Hotta, Funahashi, Bodkin, Ortmeyer, Arita, Hansen and Matsuzawa2001) Adiponectin is increased after food restriction in rodents (Berg et al. Reference Berg, Combs, Du, Brownlee and Scherer2001, Reference Benoit, Air, Coolen, Strauss, Jackman, Clegg, Seeley and Woods2002). Peripheral administration to rodents has been shown to attenuate body-weight gain, by increased O2 consumption, without affecting food intake (Berg et al. Reference Berg, Combs, Du, Brownlee and Scherer2001; Fruebis et al. Reference Fruebis, Tsao, Javorschi, Ebbets-Reed, Erickson, Yen, Bihain and Lodish2001; Yamauchi et al. Reference Yamauchi, Kamon, Ito, Tsuchida, Yokomizo and Kita2003). This effect on energy expenditure appears to be mediated by the hypothalamic melanocortin system, without affecting other neuropeptide systems regulated by leptin (Qi et al. Reference Qi, Takahashi, Hileman, Patel, Berg, Pajvani, Scherer and Ahima2004). Circulating adiponectin levels negatively correlate with insulin resistance (Hotta et al. Reference Hotta, Funahashi, Bodkin, Ortmeyer, Arita, Hansen and Matsuzawa2001), and treatment with adiponectin can reduce body-weight gain, increase insulin sensitivity and decrease lipid levels in rodents (Berg et al. Reference Berg, Combs, Du, Brownlee and Scherer2001; Yamauchi et al. Reference Yamauchi, Kamon, Waki, Terauchi, Kubota and Hara2001; Qi et al. Reference Qi, Takahashi, Hileman, Patel, Berg, Pajvani, Scherer and Ahima2004; Winzell et al. Reference Winzell, Nogueiras, Dieguez and Ahren2004). Adiponectin-knock-out mice have severe diet-induced insulin resistance (Maeda et al. Reference Maeda, Shimomura, Kishida, Nishizawa, Matsuda and Nagaretani2002). The mechanism by which adiponectin improves insulin resistance and glucose metabolism is not fully understood, but some of these effects may be mediated by activation of AMP-activated protein kinase (Yamauchi et al. Reference Yamauchi, Kamon, Minokoshi, Ito, Waki and Uchida2002).

Adiponectin binds and activates two known membrane receptors, adipoR1 and adipoR2 (Yamauchi et al. Reference Yamauchi, Kamon, Ito, Tsuchida, Yokomizo and Kita2003). AdipoR1 is highly expressed in skeletal muscle; it has a high affinity for the globular domain of adiponectin and low affinity for the full-length ligand. AdipoR2 is highly expressed in the liver and preferentially binds to the full-length ligand. Adiponectin receptors have also been detected in the hypothalamus (Qi et al. Reference Qi, Takahashi, Hileman, Patel, Berg, Pajvani, Scherer and Ahima2004) and the placenta (Caminos et al. Reference Caminos, Nogueiras, Gallego, Bravo, Tovar, García-Caballero, Casanueva and Diéguez2005). Very interestingly, it has recently been reported that adiponectin does not cross the BBB but modifies cytokine expression in the brain endothelial cells, making unlikely a direct effect of adiponectin in the CNS (Spranger et al. Reference Spranger, Verma, Gohring, Bobbert, Seifert, Sindler, Pfeiffer, Hileman, Tschop and Banks2006).

Resistin

Resistin is produced by adipose tissue and appears to be involved in the modulation of insulin sensitivity and adipocyte differentiation (Steppan et al. Reference Steppan, Bailey, Bhat, Brown, Banerjee, Wright, Patel, Ahima and Lazar2001a,Reference Steppan, Brown, Wright, Bhat, Banerjee, Dai, Enders, Silberg, Wen, Wu and Lazarb; Vidal-Puig & O'Rahilly, Reference Vidal-Puig and O'Rahilly2001; Steppan & Lazar, Reference Steppan and Lazar2002). In addition to adipose tissue, resistin is also expressed in the stomach, intestine, adrenal gland, testis and skeletal muscle (Nogueiras et al. Reference Nogueiras, Gallego, Gualillo, Caminos, Garcia-Caballero, Casanueva and Dieguez2003a,Reference Nogueiras, Gualillo, Caminos, Casanueva and Diéguezb, Reference Nogueiras, Barreiro, Caminos, Gaytan, Suominen and Navarro2004a).

Resistin expression is regulated in a tissue- and gender-specific manner. Food deprivation leads to a decrease in resistin mRNA expression only in adipose tissue (Nogueiras et al. Reference Nogueiras, Gallego, Gualillo, Caminos, Garcia-Caballero, Casanueva and Dieguez2003a,Reference Nogueiras, Gualillo, Caminos, Casanueva and Diéguezb). Circulating resistin is increased in obese rodents (Steppan et al. Reference Steppan, Bailey, Bhat, Brown, Banerjee, Wright, Patel, Ahima and Lazar2001a) and man (Savage et al. Reference Savage, Sewter, Klenk, Segal, Vidal-Puig, Considine and O'Rahilly2001) and falls after weight loss in man (Valsamakis et al. Reference Valsamakis, McTernan, Chetty, Al Daghri, Field, Hanif, Barnett and Kumar2004). Resistin-knock-out mice display increased glucose tolerance on a high-fat diet (Banerjee et al. Reference Banerjee, Rangwala, Shapiro, Rich, Rhoades and Qi2004; Sul, Reference Sul2004) and transgenic mice over-expressing a dominant negative form of resistin show increased adiposity with elevated plasma leptin and adiponectin levels, as well as enhanced glucose tolerance and insulin sensitivity (Sul, Reference Sul2004). All this evidence suggests that resistin may contribute to the development of insulin resistance and diabetes in obesity (Steppan et al. Reference Steppan, Bailey, Bhat, Brown, Banerjee, Wright, Patel, Ahima and Lazar2001a,Reference Steppan, Brown, Wright, Bhat, Banerjee, Dai, Enders, Silberg, Wen, Wu and Lazarb; Vidal-Puig & O'Rahilly, Reference Vidal-Puig and O'Rahilly2001; Steppan & Lazar, Reference Steppan and Lazar2002). In support of this role, recent evidence has shown that resistin inhibits feeding through a hypothalamic mechanism (Tovar et al. Reference Tovar, Nogueiras, Tung, Castaneda, Vázquez, Morris, Williams, Dickson and Diéguez2005). However, the molecular details of that action are not fully established.

IL-6 and IL-1

IL-6 is a multifunctional immune-modulating cytokine that has been suggested to have important functions in glucose and lipid metabolism. IL-6 is secreted from adipose tissue into the circulation, and its expression is positively correlated with BMI and total fat tissue. IL-6-knock-out mice develop obesity, which can partly be reversed by IL-6 replacement, suggesting a role for IL-6 in the long-term regulation of adipose tissue mass (Wallenius et al. Reference Wallenius, Wallenius, Ahren, Rudling, Carlsten, Dickson, Ohlsson and Jansson2002b). Furthermore, central administration of a low dose of IL-6 decreases feeding and increases energy expenditure in rats, suggesting a central site of action for IL-6 (Wallenius et al. Reference Wallenius, Wallenius, Sunter, Dickson and Jansson2002a). Supporting this hypothesis it has also been suggested that IL-6 and IL-6 receptors are expressed in the neurons in the VMH and the DMH (Schobitz et al. Reference Schobitz, De Kloet, Sutanto and Holsboer1993). IL-1 is also involved in body-weight homeostasis. IL-1 type I receptor-knock-out mice display an obese and insulin-resistant phenotype. This obese phenotype is characterised by a decrease in leptin sensitivity, fat utilization and locomotor activity (García et al. Reference García, Wernstedt, Berndtsson, Enge, Bell and Hultgren2006).

Pancreatic hormones

Insulin

Insulin is also an adiposity signal. Plasma insulin concentrations correlate with peripheral insulin sensitivity, which in turn is linked to total body fat depots and fat distribution, visceral fat being a key determinant (Schwartz et al. Reference Schwartz, Figlewicz, Baskin, Woods and Porte1992a, Reference Schwartz, Woods, Porte, Seeley and Baskin2000).

Insulin secretion by the pancreas increases rapidly after a meal, exerting an anorectic effect via the CNS (Schwartz et al. Reference Schwartz, Figlewicz, Baskin, Woods and Porte1992a, Reference Schwartz, Woods, Porte, Seeley and Baskin2000). Insulin enters the CNS via saturable receptor-mediated transport across the BBB (Woods et al. Reference Woods, Seeley, Baskin and Schwartz2003). Central administration of insulin or insulin mimetic reduces feeding and body weight in rodents and primates (Woods et al. Reference Woods, Lotter, McKay and Porte1979; Schwartz et al. Reference Schwartz, Figlewicz, Baskin, Woods and Porte1992a, Reference Schwartz, Woods, Porte, Seeley and Baskin2000; Air et al. Reference Air, Strowski, Benoit, Conarello, Salituro, Guan, Liu, Woods and Zhang2002). Administration of antisense RNA against the insulin receptor induces hyperphagia and increased fat mass (Obici et al. Reference Obici, Feng, Karkanias, Baskin and Rossetti2002a), and neuron-specific deletion of the insulin receptor results in obesity, hyperinsulinaemia and dyslipidaemia in mice (Bruning et al. Reference Bruning, Gautam, Burks, Gillette, Schubert, Orban, Klein, Krone, Muller-Wieland and Kahn2000). Insulin receptors are widespread in the brain and occur in hypothalamic nuclei involved in food intake (ARC, DMH, PVN and periventricular nucleus; Corp et al. Reference Corp, Woods, Porte, Dorsa, Figlewicz and Baskin1986; Marks et al. Reference Marks, Porte, Stahl and Baskin1990). In the hypothalamus the actions of insulin on food intake and body weight are mediated by NPY (Schwartz et al. Reference Schwartz, Sipols, Marks, Sanacora, White and Scheurink1992b) and the melanocortin system (Sipols et al. Reference Sipols, Baskin and Schwartz1995; Obici et al. Reference Obici, Feng, Tan, Liu, Karkanias and Rossetti2001; Benoit et al. Reference Benoit, Air, Coolen, Strauss, Jackman, Clegg, Seeley and Woods2002)

Pancreatic polypeptide

Pancreatic polypeptide (PP) belongs to the PP-fold family of peptides, which also includes PYY and NPY (Conlon, Reference Conlon2002). PP is mainly produced by peripheral cells of the islets of Langerhans, the exocrine pancreas and the distal gastrointestinal tract (Small & Bloom, Reference Small and Bloom2004; Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005). Plasma PP concentrations increase proportionally to energy intake (Small & Bloom, Reference Small and Bloom2004; Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005), and they appear to be inversely proportional to adiposity, with high levels in anorexic subjects and reduced levels in obese subjects (Lassmann et al. Reference Lassmann, Vague, Vialettes and Simon1980; Fujimoto et al. Reference Fujimoto, Inui, Kiyota, Seki, Koide, Takamiya, Uemoto, Nakajima, Baba and Kasuga1997).

Peripheral PP administration reduces feeding and body weight in obese rodents (Malaisse-Lagae et al. Reference Malaisse-Lagae, Carpentier, Patel, Malaisse and Orci1977) and feeding in man (Batterham et al. Reference Batterham, Le, Cohen, Park, Ellis, Patterson, Frost, Ghatei and Bloom2003). The anorectic effect of PP is exerted via brainstem pathways (in the area postrema), regulation of hypothalamic neuropeptides (NPY and OX) and modulation of ghrelin expression (Asakawa et al. Reference Asakawa, Inui, Yuzuriha, Ueno, Katsuura, Fujimiya, Fujino, Niijima, Meguid and Kasuga2003). The anorectic effect of PP is mediated by Y5 receptor. In contrast to the peripheral actions, central administration of PP increases food intake (Clark et al. Reference Clark, Kalra, Crowley and Kalra1984); the receptors mediating this action and the mechanisms involved are unclear.

Neural control of food intake

Hypothalamic regulation of food intake

The CNS receives information from the sensory experience of eating and also from the process of ingestion, absorption, metabolism and energy storage. The original theories explaining the central control of food intake were based on a ‘dual-centre hypothesis’ (Hecherington & Ranson, Reference Hecherington and Ranson1942; Anand & Brobeck, Reference Anand and Brobeck1951). In this model, based on hypothalamic-lesioning experiments, feeding is controlled by two hypothalamic areas: the lateral hypothalamic ‘feeding centres’; the ventromedial hypothalamic ‘satiety centres’. Lesions of the LHA decrease food intake and eventually lead to starvation and death. Conversely, lesions of several of the mediobasal hypothalamic nuclei result in obesity. Since then, knowledge concerning the hypothalamic regulation of feeding has increased; however, the main concept is the same, i.e. that anatomically-defined hypothalamic areas regulate food intake. These hypothalamic nuclei form interconnected neuronal circuits that respond to changes in energy status by altering the expression of specific neuropeptides, resulting in changes in energy intake and expenditure (Friedman & Halaas, Reference Friedman and Halaas1998; Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999; Schwartz et al. Reference Schwartz, Woods, Porte, Seeley and Baskin2000; Saper et al. Reference Saper, Chou and Elmquist2002; Flier, Reference Flier2004; Abizaid et al. Reference Abizaid, Gao and Horvath2006; Morton et al. Reference Morton, Cummings, Baskin, Barsh and Schwartz2006). Table 1 summarises some hypothalamic neuropeptides and neurotransmitters regulating food intake.

Table 1. Molecules with demonstrated orexigenic and/or anorexigenic effects in some animal models

CART, cocaine- and amphetamine-regulated transcript.

* The effect of somatostatin on food intake has been reported to be contradictory and very dependent of the doses used. Low doses increase feeding and high doses decrease feeding (Feifel & Vaccarino, Reference Feifel and Vaccarino1989, Reference Feifel and Vaccarino1990, Reference Feifel and Vaccarino1994).

Hypothalamic neuronal pathways regulating appetite

Arcuate nucleus

The ARC is considered as the ‘master hypothalamic centre’ for feeding control. It is situated around the base of the third ventricle and lies immediately above the median eminence. The ARC–median eminence is a circumventricular organ in which the BBB is modified, allowing the entry of peptides and proteins from the circulation, such as PYY, GLP-1, leptin and insulin (Banks et al. Reference Banks, Kastin, Huang, Jaspan and Maness1996; Kastin et al. Reference Kastin, Akerstrom and Pan2002; Nonaka et al. Reference Nonaka, Shioda, Niehoff and Banks2003; Woods et al. Reference Woods, Seeley, Baskin and Schwartz2003).

Two distinct neuronal populations in the ARC integrate peripheral nutritional and/or feeding signals. One set of neurons in the ventromedial part of the ARC express the orexigenic neuropeptides NPY and AgRP (Broberger et al. Reference Broberger, Johansen, Johansson, Schalling and Hokfelt1998b; Hahn et al. Reference Hahn, Breininger, Baskin and Schwartz1998). These neurons mostly project to the PVN. In the ventrolateral part of the ARC there is a second population of neurons that express the anorexigenic products of POMC, the precursor of α-MSH, and also CART (Elias et al. Reference Elias, Lee, Kelly, Aschkenasi, Ahima, Couceyro, Kuhar, Saper and Elmquist1998a; Kristensen et al. Reference Kristensen, Judge, Thim, Ribel, Christjansen and Wulff1998). This set of neurons projects more broadly within the CNS to hypothalamic nuclei such as the DMH, the LHA and the perifornical area, as well as the PVN. Thus, AgRP/NPY and CART/POMC neurons act as the primary hypothalamic site of action of peripheral hormones such as insulin and leptin. ARC neurons, in turn, project to secondary hypothalamic nuclei (‘second order neurons’) such as the PVN and the LHA. In these second-order neurons the release of neuropeptides is regulated to modulate energy intake (Schwartz et al. Reference Schwartz, Woods, Porte, Seeley and Baskin2000; Flier, Reference Flier2004).

Paraventricular nucleus

The PVN integrates neuropeptide signals from numerous CNS regions, including the ARC and brainstem (Sawchenko & Swanson, Reference Sawchenko and Swanson1983). Administration into the PVN of almost all of the known orexigenic and anorexigenic signalling molecules alters appetite (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999). Furthermore, CART/POMC neurons originating in the ARC potentiate inhibitory γ-aminobutyric acidergic signalling in the PVN and reduce feeding, while AgRP/NPY neurons inhibit this γ-aminobutyric acidergic signalling and stimulate food intake (Cowley et al. Reference Cowley, Pronchuk, Fan, Dinulescu, Colmers and Cone1999). Despite the large number of neuropeptides acting on the PVN, recent work suggests that they act to regulate feeding through a common mechanism involving AMP-activated protein kinase (Minokoshi et al. Reference Minokoshi, Alquier, Furukawa, Kim, Lee and Xue2004; López et al. Reference López, Lelliott, Tovar, Kimber, Gallego and Virtue2006).

The PVN also plays a major role in the integration of food intake and neuroendocrine function. AgRP/NPY and CART/POMC neurons in the ARC project to thyrotrophin-releasing hormone neurons in the PVN (Legradi & Lechan, Reference Legradi and Lechan1999; Fekete et al. Reference Fekete, Legradi, Mihaly, Huang, Tatro, Rand, Emerson and Lechan2000). AgRP/NPY inhibits pro-thyrotrophin-releasing hormone gene expression (Fekete et al. Reference Fekete, Sarkar, Rand, Harney, Emerson, Bianco and Lechan2002), while α-melanocyte-stimulating hormone stimulates pro-thyrotrophin-releasing hormone expression and inhibits the fasting-induced suppression of thyrotrophin-releasing hormone (Fekete et al. Reference Fekete, Legradi, Mihaly, Huang, Tatro, Rand, Emerson and Lechan2000). The PVH also contains corticotrophin-releasing hormone neurons, which form reciprocal circuits with NPY neurons in the ARC (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999).

Ventromedial nucleus of the hypothalamus

The VMH (as distinct from thalamic ventromedial nucleus) has long been considered as the ‘satiety centre’; since the finding that bilateral lesions in this nucleus induce hyperphagia and obesity (Hecherington & Ranson, Reference Hecherington and Ranson1942; Anand & Brobeck, Reference Anand and Brobeck1951). The VMH mainly receives projections from AgRP/NPY and CART/POMC neurons in the ARC. Additionally, the VMH neurons project their axons to the ARC, DMH and LHA, as well as brainstem regions such as the NTS (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999; Pinto et al. Reference Pinto, Roseberry, Liu, Diano, Shanabrough, Cai, Friedman and Horvath2004; Sternson et al. Reference Sternson, Shepherd and Friedman2005).

The VMH has been considered as a ‘reception nucleus’ for peripheral signals, as well as central signals. VMH neurons show a high abundance of leptin, ghrelin, oestrogen, thyroid hormone and neuropeptide receptors (Shughrue et al. Reference Shughrue, Lane and Merchenthaler1997; Roselli et al. Reference Roselli, Jorgensen, Doyle and Ronnekleiv1997; Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999; Nogueiras et al. Reference Nogueiras, Tovar, Mitchell, Rayner, Archer, Diéguez and Williams2004b; King, Reference King2006). However, despite the identification of these receptors, the molecular mechanisms regulating feeding in the VMH have not yet been well established. Some evidence suggests that brain-derived neurotrophic factor and steroidogenic factor-1 may play crucial roles in mediating body weight in this nucleus. Mice with reduced brain-derived neurotrophic factor receptor expression (Xu et al. Reference Xu, Goulding, Zang, Cepoi, Cone, Jones, Tecott and Reichardt2003), and with reduced brain-derived neurotrophic factor signalling (Xu et al. Reference Xu, Goulding, Zang, Cepoi, Cone, Jones, Tecott and Reichardt2003), and also steroidogenic factor-1-knock-out mice (Majdic et al. Reference Majdic, Young, Gomez-Sanchez, Anderson, Szczepaniak, Dobbins, McGarry and Parker2002), have increased body weight. Furthermore, activation of SF-1 neurons by leptin is required for normal body-weight homeostasis (Dhillon et al. Reference Dhillon, Zigman, Ye, Lee, McGovern and Tang2006). Finally, it has recently been reported that fatty acid synthase (FAS) and malonyl-CoA levels in this nucleus may play an important physiological role in the regulation of feeding (López et al. Reference López, Lelliott, Tovar, Kimber, Gallego and Virtue2006).

Dorsomedial nucleus of the hypothalamus

Like the VMH, the DMH has long been considered to be an integrative centre, processing information from other hypothalamic areas (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999). The DMH is located immediately dorsal to the VMH and has extensive direct connections with other hypothalamic nuclei (e.g. the PVN and the LHA), as well as the brainstem (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999; Bellinger & Bernardis, Reference Bellinger and Bernardis2002). Destruction of the DMH induces hyperphagia and obesity, although less dramatically than VMH lesions (Bellinger & Bernardis, Reference Bellinger and Bernardis2002). Very recent evidence has also demonstrated that the DMH is critical for the expression of food-entrainable circadian rhythms (Gooley et al. Reference Gooley, Schomer and Saper2006).

The DMH contains NPY-expressing cell bodies, which are involved in the hyperphagia observed in pregnant and lactating rats (Li et al. Reference Li, Chen and Smith1998; García et al. Reference García, López, Gualillo, Seoane, Diéguez and Señarís2003). Moreover, CART-expressing neurons are highly abundant in the DMH; the exact function of these cells is unknown, but they are probably involved in fasting-induced responses (Henry et al. Reference Henry, Rao, Ikenasio, Mountjoy, Tilbrook and Clarke2001).

Lateral hypothalamic area

Although the involvement of the LHA, including the perifornical area, in the regulation of feeding has been known for >60 years (Hecherington & Ranson, Reference Hecherington and Ranson1942; Anand & Brobeck, Reference Anand and Brobeck1951), the molecular mechanisms involved had remained unknown until 15 years ago when MCH was identified as the first orexigenic peptide exclusively expressed in this nucleus (Qu et al. Reference Qu, Ludwig, Gammeltoft, Piper, Pelleymounter, Cullen, Mathes, Przypek, Kanarek and Maratos-Flier1996). Other neuropeptides important in the regulation of feeding are also highly expressed in this area, such as galanin (Hakansson et al. Reference Hakansson, Brown, Ghilardi, Skoda and Meister1998), dynorphin (Chou et al. Reference Chou, Lee, Lu, Elmquist, Hara and Willie2001), CART-encoded peptides (Koylu et al. Reference Koylu, Couceyro, Lambert and Kuhar1998) and OX (de Lecea et al. Reference de Lecea, Kilduff, Peyron, Gao, Foye and Danielson1998; Sakurai et al. Reference Sakurai, Amemiya, Ishii, Matsuzaki, Chemelli and Tanaka1998; López et al. Reference López, Tena-Sempere, García-Caballero, Señarís, Diéguez, de Lecea and Sutcliffe2005b).

MCH and prepro-OX are each expressed by a different cell population, both of which receive projections from AgRP/NPY and CART/POMC neurons in the ARC (Broberger et al. Reference Broberger, de Lecea, Sutcliffe and Hokfelt1998a; Elias et al. Reference Elias, Saper, Maratos-Flier, Tritos, Lee and Kelly1998b; Horvath et al. Reference Horvath, Diano and van den Pol1999). Additionally, both sets of neurons express leptin receptors, indicating that their actions may be integrated (Hakansson et al. Reference Hakansson, Brown, Ghilardi, Skoda and Meister1998, Reference Hakansson, de Lecea, Sutcliffe, Yanagisawa and Meister1999). The LHA also contains a large number of glucose-sensing neurons (Bernardis & Bellinger, Reference Bernardis and Bellinger1996). Orexin neurons in the LHA respond to a fall in glucose levels with an increase in activity (Cai et al. Reference Cai, Widdowson, Harrold, Wilson, Buckingham, Arch, Tadayyon, Clapham, Wilding and Williams1999; Moriguchi et al. Reference Moriguchi, Sakurai, Nambu, Yanagisawa and Goto1999). LHA neurons project widely to a large number of extrahypothalamic areas. Major targets of the MCH and OX neurons include the brainstem motor systems, sympathetic and parasympathetic preganglionic nuclei in the medulla and spinal cord, the locus coeruleus, the medial raphe nucleus, the tuberomammillary nucleus and the cerebral cortex. All these areas are fundamental in different aspects of food intake regulation, from feeding-related behaviours to arousal and motor activity (Willie et al. Reference Willie, Chemelli, Sinton and Yanagisawa2001; López et al. Reference López, Diéguez, Señarís and Pandalai2001a, Reference López, Tena-Sempere, García-Caballero, Señarís, Diéguez, de Lecea and Sutcliffe2005b; Saper et al. Reference Saper, Chou and Elmquist2002; Steininger et al. Reference Steininger, Kilduff, Behan, Benca and Landry2004). Thus, these second-order neurons in the LHA play a fundamental role in integrating information from ARC neurons before sending it to other CNS areas involved in feeding control.

Hypothalamic neuropeptides regulating food intake

Neuropeptide Y

NPY is a thirty-six-amino acid peptide belonging to the PP-fold family of peptides, which also includes PYY and PP (Conlon, Reference Conlon2002). NPY is widely distributed in the CNS and is one of the most potent stimulators of food intake; repeated third ventricle or PVN administration of NPY induces striking hyperphagia and obesity (Stanley et al. Reference Stanley, Kyrkouli, Lampert and Leibowitz1986; Zarjevski et al. Reference Zarjevski, Cusin, Vettor, Rohner-Jeanrenaud and Jeanrenaud1993). Central administration of NPY also reduces brown fat thermogenesis (Billington et al. Reference Billington, Briggs, Grace and Levine1991), suppresses sympathetic nerve activity (Egawa et al. Reference Egawa, Yoshimatsu and Bray1991) and inhibits the thyroid axis (Fekete et al. Reference Fekete, Sarkar, Rand, Harney, Emerson, Bianco and Lechan2002) in order to reduce energy expenditure. Additionally, NPY induces hyperinsulinaemia (Moltz & McDonald, Reference Moltz and McDonald1985; Zarjevski et al. Reference Zarjevski, Cusin, Vettor, Rohner-Jeanrenaud and Jeanrenaud1993), hypercorticosteronaemia (Zarjevski et al. Reference Zarjevski, Cusin, Vettor, Rohner-Jeanrenaud and Jeanrenaud1993) and reduced plasma testosterone levels (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999); effects that are independent of increased food intake. NPY mRNA levels and NPY release in the ARC respond to changes in energy status, being increased after fasting and food restriction and decreased after refeeding (Sanacora et al. Reference Sanacora, Kershaw, Finkelstein and White1990; Kalra et al. Reference Kalra, Dube, Sahu, Phelps and Kalra1991; Swart et al. Reference Swart, Jahng, Overton and Houpt2002).

Regardless of its potent orexigenic effect, NPY-knock-out mice show normal body weight and adiposity (Erickson et al. Reference Erickson, Clegg and Palmiter1996), probably related to a compensatory and redundant mechanism in the orexigenic pathways, particularly in relation to AgRP. However, it has been reported recently that selective ablation of AgRP/NPY neurons in adult mice results in hypophagia and leanness, demonstrating direct evidence for a critical role of these neurons in the regulation of energy homeostasis (Gropp et al. Reference Gropp, Shanabrough, Borok, Xu, Janoschek and Buch2005; Luquet et al. Reference Luquet, Perez, Hnasko and Palmiter2005).

NPY, as part of the PP-fold family of peptides, binds and activates G-protein-coupled receptors termed Y1–Y6 (Larhammar, Reference Larhammar1996; Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999). Y1–Y5 receptors are present in rat brain, but Y6 is only active in mice, being absent in rats and inactive in primates (Inui, Reference Inui1999). The orexigenic action of NPY is thought to be mediated by hypothalamic Y1, Y2, Y4 and Y5 receptors (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999; Williams G et al. Reference Williams, Kaplan and Grill2000, Reference Williams, Bing, Cai, Harrold, King and Liu2001; Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005).

Melanocortin system (α-melanocyte-stimulating hormone/pro-opiomelanocortin and agouti-related peptide)

Among the hypothalamic neuropeptide systems that regulate feeding, melanocortins play a prominent role (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999; Cone, Reference Cone1999, Reference Cone2005; Coll et al. Reference Coll, Farooqi, Challis, Yeo and O'Rahilly2004b). The central melanocortin system modulates energy homeostasis through the anorectic actions of the agonist α-melanocyte-stimulating hormone (a POMC cleavage product) and the endogenous orexigenic antagonist AgRP (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999; Cone, Reference Cone1999, Reference Cone2005; Coll et al. Reference Coll, Farooqi, Challis, Yeo and O'Rahilly2004b). Five melanocortin receptors (MCnR, n 1–5) have been identified. The feeding-related effects of both α-melanocyte-stimulating hormone and AgRP are mediated via MC3R and MC4R. Both receptors are widely expressed in the hypothalamus and are found in the ARC, VMH and PVN (Kalra et al. Reference Kalra, Dube, Pu, Xu, Horvath and Kalra1999; Cone, Reference Cone1999, Reference Cone2005; Coll et al. Reference Coll, Farooqi, Challis, Yeo and O'Rahilly2004b).

Circulating hormones such as insulin (Kim et al. Reference Kim, Grace, Welch, Billington and Levine1999), leptin (Ahima & Flier, Reference Ahima and Flier2000b), ghrelin (Nakazato et al. Reference Nakazato, Murakami, Date, Kojima, Matsuo, Kangawa and Matsukura2001; Cowley et al. Reference Cowley, Smith, Diano, Tschop, Pronchuk and Grove2003; Seoane et al. Reference Seoane, López, Tovar, Casanueva, Señarís and Diéguez2003), PYY (Batterham et al. Reference Batterham, Cowley, Small, Herzog, Cohen and Dakin2002), glucocorticoids (Savontaus et al. Reference Savontaus, Conwell and Wardlaw2002) and oestrogens (Fodor & Delemarre-van de Waal, Reference Fodor and Delemarre-van de2001; Tritos et al. Reference Tritos, Segal-Lieberman, Vezeridis and Maratos-Flier2004) act on melanocortin AgRP and POMC neurons, providing information on energy status from the periphery. Hypothalamic POMC mRNA expression is regulated by nutritional status, with low levels during fasting that return to normal after leptin treatment or refeeding (Schwartz et al. Reference Schwartz, Seeley, Woods, Weigle, Campfield, Burn and Baskin1997; Swart et al. Reference Swart, Jahng, Overton and Houpt2002). In contrast, AgRP mRNA expression is increased by fasting, but unlike NPY mRNA levels, which are decreased after refeeding, AgRP levels remain elevated (Swart et al. Reference Swart, Jahng, Overton and Houpt2002). Recent evidence also suggests that circulating macronutrients, such as glucose (Sergeyev et al. Reference Sergeyev, Broberger, Gorbatyuk and Hokfelt2000; Fraley et al. Reference Fraley, Dinh and Ritter2002; Ibrahim et al. Reference Ibrahim, Bosch, Smart, Qiu, Rubinstein, Ronnekleiv, Low and Kelly2003) and lipids (Obici et al. Reference Obici, Feng, Morgan, Stein, Karkanias and Rossetti2002b; Morgan et al. Reference Morgan, Obici and Rossetti2004) modulate AgRP and POMC neurons.

The role of melanocortin signalling in body-weight homeostasis is fully supported by the phenotype of transgenic and knock-out mice, as well as identified human mutations. Transgenic mice over-expressing AgRP are obese (Ollmann et al. Reference Ollmann, Wilson, Yang, Kerns, Chen, Gantz and Barsh1997) and reduction of hypothalamic AgRP by RNA interference reduces body weight (Makimura et al. Reference Makimura, Mizuno, Mastaitis, Agami and Mobbs2002). However, AgRP-knock-out mice (as well as the double knock-out AgRP/NPY) show normal body weight and food intake (Qian et al. Reference Qian, Chen, Weingarth, Trumbauer, Novi and Guan2002), while selective ablation of AgRP/NPY neurons in adult mice results in hypophagia and leanness (Gropp et al. Reference Gropp, Shanabrough, Borok, Xu, Janoschek and Buch2005; Luquet et al. Reference Luquet, Perez, Hnasko and Palmiter2005). The role of AgRP in human obesity is not well defined, but a polymorphism in the human AgRP gene in man is associated with reduced body weight and fat mass (Marks et al. Reference Marks, Boucher, Lanouette, Perusse, Brookhart, Comuzzie, Chagnon and Cone2004). POMC-knock-out mice (Yaswen et al. Reference Yaswen, Diehl, Brennan and Hochgeschwender1999; Challis et al. Reference Challis, Coll, Yeo, Pinnock, Dickson and Thresher2004; Coll et al. Reference Coll, Challis, López, Piper, Yeo and O'Rahilly2005) and man (Kru de et al. Reference Krude, Biebermann, Luck, Horn, Brabant and Gruters1998) are hyperphagic and obese and display adrenal insufficiency. MC4R-knock-out mice (Huszar et al. Reference Huszar, Lynch, Fairchild-Huntress, Dunmore, Fang and Berkemeier1997; Fan et al. Reference Fan, Boston, Kesterson, Hruby and Cone1997) and man (Yeo et al. Reference Yeo, Farooqi, Aminian, Halsall, Stanhope and O'Rahilly1998; Farooqi et al. Reference Farooqi, Yeo, Keogh, Aminian, Jebb, Butler, Cheetham and O'Rahilly2000, Reference Farooqi, Yeo and O'Rahilly2003) also show hyperphagia and obesity. Finally, MC3R-knock-out mice display an increase in adiposity (Butler et al. Reference Butler, Kesterson, Khong, Cullen, Pelleymounter, Dekoning, Baetscher and Cone2000).

Melanin-concentrating hormone

MCH is an orexigenic neuropeptide expressed in the LHA–perifornical area. Central administration of MCH increases food intake and adiposity in rats and mice (Qu et al. Reference Qu, Ludwig, Gammeltoft, Piper, Pelleymounter, Cullen, Mathes, Przypek, Kanarek and Maratos-Flier1996; Marsh et al. Reference Marsh, Weingarth, Novi, Chen, Trumbauer and Chen2002). MCH receptor 1 antagonists inhibit food intake and induce weight loss (Borowsky et al. Reference Borowsky, Durkin, Ogozalek, Marzabadi, DeLeon and Lagu2002). MCH expression is regulated by nutritional status; fasting induces MCH mRNA expression and leptin decreases it (Qu et al. Reference Qu, Ludwig, Gammeltoft, Piper, Pelleymounter, Cullen, Mathes, Przypek, Kanarek and Maratos-Flier1996; Tritos et al. Reference Tritos, Mastaitis, Kokkotou and Maratos-Flier2001).

The important role of MCH in appetite regulation is supported by the phenotype of GM models. Transgenic mice over-expressing MCH display hyperphagia and obesity (Ludwig et al. Reference Ludwig, Tritos, Mastaitis, Kulkarni, Kokkotou, Elmquist, Lowell, Flier and Maratos-Flier2001; Marsh et al. Reference Marsh, Weingarth, Novi, Chen, Trumbauer and Chen2002), while MCH-knock-out mice are hypophagic and lean (Shimada et al. Reference Shimada, Tritos, Lowell, Flier and Maratos-Flier1998). Finally, the double knock-outs leptin/MCH have lower weight gain and adiposity compared with leptin-deficient ob/ob mice (Segal-Lieberman et al. Reference Segal-Lieberman, Bradley, Kokkotou, Carlson, Trombly, Wang, Bates, Myers, Flier and Maratos-Flier2003), suggesting that MCH is a downstream mediator of leptin effects on feeding. Finally, MCH receptor1-knock-out mice display a lean phenotype as a result of increased energy expenditure (Marsh et al. Reference Marsh, Weingarth, Novi, Chen, Trumbauer and Chen2002).

Orexins

The OX (OX-A and OX-B), or hypocretins (hypocretins 1 and 2), are neuropeptides derived from the common precursor prepro-OX (prepro-hyprocretin) expressed in the LHA/perifornical area (de Lecea et al. Reference de Lecea, Kilduff, Peyron, Gao, Foye and Danielson1998; Sakurai et al. Reference Sakurai, Amemiya, Ishii, Matsuzaki, Chemelli and Tanaka1998). Two different OX receptors have been cloned, termed OX 1 receptor (or hypocretin receptor 1) OX 2 receptor (or hypocretin receptor 2). Although OX expression in the brain is only located in the LHA–perifornical area (Broberger et al. Reference Broberger, de Lecea, Sutcliffe and Hokfelt1998a; Elias et al. Reference Elias, Saper, Maratos-Flier, Tritos, Lee and Kelly1998b; Horvath et al. Reference Horvath, Diano and van den Pol1999), OX receptors show a widespread distribution in the CNS, with high levels of abundance in some hypothalamic nuclei (ARC, DMH, LHA, PVN and VMH; Marcus et al. Reference Marcus, Aschkenasi, Lee, Chemelli, Saper, Yanagisawa and Elmquist2001; Backberg et al. Reference Backberg, Hervieu, Wilson and Meister2002). OX receptors are also present in the adrenal gland (López et al. Reference López, Senarís, Gallego, García-Caballero, Lago, Seoane, Casanueva and Diéguez1999), pituitary (Blanco et al. Reference Blanco, López, García-Caballero, Gallego, Vázquez-Boquete, Morel, Señaris, Casanueva, Diéguez and Beiras2001) gut (Kirchgessner & Liu, Reference Kirchgessner and Liu1999), testis, kidney, ovary and placenta (Johren et al. Reference Johren, Neidert, Kummer, Dendorfer and Dominiak2001).

OX are important regulators of the sleep–wake cycle and the absence of OX signalling causes narcolepsy (Willie et al. Reference Willie, Chemelli, Sinton and Yanagisawa2001; Taheri et al. Reference Taheri, Zeitzer and Mignot2002; Sutcliffe & de Lecea, Reference Sutcliffe and de Lecea2002). However, evidence also links OX to endocrine function (López et al. Reference López, Senarís, Gallego, García-Caballero, Lago, Seoane, Casanueva and Diéguez1999, Reference López, Seoane, Señarís and Diéguez2001b, Reference López, Seoane, Tovar, Nogueiras, Diéguez and Señarís2004, Reference López, Tena-Sempere, García-Caballero, Señarís, Diéguez, de Lecea and Sutcliffe2005b; Barreiro et al. Reference Barreiro, Pineda, Navarro, López, Suominen, Pinilla, Senarís, Toppari, Aguilar, Diéguez and Tena-Sempere2004; Seoane et al. Reference Seoane, Tovar, Pérez, Mallo, López, Señarís, Casanueva and Diéguez2004) and food intake regulation (Sakurai et al. Reference Sakurai, Amemiya, Ishii, Matsuzaki, Chemelli and Tanaka1998; López et al. Reference López, Diéguez, Señarís and Pandalai2001a, Reference López, Tena-Sempere, García-Caballero, Señarís, Diéguez, de Lecea and Sutcliffe2005b). Central administration of OX to rats stimulates feeding via a NPY-dependent mechanism (Sakurai et al. Reference Sakurai, Amemiya, Ishii, Matsuzaki, Chemelli and Tanaka1998; Dube et al. Reference Dube, Horvath, Kalra and Kalra2000; Ida et al. Reference Ida, Nakahara, Kuroiwa, Fukui, Nakazato, Murakami and Murakami2000; Jain et al. Reference Jain, Horvath, Kalra and Kalra2000; Yamanaka et al. Reference Yamanaka, Kunii, Nambu, Tsujino, Sakai, Matsuzaki, Miwa, Goto and Sakurai2000; López et al. Reference López, Seoane, García, Diéguez and Señarís2002). Other evidence has linked the feeding actions of OX-A to opioids, corticotrophin-releasing hormone (Ida et al. Reference Ida, Nakahara, Kuroiwa, Fukui, Nakazato, Murakami and Murakami2000), urocortin and melanocortins (Wang & Kotz, Reference Wang and Kotz2002). Finally, prepro-OX-knock-out mice (Willie et al. Reference Willie, Chemelli, Sinton and Yanagisawa2001) and the OX/ataxin-3 transgenic mice in which OX-containing neurons are ablated (Hara et al. Reference Hara, Beuckmann, Nambu, Willie, Chemelli, Sinton, Sugiyama, Yagami, Goto, Yanagisawa and Sakurai2001) are hypophagic.

OX neurons are also responsive to peripheral signals regulating food intake. The expression of prepro-OX is increased in fasting and restored to normal by leptin (Sakurai et al. Reference Sakurai, Amemiya, Ishii, Matsuzaki, Chemelli and Tanaka1998; López et al. Reference López, Seoane, García, Lago, Casanueva, Senarís and Diéguez2000; Zhu et al. Reference Zhu, Yamanaka, Kunii, Tsujino, Goto and Sakurai2002; Yamanaka et al. Reference Yamanaka, Beuckmann, Willie, Hara, Tsujino and Mieda2003). OX neurons in the lateral hypothalamus are also sensitive to glucose, being activated during hypoglycaemia (Cai et al. Reference Cai, Widdowson, Harrold, Wilson, Buckingham, Arch, Tadayyon, Clapham, Wilding and Williams1999; Griffond et al. Reference Griffond, Risold, Jacquemard, Colard and Fellmann1999; Moriguchi et al. Reference Moriguchi, Sakurai, Nambu, Yanagisawa and Goto1999). It has been also proposed that visceral feeding-related signals regulate OX actions, which are thought to act via the vagus nerve and the NST. Thus, stimuli acting as ‘terminate-eating’ signals, such as gastric distension and glucose concentrations in the portal vein, appear to be important in the regulation of OX (Cai et al. Reference Cai, Evans, Lister, Leslie, Arch, Wilson and Williams2001, Reference Cai, Liu, Evans, Clapham, Wilson, Arch, Morris and Williams2002).

Despite evidence supporting the orexigenic effect of OX, it has been proposed (Hagan et al. Reference Hagan, Leslie, Patel, Evans, Wattam and Holmes1999) that these effects are secondary, and are related to the state of arousal and vigilance necessary for normal feeding. However, OX-A-induced feeding that is independent of arousal activation has been reported (Kotz et al. Reference Kotz, Teske, Levine and Wang2002).

Cocaine- and amphetamine-regulated transcript

CART is the third-most-abundant transcript in the hypothalamus and is expressed in the ARC, DMH, LHA and PVN (Kristensen et al. Reference Kristensen, Judge, Thim, Ribel, Christjansen and Wulff1998; Elias et al. Reference Elias, Lee, Kelly, Ahima, Kuhar, Saper and Elmquist2001; Hunter et al. Reference Hunter, Philpot, Vicentic, Dominguez, Hubert and Kuhar2004). Food deprivation decreases ARC expression of CART, while peripheral leptin treatment in ob/ob mice increases CART expression (Kristensen et al. Reference Kristensen, Judge, Thim, Ribel, Christjansen and Wulff1998). Central administration of CART-(1–102) and CART-(82–103) inhibits feeding (Kristensen et al. Reference Kristensen, Judge, Thim, Ribel, Christjansen and Wulff1998) and CART-knock-out mice display a predisposition to become obese on a high-fat diet (Asnicar et al. Reference Asnicar, Smith, Yang, Heiman, Fox, Chen, Hsiung and Koster2001), an age-related increase in body weight and impaired glucose metabolism (Wierup et al. Reference Wierup, Richards, Bannon, Kuhar, Ahren and Sundler2005), supporting the role of CART in the hypothalamic mechanism regulating food intake.

Lipid sensing in the hypothalamus

Although circulating lipids have for some time been hypothesised as signalling molecules that inform the hypothalamus of metabolic status, this function has only recently been definitively demonstrated. Following an elegant experimental approach Rossetti and colleagues (Obici et al. Reference Obici, Feng, Morgan, Stein, Karkanias and Rossetti2002b; Morgan et al. Reference Morgan, Obici and Rossetti2004) have shown that central administration of long-chain fatty acids such as oleic acid inhibits food intake via AgRP/NPY neurons in the ARC; this effect is not produced by medium-chain fatty acids. The physiological relevance of these data is intriguing. Since circulating NEFA can access the brain it is likely that the anorectic action of long-chain fatty acids may play an important role in the regulation of energy balance by acting as a ‘nutrient abundance’ signal (Lam et al. Reference Lam, Pocai, Gutierrez-Juarez, Obici, Bryan, Aguilar-Bryan, Schwartz and Rossetti2005a,Reference Lam, Schwartz and Rossettib). Impairment of hypothalamic lipid-sensing in rats induces obesity (He et al. Reference He, Lam, Obici and Rossetti2006), as well alterations in plasma glucose (Pocai et al. Reference Pocai, Lam, Obici, Gutierrez-Juarez, Muse, Arduini and Rossetti2006), indicating that this mechanism may be important in the physiological regulation of metabolism and body-weight homeostasis.

Fatty acid synthesis pathway in the hypothalamus

Recent reports demonstrate that the enzymes of the fatty acid synthesis pathway (Fig. 2) are expressed in the hypothalamus. Acetyl-CoA carboxylase, FAS and malonyl-CoA decarboxylase mRNA and proteins have been detected in the ARC, DMH, PVN and VMH (Kim et al. Reference Kim, Miller, Landree, Borisy-Rudin, Brown, Tihan, Townsend, Witters, Moran, Kuhajda and Ronnett2002; López et al. Reference López, Lelliott, Tovar, Kimber, Gallego and Virtue2006). The anatomical location of these enzymes suggests that they may play a role in the hypothalamic mechanism regulating feeding. This notion is further supported by evidence demonstrating that peripheral and central administration of the FAS inhibitors cerulenin, C75 and tamoxifen reduces food intake and body weight through a malonyl-CoA-dependent mechanism (Loftus et al. Reference Loftus, Jaworsky, Frehywot, Townsend, Ronnett, Lane and Kuhajda2000; Hu et al. Reference Hu, Cha, Chohnan and Lane2003; Lelliott et al. Reference Lelliott, López, Curtis, Parker, Laudes and Yeo2005; López et al. Reference López, Lelliott, Tovar, Kimber, Gallego and Virtue2006). The anorectic action of FAS inhibitors is linked to decreased expression of AgRP/NPY and elevated expression of CART/POMC in the neurons of the ARC, although the molecular mechanisms of this interaction have not yet been completely defined (Loftus et al. Reference Loftus, Jaworsky, Frehywot, Townsend, Ronnett, Lane and Kuhajda2000; Shimokawa et al. Reference Shimokawa, Kumar and Lane2002; López et al. Reference López, Lelliott, Tovar, Kimber, Gallego and Virtue2006). Additionally, it has been demonstrated that nutritional status regulates hypothalamic malonyl-CoA levels and FAS expression in a nucleus-specific manner, with FAS mRNA levels down regulated by fasting and up regulated by refeeding, an effect specific to the VMH (Fig. 3; López et al. Reference López, Lelliott, Tovar, Kimber, Gallego and Virtue2006). This evidence suggests that the regulation of FAS could be a physiological mechanism of food-intake control and that the increase in malonyl-CoA induced by FAS inhibition may act as central lipid-sensing signal.

Fig. 2. Fatty acid synthesis pathway. Fatty acid synthesis is catalysed by acetyl-Co A carboxylase (ACC) and fatty acid synthase (FAS) in the cytoplasm. ACC catalyses the carboxylation of acetyl-CoA to malonyl-CoA. Acetyl-CoA and malonyl-CoA can be used as the substrates for the production of palmitate by the seven enzymic reactions catalysed by FAS. The synthesis step of malonyl-CoA is a reversible regulated mechanism, and malonyl-CoA decarboxylase (MCD) converts malonyl-CoA back to acetyl-CoA. The inhibition (⊥) of FAS (by using cerulenin, C75 or tamoxifen) increases the levels of malonyl-CoA in the hypothalamus, altering the concentration of long-chain fatty acid (LCFA)-CoA, which reduces feeding. The link between this effect and the neuropeptide changes is unknown (?). POMC, pro-opiomelanocortin; CART, cocaine- and amphetamine-regulated transcript; AgRP, agouti-related peptide; NPY, neuropeptide Y.

Fig. 3. Fatty acid synthase (FAS) expression and malonyl-CoA levels are nutritionally regulated in the rat hypothalamus. Expression of FAS in the ventromedial nucleus of the hypothalamus (VMH; a), arcuate nucleus (ARC; b) and paraventricular nucleus (PVN; c), and malonyl-CoA levels (d) of fed rats (□), fasted (F; ■) rats and F+refed (R; ) rats. Values are means with their standard errors represented by vertical bars. Mean values were significantly different from those for fed rats: *P<0·05, ***P<0·001. Mean values were significantly different from those for the corresponding F rats: ††P<0·01, †††P<0·001.

Brainstem regulation of food intake

The brainstem plays an essential role in the regulation of body-weight homeostasis. The NTS is anatomically close to the area postrema, a circumventricular organ, like the ARC, with a partial BBB (Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005). Consequently, the NTS is in a perfect location to receive peripheral circulating signals, as well as vagal afferents from the gastrointestinal tract and the glossopharyngeal nerves (Kalia & Sullivan, Reference Kalia and Sullivan1982; Sawchenko, Reference Sawchenko1983).

The NTS contains GLP-1, NPY and melanocortin neuronal circuits. GLP-1-expressing neurons comprise the main brainstem circuit modulating feeding. In the CNS GLP-1 is only synthesised in the caudal NTS. GLP-1 fibres project to the PVN and DMN, with fewer projections to the ARC (Stanley et al. Reference Stanley, Wynne, McGowan and Bloom2005). Central administration of GLP-1 into either the third ventricle or the fourth ventricle reduces food intake, and treatment with the GLP-1 receptor antagonist exendin (9-39) increases appetite (Turton et al. Reference Turton, O'Shea, Gunn, Beak, Edwards and Meeran1996). This finding suggests a role for endogenous GLP-1 in energy homeostasis. NPY neurons from the brainstem project to the PVN (Sawchenko et al. Reference Sawchenko, Swanson, Grzanna, Howe, Bloom and Polak1985), and extracellular NPY levels within the NTS are nutritionally-regulated (Yoshihara et al. Reference Yoshihara, Honma and Honma1996). Y1 and Y5 receptors are also located in the NTS (Harfstrand et al. Reference Harfstrand, Fuxe, Agnati, Benfenati and Goldstein1986; Dumont et al. Reference Dumont, Fournier and Quirion1998; Glass et al. Reference Glass, Chan and Pickel2002). POMC-derived peptides are synthesised in the NTS of the rat (Kawai et al. Reference Kawai, Inagaki, Shiosaka, Shibasaki, Ling, Tohyama and Shiotani1984; Bronstein et al. Reference Bronstein, Schafer, Watson and Akil1992; Fodor et al. Reference Fodor, Sluiter, Frankhuijzen-Sierevogel, Wiegant, Hoogerhout, De Wildt and Versteeg1996). Brainstem POMC neurons are activated by feeding and also by CCK treatment (Fan et al. Reference Fan, Boston, Kesterson, Hruby and Cone1997). MC4R are also expressed in the NTS (Mountjoy et al. Reference Mountjoy, Mortrud, Low, Simerly and Cone1994) and act to modulate energy intake. Fourth-ventricle administration of a MC3R/MC4R agonist decreases food intake, and MC3R/MC4R-antagonist administration to these areas increases food intake (Williams DL et al. Reference Williams, Harrold and Cutler2000).

Prolactin-releasing peptide is expressed in the NTS, in addition to the hypothalamic DMH (Lee et al. Reference Lee, Yang, Soares and Voogt2000). Prolactin-releasing peptide expression is reduced in fasting, and central administration of prolactin-releasing peptide decreases appetite by a corticotrophin-releasing hormone- and CCK-mediated mechanism (Seal et al. Reference Seal, Small, Dhillo, Stanley, Abbott, Ghatei and Bloom2001; Ellacott et al. Reference Ellacott, Lawrence, Rothwell and Luckman2002; Lawrence et al. Reference Lawrence, Liu, Stock and Luckman2004). Chronic administration of prolactin-releasing peptide does not affect food intake (Ellacott et al. Reference Ellacott, Lawrence, Pritchard and Luckman2003), suggesting a role in short-term appetite regulation instead of long-term control of body weight.

Reward and regulation of food intake

Even in the absence of an energy deficit, the rewarding nature of food may act as a stimulus to feeding. Several signals are able to modulate reward pathways. The reward circuitry is complex, involving interactions between several signalling systems, including the opioid, dopaminergic and endocannabinoid (EC) systems (Cota et al. Reference Cota, Marsicano, Lutz, Vicennati, Stalla, Pasquali and Pagotto2003a,Reference Cota, Marsicano, Tschop, Grubler, Flachskamm and Schubertb; Flier, Reference Flier2004; Di Marzo & Matias, Reference Di Marzo and Matias2005; Lichtman & Cravatt, Reference Lichtman and Cravatt2005; Fulton et al. Reference Fulton, Pissios, Manchon, Stiles, Frank, Pothos, Maratos-Flier and Flier2006; Hommel et al. Reference Hommel, Trinko, Sears, Georgescu, Liu, Gao, Thurmon, Marinelli and DiLeone2006).

Opioids

Opioids play an important role in the regulation of feeding. The anatomical site for opioid action is the nucleus accumbens (Zhang & Kelley Reference Zhang and Kelley2000; Zhang et al. Reference Zhang, Balmadrid and Kelley2003). Mice lacking enkephalin or β-endorphin lose the reinforcing property of food, despite the palatability. This effect is overridden after fasting, indicating that homeostatic mechanisms can overrule the hedonistic pathway (Hayward et al. Reference Hayward, Pintar and Low2002). In man opiate antagonists decrease food palatability without altering subjective hunger (Yeomans et al. Reference Yeomans, Wright, Macleod and Critchley1990; Drewnowski et al. Reference Drewnowski, Krahn, Demitrack, Nairn and Gosnell1992).

Endocannabinoids

The effect of marijuana (Cannabis sativa) to increase appetite has been known for many years (Di Marzo & Matias, Reference Di Marzo and Matias2005; Lichtman & Cravatt, Reference Lichtman and Cravatt2005). The primary constituent of cannabis is Δ9tetrahydrocannabinol; this molecule, along with other naturally-occurring and synthetic cannabinoids (CB), binds to two separate G-protein-coupled receptors: the CB1 receptor, which is located in the CNS and periphery; the CB2 receptor, which is primarily found in cells of the immune system (Matsuda et al. Reference Matsuda, Lolait, Brownstein, Young and Bonner1990; Devane et al. Reference Devane, Hanus, Breuer, Pertwee, Stevenson, Griffin, Gibson, Mandelbaum, Etinger and Mechoulam1992; Munro et al. Reference Munro, Thomas and Abu-Shaar1993). These receptors also bind endogenous ligands, the EC, which include the fatty acid amide N-arachidonoyl ethanolamine (anandamide) and the monoacylglycerol 2-arachidonoylglycerol (Di Marzo et al. Reference Di Marzo, Goparaju, Wang, Liu, Batkai, Jarai, Fezza, Miura, Palmiter, Sugiura and Kunos2001; Cota et al. Reference Cota, Marsicano, Lutz, Vicennati, Stalla, Pasquali and Pagotto2003a; Di Marzo & Matias, Reference Di Marzo and Matias2005; Lichtman & Cravatt, Reference Lichtman and Cravatt2005).

Central and peripheral administration of EC stimulates food intake (Williams et al. Reference Williams, Rogers and Kirkham1998; Koch, Reference Koch2001; Cota et al. Reference Cota, Marsicano, Lutz, Vicennati, Stalla, Pasquali and Pagotto2003a). This orexigenic effect is mediated via CB1 receptors in the hypothalamus, which co-localise with CART, corticotrophin-releasing hormone, MCH and OX (Cota et al. Reference Cota, Marsicano, Tschop, Grubler, Flachskamm and Schubert2003b). Additionally, CB1-knock-out mice display hypophagia and leanness (Cota et al. Reference Cota, Marsicano, Tschop, Grubler, Flachskamm and Schubert2003b), and leptin-deficient signalling is associated with high hypothalamic EC levels (Di Marzo et al. Reference Di Marzo, Goparaju, Wang, Liu, Batkai, Jarai, Fezza, Miura, Palmiter, Sugiura and Kunos2001). Recent evidence (Verty et al. Reference Verty, McFarlane, McGregor and Mallet2004) has also shown that the EC receptors are located downstream from the melanocortin system.

Together this evidence supports the important role of the EC system in the regulation of feeding. In fact, there is currently a CB1 selective antagonist, Rimonabant (SR141716), in use in phase III clinical trials that may be a potentially promising anti-obesity drug (Di Marzo & Matias, Reference Di Marzo and Matias2005).

Dopamine

The dopaminergic system is also important in the rewarding circuitry of feeding regulation (Fulton et al. Reference Fulton, Pissios, Manchon, Stiles, Frank, Pothos, Maratos-Flier and Flier2006; Hommel et al. Reference Hommel, Trinko, Sears, Georgescu, Liu, Gao, Thurmon, Marinelli and DiLeone2006). In fact, mice lacking tyrosine hydroxylase, the enzyme synthesising dopamine, are hypophagic (Szczypka et al. Reference Szczypka, Kwok, Brot, Marck, Matsumoto, Donahue and Palmiter2001). These actions are mediated via D1 and D2 receptors (Szczypka et al. Reference Szczypka, Kwok, Brot, Marck, Matsumoto, Donahue and Palmiter2001).

Serotonin

Serotonin plays an important role in regulating both rewarding and homeostatic mechanisms (Halford & Blundell, Reference Halford and Blundell2000b). Serotonin actions of food intake are mediated via the melanocortin system (Heisler et al. Reference Heisler, Cowley, Tecott, Fan, Low and Smart2002, Reference Heisler, Cowley, Kishi, Tecott, Fan and Low2003). In fact, the currently-discontinued anorectic agent fenfluramine mediates its actions via serotonin and melanocortins (Heisler et al. Reference Heisler, Cowley, Tecott, Fan, Low and Smart2002).

Conclusions

Multiple, redundant and complex peripheral neural circuits participate in the regulation of food intake and body-weight homeostasis. All this evidence indicates that obesity, and associated metabolic alterations, is complex, multifactorial and chronic pathology. Thus, the search for, and development of, new weight-loss drugs is made extremely complicated. In fact, the efficacy of drugs acting on a single molecular target might be limited by compensatory feedback mechanism. In the near future combined therapies that act on both peripheral and central targets should be sought. Understanding these molecular networks regulating food intake could lead to the design of better therapeutic targets for weight loss.

Acknowledgements

This work has been supported by grants from Instituto Salud Carlos III, Spanish Ministry of Health, Xunta de Galicia, DGICYT (BFU 2005-06287) and the EU (LSHM-CT-2003-503041).

References

Abizaid, A, Gao, Q & Horvath, TL (2006) Thoughts for food: brain mechanisms and peripheral energy balance. Neuron 51, 691702.CrossRefGoogle ScholarPubMed
Ahima, RS (2000) Leptin and the neuroendocrinology of fasting. Frontiers in Hormone Research 26, 4256.CrossRefGoogle ScholarPubMed
Ahima, RS & Flier, JS (2000 a) Adipose tissue as an endocrine organ. Trends in Endocrinology and Metabolism 11, 327332.CrossRefGoogle ScholarPubMed
Ahima, RS & Flier, JS (2000 b) Leptin. Annual Review of Physiology 62, 413437.CrossRefGoogle ScholarPubMed
Ahima, RS, Prabakaran, D, Mantzoros, C, Qu, D, Lowell, B, Maratos-Flier, E & Flier, JS (1996) Role of leptin in the neuroendocrine response to fasting. Nature 382, 250252.CrossRefGoogle ScholarPubMed
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 intake and body weight and prevent development of obesity. Nature Medicine 8, 179183.CrossRefGoogle ScholarPubMed
Anand, BK & Brobeck, JR (1951) Localization of a ‘feeding center’ in the hypothalamus of the rat. Proceedings of the Society for Experimental Biology and Medicine 77, 323324.CrossRefGoogle ScholarPubMed
Andersson, U, Filipsson, K, Abbott, CR, Woods, A, Smith, K, Bloom, SR, Carling, D & Small, CJ (2004) AMP-activated protein kinase plays a role in the control of food intake. Journal of Biological Chemistry 279, 1200512008.CrossRefGoogle Scholar
Arita, Y, Kihara, S, Ouchi, N, Takahashi, M, Maeda, K, Miyagawa, J et al. (1999) Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochemical and Biophysical Research Communications 257, 7983.CrossRefGoogle ScholarPubMed
Ariyasu, H, Takaya, K, Tagami, T, Ogawa, Y, Hosoda, K, Akamizu, T et al. (2001) Stomach is a major source of circulating ghrelin, and feeding state determines plasma ghrelin-like immunoreactivity levels in humans. Journal of Clinical Endocrinology and Metabolism 86, 47534758.CrossRefGoogle Scholar
Arvat, E, Di Vito, L, Broglio, F, Papotti, M, Muccioli, G, Dieguez, C, Casanueva, FF, Deghenghi, R, Camanni, F & Ghigo, E (2000) Preliminary evidence that ghrelin, the natural GH secretagogue (GHS)-receptor ligand, strongly stimulates GH secretion in humans. Journal of Endocrinological Investigation 23, 493495.CrossRefGoogle ScholarPubMed
Asakawa, A, Inui, A, Yuzuriha, H, Ueno, N, Katsuura, G, Fujimiya, M, Fujino, MA, Niijima, A, Meguid, MM & Kasuga, M (2003) Characterization of the effects of pancreatic polypeptide in the regulation of energy balance. Gastroenterology 124, 13251336.CrossRefGoogle ScholarPubMed
Asin, KE, Gore, PA Jr, Bednarz, L, Holladay, M & Nadzan, AM (1992) Effects of selective CCK receptor agonists on food intake after central or peripheral administration in rats. Brain Research 571, 169174.CrossRefGoogle ScholarPubMed
Asnicar, MA, Smith, DP, Yang, DD, Heiman, ML, Fox, N, Chen, YF, Hsiung, HM & Koster, A (2001) Absence of cocaine- and amphetamine-regulated transcript results in obesity in mice fed a high caloric diet. Endocrinology 142, 43944400.CrossRefGoogle ScholarPubMed
Backberg, M, Hervieu, G, Wilson, S & Meister, B (2002) Orexin receptor-1 (OX-R1) immunoreactivity in chemically identified neurons of the hypothalamus: focus on orexin targets involved in control of food and water intake. European Journal of Neuroscience 15, 315328.CrossRefGoogle ScholarPubMed
Badman, MK & Flier, JS (2005) The gut and energy balance: visceral allies in the obesity wars. Science 307, 19091914.CrossRefGoogle ScholarPubMed
Bado, A, Levasseur, S, Attoub, S, Kermorgant, S, Laigneau, JP, Bortoluzzi, MN, Moizo, L, Lehy, T, Guerre-Millo, M, Marchand-Brustel, Y & Lewin, MJ (1998) The stomach is a source of leptin. Nature 394, 790793.CrossRefGoogle ScholarPubMed
Banerjee, RR, Rangwala, SM, Shapiro, JS, Rich, AS, Rhoades, B, Qi, Y et al. (2004) Regulation of fasted blood glucose by resistin. Science 303, 11951198.CrossRefGoogle ScholarPubMed
Banks, WA (2001 a) Enhanced leptin transport across the blood-brain barrier by alpha1-adrenergic agents. Brain Research 899, 209217.CrossRefGoogle ScholarPubMed
Banks, WA (2001 b) Leptin transport across the blood-brain barrier: implications for the cause and treatment of obesity. Current Pharmaceutical Design 7, 125133.CrossRefGoogle ScholarPubMed
Banks, WA, Kastin, AJ, Huang, W, Jaspan, JB & Maness, LM (1996) Leptin enters the brain by a saturable system independent of insulin. Peptides 17, 305311.CrossRefGoogle ScholarPubMed
Barreiro, ML, Gaytan, F, Caminos, JE, Pinilla, L, Casanueva, FF, Aguilar, E, Dieguez, C & Tena-Sempere, M (2002) Cellular location and hormonal regulation of ghrelin expression in rat testis. Biology of Reproduction 67, 17681776.CrossRefGoogle ScholarPubMed
Barreiro, ML, Pineda, R, Navarro, VM, López, M, Suominen, JS, Pinilla, L, Senarís, R, Toppari, J, Aguilar, E, Diéguez, C & Tena-Sempere, M (2004) Orexin 1 receptor messenger ribonucleic acid expression and stimulation of testosterone secretion by orexin-a in rat testis. Endocrinology 145, 22972306.CrossRefGoogle ScholarPubMed
Batterham, RL, Cowley, MA, Small, CJ, Herzog, H, Cohen, MA, Dakin, CL et al. (2002) Gut hormone PYY(3–36) physiologically inhibits food intake. Nature 418, 650654.CrossRefGoogle ScholarPubMed
Batterham, RL, Le, Roux CW, Cohen, MA, Park, AJ, Ellis, SM, Patterson, M, Frost, GS, Ghatei, MA & Bloom, SR (2003) Pancreatic polypeptide reduces appetite and food intake in humans. Journal of Clinical Endocrinology and Metabolism 88, 39893992.CrossRefGoogle ScholarPubMed
Beglinger, C, Degen, L, Matzinger, D, D'Amato, M & Drewe, J (2001) Loxiglumide, a CCK-A receptor antagonist, stimulates calorie intake and hunger feelings in humans. American Journal of Physiology 280, R1149R1154.Google ScholarPubMed
Bellinger, LL & Bernardis, LL (2002) The dorsomedial hypothalamic nucleus and its role in ingestive behavior and body weight regulation: lessons learned from lesioning studies. Physiology and Behavior 76, 431442.CrossRefGoogle ScholarPubMed
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 by melanocortins. Journal of Neuroscience 22, 90489052.CrossRefGoogle ScholarPubMed
Berg, AH, Combs, TP, Du, X, Brownlee, M & Scherer, PE (2001) The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nature Medicine 7, 947953.CrossRefGoogle ScholarPubMed
Berg, AH, Combs, TP & Scherer, PE (2002) ACRP30/adiponectin: an adipokine regulating glucose and lipid metabolism. Trends in Endocrinology and Metabolism 13, 8489.CrossRefGoogle ScholarPubMed
Bernardis, LL & Bellinger, LL (1996) The lateral hypothalamic area revisited: ingestive behavior. Neuroscience and Biobehavioral Reviews 20, 189287.CrossRefGoogle ScholarPubMed
Berthoud, HR, Jedrzejewska, A & Powley, TL (1990) Simultaneous labeling of vagal innervation of the gut and afferent projections from the visceral forebrain with dil injected into the dorsal vagal complex in the rat. Journal of Comparative Neurology 301, 6579.CrossRefGoogle ScholarPubMed
Bi, S, Ladenheim, EE, Schwartz, GJ & Moran, TH (2001) A role for NPY overexpression in the dorsomedial hypothalamus in hyperphagia and obesity of OLETF rats. American Journal of Physiology 281, R254R260.Google ScholarPubMed
Billington, CJ, Briggs, JE, Grace, M & Levine, AS (1991) Effects of intracerebroventricular injection of neuropeptide Y on energy metabolism. American Journal of Physiology 260, R321R327.Google ScholarPubMed
Blanco, M, López, M, García-Caballero, T, Gallego, R, Vázquez-Boquete, A, Morel, G, Señaris, RM, Casanueva, F, Diéguez, C & Beiras, A (2001) Cellular localization of orexin receptors in human pituitary. Journal of Clinical Endocrinology and Metabolism 86, 16161619.CrossRefGoogle ScholarPubMed
Borowsky, B, Durkin, MM, Ogozalek, K, Marzabadi, MR, DeLeon, J, Lagu, B et al. (2002) Antidepressant, anxiolytic and anorectic effects of a melanin-concentrating hormone-1 receptor antagonist. Nature Medicine 8, 825830.CrossRefGoogle ScholarPubMed
Bouret, SG, Draper, SJ & Simerly, RB (2004 a) Formation of projection pathways from the arcuate nucleus of the hypothalamus to hypothalamic regions implicated in the neural control of feeding behavior in mice. Journal of Neuroscience 24, 27972805.CrossRefGoogle ScholarPubMed
Bouret, SG, Draper, SJ & Simerly, RB (2004 b) Trophic action of leptin on hypothalamic neurons that regulate feeding. Science 304, 108110.CrossRefGoogle ScholarPubMed
Bouret, SG & Simerly, RB (2004) Leptin and development of hypothalamic feeding circuits. Endocrinology 145, 26212626.CrossRefGoogle ScholarPubMed
Bray, GA (2000) Afferent signals regulating food intake. Proceedings of the Nutrition Society 59, 373384.CrossRefGoogle ScholarPubMed
Broberger, C, de Lecea, L, Sutcliffe, JG & Hokfelt, T (1998 a) Hypocretin/orexin- and melanin-concentrating hormone-expressing cells form distinct populations in the rodent lateral hypothalamus: relationship to the neuropeptide Y and agouti gene-related protein systems. Journal of Comparative Neurology 402, 460474.3.0.CO;2-S>CrossRefGoogle Scholar
Broberger, C, Johansen, J, Johansson, C, Schalling, M & Hokfelt, T (1998 b) The neuropeptide Y/agouti gene-related protein (AGRP) brain circuitry in normal, anorectic, and monosodium glutamate-treated mice. Proceedings of the National Academy of Sciences USA 95, 1504315048.CrossRefGoogle ScholarPubMed
Broberger, C, Landry, M, Wong, H, Walsh, JN & Hokfelt, T (1997) Subtypes Y1 and Y2 of the neuropeptide Y receptor are respectively expressed in pro-opiomelanocortin- and neuropeptide-Y-containing neurons of the rat hypothalamic arcuate nucleus. Neuroendocrinology 66, 393408.CrossRefGoogle ScholarPubMed
Bronstein, DM, Schafer, MK, Watson, SJ & Akil, H (1992) Evidence that beta-endorphin is synthesized in cells in the nucleus tractus solitarius: detection of POMC mRNA. Brain Research 587, 269275.CrossRefGoogle ScholarPubMed
Bruning, JC, Gautam, D, Burks, DJ, Gillette, J, Schubert, M, Orban, PC, Klein, R, Krone, W, Muller-Wieland, D & Kahn, CR (2000) Role of brain insulin receptor in control of body weight and reproduction. Science 289, 21222125.CrossRefGoogle ScholarPubMed
Bullock, BP, Heller, RS & Habener, JF (1996) Tissue distribution of messenger ribonucleic acid encoding the rat glucagon-like peptide-1 receptor. Endocrinology 137, 29682978.CrossRefGoogle ScholarPubMed
Burguera, B, Couce, ME, Curran, GL, Jensen, MD, Lloyd, RV, Cleary, MP & Poduslo, JF (2000) Obesity is associated with a decreased leptin transport across the blood-brain barrier in rats. Diabetes 49, 12191223.CrossRefGoogle ScholarPubMed
Butler, AA, Kesterson, RA, Khong, K, Cullen, MJ, Pelleymounter, MA, Dekoning, J, Baetscher, M & Cone, RD (2000) A unique metabolic syndrome causes obesity in the melanocortin-3 receptor-deficient mouse. Endocrinology 141, 35183521.CrossRefGoogle ScholarPubMed
Cai, XJ, Evans, ML, Lister, CA, Leslie, RA, Arch, JR, Wilson, S & Williams, G (2001) Hypoglycemia activates orexin neurons and selectively increases hypothalamic orexin-B levels: responses inhibited by feeding and possibly mediated by the nucleus of the solitary tract. Diabetes 50, 105112.CrossRefGoogle ScholarPubMed
Cai, XJ, Liu, XH, Evans, M, Clapham, JC, Wilson, S, Arch, JR, Morris, R & Williams, G (2002) Orexins and feeding: special occasions or everyday occurrence? Regulatory Peptides 104, 19.CrossRefGoogle ScholarPubMed
Cai, XJ, Widdowson, PS, Harrold, J, Wilson, S, Buckingham, RE, Arch, JR, Tadayyon, M, Clapham, JC, Wilding, J & Williams, G (1999) Hypothalamic orexin expression: modulation by blood glucose and feeding. Diabetes 48, 21322137.CrossRefGoogle ScholarPubMed
Calle, EE & Kaaks, R (2004) Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nature Reviews Cancer 4, 579591.CrossRefGoogle ScholarPubMed
Caminos, JE, Nogueiras, R, Blanco, M, Seoane, LM, Bravo, S, Alvarez, CV, Garcia-Caballero, T, Casanueva, FF & Dieguez, C (2003 a) Cellular distribution and regulation of ghrelin messenger ribonucleic acid in the rat pituitary gland. Endocrinology 144, 50895097.CrossRefGoogle ScholarPubMed
Caminos, JE, Nogueiras, R, Gallego, R, Bravo, S, Tovar, S, García-Caballero, T, Casanueva, FF & Diéguez, C (2005) Expression and regulation of adiponectin and receptor in human and rat placenta. Journal of Clinical Endocrinology and Metabolism 90, 42764286.CrossRefGoogle ScholarPubMed
Caminos, JE, Tena-Sempere, M, Gaytan, F, Sanchez-Criado, JE, Barreiro, ML, Nogueiras, R, Casanueva, FF, Aguilar, E & Dieguez, C (2003 b) Expression of ghrelin in the cyclic and pregnant rat ovary. Endocrinology 144, 15941602.CrossRefGoogle ScholarPubMed
Campfield, LA, Smith, FJ & Burn, P (1996) The OB protein (leptin) pathway – a link between adipose tissue mass and central neural networks. Hormone and Metabolic Research 28, 619632.CrossRefGoogle ScholarPubMed
Campfield, LA, Smith, FJ, Guisez, Y, Devos, R & Burn, P (1995) Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science 269, 546549.CrossRefGoogle ScholarPubMed
Casanueva, FF & Diéguez, C (1999) Neuroendocrine regulation and actions of leptin. Frontiers in Neuroendocrinology 20, 317363.CrossRefGoogle ScholarPubMed
Challis, BG, Coll, AP, Yeo, GS, Pinnock, SB, Dickson, SL, Thresher, RR et al. (2004) Mice lacking pro-opiomelanocortin are sensitive to high-fat feeding but respond normally to the acute anorectic effects of peptide-YY(3–36). Proceedings of the National Academy of Sciences USA 101, 46954700.CrossRefGoogle Scholar
Challis, BG, Pinnock, SB, Coll, AP, Carter, RN, Dickson, SL & O'Rahilly, S (2003) Acute effects of PYY3–36 on food intake and hypothalamic neuropeptide expression in the mouse. Biochemical and Biophysical Research Communications 311, 915919.CrossRefGoogle ScholarPubMed
Chen, H, Charlat, O, Tartaglia, LA, Woolf, EA, Weng, X, Ellis, SJ et al. (1996) Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell 84, 491495.CrossRefGoogle ScholarPubMed
Chen, HY, Trumbauer, ME, Chen, AS, Weingarth, DT, Adams, JR, Frazier, EG et al. (2004) Orexigenic action of peripheral ghrelin is mediated by neuropeptide Y (NPY) and Agouti-related protein (AgRP). Endocrinology 145, 26072612.CrossRefGoogle ScholarPubMed
Cheung, CC, Clifton, DK & Steiner, RA (1997) Proopiomelanocortin neurons are direct targets for leptin in the hypothalamus. Endocrinology 138, 44894492.CrossRefGoogle ScholarPubMed
Chou, TC, Lee, CE, Lu, J, Elmquist, JK, Hara, J, Willie, JT et al. (2001) Orexin (hypocretin) neurons contain dynorphin. Journal of Neuroscience 21, RC168.CrossRefGoogle ScholarPubMed
Chua, SC Jr, Koutras, IK, Han, L, Liu, SM, Kay, J, Young, SJ, Chung, WK & Leibel, RL (1997) Fine structure of the murine leptin receptor gene: splice site suppression is required to form two alternatively spliced transcripts. Genomics 45, 264270.CrossRefGoogle ScholarPubMed
Clark, JT, Kalra, PS, Crowley, WR & Kalra, SP (1984) Neuropeptide Y and human pancreatic polypeptide stimulate feeding behavior in rats. Endocrinology 115, 427429.CrossRefGoogle ScholarPubMed
Clark, JT, Sahu, A, Kalra, PS, Balasubramaniam, A & Kalra, SP (1987) Neuropeptide Y (NPY)-induced feeding behavior in female rats: comparison with human NPY ([Met17]NPY), NPY analog ([norLeu4]NPY) and peptide YY. Regulatory Peptides 17, 3139.CrossRefGoogle ScholarPubMed
Clement, K, Vaisse, C, Lahlou, N, Cabrol, S, Pelloux, V, Cassuto, D et al. (1998) A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392, 398401.CrossRefGoogle ScholarPubMed
Clement, K, Vega, N, Laville, M, Pelloux, V, Guy-Grand, B, Basdevant, A & Vidal, H (2002) Adipose tissue gene expression in patients with a loss of function mutation in the leptin receptor. International Journal of Obesity and Related Metabolic Disorders 26, 15331538.CrossRefGoogle ScholarPubMed
Coll, AP, Challis, BG, López, M, Piper, S, Yeo, GS & O'Rahilly, S (2005) Proopiomelanocortin-deficient mice are hypersensitive to the adverse metabolic effects of glucocorticoids. Diabetes 54, 22692276.CrossRefGoogle Scholar
Coll, AP, Challis, BG & O'Rahilly, S (2004 a) Peptide YY3–36 and satiety: clarity or confusion? Endocrinology 145, 25822584.CrossRefGoogle ScholarPubMed
Coll, AP, Farooqi, IS, Challis, BG, Yeo, GS & O'Rahilly, S (2004 b) Proopiomelanocortin and energy balance: insights from human and murine genetics. Journal of Clinical Endocrinology and Metabolism 89, 25572562.CrossRefGoogle ScholarPubMed
Cone, RD (1999) The central melanocortin system and energy homeostasis. Trends in Endocrinology and Metabolism 10, 211216.CrossRefGoogle ScholarPubMed
Cone, RD (2005) Anatomy and regulation of the central melanocortin system. Nature Neuroscience 8, 571578.CrossRefGoogle ScholarPubMed
Conlon, JM (2002) The origin and evolution of peptide YY (PYY) and pancreatic polypeptide (PP). Peptides 23, 269278.CrossRefGoogle ScholarPubMed
Considine, RV, Sinha, MK, Heiman, ML, Kriauciunas, A, Stephens, TW, Nyce, MR, Ohannesian, JP, Marco, CC, McKee, LJ & Bauer, TL (1996) Serum immunoreactive-leptin concentrations in normal-weight and obese humans. New England Journal of Medicine 334, 292295.CrossRefGoogle ScholarPubMed
Corp, ES, Woods, SC, Porte, D Jr, Dorsa, DM, Figlewicz, DP & Baskin, DG (1986) Localization of 125I-insulin binding sites in the rat hypothalamus by quantitative autoradiography. Neuroscience Letters 70, 1722.CrossRefGoogle ScholarPubMed
Corpa, ES, McQuade, J, Krasnicki, S & Conze, DB (2001) Feeding after fourth ventricular administration of neuropeptide Y receptor agonists in rats. Peptides 22, 493499.CrossRefGoogle Scholar
Cota, D, Marsicano, G, Lutz, B, Vicennati, V, Stalla, GK, Pasquali, R & Pagotto, U (2003 a) Endogenous cannabinoid system as a modulator of food intake. International Journal of Obesity and Related Metabolic Disorders 27, 289301.CrossRefGoogle ScholarPubMed
Cota, D, Marsicano, G, Tschop, M, Grubler, Y, Flachskamm, C, Schubert, M et al. (2003 b) The endogenous cannabinoid system affects energy balance via central orexigenic drive and peripheral lipogenesis. Journal of Clinical Investigation 112, 423431.CrossRefGoogle ScholarPubMed
Cowley, MA, Pronchuk, N, Fan, W, Dinulescu, DM, Colmers, WF & Cone, RD (1999) Integration of NPY, AGRP, and melanocortin signals in the hypothalamic paraventricular nucleus: evidence of a cellular basis for the adipostat. Neuron 24, 155163.CrossRefGoogle ScholarPubMed
Cowley, MA, Smith, RG, Diano, S, Tschop, M, Pronchuk, N, Grove, KL et al. (2003) The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron 37, 649661.CrossRefGoogle ScholarPubMed
Cummings, DE, Clement, K, Purnell, JQ, Vaisse, C, Foster, KE, Frayo, RS, Schwartz, MW, Basdevant, A & Weigle, DS (2002 a) Elevated plasma ghrelin levels in Prader Willi syndrome. Nature Medicine 8, 643644.CrossRefGoogle ScholarPubMed
Cummings, DE, Frayo, RS, Marmonier, C, Aubert, R & Chapelot, D (2004) Plasma ghrelin levels and hunger scores in humans initiating meals voluntarily without time- and food-related cues. American Journal of Physiology 287, E297E304.Google ScholarPubMed
Cummings, DE, Purnell, JQ, Frayo, RS, Schmidova, K, Wisse, BE & Weigle, DS (2001) A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 50, 17141719.CrossRefGoogle ScholarPubMed
Cummings, DE & Shannon, MH (2003) Roles for ghrelin in the regulation of appetite and body weight. Archives of Surgery 138, 389396.CrossRefGoogle ScholarPubMed
Cummings, DE, Weigle, DS, Frayo, RS, Breen, PA, Ma, MK, Dellinger, EP & Purnell, JQ (2002 b) Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. New England Journal of Medicine 346, 16231630.CrossRefGoogle ScholarPubMed
Dakin, CL, Small, CJ, Batterham, RL, Neary, NM, Cohen, MA, Patterson, M, Ghatei, MA & Bloom, SR (2004) Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocrinology 145, 26872695.CrossRefGoogle ScholarPubMed
Date, Y, Kojima, M, Hosoda, H, Sawaguchi, A, Mondal, MS, Suganuma, T, Matsukura, S, Kangawa, K & Nakazato, M (2000) Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology 141, 42554261.CrossRefGoogle Scholar
de Lecea, L, Kilduff, TS, Peyron, C, Gao, X, Foye, PE, Danielson, PE et al. (1998) The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proceedings of the National Academy of Sciences USA 95, 322327.CrossRefGoogle ScholarPubMed
Devane, WA, Hanus, L, Breuer, A, Pertwee, RG, Stevenson, LA, Griffin, G, Gibson, D, Mandelbaum, A, Etinger, A & Mechoulam, R (1992) Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 258, 19461949.CrossRefGoogle Scholar
Dhillon, H, Zigman, JM, Ye, C, Lee, CE, McGovern, RA, Tang, V et al. (2006) Leptin directly activates SF1 neurons in the VMH, and this action by leptin is required for normal body-weight homeostasis. Neuron 49, 191203.CrossRefGoogle ScholarPubMed
Di Marzo, V, Goparaju, SK, Wang, L, Liu, J, Batkai, S, Jarai, Z, Fezza, F, Miura, GI, Palmiter, RD, Sugiura, T & Kunos, G (2001) Leptin-regulated endocannabinoids are involved in maintaining food intake. Nature 410, 822825.CrossRefGoogle ScholarPubMed
Di Marzo, V & Matias, I (2005) Endocannabinoid control of food intake and energy balance. Nature Neuroscience 8, 585589.CrossRefGoogle ScholarPubMed
Drewnowski, A, Krahn, DD, Demitrack, MA, Nairn, K & Gosnell, BA (1992) Taste responses and preferences for sweet high-fat foods: evidence for opioid involvement. Physiology and Behavior 51, 371379.CrossRefGoogle ScholarPubMed
Dube, MG, Horvath, TL, Kalra, PS & Kalra, SP (2000) Evidence of NPY Y5 receptor involvement in food intake elicited by orexin A in sated rats. Peptides 21, 15571560.CrossRefGoogle ScholarPubMed
Dumont, Y, Fournier, A & Quirion, R (1998) Expression and characterization of the neuropeptide Y Y5 receptor subtype in the rat brain. Journal of Neuroscience 18, 55655574.CrossRefGoogle ScholarPubMed
Egawa, M, Yoshimatsu, H & Bray, GA (1991) Neuropeptide Y suppresses sympathetic activity to interscapular brown adipose tissue in rats. American Journal of Physiology 260, R328R334.Google ScholarPubMed
El Haschimi, K, Pierroz, DD, Hileman, SM, Bjorbaek, C & Flier, JS (2000) Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. Journal of Clinical Investigation 105, 18271832.CrossRefGoogle ScholarPubMed
Elias, CF, Aschkenasi, C, Lee, C, Kelly, J, Ahima, RS, Bjorbaek, C, Flier, JS, Saper, CB & Elmquist, JK (1999) Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area. Neuron 23, 775786.CrossRefGoogle Scholar
Elias, CF, Lee, C, Kelly, J, Aschkenasi, C, Ahima, RS, Couceyro, PR, Kuhar, MJ, Saper, CB & Elmquist, JK (1998 a) Leptin activates hypothalamic CART neurons projecting to the spinal cord. Neuron 21, 13751385.CrossRefGoogle ScholarPubMed
Elias, CF, Lee, CE, Kelly, JF, Ahima, RS, Kuhar, M, Saper, CB & Elmquist, JK (2001) Characterization of CART neurons in the rat and human hypothalamus. Journal of Comparative Neurology 432, 119.CrossRefGoogle ScholarPubMed
Elias, CF, Saper, CB, Maratos-Flier, E, Tritos, NA, Lee, C, Kelly, J et al. (1998 b) Chemically defined projections linking the mediobasal hypothalamus and the lateral hypothalamic area. Journal of Comparative Neurology 402, 442459.3.0.CO;2-R>CrossRefGoogle ScholarPubMed
Ellacott, KL, Lawrence, CB, Pritchard, LE & Luckman, SM (2003) Repeated administration of the anorectic factor prolactin-releasing peptide leads to tolerance to its effects on energy homeostasis. American Journal of Physiology 285, R1005R1010.Google ScholarPubMed
Ellacott, KL, Lawrence, CB, Rothwell, NJ & Luckman, SM (2002) PRL-releasing peptide interacts with leptin to reduce food intake and body weight. Endocrinology 143, 368374.CrossRefGoogle ScholarPubMed
Elmquist, JK, Ahima, RS, Maratos-Flier, E, Flier, JS & Saper, CB (1997) Leptin activates neurons in ventrobasal hypothalamus and brainstem. Endocrinology 138, 839842.CrossRefGoogle ScholarPubMed
Elmquist, JK, Bjorbaek, C, Ahima, RS, Flier, JS & Saper, CB (1998) Distributions of leptin receptor mRNA isoforms in the rat brain. Journal of Comparative Neurology 395, 535547.3.0.CO;2-2>CrossRefGoogle ScholarPubMed
English, PJ, Ghatei, MA, Malik, IA, Bloom, SR & Wilding, JP (2002) Food fails to suppress ghrelin levels in obese humans. Journal of Clinical Endocrinology and Metabolism 87, 2984.CrossRefGoogle ScholarPubMed
Erickson, JC, Clegg, KE & Palmiter, RD (1996) Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y. Nature 381, 415421.CrossRefGoogle ScholarPubMed
Fan, W, Boston, BA, Kesterson, RA, Hruby, VJ & Cone, RD (1997) Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 385, 165168.CrossRefGoogle ScholarPubMed
Farooqi, IS, Jebb, SA, Langmack, G, Lawrence, E, Cheetham, CH, Prentice, AM, Hughes, IA, McCamish, MA & O'Rahilly, S (1999) Effects of recombinant leptin therapy in a child with congenital leptin deficiency. New England Journal of Medicine 341, 879884.CrossRefGoogle Scholar
Farooqi, IS & O'Rahilly, S (2005) Monogenic obesity in humans. Annual Review of Medicine 56, 443458.CrossRefGoogle ScholarPubMed
Farooqi, IS, Yeo, GS, Keogh, JM, Aminian, S, Jebb, SA, Butler, G, Cheetham, T & O'Rahilly, S (2000) Dominant and recessive inheritance of morbid obesity associated with melanocortin 4 receptor deficiency. Journal of Clinical Investigation 106, 271279.CrossRefGoogle ScholarPubMed
Farooqi, IS, Yeo, GS & O'Rahilly, S (2003) Binge eating as a phenotype of melanocortin 4 receptor gene mutations. New England Journal of Medicine 349, 606609.Google ScholarPubMed
Fei, H, Okano, HJ, Li, C, Lee, GH, Zhao, C, Darnell, R & Friedman, JM (1997) Anatomic localization of alternatively spliced leptin receptors (Ob-R) in mouse brain and other tissues. Proceedings of the National Academy of Sciences USA 94, 70017005.CrossRefGoogle ScholarPubMed
Feifel, D & Vaccarino, FJ (1989) Feeding effects of growth hormone-releasing factor in rats are photoperiod sensitive. Behavioral Neuroscience 103, 824830.CrossRefGoogle ScholarPubMed
Feifel, D & Vaccarino, FJ (1990) Central somatostatin: a re-examination of its effects on feeding. Brain Research 535, 189194.CrossRefGoogle ScholarPubMed
Feifel, D & Vaccarino, FJ (1994) Growth hormone-regulatory peptides (GHRH and somatostatin) and feeding: a model for the integration of central and peripheral function. Neuroscience and Biobehavioral Reviews 18, 421433.CrossRefGoogle Scholar
Fekete, C, Legradi, G, Mihaly, E, Huang, QH, Tatro, JB, Rand, WM, Emerson, CH & Lechan, RM (2000) α-Melanocyte-stimulating hormone is contained in nerve terminals innervating thyrotropin-releasing hormone-synthesizing neurons in the hypothalamic paraventricular nucleus and prevents fasting-induced suppression of prothyrotropin-releasing hormone gene expression. Journal of Neuroscience 20, 15501558.CrossRefGoogle ScholarPubMed
Fekete, C, Sarkar, S, Rand, WM, Harney, JW, Emerson, CH, Bianco, AC & Lechan, RM (2002) Agouti-related protein (AGRP) has a central inhibitory action on the hypothalamic-pituitary-thyroid (HPT) axis; comparisons between the effect of AGRP and neuropeptide Y on energy homeostasis and the HPT axis. Endocrinology 143, 38463853.CrossRefGoogle Scholar
Flier, JS (2004) Obesity wars: molecular progress confronts an expanding epidemic. Cell 116, 337350.CrossRefGoogle ScholarPubMed
Flint, A, Raben, A, Astrup, A & Holst, JJ (1998) Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. Journal of Clinical Investigation 101, 515520.CrossRefGoogle ScholarPubMed
Flint, A, Raben, A, Ersboll, AK, Holst, JJ & Astrup, A (2001) The effect of physiological levels of glucagon-like peptide-1 on appetite, gastric emptying, energy and substrate metabolism in obesity. International Journal of Obesity and Related Metabolic Disorders 25, 781792.CrossRefGoogle ScholarPubMed
Flint, A, Raben, A, Rehfeld, JF, Holst, JJ & Astrup, A (2000) The effect of glucagon-like peptide-1 on energy expenditure and substrate metabolism in humans. International Journal of Obesity and Related Metabolic Disorders 24, 288298.CrossRefGoogle ScholarPubMed
Fodor, M & Delemarre-van de, Waal HA (2001) Are POMC neurons targets for sex steroids in the arcuate nucleus of the rat? Neuroreport 12, 39893991.CrossRefGoogle ScholarPubMed
Fodor, M, Sluiter, A, Frankhuijzen-Sierevogel, A, Wiegant, VM, Hoogerhout, P, De Wildt, DJ & Versteeg, DH (1996) Distribution of Lys-gamma 2-melanocyte-stimulating hormone-(Lys-gamma 2-MSH)-like immunoreactivity in neuronal elements in the brain and peripheral tissues of the rat. Brain Research 731, 182189.CrossRefGoogle ScholarPubMed
Fogteloo, AJ, Pijl, H, Frolich, M, McCamish, M & Meinders, AE (2003) Effects of recombinant human leptin treatment as an adjunct of moderate energy restriction on body weight, resting energy expenditure and energy intake in obese humans. Diabetes Nutrition and Metabolism 16, 109114.Google ScholarPubMed
Fraley, GS, Dinh, TT & Ritter, S (2002) Immunotoxic catecholamine lesions attenuate 2DG-induced increase of AGRP mRNA. Peptides 23, 10931099.CrossRefGoogle ScholarPubMed
Frederich, RC, Lollmann, B, Hamann, A, Napolitano-Rosen, A, Kahn, BB, Lowell, BB & Flier, JS (1995) Expression of ob mRNA and its encoded protein in rodents. Impact of nutrition and obesity. Journal of Clinical Investigation 96, 16581663.CrossRefGoogle Scholar
Friedman, JM (2000) Obesity in the new millennium. Nature 404, 632634.CrossRefGoogle ScholarPubMed
Friedman, JM (2003) A war on obesity, not the obese. Science 299, 856858.CrossRefGoogle Scholar
Friedman, JM & Halaas, JL (1998) Leptin and the regulation of body weight in mammals. Nature 395, 763770.CrossRefGoogle ScholarPubMed
Fruebis, J, Tsao, TS, Javorschi, S, Ebbets-Reed, D, Erickson, MR, Yen, FT, Bihain, BE & Lodish, HF (2001) Proteolytic cleavage product of 30-kDa adipocyte complement-related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proceedings of the National Academy of Sciences USA 98, 20052010.CrossRefGoogle ScholarPubMed
Fujimoto, S, Inui, A, Kiyota, N, Seki, W, Koide, K, Takamiya, S, Uemoto, M, Nakajima, Y, Baba, S & Kasuga, M (1997) Increased cholecystokinin and pancreatic polypeptide responses to a fat-rich meal in patients with restrictive but not bulimic anorexia nervosa. Biological Psychiatry 41, 10681070.CrossRefGoogle Scholar
Fulton, S, Pissios, P, Manchon, RP, Stiles, L, Frank, L, Pothos, EN, Maratos-Flier, E & Flier, JS (2006) Leptin regulation of the mesoaccumbens dopamine pathway. Neuron 51, 811822.CrossRefGoogle ScholarPubMed
Furuhata, Y, Kagaya, R, Hirabayashi, K, Ikeda, A, Chang, KT, Nishihara, M & Takahashi, M (2000) Development of obesity in transgenic rats with low circulating growth hormone levels: involvement of leptin resistance. European Journal of Endocrinology 143, 535541.CrossRefGoogle ScholarPubMed
García, A, Alvarez, CV, Smith, RG & Diéguez, C (2001) Regulation of pit-1 expression by ghrelin and ghrp-6 through the gh secretagogue receptor. Molecular Endocrinology 15, 14841495.CrossRefGoogle ScholarPubMed
García, MC, Casanueva, FF, Diéguez, C & Señarís, R (2000) Gestational profile of leptin messenger ribonucleic acid (mRNA) content in the placenta and adipose tissue in the rat, and regulation of the mRNA levels of the leptin receptor subtypes in the hypothalamus during pregnancy and lactation. Biology of Reproduction 62, 698703.CrossRefGoogle ScholarPubMed
García, MC, López, M, Gualillo, O, Seoane, L, Diéguez, C & Señarís, R (2003) Hypothalamic levels of NPY, MCH, and prepro-orexin mRNA during pregnancy and lactation in the rat: role of prolactin. FASEB Journal 17, 13921400.CrossRefGoogle ScholarPubMed
García, MC, Wernstedt, I, Berndtsson, A, Enge, M, Bell, M, Hultgren, O et al. (2006) Mature-onset obesity in interleukin-1 receptor I knockout mice. Diabetes 55, 12051213.CrossRefGoogle ScholarPubMed
Gaytan, F, Barreiro, ML, Chopin, LK, Herington, AC, Morales, C, Pinilla, L, Casanueva, FF, Aguilar, E, Dieguez, C & Tena-Sempere, M (2003) Immunolocalization of ghrelin and its functional receptor, the type 1a growth hormone secretagogue receptor, in the cyclic human ovary. Journal of Clinical Endocrinology and Metabolism 88, 879887.CrossRefGoogle ScholarPubMed
Ghatei, MA, Uttenthal, LO, Christofides, ND, Bryant, MG & Bloom, SR (1983) Molecular forms of human enteroglucagon in tissue and plasma: plasma responses to nutrient stimuli in health and in disorders of the upper gastrointestinal tract. Journal of Clinical Endocrinology and Metabolism 57, 488495.CrossRefGoogle ScholarPubMed
Gibbs, J, Fauser, DJ, Rowe, EA, Rolls, BJ, Rolls, ET & Maddison, SP (1979) Bombesin suppresses feeding in rats. Nature 282, 208210.CrossRefGoogle ScholarPubMed
Gibbs, J, Young, RC & Smith, GP (1973) Cholecystokinin decreases food intake in rats. Journal of Comparative Physiology and Psychology 84, 488495.CrossRefGoogle ScholarPubMed
Glass, MJ, Chan, J & Pickel, VM (2002) Ultrastructural localization of neuropeptide Y Y1 receptors in the rat medial nucleus tractus solitarius: relationships with neuropeptide Y or catecholamine neurons. Journal of Neuroscience Research 67, 753765.CrossRefGoogle ScholarPubMed
Gooley, JJ, Schomer, A & Saper, CB (2006) The dorsomedial hypothalamic nucleus is critical for the expression of food-entrainable circadian rhythms. Nature Neuroscience 9, 398407.CrossRefGoogle ScholarPubMed
Grandt, D, Schimiczek, M, Beglinger, C, Layer, P, Goebell, H, Eysselein, VE & Reeve, JR Jr (1994) Two molecular forms of peptide YY (PYY) are abundant in human blood: characterization of a radioimmunoassay recognizing PYY 1–36 and PYY 3–36. Regulatory Peptides 51, 151159.CrossRefGoogle ScholarPubMed
Griffond, B, Risold, PY, Jacquemard, C, Colard, C & Fellmann, D (1999) Insulin-induced hypoglycemia increases preprohypocretin (orexin) mRNA in the rat lateral hypothalamic area. Neuroscience Letters 262, 7780.CrossRefGoogle ScholarPubMed
Gropp, E, Shanabrough, M, Borok, E, Xu, AW, Janoschek, R, Buch, T et al. (2005) Agouti-related peptide-expressing neurons are mandatory for feeding. Nature Neuroscience 8, 12891291.CrossRefGoogle ScholarPubMed
Grove, KL & Cowley, MA (2005) Is ghrelin a signal for the development of metabolic systems? Journal of Clinical Investigation 115, 33933397.CrossRefGoogle ScholarPubMed
Gualillo, O, Caminos, J, Blanco, M, García-Caballero, T, Kojima, M, Kangawa, K, Diéguez, C & Casanueva, F (2001) Ghrelin, a novel placental-derived hormone. Endocrinology 142, 788794.CrossRefGoogle ScholarPubMed
Guan, XM, Yu, H, Palyha, OC, McKee, KK, Feighner, SD, Sirinathsinghji, DJ, Smith, RG, Van Der Ploeg, LH & Howard, AD (1997) Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues. Brain Research Molecular Brain Research 48, 2329.CrossRefGoogle ScholarPubMed
Gutzwiller, JP, Goke, B, Drewe, J, Hildebrand, P, Ketterer, S, Handschin, D, Winterhalder, R, Conen, D & Beglinger, C (1999) Glucagon-like peptide-1: a potent regulator of food intake in humans. Gut 44, 8186.CrossRefGoogle ScholarPubMed
Hagan, JJ, Leslie, RA, Patel, S, Evans, ML, Wattam, TA, Holmes, S et al. (1999) Orexin A activates locus coeruleus cell firing and increases arousal in the rat. Proceedings of the National Academy of Sciences USA 96, 1091110916.CrossRefGoogle ScholarPubMed
Hagan, MM, Castaneda, E, Sumaya, IC, Fleming, SM, Galloway, J & Moss, DE (1998) The effect of hypothalamic peptide YY on hippocampal acetylcholine release in vivo: implications for limbic function in binge-eating behavior. Brain Research 805, 2028.CrossRefGoogle ScholarPubMed
Hahn, TM, Breininger, JF, Baskin, DG & Schwartz, MW (1998) Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nature Neuroscience 1, 271272.CrossRefGoogle ScholarPubMed
Hakansson, M, de Lecea, L, Sutcliffe, JG, Yanagisawa, M & Meister, B (1999) Leptin receptor- and STAT3-immunoreactivities in hypocretin/orexin neurones of the lateral hypothalamus. Journal of Neuroendocrinology 11, 653663.CrossRefGoogle ScholarPubMed
Hakansson, ML, Brown, H, Ghilardi, N, Skoda, RC & Meister, B (1998) Leptin receptor immunoreactivity in chemically defined target neurons of the hypothalamus. Journal of Neuroscience 18, 559572.CrossRefGoogle ScholarPubMed
Halaas, JL, Gajiwala, KS, Maffei, M, Cohen, SL, Chait, BT, Rabinowitz, D, Lallone, RL, Burley, SK & Friedman, JM (1995) Weight-reducing effects of the plasma protein encoded by the obese gene. Science 269, 543546.CrossRefGoogle ScholarPubMed
Halatchev, IG & Cone, RD (2005) Peripheral administration of PYY3–36 produces conditioned taste aversion in mice. Cell Metabolism 1, 159168.CrossRefGoogle ScholarPubMed
Halford, JC & Blundell, JE (2000 a) Pharmacology of appetite suppression. Progress in Drug Research 54, 2558.CrossRefGoogle ScholarPubMed
Halford, JC & Blundell, JE (2000 b) Separate systems for serotonin and leptin in appetite control. Annals of Medicine 32, 222232.CrossRefGoogle ScholarPubMed
Hansen, TK, Dall, R, Hosoda, H, Kojima, M, Kangawa, K, Christiansen, JS & Jorgensen, JO (2002) Weight loss increases circulating levels of ghrelin in human obesity. Clinical Endocrinology 56, 203206.CrossRefGoogle ScholarPubMed
Hara, J, Beuckmann, CT, Nambu, T, Willie, JT, Chemelli, RM, Sinton, CM, Sugiyama, F, Yagami, K, Goto, K, Yanagisawa, M & Sakurai, T (2001) Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity. Neuron 30, 345354.CrossRefGoogle ScholarPubMed
Harfstrand, A, Fuxe, K, Agnati, LF, Benfenati, F & Goldstein, M (1986) Receptor autoradiographical evidence for high densities of 125I-neuropeptide Y binding sites in the nucleus tractus solitarius of the normal male rat. Acta Physiologica Scandinavica 128, 195200.CrossRefGoogle ScholarPubMed
Hayward, MD, Pintar, JE & Low, MJ (2002) Selective reward deficit in mice lacking beta-endorphin and enkephalin. Journal of Neuroscience 22, 82518258.CrossRefGoogle ScholarPubMed
He, W, Lam, TK, Obici, S & Rossetti, L (2006) Molecular disruption of hypothalamic nutrient sensing induces obesity. Nature Neuroscience 9, 227233.CrossRefGoogle ScholarPubMed
Hecherington, A & Ranson, S (1942) The spontaneous activity and food intake of rats with hypothalamic lesions. American Journal of Physiology 136, 609617.CrossRefGoogle Scholar
Heisler, LK, Cowley, MA, Kishi, T, Tecott, LH, Fan, W, Low, MJ et al. (2003) Central serotonin and melanocortin pathways regulating energy homeostasis. Annals of the New York Academy of Sciences 994, 169174.CrossRefGoogle ScholarPubMed
Heisler, LK, Cowley, MA, Tecott, LH, Fan, W, Low, MJ, Smart, JL et al. (2002) Activation of central melanocortin pathways by fenfluramine. Science 297, 609611.CrossRefGoogle ScholarPubMed
Henry, BA, Rao, A, Ikenasio, BA, Mountjoy, KG, Tilbrook, AJ & Clarke, IJ (2001) Differential expression of cocaine- and amphetamine-regulated transcript and agouti related-protein in chronically food-restricted sheep. Brain Research 918, 4050.CrossRefGoogle ScholarPubMed
Herrmann, C, Goke, R, Richter, G, Fehmann, HC, Arnold, R & Goke, B (1995) Glucagon-like peptide-1 and glucose-dependent insulin-releasing polypeptide plasma levels in response to nutrients. Digestion 56, 117126.CrossRefGoogle ScholarPubMed
Heymsfield, SB, Greenberg, AS, Fujioka, K, Dixon, RM, Kushner, R, Hunt, T, Lubina, JA, Patane, J, Self, B, Hunt, P & McCamish, M (1999) Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. Journal of the American Medical Association 282, 15681575.CrossRefGoogle ScholarPubMed
Hirasawa, A, Tsumaya, K, Awaji, T, Katsuma, S, Adachi, T, Yamada, M, Sugimoto, Y, Miyazaki, S & Tsujimoto, G (2005) Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nature Medicine 11, 9094.CrossRefGoogle ScholarPubMed
Holst, B, Egerod, KL, Schild, E, Vickers, SP, Cheetham, S, Gerlach, LO, Storjohann, L, Stidsen, CE, Jones, R, Beck-Sickinger, AG & Schwartz, TW (2006) GPR39 signaling is stimulated by zinc ions but not by obestatin. Endocrinology (Epublication ahead of print version).Google Scholar
Hommel, JD, Trinko, R, Sears, RM, Georgescu, D, Liu, ZW, Gao, XB, Thurmon, JJ, Marinelli, M & DiLeone, RJ (2006) Leptin receptor signaling in midbrain dopamine neurons regulates feeding. Neuron 51, 801810.CrossRefGoogle ScholarPubMed
Horvath, TL & Diano, S (2004) Opinion: The floating blueprint of hypothalamic feeding circuits. Nature Reviews Neuroscience 5, 662667.CrossRefGoogle Scholar
Horvath, TL, Diano, S, Sotonyi, P, Heiman, M & Tschop, M (2001) Minireview: ghrelin and the regulation of energy balance – a hypothalamic perspective. Endocrinology 142, 41634169.CrossRefGoogle ScholarPubMed
Horvath, TL, Diano, S & Tschop, M (2004) Brain circuits regulating energy homeostasis. Neuroscientist 10, 235246.CrossRefGoogle ScholarPubMed
Horvath, TL, Diano, S & van den Pol, AN (1999) Synaptic interaction between hypocretin (orexin) and neuropeptide Y cells in the rodent and primate hypothalamus: a novel circuit implicated in metabolic and endocrine regulations. Journal of Neuroscience 19, 10721087.CrossRefGoogle ScholarPubMed
Hotta, K, Funahashi, T, Bodkin, NL, Ortmeyer, HK, Arita, Y, Hansen, BC & Matsuzawa, Y (2001) Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys. Diabetes 50, 11261133.CrossRefGoogle ScholarPubMed
Howard, JK, Cave, BJ, Oksanen, LJ, Tzameli, I, Bjorbaek, C & Flier, JS (2004) Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of Socs3. Nature Medicine 10, 734738.CrossRefGoogle ScholarPubMed
Hu, E, Liang, P & Spiegelman, BM (1996) AdipoQ is a novel adipose-specific gene dysregulated in obesity. Journal of Biological Chemistry 271, 1069710703.CrossRefGoogle ScholarPubMed
Hu, Z, Cha, SH, Chohnan, S & Lane, MD (2003) Hypothalamic malonyl-CoA as a mediator of feeding behavior. Proceedings of the National Academy of Sciences USA 100, 1262412629.CrossRefGoogle ScholarPubMed
Hunter, RG, Philpot, K, Vicentic, A, Dominguez, G, Hubert, GW & Kuhar, MJ (2004) CART in feeding and obesity. Trends in Endocrinology and Metabolism 15, 454459.CrossRefGoogle ScholarPubMed
Huszar, D, Lynch, CA, Fairchild-Huntress, V, Dunmore, JH, Fang, Q, Berkemeier, LR et al. (1997) Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 88, 131141.CrossRefGoogle ScholarPubMed
Ibrahim, N, Bosch, MA, Smart, JL, Qiu, J, Rubinstein, M, Ronnekleiv, OK, Low, MJ & Kelly, MJ (2003) Hypothalamic proopiomelanocortin neurons are glucose responsive and express K(ATP) channels. Endocrinology 144, 13311340.CrossRefGoogle ScholarPubMed
Ida, T, Nakahara, K, Kuroiwa, T, Fukui, K, Nakazato, M, Murakami, T & Murakami, N (2000) Both corticotropin releasing factor and neuropeptide Y are involved in the effect of orexin (hypocretin) on the food intake in rats. Neuroscience Letters 293, 119122.CrossRefGoogle ScholarPubMed
Iglesias, MJ, Pineiro, R, Blanco, M, Gallego, R, Dieguez, C, Gualillo, O, Gonzalez-Juanatey, JR & Lago, F (2004) Growth hormone releasing peptide (ghrelin) is synthesized and secreted by cardiomyocytes. Cardiovascular Research 62, 481488.CrossRefGoogle ScholarPubMed
Inui, A (1999) Neuropeptide Y feeding receptors: are multiple subtypes involved? Trends in Pharmacological Sciences 20, 4346.CrossRefGoogle ScholarPubMed
Jain, MR, Horvath, TL, Kalra, PS & Kalra, SP (2000) Evidence that NPY Y1 receptors are involved in stimulation of feeding by orexins (hypocretins) in sated rats. Regulatory Peptides 87, 1924.CrossRefGoogle ScholarPubMed
Johren, O, Neidert, SJ, Kummer, M, Dendorfer, A & Dominiak, P (2001) Prepro-orexin and orexin receptor mRNAs are differentially expressed in peripheral tissues of male and female rats. Endocrinology 142, 33243331.CrossRefGoogle ScholarPubMed
Kalia, M & Sullivan, JM (1982) Brainstem projections of sensory and motor components of the vagus nerve in the rat. Journal of Comparative Neurology 211, 248265.CrossRefGoogle ScholarPubMed
Kalra, SP, Dube, MG, Pu, S, Xu, B, Horvath, TL & Kalra, PS (1999) Interacting appetite-regulating pathways in the hypothalamic regulation of body weight. Endocrine Reviews 20, 68100.Google ScholarPubMed
Kalra, SP, Dube, MG, Sahu, A, Phelps, CP & Kalra, PS (1991) Neuropeptide Y secretion increases in the paraventricular nucleus in association with increased appetite for food. Proceedings of the National Academy of Sciences USA 88, 1093110935.CrossRefGoogle ScholarPubMed
Kamegai, J, Tamura, H, Shimizu, T, Ishii, S, Sugihara, H & Wakabayashi, I (2001) Chronic central infusion of ghrelin increases hypothalamic neuropeptide Y and Agouti-related protein mRNA levels and body weight in rats. Diabetes 50, 24382443.CrossRefGoogle ScholarPubMed
Kastin, AJ, Akerstrom, V & Pan, W (2002) Interactions of glucagon-like peptide-1 (GLP-1) with the blood-brain barrier. Journal of Molecular Neuroscience 18, 714.CrossRefGoogle ScholarPubMed
Kawai, Y, Inagaki, S, Shiosaka, S, Shibasaki, T, Ling, N, Tohyama, M & Shiotani, Y (1984) The distribution and projection of gamma-melanocyte stimulating hormone in the rat brain: an immunohistochemical analysis. Brain Research 297, 2132.CrossRefGoogle Scholar
Kim, EK, Miller, I, Landree, LE, Borisy-Rudin, FF, Brown, P, Tihan, T, Townsend, CA, Witters, LA, Moran, TH, Kuhajda, FP & Ronnett, GV (2002) Expression of FAS within hypothalamic neurons: a model for decreased food intake after C75 treatment. American Journal of Physiology 283, E867E879.Google Scholar
Kim, EM, Grace, MK, Welch, CC, Billington, CJ & Levine, AS (1999) STZ-induced diabetes decreases and insulin normalizes POMC mRNA in arcuate nucleus and pituitary in rats. American Journal of Physiology 276, R1320R1326.Google ScholarPubMed
King, BM (2006) The rise, fall, and resurrection of the ventromedial hypothalamus in the regulation of feeding behavior and body weight. Physiology and Behavior 87, 221244.CrossRefGoogle ScholarPubMed
Kirchgessner, AL & Liu, M (1999) Orexin synthesis and response in the gut. Neuron 24, 941951.CrossRefGoogle ScholarPubMed
Kissileff, HR, Pi-Sunyer, FX, Thornton, J & Smith, GP (1981) C-terminal octapeptide of cholecystokinin decreases food intake in man. American Journal of Clinical Nutrition 34, 154160.CrossRefGoogle ScholarPubMed
Koch, JE (2001) Delta(9)-THC stimulates food intake in Lewis rats: effects on chow, high-fat and sweet high-fat diets. Pharmacology, Biochemistry, and Behavior 68, 539543.CrossRefGoogle ScholarPubMed
Kojima, M, Hosoda, H, Date, Y, Nakazato, M, Matsuo, H & Kangawa, K (1999) Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402, 656660.CrossRefGoogle ScholarPubMed
Kola, B, Hubina, E, Tucci, SA, Kirkham, TC, Garcia, EA, Mitchell, SE, Williams, LM, Hawley, SA, Hardie, DG, Grossman, AB & Korbonits, M (2005) Cannabinoids and ghrelin have both central and peripheral metabolic and cardiac effects via AMP-activated protein kinase. Journal of Biological Chemistry 280, 2519625201.CrossRefGoogle ScholarPubMed
Konturek, SJ, Konturek, JW, Pawlik, T & Brzozowski, T (2004) Brain-gut axis and its role in the control of food intake. Journal of Physiology and Pharmacology 55, 137154.Google ScholarPubMed
Kotz, CM, Teske, JA, Levine, JA & Wang, C (2002) Feeding and activity induced by orexin A in the lateral hypothalamus in rats. Regulatory Peptides 104, 2732.CrossRefGoogle ScholarPubMed
Kola, B, Hubina, E, Tucci, SA, Kirkham, TC, Garcia, EA, Mitchell, SE, Williams, LM, Hawley, SA, Hardie, DG, Grossman, AB & Korbonits, M (2005) Cannabinoids and ghrelin have both central and peripheral metabolic and cardiac effects via AMP-activated protein kinase. Journal of Biological Chemistry 280, 2519625201.CrossRefGoogle ScholarPubMed
Koylu, EO, Couceyro, PR, Lambert, PD & Kuhar, MJ (1998) Cocaine- and amphetamine-regulated transcript peptide immunohistochemical localization in the rat brain. Journal of Comparative Neurology 391, 115132.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Kristensen, P, Judge, ME, Thim, L, Ribel, U, Christjansen, KN, Wulff, BS et al. (1998) Hypothalamic CART is a new anorectic peptide regulated by leptin. Nature 393, 7276.CrossRefGoogle ScholarPubMed
Krude, H, Biebermann, H, Luck, W, Horn, R, Brabant, G & Gruters, A (1998) Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nature Genetics 19, 155157.CrossRefGoogle ScholarPubMed
Ladenheim, EE, Moore, KA, Salorio, CF, Mantey, SA, Taylor, JE, Coy, DH, Jensen, RT & Moran, TH (1997) Characterization of bombesin binding sites in the rat stomach. European Journal of Pharmacology 319, 245251.CrossRefGoogle ScholarPubMed
Ladyman, SR & Grattan, DR (2004) Region specific reduction in leptin-induced phosphorylation of STAT3 in the rat hypothalamus is associated with leptin resistance during pregnancy. Endocrinology 145, 37043711.CrossRefGoogle ScholarPubMed
Lam, TK, Pocai, A, Gutierrez-Juarez, R, Obici, S, Bryan, J, Aguilar-Bryan, L, Schwartz, GJ & Rossetti, L (2005 a) Hypothalamic sensing of circulating fatty acids is required for glucose homeostasis. Nature Medicine 11, 320327.CrossRefGoogle ScholarPubMed
Lam, TK, Schwartz, GJ & Rossetti, L (2005 b) Hypothalamic sensing of fatty acids. Nature Neuroscience 8, 579584.CrossRefGoogle ScholarPubMed
Langley, K (1994) The neuroendocrine concept today. Annals of the New York Academy of Sciences 733, 117.CrossRefGoogle ScholarPubMed
Larhammar, D (1996) Structural diversity of receptors for neuropeptide Y, peptide YY and pancreatic polypeptide. Regulatory Peptides 65, 165174.CrossRefGoogle ScholarPubMed
Larsson, LI & Rehfeld, JF (1978) Distribution of gastrin and CCK cells in the rat gastrointestinal tract. Evidence for the occurrence of three distinct cell types storing COOH-terminal gastrin immunoreactivity. Histochemistry 58, 2331.CrossRefGoogle ScholarPubMed
Lassmann, V, Vague, P, Vialettes, B & Simon, MC (1980) Low plasma levels of pancreatic polypeptide in obesity. Diabetes 29, 428430.CrossRefGoogle ScholarPubMed
Lawrence, CB, Liu, YL, Stock, MJ & Luckman, SM (2004) Anorectic actions of prolactin-releasing peptide are mediated by corticotropin-releasing hormone receptors. American Journal of Physiology 286, R101R107.Google ScholarPubMed
Lawrence, CB, Snape, AC, Baudoin, FM & Luckman, SM (2002) Acute central ghrelin and GH secretagogues induce feeding and activate brain appetite centers. Endocrinology 143, 155162.CrossRefGoogle ScholarPubMed
Le Quellec, A, Kervran, A, Blache, P, Ciurana, AJ & Bataille, D (1992) Oxyntomodulin-like immunoreactivity: diurnal profile of a new potential enterogastrone. Journal of Clinical Investigation 74, 14051409.Google ScholarPubMed
Lee, GH, Proenca, R, Montez, JM, Carroll, KM, Darvishzadeh, JG, Lee, JI & Friedman, JM (1996) Abnormal splicing of the leptin receptor in diabetic mice. Nature 379, 632635.CrossRefGoogle ScholarPubMed
Lee, Y, Yang, SP, Soares, MJ & Voogt, JL (2000) Distribution of prolactin-releasing peptide mRNA in the rat brain. Brain Research Bulletin 51, 171176.CrossRefGoogle ScholarPubMed
Legradi, G & Lechan, RM (1999) Agouti-related protein containing nerve terminals innervate thyrotropin- releasing hormone neurons in the hypothalamic paraventricular nucleus. Endocrinology 140, 36433652.CrossRefGoogle ScholarPubMed
Lelliott, CJ, López, M, Curtis, RK, Parker, N, Laudes, M, Yeo, G et al. (2005) Transcript and metabolite analysis of the effects of tamoxifen in rat liver reveals inhibition of fatty acid synthesis in the presence of hepatic steatosis. FASEB Journal 19, 11081119.CrossRefGoogle ScholarPubMed
Levin, BE & Dunn-Meynell, AA (2002) Reduced central leptin sensitivity in rats with diet-induced obesity. American Journal of Physiology 283, R941R948.Google ScholarPubMed
Levin, BE, Dunn-Meynell, AA & Banks, WA (2004) Obesity-prone rats have normal blood-brain barrier transport but defective central leptin signaling before obesity onset. American Journal of Physiology 286, R143R150.Google ScholarPubMed
Li, C, Chen, P & Smith, MS (1998) The acute suckling stimulus induces expression of neuropeptide Y (NPY) in cells in the dorsomedial hypothalamus and increases NPY expression in the arcuate nucleus. Endocrinology 139, 16451652.CrossRefGoogle ScholarPubMed
Lichtman, AH & Cravatt, BF (2005) Food for thought: endocannabinoid modulation of lipogenesis. Journal of Clinical Investigation 115, 11301133.CrossRefGoogle ScholarPubMed
Licinio, J, Caglayan, S, Ozata, M, Yildiz, BO, de Miranda, PB, O'Kirwan, F et al. (2004) Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proceedings of the National Academy of Sciences USA 101, 45314536.CrossRefGoogle ScholarPubMed
Loftus, TM, Jaworsky, DE, Frehywot, GL, Townsend, CA, Ronnett, GV, Lane, MD & Kuhajda, FP (2000) Reduced food intake and body weight in mice treated with fatty acid synthase inhibitors. Science 288, 23792381.CrossRefGoogle ScholarPubMed
López, M, Diéguez, C & Señarís, R (2001 a) The hypocretins/orexins; a new bridge between sleep and feeding. In Recent Research in Endocrinology, pp. 8798 [Pandalai, SG editor]. Kerala, India: Transworld Research Network.Google Scholar
López, M, Lelliott, CJ, Tovar, S, Kimber, W, Gallego, R, Virtue, S et al. (2006) Tamoxifen-induced anorexia is associated with fatty acid synthase inhibition in the ventromedial nucleus of the hypothalamus and accumulation of malonyl-CoA. Diabetes 55, 13271336.CrossRefGoogle ScholarPubMed
López, M, Senarís, R, Gallego, R, García-Caballero, T, Lago, F, Seoane, L, Casanueva, F & Diéguez, C (1999) Orexin receptors are expressed in the adrenal medulla of the rat. Endocrinology 140, 59915994.CrossRefGoogle ScholarPubMed
López, M, Seoane, L, García, MC, Lago, F, Casanueva, FF, Senarís, R & Diéguez, C (2000) Leptin regulation of prepro-orexin and orexin receptor mRNA levels in the hypothalamus. Biochemical and Biophysical Research Communications 269, 4145.CrossRefGoogle ScholarPubMed
López, M, Seoane, LM, García, MC, Diéguez, C & Señarís, R (2002) Neuropeptide Y, but not agouti-related peptide or melanin-concentrating hormone, is a target peptide for orexin-a feeding actions in the rat hypothalamus. Neuroendocrinology 75, 3444.CrossRefGoogle ScholarPubMed
López, M, Seoane, L, Señarís, R & Diéguez, C (2001 b) Prepro-orexin mRNA levels in the rat hypothalamus, and orexin receptors mRNA levels in the rat hypothalamus and adrenal gland are not influenced by the thyroid status. Neuroscience Letters 300, 171175.CrossRefGoogle Scholar
López, M, Seoane, LM, Tovar, S, García, MC, Nogueiras, R, Diéguez, C & Señarís, RM (2005 a) A possible role of neuropeptide Y, agouti-related protein and leptin receptor isoforms in hypothalamic programming by perinatal feeding in the rat. Diabetologia 48, 140148.CrossRefGoogle ScholarPubMed
López, M, Seoane, LM, Tovar, S, Nogueiras, R, Diéguez, C & Señarís, R (2004) Orexin-A regulates growth hormone releasing hormone mRNA content in a nucleus specific manner and somatostatin mRNA content in a growth hormone-dependent fashion in the rat hypothalamus. European Journal of Neuroscience 19, 20802088.CrossRefGoogle Scholar
López, M, Tena-Sempere, M, García-Caballero, T, Señarís, R & Diéguez, C (2005 b) Hypocretins in endocrine regulation. In Hypocretins. Integrators of Physiological Functions, pp. 393421 [de Lecea, L and Sutcliffe, JG editors]. New York: Springer Science.Google Scholar
Ludwig, DS, Tritos, NA, Mastaitis, JW, Kulkarni, R, Kokkotou, E, Elmquist, J, Lowell, B, Flier, JS & Maratos-Flier, E (2001) Melanin-concentrating hormone overexpression in transgenic mice leads to obesity and insulin resistance. Journal of Clinical Investigation 107, 379386.CrossRefGoogle ScholarPubMed
Luquet, S, Perez, FA, Hnasko, TS & Palmiter, RD (2005) NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 310, 683685.CrossRefGoogle ScholarPubMed
Maeda, N, Shimomura, I, Kishida, K, Nishizawa, H, Matsuda, M, Nagaretani, H et al. (2002) Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nature Medicine 8, 731737.CrossRefGoogle ScholarPubMed
Maffei, M, Halaas, J, Ravussin, E, Pratley, RE, Lee, GH, Zhang, Y et al. (1995) Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nature Medicine 1, 11551161.CrossRefGoogle ScholarPubMed
Maggi, CA (1991) The pharmacology of the efferent function of sensory nerves. Journal of Autonomic Pharmacology 11, 173208.CrossRefGoogle ScholarPubMed
Majdic, G, Young, M, Gomez-Sanchez, E, Anderson, P, Szczepaniak, LS, Dobbins, RL, McGarry, JD & Parker, KL (2002) Knockout mice lacking steroidogenic factor 1 are a novel genetic model of hypothalamic obesity. Endocrinology 143, 607614.CrossRefGoogle ScholarPubMed
Makimura, H, Mizuno, TM, Mastaitis, JW, Agami, R & Mobbs, CV (2002) Reducing hypothalamic AGRP by RNA interference increases metabolic rate and decreases body weight without influencing food intake. BMC Neuroscience 3, 18.CrossRefGoogle ScholarPubMed
Malaisse-Lagae, F, Carpentier, JL, Patel, YC, Malaisse, WJ & Orci, L (1977) Pancreatic polypeptide: a possible role in the regulation of food intake in the mouse. Hypothesis. Experientia 33, 915917.CrossRefGoogle ScholarPubMed
Marcus, JN, Aschkenasi, CJ, Lee, CE, Chemelli, RM, Saper, CB, Yanagisawa, M & Elmquist, JK (2001) Differential expression of orexin receptors 1 and 2 in the rat brain. Journal of Comparative Neurology 435, 625.CrossRefGoogle ScholarPubMed
Marks, DL, Boucher, N, Lanouette, CM, Perusse, L, Brookhart, G, Comuzzie, AG, Chagnon, YC & Cone, RD (2004) Ala67Thr polymorphism in the Agouti-related peptide gene is associated with inherited leanness in humans. American Journal of Medical Genetics 126 A, 267271.CrossRefGoogle Scholar
Marks, JL, Porte, D Jr, Stahl, WL & Baskin, DG (1990) Localization of insulin receptor mRNA in rat brain by in situ hybridization. Endocrinology 127, 32343236.CrossRefGoogle ScholarPubMed
Marsh, DJ, Weingarth, DT, Novi, DE, Chen, HY, Trumbauer, ME, Chen, AS et al. (2002) Melanin-concentrating hormone 1 receptor-deficient mice are lean, hyperactive, and hyperphagic and have altered metabolism. Proceedings of the National Academy of Sciences USA 99, 32403245.CrossRefGoogle ScholarPubMed
Masuzaki, H, Ogawa, Y, Sagawa, N, Hosoda, K, Matsumoto, T, Mise, H, Nishimura, H, Yoshimasa, Y, Tanaka, I, Mori, T & Nakao, K (1997) Nonadipose tissue production of leptin: leptin as a novel placenta-derived hormone in humans. Nature Medicine 3, 10291033.CrossRefGoogle ScholarPubMed
Matsuda, LA, Lolait, SJ, Brownstein, MJ, Young, AC & Bonner, TI (1990) Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 346, 561564.CrossRefGoogle ScholarPubMed
Meeran, K, O'Shea, D, Edwards, CM, Turton, MD, Heath, MM, Gunn, I et al. (1999) Repeated intracerebroventricular administration of glucagon-like peptide-1-(7–36) amide or exendin-(9–39) alters body weight in the rat. Endocrinology 140, 244250.CrossRefGoogle ScholarPubMed
Meier, JJ, Gallwitz, B, Schmidt, WE & Nauck, MA (2002) Glucagon-like peptide 1 as a regulator of food intake and body weight: therapeutic perspectives. European Journal of Pharmacology 440, 269279.CrossRefGoogle ScholarPubMed
Mercer, JG, Hoggard, N, Williams, LM, Lawrence, CB, Hannah, LT, Morgan, PJ & Trayhurn, P (1996) Coexpression of leptin receptor and preproneuropeptide Y mRNA in arcuate nucleus of mouse hypothalamus. Journal of Neuroendocrinology 8, 733735.CrossRefGoogle ScholarPubMed
Mercer, JG, Moar, KM & Hoggard, N (1998) Localization of leptin receptor (Ob-R) messenger ribonucleic acid in the rodent hindbrain. Endocrinology 139, 2934.CrossRefGoogle ScholarPubMed
Minokoshi, Y, Alquier, T, Furukawa, N, Kim, YB, Lee, A, Xue, B et al. (2004) AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 428, 569574.CrossRefGoogle ScholarPubMed
Miyawaki, K, Yamada, Y, Ban, N, Ihara, Y, Tsukiyama, K, Zhou, H et al. (2002) Inhibition of gastric inhibitory polypeptide signaling prevents obesity. Nature Medicine 8, 738742.CrossRefGoogle ScholarPubMed
Moltz, JH & McDonald, JK (1985) Neuropeptide Y: direct and indirect action on insulin secretion in the rat. Peptides 6, 11551159.CrossRefGoogle ScholarPubMed
Montague, CT, Farooqi, IS, Whitehead, JP, Soos, MA, Rau, H, Wareham, NJ et al. (1997) Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387, 903908.CrossRefGoogle ScholarPubMed
Moran, TH, Baldessarini, AR, Salorio, CF, Lowery, T & Schwartz, GJ (1997) Vagal afferent and efferent contributions to the inhibition of food intake by cholecystokinin. American Journal of Physiology 272, R1245R1251.Google Scholar
Moran, TH, Norgren, R, Crosby, RJ & McHugh, PR (1990) Central and peripheral vagal transport of cholecystokinin binding sites occurs in afferent fibers. Brain Research 526, 95102.CrossRefGoogle ScholarPubMed
Moran, TH, Robinson, PH, Goldrich, MS & McHugh, PR (1986) Two brain cholecystokinin receptors: implications for behavioral actions. Brain Research 362, 175179.CrossRefGoogle ScholarPubMed
Moran, TH, Smedh, U, Kinzig, KP, Scott, KA, Knipp, S & Ladenheim, EE (2005) Peptide YY(3–36) inhibits gastric emptying and produces acute reductions in food intake in rhesus monkeys. American Journal of Physiology 288, R384R388.Google ScholarPubMed
Morgan, K, Obici, S & Rossetti, L (2004) Hypothalamic responses to long-chain fatty acids are nutritionally regulated. Journal of Biological Chemistry 279, 3113931148.CrossRefGoogle ScholarPubMed
Moriguchi, T, Sakurai, T, Nambu, T, Yanagisawa, M & Goto, K (1999) Neurons containing orexin in the lateral hypothalamic area of the adult rat brain are activated by insulin-induced acute hypoglycemia. Neuroscience Letters 264, 101104.CrossRefGoogle ScholarPubMed
Morton, GJ, Cummings, DE, Baskin, DG, Barsh, GS & Schwartz, MW (2006) Central nervous system control of food intake and body weight. Nature 443, 289295.CrossRefGoogle ScholarPubMed
Mountjoy, KG, Mortrud, MT, Low, MJ, Simerly, RB & Cone, RD (1994) Localization of the melanocortin-4 receptor (MC4-R) in neuroendocrine and autonomic control circuits in the brain. Molecular Endocrinology 8, 12981308.Google ScholarPubMed
Munro, S, Thomas, KL & Abu-Shaar, M (1993) Molecular characterization of a peripheral receptor for cannabinoids. Nature 365, 6165.CrossRefGoogle ScholarPubMed
Munzberg, H, Flier, JS & Bjorbaek, C (2004) Region-specific leptin resistance within the hypothalamus of diet-induced-obese mice. Endocrinology 145, 48804889.CrossRefGoogle ScholarPubMed
Munzberg, H & Myers, MG (2005) Molecular and anatomical determinants of central leptin resistance. Nature Neuroscience 8, 566570.CrossRefGoogle ScholarPubMed
Muurahainen, N, Kissileff, HR, DeRogatis, AJ & Pi-Sunyer, FX (1988) Effects of cholecystokinin-octapeptide (CCK-8) on food intake and gastric emptying in man. Physiology and Behavior 44, 645649.CrossRefGoogle ScholarPubMed
Nakazato, M, Murakami, N, Date, Y, Kojima, M, Matsuo, H, Kangawa, K & Matsukura, S (2001) A role for ghrelin in the central regulation of feeding. Nature 409, 194198.CrossRefGoogle ScholarPubMed
Naslund, E, Barkeling, B, King, N, Gutniak, M, Blundell, JE, Holst, JJ, Rossner, S & Hellstrom, PM (1999) Energy intake and appetite are suppressed by glucagon-like peptide-1 (GLP-1) in obese men. International Journal of Obesity and Related Metabolic Disorders 23, 304311.CrossRefGoogle ScholarPubMed
Nogueiras, R, Barreiro, ML, Caminos, JE, Gaytan, F, Suominen, JS, Navarro, VM et al. (2004 a) Novel expression of resistin in rat testis: functional role and regulation by nutritional status and hormonal factors. Journal of Cell Science 117, 32473257.CrossRefGoogle ScholarPubMed
Nogueiras, R, Gallego, R, Gualillo, O, Caminos, JE, Garcia-Caballero, T, Casanueva, FF & Dieguez, C (2003 a) Resistin is expressed in different rat tissues and is regulated in a tissue- and gender-specific manner. FEBS Letters 548, 2127.CrossRefGoogle Scholar
Nogueiras, R, Gualillo, O, Caminos, JE, Casanueva, FF & Diéguez, C (2003 b) Regulation of resistin by gonadal, thyroid hormone, and nutritional status. Obesity Research 11, 408414.CrossRefGoogle ScholarPubMed
Nogueiras, R, Pfluger, P, Tovar, S, Myrtha, A, Mitchell, S, Morris, A et al. (2006) Effects of obestatin on energy balance and growth hormone secretion in rodents. Endocrinology (Epublication ahead of print version).Google ScholarPubMed
Nogueiras, R, Tovar, S, Mitchell, SE, Rayner, DV, Archer, ZA, Diéguez, C & Williams, LM (2004 b) Regulation of growth hormone secretagogue receptor gene expression in the arcuate nuclei of the rat by leptin and ghrelin. Diabetes 53, 25522558.CrossRefGoogle ScholarPubMed
Nogueiras, R & Tschop, M (2005) Biomedicine. Separation of conjoined hormones yields appetite rivals. Science 310, 985986.CrossRefGoogle ScholarPubMed
Nonaka, N, Shioda, S, Niehoff, ML & Banks, WA (2003) Characterization of blood-brain barrier permeability to PYY3–36 in the mouse. Journal of Pharmacology and Experimental Therapeutics 306, 948953.CrossRefGoogle ScholarPubMed
Obici, S, Feng, Z, Karkanias, G, Baskin, DG & Rossetti, L (2002 a) Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats. Nature Neuroscience 5, 566572.CrossRefGoogle ScholarPubMed
Obici, S, Feng, Z, Morgan, K, Stein, D, Karkanias, G & Rossetti, L (2002 b) Central administration of oleic acid inhibits glucose production and food intake. Diabetes 51, 271275.CrossRefGoogle ScholarPubMed
Obici, S, Feng, Z, Tan, J, Liu, L, Karkanias, G & Rossetti, L (2001) Central melanocortin receptors regulate insulin action. Journal of Clinical Investigation 108, 10791085.CrossRefGoogle ScholarPubMed
Ohki-Hamazaki, H, Watase, K, Yamamoto, K, Ogura, H, Yamano, M, Yamada, K, Maeno, H, Imaki, J, Kikuyama, S, Wada, E & Wada, K (1997) Mice lacking bombesin receptor subtype-3 develop metabolic defects and obesity. Nature 390, 165169.CrossRefGoogle ScholarPubMed
Ollmann, MM, Wilson, BD, Yang, YK, Kerns, JA, Chen, Y, Gantz, I & Barsh, GS (1997) Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 278, 135138.CrossRefGoogle ScholarPubMed
Pajvani, UB, Du, X, Combs, TP, Berg, AH, Rajala, MW, Schulthess, T, Engel, J, Brownlee, M & Scherer, PE (2003) Structure-function studies of the adipocyte-secreted hormone Acrp30/adiponectin. Implications for metabolic regulation and bioactivity. Journal of Biological Chemistry 278, 90739085.CrossRefGoogle ScholarPubMed
Pelleymounter, MA, Cullen, MJ, Baker, MB, Hecht, R, Winters, D, Boone, T & Collins, F (1995) Effects of the obese gene product on body weight regulation in ob/ob mice. Science 269, 540543.CrossRefGoogle ScholarPubMed
Perez-Tilve, D, Nogueiras, R, Mallo, F, Benoit, SC & Tschoep, M (2006) Gut hormones ghrelin, PYY, and GLP-1 in the regulation of energy, balance, and metabolism. Endocrine 29, 6172.CrossRefGoogle Scholar
Pi-Sunyer, X, Kissileff, HR, Thornton, J & Smith, GP (1982) C-terminal octapeptide of cholecystokinin decreases food intake in obese men. Physiology and Behavior 29, 627630.CrossRefGoogle ScholarPubMed
Pineiro, R, Iglesias, MJ, Gallego, R, Raghay, K, Eiras, S, Rubio, J, Dieguez, C, Gualillo, O, Gonzalez-Juanatey, JR & Lago, F (2005) Adiponectin is synthesized and secreted by human and murine cardiomyocytes. FEBS Letters 579, 51635169.CrossRefGoogle ScholarPubMed
Pinto, S, Roseberry, AG, Liu, H, Diano, S, Shanabrough, M, Cai, X, Friedman, JM & Horvath, TL (2004) Rapid rewiring of arcuate nucleus feeding circuits by leptin. Science 304, 110115.CrossRefGoogle ScholarPubMed
Pocai, A, Lam, TK, Obici, S, Gutierrez-Juarez, R, Muse, ED, Arduini, A & Rossetti, L (2006) Restoration of hypothalamic lipid sensing normalizes energy and glucose homeostasis in overfed rats. Journal of Clinical Investigation 116, 10811091.CrossRefGoogle ScholarPubMed
Qi, Y, Takahashi, N, Hileman, SM, Patel, HR, Berg, AH, Pajvani, UB, Scherer, PE & Ahima, RS (2004) Adiponectin acts in the brain to decrease body weight. Nature Medicine 10, 524529.CrossRefGoogle ScholarPubMed
Qian, S, Chen, H, Weingarth, D, Trumbauer, ME, Novi, DE, Guan, X et al. (2002) Neither agouti-related protein nor neuropeptide Y is critically required for the regulation of energy homeostasis in mice. Molecular and Cellular Biology 22, 50275035.CrossRefGoogle ScholarPubMed
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. Nature 380, 243247.CrossRefGoogle ScholarPubMed
Reidelberger, RD, Hernandez, J, Fritzsch, B & Hulce, M (2004) Abdominal vagal mediation of the satiety effects of CCK in rats. American Journal of Physiology 286, R1005R1012.Google ScholarPubMed
Reidelberger, RD & Solomon, TE (1986) Comparative effects of CCK-8 on feeding, sham feeding, and exocrine pancreatic secretion in rats. American Journal of Physiology 251, R97R105.Google ScholarPubMed
Roselli, CE, Jorgensen, EZ, Doyle, MW & Ronnekleiv, OK (1997) Expression of the orphan receptor steroidogenic factor-1 mRNA in the rat medial basal hypothalamus. Brain Research Molecular Brain Research 44, 6672.CrossRefGoogle ScholarPubMed
Sakata, I, Nakamura, K, Yamazaki, M, Matsubara, M, Hayashi, Y, Kangawa, K & Sakai, T (2002) Ghrelin-producing cells exist as two types of cells, closed- and opened-type cells, in the rat gastrointestinal tract. Peptides 23, 531536.CrossRefGoogle ScholarPubMed
Sakurai, T, Amemiya, A, Ishii, M, Matsuzaki, I, Chemelli, RM, Tanaka, H et al. (1998) Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92, 573585.CrossRefGoogle Scholar
Sanacora, G, Kershaw, M, Finkelstein, JA & White, JD (1990) Increased hypothalamic content of preproneuropeptide Y messenger ribonucleic acid in genetically obese Zucker rats and its regulation by food deprivation. Endocrinology 127, 730737.CrossRefGoogle ScholarPubMed
Saper, CB, Chou, TC & Elmquist, JK (2002) The need to feed: homeostatic and hedonic control of eating. Neuron 36, 199211.CrossRefGoogle ScholarPubMed
Savage, DB, Sewter, CP, Klenk, ES, Segal, DG, Vidal-Puig, A, Considine, RV & O'Rahilly, S (2001) Resistin/Fizz3 expression in relation to obesity and peroxisome proliferator-activated receptor-gamma action in humans. Diabetes 50, 21992202.CrossRefGoogle ScholarPubMed
Savontaus, E, Conwell, IM & Wardlaw, SL (2002) Effects of adrenalectomy on AGRP, POMC, NPY and CART gene expression in the basal hypothalamus of fed and fasted rats. Brain Research 958, 130138.CrossRefGoogle ScholarPubMed
Sawchenko, PE (1983) Central connections of the sensory and motor nuclei of the vagus nerve. Journal of the Autonomic Nervous System 9, 1326.CrossRefGoogle ScholarPubMed
Sawchenko, PE & Swanson, LW (1983) The organization and biochemical specificity of afferent projections to the paraventricular and supraoptic nuclei. Progress in Brain Research 60, 1929.CrossRefGoogle Scholar
Sawchenko, PE, Swanson, LW, Grzanna, R, Howe, PR, Bloom, SR & Polak, JM (1985) Colocalization of neuropeptide Y immunoreactivity in brainstem catecholaminergic neurons that project to the paraventricular nucleus of the hypothalamus. Journal of Comparative Neurology 241, 138153.CrossRefGoogle Scholar
Scherer, PE, Williams, S, Fogliano, M, Baldini, G & Lodish, HF (1995) A novel serum protein similar to C1q, produced exclusively in adipocytes. Journal of Biological Chemistry 270, 2674626749.CrossRefGoogle ScholarPubMed
Schmidt, PT, Naslund, E, Gryback, P, Jacobsson, H, Hartmann, B, Holst, JJ & Hellstrom, PM (2003) Peripheral administration of GLP-2 to humans has no effect on gastric emptying or satiety. Regulatory Peptides 116, 2125.CrossRefGoogle ScholarPubMed
Schobitz, B, De Kloet, ER, Sutanto, W & Holsboer, F (1993) Cellular localization of interleukin 6 mRNA and interleukin 6 receptor mRNA in rat brain. European Journal of Neuroscience 5, 14261435.CrossRefGoogle ScholarPubMed
Schwartz, GJ & Moran, TH (1994) CCK elicits and modulates vagal afferent activity arising from gastric and duodenal sites. Annals of the New York Academy of Sciences 713, 121128.CrossRefGoogle ScholarPubMed
Schwartz, MW, Figlewicz, DP, Baskin, DG, Woods, SC & Porte, D Jr (1992 a) Insulin in the brain: a hormonal regulator of energy balance. Endocrine Reviews 13, 387414.Google Scholar
Schwartz, MW, Seeley, RJ, Woods, SC, Weigle, DS, Campfield, LA, Burn, P & Baskin, DG (1997) Leptin increases hypothalamic pro-opiomelanocortin mRNA expression in the rostral arcuate nucleus. Diabetes 46, 21192123.CrossRefGoogle ScholarPubMed
Schwartz, MW, Sipols, AJ, Marks, JL, Sanacora, G, White, JD, Scheurink, A et al. (1992 b) Inhibition of hypothalamic neuropeptide Y gene expression by insulin. Endocrinology 130, 36083616.CrossRefGoogle ScholarPubMed
Schwartz, MW, Woods, SC, Porte, D Jr, Seeley, RJ & Baskin, DG (2000) Central nervous system control of food intake. Nature 404, 661671.CrossRefGoogle ScholarPubMed
Scrocchi, LA, Brown, TJ & Drucker, DJ (1997) Leptin sensitivity in nonobese glucagon-like peptide I receptor -/- mice. Diabetes 46, 20292034.CrossRefGoogle ScholarPubMed
Scrocchi, LA, Brown, TJ, Maclusky, N, Brubaker, PL, Auerbach, AB, Joyner, AL & Drucker, DJ (1996) Glucose intolerance but normal satiety in mice with a null mutation in the glucagon-like peptide 1 receptor gene. Nature Medicine 2, 12541258.CrossRefGoogle Scholar
Scrocchi, LA, Hill, ME, Saleh, J, Perkins, B & Drucker, DJ (2000) Elimination of glucagon-like peptide 1R signaling does not modify weight gain and islet adaptation in mice with combined disruption of leptin and GLP-1 action. Diabetes 49, 15521560.CrossRefGoogle Scholar
Seal, LJ, Small, CJ, Dhillo, WS, Stanley, SA, Abbott, CR, Ghatei, MA & Bloom, SR (2001) PRL-releasing peptide inhibits food intake in male rats via the dorsomedial hypothalamic nucleus and not the paraventricular hypothalamic nucleus. Endocrinology 142, 42364243.CrossRefGoogle Scholar
Seeber, RM, Smith, JT & Waddell, BJ (2002) Plasma leptin-binding activity and hypothalamic leptin receptor expression during pregnancy and lactation in the rat. Biology of Reproduction 66, 17621767.CrossRefGoogle ScholarPubMed
Segal-Lieberman, G, Bradley, RL, Kokkotou, E, Carlson, M, Trombly, DJ, Wang, X, Bates, S, Myers, MG Jr, Flier, JS & Maratos-Flier, E (2003) Melanin-concentrating hormone is a critical mediator of the leptin-deficient phenotype. Proceedings of the National Academy of Sciences USA 100, 1008510090.CrossRefGoogle ScholarPubMed
Señarís, R, García-Caballero, T, Casabiell, X, Gallego, R, Castro, R, Considine, RV, Diéguez, C & Casanueva, FF (1997) Synthesis of leptin in human placenta. Endocrinology 138, 45014504.CrossRefGoogle ScholarPubMed
Seoane, LM, López, M, Tovar, S, Casanueva, F, Señarís, R & Diéguez, C (2003) Agouti-related peptide, neuropeptide Y, and somatostatin-producing neurons are targets for ghrelin actions in the rat hypothalamus. Endocrinology 144, 544551.CrossRefGoogle ScholarPubMed
Seoane, LM, Tovar, S, Baldelli, R, Arvat, E, Ghigo, E, Casanueva, FF & Diéguez, C (2000) Ghrelin elicits a marked stimulatory effect on GH secretion in freely-moving rats. European Journal of Endocrinology 143, R7R9.CrossRefGoogle ScholarPubMed
Seoane, LM, Tovar, S, Pérez, D, Mallo, F, López, M, Señarís, R, Casanueva, FF & Diéguez, C (2004) Orexin A suppresses in vivo GH secretion. European Journal of Endocrinology 150, 731736.CrossRefGoogle ScholarPubMed
Sergeyev, V, Broberger, C, Gorbatyuk, O & Hokfelt, T (2000) Effect of 2-mercaptoacetate and 2-deoxy-D-glucose administration on the expression of NPY, AGRP, POMC, MCH and hypocretin/orexin in the rat hypothalamus. Neuroreport 11, 117121.CrossRefGoogle ScholarPubMed
Shimada, M, Tritos, NA, Lowell, BB, Flier, JS & Maratos-Flier, E (1998) Mice lacking melanin-concentrating hormone are hypophagic and lean. Nature 396, 670674.CrossRefGoogle ScholarPubMed
Shimokawa, T, Kumar, MV & Lane, MD (2002) Effect of a fatty acid synthase inhibitor on food intake and expression of hypothalamic neuropeptides. Proceedings of the National Academy of Sciences USA 99, 6671.CrossRefGoogle ScholarPubMed
Shintani, M, Ogawa, Y, Ebihara, K, Aizawa-Abe, M, Miyanaga, F, Takaya, K et al. (2001) Ghrelin, an endogenous growth hormone secretagogue, is a novel orexigenic peptide that antagonizes leptin action through the activation of hypothalamic neuropeptide Y/Y1 receptor pathway. Diabetes 50, 227232.CrossRefGoogle ScholarPubMed
Shughrue, PJ, Lane, MV & Merchenthaler, I (1996) Glucagon-like peptide-1 receptor (GLP1-R) mRNA in the rat hypothalamus. Endocrinology 137, 51595162.CrossRefGoogle ScholarPubMed
Shughrue, PJ, Lane, MV & Merchenthaler, I (1997) Comparative distribution of estrogen receptor-alpha and -beta mRNA in the rat central nervous system. Journal of Comparative Neurology 388, 507525.3.0.CO;2-6>CrossRefGoogle ScholarPubMed
Sipols, AJ, Baskin, DG & Schwartz, MW (1995) Effect of intracerebroventricular insulin infusion on diabetic hyperphagia and hypothalamic neuropeptide gene expression. Diabetes 44, 147151.CrossRefGoogle ScholarPubMed
Small, CJ & Bloom, SR (2004) Gut hormones and the control of appetite. Trends in Endocrinology and Metabolism 15, 259263.CrossRefGoogle ScholarPubMed
Smith, GP, Gibbs, J, Jerome, C, Pi-Sunyer, FX, Kissileff, HR & Thornton, J (1981 a) The satiety effect of cholecystokinin: a progress report. Peptides 2, Suppl. 2, 5759.CrossRefGoogle ScholarPubMed
Smith, GP, Jerome, C & Gibbs, J (1981 b) Abdominal vagotomy does not block the satiety effect of bombesin in the rat. Peptides 2, 409411.CrossRefGoogle Scholar
Spranger, J, Verma, S, Gohring, I, Bobbert, T, Seifert, J, Sindler, AL, Pfeiffer, A, Hileman, SM, Tschop, M & Banks, WA (2006) Adiponectin does not cross the blood-brain barrier but modifies cytokine expression of brain endothelial cells. Diabetes 55, 141147.CrossRefGoogle Scholar
Stanley, BG, Daniel, DR, Chin, AS & Leibowitz, SF (1985) Paraventricular nucleus injections of peptide YY and neuropeptide Y preferentially enhance carbohydrate ingestion. Peptides 6, 12051211.CrossRefGoogle ScholarPubMed
Stanley, BG, Kyrkouli, SE, Lampert, S & Leibowitz, SF (1986) Neuropeptide Y chronically injected into the hypothalamus: a powerful neurochemical inducer of hyperphagia and obesity. Peptides 7, 11891192.CrossRefGoogle ScholarPubMed
Stanley, S, Wynne, K, McGowan, B & Bloom, S (2005) Hormonal regulation of food intake. Physiological Reviews 85, 11311158.CrossRefGoogle ScholarPubMed
Steininger, TL, Kilduff, TS, Behan, M, Benca, RM & Landry, CF (2004) Comparison of hypocretin/orexin and melanin-concentrating hormone neurons and axonal projections in the embryonic and postnatal rat brain. Journal of Chemical Neuroanatomy 27, 165181.CrossRefGoogle ScholarPubMed
Stephens, TW, Basinski, M, Bristow, PK, Bue-Valleskey, JM, Burgett, SG, Craft, L et al. (1995) The role of neuropeptide Y in the antiobesity action of the obese gene product. Nature 377, 530532.CrossRefGoogle ScholarPubMed
Steppan, CM, Bailey, ST, Bhat, S, Brown, EJ, Banerjee, RR, Wright, CM, Patel, HR, Ahima, RS & Lazar, MA (2001 a) The hormone resistin links obesity to diabetes. Nature 409, 307312.CrossRefGoogle ScholarPubMed
Steppan, CM, Brown, EJ, Wright, CM, Bhat, S, Banerjee, RR, Dai, CY, Enders, GH, Silberg, DG, Wen, X, Wu, GD & Lazar, MA (2001 b) A family of tissue-specific resistin-like molecules. Proceedings of the National Academy of Sciences USA 98, 502506.CrossRefGoogle ScholarPubMed
Steppan, CM & Lazar, MA (2002) Resistin and obesity-associated insulin resistance. Trends in Endocrinology and Metabolism 13, 1823.CrossRefGoogle ScholarPubMed
Sternson, SM, Shepherd, GM & Friedman, JM (2005) Topographic mapping of VMH – arcuate nucleus microcircuits and their reorganization by fasting. Nature Neuroscience 8, 13561363.CrossRefGoogle ScholarPubMed
Strobel, A, Issad, T, Camoin, L, Ozata, M & Strosberg, AD (1998) A leptin missense mutation associated with hypogonadism and morbid obesity. Nature Genetics 18, 213215.CrossRefGoogle ScholarPubMed
Sul, HS (2004) Resistin/ADSF/FIZZ3 in obesity and diabetes. Trends in Endocrinology and Metabolism 15, 247249.Google ScholarPubMed
Sun, Y, Ahmed, S & Smith, RG (2003) Deletion of ghrelin impairs neither growth nor appetite. Molecular and Cellular Biology 23, 79737981.CrossRefGoogle ScholarPubMed
Sun, Y, Asnicar, M, Saha, PK, Chan, L & Smith, RG (2006) Ablation of ghrelin improves the diabetic but not obese phenotype of ob/ob mice. Cell Metabolism 3, 379386.CrossRefGoogle Scholar
Sun, Y, Wang, P, Zheng, H & Smith, RG (2004) Ghrelin stimulation of growth hormone release and appetite is mediated through the growth hormone secretagogue receptor. Proceedings of the National Academy of Sciences USA 101, 46794684.CrossRefGoogle ScholarPubMed
Sutcliffe, JG & de Lecea, L (2002) The hypocretins: setting the arousal threshold. Nature Reviews Neuroscience 3, 339349.CrossRefGoogle ScholarPubMed
Swart, I, Jahng, JW, Overton, JM & Houpt, TA (2002) Hypothalamic NPY, AGRP, and POMC mRNA responses to leptin and refeeding in mice. American Journal of Physiology 283, R1020R1026.Google ScholarPubMed
Szczypka, MS, Kwok, K, Brot, MD, Marck, BT, Matsumoto, AM, Donahue, BA & Palmiter, RD (2001) Dopamine production in the caudate putamen restores feeding in dopamine-deficient mice. Neuron 30, 819828.CrossRefGoogle ScholarPubMed
Taheri, S, Zeitzer, JM & Mignot, E (2002) The role of hypocretins (orexins) in sleep regulation and narcolepsy. Annual Review of Neuroscience 25, 283313.CrossRefGoogle ScholarPubMed
Tamura, H, Kamegai, J, Shimizu, T, Ishii, S, Sugihara, H & Oikawa, S (2002) Ghrelin stimulates GH but not food intake in arcuate nucleus ablated rats. Endocrinology 143, 32683275.CrossRefGoogle Scholar
Tang-Christensen, M, Vrang, N & Larsen, PJ (2001) Glucagon-like peptide containing pathways in the regulation of feeding behaviour. International Journal of Obesity and Related Metabolic Disorders 25, Suppl. 5, S42S47.CrossRefGoogle ScholarPubMed
Tannenbaum, GS, Lapointe, M, Beaudet, A & Howard, AD (1998) Expression of growth hormone secretagogue-receptors by growth hormone-releasing hormone neurons in the mediobasal hypothalamus. Endocrinology 139, 44204423.CrossRefGoogle ScholarPubMed
Tartaglia, LA (1997) The leptin receptor. Journal of Biological Chemistry 272, 60936096.CrossRefGoogle ScholarPubMed
Tartaglia, LA, Dembski, M, Weng, X, Deng, N, Culpepper, J, Devos, R et al. (1995) Identification and expression cloning of a leptin receptor, OB-R. Cell 83, 12631271.CrossRefGoogle ScholarPubMed
Tena-Sempere, M, Barreiro, ML, Gonzalez, LC, Gaytan, F, Zhang, FP, Caminos, JE, Pinilla, L, Casanueva, FF, Dieguez, C & Aguilar, E (2002) Novel expression and functional role of ghrelin in rat testis. Endocrinology 143, 717725.CrossRefGoogle ScholarPubMed
Thiele, TE, van Dijk, G, Campfield, LA, Smith, FJ, Burn, P, Woods, SC, Bernstein, IL & Seeley, RJ (1997) Central infusion of GLP-1, but not leptin, produces conditioned taste aversions in rats. American Journal of Physiology 272, R726R730.Google Scholar
Toshinai, K, Date, Y, Murakami, N, Shimada, M, Mondal, MS, Shimbara, T et al. (2003) Ghrelin-induced food intake is mediated via the orexin pathway. Endocrinology 144, 15061512.CrossRefGoogle ScholarPubMed
Tovar, S, Nogueiras, R, Tung, LY, Castaneda, TR, Vázquez, MJ, Morris, A, Williams, LM, Dickson, SL & Diéguez, C (2005) Central administration of resistin promotes short-term satiety in rats. European Journal of Endocrinology 153, R1R5.CrossRefGoogle ScholarPubMed
Tritos, NA, Mastaitis, JW, Kokkotou, E & Maratos-Flier, E (2001) Characterization of melanin concentrating hormone and preproorexin expression in the murine hypothalamus. Brain Research 895, 160166.CrossRefGoogle ScholarPubMed
Tritos, NA, Segal-Lieberman, G, Vezeridis, PS & Maratos-Flier, E (2004) Estradiol-induced anorexia is independent of leptin and melanin-concentrating hormone. Obesity Research 12, 716724.CrossRefGoogle ScholarPubMed
Tsao, TS, Lodish, HF & Fruebis, J (2002) ACRP30, a new hormone controlling fat and glucose metabolism. European Journal of Pharmacology 440, 213221.CrossRefGoogle ScholarPubMed
Tschop, M, Castaneda, TR, Joost, HG, Thone-Reineke, C, Ortmann, S, Klaus, S et al. (2004) Physiology: does gut hormone PYY3–36 decrease food intake in rodents? Nature 430, 1.CrossRefGoogle ScholarPubMed
Tschop, M, Smiley, DL & Heiman, ML (2000) Ghrelin induces adiposity in rodents. Nature 407, 908913.CrossRefGoogle ScholarPubMed
Tschop, M, Wawarta, R, Riepl, RL, Friedrich, S, Bidlingmaier, M, Landgraf, R & Folwaczny, C (2001 a) Post-prandial decrease of circulating human ghrelin levels. Journal of Endocrinological Investigation 24, RC19RC21.CrossRefGoogle ScholarPubMed
Tschop, M, Weyer, C, Tataranni, PA, Devanarayan, V, Ravussin, E & Heiman, ML (2001 b) Circulating ghrelin levels are decreased in human obesity. Diabetes 50, 707709.CrossRefGoogle ScholarPubMed
Turton, MD, O'Shea, D, Gunn, I, Beak, SA, Edwards, CM, Meeran, K et al. (1996) A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379, 6972.CrossRefGoogle ScholarPubMed
Uttenthal, LO, Toledano, A & Blazquez, E (1992) Autoradiographic localization of receptors for glucagon-like peptide-1 (7–36) amide in rat brain. Neuropeptides 21, 143146.CrossRefGoogle ScholarPubMed
Vaisse, C, Halaas, JL, Horvath, CM, Darnell, JE Jr, Stoffel, M & Friedman, JM (1996) Leptin activation of Stat3 in the hypothalamus of wild-type and ob/ob mice but not db/db mice. Nature Genetics 14, 9597.CrossRefGoogle Scholar
Valsamakis, G, McTernan, PG, Chetty, R, Al Daghri, N, Field, A, Hanif, W, Barnett, AH & Kumar, S (2004) Modest weight loss and reduction in waist circumference after medical treatment are associated with favorable changes in serum adipocytokines. Metabolism 53, 430434.CrossRefGoogle ScholarPubMed
Van Heek, M, Compton, DS, France, CF, Tedesco, RP, Fawzi, AB, Graziano, MP, Sybertz, EJ, Strader, CD & Davis, HRJ (1997) Diet-induced obese mice develop peripheral, but not central, resistance to leptin. Journal of Clinical Investigation 99, 385390.CrossRefGoogle Scholar
Verdich, C, Flint, A, Gutzwiller, JP, Naslund, E, Beglinger, C, Hellstrom, PM, Long, SJ, Morgan, LM, Holst, JJ & Astrup, A (2001) A meta-analysis of the effect of glucagon-like peptide-1 (7–36) amide on ad libitum energy intake in humans. Journal of Clinical Endocrinology and Metabolism 86, 43824389.Google ScholarPubMed
Verty, AN, McFarlane, JR, McGregor, IS & Mallet, PE (2004) Evidence for an interaction between CB1 cannabinoid and melanocortin MCR-4 receptors in regulating food intake. Endocrinology 145, 32243231.CrossRefGoogle ScholarPubMed
Vidal-Puig, A & O'Rahilly, S (2001) Resistin: a new link between obesity and insulin resistance? Clinical Endocrinology 55, 437438.CrossRefGoogle ScholarPubMed
Wallenius, K, Wallenius, V, Sunter, D, Dickson, SL & Jansson, JO (2002 a) Intracerebroventricular interleukin-6 treatment decreases body fat in rats. Biochemical and Biophysical Research Communications 293, 560565.CrossRefGoogle ScholarPubMed
Wallenius, V, Wallenius, K, Ahren, B, Rudling, M, Carlsten, H, Dickson, SL, Ohlsson, C & Jansson, JO (2002 b) Interleukin-6-deficient mice develop mature-onset obesity. Nature Medicine 8, 7579.CrossRefGoogle ScholarPubMed
Wang, C & Kotz, CM (2002) Urocortin in the lateral septal area modulates feeding induced by orexin A in the lateral hypothalamus. American Journal of Physiology 283, R358R367.Google ScholarPubMed
Wank, SA, Harkins, R, Jensen, RT, Shapira, H, de Weerth, A & Slattery, T (1992 a) Purification, molecular cloning, and functional expression of the cholecystokinin receptor from rat pancreas. Proceedings of the National Academy of Sciences USA 89, 31253129.CrossRefGoogle ScholarPubMed
Wank, SA, Pisegna, JR & de Weerth, A (1992 b) Brain and gastrointestinal cholecystokinin receptor family: structure and functional expression. Proceedings of the National Academy of Sciences USA 89, 86918695.CrossRefGoogle ScholarPubMed
Wei, Y & Mojsov, S (1995) Tissue-specific expression of the human receptor for glucagon-like peptide-I: brain, heart and pancreatic forms have the same deduced amino acid sequences. FEBS Letters 358, 219224.CrossRefGoogle ScholarPubMed
West, DB, Fey, D & Woods, SC (1984) Cholecystokinin persistently suppresses meal size but not food intake in free-feeding rats. American Journal of Physiology 246, R776R787.Google Scholar
Wierup, N, Richards, WG, Bannon, AW, Kuhar, MJ, Ahren, B & Sundler, F (2005) CART knock out mice have impaired insulin secretion and glucose intolerance, altered beta cell morphology and increased body weight. Regulatory Peptides 129, 203211.CrossRefGoogle ScholarPubMed
Willesen, MG, Kristensen, P & Romer, J (1999) Co-localization of growth hormone secretagogue receptor and NPY mRNA in the arcuate nucleus of the rat. Neuroendocrinology 70, 306316.CrossRefGoogle ScholarPubMed
Williams, CM, Rogers, PJ & Kirkham, TC (1998) Hyperphagia in pre-fed rats following oral delta9-THC. Physiology and Behavior 65, 343346.CrossRefGoogle ScholarPubMed
Williams, DL, Kaplan, JM & Grill, HJ (2000) The role of the dorsal vagal complex and the vagus nerve in feeding effects of melanocortin-3/4 receptor stimulation. Endocrinology 141, 13321337.CrossRefGoogle ScholarPubMed
Williams, G, Bing, C, Cai, XJ, Harrold, JA, King, PJ & Liu, XH (2001) The hypothalamus and the control of energy homeostasis: different circuits, different purposes. Physiology and Behavior 74, 683701.CrossRefGoogle ScholarPubMed
Williams, G, Harrold, JA & Cutler, DJ (2000) The hypothalamus and the regulation of energy homeostasis: lifting the lid on a black box. Proceedings of the Nutrition Society 59, 385396.CrossRefGoogle ScholarPubMed
Willie, JT, Chemelli, RM, Sinton, CM & Yanagisawa, M (2001) To eat or to sleep? Orexin in the regulation of feeding and wakefulness. Annual Review of Neuroscience 24, 429458.CrossRefGoogle ScholarPubMed
Winzell, MS, Nogueiras, R, Dieguez, C & Ahren, B (2004) Dual action of adiponectin on insulin secretion in insulin-resistant mice. Biochemical and Biophysical Research Communications 321, 154160.CrossRefGoogle ScholarPubMed
Woods, SC, Lotter, EC, McKay, LD & Porte, D Jr (1979) Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature 282, 503505.CrossRefGoogle ScholarPubMed
Woods, SC, Seeley, RJ, Baskin, DG & Schwartz, MW (2003) Insulin and the blood-brain barrier. Current Pharmaceutical Design 9, 795800.CrossRefGoogle ScholarPubMed
Wortley, KE, Del Rincon, JP, Murray, JD, García, K, Iida, K, Thorner, MO & Sleeman, MW (2005) Absence of ghrelin protects against early-onset obesity. Journal of Clinical Investigation 115, 35733578.CrossRefGoogle ScholarPubMed
Wren, AM, Seal, LJ, Cohen, MA, Brynes, AE, Frost, GS, Murphy, KG, Dhillo, WS, Ghatei, MA & Bloom, SR (2001 a) Ghrelin enhances appetite and increases food intake in humans. Journal of Clinical Endocrinology and Metabolism 86, 5992.CrossRefGoogle ScholarPubMed
Wren, AM, Small, CJ, Abbott, CR, Dhillo, WS, Seal, L, Cohen, MA, Batterham, RL, Taheri, S, Stanley, SA, Ghatei, MA & Bloom, SR (2001 b) Ghrelin causes hyperphagia and obesity in rats. Diabetes 50, 25402547.CrossRefGoogle ScholarPubMed
Wren, AM, Small, CJ, Ward, HL, Murphy, KG, Dakin, CL, Taheri, S, Kennedy, AR, Roberts, GH, Morgan, DG, Ghatei, MA & Bloom, SR (2000) The novel hypothalamic peptide ghrelin stimulates food intake and growth hormone secretion. Endocrinology 141, 43254328.CrossRefGoogle ScholarPubMed
Wynne, K, Stanley, S & Bloom, S (2004) The gut and regulation of body weight. Journal of Clinical Endocrinology and Metabolism 89, 25762582.CrossRefGoogle ScholarPubMed
Xu, B, Goulding, EH, Zang, K, Cepoi, D, Cone, RD, Jones, KR, Tecott, LH & Reichardt, LF (2003) Brain-derived neurotrophic factor regulates energy balance downstream of melanocortin-4 receptor. Nature Neuroscience 6, 736742.CrossRefGoogle ScholarPubMed
Yamamoto, H, Lee, CE, Marcus, JN, Williams, TD, Overton, JM, Lopez, ME, Hollenberg, AN, Baggio, L, Saper, CB, Drucker, DJ & Elmquist, JK (2002) Glucagon-like peptide-1 receptor stimulation increases blood pressure and heart rate and activates autonomic regulatory neurons. Journal of Clinical Investigation 110, 4352.CrossRefGoogle ScholarPubMed
Yamanaka, A, Beuckmann, CT, Willie, JT, Hara, J, Tsujino, N, Mieda, M et al. (2003) Hypothalamic orexin neurons regulate arousal according to energy balance in mice. Neuron 38, 701713.CrossRefGoogle ScholarPubMed
Yamanaka, A, Kunii, K, Nambu, T, Tsujino, N, Sakai, A, Matsuzaki, I, Miwa, Y, Goto, K & Sakurai, T (2000) Orexin-induced food intake involves neuropeptide Y pathway. Brain Research 859, 404409.CrossRefGoogle ScholarPubMed
Yamauchi, T, Kamon, J, Ito, Y, Tsuchida, A, Yokomizo, T, Kita, S et al. (2003) Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 423, 762769.CrossRefGoogle ScholarPubMed
Yamauchi, T, Kamon, J, Minokoshi, Y, Ito, Y, Waki, H, Uchida, S et al. (2002) Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nature Medicine 8, 12881295.CrossRefGoogle ScholarPubMed
Yamauchi, T, Kamon, J, Waki, H, Terauchi, Y, Kubota, N, Hara, K et al. (2001) The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nature Medicine 7, 941946.CrossRefGoogle ScholarPubMed
Yaswen, L, Diehl, N, Brennan, MB & Hochgeschwender, U (1999) Obesity in the mouse model of pro-opiomelanocortin deficiency responds to peripheral melanocortin. Nature Medicine 5, 10661070.CrossRefGoogle ScholarPubMed
Yeo, GS, Farooqi, IS, Aminian, S, Halsall, DJ, Stanhope, RG & O'Rahilly, S (1998) A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nature Genetics 20, 111112.CrossRefGoogle ScholarPubMed
Yeomans, MR, Wright, P, Macleod, HA & Critchley, JA (1990) Effects of nalmefene on feeding in humans. Dissociation of hunger and palatability. Psychopharmacology 100, 426432.CrossRefGoogle ScholarPubMed
Yoshihara, T, Honma, S & Honma, K (1996) Effects of restricted daily feeding on neuropeptide Y release in the rat paraventricular nucleus. American Journal of Physiology 270, E589E595.Google ScholarPubMed
Zarjevski, N, Cusin, I, Vettor, R, Rohner-Jeanrenaud, F & Jeanrenaud, B (1993) Chronic intracerebroventricular neuropeptide-Y administration to normal rats mimics hormonal and metabolic changes of obesity. Endocrinology 133, 17531758.CrossRefGoogle ScholarPubMed
Zhang, JV, Ren, PG, Avsian-Kretchmer, O, Luo, CW, Rauch, R, Klein, C & Hsueh, AJ (2005) Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin's effects on food intake. Science 310, 996999.CrossRefGoogle ScholarPubMed
Zhang, M, Balmadrid, C & Kelley, AE (2003) Nucleus accumbens opioid, GABaergic, and dopaminergic modulation of palatable food motivation: contrasting effects revealed by a progressive ratio study in the rat. Behavioral Neuroscience 117, 202211.CrossRefGoogle ScholarPubMed
Zhang, M & Kelley, AE (2000) Enhanced intake of high-fat food following striatal mu-opioid stimulation: microinjection mapping and fos expression. Neuroscience 99, 267277.CrossRefGoogle ScholarPubMed
Zhang, Y, Proenca, R, Maffei, M, Barone, M, Leopold, L & Friedman, JM (1994) Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425432.CrossRefGoogle ScholarPubMed
Zhu, Y, Yamanaka, A, Kunii, K, Tsujino, N, Goto, K & Sakurai, T (2002) Orexin-mediated feeding behavior involves both leptin-sensitive and -insensitive pathways. Physiology and Behavior 77, 251257.CrossRefGoogle ScholarPubMed
Zigman, JM, Jones, JE, Lee, CE, Saper, CB & Elmquist, JK (2006) Expression of ghrelin receptor mRNA in the rat and the mouse brain. Journal of Comparative Neurology 494, 528548.CrossRefGoogle ScholarPubMed
Zigman, JM, Nakano, Y, Coppari, R, Balthasar, N, Marcus, JN, Lee, CE et al. (2005) Mice lacking ghrelin receptors resist the development of diet-induced obesity. Journal of Clinical Investigation 115, 35643572.CrossRefGoogle ScholarPubMed