21 - Aracne · tryptophan is made available according to Le Châtelier’s equilibrium (367,402)....

84
A05 21

Transcript of 21 - Aracne · tryptophan is made available according to Le Châtelier’s equilibrium (367,402)....

A0521

ACKNOWLEDGMENTSWe thank Michele Colletti for the assistence and we were grateful to the subjects who participated inthe study

ARACNE

Role of the Neurotransmitters and Peptides Involved in Feeding Behaviour Control

Maurizio La Guardia / Santo Giammanco / Marco Giammanco

UNIVERSITÀ DI PALERMOFISIOLOGIA E NUTRIZIONE UMANASezione del Dipartimento DIMPEFINU

ASSISTED BY THE FOLLOWING MEMBERS:Cecconi M. PHD – University of Palermo

Chiazzese F. – Graduated PharmacyDi Majo D. PHD – University of Palermo

Polidori P. – Graduated Pharmacy

Copyright © MMVIIARACNE editrice S.r.l.

[email protected]

via Raffaele Garofalo, 133 A/B00173 Roma

(06) 93781065

ISBN 978–88–548–1010-5

I diritti di traduzione, di memorizzazione elettronica,di riproduzione e di adattamento anche parziale,

con qualsiasi mezzo, sono riservati per tutti i Paesi.

Non sono assolutamente consentite le fotocopiesenza il permesso scritto dell’Editore.

I edizione: febbraio 2007

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

5

CONTENTS

I. INTRODUCTION pag. 7 II. CENTRAL FEEDING BEHAVIOUR CONTROL pag. 8 III. NEUROTRANSMITTERS

A. Tryptophan and Serotonin pag 10 B. Noradrenaline pag. 15 C. Dopamine pag. 16 D. γ-aminobutyric Acid pag. 17 E. Nitric Oxide pag. 18

IV. PEPTIDES 1- PEPTIDES ACTING AS PERIPHERAL SIGNALS A. Leptin pag. 21 B. Insulin pag. 24 C. Glucagon and Glucagon-like Peptides pag. 26 D. Cholecyistokinin pag. 27 E. Bombesin Homologous Peptides pag. 28 F. Amylin and Calcitonin Gene Related Peptide pag. 29 G. Ghrelin pag. 30

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

6

2- ANOREXANT PEPTIDES FROM NEURONS IN THE CNS

A. Melanocytes Stimulating Hormone pag. 31 B. Cocaine and Amphetamine Regulated Transcript pag. 32 C. Corticotropin Releasing Factor and Glycocorticoids

pag. 33

D. Neurotensin pag. 37 E. Oxytoicin pag. 37 F. Thyrotropin-releasing hormone pag. 38

3- PEPTIDES INDUCING FOOD INTAKE

A. Neuropeptide Y pag. 39 B. “Agouti” Related Protein pag. 40 C. Endogen Opioids pag. 41 D. Orexins A and B (Respectively Hypocretin 1 and 2) pag. 42 E. Melanine Concentrating Hormone pag. 44 F. Growth Hormone Releasing Hormone pag. 45 G. Galanin pag. 45

V. OTHER FACTORS AFFECTING FOOD INTAKE IN DIFFERENT PATHOLOGIES: CYTOKINES

pag. 46

VI. CONCLUSIONS pag. 48 VII. REFERENCES pag. 49

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

7

I. INTRODUCTION

Recently, researchers have invested greater interest in studies concerning body-weight, appetite, and feeding behaviour illnesses. The development of new theoretical models has rendered problems more complex, especially in relation to etiopathogenesis, nosographic classification and to therapeutic strategies. A great number of clinical and experimental studies have brought to light many scarcely known aspects of feeding behaviour, showing the essential neuropeptide and neurotransmitter role. In particular, the so called aminostatic mechanism has been widely accepted, highlighting the close relationship between the serotoninergic system and feeding behaviour, thus allowing an effective therapeutic approach to some clinical conditions. Also the role of hypothalamic peptides in food intake control and energy consumption has recently received greater attention. This work analyses the central regulation feeding behaviour mechanisms; the neurotransmitter and neuropeptide roles are treated in this context given that the great amount of data leads to the expectation of future developments of feeding behaviour regulating substances.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

8

II. CENTRAL FEEDING BEHAVIOUR CONTROL

As known, animals are able to control food intake according to their own energy requirements. Although not everything is known yet on the control mechanisms, it is commonly believed that such ability is part of the control mechanisms of the energy balance. Now classic experimental studies have shown that food intake is controlled by hypothalamic suprahypothalamic centers. There are hypothalamic areas in: the lateral area (LHA), ‘hunger center’ and the ventromedial nucleus (VMN), ‘satiety center’; inhibition of the hunger center after feeding causes ‘satiety sensation’ (6, 161). Researchers, concerning feeding behaviour, have not only focused their attention on the hypothalamus, but they have also studied the effects of electrical stimulations on limbic system structures. In particular electrical stimulations of the dorsal hippocampus induce, in experimental animals (rabbits or monkeys), both short term and long term increment of food intake and body weight as well as (124, 125, 126, 127, 129, 130, 131, 132, 133, 135, 136). The hippocampus is part of Papez’s emotional circuit (hippocampus, via fornix, mammillary bodies and hypothalamus, via Vicq D’Azyr’s mammillo-thalamic bundle, anterior thalamus, via thalamic projections to the limbic cortex, V temporal convolution, hippocampus) (302); such a circuit represents the anatomical substrate that explains the limbic system influences on the hypothalamus. On the other hand, also the hippocampus is sensible to glycemic levels: in fact this structure has shown electrical activity changes during hypoglycemia (128, 134). As said before, although neurophysiological studies have the great merit of having allowed to locate the anatomical structures involved in feeding behaviour control and to explain –though roughly, their functions, they could not shed any light on the finer food intake mechanisms. In particular it was not possible to find out how the hypothalamus is able to keep long-term energy reserve amounts fairly constant. Even before food is tasted, smell, food appearance, thinking about it, along with factors connected to the environment trigger reflexes (conditioned reflexes) that have noticeable effects on numerous functions of the gastro-intestinal apparatus. Subsequently even food taste and texture influence feeding behaviour (25, 77). Such factors add up to the responses –the nervous inputs, coming from the mechanoceptors and chemoceptors of the

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

9

digestive system and are determined by metabolic and endocrine variations, which induce food intake or lead to it (31). Nervous inputs are sent to the central nervous system (CNS) through the glossopharyngeal, facial, trigeminal and vagus nerves; such information represents a first class of signals acting on the CNS, which, as reply, induces food intake reduction (‘post-ingestive’ control of feeding behaviour) (31,275,399). Food intake interruption is influenced by gastric wall distention, always through the vagus afferences, which are followed by the release of peptide intestinal factors (cholecystokinin, CCK), able to influence the CNS (31,275,343,399). During the ‘post-absorptive’ phase, the nutrients carried to the peripheral tissue constitute a second class of signals (metabolic signals) for the CNS. Here we have: glucose, free fatty acids (FFA), and several amino acids (AA) like tryptophan and large neutral AA (LNAA) (31,218,246). Glucose represents the most classical example. According to Mayer’s glycostatic theory, a large artero-venous difference in glycemic levels determines fullness sensation due to the VMN glycoceptor stimulations; instead its reduction induces hunger, independently of the absolute glycemic levels (246). The AA role in regulating food intake has been confirmed by experimental data supporting the hypothesis where variations in AA plasma levels can affect the availability of AA –cerebral neurotransmitter precursors, involved in feeding behaviour (218,401,402,403). Tryptophan –precursor of the neurotransmitter serotonin or 5-hydroxytriptamine (5-HT), is considered an anorexiant substance (56,218). We will see how the role covered by (essential) phenylalanine and tyrosine in feeding behaviour control is not as important as that carried by tryptophan, though these AA, being noradrenaline adrenaline and dopamine (DA) precursors, are also food intake regulating neurotransmitters. Also pancreas hormones as leptin, which will be discussed in the following pages, are to be considered as ‘post-absorptive’ signals. The CNS reacts to all these signals through peptide synthesis, inducing or inhibiting energy dispersion (through the vegetative nervous system) thus controlling food intake (199,230,318). In synthesis, nerve and humoral afferences produced in the ‘post-ingestive’ and ‘post-absorptive’ phase induce CNS synthesis and release of numerous neurotransmitters and neuropeptides involved in feeding behaviour control.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

10

III. NEUROTRANSMITTERS

A. Tryptophan and Serotonin

Tryptophan is a neutral, essential, monoamino, monocarboxyl, heterocyclic amino acid. Besides being needed for protein synthesis, it can be converted in nicotinic acid (PP vitamin) and in serotonin (5-HT). It’s mainly metabolized by the liver (therefore influenced by hepatocyte functionality) and to a lesser extent by the brain (19). Tryptophan concentration in plasma, along with other factors (see ahead) also depends on the hepatic activity of the tryptophan pyrrolase (TRPO) involved in the metabolism of tryptophan leading to kynurenine and nicotinic acid formation (19). In normal subjects, about 1-2% of the amino acid ingested with food is transformed in 5-HT. However such a quota can reach 60% in patients suffering from intestinal carcinoma (304). Drugs capable of increasing hepatic TRPO activity, such as glucocorticoids, or conditions where the secretionary increment of the latter occurs (stress), take tryptophan from the metabolic pathway leading to 5-HT synthesis to the one leading to kynurenine nicotinic acid formation (304). Circulating tryptophan binds through a non covalent-bound with plasma albumin in such a way that only 10% of the tryptophan is free and readily diffusible (152); the bounding is fairly specific, though some metabolites compete for it, e.g. FFA: a variation of the plasma levels of the latter is capable of changing the free tryptophan amount, which, in turn, can go in the brain to form 5-HT; it is then believed that FFA are, at least in part, intermediaries of the insulin effect on the plasma tryptophan levels (106, 321). After feeding, the increment of insulin plasma levels allows AA to enter muscle tissue; however being tryptophan the only albumin bound amino acid, it will be less liable to enter muscles tissue; this implies a relative increment of the total tryptophan proportion but not of the free tryptophan; furthermore insulin frees albumin from its FFA bounds (facilitating FFA entrance in the adipocytes) thus allowing free tryptophan molecules to bind with albumin. As the concentration of free plasma tryptophan decreases, it is partially reconstituted through other tryptophan molecules spreading from other tissues as the skeleton muscle; furthermore part of the albumin-bound tryptophan is made available according to Le Châtelier’s equilibrium (367,402). In any case the concentration of free and total tryptophan is

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

11

greater than the other neutral AA, due to the fact that the latter goes in muscle tissue. Only the free tryptophan present in the blood is able to cross the hematoencephalic barrier (HEB). Tryptophan entrance in the brain is also influenced by the concentrations of other neutral amino acids: veline, leucine, isoleucine, phenylalanin, tyrosine and methiosine, which share in a competitive way with tryptophan the same specific transportation system across the HEB (106,322,401,402,403). The brain amino acid pool is very different than the plasma pool; not only some AA are only present in the brain (γ-aminobutyrric acid), but also other AA are present in the brain a remarkably greater concentration than in the plasma (glutamate, glutamine, aspartate). The brain captures plasma AA at a fast rate yet it discharges them at a similar rate, making sure that the pool composition remains constant. Amino acid uptake is an active transportation phenomenon; as far as the plasma secretion is concerned, although is an active mechanism, transportations is largely mediated by a carrier exchange in the uptake without energy consumption. Five AA transportation systems were identified on the basis of in vitro studies: one for acidic AA, one for basic AA, one for β-AA, two for neutral AA, in a similar way to what happens in the kidneys and intestine. As far as neutral AA are concerned, the systems are three but one of those (A-alanine system) involves only secretion not uptake (322). In the CNS, the tryptophan captured within the serotoninergic termination is hydroxylated to 5-OH-tryptophan and then decarboxylated to 5-HT (19). There are two different tryptophan hydroxylase (TRPH) isoforms –the enzyme considered the factor limiting 5-HT synthesis, the first isoform is found in the epiphysis, retina, stomach, spleen and thymus, the second (neuronal) is typical of the raphe (386). In physiological conditions, the 5-HT synthesis rate mainly depends on its cerebral precursor’s concentration. A controlling factor for cerebral 5-HT cerebral biosynthesis is the plasma ratio between tryptophan and LNAA –being competitors for the same carrier which transports them from the plasma to the brain (367,401,402,403). Tryptophan systemic administration increases of the cerebral concentration of 5-HT and its metabolite 5-OH-indolacetic acid (5-HIAA) (218). Summarizing, tryptophan cerebral levels are determined by at least three factors: 1. Plasma free tryptophan levels (not albumin bound); 2. Plasma molar ratio between free tryptophan and other LNAA competing

with tryptophan for access to the brain;

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

12

3. Activity of the common transportation system across the HEB. Consequently changes may take place to the feeding behaviour when some clinical or experimental conditions, influencing one or more of the above factors, affect tryptophan availability to the brain (1,48,206,225,323). In fact the serotoninergic system seems to have the more relevant action of the two neurotransmission systems, the catecholaminergic and the serotoninergic, involved in hypothalamic feeding behaviour control (Table-1). The reason lies in the different characteristics of the serotoninergic hydroxylases which transform 5-HT and tyrosine respectively into 5-OH-tryptophan and DOPA. Unlike tyrosine hydroxylase, TRPH, due to its high dissociation constant, cannot be saturated to tryptophan physiological concentrations; this implies that the amount of 5-HT synthesis depends on the tryptophan available and, since the latter car vary remarkably, 5-HT levels undergo substantial variations even in physiological conditions (107). It has been known for sometime that 5-HT inhibits feeding behaviour (219,222,225). Tests carried through the central administration of exogenous 5-HT or drugs releasing endogenous 5-HT or one of its precursors –tryptophan or 5-OH-tryptophan, strongly support the idea that the serotoninergic system is able to reduce the amount of food intake by increasing the sensation of fullness (219). In particular it is known that 5-HT affects both the energetic balance and the circadian food intake rhythm activating satiety neurons sited in the medial hypothalamus (219,225). 5-HT reduces the amount of food intake, the length and speed of meals, while it does not influences the latency of the beginning of meals or the meal frequency (219,225); this suggests that 5-HT produces sense of satiety, rather than appetite inhibition, that is the beginning of the meal. Besides, the serotoninergic antagonists preferably reduce carbohydrates intake scarcely effecting protein intake (219,225,402). Actually, carbohydrate intake, through an increment of the insulinemia levels increases plasma tryptophan and then cerebral 5-HT inducing, in turn, meal cessation or, at least, guiding the next feeding choice towards proteins (222). On the contrary, protein ingestion lowers 5-HT cerebral levels. This can be explained since tryptophan is the least abundant of the amino acids among the proteins ingested as food so its relative plasma increment is less that the increment of the competitive amino acids (105).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

13

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

14

The aforementioned data support the so called ‘aminostatic hypothesis’ for feeding behaviour control; that is, besides glycemia, also changes in plasma amino acid concentration give rise to metabolic signals affecting feeding behaviour. However also the aminostatic hypothesis does not explain how the hypothalamus can regulate long term body composition. There is, as brought out by specific studies, a different action of 5-HT on feeding behaviour, according to the receptor subtypes set in different cerebral areas: in fact, there are presynaptic receptors 5-HT1A in the raphe median nucleus, which, when activated, determine hyperphagia (20,87,384), while receptors (5-HT1B/2C) responsible for food consumption reduction are to be found in the hypothalamus shown in Table 1 (32,81,87,216,222) . Mice –knockout for the gene of receptor 5-HT1B, eat and weigh more than the control group (28). The 5-HT effect on feeding behaviour must be understood as part of a wider influence of this neurotransmitter on energy balance. In fact, its administration in the VMN determines an increment of the orthosympathetic system activity with respect to the brown adipose tissue (328) and also increases O2 consumption (215). Besides the ‘classical’ hypothalamic control centers for food intake and energy consumption, there are other diencephalic structures involved in feeding behaviour control. The hippocampus is connected to the hypothalamus through the fornix –a nervous pathway that is part of Papez’s emotional circuit and is formed by the following structures: hippocampus, via fornix, mammillary bodies and hypothalamus, via Vicq D’Azyr’s mammillo-thalamic bundle, anterior thalamus, via thalamic projections to the limbic cortex, V temporal convolution, hippocampus (302). The electrical stimulation of one of these structures induces an increment of food consumption in experiment animals; in particular, electrical stimulation of the dorsal hippocampus in rabbits (125) and monkeys (124,130), as well as irritative alterations of the rabbit hippocampus (109), remarkably increase food intake and body weight. Posterodorsal amygdala damage induces food intake in rats; such a condition reduces the orexia effect of 5-HT1A agonists (72), implying that this structure can be considered a ‘satiety center’ activated by 5-HT pathways; if the center is damaged the orexia effect of the drug is reduced since it only acts on the hypothalamus ‘satiety center’. 5-HT inhibits food consumption also by integrating with feeding behaviour control systems; in fact serotoninergic receptors are present in several

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

15

neurons that release other substance involved in feeding behaviour (66). Clinical studies showed that oral 5-OH-tryptophan administration can reduce food intake in obese patients not subject to caloric diets (56), can favour compliance to hypocaloric diets (48) and reduce carbohydrate craving in non insulin-dependent diabetic patients, thus confirming the 5-HT role in the choice of macronutrients (225). Finally, anorexia onset in cancer patients seems to be correlated to tryptophan increment of the plasma and the liquor (47,253,277).

B. Noradrenaline

As early as the 60’s, some studies showed food intake increments in sated rats that had been administered noradrenaline (NA) in the anterior lateral hypothalamus (142). Through successive pharmacological studies, the noradrenaline system has shown opposite effects on feeding behaviour control according to where it acts. NA promotes food intake when injected in the hypothalamus* paraventricular nucleus (PVN) (Table-1); such effect is linked to the activity of α2-postsynaptic adrenergic receptors; α2-adrenergic stimulation induces food intake during meals and determines a preferential ingestion of food rich of carbohydrates (224). Given these effects NA, through its α2-noradrenergic receptors, interacts antagonistically with 5-HT (225). α2-agonists drugs (clonidine) inhibit adenylate cyclase in the PVN neurons and such effect is counteracted by selective antagonists (54). Instead α-blocking drugs cause a

* After pharmacological studies on the administration of agonists and antagonists in the rat

intracerebro-venticular area or directly on the hypothalamus, many consider the PVN the ‘satiety center’ (45). Actually it is very difficult to establish whether the VMN or the PVN is the ‘satiety center’; in fact, it is true that VMN damage could be associated with the interruption of important nerve pathways and this could invalidate Heterington and Ranson remarkable results obtained in the 40’s, but it is true as well that neurotransmitter or drug central administrations do not demonstrate that the PVN is the ‘satiety center’ for at least two reasons: a) the drug or neurotransmitter administered dose could be excessive or, on the contrary, insufficient; b) drug diffusion to the nearby centers dare not avoidable no matter how skilled the researcher be, the observed effect could be caused by the activation or inhibition of other hypothalamus nuclei instead of the action of the substance on the PVN; in particular, this is relevant in rats, where the PVN is remarkably thin. Such limitations were highlighted by Leibowits herself (45) who sustained the importance of the realization of this type of experiments.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

16

reduction in carbohydrate ingestion (4). Then it is possible to hypothesize that PVN α-adrenergic receptors are involved in controlling diet composition and in particular carbohydrate percentages with respect to proteins, slowing down or inhibiting carbohydrate satiety rather then stimulating hunger or meal onset. The afferent pathways of this noradrenergic system come from the PVN level on dorsomedial direction, cross the thalamus preventricular area then descend to the central midbrain gray matter, through the pons and then continue towards the dorsal vagus complex (388). Moreover the very same NA causes the suppression of feeding behaviour in β-adrenergic mediated circuits set in the lateral hypothalamus; administering agonists on β2-adrenergic receptors reduces food intake in rats and such effect is blocked if these receptor antagonists are administered in the perifornix area (26). NA and adrenaline stimulate food intake when they act on the hypothalamus paraventricular nucleus, but they are anorexiant when they act on the adrenergic receptors in the hypothalamus perifornix area (219). Both 5-HT and NA, not only reduce food intake, but also they increase sympathetic activity (32). The relation ‘food intake – sympathetic activity increment’ suggests that effects of the neurotransmitters on feeding behaviour must be seen within the wider function of energetic balance regulation: the sympathetic activity increment following food intake aims at dispersing the introduced energy. Instead, when energy is low, sympathetic activity does not increase thus contributing to limit energy dispersion.

C. Dopamine

Up to now dopamine (DA) action on food intake control has not been well defined since DA researches are difficult because the neurotransmitter is also involved in animal motorial behaviour (272). It is fundamental to consider DA action site. Experimental trials carried by injecting DA in hypothalamus perifornix area have produced anorexia mainly for proteins, as well as a specific interference with meal onset (219,220,223). Initially experimental data indicated the receptor D2 as responsible for anorexia; in fact the administration of a D2 agonist led to food intake reduction; while a D1 agonist did not cause any effect on feeding

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

17

behaviour; furthermore administrations of D2 antagonists (Table-1) in the perifornix area increased food intake (54, 303). Recently it has been shown D1-D2 cooperation in mediating the aforementioned effect (204). Finally, using knockout mice for the gene of the receptor D3, it was brought out that also this receptor has a function in food intake control, above all by inhibiting lipid ingestion (252). The DA function on the base nuclei and in the accumbens nucleus in particular has altogether a different meaning: this was shown by using knockout mice for the gene of tyrosine hydroxylase. These hypokinetic, aphagic and adipsic animals, after having been administered l-DOPA, temporarily acquired the motorial ability needed to feed themselves, however they ingested a definitely insufficient amount of food (366,411). Genic therapy restores the ability to normally feed (365). Furthermore DA turns out to be essential for animal survival.

D. γ-aminobutyric Acid

The inhibitory neurotransmitter gamma-aminobutyric acid (GABA) is present in high concentrations in the lateral hypothalamic area (79). It stimulates food intake when injected in the VMN, and reduces it when injected in the LHA, as shown in Table 1 (187,188). Insulin induced hypoglycemia determines GABA reduction in the lateral hypothalamic area (thus disinhibiting the hunger center) and determines GABA increment in the vetromedial hypothalamus (increasing inhibition in the neuronal system) (188,194); thus it has been thought this GABAergic system could be the one that connects the hunger to the satiety center. Muscimol injections (GABAergic agonist) in the vetromedial nucleus lead to an increment of food consumption (219); bicuculline (GABAergic antagonist) antagonizes muscimol and also noradrenaline hyperphagic outcomes (189). Though partially, bicuculline antagonizes ß-endorphine induced hyperphagia (71). Given that there is specificity in removing inhibition on the satiety center from phentolone (α-adrenergic antagonist) for noradrenaline and from naloxone for ß-endorphine while the bicuculline effect is aspecific, it is supposed that GABA is the terminal neurotransmitter of an interconnection system between the hunger and the satiety center (53). Recently, a population was found with an association between the increment of activity of the gene encoding for the glutamic acid decarboxylase enzyme

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

18

(GAD2) and favors obesity; it has been hypothesized that the increment in the hypothalamus GABA pool and its consequent orexia, could be responsible for changes in feeding behaviour and that GAD2 could be the obesity gene (27).

E. Nitric Oxide

More than 15 years ago it was shown that endothelial cells synthesize and release several factors, Nitric Oxide (NO) is among those (174). It is a highly labile gaseous substance with a short half-life (3-5”) and a powerful vasodilator and platelet-aggregator. Since then a wide array of data has shown how also cells from other tissues, neurons included, release this substance. The enzyme addressed to its synthesis is NO synthase (NOS) that is functional starting from the amino acid arginine (266). NO is not enclosed in vesicles, but being a gas is able to easily cross cell membranes and quickly spread without any specialized carriers and without the interaction of membrane receptors. This way, not needing a close anatomical juxtaposition, NO affects many nearby cellular elements. NO stimulates cyclic guanosine monophosphate (cGMP) synthesis, which, in turn, determines effects that depend on the cell in which it resides. The discovery that NO works as a CNS neurotransmitter (116) has changed the classical concept of chemical neurotransmission where information is passed from a presynaptic neuron to a postsynaptic neuron; in fact according to the most accredited hypothesis the postsynaptic cell releases NO in the CNS in reply to an intracellular Ca++ increment (113). In this context the glutamatergic receptors, N-metyl- D-aspartate (NMDA) exitors that have an ionic Ca++ gain a particular relevance. One of the functions ascribed to NO in the CNS is feeding behaviour control. NO is thought to induce food intake (274). In rats, lack of food increases nNOS activity (355) and administering nNOS inhibitors reduces food intake (357), even in fasting animals (45). NO anorexigenic activity can be mediated by several neurotransmitter systems. Administering nNOS inhibitors increases cerebral 5-HT (355,356). Furthermore NO irreversibly inactivates tryptophan hydroxylase (203). Experimental results show that high concentration of cerebral NO increase HEB permeability to GABA (351). It has been reported that NO stimulates GABA release through the increment of K+ (341) or through peroxynitrite

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

19

formation, which, in turn is formed by a reaction of NO with a superoxide (292). Leptin, an anorexigenic peripheral peptide, inhibits NOS activity; L-arginine, a NO precursor, counteracts leptin effects on food intake and body weight, implying that it may modulate the serotoninergic function by inhibiting the activity of diencephalic nNOS. In fact L-arginine icv administrations counteract leptin action on 5-HT turnover, while peripheral L-arginine administrations do not alter leptin effect on food intake, indicating that leptin effects are mediated by the inhibition of NO formation in the brain. Furthermore both peripheral and central leptin administrations increase 5-HT turnover thanks to nNOS activity (44). In knockout mice for the nNOS gene, leptin inhibiting effects on food intake are less than in wild animals, implying that such inhibiting effects are partly mediated by the inhibition of NO synthesis in the brain (45).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

20

IV. PEPTIDES

In the last 10 years, a noteworthy amount of work has brought up the importance of several peptides in feeding behaviour control. The remarkable increase of knowledge in this field has brought numerous researches to individualize agonists and antagonists for these peptide receptors with the purpose of gaining new weapons for the treatment of feeding behaviour disorders; most likely in a near future, anorexia and bulimia therapy will employ these very same drugs. Many peptides are present in peripheral cerebral regions normally associated with feeding behaviour control. Some of the above are synthesized and released by peripheral structures above all in the digestive tract: the presence of food in the gastro-intestinal tract acts as a satiety signal and several indications suggest that hormones released by stomach and intestine could act as peripheral satiety signals CNS (30,31,275,343). Prevalently hypothalamic neurons synthesize and secrete several peptides; micro-inoculating some peptides in specific cerebral areas can cause feeding-behavioural changes (30). In synthesis, leptin, insulin, glucagon, CCK, bombesin, somatostatin, melanocyte stimulating hormone (α-MSH), the Cocaine peptide and Amphetamine Regulated Transcript (CART), corticotrophic releasing hormone (CRH), the thyrocotrophic releasing factor (TRH), neurotensin and oxyticin inhibit food intake; opiates, the neuropetide Y (NPY), the orexins, galamin, the melanin concentrating hormone (MCH), ghrelin and the growth hormone releasing hormone (GHRH) increase food intake. The interaction that the neuropeptides have among themselves and with neurotransmitters must be fully recognized in order to fully understand their functions in food consumption.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

21

1- PEPTIDES ACTING AS PERIPHERAL SIGNALS

A. Leptin

As said, body weight is controlled by counter-regulating physiological mechanisms determining reductions in food ingestions whenever increments in food assumption take place. Since animals, in normal conditions, are able to regulate their own body weight, a peripheral signal is needed to inform the CNS on the entity of the energy reserves of the organism. The glucostatic and the aminostatic hypothesis explain the reduction in food intake after feeding, but they cannot explain how peripheral signals inform the hypothalamus on the entity of the organism’s energy reserves. The answer to this fundamental problem came with the identification of leptin –protein produced by the mutated gene of the obese ob/ob mice (111) (gene also present in man) (232); in fact it brings this type of mice to reduce food ingestion and body weight (408). Such a peptide is secreted by the white adipocytes; it goes in the CNS through the HEB (46) and acts on CNS as a peripheral signal on the energy reserves, inhibiting food intake, reducing body weight and regulating energy balance (lipostatic hypothesis) (31,32,46,149,325,399). Leptin receptors are widely found in the VMN and the LHA as shown in Table 2 (31,257).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

22

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

23

Leptin administrations, besides reducing food intake and body weight in ob/ob mice, increase thermogenesis, physical activity and also normalize hyperglycemia and hyperinsulinemia (306); administrating icv leptin in normal rats reduces insulinemia and the ability to store lipids in adipose tissues, it also increases glucose availability and the tendency to oxidation; it favours uncoupling protein manifestation and then energy dissipation (318); body weight loss, more than anything else is tied to adipose tissue reduction (61), as it has also been demonstrated by leptin-treated animals (325); in vitro studies show also that leptin directly acts on adipocytes, increasing fat mobility (244). The above data suggest that food intake reduction is part of a more general leptin-induced tendency aiming to disperse the energy present in organisms and to prevent its further assumption (46,306,318) (catabolizing action); many of the above effects are mediated by the activations of the autonomous nervous system (32,46). Then, leptin regulates the individuals’ nutritional state (46, 318, 325). Gene mutations for leptin could also determine human obesity as it happens in ob/ob mice. Although such mechanism could theoretically be possible, very few mutations have been described for the homologous human ob gene: therefore it seems unlikely that it may occur in most of the human cases of obesity (51, 92, 267). In fact plasma leptin levels –far from being reduced in obese men (68), are correlated with adipose tissue reserves (232). The manifestations of the human ob gene have been studied through “in situ” hystochemical hybridizations techniques on subcutaneous and omental adipose tissue of thin and large obese subjects, highlighting a massive hyperconcentration in large obese. This fact introduces the concept of leptino-resistence at the hypothalamus level for some types of obesity. Under these circumstances, the abundant adipose tissue remarkably increases plasma leptin, probably in the attempt to carry out, at the hypothalamus level, metabolism adjustments towards some sort of energetic consumption and a reduced calorie input. In reality, the expected outcome does not take place in the hypothalamus due to a resistance towards the ob gene production (151,232). Therefore in obesity, the fault does not lie in the synthesis of adequate leptin amounts, but in the leptin receptor functionality or in alterations of the transductional postreceptor mechanisms of the message. For this matter, db/db mice, also genetically obese, but with high leptin levels, present a genetic alteration in leptin receptors (257,318); anyhow, other genetic leptin forms of obesity resistance are observed in rodents (173) that can also be induced by feeding (110), since hyperleptinemia induces leptin receptor down-regulation that

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

24

initially only concerns peripheral receptors (382) and those for transportation in the CNS (52), and subsequently also the hypothalamic receptors (228). These obesity models get closer to human obesity. Leptin secretions, though being correlated to amounts of the adipose tissue, vary during the day; food intake, carbohydrates above all, increases it; furthermore plasma leptin levels seem to be correlated to insulin amounts (331); however, in this case, such variations cannot be easily related to the “short term” variations of the satiety sensations (305; 320). Then, according to the above data, leptin function is to reduce “long term” food intake (17, 158). Leptin acts on feeding behaviour inhibiting secretion and release of other orexiant peptides and increasing anorexiant factors. For example, leptin anorexiant effect in part depends on its action on the NPY system (the main orexiant central peptide: see ahead); in fact it has been observed that it inhibits hypothalamus NPY release (362). Several authors have noticed relationships between leptin and the serotoninergic system. 5-OH-tryptophan administrations increment plasma leptin levels in mice (405); icv or intraperitoneal (ip) leptin administrations increase diencephalic 5-HT metabolism (45) even in ob/ob mice (156); leptin affects 5-HT metabolism through a mechanism which involves nNOS activities; in fact icv leptin administrations reduce NOS diencephalic activity for a long time (44); the increment of leptin-induced diencephalic 5-HT is antagonized by L-arginin icv administrations; in knockout mice for the nNOS gene, the leptin anorexiant effect is reduced and no increment is observed in diencephalic 5-HT metabolism (45). Leptin receptors have been observed in the nuclei of the dorsal raphe for rats and in the caudal median and dorsal raphe for monkeys, being the raphe the well known origin serotoninergic pathways. In ob/ob mice, the 5-HT carrier levels in the neurons of the raphe dorsal nucleus are low (67), therefore 5-HT release towards the superior centres is reduced; the above is in agreement with the behavioural effects increasing food ingestion; furthermore, always in ob/ob mice, leptin ip administrations increase 5-HT metabolism in the midbrain and in the hypothalamus (Harris, et al, 1998). Other authors, instead, though they remark that both 5-HT and leptin reduce food intake, reject a relationship between the two systems (150). These data show how alterations concerning leptin and its complex mechanisms, which regulate its action, may actually be the primum movens for some obesity forms. On the other hand further research must be carried out before being able to think about using leptin clinically, at least in those

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

25

cases characterized by an insufficiency or structural alterations of the hormone produced in the adipocytes.

B. Insulin

The severe effects of insulin administrations mainly set this hormone as an appetite stimulator with a mechanism that involves both liver and CNS as shown in Table 2 (142). In reality this is due to its hypoglycemizing action rather than a proper orexiant action, since its peripheral administration, carried together with glucose-controlled administrations, so to keep glycemia constant (euglycemic clamp), determine reduction in food ingestion (398). Hypothalamic insulin release or its icv administrations inhibit food intake and produce permanent body weight loss in rodents (249, 338) and primates (398). Injections of anti-insulin anticorps in the rat ventromedial hypothalamus increases food intake and then body weight (249, 364); Furthermore knockout mice for the neuronal insulin receptor are hyperphagic, obese and show an imperfection in the reproductive central control (37,290). Then, like leptin, insulin is considered a satiety signal acting on the CNS and inhibiting those central systems that favour food consumption. Insulin can cross the HEB and directly interact with the CNS cells (17, 398, 399). A number of experiments suggests that insulin interacts in the brain with both orexiant and anorexiant peptides to regulate feeding behaviour, body weight and energy homeostasis. Insulin and leptin have a tight relationship; both are satiety signals and both are hyperexpressed in a state of obesity; Insulin increases leptin secretion (305), while leptin inhibits insulin secretion (103). A convergent mechanism mediating responses to cellular leptin and insulin in the hypothalamic neurons has been suggested on the basis of cerebral insulin and leptin similarities (55, 287,288, 291, 311, 409, 410). Insulin and leptin together regulate hypothalamus NPY synthesis (181, 339). NPY manifestations in the arcuate nucleus (ARC) neurons diminish after central or systemic administrations of insulin and leptin, while NPY hyperactivity is present when those hormones diminish (339, 352, 392). Furthermore cerebral insulin has been proposed as a neuromodulator involved in cognitive feeding processes (121,122). An in vivo a

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

26

microdialysis technique showed hypothalamic insulin increments in rats accustomed to a specific “feeding-time” in the 30 minutes preceding mealtimes, while peripheral insulin levels remained unaltered (120,121; 298). Finally, other regulator peptides, such as leptin and galanin (2,361), interacting with insulin, revealed a possible involvement in such cognitive processes (122).

C. Glucagon and Glucagon-like Peptides

Glucagon ip administrations reduce the amount of food intake in rats, but it does not affect meal starting times or the other behavioural characteristics of postprandial normal satiety (118). Such effect could be due to hyperglycemia caused by increased hepatic carbohydrates mobilization, but that small icv injected doses of glucagon reduce food intake suggests a direct action on the CNS (142,176). Anyhow, more recent studies have reconsidered glucagon function in feeding behaviour; in fact, knockout mice for glucagon receptors have normal feeding behaviour and body weight, even though they show a smaller amount of fat mass and a thinner amount of thin mass (119). Glucagon comes from cleavage of a precursor 160 amino acid peptide precursor: pre-proglucagon. At the same time, such a peptide synthesises several other peptides called glucagon-like peptides (GLP), involved in many peripheral functions of the organism like glucose homeostasis, gastric emptying, insulin secretion and food intake (90, 368). The most important among the GLP’s is GLP-1 (Table-2). GLP-1 is released by L cells present in the digestive system; in man, such cells only release GLP-1 (166), while in other animals (rats, dogs, cats) they also release glucagon (85,210). Although they show 50% homology, GPL-1 and glucagon act through different receptors (372). As early as 1996, the discovery of specific GLP1 receptors and glucose sensible proteins (GLUT2 and glucokinase) supported the hypothesis that this peptide function is the hypothalamic regulation of macronutrients and water intake (280). GLP-1 works through specific receptors (GLP1-R) set in the temporal cortex, amygdala, ARC and PVN (164). It inhibits gastric emptying (279), glucagon secretion, stimulates biosynthesis and secretes

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

27

glucose-dependent insulin (299), furthermore it has short-term effects on feeding behaviour control in the CNS of rats (379) and man (145). The exact mechanism through which GLP-1 influences feeding behaviour is not properly understood, however it seems its anorexiant effect is due to an influence on NPY (376). GLP-1 central administrations also activate neurons containing CRH and oxyticin; therefore this activation could be responsible for its anorexiant effect (209). GLP iv administrations not only diminish food intake but also lead to the dose-dependent inhibition of water intake (207). While CNS leptin and GLP-1 function synergically in the CNS, since leptin activates GLP-1 that, in turn inhibits food intake (139), those two peptides carry on opposite functions in the endocrine pancreas: in fact GLP-1 stimulates insulin biosynthesis and secretion (299), instead leptin inhibits the gene transcription for insulin and its glucose-dependent secretion (191). GLP-2 is a powerful neurotransmitter that inhibits feeding behaviour in rodents and has long-term effects homeostasis body weight homeostasis (368); this is important since any possible long-term effect of GLP-1 has been previously excluded (86). The GLP-2 effects are mediated by specific GLP-2R receptors present in the intestine, dorsal median hypothalamus, in the thalamic regions, hippocampus and cortex regions (276).

D. Cholecystokinin

Cholecystokinin (CCK) is a hormonal peptide produced by the gastrointestinal tract, affects the very same gastrointestinal tract and the CNS. Its ip administration inhibits food intake in rats (7, 137). Since CCK plasma concentrations are very low and its effects are abolished by vagotomy (115,233,315,353) and by damage in the nucleus of the tractus solitarius (95), it is thought that this peptide very likely acts through nervous pathways activating ascendant vagal fibers that synapse with the nucleus of the tractus solitarius, where fibers set off for the hypothalamus PVN and other higher centers (31,200,316,343). Such hypothesis is confirmed by the presence of CCK receptors in the afferent vagal neurons (269,270,271). Besides both gastric relaxation and CCK activate afferent vagal pathways (334). There are two types of receptors for CCK: A (or 1), mainly present at

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

28

the peripheral level, but also in the CNS; B (or 2), mainly present in the CNS and in the gastric mucous (Table-2) (73, 282). CCK-A receptors mediate feeding inhibition; CCK-B receptors carry an important function for anxiety (108). It is thought that CCK acts on feeding behaviour mostly by activating CCK-1 receptors present on the afferent neurons travelling along the vagus nerve (335). OLETF rats (Otsuka Long-Evans Tokushima Fatty), genetically lacking CCK-1 receptors, are hyperfagic, diabetic and obese (112). Even if CCK-2 receptors in the hypothalamic nuclei are involved in food intake control (258), their function in feeding behaviour is controversial. Ip administrations of antagonists for the latter receptors increase food intake in rats (88), agonist administrations affect only the motivational aspects of food intake (180); however ip administrations of CCK-2 receptor agonists determine the activation of hypothalamic neurons involved in food intake control not followed by the activation of cerebral trunk neurons (50). In spite that CCK is considered one of the earliest signals just after the occurrence of food intake leaving the periphery for the CNS (31,275,399); its function on feeding behaviour, as the one of the other peptides released by the digestive apparatus (bombesin, see ahead), is very different than leptin: mainly it informs the CNS that food intake has occurred but does not provide any information on the entity of the energy reserves (230), in so much that it controls meal size but not the total food intake (389). Anyway leptin and insulin increase CNS sensibility to CCK mediated information (17); in fact the lack of leptin (251) or of its receptors (78) annul CCK effect on feeding behaviour. In the end, administrations of icv leptin mixed with i.p CCK determine a meaningful body weight reduction, greater than the one produced by leptin alone (245). Several pharmacological studies show synergism between CCK and 5-HT; administrations of an antagonist of CCK-1 receptors inhibit the anorexiant effect of fenfluoroamine (a releaser and 5-HT reuptake inhibitor) and of the very same 5-HT (70). Furthermore an antagonist of the CCK-1 receptor partially antagonizes fenfluoroamine and metergolin (5-HT antagonist) antagonizes CCK administrations (141). In man, CCK has shown to be effective in reducing the amount of food intake in normal and obese subjects (31,32,343,399), while its high iv administrations do not have any effect on subjects suffering from nervous bulimia (262).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

29

E. Bombesin Homologous Peptides

It is a peptide group presenting noteworthy structural analogies with bombesin, a peptide extracted from the skin of a fog (Bombina bombina). The homologous in mammals are gastrin releasing peptide (GRP) and neuromedines B (NMB) (261, 297). Three different receptors have been identified for these peptides: a receptor for GRP, one for the neuromedines B and a third (with a 50% homology degree with the GRP receptors) for a still unidentified binder (orphan receptor) (18,140). Both gastric cells (248) and CNS neurons (261) synthesize GRP and NMB. Nerve fibers in the tractus solitarius nucleus (TSN) and in dorsal motorial nucleus of the vagus (DMV) have been immunoreactive to these peptides (240). Furthermore, their relative receptors were found in the same nervous centers (18). Peripheral administrations, mainly of GRP, reduce food intake both in experimental animals and man (101,138,146). The same can be held true for administrations in the rat fourth ventricle; actually central administrations produces better results (205). In the end, mice lacking the GRP gene do not show any glucose intake reduction when GRP has been peripherally administered (153). However at least two points must be clarified: 1) how do peripheral peptides act in the CNS, since they can hardly cross the HEB (13) and since the effect of peripheral administrations is not abolished by vagotomy as it happens for CCK (336) 2) Where from do the nerve fibers innervating zones of the TSN come, since the zones that receive projections from the digestive apparatus do not show any immunoreactivity to the above peptides (240) Several studies concerning the first point suggest that these peptides activate some capsaicin-sensible non-vagal nerves (260) instead they supply the celiac artery (196). Concerning the TSN afferences, it has been hypothesized that they may come from the PVN, the amygdala central nucleus and the stria terminalis, centers all involved in feeding behaviour control (227).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

30

F. Amylin and Calcitonin Gene Related Peptide

Amylin is a peptide released by the pancreas β cells together with insulin as response to feeding (42; 268). It reduces food intake in rats though in man its anorexiant effects is not as strong (12). They both act at the level of the last area (236,239,264) binding to a common receptor (69). These peptides’ anorexiant effect is synergic with CCK and GRP (23, 238). Amylin mechanism is still poorly understood. According to some authors, amylin mediates CCK and gene related peptide (GRP) anorexiant effect, since in mice lacking the amylin gene the effect of the aforementioned two peptides is not shown (265). However, if it were so, the amylin effect should be abolished by vagotomy as it happens for CCK (233) and/or the pharmacological destruction of the splanchnic vagal and non vagal afferences to the CNS and as it happens GRP (260); instead neither bilateral vagotomy (237) nor capsaicin treatment (236,239) reduce the peripherally administered amylin anorexiant effect.

G. Ghrelin

It is a 28 amino acid peptide released by the stomach that, unlike other gastrointestinal peptides, favours food intake increasing body weight through adipose tissue storage (377, 400). As it is logically expected, ghrelin plasma levels diminish after meals and increase during the inter-prandial periods (75). Ghrelin administrations activate neurons NPY and Agouti Related Protein (AGRP) secrete neurons in the ARC (183, 348). Reciprocal inhibition with leptin has been observed (15,74): on one hand leptin does not only acts centrally inhibiting orexiant peptide secretions like ghrelin; on the other hand ghrelin acts anorexiantly inhibiting leptin release.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

31

In agreement with the above hypothesis it has been observed that plasma ghrelin levels are high in anorexic individuals (9) and very low in the obese (344); variations related to food intake have been observed in the latter group (98). Some authors have observed that several polymorphisms of the ghrelin gene can contribute to genetic predisposition for obesity (380).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

32

2- ANOREXIANT PEPTIDES FROM NEURONS IN THE CNS

A. Melanocytes Stimulating Hormone

The discovery that α-MSH, deriving from the cleavage of proopiomelanocortin (POMC) and being secreted in the arcuate nucleus, reduces food intake, comes from studying a particular line of genetically yellow and obese mice: the agouti mice (346). These animals produce excessive amounts of the peptide AGRP, normally present in healthy animals (see ahead), which counteracts α -MSH both in the skin, interacting with melanocortin-1 (MC1) receptors, and in the hypothalamus, interacting with melanocortin-4 (MC4) receptors (Table-3) (102,293,350). AGRP hyperexpression counteracts excessively with α -MSH in the skin (yellow hair) and in the CNS (obesity). Mice with mutations for MC4 receptors or knockout mice for the same receptor are obese but not yellow (171,374). In rats, α -MSH icv administrations reduce food intake while AGRP icv administrations block α -MSH anorexiant effect (93). α -MSH also increases the orthosympathetic system activity (32); then it produces, as all the other food intake-reducing factors, a negative effect on energy balance, where food intake inhibition is only a single aspect (399). Endogen agonist and antagonist expression for α -MSH receptors in the CNS is controlled by energy balance variations and by signals coming from adipose tissues (21). Leptin icv administrations increase POMC expression in the hypothalamus ARC (17,263,327); in fact, leptin receptors, in the ARC neurons synthesize POMC and other anorexiant peptides (256). When leptin is lacking (fasting or ob/ob mice), α -MSH expression drops (263). However, unlike ob/ob mice, knockout mice for MC4 receptors turn out obese only when fed with lipid-rich diets (41,197); this suggests that α-MSH could be more than just a simple “effector” for leptin action. Furthermore a gene mutation for MC4 receptors has been found in 3.8% of a group 209 obese subjects (381). Finally, it must be pointed out that α-MSH-releasing neurons also have 5-HT2C receptors and that the anorexiant effect of the serotoninergic drugs needs the functional integrity of melanocortin neurons and receptors (160).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

33

Then α-MSH can be considered not only an effector for leptin but also for 5-HT.

B. Cocaine and Amphetamine Regulated Transcript

This protein, the peptide CART, inhibits feeding behaviour and stimulates the orthosympathetic system (Table-3) (32,202,374). CART and its mRNA are found in hypothalamic sites as the paraventricular nucleus, the supraoptic nucleus, the LHA, the hypothalamus dorsomedial nucleus, the ARC, the periventricular nucleus and the premammillar ventral nucleus. The CART neurons in the magnocellular paraventricular nuclei and in the supraoptic area (SON) express also dynorphin; those in the LHA and in the posterior periventricular area express also the Melanin-Concentrating Hormone; many CART neurons in the ARC express also mRNA for neurotensin; those in the parvicellular paraventricular area, in the dorsomedial hypothalamus and in the anterior periventricular area express also mRNA for TRH. Then CART neurons produce neuropeptides involved in energy homeostasis (Melanin-Concentrating Hormone, TRH, dynorphin and neurotensin) (96). CART affects feeding behaviour at lower doses than α -MSH (93). As shown for α -MSH, CART-secreting neurons also express leptin receptors (257); since CART genic expression is increased by leptin and reduced in ob/ob mice and by fasting, also in this case, this peptide can be considered a leptin intermediary (257; 202). However, CART and α -MSH act through different pathways since AGRP does not contrast CART anorexiant effects (93). CART also acts on lipid metabolism: in fact both normal and obese rats –chronically administered with icv CART, have a lower body weight and food intake as well as lower insulin and leptin plasma levels; besides it increases lipid oxidation –in obese rats above all (317). Several experimental results lead to the hypothesis that leptin may increase CART expression. In fact, due to lipid-rich diets, hypothalamic CART increases (317) while its mRNA is almost absent in obese and hyperleptinemic rats (202). Furthermore, animals, undergoing food restrictions, show a decrement for CART and its mRNA in the ARC, while peripheral leptin administrations increase their expression in obese mice (202).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

34

How CART functions on feeding behaviour is still not properly understood. In fact, in spite of its effects, CART knockout mice have normal body weight and food intake, but appear less sensible to pain (14). CART icv administrations activate neurons releasing CRH, as well as oxitonergic neurons in the magnocellular and parvocellular PVN. In particular, CART effects on its CRH neurons make the epinephral gland secrete corticosterone; at least in part, this could explain CART inhibition on feeding behaviour (385). Besides it seems that CART effects on locomotion and feeding could be mediated by a direct or indirect neuronal dopaminergic activity, on the ventral tegmental area (76). In the end, it also has been hypothesized that CART role on feeding could be tied, in part, to its role mediating CCK-induced postprandial satiety. In fact, several experimental results suggest that CART could be the neuropeptide released to tractus solitarius nucleus by afferent vagal neurons (33, 34).

C. Corticotropin Releasing Factor and Glycocorticoids

CRH is a powerful anorexiant agent whose mechanism seems to be related to the one of the neurotransmitters (32; 215; 374; 39); in fact, 5-HT inhibits food consumption and stimulates CRH release; instead, NA increases food intake and inhibits CRH release (39). CRH receptors are present in the ventromedial hypothalamus (286). There are two types of CRH receptors called CRH1 and CRH2; both receptors show some homologies, though they have a different anatomic distribution. CRH2 has two variants: CRH2α and CRH2β (Table-3). CRH2α seems to be involved in the effects produced by CRH on food intake (360). Also urocortin, a 40 amminoacid peptide strictly linked to CRH, found in many brain regions as well as other organs, is an appetite suppressor (22, 211); central and peripheral urocortin administrations influence satiety and modify the amount of introduced food (22,211,354). Urocortin binds to both CRH receptors; in particular, it is a very powerful binder for CRH2-R, more than for CRH (Table-3) (354). Since according to the animal nutritional state, leptin can increase (fed state) o diminish (fasted state) CRH expression and the activity of the neurons

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

35

containing it, it has been hypothesized that CRH is a leptin effector (170,286), attributing it an analogous role as the one of α -MSH and CART. Anyway, CRH mechanism, as seen also for CART, is not related with α -MSH, since AGRP does not contrast CRH anorexiant effects (93); instead, paradoxically, α -MSH administrations attenuate CRH anorexigenic effects (296). Other authors hypothesize antagonism between CRH and NPY. In fact, it has been observed that diabetic insulin-deficient rats –fed with hyperglycidic diets, increase NPY expression in the ARC nucleus and diminish CRH in the paraventricular nucleus; besides, such rats are hyperphagic. On the contrary, in insulin-deficient diabetic rats –fed with hyperlipidic diets, NPY and CRH expressions have opposite profiles; these animals feed normally. These observations support hypothesis that food intake normalization seen in diabetic rats fed with hyperlipidic diets could be, at least in part, mediated by a drop in NPY and by an increase in CRH (60). The antagonism between CRH and NPY has also been shown by pharmacological studies (159).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

36

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

37

While dealing with CRH, it seems proper to briefly discuss glycocorticoid effects on feeding behaviour. The description of feeding behaviour outcomes in patients affected by both Cushing and Addison disease is classical. Glycocorticoids increase food intake, carbohydrates above all, reduce hypothalamic CRH levels and increase NA activity in the PVN; in turn, PVN α-adrenergic stimulations increase circulating glycocorticoids (224). Other authors have stressed how elevated glycocorticoids levels reduce leptin anorexiant effects and determine leptin-resistance (407). Several observations suggest that glucocorticoids stimulate hypothalamic NPY secretions (163,208,250), also in ob/ob mice (62). Epinephrectomy reduces NPY in the paraventricular nucleus and preproNPY in the ARC nucleus (390); it also reduces NPY receptors in the VMN (395). Furthermore, glucocorticoids have a stimulating action on the levels of the mRNA for receptors Y1 (208). Finally, epinephrectomy compromises the orexiant effect caused by icv NPY administrations, (359). Since central NPY administrations stimulate glucocorticoid secretions (154,319), it may be possible that there is a positive feedback between hypothalamus NPY and glucocorticoid plasma levels. Furthermore there is a correlation between the glucocorticoid circadian rhythm and the NPY circadian rhythm and this could explain the circadian rhythmical choice of carbohydrates normally observed also in animals (3). These data are certainly important for their possible clinical outcomes since they may show central pathogenetic mechanisms determining and supporting overweight in pathologies caused by glucocorticoid overproduction, although the peripheral effects from hypercortisolism on body fat distribution and from insulin-resistance must be taken in consideration. Besides they could explain the different attitudes towards food observed in the pathologies caused by the excess and from the defect of glucocorticoid production.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

38

D. Neurotensin

Neurotensin, a neurotransmitter known as an antinociceptive (281), inhibits food consumption (235). In rats, it interferes with NA release when administered in the PVN and in the LHA (216). According to some authors, neurotensin is a leptin mediator (327); in fact, its genic expression, reduced in ob/ob mice (394) and in neurotensin-releasing neurons, present in hypothalamic nuclei involved in feeding behaviour regulation (172), expresses leptin receptors (148). In the end, a study, carried out using homozygote mice lacking the gene for neurotensin-receptors1 (NT-1R), showed that this subtype of neurotensin receptors is implied in feeding behaviour control but not in the antinociceptive effect.

E. Oxytoicin

Oxyticin (OT) is a 9 amino acid peptide expressed in the SON, magnocellular hypothalamic neurons and hypothalamus parvocellular neurons (PVN) (Table-3). Magnocellular neurons come out on the posterior part of the pituitary gland, where OT secretion takes place; projections from the parvocellular neurons reach out to the brain central regions and external parts of the median eminence (212). OT, besides the well known effects on milk ejection and on uterine motility, has anorexiant effects (10, 11, 294); also it controls centrally and peripherally the electrolytic balance and amount of fluids (383). Ip or icv OT administrations reduce liquid and food intake and the time spent in eating both in fat and fasting animals for 21 hours; besides OT increases the latency period to the next mealtime. The effect is not observed if vasotocin, an OT antagonist, is icv injected. This shows that OT effects are mediated by specific cerebral receptors (11). However, anorexiant effects are not observed in non-fasted animals (231). A drop in food intake characterizes the OT inhibitory effect, which is not associated to plasma level alterations of hormones involved in caloric homeostasis or changes of the glycemic levels. OT agonists, administered in doses able to meaningfully inhibit food intake, have only a slight effect on water intake (294). Lack of food does not modify OT content in the SON

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

39

and in the PVN, while OT is reduced in the median eminence; the latter effect is not modified when feeding starts up again (40). The oxytoninergic system appears to be linked to CRH, as shown by the presence of CRH1-R and CRH2-R in the OT neurons of the PVN and SON (8) and by OT secretions in the pituitary gland right after CRH icv administrations (36); besides, pre-treating with oxytoninergic antagonists blocks the CRH effects on food intake, suggesting that OT is a mediator for CRH-induced hypophagia (295).

F. Thyrotropin-releasing hormone

Thyrotropin-releasing hormone (TRH) is an anorexiant peptide, whose main function is stimulating thyroid-stimulating hormone (TSH) release, but also it has other effects on the CNS. Such effects are mediated by two receptors with different anatomic distributions: TRH-R1 and TRH-R2. TRH-R1 is mainly found in the hypothalamus, while TRH-R2 is found in the thalamus, in the cerebral and cerebellar cortex and through the reticular formation (Table-3). In particular the latter is involved in pain perception and in some superior cognitive functions (162). TRH inhibits food intake acting in concomitance with leptin and melanocortin. Low leptin levels lead to a drop of the thyroid hormones as a consequence of a reduction of TRH synthesis and TSH secretion; while, in rats, TRH hyperexpression reduces leptin plasma levels (114). Not only leptin but also the melanocortins, as α-MSH, affect TRH expression and secretion (342) both in a direct way (64,143,284), and through POMC fibers linked to the TRH neurons in the PVN (155,217). In particular, the melanocortins increase TRH release, while antagonists of MC3/MC4 receptors block it (193). Instead, NPY has an inhibitory effect on TRH synthesis (64,104) and, consequently, even through this pathway, it carries out its orexiant action.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

40

3- PEPTIDES INDUCING FOOD INTAKE

A. Neuropeptide Y

NPY, polypeptide extracted for the first time in 1982 from pig brain (369), is one of the most natural orexiant agents ever obtained. In fact, its icv administrations in the hypothalamus determine hyperphagia and even food intake in already sated rats; it also determines ponderal increment (as in the syndrome from VMN damage) (32,289,317,358); on the contrary, anti-NPY antibody administrations in the paraventricular nucleus, reduce carbohydrates intake (Shibasaki, et al, 1993). NPY is synthesized by ARC neurons and produces its effects on food consumption by acting on the CNS sites responsible for body weight and energetic consumption control, in particular in the paraventricular nucleus and the dorsomedial hypothalamus (182,256). Besides, it diminishes the orthosympathetic system activity (32,199), thermogenesis (289,362), and increases insulinemia, corticosteroid plasma levels (318,362) and leptin release from the adipose tissue (318); studies in vitro show that NPY also directly affects both white adipocytes, reducing fat mobility (244), and brown adipocytes, inhibiting the effects of the orthosympathetic stimulation (Table-4) (24). Such reasons make it the most important CNS anabolizing peptide (32,318,399). At least six different NPY receptors are known; initially, the one considered responsible for the orexiant effect was the Y1 receptor (221); instead, according to more recent studies also the Y5 receptor induces food intake (184). Leptin (17,318,362) or insulin (17) central administrations reduce the expression of NPY mRNA in those nuclei where leptin (257) and insulin (406) receptors are present. It is thought that, at least in part, leptin (17,362) and insulin (17) act through the inhibition of the biosynthesis and release of NPY. Lack of leptin (ob/ob mice) or alterations of its receptors (db/db mice) determine disinhibition of NPY-releasing neurons (393). NPY-secreting neurons also have glucoreceptors; lack of glucose activates NPY release (393). In any case, NPY secretion depends from food intake: its hypothalamic levels increase during fasting and decrease after feeding (326); besides, stimulating the ARC–PVN pathway through catabolic signals (fasting, physical exercise, glicosuria) induces food intake, orthosympathetic activity and energetic consumption reduction and, parasympathetic activity

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

41

increment (199). It is thought that the effect of the catabolic signals on NPY synthesis is mediated by leptin levels reduction (337); the inverse connection between leptin and NPY is confirmed by the fact that anorexia induced by some α-MSH receptor antagonists (that, as said before, is considered the main leptin effector) is mediated by NPY (185). Therefore the studies, undertaken by administering NPY centrally or peripherally, have highlighted the orexiant effects of this peptide; on the contrary, lack of NPY, obtained in mice through inactivation of the gene producing it, does not determine any behavioural or metabolic alteration (99,301); a partial improvement of the obesity and the metabolic syndrome is observed (100, 301) only if the mice also lack leptin (ob/ob). This leads to think that the role of NPY is to have a tonically inhibitory action on the anorexiant leptin effects and/or that other orexiant peptides have the aforementioned function (243,301). These data lead to view NPY as a peptide useful in times of shortage: an increment of its secretion, induced by the lack of energetic substrates, determines metabolic effects, mainly aiming to limit energetic consumption, and behavioural effects, aiming to seek out food. The remarkable amount of data concerning the hypothalamic peptide effects on feeding behaviour starts to give shape to an adipocyte-hypothalamus axe; leptin and insulin plasma levels, somewhat directly proportional to the white adipose mass, are seen as signals informing the CNS on the entity of the energetic reserves; the hypothalamus synthesizes stimulator and inhibitor peptides affecting - energy consumption (through sympathetic activity, then to be considered, as an efferent pathway) and feeding behaviour in a direct or inverse way to leptin and insulin plasma levels (199,230,318,327); in turn, the orthosympathetic nervous system, through β3 receptors, activates the brown adipocytes inducing thermodispersion and reducing leptin expression (242).

B. “Agouti” Related Protein

As said before, the peptide AGRP antagonizes α-MSH at its receptor level. ARC neurons secrete this peptide; to be specific, 90% of the NPY-releasing neurons also release AGRP (35). Then, these neurons’ role is really relevant;

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

42

in fact they release both an orexiant peptide (NPY), and a peptide that inhibits α-MSH anorexiant activities (AGRP). Leptin reduces AGRP synthesis while fasting increases it in ob/ob mice (350). The AGRP/α-MSH system is considered one of the most important mechanisms through which leptin reduces food intake (230).

C. Endogen Opioids

In rats, central administrations of opioid peptides increase food intake, while their antagonists reduce it (226). In man, opiate antagonists reduce food intake in bulimic patients during crises (262); they are also effective in obese patients and those having a normal weight (82). Anyhow opiate orexiant effects are not as marked at NPY (226). They involve the µ receptors (186,313) and, according to some authors, also the δ (313), even if initially also κ (226) seemed involved in this action. The effects of the opioid system on feeding behaviour show some differences when compared to neuropeptide effects. While strong antagonist administrations reduce food intake, chronic administrations have shown to be ineffective in controlling body weight (226). According to some authors, more than controlling food intake as source of energy and nutrients, opioids are linked to the pleasing/displeasing sensation associated with food (89). Although the nervous centers involved in the opioid effects on feeding behaviour are different than those of other peptides, numerous experimental data show that their action site is the accumbens nucleus (313). According to some authors such a neural network involves LHA, the dorsomedial hypothalamus, the tegmental ventral area and the intermediate region of the tractus solitarius nucleus (391), all nuclei that co-ordinate metabolic, motivational, motorial and vegetative activities (Table-4). According to other authors, endogen opiods carry on a stimulating action with respect to feeding behaviour thanks to a mechanism interacting with the NPYergic pathways; this is suffraged by the fact that in sated rats, naloxone reduces NPY-induced food intake (332) and that there are anatomical relationships between NPY nervous immunopositive fibers and neurons containing β-endorphin (169).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

43

D. Orexins A and B (Respectively Hypocretin 1 and 2)

They are orexiant peptides (32,80,374) deriving from the same gene, whose product (preprohypocretin) cleaves in a different way, originating the two orexins. They interact with 2 different receptors (1 and 2); orexin A has greater affinity for receptor 1, orexin B for receptor 2 (Table-4) (329). Of the two orexins, A is more effective on feeding behaviour (94). Their intracerebroventricular administrations increase food ingestion (91), while fasting increases these proteins’ genic expression (241). The orexins are secreted by lateral hypothalamic neurons (177,256); ARC neurons releasing NPY innervate orexin-releasing neurons containing NPY1 and NPY5 receptors (167). These substances are supposed to interact with NPY (80,91,178).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

44

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

45

All the orexin-secreting neurons also secrete dynorphin (63), some also galanin (147) and, in man, also CART (307). The link with the opioid system is also supported by the fact that orexin administrations induce animals to choose tastier food (65). Orexin A reduces the activity of VMN glucosensible neurons (349) and increases the activity of the lateral hypothalamic neurons (229); these neurons contain receptors for orexin A (277). Orexinergic neurons, besides innervating the hypothalamus, they also innervate the tractus solitarius nucleus that receives vagal information on gastric distention (371). The presence of food in the stomach reduces neuronal orexinergic activity (42). Finally (but not for this less important) orexins are involved in the regulation of the sleep-awake rhythm and of locomotory activity (330). The reduction of their secretion favours sleeping (192). This could explain why is harder to fall asleep when fasting: fasting increases orexin release, in turn, making subjects more awake. Narcolepsy in man is attributed to the destruction of hypothalamic orexinergic neurons (285). Anyhow the effects on the sleep-awake rhythm and locomotory activity, at least in part, put in discussion the physiological role of the orexins on feeding behaviour. The increased ingestion of food, determined by these peptides, could be tied to a more general increment of the locomotory activity rather than a real orexiant action. In fact increments in orexin synthesis caused by fasting is not rapid, furthermore lack of orexins determines narcolepsy rather than anorexia (375). Furthermore, total fasting, not the simple reduction of food intake, increases orexin expression (42); at least in part, this could be due to the presence of food in the stomach. Finally, a single icv orexin administration is not sufficient in order to manifest hyperphagia and, chronic administrations do not induce obesity (157).

E. Melanine Concentrating Hormone

Strong MCH icv administrations transitorily increase food intake in rats; chronic administrations keep food intake constant and produce ponderal increment (83). Instead MCH knock-out mice show a drop in food intake and

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

46

body weight but an increase in energy consumption (345). Also in this case, MCH-secreting neurons are found in the lateral hypothalamus and express leptin receptors (257); these neurons also release CART peptides (76). The control of MCH synthesis is prevalently carried out by the interaction between leptin and its receptors; in fact MCH synthesis is high in ob/ob mice (312) and obese Zucker rats (363), while leptin administrations make it drop in ob/ob mice (198). MCH is considered a α-MSH antagonist, then it increases food intake and promotes energy reserve conservation (234, 374). More recent studies suggest that MCH main function is to increase energy consumption, rather than reduce food intake; in fact, ob/ob mice also lacking the MCH gene are thin but their food intake is always high; these animals show increased oxygen consumption, body temperature, resistance to cold and expression of uncoupling proteins typical of the brown adipose tissue (UCP-1) (340). According to some authors, MCH action on energetic consumption is tied to its inhibitory action on the hypothalamus- hypophysis -thyroid axis (190). MCH is also found in the plasma and its receptors are also present in the adipose tissue, where it increases leptin (29).

F. Growth Hormone Releasing Hormone

GHRH administrations intake in the hypothalamus pre-optic medial area stimulate food (84); such an effect presents a U inverted dose-response curve, since higher doses inhibit food consumption (175). GHRH favours food intake of proteins more than carbohydrates (84). According to some authors, the GHRH effect is related to opioid system effect (84). Since both GHRH, and ghrelin increase somatotrophic hormone release, it could be hypothesized that both hormones act just through the growth hormone (GH). Instead the action mechanisms are independent, because ghrelin favours food intake also in rats lacking the GH gene (348). Besides, the body weight increment caused by ghrelin is tied for a good part to its inhibition of fat utilization (377), while, as known, the somatotrophic hormone acts catabolically on adipose tissues.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

47

G. Galanin

Galanin is a peptide isolated in the gastrointestinal tract, found in high concentrations also in the hypothalamus, and in particular in PVN neurons also expressing leptin receptors (259). It stimulates food intake, above all fat (32,327,370,374). Anyway galanin seems not to be as effective on feeding behaviour as other neuropeptides; in fact, body weight, both in galanin knockout rats and in those hyperexpressing it, is not different from the control groups (165). According to some authors, also galanin effects would be mediated by the opioid system, since considerable synaptic connections are found in the hypothalamus among galanin immunopositive nervous fibers and β-endorphin-containing neurons (168).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

48

V. OTHER FACTORS AFFECTING FOOD INTAKE IN DIFFERENT PATHOLOGIES: CYTOKINES Several severe pathologies, etiopathogenetically different among themselves (tumors, cirrhosis, uremia, cardiac insufficiency, chronic obstructive lung disease), have anorexia as common symptomatologic characteristic. The consequent food intake reduction leads to severe malnutrition states co-responsible for the hypercatabolic conditions that, in turn, contribute to meaningfully increase the disease morbidity and mortality (123,300,324,333,396). Also, a poor nutritive state, besides worsening the quality of life, can limit the use therapeutic tools, retarding or impeding an adequate treatment. The above show how important is to know the relationships among these pathologies and the mechanisms controlling feeling behaviour. The aforementioned relationships are nowadays still very controversial. Initially, researchers mainly focused their attention on two different factors: free tryptophan plasma levels and cytokine actions on the hypothalamus nervous centers. According to some authors, the increment of CNS tryptophan availability (expressed by the ratio free plasma tryptophan/LNAA), followed by cerebral 5-HT increment, represents the common pathogenic anorexic mechanism in different diseases (1,48,49, 206, 323); when considering 5-HT action on feeding behaviour, is comprehensible how reducing tryptophan transportation in the CNS, with the consequent reduction of cerebral 5-HT synthesis and activity, could alleviate anorexic states (49). The cytokines released by the immune system cells inhibit food intake in some pathologies (1,179,397). Following interferon treatment (144), anorexia onset has been observed for sometime but the first cytokines to be specifically studied for this purpose were the Tumor Necrosis Factor (TNF) (57) and interleukin (IL)-1 (247). Successivly, anorexia onset was also liked to IL-6 (310) and IL-2 (58). Knowing how cytokines affect feeding behaviour has a considerable importance since it can be the physiopathologic base for the development of new specific drugs able to contrast anorexia onset and, then, increase the effectiveness of specific therapies, improve the quality of life reducing morbidity and mortality rate in numerous chronic diseases (213). For example, the use of anticorps IL-6 alleviates anorexia and malnutrition in patients affected by some cancer types (373). Up to now, it is not completely clear how cytokines induce anorexia. It is obviously supposed that they may act by interfering with some of the peptidergic mechanisms described above (314).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

49

IL-1 and Ciliary Neurotropic Factor determine the reduction of hypothalamic NPY synthesis and of its post-receptor effects (117,195,404). Besides, over-regulations of the cerebral receptors for IL-1 has been observed in murine tumors (309, 378). It has been hypothesized that TNF and IL-2 reduce the responsivity of the α-MSH/AGRP system (59); in practice, these cytokines constantly activate α-MSH release and consequently inhibit NPY (214). According to other authors, cytokines also act on the serotoninergic system controlling feeding behaviour (1,49,255,308). TNF induces the oxidation of the ramified chain AA, which compete with tryptophan for HEB crossing; this way, more tryptophan reaches the CNS and more 5-HT is formed. In the end, it has been hypothesized that cytokines may also act by activating afferent vagal fibres that inform the CNS of the occurrence of food intake (38,97,283). Anyhow, given the close relationships among the different mechanisms of feeding behaviour control, it can be supposed that all these different hypothesis may not be valid, and at the same time that they may also be integrating among themselves. The results from different studies support such supposition (16,49,254, 347, 387).

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

50

VI. CONCLUSIONS Knowing the exact mechanisms through which neuropeptides and cytokines as well as the respective receptor antagonists and agonists can increase or reduce food intake represents the first step for the development of new pharmacological interventions to be used in feeding behaviour disorders. Today ulterior studies are needed on the matter, not only to know more on food intake control systems, but also to evaluate the affects that the administrations of agonists and antagonists of neuropeptides and cytokines may have on other functions. The amount of commitment that today is being applied in merit is prevalently aimed to the treatment of obesity and primary feeding behaviour disorders, but the results of these researches will certainly have important consequences not only on the treatment of obesity and on feeding behaviour disorders, but also on the treatment for anorexia that is manifested in pathologies such as cancers, chronic kidney insufficiency and hepatic cirrhosis.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

51

VII. REFERENCES 1. Aguilera A, Selgas R, Codoceo R, and Bajo A. Uremic anorexia: a consequence

of persistently high brain serotonin levels? The tryptophan/serotonin disorder hypothesis. Perit Dial Int 20: 810-6, 2000.

2. Air EL, Benoit SC, Clegg DJ, Seeley RJ, and Woods SC. Insulin and leptin combine additively to reduce food intake and body weight in rats. Endocrinology 143: 2449-2452, 2002.

3. Akabayashi A, Levin N, Paez X, Alexander JT, and Leibowitz SF. Hypothalamic neuropeptide Y and its gene expression: relation to light/dark cycle and circlulating corticosterone. Mol Cell Neurosci 5: 210-8, 1994.

4. Alexander JT, Cheung WK, Dietz CB, and Leibowitz SF. Meal patterns and macronutrient intake after peripheral and PVN injections of the alpha 2-receptor antagonist idazoxan. Physiol Behav 53: 623-30, 1993.

5. Amara SG, Jonas V, Rosenfeld MG, Ong ES, and Evans RM. Alternative RNA processing in calcitonin gene expression generates mRNAs encoding different polypeptide products. Nature 298: 240-4, 1982.

6. Anand BK and Brobeck JR. Hypothalamic control of food intake. J Biol Med 24: 123-140, 1951.

7. Antin J, Gibbs J, Holt J, Young RC, and Smith GP. Cholecystokinin elicits the complete behavioral sequence of satiety in rats. J Comp Physiol Psychol 89: 784-90, 1975.

8. Arima H and Aguilera G. Vasopressin and oxytocin neurones of hypothalamic supraoptic and paraventricular nuclei co-express mRNA for Type-1 and Type-2 corticotropin-releasing hormone receptors. J Neuroendocrinol 12: 833-42, 2000.

9. Ariyasu H, Takaya K, Tagami T, Ogawa Y, Hosoda K, Akamizu T, Suda M, Koh T, Natsui K, Toyooka S, Shirakami G, Usui T, Shimatsu A, Doi K, Hosoda H, Kojima M, Kangawa K, and Nakao K. Stomach is a major source of circulating ghrelin, and feeding state determines plasma ghrelin-like immunoreactivity levels in humans. J Clin Endocrinol Metab 86: 4753-8, 2001.

10. Arletti R, Benelli A, and Bertolini A. Influence of oxytocin on feeding behavior in the rat. Peptides 10: 89-93, 1989.

11. Arletti R, Benelli A, and Bertolini A. Oxytocin inhibits food and fluid intake in rats. Physiol Behav 48: 825-30, 1990.

12. Balasubramaniam A, Renugopalakrishnan V, Stein M, Fischer JE, and Chance WT. Syntheses, structures and anorectic effects of human and rat amylin. Peptides 12: 919-24, 1991.

13. Banks WA and Kastin AJ. Quantifying carrier-mediated transport of peptides from the brain to the blood. Methods Enzymol 168: 652-60, 1989.

14. Bannon AW, Seda J, Carmouche M, Richards WG, Fan W, Jarosinski M, and McKinzie AA. Douglass J. Biological functions of cocaine- and

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

52

arnphetamine-regulated transcript (CART): data with CART peptides and CART knockout mice. Soc Neurosci Abstr 26: 2041 (761.10), 2000.

15. Barazzoni R, Zanetti M, Stebel M, Biolo G, Cattin L, and Guarnieri G. Hyperleptinemia prevents increased plasma ghrelin concentration during short-term moderate caloric restriction in rats. Gastroenterology 124: 1188-9, 2003.

16. Bartholomew SA and Hoffman SA. Effects of peripheral cytokine injections on multiple unit activity in the anterior hypothalamic area of the mouse. Brain Behav Immun 7: 301-16, 1993.

17. Baskin DG, Figlewicz Lattemann D, Seeley RJ, Woods SC, Porte D Jr, and Schwartz MW. Insulin and leptin: dual adiposity signals to the brain for the regulation of food intake and body weight. Brain Res 848: 114-23, 1999.

18. Battey J and Wada E. Two distinct receptor subtypes for mammalian bombesin-like peptides. Trends Neurosci 14: 524-8, 1991.

19. Bender DA. Amino acid metabolism. In: 2nd edition, John Wiley & Sons, 221-34, 1985.

20. Bendotti C and Samanin R. 8-hydroxy-2-(n-propylamino) tetralin (8-OH-DPAT) elicits eating in free feeding rats by acting on central serotonin neurons. Eur J Pharmacol 121: 147-150, 1986.

21. Benoit S, Schwartz M, Baskin D, Woods SC, and Seeley RJ. CNS melanocortin system involvement in the regulation of food intake. Horm Behav 37: 299-305, 2000.

22. Benoit SC, Thiele TE, Heinrichs SC, Rushing PA, Blake KA, and Steeley RJ. Comparison of central administration of corticotropin-releasing hormone and urocortin on food intake, conditioned taste aversion, and c-Fos expression. Peptides 21: 345-51, 2000.

23. Bhavsar S, Watkins J, and Young A. Synergy between amylin and cholecystokinin for inhibition of food intake in mic. Physiol Behav 64: 557-61, 1998.

24. Billington CJ, Briggs JE, Grace M, and Levine AS. Effects of intracerebroventricular injection of neuropeptide Y on energy metabolism. Am J Physiol 260: 321-7, 1991.

25. Blundell JE. The control of appetite: basic concepts and practical implications. Schweiz Med Wochenschr; 129: 182-8, 1999.

26. Borsini F, Bendotti C, Thurbly P, and Samanin R. Evidence that systemically administered salbutamol reduces food intake in rats by acting on central β-adrenergic sites. Life Sc 30: 277-83, 1982.

27. Boutin P, Dina C, Vasseur F, Dubois S S, Corset L, Seron K, Bekris L, Cabellon J, Neve B, Vasseur-Delannoy V, Chikri M, Charles MA, Clement K, Lernmark A, and Froguel P. GAD2 on Chromosome 10p12 Is a Candidate Gene for Human Obesity. PLoS Biol 1: 361-371, 2003.

28. Bouwknecht JA, van der Gugten J, Hijzen TH, Maes RA, Hen R, and Olivier B. Male and female 5-HT(1B) receptor knockout mice have higher body weights than wildtypes. Physiol Behav 74: 507-16, 2001.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

53

29. Bradley RL, Kokkotou EG, Maratos-Flier E, and Cheatham B. Melanin-concentrating hormone regulates leptin synthesis and secretion in rat adipocytes. Diabetes 49: 1073-7, 2000.

30. Brambilla F. Alterazioni neurotrasmettitoriali-neuropeptidiche nell'anoressia nervosa e bulimia nervosa. In Muller EE: Disordini del comportamento alimentare. Roma Milano: Ed. Pythagora Press, 81-97, 1990.

31. Bray GA. Afferent signals regulating food intake. Proc Nutr Soc 59: 373-84, 2000.

32. Bray GA. Reciprocal relation of food intake and sympathetic activity: experimental observations and clinical implications. Int J Obes Relat Metab Disord 24:8-17, 2000.

33. Broberger C and Hökfelt T. Hypothalamic and vagal neuropeptide circuitries regulating food intake. Physiol Behav 74: 669-82, 2001.

34. Broberger C, Holmberg K, Kuhar MJ, and Hökfelt T. Cocaine and amphetamine-regulated transcript in the rat vagus nerve: A putative mediator of cholecystokinin-induced satiety. Proc Natl Acad Sci U S A 96: 13506-11, 1999.

35. Broberger C, Johansen J, Johansson C, Schalling M, and Hökfelt T. The neuropeptide Y/agouti gene-related protein (AGRP) brain circuitry in normal, anorectic, and monosodium glutamate-treated mice. Proc Natl Acad Sci U S A 95: 15043-8, 1998.

36. Bruhn TO, Sutton SW, Plotsky PM, and Vale WW. Central administration of corticotropin-releasing factor modulates oxytocin secretion in the rat. Endocrinology l19: 1558-63, 1986.

37. Bruning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban PC, Klein R, Krone W, Muller-Wieland D, and Kahn CR. Role of brain insulin receptor in control of body weight and reproduction. Science 289: 2122-2125, 2000.

38. Bucinskaite V, Kurosawa M, Miyasaka K, Funakoshi A, and Lundeberg T. Interleukin-1beta sensitizes the response of the gastric vagal afferent to cholecystokinin in rat. Neurosci Lett 229: 33-6, 1997.

39. Buckingham JC. Corticotropin-releasing factor. Pharmacol Rev 31: 253-274, 1979.

40. Burlet AJ, Jhanwar-Uniyal M, Chapleur-Chateau M, Burlet CR, and Leibowitz SF. Effect of food deprivation and refeeding on the concentration of vasopressin and oxytocin in discrete hypothalamic sites. Pharmacol Biochem Behav 43: 897-905, 1992.

41. Butler AA, Marks DL, Fan W, Kuhn CM, Bartolome M, and Cone RD. Melanocortin-4 receptor is required for acute homeostatic responses to increased dietary fat. Nat Neurosci 4: 605-11, 2001.

42. Butler PC, Chou J, Carter WB, Wang YN, Bu BH, Chang D, Chang JK, and Rizza RA. Effects of meal ingestion on plasma amylin concentration in NIDDM and nondiabetic humans. Diabetes 39: 752-6, 1990.

43. Cai XJ, Widdowson PS, Harrold J, Wilson S, Buckingham RE, Arch JR, Tadayyon M, Clapham JC, Wilding J, and Williams G. Hypothalamic orexin

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

54

expression: modulation by blood glucose and feeding. Diabetes 48: 2132-7, 1999.

44. Calapai G, Corica F, Allegra A, Corsonello A, Sautebin L, De Gregorio T, Di Rosa M, Costantino G, Buemi M, and Caputi AP. Effects of intracerebroventricular leptin administration on food intake, body-weight gain and diencephalic nitric oxide synthase activity in the mouse. Br J Pharamcol 125: 798-802, 1998.

45. Calapai G, Corica F, Corsonello A, Sautebin L, Di Rosa M, Campo GM, Buemi M, Nicita Mauro V, and Caputi AP. Leptin increases serotonin turnover by inhibition of brain nitric oxide synthesis. J Clin Invest, 104: 975-82, 1999.

46. Campfield LA, Smith FJ, and Burn P. The OB protein (leptin) pathway - a link between adipose tissue mass and central neural networks. Horm Metab Res 28: 619-632, 1996.

47. Cangiano C, Cascio A, Ceci F. Plasma and CSF tryptophan and anorexia in cancer anorexia. J Neural Transm 81: 225, 1990.

48. Cangiano C, Laviano A, Del Ben M, Preziosa I, Angelico F, Cascino A, and Rossi-Fanelli F. Effects of oral 5-hydroxy-tryptophan on energy intake and macronutrient selection in non-insulin dependent diabetic patients. Int J Obes Relat Metab Disord 22: 648-54, 1998.

49. Cangiano C, Laviano A, Muscaritoli M, Meguid MM, Cascino A, and Rossi Fanelli F. Cancer Anorexia: new pathogenic and therapeutic insights. Nutrition 12 Suppl 1: S48-51, 1996.

50. Cano V, Caicoya E, and Ruiz-Gayo M. Effect of peripheral cholecystokinin receptor agonists on c-Fos expression in brain sites mediating food consumption in rats. Neurosci Lett 343: 13-6, 2003.

51. Carlsson B, Lindell K, Gabrielsson B, Karlsson C, Bjarnason R, Westphal O, Karlsson U, Sjostrom L, and Carlsson LM. Obese (ob) gene defects are rare in human obesity. Obes Res 5: 30-5, 1997.

52. Caro JR, Kolaczynski JW, Nyce MR, Ohannesian JP, Opentanova I, Goldman WH, Lynn RB, Zhang PL, Sinha MK, and Considine RV. Decreased cerebrospinal-fluid/serum leptin ratio in obesity: a possible mechanism for leptin resistance. Lancet 348: 159-61, 1996.

53. Carruba MO and Garosi V. Controllo centrale dell’assunzione di cibo. In “Disordini del comportamento alimentare-Anoressia nervosa e bulimia nervosa”, a cura di E. Muller e F. Brambilla, Pythagora Press Roma, Milano, pag. 77,1990.

54. Carruba MO, Nisoli E, Garosi V, Pizzi M, Memo M, and Spano P. Characterization of monoaminergic receptors in hypothalamic nuclei mediating feeding behavior in rats. J Pharmacol Exp Ther 259: 124-130, 1991.

55. Carvalheira JB, Siloto RM, Ignacchitti I, Brenelli SL, Carvalho CR, Leite A, Velloso LA, Gontijo JA, and Saad MJ. Insulin modulates leptin-induced STAT3 activation in rat hypothalamus. FEBS Lett 500: 119-24, 2001.

56. Ceci F, Cangiano C, Cairella M, Cascino A, Del Ben M, Muscaritoli M, Sibilia

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

55

L, and Rossi-Fanelli F. The effects of oral 5-hydroxytryptophan administration on feeding behavior in obese adult female subjects. J Neural Transm 76: 109-117, 1989.

57. Cerami A, Ikeda Y, Le Trang N, Hotez PJ, and Beutler B. Weight loss associated with an endotoxin-induced mediator from peritoneal macrophages: the role of cachectin (tumor necrosis factor). Immunol Lett 11: 173-7, 1985.

58. Cesario TC, Vaziri ND, Ulich TR, Khamiseh G, Oveisi F, Rahimzadeh M, Yousefi S, and Pandian MR. Functional, biochemical, and histopathologic consequences of high-dose interleukin-2 administration in rats. J Lab Clin Med 118: 81-8, 1991.

59. Chance WT, Sheriff S, Dayal R, and Balasubramaniam A. Refractory hypothalamic alpha-mSH satiety and AGRP feeding systems in rats bearing MCA sarcomas. Peptides 24: 1909-19, 2003.

60. Chavez M, Seeley RJ, Havel PJ, Friedman MI, Matson CA, Woods SC, and Schwartz MW. Effect of a high-fat diet on food intake and hypothalamic neuropeptide gene expression in streptozotocin diabetes. J Clin Invest 102: 340-6, 1998.

61. Chen G, Koyama K, Yuan X, Lee Y, Zhou YT, O’Doherty R, Newgard CB, and Unger RH. Disappearance of body fat in normal rats induced by adenovirus-mediated leptin gene therapy. Proc Natl Acad Sci USA 93: 14795-9, 1996.

62. Chen HL and Romsos DR. Dexamethasone rapidly increases hypothalamic neuropeptide Y secretion in adrenalectomized ob/ob mice. Am J Physiol 271: 151-8, 1996.

63. Chou TC, Lee CE, Lu J, Elmquist JK, Hara J, Willie JT, Beuckmann CT, Chemelli RM, Sakurai T, Yanagisawa M, Saper CB, and Scammell TE. Orexin (hypocretin) neurons contain dynorphin. J Neurosci 21: 168, 2001.

64. Chowdhury I, Chien JT, Chatterjee A, and Yu JY. Effects of leptin and neuropeptide-Y on transcript levels of thyrotropin beta and common alpha subunits of rat pituitary cells in vitro. Life Sci 75: 2897-909, 2004.

65. Clegg DJ, Air EL, Woods SC, and Seeley RJ. Eating elicited by orexin-a, but not melanin-concentrating hormone, is opioid mediated. Endocrinology 143: 2995-3000, 2002.

66. Collin M, Backberg M, Onnestam K, and Meister B. 5-HT1A receptor immunoreactivity in hypothalamic neurons involved in body weight control. Neuroreport 13: 945-51, 2002.

67. Collin M, Hakansson-Ovesjo M, Misane I, Ogren SO, and Meister B. Decreased 5-HT transporter mRNA in neurons of the dorsal raphe nucleus and behavioral depression in the obese leptin-deficient ob/ob mouse. Brain Res Mol Brain Res 81: 51-61, 2000.

68. Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med 334: 292-5, 1996.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

56

69. Cooper GJ. Amylin compared with calcitonin gene-related peptide: structure, biology, and relevance to metabolic disease. Endocr Rev 15: 163-201, 1994.

70. Cooper SJ, Dourish CT, and Clifton PG. CCK antagonists and CCK-monoamine interactions in the control of satiety. Am J Clin Nutr 55 Suppl 1: 291S-295S, 1992.

71. Cooper SJ. Minireview: Benzodiazepine-opiate antagonist interaction in relation to feeding and drinking reward. Life Sci 32: 1043-1051, 1983.

72. Coscina DV, Currie PJ, Bishop C, Parker GC, Rollins BL, and King BM. Posterodorsal amygdala lesions reduce feeding stimulated by 8-OH-DPAT. Brain Res 883: 243-9, 2000.

73. Crawley JN and Corwin RL. Biological actions of cholecystokinin. Peptides 15: 731-55, 1994.

74. Cummings DE and Foster KE. Ghrelin-leptin tango in body-weight regulation. Gastroenterology 124: 1532-5, 2003.

75. Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, and Weigle DS. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 50: 1714-9, 2001.

76. Dallvechia-Adams S, Kuhar MJ, and Smith Y. Cocaine- and amphetamine-regulated transcript peptide projections in the ventral midbrain: colocalization with gamma-aminobutyric acid, melanin-concentrating hormone, dynorphin, and synaptic interactions with dopamine neurons. J Comp Neurol 448: 360-72, 2002.

77. De Castro JM. Socio-cultural determinants of meal size and frequency. Br J Nutr 77: S39-54, 1997.

78. De Fanti BA, Backus RC, Hamilton JS, Gietzen DW, and Horwitz BA. Lean (Fa/Fa) but not obese (fa/fa) Zucker rats release cholecystokinin at PVN after a gavaged meal. Am J Physiol Endocrinol Metab 275: E1-E5, 1998.

79. De Feudis F.V. Central GABAergic system and feeding behavior. Neurochem Int 3: 273-279, 1981.

80. De Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, Fukuhara C, Battenberg EL, Gautvik VT, Bartlett FS 2nd, Frankel WN, van den Pol AN, Bloom FE, Gautvik KM, and Sutcliffe JG. The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci USA 95: 322-327, 1998.

81. De Vry J and Schreiber R. Effects of selected serotonin 5-HT(1) and 5-HT(2) receptor agonists on feeding behavior: possible mechanisms of action. Neurosci Biobehav Rev 24: 341-53, 2000.

82. De Zwaan M and Mitchell JE. Opiate antagonists and eating behavior in humans: a review. J Clin Pharmacol 32: 1060-72, 1992.

83. Della-Zuana O, Presse F, Ortola C, Duhault J, Nahon JL, and Levens N. Acute and chronic administration of melanin-concentrating hormone enhances food intake and body weight in Wistar and Sprague-Dawley rats. Int J Obes Relat Metab Disord 26: 1289-95, 2002.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

57

84. Dickson PR and Vaccarino FJ. GRF-induced feeding: evidence for protein selectivity and opiate involvement. Peptides 15: 1343-52, 1994.

85. Doi K, Prentki M, and Yip C. Identical biological effects of pancreatic glucagon and a purified moiety of canine gastric immunoreactive glucagon. J Clin Invest 63: 525-31, 1979.

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

87. Dourish CT, Hutson PH, Kennet GA, and Curzon G. 8-OH-DPAT-induced hyperphagia: its neural basis and possible therapeutic relevance. Appetite 7 (supp.): 127-140, 1986.

88. Dourish CT, Rycroft W, and Iversen SD. Postponement of satiety by blockade of brain cholecystokinin (CCK-B) receptors. Science 245: 1509-11, 1989.

89. Doyle TG, Berridge KC, and Gosnell BA. Morphine enhances hedonic taste palatability in rats. Pharmacol Biochem Behav 46: 745-9, 1993.

90. Drucker DJ. Glucagon-like peptides. Diabetes 47: 159-69, 1998. 91. Dube MG, Kalra SP, and Kalra PS. Food-intake elicited by central

administration of orexin/hypocretins: identification of hypothalamic site of action. Brain Res 140: 5171-7, 1999.

92. Echwald SM, Rasmussen SB, Sorensen TI, Andersen T, Tybjaerg-Hansen A, Clausen JO, Hansen L, Hansen T, and Pedersen O. Identification of two novel missense mutations in the human OB gene. Int J Obes Relat Metab Disord 21: 321-6, 1997.

93. Edwards CM, Abbott CR, Sunter D, Kim M, Dakin CL, Murphy KG, Abusnana S, Taheri S, Rossi M, and Bloom SR. Cocaine- and amphetamine-regulated transcript, glucagon-like peptide-1 and corticotrophin releasing factor inhibit feeding via agouti-related protein independent pathways in the rat. Brain Res 866: 128-34, 2000.

94. Edwards CM, Abusnana S, Sunter D, Murphy KG, Ghatei MA, and Bloom SR. The effect of the orexins on food intake: comparison with neuropeptide Y, melanin-concentrating hormone and galanin. J Endocrinol 160: 7-12, 1999.

95. Edwards GL, Ladenheim EE, and Ritter RC. Dorsomedial hindbrain participation in cholecystokinin-induced satiety. Am J Physiol 251: 971-7, 1986.

96. Elias CF, Lee CE, Kelly JF, Ahima RS, Kuhar M, Saper CB, and Elmquist JK. Characterization of CART neurons in the rat and human hypothalamus. J Comp Neurol 432: 1-19, 2001.

97. Emch GS, Hermann GE, and Rogers RC. TNF-alpha activates solitary nucleus neurons responsive to gastric distension. Am J Physiol Gastrointest Liver Physiol 279: 582-6, 2000.

98. English PJ, Ghatei MA, Malik IA, Bloom SR, and Wilding JP. Food fails to suppress ghrelin levels in obese humans. J Clin Endocrinol Metab 87: 2984-94, 2002.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

58

99. Erickson JC, Clegg KE, and Palmiter RD. Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y. Nature 381: 415-21, 1996.

100. Erickson JC and Hollopeter G. Attenuation of the obesity syndrome of ob/ob mice by the loss of neuropeptide Y. Science 274: 1704-7, 1996.

101. Ervin GN, Birkemo LS, Johnson MF, Conger LK, Mosher JT, and Menius JA Jr. The effects of anorectic and aversive agents on deprivation-induced feeding and taste aversion conditioning in rats. J Pharmacol Exp Ther 273: 1203-10, 1995.

102. Fan W, Kesterson RA, Hruby VJ, and Cone RD. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 385: 165-8, 1997.

103. Fehmann HC, Peiser C, Bode HP, Stamm M, Staats P, Hedetoft C, Lang RE, and Goke B. Leptin: a potent inhibitor of insulin secretion. Peptides 18: 1267-73, 1997.

104. Fekete C, Kelly J, Mihaly E, Sarkar S, Rand WM, Legradi G, Emerson CH, and Lechan RM. Neuropeptide Y has a central inhibitory action on the hypothalamic-pituitary-thyroid axis. Endocrinology 142: 2606-13, 2001.

105. Fernstrom JD and Wurtman RJ. Brain serotonin content: physiological regulation by plasma neutral amino acids Science 178: 414-416, 1972.

106. Fernstrom JD. Diet-induced changes in plasma amino acid pattern: effect on the brain uptake of large neutral amino acid, and on brain serotonin synthesis. J Neural Trasmission, 15: 55-67, 1979.

107. Fernstrom JD. Effects on the diet on brain neurotransmitters. Metabolism 26: 207-23, 1977.

108. Fink H, Rex A, Voits M, and Voigt JP. Major biological actions of CCK--a critical evaluation of research findings. Exp Brain Res 123: 77-83, 1998.

109. Forloni G, Fisone G, Guaitani A, Ladinsky H, and Consolo S. Role of the hippocampus in the sex-dependent regulation of eating behavior: studies with kainic acid. Physiol Behav 38 : 321-6, 1986.

110. Frederich RC, Hamann A, Anderson S, Lollman B, Lowell BB, and Flier JS. Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin action. Nature Med 1: 1311-4, 1995.

111. Friedman JM, Leibel RD, Siegel DS, Walsh J, and Bahery N. Molecular mapping of the mouse ob mutation. Genomics 11: 1054-63, 1991.

112. Funakoshi A, Miyasaka K, Jimi A, Kawanai T, Takata Y, and Kono A. Little or no expression of the cholecystokinin-A receptor gene in the pancreas of diabetic rats (Otsuka Long-Evans Tokushima Fatty = OLETF rats). Biochem Biophys Res Commun 199: 482-8, 1994.

113. Gally JA, Montague PR, Reeke GN Jr, and Edelman GM. The NO hypothesis: possible effects of a short-lived rapidly diffusible signal in the development and function of the nervous system. Proc Natl Acad Sci USA 87: 3547-51, 1990.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

59

114. Garcia SI, Landa MS, Porto PI, Alvarez AL, Schuman M, Finkielman S, and Pirola CJ. Thyrotropin-releasing hormone decreases leptin and mediates the leptin-induced pressor effect. Hypertension 39: 491-5, 2002.

115. Garlicki J, Konturek PC, Majka J, Kwiecien N, and Konturek SJ. Cholecystokinin receptors and vagal nerves in control of food intake in rats. Am J Physiol 258: 40-5, 1990.

116. Garthwaite J. Glutamate, nitric oxide and cell-cell signalling in the nervous system. Trends Neurosci 14: 60-61, 1991.

117. Gayle D, Ilyin SE, and Plata-Salaman CR. Central nervous system IL-1 beta system and neuropeptide Y mRNAs during IL-1 beta-induced anorexia in rats. Brain Res Bull 44: 311-7, 1997.

118. Geary N and Smith GP. Pancreatic glucagon and postprandial satiety in the rat. Physiol Behav 28: 313-322, 1982.

119. Gelling RW, Du XQ, Dichmann DS, Romer J, Huang H, Cui L, Obici S, Tang B, Holst JJ, Fledelius C, Johansen PB, Rossetti L, Jelicks LA, Serup P, Nishimura E, and Charron MJ. Lower blood glucose, hyperglucagonemia, and pancreatic alpha cell hyperplasia in glucagon receptor knockout mice. Proc Natl Acad Sci U S A 100: 1438-43, 2003.

120. Gerozissis K, Rouch C, Lemierre S, Meile MJ, and Orosco M. Brain insulin and cognition related to feeding. 5th Intemational Congress of Neuroendocrinology, Bristol (UK), August 2002.

121. Gerozissis K, Rouch C, Lemierre S, Nicolaidis S, and Orosco M. A potential role of central insulin in leaming and memory related to feeding. Cell Mol Neurobiol 21: 389-401, 2001.

122. Gerozissis K. Brain insulin: regulation, mechanisms of action and functions. Cell Mol Neurobiol 23: 1-25, 2003.

123. Gerstner C, Schütz T, von Baer V, Roske AE, Volk HD, Lochs H, and Plauth M. Correlation between energy expenditure, nutrient intake, malnutrition and activation of the inflammatory system in patients with liver cirrhosis (LC). Clin Nutr 19 Suppl 1: 7, 2000.

124. Giammanco S, Delgado JMR, Paderni MA, and Carollo A. A study of the feeding and social behaviour of the “Macacus rhesus” monkey after direct electrical stimulation and telestimulation of different points of the amygdala and the hippicampus. Archivio di Fisiologia 70: 243-269, 1973.

125. Giammanco S and La Grutta V. Effetti immediati e a distanza della stimolazione dell’ippocampo dorsale sul comportamento alimentare del coniglio. Archivio di Fisiologia 65 : 188-218, 1967.

126. Giammanco S. Effect of electric stimulation, direct and remote, of the amigdala and hippocampus, upon alimentary and social behaviour in the monkey. Proc Int Union of Physiological sciences 9: 1871, 1971.

127. Giammanco S. Effetti della stimolazione elettrica diretta e della telestimolazione dell’ippocampo sul comportamento alimentare del coniglio e della scimmia (Macacus Rhesus). Arch Fisiol 68: 329, 1971.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

60

128. Giammanco S. Sulle modificazioni dell’attività elettrica dell’ipotalamo indotte da variazioni glicemiche. Arch Fisiologia 64: 1, 1965.

129. Giammanco S and Delgado JMR Effetti della stimolazione diretta e della telestimolazione dell’amigdala e dell’ippocampo sul comportamento alimentare e sociale della scimmia (Macacus Rhesus). Boll Soc Ital Biol Sperim 48: 303, 1972.

130. Giammanco S, Delgado JMR, Paderni MA, and Carollo A. A study of the feeling and social behavior of the “Macacus Rhesus” monkey after direct electrical stimulation and telestimulation of different points of the amigdala and the hippocampus. Arch Fisiol 70: 243, 1973.

131. Giammanco S and La Grutta V. Stimolazione elettrica e chimica dell’ippocampo nel coniglio portatore di elettrodi e cannula a permanenza ed analisi del comportamento alimentare. Boll Soc Ital Biol Sper 43: 217, 1967.

132. Giammanco S and La Grutta V. Comportamento alimentare del coniglio con elettrodi e con cannula a permanenza in seguito a stimolazione elettrica o chimica dell’ippocampo dorsale. Boll Soc Ital Biol Sper, 43: 1290, 1967.

133. Giammanco S and La Grutta V. Effetti immediati e a distanza della stimolazione dell’ippocampo dorsale sul comportamento alimentare del coniglio. Arch Fisiol 65: 188, 1967.

134. Giammanco S, La Grutta V, and Cajola G. Analisi dell’attività bioelettrica spontanea e dell’eccitabilità dell’ippocampo dorsale nel corso dell’ipoglicemia, nel coniglio con elettrodi a permanenza nel corso dell’ipoglicemia. Boll Soc Ital Biol Sper 43: 1685, 1967.

135. Giammanco S and Raspanti G. Aumento del peso corporeo in seguito a stimolazione dell’ippocampo dorsale nel coniglio con elettrodi a permanenza. Boll Soc It Biol Sper 43: 1286, 1967.

136. Giammanco S and Raspanti G. Aumento di peso da epilessia per stimolazione dell’ippocampo dorsale nel coniglio con elettrodi a permanenza. Boll Soc It Biol Sper 42: 24, 1966.

137. Gibbs J, Young RC, and Smith GP. Cholecystokinin decreases food intake in rats. J Comp Physiol Psychol 84: 488-95, 1973.

138. Gibbs J. Effect of bombesin on feeding behavior. Life Sci 37: 147-53, 1985. 139. Goldstone AP, Morgan I, Mercer JG, Morgan DG, Moar KM, Ghatei MA, and

Bloom SR. Effect of leptin on hypothalamic GLP-1 peptide and brain-stem pre-proglucagon mRNA. Biochem Biophys Res Commun 269: 331-5, 2000.

140. Gorbulev V, Akhundova A, Buchner H, and Fahrenholz F. Molecular cloning of a new bombesin receptor subtype expressed in uterus during pregnancy. Eur J Biochem 208: 405-10, 1992.

141. Grignaschi G, Mantelli B, Fracasso C, Anelli M, Caccia S, and Samanin R. Reciprocal interaction of 5-hydroxytryptamine and cholecystokinin in the control of feeding patterns in rats. Br J Pharmacol 109: 491-4, 1993.

142. Grossman SP. Direct adrenergic and cholinergic stimulation of hypotalamic mechanism. Am J Physiol 202: 872-882, 1962.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

61

143. Guo F, Bakal K, Minokoshi Y, and Hollenberg AN. Leptin signaling targets the thyrotropin-releasing hormone gene promoter in vivo. Endocrinology 145: 2221-7, 2004.

144. Gutterman JU, Blumenschein GR, Alexanian R, Yap HY, Buzdar AU, Cabanillas F, Hortobagyi GN, Hersh EM, Rasmussen SL, Harmon M, Kramer M, and Pestka S. Leukocyte interferon-induced tumor regression in human metastatic breast cancer, multiple myeloma, and malignant lymphoma, Ann Intern Med 93: 399-406, 1980.

145. Gutzwiller JP, Drewe J, Goke B, Schmidt H, Rohrer B, Lareida J, and Beglinger C. Glucagon-like peptide-1 promotes satiety and reduces food intake in patients with diabetes mellitus type 2. Am J Physiol 276: 1541-4, 1999.

146. Gutzwiller JP, Drewe J, Hildebrand P, Rossi L, Lauper JZ, and Beglinger C. Effect of intravenous human gastrin-releasing peptide on food intake in humans. Gastroenterology 106: 1168-73, 1994.

147. Hakansson M, de Lecea L, Sutcliffe JG, Yanagisawa M, and Meister B. Leptin receptor- and STAT3-immunoreactivities in hypocretin/orexin neurones of the lateral hypothalamus. J Neuroendocrinol;11(8):653-63, 1999.

148. Hakansson ML, Brown H, Ghilardi N, Skoda RC, and Meister B. Leptin receptor immunoreactivity in chemically defined target neurons of the hypothalamus. J Neurosci 18: 559-72, 1998.

149. Halaas J, Boozer C, Blair-West J, Fidahusein N, Denton D, and Friedman J. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc Natl Acad Sci USA 94: 8878-83, 1997.

150. Halford JC and Blundell JE. Separate systems for serotonin and leptin in appetite control. Ann Med 32: 222-32, 2000.

151. Hamilton S, Paglia D, Kwan A, and Deitel M. Increased obese mRNA expression in omental fat cells from massively obese humans. Nature Med 1: 953, 1995.

152. Hamon M and Bourgoin S. Ontogenesis of tryptophan transport in the rat brain. J Neural Transmission, 15: 93-105, 1979.

153. Hampton LL, Ladenheim EE, Akeson M, Way JM, Weber HC, Sutliff VE, Jensen RT, Wine LJ, Arnheiter H, and Battey JF. Loss of bombesin-induced feeding suppression in gastrin-releasing peptide receptor-deficient mice. Proc Natl Acad Sci U S A 95: 3188-92, 1998.

154. Hanson ES and Dallman MF. Neuropeptide Y (NPY) may integrate responses of hypothalamic feeding systems and the hypothalamo-pituitary-adrenal axis. J Neuroendocrinol 7: 273-9, 1995.

155. Harris M, Aschkenasi C, Elias CF, Chandrankunnel A, Nillni EA, Bjoorbaek C, Elmquist JK, Flier JS, and Hollenberg AN. Transcriptional regulation of the thyrotropin-releasing hormone gene by leptin and melanocortin signaling. J Clin Invest 107: 111-20, 2001.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

62

156. Harris RB, Zhou J, Redmann SM Jr, Smagin GN, Smith SR, Rodgers E, and Zachwieja JJ. A leptin dose-response study in obese (ob/ob) and lean (+/?) mice. Endocrinology 139: 8-19, 1998.

157. Haynes AC, Jackson B, Overend P, Buckingham RE, Wilson S, Tadayyon M, and Arch JR. Effects of single and chronic intracerebroventricular administration of the orexins on feeding in the rat. Peptides 20: 1099-105, 1999.

158. Heini AF, Lara-Castro C, Kirk KA, Considine RV, Caro JF, and Weinsier RL. Association of leptin and hunger-satiety ratings in obese women. Int J Obes Relat Metab Disord 22: 1084-7, 1998.

159. Heinrichs SC, Menzaghi F, Pich EM, Hauger RL, and Koob GF. Corticotropin-releasing factor in the paraventricular nucleus modulates feeding induced by neuropeptide Y. Brain Res 611: 18-24, 1993.

160. Heisler LK, Cowley MA, Kishi T, Tecott LH, Fan W, Low MJ, Smart JL, Rubinstein M, Tatro JB, Zigman JM, Cone RD, and Elmquist JK. Central serotonin and melanocortin pathways regulating energy homeostasis. Ann N Y Acad Sci 994: 169-74, 2003.

161. Hetherigton AW and Ranson SW. Hypothalamic lesion and adiposity in the rat. Anat Rec 78: 149-54, 1940.

162. Heuer H, Schafer MK, O’Donnell D, Walker P, and Bauer K. Expression of thyrotropin-releasing hormone receptor 2 (TRH-R2) in the central nervous system of rats. J Comp Neurol 428: 319-36, 2000.

163. Higuchi H, Yang HY, and Sabol SL. Rat neuropeptide Y precursor gene expression. mRNA structure, tissue distribution, and regulation by glucocorticoids, cyclic AMP, and phorbol ester. J Biol Chem 263: 6288-95, 1988.

164. Hillebrand JJ, de Wied D and Adan RA. Neuropeptides, food intake and body weight regulation: a hypothalamic focus. Peptides 23: 2283-306, 2002.

165. Homann JG, Krasnow SM, Teklemichael DN, Clifton DK, Wynick D, and Steiner RA. Neuroendocrine profiles in galanin-overexpressing and knockout mice. Neuroendocrinology 77: 354-66, 2003.

166. Holst JJ, and Orskov C. Glucagon and other proglucagon-derived peptides. In: “Gut peptides: Biochemistry and Physiology”, Walsh JH and Dockray GJ (eds), New York: Raven, pagg. 305-40, 1994.

167. Horvath TL, Diano S, and van den Pol AN. Synaptic interaction between hypocretin (orexin) and neuropeptide Y cells in the rodent and primate hypothalamus: a novel circuit implicated in metabolic and endocrine regulations. J Neurosci 19: 1072-87, 1999.

168. Horvath TL, Kalra SP, Naftolin F, and Leranth C. Morphological evidence for a galanin-opiate interaction in the rat mediobasal hypothalamus. J Neuroendocrinol 7: 579-88, 1995.

169. Horvath TL, Naftolin F, Kalra SP, and Leranth C. Neuropeptide-Y innervation of beta-endorphin containing cells in the rat mediobasal hypothalamus: a light and electron microscopic double immunostaining analysis.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

63

Endocrinology 131: 2461-7, 1992. 170. Huang Q, Rivest R, and Richard D. Effects of leptin on corticotropin-releasing

factor (CRF) synthesis and CRF neuron activation in the paraventricular hypothalamic nucleus of obese (ob/ob) mice. Endocrinology 139: 1524-32, 1998.

171. Huszar D, Lynch C, Fairchild-Huntress V, Dunmore L, Fang Q, Berkemeier L, Gu W, Kesterson R, Boston B, Cone R, Smith F, Campfield L, Burn P, and Lee F. Targeted disruption of the melanocortin-4 receptor results in obesity. Cell 88: 131-41, 1997.

172. Ibata Y, Kawakami F, Fukui K, Obata-Tsuto HL, Tanaka M, Kubo T, Okamura H, Morimoto N, Yanaihara C, and Yanaihara N. Light and electron microscopic immunocytochemistry of neurotensin-like immunoreactive neurons in the rat hypothalamus. Brain Res 302: 221-30, 1984.

173. Igel M, Becker W, Herberg L, and Joost HG. Hyperleptinemia, leptin resistance, and polymorphic leptin receptor in New Zealand obese mouse. Endocrinology 138: 4234-9, 1997.

174. Ignarro LJ, Byrns RE, Buga GM, and Wood KS. Endothelium-derived relaxing factor from pulmonary artery and vein possesses pharmacological and chemical properties identical to those of nitric oxide radical. Circulation Research 61: 866-79, 1987.

175. Imaki T, Shibasaki T, Hotta M, Masuda A, Demura H, Shizume K, and Ling N. The satiety effect of growth hormone-releasing factor in rats. Brain Res 340: 186-8, 1985.

176. Inokuchi A, Oomura Y, and Nishimura H. Effect of intracerebroventricularly infused glucagon on feeding behavior. Physiol Behav 33: 397-400, 1984.

177. Iqbal J, Pompolo S, Murakami T, Grouzmann E, Sakurai T, Meister B, and Clarke IJ. Immunohistochemical characterization of localization of long-form leptin receptor (OB-Rb) in neurochemically defined cells in the ovine hypothalamus. Brain Res 920: 55-64, 2001.

178. Jain MR, Horvath TL, Kalra PS, and Kalra SP. Evidence that NPY Y1 receptors are involved in stimulation of feeding by orexins (hypocretins) in sated rats. Regu Pept 87: 19-24, 2000.

179. Jansson JO, Wallenius K, Wernstedt I, Ohlsson C, Dickson SL, and Wallenius V. On the site and mechanism of action of the anti-obesity effects of interleukin-6. Growth Horm IGF Res 13 Suppl A: S28-32, 2003.

180. Kadar T, Penke B, Kovacs K, and Telegdy G. Inhibition of feeding by the C-terminal tetrapeptide fragment of cholecystokinin in a novel environment. Neuropeptides 7: 97-108, 1986.

181. Kalra SP, Dube MG, Fournier A, and Kalra PS. Structure-function analysis of stimulation of food intake by neuropeptide Y: effects of receptor agonists. Physiol Behav 50: 5-9, 1991.

182. Kalra SP, Dube MG, Xu B, Horvath TL, and Kalra PS. Interacting appetite-regulating pathways in the hypothalamic regulation of body weight. Endocr Rev 20: 68-100, 1999.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

64

183. Kamegai J, Tamura H, Shimizu T, Ishii S, Sugihara H, and Wakabayashi I. Chronic central infusion of ghrelin increases hypothalamic neuropeptide Y and Agouti-related protein mRNA levels and body weight in rats. Diabetes 50: 2438-43, 2001.

184. Kanatani A, Ishihara A, Iwaasa H, Nakamura K, Okamoto O, Hidaka M, Ito J, Fukuroda T, MacNeil DJ, Van der Ploeg LH, Ishii Y, Okabe T, Fukami T, and Ihara M. L-152,804: orally active and selective neuropeptide Y Y5 receptor antagonist. Biochem Biophys Res Commun 272: 169-73, 2000.

185. Kask A, Rago L, Korrovits R, Wikberg J, and Schioth H. Evidence the orexigenic effects of melanocortin 4 receptor antagonist HS014 are mediated by neuropeptide Y. Biochem Biophys Res Comm 248: 245-9, 1998.

186. Kelley AE, Bakshi VP, Fleming S, and Holahan MR. A pharmacological analysis of the substrates underlying conditioned feeding induced by repeated opioid stimulation of the nucleus accumbens. Neuropsychopharmacology 23: 455-67, 2000.

187. Kelly J, Alheid GF, Newberg A, and Grossman SP. GABA stimulation and blockade in the hypothalamus and midbrain: effects on feeding and locomotor activity. Pharmacol Biochem Behav 7: 537-41, 1977.

188. Kelly J and Grossman SP. GABA and hypothalamic feeding systems. II. A comparison of GABA, glycine and acetylcholine agonists and their antagonists. Pharmacol Biochem Behav 11: 647-652, 1979.

189. Kelly J, Rothstine J, and Grossman SP. GABA and hypotalamic feeding system. I) Topographic analysis of the effects of microinjection of muscimol. Physiol Behav 23: 1123-1134, 1979.

190. Kennedy AR, Todd JF, Stanley SA, Abbott CR, Small CJ, Ghatei MA, and Bloom SR. Melanin-concentrating hormone (MCH) suppresses thyroid stimulating hormone (TSH) release, in vivo and in vitro, via the hypothalamus and the pituitary. Endocrinology 142: 3265-8, 2001.

191. Kieffer TJ and Habener JF. The adipoinsular axis: effects of leptin on pancreatic beta-cells. Am J Physiol Endocrinol Metab 278: 1-14, 2000.

192. Kilduff TS and Peyron C. The hypocretin/orexin ligand-receptor system: implications for sleep and sleep disorders. Trends Neurosci 23: 359-65, 2000.

193. Kim MS, Small CJ, Stanley SA, Morgan DG, Seal LJ, Kong WM, Edwards CM, Abusnana S, Sunter D, Ghatei MA, and Bloom SR. The central melanocortin system affects the hypothalamo-pituitary thyroid axis and may mediate the effect of leptin. J Clin Invest 105: 1005-11, 2000.

194. Kimura H and Kuriyama K. Distribution of gamma-aminobutyric acid (GABA) in the rat hypothalamus: functional correlates of GABA with activities of appetite controlling mechanisms. J Neurochem 24: 903-7, 1975.

195. King PJ, Widdowson PS, Doods H, and Williams G. Effect of cytokines on hypothalamic neuropeptide Y release in vitro. Peptides 21: 143-6, 2000.

196. Kirkham TC, Gibbs J, and Smith GP. Satiating effect of bombesin is mediated by receptors perfused by celiac artery. Am J Physiol 261: 614-8, 1991.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

65

197. Koegler FH, Schaffhauser RO, Mynatt RL, York DA, and Bray GA. Macronutrient diet intake of the lethal yellow agouti (Ay/a) mouse. Physiol Behav 67: 809-12, 1999.

198. Kokkotou EG, Tritos NA, Mastaitis JW, Slieker L, and Maratos-Flier E. Melanin-concentrating hormone receptor is a target of leptin action in the mouse brain. Endocrinology 142: 680-6, 2001.

199. Kokot F and Ficek R. Effects of neuropeptide Y on appetite. Miner Electrolyte Metab 25: 303-5, 1999.

200. Konturek SJ, Konturek JW, Pawlik T, and Brzozowki T. Brain-gut axis and its role in the control of food intake. J Physiol Pharmacol 55: 137-54, 2004.

201. Krahn DD, Gosnell BA, Levine AS, and Morley JE. Effects of calcitonin gene-related peptide on food intake. Peptides 5: 861-4, 1984.

202. Kristensen R, Judge M, Thim L, Ribel U, Christjansen K, Wulff B, Clausen J, Jensen P, Madsen O, Vrang N, Larsen P, and Hastrup S. Hypothalamic CART is a new anorectic peptide regulated by leptin. Nature 393: 72-6, 1998.

203. Kuhn DM and Arthur R Jr. Molecular mechanism of the inactivation of tryptophan hydroxylase by nitric oxide: attack on critical sulfhydryls that spare the enzyme iron center. J Neurosci 17: 7245-51, 1997.

204. Kuo DY. Co-administration of dopamine D1 and D2 agonists additively decreases daily food intake, body weight and hypothalamic neuropeptide Y level in rats. J Biomed Sci 9: 126-32, 2002.

205. Ladenheim EE, Ritter RC. Low-dose fourth ventricular bombesin selectively suppresses food intake. Am J Physiol 255: 988-93, 1988.

206. Lambert GW, Vaz M, Cox HS, Turner AG, Kaye DM, Jennings GL, and Esler MD. Human obesity is associated with a chronic elevation in brain 5-hydroxytryptamine turnover. Clin Sci 96: 191-7, 1999.

207. Larsen PJ, Fledelius C, Knudsen LB, and Tang-Christensen M. Systemic administration of the long-acting GLP-1 derivative NN2211 induces lasting and reversible weight loss in both normal and obese rats. Diabetes 50: 2530-9, 2001.

208. Larsen PJ, Jessop DS, Chowdrey HS, Lightman SL, and Mikkelsen JD. Chronic administration of glucocorticoids directly upregulates prepro-neuropeptide Y and Y1-receptor mRNA levels in the arcuate nucleus of the rat. J Neuroendocrinol 6: 153-9, 1994.

209. Larsen PJ, Tang-Christensen M, and Jessop DS. Central administration of glucagon-like peptide-1 activates hypothalamic neuroendocrine neurons in the rat. Endocrinology 138: 4445-55, 1997.

210. Larsson LI, Holst J, Hakanson R, and Sundler F. Distribution and properties of glucagon immunoreactivity in the digestive tract of various mammals: an immunohistochemical and immunochemical study. Histochemistry 44: 281-90, 1975.

211. Latchman DS. Urocortin. Int J Biochem Cell Biol 34: 907-10, 2002.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

66

212. Laurent FM, Hindelang C, Klein MI, Stoeckel ME, and Felix IM. Expression of the oxytocin and vasopressin genes in the rat hypothalamus during development: an in situ hybridization study. Brain Res Dev Brain Res 46: 145-54, 1989.

213. Laviano A, Meguid MM, and Rossi-Fanelli F. Cancer anorexia: clinical implications, pathogenesis, and therapeutic strategies. Lancet Oncol 4: 686-94, 2003.

214. Laviano A, Meguid MM, and Rossi-Fanelli F. Improving food intake in anorectic cancer patients. Curr Opin Clin Nutr Metab Care 6: 421-6, 2003.

215. Le Feuvre RA, Aisenthal L, and Rothwell NJ. Involvement of corticotrophin releasing factor (CRF) in the thermogenic and anorexic actions of serotonin (5-HT) and related compounds. Brain Res 555: 245-50, 1991.

216. Lee MD, Kennett GA, Dourish CT, and Clifton PG. 5-HT1B receptors modulate components of satiety in the rat: behavioural and pharmacological analyses of the selective serotonin1B agonist CP-94,253. Psychopharmacology 164: 49-60, 2002.

217. Legradi G, Emerson CH, Ahima RS, Flier JS, and Lechan RM. Leptin prevents fasting-induced suppression of prothyrotropin-releasing hormone messenger ribonucleic acid in neurons of the hypothalamic paraventricular nucleus. Endocrinology 138: 2569-76, 1997.

218. Lehnert H and Wurtman RJ. Amino acid control of neurotransmitter synthesis and release: physiological and clinical implications. Psychother Psychosom 60: 18-32, 1993.

219. Leibowitz SF. Neurochemical systems of the hypothalamus: control of feeding and drinking behavior and water-electrolyte excretion. In: “Behavioral Studies of the Hypothalamus. Handbook of the Hypothalamus”, vol. 3 A., Morgane P.J., Panksepp J. (Eds.), Marcel Dekker, New York, pp.299-437, 1980.

220. Leibowitz SF and Shor-Posner G. Brain serotonin and eating behaviour. Appetite 7: 14, 1986.

221. Leibowitz SF and Alexander JT. Analysis of neuropeptide Y-induced feeding: dissociation of Y1 and Y2 receptor effects on natural meal patterns. Peptides 12: 1251-60, 1991.

222. Leibowitz SF and Alexander JT. Hypothalamic serotonin in control of eating behavior, meal size, and body weight. Biol Psychiatry 44: 851-64, 1998.

223. Leibowitz SF. Catecholaminergic mechanisms of the amphetamine anorexia. Brain Res 98: 529-45, 1975.

224. Leibowitz SF. Hypothalamic paraventricular nucleus: interaction between alpha 2-noradrenergic system and circulating hormones and nutrients in relation to energy balance. Neurosci Biobehav Rev 12: 101-9, 1988.

225. Leibowitz SF. The role of serotonin in eating disorders. Drugs 39 Suppl. 3: 33-48, 1990.

226. Levine AS and Billington CJ. Opioids. Are they regulators of feeding? Ann NY Acad Sci 575: 209-19, 1989.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

67

227. Li BH and Rowland NE. Peripherally and centrally administered bombesin induce Fos-like immunoreactivity in different brain regions in rats. Regul Pept 62: 167-72, 1996.

228. Lin S, Storlien LH, and Huang X. Leptin receptor, NPY, POMC mRNA expression in the diet-induced obese mouse brain Brain Res 875: 89-95, 2000.

229. Liu XH, Morris R, Spiller D, White M, and Williams G. Orexin-A preferentially excites glucose-sensitive neurons in the lateral hypothalamus of the rat in vitro. Diabetes 50: 2431-7, 2001.

230. Loftus TM. An adipocyte-central nervous system regulatory loop in the control of adipose homeostasis. Cell Develop Biol 10: 11-8, 1999.

231. Lokrantz CM, Uvnas-Moberg K, and Kaplan JM. Effects of central oxytocin administration on intraoral intake of glucose in deprived and nondeprived rats. Physiol Behav 62: 347-52, 1997.

232. Lonnqvist F, Arner P, Nordfors L, and Schalling M. Overexpression of the obese (ob) gene in the adipose tissue of human obese subjects. Nature Med 1: 950-2, 1995.

233. Lorenz DN and Goldman SA. Vagal mediation of the cholecystokinin satiety effect in rats. Physiol Behav 29: 599-604, 1982.

234. Ludwig D, Mountjoy K, Tatro J, Gillette J, Frederich R, Flier J, and Maratos-Flier E. Melanin-concentrating hormone: a functional melanocortin antagonist in the hypothalamus. Am J Physiol 274: , 1998.

235. Luttinger D, King RA, Sheppard D, Strupp J, Nemeroff CB, and Prange AJ Jr. The effect of neurotensin on food consumption in the rat. Eur J Pharmacol 81: 499-503, 1982.

236. Lutz TA, Althaus J, Rossi R, and Scharrer E. Anorectic effect of amylin is not transmitted by capsaicin-sensitive nerve fibers. Am J Physiol 274: 1777-82, 1998.

237. Lutz TA, Del Prete E, and Scharrer E. Subdiaphragmatic vagotomy does not influence the anorectic effect of amylin. Peptides 16: 457-62, 1995.

238. Lutz TA, Del Prete E, Szabady MM, and Scharrer E. Attenuation of the anorectic effects of glucagon, cholecystokinin, and bombesin by the amylin receptor antagonist CGRP(8-37). Peptides 17: 119-24, 1996.

239. Lutz TA, Senn M, Althaus J, Del Prete E, Ehrensperger F, and Scharrer E. Lesion of the area postrema/nucleus of the solitary tract (AP/NTS) attenuates the anorectic effects of amylin and calcitonin gene-related peptide (CGRP) in rats. Peptides 19: 309-17, 1998.

240. Lynn RB, Hyde TM, Cooperman RR, and Miselis RR. Distribution of bombesin-like immunoreactivity in the nucleus of the solitary tract and dorsal motor nucleus of the rat and human: colocalization with tyrosine hydroxylase. J Comp Neurol 369: 552-70, 1996.

241. Mandal MS, Nakazato M, Date Y, Murakami N, Yanagisawa M, and Matsukura S. Widespread distribution of orexin in rat brain and its regulation upon fasting. Biochem Biophys Res Commun 256: 495-9, 1999.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

68

242. Mantzoros CS, Qu D, Frederich RC, Susulic VS, Lowell BB, Maratos-Flier E, and Flier JS. Activation of beta(3) adrenergic receptors suppresses leptin expression and mediates a leptin-independent inhibition of food intake in mice. Diabetes 45: 909-14, 1996.

243. Marsh DJ, Miura GI, Yagaloff KA, Schwartz MW, Barsh GS, and Palmiter RD. Effects of neuropeptide Y deficiency on hypothalamic agouti-related protein expression and responsiveness to melanocortin analogues. Brain Res 848: 66-77, 1999.

244. Martinez JA, Aguado M, and Fruhbeck G. Interactions between leptin and NPY affecting lipid mobilization in adipose tissue. J Physiol Biochem 56: 1-8, 2000.

245. Matson CA, Reid DF, Cannon TA, and Ritter RC. Cholecystokinin and leptin act synergistically to reduce body weight. Am J Physiol Regul Integr Comp Physiol 278: 882-90, 2000.

246. Mayer J. Glucostatic mechanisms in regulation of food intake. N Engl J Med 249: 13-16, 1953.

247. McCarthy DO, Kluger MJ, and Vander AJ. Suppression of food intake during infection: is interleukin-1 involved? Am J Clin Nutr 42: 1179-82, 1985.

248. McDonald TJ, Nilsson G, Vagne M, Ghatei M, Bloom SR, and Mutt V. A gastrin releasing peptide from the porcine nonantral gastric tissue. Gut 19: 767-74, 1978.

249. McGowan MK, Andrews KM, Kelly J, and Grossman SP. Effects of chronic intrahypothalamic infusion of insulin on food intake and diurnal meal patterning in the rat. Behav Neurosci 104: 371-83, 1990.

250. McKibbin PE, Cotton SJ, McCarthy HD, and Williams G. The effect of dexamethasone on neuropeptide Y concentrations in specific hypothalamic regions. Life Sci 51: 1301-7, 1992.

251. McMinn JE, Sindelar DK, Havel PJ, and Schwartz MW. Leptin deficiency induced by fasting impairs the satiety response to cholecystokinin. Endocrinology 141: 4442-8, 2000.

252. McQuade JA, Benoit SC, Xu M, Woods SC, and Seeley RJ. High-fat diet induced adiposity in mice with targeted disruption of the dopamine-3 receptor gene. Behav Brain Res 2004;151(1-2):313-9

253. Meguid MM, Muscaritoli M, Beverly JL, The early cancer anorexia paradigm: changes in plasma free tryptophan and feeding indexes. JPEN; 16: 56-63, 1992.

254. Meguid MM, Ramos EJB, Laviano A, Varma M, Sato T, Chen C, Qi Y, and Das UN. Tumor anorexia: effects on neuropeptide Y and monoamines in paraventricular nucleus. Peptides 25: 261-6, 2004.

255. Meguid MM, Yang ZJ, and Gleason JR. The gut-brain axis on anorexia: toward an understanding of food intake regulation. Nutrition 12 Suppl 1: S57-S62, 1996.

256. Meister B, Ceccatelli S, Hokfelt T, Anden NE, Anden M, and Theodorsson E. Neurotransmitters, neuropeptides and binding sites in the rat mediobasal

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

69

hypothalamus: effects of monosodium glutamate (MSG) lesions. Exp Brain Res 76: 343-68, 1989.

257. Meister B. Control of food intake via leptin receptors in the hypothalamus. Vitam Horm 59: 265-304, 2000.

258. Mercer LD, Le VQ, Nunan J, Jones NM, and Beart PM. Direct visualization of cholecystokinin subtype2 receptors in rat central nervous system using anti-peptide antibodies. Neurosci Lett 293: 167-70, 2000.

259. Merchenthaler I, Lopez FJ, and Negro-Vilar A. Anatomy and physiology of central galanin-containing pathways. Prog Neurobiol 40: 711-69, 1993.

260. Michaud D, Anisman H, and Merali Z. Capsaicin-sensitive fibers are required for the anorexic action of systemic but not central bombesin. Am J Physiol 276: 1617-22, 1999.

261. Minamino N, Kangawa K, and Matsuo H. Neuromedin B and neuromedin C: Two mammalian bombesin-like peptides identified in porcine spinal cord and brain. Ann N Y Acad Sci 547: 373-90, 1988.

262. Mitchell JE, Lane DE, Morley JE, and Levine AS. Naloxone but not CCK-8 attenuates binge-eating in patients with the bulimia syndrome. Biol Psychiatry 21: 1399-1406, 1986.

263. Mizuno T, Kleopoulos S, Bergen H, Roberts L, Priest C, and Mobbs C. Hypothalamic pro-opiomelanocortin mRNA is reduced by fasting and in ob/ob and db/db mice, but is stimulated by leptin. Nature 47: 294-7, 1997.

264. Mollet A, Gilg S, Riediger T, and Lutz TA. Infusion of the amylin antagonist AC 187 into the area postrema increases food intake in rats. Physiol Behav 81: 149-55, 2004.

265. Mollet A, Meier S, Grabler V, Gilg S, Scharrer E, and Lutz TA. Endogenous amylin contributes to the anorectic effects of cholecystokinin and bombesin. Peptides 24: 91-8, 2003.

266. Moncada S, Palmer R, and Higgs EA. Biosynthesis of nitric oxide from L-arginine: a pathway for the regulation of cell function and communication. Biochem Pharmacol 38: 1709-1715, 1989.

267. Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH, Earley AR, Barnett AH, Prins JB, and O'Rahilly S. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387: 903-8, 1997.

268. Moore CX and Cooper GJ. Co-secretion of amylin and insulin from cultured islet beta-cells:modulation by nutrient secretagogues, islet hormones and hypoglycemic agents. Biochem Biophys Res Commun 179: 1-9, 1991.

269. Moran TH, Norgren R, Crosby RJ, and McHugh PR. Central and peripheral vagal transport of cholecystokinin binding sites occurs in afferent fibers. Brain Res 526: 95-102, 1990.

270. Moran TH, Smith GP, Hostetler AM, and McHugh PR. Transport of cholecystokinin (CCK) binding sites in subdiaphragmatic vagal branches. Brain Res 415: 149-52, 1987.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

70

271. Moriarty P, Dimaline R, Thompson DG, and Dockray GJ. Characterization of cholecystokininA and cholecystokininB receptors expressed by vagal afferent neurons. Neuroscience 79: 905-13, 1997.

272. Morley JE and Blundell JE. The neurobiological basis of eating disorders: some formulations. Biol Psychiatry 23: 53-78, 1988.

273. Morley JE, Farr SA, and Flood JF. Peripherally administered calcitonin gene-related peptide decreases food intake in mice. Peptides 17: 511-6, 1996.

274. Morley JE and Flood JF. Evidence that nitric oxide modulates food intake in mice. Life Sci 49: 707-11, 1991.

275. Morley JE, Levine AS, Gosnell BA, and Billington CJ. Neuropeptides and appetite: contribution of neuropharmacological modeling. Fed Proc 43: 2903-7, 1984.

276. Munroe DG, Gupta AK, Kooshesh F, Vyas TB, Rizkalla G, Wang H, Demchyshyn L, Yang ZJ, Kamboj RK, Chen H, McCallum K, Sumner-Smith M, Drucker DJ, and Crivici A. Prototypic G protein-coupled receptor for the intestinotrophic factor glucagon-like peptide 2. Proc Natl Acad Sci U S A 96: 1569-73, 1999.

277. Muroya S, Uramura K, Sakurai T, Takigawa M, and Yada T. Lowering glucose concentrations increases cytosolic Ca2+ in orexin neurons of the rat lateral hypothalamus. Neurosci Lett 309: 165-8, 2001.

278. Muscaritoli M, Meguid MM, Beverly JL. Mechanism of early tumor anorexia. J Surg Res 60: 389, 1996.

279. Nauck MA, Niedereichholz U, Ettler RI, Holst JJ, Orskov C, Ritzel RI, and Schmiegel WH. Glucagon-like peptide I inhibition of gastric emptying outweighs its insulinotropic effects in healthy humans. Am J Physiol 273: 981-8, 1997.

280. Navarro M, Rodriquez de Fonseca F, Alvarez E, Chowen JA, Zueco JA, Gomez R, Eng J, and Blazquez E. Colocalization of glucagon-like peptide-1 (GLP-1) receptors, glucose transporter GLUT-2, and glucokinase mRNAs in rat hypothalamic cells: evidence for a role of GLP-1 receptor agonists as an inhibitory signal for food and water intake. J Neurochem 67: 1982-91, 1996.

281. Nemeroff CB, Osbahr AJ 3rd, Manberg PJ, Ervin GN, and Prange AJ Jr. Alterations in nociception and body temperature after intracisternal administration of neurotensin, beta-endorphin, other endogenous peptides, and morphine. Proc Natl Acad Sci U S A 76: 5368-71, 1979.

282. Niehoff DL. Quantitative autoradiographic localization of cholecystokinin receptors in rat and guinea pig brain using 125I-Bolton-Hunter-CCK8. Peptides 10: 265-74, 1989.

283. Niijima A. The afferent discharges from sensors for interleukin 1 beta in the hepatoportal system in the anesthetized rat. J Auton Nerv Syst 61: 287-91, 1996.

284. Nillni EA, Vaslet C, Harris M, Hollenberg A, Bjorbak C, and Flier JS. Leptin regulates prothyrotropin-releasing hormone biosynthesis. Evidence for direct and indirect pathways. J Biol Chem 275: 36124-33, 2000.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

71

285. Nishino S, Ripley B, Overeem S, Lammers GJ, and Mignot E. Hypocretin (orexin) deficiency in human narcolepsy. Lancet 355: 39-40, 2000.

286. Nishiyama M, Makino S, Asaba K, and Hashimoto K. Leptin effects on the expression of type-2 CRH receptor mRNA in the ventromedial hypothalamus in the rat. J Neuroendocrinol 11: 307-14, 1999.

287. Niswender KD, Gallis B, Blevins JE, Corson MA, Schwartz MW, and Baskin DG. Immunocytochemical Detection of Phosphatidylinositol 3-kinase Activation by Insulin and Leptin. J Histochem Cytochem 51: 275-83, 2003.

288. Niswender KD and Schwartz MW. Insulin and leptin revisited: adiposity signals with overlapping physiological and intracellular signaling capabilities. Front Neuroendocrinol 24:1-10, 2003.

289. O’Shea D, Morgan D, Meeran K, Edwards C, Turton M, Choi S, Heath M, Gunn I, Taylor G, Howard J, Bloom C, Small C, Haddo O, Ma J, Callinan W, Smith D, Ghatei M, and Bloom S. Neuropeptide Y induced feeding in the rat is mediated by a novel receptor. Endocrinology 138: 196-202, 1997.

290. Obici S, Feng Z, Karkanias G, Baskin DG, and Rossetti L. Decreasing hypothalamic insulin receptors cause hyperphagia and insulin resistance in rats. Nat Neurosci 5: 566-72, 2002.

291. Obici S, Feng Z, Morgan K, Stein D, Karkanias G, and Rossetti L. Central administration of oleic acid inhibits glucose production and food intake. Diabetes 51: 271-75, 2002.

292. Ohkuma S, Narihara H, Katsura M, Hasegawa T, and Kuryama K. Nitric oxide-induced [3H]GABA release from cerebral cortical neurons is mediated by peroxynitrite. J Neurochem 65: 1109-14, 1995.

293. Ollmann M, Wilson B, Yang Y, Kerns J, Chen Y, Gantz I, and Barsh G. Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 278: 135-8, 1997.

294. Olson BR, Drutarosky MD, Chow MS, Hruby VJ, Stricker EM, and Verbalis JG. Oxytocin and an oxytocin agonist administered centrally decrease food intake in rats. Peptides 12: 113-8, 1991.

295. Olson BR, Drutarosky MD, Stricker EM, and Verbalis JG. Brain oxytocin receptors mediate corticotropin-releasing hormone-induced anorexia. Am J Physiol 260: 448-52, 1991.

296. Oohara M, Negishi M, Shimizu H, Sato N, and Mori M. Alpha-melanocyte stimulating hormone (MSH) antagonizes the anorexia by corticotropin releasing factor (CRF). Life Sci 53: 1473-7, 1993.

297. Orloff MS, Reeve JR Jr, Ben-Avram CM, Shively JE, and Walsh JH. Isolation and sequence analysis of human bombesin-like peptides. Peptides 5: 865-70, 1984.

298. Orosco M, Gerozissis K, Rouch C, and Nicolaidis S. Feeding-related immunoreactive insulin changes in the PVN-VMH revealed by microdialysis. Brain Res 671: 149-58, 1995.

299. Orskov C. Glucagon-like peptide-1, a new hormone of the entero-insular axis. Diabetologia 35: 701-11, 1992.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

72

300. Owen W, Lew N, Luì Y, and Lazarus J. The urea reduction ratio and serum albumin concentration as predictors of mortality in patients undergoing hemodialysis. N. Engl. J. Med 329: 1001-6, 1993.

301. Palmiter RD, Erickson JC, Hollopeter G, Baraban SC, and Schwartz MW. Life without neuropeptide Y. Recent Prog Horm Res 53: 163-99, 1998.

302. Papez J.W. A proposed mechanism of emotions. Arch Neurol Psychiat Chicago 8: 725-43, 1937.

303. Parada MA, Hernandez L, and Hoebel BG. Sulpiride injections in the lateral hypothalamus induce feeding and drinking in rats. Pharmacol Biochem Behav 30: 917-23, 1988.

304. Paroli E. Farmacologia generale speciale e clinica tossicologica. Roma: Soc Ed Universo, 1997.

305. Patel B, Koenig L, Kaplan L, and Hooi S. Increase in plasma leptin and Lep mRNA concentrations by food intake is dependent on insulin. Metab Clin Exp 47: 603-7, 1998.

306. Pelleymounter MA, Cullen MJ, Baker MB, Hetch R, Winters D, Boone T, and Collins F. Effect of the obese gene product on body weight regulation in ob/ob mice. Science 269: 540-543, 1995.

307. Peyron C and Charnay Y. Hypocretin (orexin) and CART peptides are co-expressed in human but not in rodents’ hypothalamus. FENS Meet Abstr Paris, 2002.

308. Plata-Salaman C. Cytokines and feeling suppression: an integrative view from neurologic to molecular levels. Nutrition 11: S674-7, 1995.

309. Plata-Salaman CR, Ilyin SE, and Gayle D. Brain cytokine mRNAs in anorectic rats bearing prostate adenocarcinoma tumor cells. Am J Physiol 275: 566-73, 1998.

310. Plata-Salaman. Food intake suppression by immunomodulators. Neurosi Res Commun 3: 159-65, 1988.

311. Porte Jr D, Baskin DG, and Schwartz MW. Leptin and insulin action in the central nervous system. Nutr Rev 60: S20-S29, 2002.

312. Qu D, Ludwig DS, Gammeltoft S, Piper M, Pelleymounter MA, Cullen MJ, Mathes WF, Przypek R, Kanarek R, and Maratos-Flier E. A role for melanin-concentrating hormone in the central regulation of feeding behaviour. Nature 380: 243-7, 1996.

313. Ragnauth A, Moroz M, and Bodnar RJ. Multiple opioid receptors mediate feeding elicited by mu and delta opioid receptor subtype agonists in the nucleus accumbens shell in rats. Brain Res 876: 76-87, 2000.

314. Ramos EJ, Suzuki S, Marks D, Inui A, Asakawa A, and Meguid MM. Cancer anorexia-cachexia syndrome: cytokines and neuropeptides. Curr Opin Clin Nutr Metab Care 7: 427-434, 2004.

315. Rehfeld JF. Cholecystokinin as satiety signal. Int J Obes 5: 465-9, 1981. 316. Rinaman L, Hoffman GE, Dohanics J, Le WW, Stricker EM, and Verbalis JG.

Cholecystokinin activates catecholaminergic neurons in the caudal medulla

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

73

that innervate the paraventricular nucleus of the hypothalamus in rats. J Comp Neurol 360: 246-56, 1995.

317. Rohner-Jeanrenaud F, Craft LS, Bridwell J, Suter TM, Tinsley FC, Smiley DL, Burkhart DR, Statnick MA, Heiman ML, Ravussin E, and Caro JF. Chronic central infusion of cocaine- and amphetamine-regulated transcript (CART 55-102): effects on body weight homeostasis in lean and high-fat-fed obese rats. Int J Obes Relat Metab Disord 26: 143-9, 2002.

318. Rohner-Jeanrenaud F. Hormonal regulation of energy partitioning. Int J Obes Relat Metab Disord 24 Suppl 2: S4-7, 2000.

319. Rohner-Jeanrenaud F. Neuroendocrine regulation of nutrient partitioning. Ann N Y Acad Sci 892: 261-71, 1999.

320. Romon M, Lebel P, Velly C, Marecaux N, Fruchart JC, and Dallongeville J. Leptin response to carbohydrate or fat meal and association with subsequent satiety and energy intake. Am J Physiol 277: 855-61, 1999.

321. Rossi Fanelli F, Cangiano C, Ceci F, Cellerino R, Fraschi F, Menichetti ET. Plasma tryptophan and anorexia in human cancer. Eur J Cancer Clin Oncol 22: 89-95, 1986.

322. Rossi Fanelli F and Cangiano C. Fisiopatologia del comportamento alimentare. In: “Manuale di nutrizione artificiale” di Bozzetti F, Guarnieri G, Milano: Masson, pp.55-61, 1992.

323. Rossi Fanelli F and Cangiano C. Increased availability of tryptophan in brain as common pathogenic mechanism for anorexia associated with different diseases. Nutrition 7: 364-367, 1991.

324. Rossi Fanelli F and Laviano A. Cancer anorexia: a model for the understanding and treatment of secondary anorexia. Int J Cardiol 85: 67-72, 2002.

325. Ruffin M and Nicolaidis S. Intracerebroventricular injection of murine leptin enhances the postprandial metabolic rate in the rat. Brain Res 874: 30-6, 2000.

326. Sahu A, Kalra PS, and Kalra SP. Food deprivation and ingestion induce reciprocal chenges in neuropeptide Y concentrations in the periventricular nucleus. Peptides 9: 83-6, 1988.

327. Sahu A. Evidence suggesting that galanin (GAL), melanin-concentrating hormone (MCH), neurotensin (NT), proopiomelanocortin (POMC) and neuropeptide Y (NPY) are targets of leptin signaling in the hypothalamus. Endocrinology 139: 795-798, 1998.

328. Sakaguchi T and Bray GA. Effect of norepinephrine, 5-HT and tryptophan on the firing rate of sympathetic nerves. Brain Res 492: 271-80, 1989.

329. Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, Williams SC, Richardson JA, Kozlowski GP, Wilson S, Arch JR, Buckingham RE, Haynes AC, Carr SA, Annan RS, McNulty DE, Liu WS, Terrett JA, Elshourbagy NA, Bergsma DJ, and Yanagisawa M. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92: 573-85, 1998.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

74

330. Sakurai T. Orexin: a link between energy homeostasis and adaptive behaviour. Curr Opin Clin Nutr Metab Care 6: 353-60, 2003.

331. Saladin R, De Vos P, Guerre-Millo M, Leturque A, Girard J, Staels B, and Auwerx J. Transient increase in obese gene expression after food intake or insulin administration. Nature 377: 527-9, 1995.

332. Schick RR, Schusdziarra V, Nussbaumer C, and Classen M. Neuropeptide Y and food intake in fasted rats: effect of naloxone and site of action. Brain Res 552: 232-9, 1991.

333. Schols AM, Soeters PB, Mostert R, Saris WH, and Wouters EF. Energy balance in chronic obstructive pulmonary disease. Am Rev Respir Dis 143: 1248-52, 1991.

334. Schwartz GJ, McHugh PR, and Moran TH. Gastric loads and cholecystokinin synergistically stimulate rat gastric vagal afferents. Am J Physiol 265: 872-6, 1993.

335. Schwartz GJ, McHugh PR, and Moran TH. Integration of vagal afferent responses to gastric loads and cholecystokinin in rats. Am J Physiol 261: 64-9, 1991.

336. Schwartz GJ, Moran TH, White WO, and Ladenheim EE. Relationships between gastric motility and gastric vagal afferent responses to CCK and GRP in rats differ. Am J Physiol 272: 1726-33, 1997.

337. Schwartz M, Erickson J, Baskin D, and Palmiter R. Effect of fasting and leptin deficiency on hypothalamic neuropeptide Y gene transcription in vivo revealed by expression of a lacZ reporter gene. Endocrinology 139: 2629-35, 1998.

338. Schwartz MW, Figlewicz DP, Baskin DG, Woods SC , and Porte Jr D. Insulin in the brain: a hormonal regulator of energy balance. Endocr Rev 13: 387-414, 1992.

339. Schwartz MW, Sipols AJ, Marks JL, Sanacora G, White JD, Scheurink A, Kahn SE, Baskin DG, Woods SC, Figlewicz D, and Porte Jr D. Inhibition of hypothalamic NPY gene expression by insulin. Endocrinology 130: 3608-16, 1992.

340. Segal-Lieberman G, Bradley RL, Kokkotou E, Carlson M, Trombly DJ, Wang X, Bates S, Myers MG Jr, Flier JS, and Maratos-Flier E. Melanin-concentrating hormone is a critical mediator of the leptin-deficient phenotype. Proc Natl Acad Sci U S A 100: 10085-90, 2003.

341. Seilicovich A, Duvilanski BH, Pisera D, Theas S, Gimeno M, Rettori V, and McCann SM. Nitric oxide inhibits hypothalamic luteinizing hormone-releasing hormone release by releasing gamma-aminobutyric acid. Proc Natl Acad Sci U S A 92: 3421-4, 1995.

342. Seoane LM, Carro E, Tovar S, Casanueva FF, and Dieguez C. Regulation of in vivo TSH secretion by leptin. Regul Pept 92: 25-9, 2000.

343. Shaw MJ, Hughes JJ, Morley JE, Levine AS, Silvis SE, and Shafer RB. Cholecystokinin octapeptide action on gastric emptying and food intake in normal and vagotomized man. Ann NY Acad Sci 448: 640-651, 1985.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

75

344. Shiiya T, Nakazato M, Mizuta M, Date Y, Mondal MS, Tanaka M, Nozoe S, Hosoda H, Kangawa K, and Matsukura S. Plasma ghrelin levels in lean and obese humans and the effect of glucose on ghrelin secretion. J Clin Endocrinol Metab 87: 240-4, 2002.

345. Shimada M, Tritos NA, Lowell BB, Flier JS, and Maratos-Flier E. Mice lacking melanin-concentrating hormone are hypophagic and lean. Nature 396: 670-4, 1998.

346. Shimizu H, Shargill N, Bray G, Yen T, and Gesellchen P. Effects of MSH on food intake, body weight and coat color of the yellow obese mouse. Life Sci 45: 543-52, 1989.

347. Shintani F, Kanba S, Nakaki T, Nibuya M, Kinoshita N, Suzuki E, Yagi G, Kato R, and Asai M. Interleukin-1 beta augments release of norepinephrine, dopamine, and serotonin in the rat anterior hypothalamus. J Neurosci 13: 3574-81, 1993.

348. Shintani M, Ogawa Y, Ebihara K, Aizawa-Abe M, Miyanaga F, Takaya K, Hayashi T, Inoue G, Hosoda K, Kojima M, Kangawa K, and Nakao K. 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: 227-32, 2001.

349. Shiraishi T, Oomura Y, Sasaki K, and Wayner MJ. Effects of leptin and orexin-A on food intake and feeding related hypothalamic neurons. Physiol Behav 71: 251-61, 2000.

350. Shutter JR, Kinsey AC, Scully S, Luthy R, and Stark KL. Hypothalamic expression of ART, a novel gene related to agouti, is up-regulated in obese and diabetic mutant mice. Genes Dev 11: 593-602, 1997.

351. Shyamaladevi N, Jayakumar AR, Sujatha R, Paul V, and Subramanian EH. Evidence that nitric oxide production increases gamma-amino butyric acid permeability of blood-brain barrier. Brain Res Bull 57: 231-6, 2002.

352. Sipols AJ, Baskin DG, and Schwartz MW. Effect of intracerebroventricular insulin infusion on diabetic hyperphagia and hypothalamic neuropeptide gene expression. Diabetes 44: 147-51, 1995.

353. Smith GP, Jerome C, Cushin BJ, Eterno R, and Simansky KJ. Abdominal vagotomy blocks the satiety effect of cholecystokinin in the rat. Science 213: 1036-7, 1981.

354. Spina M, Merlo-Pich E, Chan RK, Basso AM, Rivier J, Vale W, and Koob GF. Appetite-suppressing effects of urocortin, a CRF-related neuropeptide. Science 273: 1561-4, 1996.

355. Squadrito F, Calapai G, Altavilla D, Cucinotta D, Zingarelli B, Arcoraci V, Campo GM, and Caputi AP.Central serotoninergic system involvement in the anorexia induced by Nω-nitro-L-arginine, an inhibitor of nitric oxide synthase. Eur J Pharmacol 255: 51-5, 1994.

356. Squadrito F, Calapai G, Altavilla D, Cucinotta D, Zingarelli B, Campo GM, Arcoraci V, Sautebin L, Mazzaglia G, and Caputi AP. Food deprivation

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

76

increases brain nitric oxide synthase and depresses brain 5-HT levels in rats. Neuropharmacology 33: 83-6, 1994.

357. Squadrito F, Calapai G, Cucinotta D, Altavilla D, Zingarelli B, Ioculano M, Urna G, Sardella A, Campo GM, and Caputi AP. Anorectic activity of Nω-nitro-L-arginine, an inhibitor of brain nitric oxide synthase, in obese Zucker rats. Eur J Pharmacol 230: 125-8, 1993.

358. Stanley B, Kyrkouli S, Lampert S, and Leibowitz S. Neuropeptide Y chronically injected into the hypothalamus: a powerful neurochemical inducer of hyperphagia and obesity. Peptides 7: 1189-92, 1986.

359. Stanley BG, Lanthier D, Chin AS, and Leibowitz SF. Suppression of neuropeptide Y-elicited eating by adrenalectomy or hypophysectomy: reversal with corticosterone. Brain Res 501: 32-6, 1989.

360. Steckler T and Holsboer F. Corticotropin-releasing hormone receptor subtypes and emotion. Biol Psychiatry 46: 1480-508, 1999.

361. Steiner RA, Hohmann JG, Holmes AN, Wrenn CC, Cadd G, Jureus A, Clifton OK, Luo M, Gutahall M, Ma SY, Mufton EJ, and Crawley JN. Galanin transgenic mice display cognitive and neurochemical deficits characteristic of Alzheimer’s disease. Proc Natl Acad Sci 98: 4184-9, 2001.

362. Stephens TW, Basinski M, Bristow PK, Bue-Vallesky JM, Burgett SG, Craft L, Hale J, Hoffmann J, Hsiung HM, Kriauciunas A, et al. The role of neuropeptide Y in antiobesity action of the obese gene product. Nature 377: 530-532, 1995.

363. Stricker-Krongrad A, Dimitrov T, and Beck B. Central and peripheral dysregulation of melanin-concentrating hormone in obese Zucker rats. Brain Res Mol Brain Res 92: 43-8, 2001.

364. Strubbe JH and Mein CG. Increased feeding in response to bilateral injection of insulin antibodies in the VMH. Physiol Behav 19: 309-13, 1977.

365. Szczypka MS, Kwok K, Brot MD, Marck BT, Matsumoto AM, Donahue BA, and Palmiter RD. Dopamine production in the caudate putamen restores feeding in dopamine-deficient mice. Neuron 30: 819-28, 2001.

366. Szczypka MS, Rainey MA, Kim DS, Alaynick WA, Marck BT, Matsumoto AM, and Palmiter RD. Feeding behavior in dopamine-deficient mice. Proc Natl Acad Sci U S A 96: 12138-43, 1999.

367. Tagliamonte A, Biggio G, Vargiu L, and Gessa GL. Free tryptophan in serum controls brain tryptophan levels and serotonin synthesis. Life Sci 12: 277-287, 1973.

368. Tang-Christensen M, Larsen P1, Thulesen I, Romer I, and Vrang N. The proglucagon-derived peptide, glucagon-like peptide-2, is a neurotransmitter involved in the regulation of food intake. Nat Med 6: 802-7, 2000.

369. Tatemoto K, Carlquist M, and Mutt V. Neuropeptide Y: a novel brain peptide with structural similarities to peptide YY and pancreatic polypeptide. Nature 89: 1174, 1982.

370. Tempel DL, Leibowitz KJ, Smith D, and Leibowitz SF. Galanin in the paraventricular nucleus preferentially enhances fat intake. Proc Soc Neurosci

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

77

12: 59-600, 1986. 371. Ter Horst GJ, de Boer P, Luiten PG, and van Willigen JD. Ascending

projections from the solitary tract nucleus to the hypothalamus. A Phaseolus vulgaris lectin tracing study in the rat. Neuroscience 31: 785-97, 1989.

372. Thorens B. Expression cloning of the pancreatic beta cell receptor for the gluco-incretin hormone glucagon-like peptide 1. Proc Natl Acad Sci USA 89: 8641-5, 1992.

373. Trikha M, Corringham R, Klein B, and Rossi JF. Targeted anti-interleukin-6 monoclonal antibody therapy for cancer: a review of the rationale and clinical evidence. Clin Cancer Res 9: 4653-65, 2003.

374. Tritos NA and Maratos-Flier E. Two important systems in energy homeostasis: melanocortins and melanin-concentrating hormone. Neuropeptides 33: 339-49, 1999.

375. Tritos NA, Mastaitis JW, Kokkotou E, and Maratos-Flier E. Characterization of melanin concentrating hormone and preproorexin expression in the murine hypothalamus. Brain Res 895: 160-6, 2001.

376. Tritos NA, Vicent D, Gillette J, Ludwig DS, Flier ES, and Maratos-Flier E. Functional interactions between melanin-concentrating hormone, neuropeptide Y, and anorectic neuropeptides in the rat hypothalamus. Diabetes Nov 47: 1687-92, 1998.

377. Tschop M, Smiley DL, and Heiman ML. Ghrelin induces adiposity in rodents. Nature 407: 908-13, 2000.

378. Turrin NP, Ilyin SE, Gayle DA, Plata-Salaman CR, Ramos EJ, Laviano A, Das UN, Inui A, and Meguid MM. Interleukin-1beta system in anorectic catabolic tumor-bearing rats. Curr Opin Clin Nutr Metab Care 7: 419-426, 2004.

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

380. Ukkola O, Ravussin E, Jacobson P, Perusse L, Rankinen T, Tschop M, Heiman ML, Leon AS, Rao DC, Skinner JS, Wilmore JH, Sjostrom L, and Bouchard C. Role of ghrelin polymorphisms in obesity based on three different studies. Obes Res 10: 782-91, 2002.

381. Vaisse C, Clement K, Durand E, Hercberg S, Guy-Grand B, and Froguel P. Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity. J Clin Invest 106: 253-62, 2000.

382. Van Heek M, Compton DS, France CF, Tedesco RP, Fawzi AB, Graziano MP, Sybertz EJ, Strader CD, and Davis HR Jr. Diet-induced obese mice develop peripheral, but not central resistance to leptin. J Clin Invest 99: 385-90, 1997.

383. Verbalis JG, Mangione MP, and Stricker EM. Oxytocin produces natriuresis in rats at physiological plasma concentrations. Endocrinology 128: 1317-22, 199l.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

78

384. Voigt JP, Kienzle F, Sohr R, Rex A, and Fink H. Feeding and 8-OH-DPAT-related release of serotonin in the rat lateral hypothalamus. Pharmacol Biochem Behav 65: 183-9, 2000.

385. Vrang N, Larsen PJ, Kristensen P, and Tang-Christensen M. Central administration of cocaine-amphetamine-regulated transcript activates hypothalamic neuroendocrine neurons in the rat. Endocrinology 141: 794-801, 2000.

386. Walther DJ and Bad$er M. A unique central tryptophan hydroxylase isoform. Biochem Pharmacol 66: 1673-80, 2003.

387. Wang J and Dunn AJ. Mouse interleukin-6 stimulates the HPA axis and increases brain tryptophan and serotonin metabolism. Neurochem Int 33: 143-54, 1998.

388. Weiss GF and Leibowitz SF. Efferent projection from the paraventricular nucleus mediating alpha-2 noradrenergic feeding. Brain Res 347: 225-238, 1985.

389. West DB, Fey D, and Woods SC. Cholecystokinin persistently suppresses meal size but not food intake in free-feeding rats. Am J Physiol 246: 776-87, 1984.

390. White BD, Dean RG, and Martin RJ. Adrenalectomy decreases neuropeptide Y mRNA levels in the arcuate nucleus. Brain Res Bull 25: 711-5, 1990.

391. Will MJ, Franzblau EB, and Kelley AE. Nucleus accumbens mu-opioids regulate intake of a high-fat diet via activation of a distributed brain network. J Neurosci 23: 2882-8, 2003.

392. Williams G, Bing C, Cai XJ, Harrold JA, King JP, and Liu XH. The hypothalamus and the control of energy homeostasis, Different circuits, different perpuses. Physiol Behav 74: 683-701, 2001.

393. Williams G, Cai XJ, Elliott JC, and Harrold JA. Anabolic neuropeptides. Physiol Behav 81: 211-22, 2004.

394. Williams G, Cardoso H, Lee YC, Ghatei MA, Flatt PR, Bailey CJ, and Bloom SR. Reduced hypothalamic neurotensin concentrations in the genetically obese diabetic (ob/ob) mouse: possible relationship to obesity. Metabolism 40: 1112-6, 1991.

395. Wisialowski T, Parker R, Preston E, Sainsbury A, Kraegen E, Herzog H, and Cooney G. Adrenalectomy reduces neuropeptide Y-induced insulin release and NPY receptor expression in the rat ventromedial hypothalamus. J Clin Invest 105: 1253-9, 2000.

396. Witte KKA and Clark AL. Nutritional abnormalities contributing to cachexia in chronic illness. Int J Cardiol 85: 23-31, 2002.

397. Wong S and Pinkney J. Role of cytokines in regulating feeding behaviour. Curr Drug Targets ;5: 251-63, 2004.

398. Woods SC, Lotter EC, McKay D, and Porte Jr D. Chronic intracerebroventricular infusion of insulin reduces food intake and body weight in baboons. Nature 282: 503-5, 1979.

399. Woods SC and Seeley RJ. Adiposity signals and the control of energy homeostasis. Nutrition 16: 894-902, 2000.

Role of the Neurotransmitters and Peptides Involved in feeding behaviour control

79

400. Wren AM, Seal LJ, Cohen MA, Brynes AE, Frost GS, Murphy KG, Dhillo WS, Ghatei MA, and Bloom SR. Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab 86: 5992, 2001.

401. Wurtman RJ and Fernström JD. Control of brain neurotransmitter synthesis by precursor availability and nutritional state. Biochem Pharmacol 25: 1691, 1976.

402. Wurtman RJ and Wurtman JJ. Nutrients, neurotrasmitter synthesis, and the control of food intake. In Stunkard A, Stellar GE: Eating and its disorders. New York: Raven Press, 1985:77-86.

403. Wurtman RJ. When and why should nutritional state control neurotransmitter synthesis? J Neural Transmission Suppl 15:69, 1979.

404. Xu B, Dube MG, Kalra PS, Farmerie WG, Kaibara A, Moldawer LL, Martin D, and Kalra SP. Anorectic effects of the cytokines, Ciliary Neurotropic Factor, are mediated by hypothalamic Neuropeptide Y: comparison with leptin. Endocrinology 139: 466-73, 1998.

405. Yamada J, Sugimoto Y, and Ujikawa M. The serotonin precursor 5-hydroxytryptophan elevates serum leptin levels in mice. Eur J Pharmacol 383: 49-51, 1999.

406. Zahniser NR, Goens MB, Hanaway PJ, and Vinych JV. Characterization and regulation of insulin receptors in rat brain. J Neurochem 42: 1354-62, 1984.

407. Zakrzewska KE, Cusin I, Sainsbury A, Rohner Jeanrenaud F, and Jeanrenaud B. Glucocorticoids as counterregulatory hormones of leptin: toward an understanding of leptin resistance. Diabetes 46: 717-9, 1997.

408. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, and Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature 372: 425-432, 1994.

409. Zhao AZ, Huan JN, Gupta S, Pal R, and Sahu A. A phosphatidylinositol 3-kinase phosphodiesterase 3B-cyclic AMP pathway in hypothalamic action of leptin on feeding. Nat Neurosci 5: 727-28, 2002.

410. Zhao WQ, Don JT, Liu QW, and Alkon DC. Evidence for locally produced insulin in the adult rat brain as a neuroactive peptide. 32nd SEN meeting, November 2002b, Orlando, FL, USA. 375.12

411. Zhou QY and Palmiter RD. Dopamine-deficient mice are severely hypoactive, adipsic, and aphagic. Cell 83: 1197-209, 1995.

AREE SCIENTIFICO–DISCIPLINARI

Area 01 – Scienze matematiche e informatiche

Area 02 – Scienze fisiche

Area 03 – Scienze chimiche

Area 04 – Scienze della terra

Area 05 – Scienze biologiche

Area 06 – Scienze mediche

Area 07 – Scienze agrarie e veterinarie

Area 08 – Ingegneria civile e Architettura

Area 09 – Ingegneria industriale e dell’informazione

Area 10 – Scienze dell’antichità, filologico–letterarie e storico–artistiche

Area 11 – Scienze storiche, filosofiche, pedagogiche e psicologiche

Area 12 – Scienze giuridiche

Area 13 – Scienze economiche e statistiche

Area 14 – Scienze politiche e sociali

Le pubblicazioni di Aracne editrice sono su

www.aracneeditrice.it

Finito di stampare nel mese di febbraio del 2007dalla tipografia « Braille Gamma S.r.l. » di Santa Rufina di Cittaducale (Ri)

per conto della « Aracne editrice S.r.l. » di Roma