Food Intake Review 2005

download Food Intake Review 2005

of 27

Transcript of Food Intake Review 2005

  • 8/12/2019 Food Intake Review 2005

    1/27

    REVIEW

    Brain regulation of food intake and appetite: molecules and

    networks

    C . B R O B E R G E RFrom the Department of Neuroscience, Karolinska Institute, Stockholm, Sweden

    Abstract. Broberger C (Karolinska Institute,

    Stockholm, Sweden). Brain regulation of foodintake and appetite: molecules and networks

    (Review). J Intern Med2005; 258: 301327.

    In the clinic, obesity and anorexia constitute

    prevalent problems whose manifestations are

    encountered in virtually every field of medicine.

    However, as the command centre for regulating food

    intake and energy metabolism is located in the

    brain, the basic neuroscientist sees in the same

    disorders malfunctions of a model network for how

    integration of diverse sensory inputs leads to a

    coordinated behavioural, endocrine and autonomicresponse. The two approaches are not mutually

    exclusive; rather, much can be gained by combining

    both perspectives to understand the pathophysiology

    of over- and underweight. The present review

    summarizes recent advances in this field including

    the characterization of peripheral metabolic signals

    to the brain such as leptin, insulin, peptide YY,

    ghrelin and lipid mediators as well as the vagusnerve; signalling of the metabolic sensors in the

    brainstem and hypothalamus via, e.g. neuropeptide

    Y and melanocortin peptides; integration and

    coordination of brain-mediated responses to

    nutritional challenges; the organization of food

    intake in simple model organisms; the mechanisms

    underlying food reward and processing of the

    sensory and metabolic properties of food in the

    cerebral cortex; and the development of the central

    metabolic system, as well as its pathological

    regulation in cancer and infections. Finally,

    recent findings on the genetics of human obesityare summarized, as well as the potential for novel

    treatments of body weight disorders.

    Keywords: brainstem, cerebral cortex, feeding,

    hypothalamus, metabolic, reward.

    Introduction

    Few tasks executed by the brain hold greater

    survival value than keeping us fed and in adequate

    nutritional state. It is not surprising then that the

    central nervous system has developed a meticu-

    lously interconnected circuitry to meet this chal-lenge. A consequence of this organization is that an

    energy-dense environment favours the development

    of obesity, whilst overcompensation may shut down

    the drive to feed. In todays society where evolving

    disease demographics and lifestyle allow for a

    greater diversity of metabolic phenotypes than

    perhaps ever before [1] disorders of both extremes

    of energy intake are common in health care.

    This paper builds partly on presentations made at a

    Nobel Conference on Brain Control of Feeding Behaviour

    organized at the Karolinska Institute, Stockholm, Sweden,

    in September 911, 2004.

    Journal of Internal Medicine 2005; 258: 301327 doi:10.1111/j.1365-2796.2005.01553.x

    2005 Blackwell Publishing Ltd 301

  • 8/12/2019 Food Intake Review 2005

    2/27

    Obesity is increasing at an alarming rate in

    industrialized and developing countries alike [2]

    and is associated with a wealth of conditions

    afflicting virtually all organ systems [3, 4]. Exam-

    ples diverge widely to include cholelithiasis [5],

    osteoarthritis [6], infertility [7], stroke [8], cutane-ous infections [9], wound healing deficiencies [10],

    as well as a general increase in mortality [11]

    and social and professional stigmatization [12].

    The urgency of the problem is illustrated dramat-

    ically by the previous rarity of paediatric obesity-

    associated type 2 diabetes, which is increasing to

    the point of taking over as the leading cause of

    childhood diabetes [13]. The opposite extreme of

    anorexia and hypophagia includes not only anor-

    exia nervosa [14] but is also a common and

    potentially fatal complication of infections [15],

    malignancies [16] and ageing [17].Unlike many other common diseases, these disor-

    ders have an obvious solution: adjusting food intake

    and exercise until normal body weight has been

    restored. However, it is no great revelation that this

    solution is as simple as it has repeatedly proved

    elusive [18]. Experimental studies confirm the com-

    mon knowledge that weight loss almost always is

    followed by a rapid return to initial weight once the

    anorexigenic regimen is terminated [19]. Notably,

    the same applies to humans subjected to voluntary

    overfeeding [20], supporting the concept of a tightly

    regulated set-point for body weight. Treatment ofeating disorders has been remarkably unsuccessful

    a consequence possibly of that we are battling

    ancient systems maintained by thrifty genes that

    favour the preservation of energy stores [21]. Avail-

    able options for pharmacological therapy leave much

    to be desired, and compounds that have been

    introduced for obesity management have subse-

    quently often been withdrawn due to intolerable

    side-effects [22]. The most effective obesity treatment

    at present is bariatric surgery, but this is a

    complicated procedure not without adverse effects

    [23]. Preventive measures have frustratingly lim-ited effect. It has proved even more difficult to

    devise strategies for increasing food intake in cases

    of anorexia. Although some success has been

    reported with behavioural approaches for anorexia

    nervosa [24], the more common forms of cancer-

    and inflammation-associated anorexia remain a

    major therapeutic challenge. Novel treatments are

    greatly needed.

    But what systems should such treatments target?

    Early clinical observations that patients with pituit-

    ary tumours and accompanying injury to the base of

    the brain develop obesity [2528], inspired experi-

    mental lesion studies [2933], which demonstrated

    that damage to particular regions of the hypotha-lamus and brainstem lead to profound, often fatal,

    alterations of feeding behaviour. It also became

    apparent that signals from the peripheral energy

    stores [34] and gastrointestinal canal [35] provide

    essential cues for appetite and satiety. Based on

    these and other findings, Stellar [36] half a century

    ago proposed a dual centre hypothesis for the

    initiation of motivated behaviour. The hypothesis

    included both mechanisms for sensing peripheral

    cues, separate nuclei (i.e. the dual centres) for

    stimulating and inhibiting behaviour, and connec-

    tions between the hypothalamus and higher brainregions to allow for internal states to determine the

    threshold for initiating behaviour. Of all motivated

    behaviours, the model is perhaps most applicable for

    food intake. Yet, for all its heuristic value, the dual

    centre hypothesis was as low on specifics as it was

    laden with theory. Research dating in particular

    from the last decade has changed this. Today, we

    have an understanding of the circuitry and neuro-

    pharmacology of feeding behaviour that can be

    probed for therapeutic targets. The present article is

    not an exhaustive review of the central control of

    energy metabolism [37, 38], but summarizes recentadvances, which have brought new insight into the

    peripheral signals describing the metabolic state to

    the brain; the input stations in the hypothalamus

    and brainstem sensitive to these signals, the organ-

    ization of feeding behaviour in simple and complex

    organisms; the link between food intake and energy

    expenditure; the neural framework for integrating

    metabolic cues and reward properties; the mecha-

    nisms of infection- and cancer-associated anorexia;

    developmental and genetic causes of obesity and

    novel therapeutic strategies.

    A central framework for sensing andorchestrating energy metabolism

    The regulation of energy metabolism presents a

    prototypical homeostatic system, with the brain

    acting as the central coordinator and rectifier

    (Fig. 1). It is one of the great wonders of the brain

    that body weight stays remarkably fixed (as a

    2005 Blackwell Publishing Ltd Journal of Internal Medicine258: 301327

    3 02 C . B RO BE RG ER

  • 8/12/2019 Food Intake Review 2005

    3/27

    set-point) most of the time in most people [39]. The

    first step in maintaining this homeostasis is for

    the brain to inform itself of the metabolic status of

    the individual, which it does through two main

    channels. First, hormonal signals reflecting the

    availability and demand for metabolic fuel is relayed

    via neurones in the hypothalamus. The receptors for

    these signals are primarily expressed on two neuro-

    chemically distinct sets of neurones located in thearcuate nucleus (Arc) in the mediobasal hypothala-

    mus, alongside the third ventricle [40]. One neurone

    group expresses neuropeptide Y (NPY); increasing

    NPY release or activation of these neurones by a

    variety of approaches results in increased food

    intake and decreased energy expenditure. The other

    group expresses the neuropeptide precursor pro-

    opiomelanocortin (POMC), which is processed to

    melanocortin peptides; activation of these neurones

    has the opposite effect of triggering the NPY cells, i.e.

    decreased food intake and increased energy expen-

    diture. The yinyang relationship between the twoArc groups is further underscored by their opposite

    regulation by leptin and insulin, hormones signal-

    ling metabolic affluence, which decrease the expres-

    sion of NPY, whilst they increase the expression of

    POMC. The second main input for information

    pertaining to energy balance is the brainstem,

    classically viewed as a channel for visceroceptive

    information as cranial nerves, in particular the

    vagus nerve, carrying information from the aliment-

    ary organs enter the brain here. Vagal afferents

    synapse onto [41, 42] and excite [43] neurones in

    the brainstem nucleus tractus solitarii (nTS). Fromboth the hypothalamus and the brainstem, projec-

    tions fan further into the brain to engage other brain

    regions in the initiation and organization of food

    intake. As in all homeostatic systems, the brain has

    at its disposal three effector pathways to activate

    when the controlled variable (i.e. body weight)

    needs to be adjusted: behaviour (i.e. food intake), the

    endocrine system and the autonomic nervous sys-

    tem [44]. Importantly, all three of these systems are

    engaged downstream of the Arc and nTS stations to

    provide a synchronized response to fluctuations in

    energy balance; the first primarily in the volitionalcontrol of intake, the latter two regulate energy

    expenditure.

    Peripheral control of feeding behaviour

    Metabolic state is reflected in a diverse array of

    signals of the brain. Recent investigations have shed

    light on some of the key hormones and the vagal

    ARC

    POMC

    NPY

    GHRELIN

    OEA

    PYY

    VAGUS

    LEPTIN

    DMX

    IML

    INSULIN

    nTS

    Fig. 1 The central metabolic circuitry is regulated by numerous

    endocrine and neural inputs. Schematic illustration of how brain

    networks regulating ingestive behaviour communicate with per-

    ipheral organs. Hormones supplying information about the per-

    ipheral metabolic state to the brain include the gastrointestinalpeptides ghrelin and PYY(3-36), insulin from the pancreas and

    leptin from adipose tissue. Ghrelin and leptin act both on the

    hypothalamus (Arc) and the brainstem (nTS). The afferent por-

    tion of the vagus nerve innervates most of the gastrointestinal

    tract where it collects information about the immediate aliment-

    ary state, and terminates in the nTS. The lipid mediator OEA is

    produced in the duodenum and activates the brainstem, possibly

    via the vagus nerve. Both the Arc (via antagonistic NPY- and

    POMC-expressing cells) and the nTS project further into the brain

    in parallel pathways to engage higher brain regions into ingestive

    behaviour. Outputs from the brain regulating energy expenditure

    include both branches of the autonomic nervous system; the

    sympathetic system whose preganglionic neurones are located in

    the intermediolateral cell column (IML), which is directly inner-

    vated by POMC neurones from the Arc, as well as the parasym-pathetic system with preganglionic neurones for the efferent

    portion of the vagus nerve located in the dorsal motor nucleus of

    the vagus (DMX). The efferent autonomic innervation regulates,

    e.g. glucose homeostasis via actions in liver and skeletal muscle.

    See text for details.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine 258: 301327

    R E V I EW : B R A I N C O N T RO L O F F O O D I N T A KE 3 0 3

  • 8/12/2019 Food Intake Review 2005

    4/27

    mechanisms that shape the central response to

    nutritional challenges (Fig. 1).

    The vagus nerve

    The gastrointestinal canal is equipped with a myriadof sensory receptors along its full crown-rump

    extension [45]. Thus, the taste, texture and

    mechanic stress of food is reported to the brain to

    provide an online description of the immediate

    alimentary state. This information is routed to the

    nTS primarily via the afferent portion of the vagus

    nerve, so that vagal activation causes satiation and

    meal termination. (Parallel neural feedback is also

    supplied by sensory neurones innervating the oral

    cavity mediating, e.g. taste, and lesser-studied

    splanchnic nerves [46].) Vagal afferents are broadly

    sensitive to gastrointestinal signals, including gas-troduodenal distension, the presence of chemically

    distinct nutrients within the gastrointestinal tract as

    well as peptides produced by endocrine cells in the

    gut wall, most prominently cholecystokinin (CCK), a

    well-characterized satiety signal [47, 48]. Import-

    antly, these signals are integrated within the indi-

    vidual vagal sensory neurone prior to the signal

    being relayed in the nTS [49, 50]. The neurochem-

    ical identity of viscerosensory vagal neurones has

    remained enigmatic, but these cells likely signal via

    glutamate [51] and the neuropeptide cocaine- and

    amphetamine-regulated transcript [52], whichinhibits feeding upon brainstem administration [53].

    Leptin

    An appetite-regulating endocrine signal from fat

    tissue maintaining energy homeostasis had been

    hypothesized already with parabiosis experiments

    in the 1950s [34]. The seminal discovery of leptin,

    the adipocyte-derived protein hormone providing

    this signal, by Friedman and collaborators in 1994

    [54] was a decisive catalyst for much of the current

    investigation on peripheral modulation of centralnetworks. A little more than a decade later, leptin

    has been shown to modulate several aspects of

    energy balance through several different mecha-

    nisms and across a wide spectrum of timeframes,

    alerting the brain to the state of body adiposity [55]

    and acting as a fat-o-stat. It is now well estab-

    lished that the pronounced obesity in genetically

    leptin-deficient ob/ob mice is due to the loss of a

    centrally active feeding-inhibitory messenger, asrestitution of the leptin signal in these animals

    normalizes food intake and body weight [56]

    (Fig. 2). Serum leptin correlates well to the size of

    the body fat deposit, and falls with weight loss [57].

    This relationship is seen also in obesity, where the

    combination of hyperleptinaemia and hyperphagia

    has led to the suggestion that overweight is

    characterized by leptin resistance [58]. Central

    actions underlie both leptin-mediated feeding sup-

    pression as well as the extensive peripheral meta-

    bolic effects of this hormone; thus, e.g. replacement

    of leptin receptors selectively in the brain of ob/obmice is sufficient to prevent hepatic steatosis [59].

    Insulin

    Insulin is well recognized as the key glucostatic

    regulator.Recent data demonstrate that in addition to

    the control of peripheral glucose uptake, this role also

    encompasses powerful central effects, in synergism

    with leptin. First, intracerebroventricular (i.c.v.)

    administration of insulin decreases food intake [60]

    via insulin receptors expressed on Arc neurones [61].

    The role of insulin in feeding is complicatedby thefactthat the hypoglycaemia resulting from elevations in

    serum insulin in itself stimulates food intake. How-

    ever, when blood glucose changes are compensated

    for, hypophagia is seen also with increases in periph-

    eral insulin [62, 63], suggesting that the central

    effects of insulin are anabolic. (This secondary hypo-

    glycaemia may also explain why the insulin secretion

    triggered already at the sight of a palatable-looking

    Fig. 2 Genetically leptin-deficient ob/ob mice treated subcutane-

    ously (s.c.) with saline (left) or with leptin (right). The severe

    obesity in these animals is abolished with leptin replacement

    therapy. Figure generously provided by Dr Jeffrey M. Friedman.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine258: 301327

    3 04 C . B RO BE RG ER

  • 8/12/2019 Food Intake Review 2005

    5/27

    meal stimulates appetite (the cephalic phase [64]),

    an indication that direct sensory input relayed via the

    cortex can set off powerful appetitive mechanisms.)

    Secondly, and again similar to leptin, insulin also

    modulates peripheral energy metabolism via central

    effects by inhibiting liver gluconeogenesis [65, 66].Thus, whilst the brain does not depend on insulin for

    glucose uptake, it is very much interested in what

    insulin has to say about the metabolic state of the

    body.

    Peptide YY (3-36)

    In addition to CCK, several gut-derived peptides

    provide alimentary feedback to the central metabolic

    circuits [67]. Peptide tyrosine-tyrosine (3-36)

    [PYY(3-36)], a member of the NPY peptide family

    produced by enteroendocrine cells [68], has recentlybeen shown to act as an important feedback signal

    from the gut to the hypothalamus. Following a

    meal, PYY(3-36) is released into the circulation

    [69], specifically stimulated by the presence of lipids

    and carbohydrates in the lumen of the distal ileum

    and colon [70, 71]. Peripheral administration of this

    hormone inhibits food intake and causes weight loss

    [72, 73]. While some laboratories initially were

    unable to replicate this effect [74], this may partly be

    due to discrepancies in technique [75] and the

    results have since been independently confirmed

    [76, 77]. The satiety effect of PYY(3-36) is compar-atively modest but, importantly, is observed also in

    humans, including obese patients [73, 78]. Plasma

    levels of PYY(3-36) rise markedly following ileal

    resection [79, 80], an observation that has been

    linked to the weight loss observed in patients

    undergoing this procedure (S.R. Bloom and C. Le

    Roux, personal communication).

    Ghrelin

    Thus, the gastrointestinal-brain axis has long been

    viewed as a key channel subserving meal termin-ation with CCK and PYY(3-36) as prime mediator

    examples. Novel findings on the hormone ghrelin

    (produced in stomach and small intestine epithelia

    [81], see [82]) are challenging this doctrine. Ghrelin

    is unique as the first gut hormone shown to

    stimulate food intake [67]. Both peripheral and

    central injections of ghrelin result in increased

    feeding as well as fat mass [8386]. Ghrelin levels

    peak sharply in anticipation of a meal in humans as

    well as experimental animals [87], resulting in

    stimulation of both feeding and gastric emptying

    [88] through actions possibly involving the vagus

    nerve [89], and may thus provide a meal initiation

    signal. In the hypothalamus, peripherally adminis-tered ghrelin mainly activates the NPY neurones

    [85, 90] and antagonizing the actions of these cells

    inhibits the orexigenic effect of ghrelin administra-

    tion [85, 91]. In contrast, the melanocortin pathway

    does not appear to be primarily involved [90]. Recent

    reports have proposed that ghrelin is synthesized also

    in hypothalamic neurones, but this issue remains

    controversial, in part due to the contradictory claims

    of the site of central ghrelinergic neurones and the

    failure to demonstrate ghrelin mRNA in the brain (cf.

    [92] and [93]).

    Importantly, a loss of the hunger message relayedby ghrelin has been suggested as the mechanism

    behind the weight-reducing effects of bariatric sur-

    gery [94]. The initial rationale for gastric bypass

    [95] was that the procedure would produce weight

    loss by means of malabsorption. However, this turns

    out to be a transient effect due to the considerable

    compensatory potential of the digestive system.

    Nevertheless, weight loss persists, caused instead

    by a loss of appetite and hypophagia [96]. Concom-

    itant with this effect, a fall in plasma ghrelin is

    observed following the bypass procedure, in contrast

    to the ghrelin increase associated with nonsurgicalweight reduction, where weight relapse is common

    [86, 97]. (Note, however, findings that argue

    against such a relationship, see [67].) Furthermore,

    clinical data tie the hyperphagia observed in Prader

    Willi syndrome to strikingly high plasma ghrelin

    levels [98]. These results, coupled with the discovery

    that elevated plasma ghrelin is a marker for future

    weight gain (D.E. Cummings and J. Krakoff, personal

    communication) indicate that interfering with ghre-

    lin signalling offers a clinically promising approach

    to treating eating disorders.

    Oleoylethanolamide

    A role for endogenous cannabinoids in appetite

    regulation has long been suspected from the carbo-

    hydrate craving observed in marijuana smoking

    [99]; indeed, increased appetite is a diagnostic

    criterion for cannabis intoxication [100]. Neuronal

    production of cannabinoids is widespread and these

    2005 Blackwell Publishing Ltd Journal of Internal Medicine 258: 301327

    R E V I EW : B R A I N C O N T RO L O F F O O D I N T A KE 3 0 5

  • 8/12/2019 Food Intake Review 2005

    6/27

    mediators play an important and general role in the

    modulation of synaptic transmission [101], with the

    orexigenic effects likely mediated via central cann-

    abinoid CB1 receptors [102, 103]. However, the

    lipid family to which the cannabinoids belong also

    includes other members with opposite and periph-eral effects on energy metabolism. Piomelli and

    colleagues have accumulated evidence that the fatty

    acid oleoylethanolamide (OEA), chemically but not

    pharmacologically similar to the cannabinoids, is

    produced in the duodenum and acts via the vagus

    nerve to decrease body weight through activation of

    the nTS [104]. OEA increases the inter-meal latency,

    an effect distinct from that of, e.g. CCK, which

    primarily decreases meal size [105]. However, chan-

    ges in energy expenditure also underlie the OEA-

    mediated weight reduction and are especially pro-

    nounced in models of obesity, involving in particularincreased fat utilization, whereas glucose home-

    ostasis is relatively unaffected [106]. The catabolic

    effects are most noticeable as a slowing of body

    weight gain in growing rats, with OEA synthesis

    reduced by food deprivation and stimulated in

    response to increased demands on energy availability

    such as cold exposure [104]. The metabolic actions

    of OEA depend selectively and critically on genomic

    as well as nongenomic actions of the ubiquitous

    nuclear peroxisome proliferator-activated receptor-

    alpha (PPAR-a) [107]. These results add obesity to

    the growing list of potential therapeutic applicationsfor nuclear receptor pharmaceuticals. Notably, drugs

    that target PPAR-a, e.g. gemfibrozil, are already in

    clinical use to treat hypercholesterolaemia [108].

    Integration of peripheral signals in the Arc

    The peripheral signals described above thus act upon

    the Arc (and nTS, see below) to influence the central

    pathways regulating energy balance. In the Arc,

    receptors for leptin and insulin found on NPY and

    POMC neurones serve to inhibit transcription of NPY

    [109, 55] and increase POMC mRNA levels [110112] via differential second messenger systems [113].

    It is becoming evident that insulin, leptin and other

    metabolically relevant hormones eventually con-

    verge not only on a common set of neurones, but

    indeed also on the same molecules. Recent reports

    highlight the role of the ATP-dependent potassium

    current, IK(ATP), as a molecular target mediating

    rapid, electrophysiological effects, of peripheral

    hormones. This K+ conductance is a priori sensitive

    to the availability of metabolic fuel as a fall in

    intracellular levels of the energy donor ATP causes

    the channel to open, leading to K+ influx and

    hyperpolarization; this mechanism enables neurones

    expressing IK(ATP) to vary their excitability in responseto changes in glucose concentration[114].Leptin and

    insulin both hyperpolarize Arc neurones by enhan-

    cing IK(ATP) [115, 116], by activating a common

    enzyme, phosphoinositide 3 (PI3) kinase [116, 117].

    It should be emphasized that the transmitter pheno-

    type of Arc neurones expressing IK(ATP) is a contro-

    versial issue which remains to be conclusively

    resolved [118120]. Additional signals likely weigh

    in on IK(ATP); this current is augmented when the

    concentration of fatty acid derivatives is increased

    locally within the Arc by inhibition of lipid oxidation,

    a message of energy surplus that also decreases foodintake [66, 121]. This convergence of nutrient

    information makes the PI3-kinase/IK(ATP) a key

    integration node within the metabolic signalling

    chain, attractive as a therapeutic target.

    Modulation of the membrane potential of Arc

    neurones has recently been demonstrated to control

    glucose homeostasis. Opening of Arc K(ATP) chan-

    nels via either hyperinsulinaemia or central inhibi-

    tion of lipid oxidation inhibits vagal efferent (i.e.

    parasympathetic) gluconeogenic signals to the liver,

    promoting the use of fat as metabolic fuel [66, 122].

    The Arc is also the site of central leptin regulation ofglucose homeostasis as selective restoration of Arc

    leptin receptor expression in otherwise leptin recep-

    tor-deficient mice is sufficient to correct their

    hyperglycaemia [123]. These results show that

    insulin modulates glucose homeostasis by independ-

    ent peripheral and central mechanisms and empha-

    size that interconnectivity within brain metabolic

    regions serve to switch the body between different

    fuel sources, in parallel to controlling food intake.

    Interestingly, in obese rats, hypothalamic IK(ATP)channels fail to respond to leptin and insulin [115,

    116]. Whether similar defects underlie insulin and/or leptin resistance in human diabetes and obesity is

    an interesting possibility, which remains to be

    investigated.

    Output from the Arc

    NPY neurones. Neuropeptide Y is one of the most

    potent stimulators of feeding known [124], an

    2005 Blackwell Publishing Ltd Journal of Internal Medicine258: 301327

    3 06 C . B RO BE RG ER

  • 8/12/2019 Food Intake Review 2005

    7/27

    effect that has been confirmed by various approa-

    ches [40]. While there is conflicting data on whe-

    ther deletion of the NPY gene produces hypophagia

    (cf. [125] and [126]), the obesity of ob/ob mice is

    attenuated when combined with an NPY)/) geno-

    type [127], suggesting that NPY is an importantmediator of central leptin signalling. Stimulation of

    feeding appears to be transduced predominantly via

    postsynaptic NPY Y1 receptors, as determined from

    pharmacological and genetic engineering studies

    (reviewed in [128], Fig. 3a). However, the syner-

    gistic actions of multiple NPY receptor subtypes

    participate to produce orexigenic effects in vivo

    [129]. Detailed behavioural analysis of those effects

    suggests that NPY primarily stimulates appetitive

    rather than consummatory behaviour [130].

    POMC neurones. Pro-opiomelanocortin is a large

    precursor protein which gives rise to several bioac-

    tive peptides. Among these, the melanocortin pep-tides, in particular a- and c-melanocyte-stimulating

    hormone, have been shown to exert potent ano-

    rexigenic effects when administered i.c.v. [131,

    132]. Central melanocortin effects are mediated by

    the melanocortin 3 and 4 receptors (MC3R and

    MC4R, respectively; Fig. 3b). Deletion of the genes

    for either POMC, MC3R or MC4R result in obesity in

    mice, suggesting that the melanocortin system is

    crucial in maintaining body weight [133135] as

    supported by similar findings in humans (see below).

    MC4R)/) mice also increase their feeding in

    response to a high fat diet, in contrast to wild-typelittermates where a reduction is seen andob/obmice,

    which maintain the same intake as with regular

    chow [136], underlining the importance of the

    melanocortin system for adjusting food intake in

    response to caloric variations. In addition, the hall-

    mark hypophagia seen in disease models as diverse

    as renal failure, immunological challenge with

    lipopolysaccharide (LPS) and tumour implants is

    ablated. The obesity in MC4R-deficient animals is

    partly due to changes in energy expenditure, such as

    deficient diet-induced thermogenesis [136]. The

    anatomical substrate for this effect may be a directprojection from the POMC neurones in the Arc to

    the preganglionic sympathetic neurones in the spi-

    nal cord [137139] constituting a link between the

    metabolic integrator and the autonomic effector

    system. Interestingly, the spinal projection sets the

    POMC neurones apart from the neighbouring NPY

    neurones which otherwise exhibit very parallel

    innervation patterns. However, it should be pointed

    out, that in humans, the melanocortin system

    appears to be more geared towards regulating feed-

    ing behaviour, with a proportionately smaller role in

    peripheral metabolism [140].

    NPYPOMC interactions. Interactions between the

    Arc populations allow the NPY neurones to control

    the activity of the POMC cells via two mechanisms.

    First, NPY neurones coexpress agouti gene-related

    peptide (AGRP), an endogenous melanocortin ant-

    agonist [141143]. Thus, at the axon terminal,

    melanocortin action can be blocked by simultaneous

    Fig. 3 Expression of NPY and melanocortin receptors in the

    mouse brain. In situ hybridization histochemistry (a) shows the

    distribution of NPY Y1 receptor mRNA detected as silver grains in

    a coronal section, revealing dense expression in the cerebral

    cortex and nuclei in the amygdala, thalamus and hypothalamus.

    In panel b, green fluorescent protein (GFP) is expressed in a

    neurone under control of the melanocortin (MC) 4 receptor pro-

    moter; note strong immunoreactivity throughout cell soma and

    dendrites. A Nissl-stained coronal section (c) shows neurones

    clustered to form the paraventricular hypothalamic nucleus

    (PVH) alongside the third ventricle. The PVH constitutes a central

    integrative hub within the metabolic circuitry. (d) Immunoh-

    istochemical for GFP (indicating the presence of the MC4 receptor)

    and in situ hybridization for NPY Y1 receptor mRNA have beencombined in a section from the amygdala, revealing the coexist-

    ence of these receptors in neurones downstream of the Arc. Figure

    produced by Drs Toshiro Kishi and Joel K. Elmquist. Reprinted

    with permission from Macmillan Publishers Ltd.; Molecular Psy-

    chiatry 2005;10:132146.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine 258: 301327

    R E V I EW : B R A I N C O N T RO L O F F O O D I N T A KE 3 0 7

  • 8/12/2019 Food Intake Review 2005

    8/27

    release of AGRP, and in agreement with such an

    arrangement, a single i.c.v. administration of AGRP

    causes an impressively long-lasting (one week)

    suppression of food intake [144]. Secondly, at the

    cell body level, POMC neurones are innervated by

    NPY-ergic terminals [145] and express the Y1receptor [146], through which NPY causes a

    powerful membrane potential hyperpolarization (i.e.

    inhibition) [147]. Surprisingly, no reciprocal inner-

    vation has yet been described and Roseberry et al.

    [147] did not detect any changes in the electrical

    properties of NPY neurones using a melanocortin

    analogue. Thus, there may exist an asymmetrical

    interaction in the Arc favouring the orexigenic

    NPY/AGRP message over anorexigenic melanocor-

    tin signalling. However, an inhibitory influence over

    the NPY neurones may be provided by PYY (3-36),

    which is a selective agonist of the inhibitory Y2autoreceptors [148] expressed by these cells [146].

    Such gastrointestinal negative feedback has been

    proposed as the mechanism whereby PYY(3-36)

    inhibits feeding as no such effect is observed in mice

    genetically deficient for the Y2 receptor and appli-

    cation of the peptide inhibits the electrical activity of

    Arc NPY terminals [73]. This effect is relatively

    selective as disruption of other relevant metabolic

    pathways does not affect the satiety effect; the

    PYY(3-36) effect persists both after vagotomy and in

    MC4)/) mice [76], suggesting that neither the nTS

    nor the Arc POMC neurones are directly involved.

    Classical transmitters: glutamate and GABA. While

    much of the current research on the central regula-

    tion of energy balance focuses on the role of peptides,

    it should be emphasized that in the hypothalamus, as

    in the rest of the brain, the key chemical mode of

    communication between neurones is via amino acid

    transmitters, i.e. excitatory glutamate and inhibitory

    c-amino butyric acid (GABA). Indeed, in the absence

    of glutamate and GABA-mediated transmission, little

    remains of hypothalamic synaptic activity [149,

    150]. The major function of peptides, in addition totheir genomic effects, is likely to modulate the syn-

    aptic transmission of classical transmitters [151]

    with which they coexist [152]. Interestingly, glu-

    tamate N-methyl-d-aspartate (NMDA) receptors

    have been found to stimulate feeding with remark-

    able anatomical specificity within the lateral hy-

    pothalamic area (LHA), in comparison with other

    hypothalamic regions tested and the amygdala

    [153]. Infusion of NMDA antagonists locally within

    the LHA blocks both agonist-induced and depriva-

    tion-induced food intake, indicating the involvement

    of endogenous glutamatergic tone in natural feeding

    [154]. Histochemical studies suggest that within the

    Arc, NPY neurones largely contain GABA, whereasPOMC neurones signal via glutamate [155, 156].

    Downstream targets of the Arc. The downstream

    cellular effects of NPY are still mysterious. It was

    initially assumed that feeding-promoting neurones

    in loci sensitive to NPY orexigenesis were excited by

    NPY. However, all known members of the NPY

    receptor family couple to inhibitory second mes-

    senger systems [128]. Electrical excitation has been

    proposed to come about in the form of disinhibition

    via NPY-mediated suppression of GABA-dependent

    inhibitory postsynaptic currents [157, 158], withmelanocortin stimulation producing the opposite

    result, i.e. inhibition via stimulation of GABA release

    [157]. However, that does not explain the role of

    postsynaptic Y1 receptors, which exist throughout

    the hypothalamus [146, 159, 160] (Fig. 3a). The

    most potent orexigenic effects of NPY are seen

    within the perifornical region/LHA [124], where

    NPY/AGRP-ergic terminals appear to target two

    separate populations of neurones expressing the

    neuropeptides hypocretin (Hcrt; also known as

    orexin) and melanin-concentrating hormone (MCH;

    Fig. 4, [142, 161, 162]). This pathway is of interestas Hcrt and MCH potently modulate wakefulness

    [163167], providing a means for metabolic signals

    to control arousal state. Surprisingly, in a recent

    investigation of the electrophysiology of the LHA

    neurones, melanocortin stimulation did not affect

    the electrical properties of MCH-expressing cells,

    whereas both these and Hcrt-expressing cells were

    inhibited by NPY [168, 169]. Furthermore, micro-

    injection of NPY into the LHA appears to activate a

    group of neurones distinct from those expressing

    Hcrt or MCH [170]. As with all neural interactions,

    it is important to bear in mind that the activity ofneurones can be influenced via several independent

    mechanisms, including (but not exclusively) elec-

    trical and transcriptional effects, and that different

    changes proceed along different temporal scales.

    Thus, a functional Arc-LHA pathway cannot be

    excluded. Nevertheless, these data invite a re-eval-

    uation of the role of Hcrt and MCH as downstream

    mediators of the NPY and POMC neurones.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine258: 301327

    3 08 C . B RO BE RG ER

  • 8/12/2019 Food Intake Review 2005

    9/27

    Circadian regulation of metabolic processes. In addition

    to the various controls summarized above, metabolicprocesses also follow strict circadian variations as

    recently underscored by the demonstration that

    inactivation of key genes maintaining circadian

    rhythmicity results in manifest metabolic syndrome

    in mice [171]. Thus, for example, in rats the active

    period of the day is immediately preceded by

    coordinated peaks in hepatic glucose output (via the

    sympathetic nervous system) and glucose uptake in

    striated muscle (a parasympathetic effect; see [172]).

    Buijs et al. [173] have investigated which brain re-gions are responsible for this synchronization. Using

    anatomical tracing they find that the chains of

    neurones innervating liver and muscle are separated

    all the way through brainstem and hypothalamus to

    distinct populations of preautonomic master neu-

    rones in the suprachiasmatic nucleus [173], the

    brain region maintaining circadian rhythmicity and

    entrained by direct retinal input [174, 175]. The

    CerebralCortex

    AcbSh

    PFCx

    MCH Hcrt

    LHA

    Othersubcortical nuclei,

    incl. BST,MPO, PVT,

    DMH,Amgdl,Raphe, PAG

    and PBN

    NPY POMC

    Arc

    DMX

    IML

    FOODINTAKE

    ENERGYEXPENDITURE

    INGESTIVE(Motivated)BEHAVIOUR

    Sympathetic

    ParasympatheticANS

    Endocrine

    regulationincl. thyroid andadrenocortical axes

    PVH

    Pituitary

    Fig. 4 Integration in higher brain regions determines the central response to changes in peripheral metabolic state. Schematic illustration

    of connections between brain regions responsible for coordinating the behavioural somatomotor (i.e. food intake), autonomic and endocrine

    (the latter two regulating energy expenditure) responses that together constitute the motivated ingestive behaviour used by the nervous

    system to meet nutritional challenges. The antagonistic orexigenic NPY and anorexigenic POMC neurones in the Arc project in parallel

    paths to numerous subcortical nuclei [including the bed nucleus of the stria terminals (BST), the medial preoptic area (MPO), the

    paraventricular nucleus of the thalamus (PVT), several hypothalamic nuclei, e.g. the dorsomedial nucleus (DMH), the amygdala (Amgdl),

    the serotonin-containing system in the raphe nuclei, the periacqueductal grey area (PAG) and the parabrachial nucleus (PBN)] distributedthroughout the brain. A projection to neurones expressing melanin-concentrating hormone (MCH) or hypocretin (Hcrt) in the lateral

    hypothalamic area (LHA) provides an indirect pathway to the cerebral cortex for metabolic signals relayed via the Arc. The cortex in turn

    projects back heavily to both the LHA and other feeding-regulatory regions. In addition, the LHA also receives an inhibitory input from the

    shell of the nucleus accumbens (AcbSh), which in turn is modulated via prominent excitatory inputs from the prefrontal cortex (PFCx).

    Thus, the LHA is positioned to integrate both homeostatic and reward-related signals in the gating of food intake. Energy expenditure is

    modulated via outputs from the Arc to neuroendocrine neurones in the paraventricular hypothalamic nucleus (PVH), which control

    release of, e.g. thyrotropin-releasing hormone and adrenocorticotropic hormone from the pituitary gland. Energy expenditure is also

    regulated by projections from POMC neurones in the Arc and descending pathways from the PVH to autonomic preganglionic neurones in,

    e.g. the dorsal motor nucleus of the vagus (DMX; parasympathetic) and spinal cord intermediolateral cell column (IML; sympathetic).

    Note that ascending projections from the brainstem, which provide parallel important metabolic inputs to the brain, have not been included

    in the figure. See text for details.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine 258: 301327

    R E V I EW : B R A I N C O N T RO L O F F O O D I N T A KE 3 0 9

  • 8/12/2019 Food Intake Review 2005

    10/27

    distinct pathways originating in the suprachias-

    matic nucleus are particularly interesting in con-

    junction with the discovery that the autonomic

    inputs to intra-abdominal and subcutaneous fat

    stores are also separate [176]. In humans, shift work

    [177] and sleep deprivation [178] are associatedwith increased adiposity, findings that have been

    linked to the sleep-associated peak in leptin secretion

    [179]. However, this anatomically separate inner-

    vation indicates that loss of periodicity in the circa-

    dian input to adipose tissue may disrupt the balance

    between different fat compartments leading, in turn,

    to manifestations of the metabolic syndrome, which

    is correlated to abdominal but not subcutaneous fat

    accumulation [180].

    Contributions of hind- and forebrain to

    feeding regulation

    As mentioned above, the brainstem provides a port

    for vagal and other neural sensory signals into the

    brain. Classical accounts of brain regulation of

    feeding described two systems balancing each other:

    the hypothalamus, monitoring the periphery for

    signals alerting central circuits to diminishing

    energy stores, and the brainstem, receiving oral

    and gastrointestinal information as an online signal

    of the amounts and qualities of the food that was

    being ingested. This arrangement would allow the

    hypothalamus to function as a long-term controlorchestrating meal initiation and the brainstem

    served as a short-term control for meal termination.

    Much of our knowledge on the different contribu-

    tions of the fore- and hindbrain in meal regulation

    comes from a lesion model developed by Grill and

    Norgren [33]. Disconnecting the forebrain (which

    includes the hypothalamus) produces a rat inca-

    pable of the motor activation necessary for normal

    feeding. However, if this animal whose brainstem

    remains intact is provided sucrose solution via an

    intraoral cannula, intake can be measured as the

    solution consumed until the meal is terminated asthe animal lets solution drip out of the mouth. These

    decerebrated rats maintain the ability to terminate

    their meal in response to changes in gastrointestinal

    feedback, but are unable to compensate for varia-

    tions in the caloric value of the fed solution,

    resulting in anorexia if the sucrose concentration

    is reduced. Similarly, removal of the post-oral

    feedback (by e.g. vagus nerve transection or gastric

    drainage) leads to increases in meal size as well as

    duration [181, 182], although there is a compen-

    satory delay in the latency to meal initiation,

    possibly mediated by the hypothalamus. These

    results underscore the role of the Arc as a metabolic

    sensor. It should be pointed out that an intact Arc isnot necessary for meal initiation humans and

    animals with selective lesion of this region not only

    eat, they eat copiously [29, 183185].

    It is now becoming evident that the brainstem can

    integrate much the same signals as have been

    shown to modulate hypothalamic activity. Leptin

    receptors are expressed at several strategically

    located brainstem sites, and selective stimulation of

    these receptors suppresses food intake at doses

    comparable with those used in forebrain injections

    [186, 187]. Here, leptin activates the same medial

    region of the nTS that is stimulated by gastricdistension [188], suggesting an anatomical site of

    integration of long- and short-term feeding controls.

    Likewise, melanocortin agonists can reduce feeding

    and body weight by brainstem mechanisms [189].

    Interestingly, the neurones in the nTS mediating the

    viscerosensory signal may also be POMC-encoded

    [43, 190], a finding that puts our understanding of

    melanocortin-mediated meal suppression in a new

    light. Orexigenic effects of ghrelin are also seen with

    selective local administration both in the hypotha-

    lamus [84] and in the brainstem [191] (Fig. 5a,b).

    Finally, glucosensitive neurones have been recordedin the nTS [192]. Thus, the nTS is in no way a

    Fig. 5 Ghrelin increases food intake following brainstem admin-

    istration. Unilateral injection of 10 pmol (but not 5) of ghrelin

    (black bars) into the dorsal vagal complex, including the nucleus

    tractus solitarii, results in a significant increase of food intake both

    1.5 and 3 h after drug administration compared with vehicle

    (white bars), in an experiment by Faulconbridge et al. [191].

    Reprinted with permission from the American Diabetes Associ-

    ation;Diabetes 2003;52:22602265.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine258: 301327

    3 10 C . B RO BE RG ER

  • 8/12/2019 Food Intake Review 2005

    11/27

    passive transducer of viscerosensory signals, but

    serves to integrate numerous indices of the animals

    metabolic state. This conclusion is supported by the

    observation that the hyperphagia of rats that lack

    leptin receptors is caused by larger meal size (i.e.

    meal termination delay) rather than an increasednumber of feeding bouts [193], suggesting an action

    localized in the hindbrain. However, the response to

    CCK (i.e. brainstem satiety signalling) as well as

    normal meal size is restored in these animals

    following selective re-establishment of leptin recep-

    tor expression in the Arc. Weighed together, these

    data emphasize that actions of metabolically rele-

    vant hormone take place at a few, but distributed,

    sites in the brain contributing to a coordinated

    feeding response.

    Beyond the primary sensors: CNSintegration of hunger and satiety signals

    As in all matters involving the brain, behaviour begs

    the question: What are the pathways? For the

    metabolic signals to produce behaviour they need to

    proceed further into the brain beyond the primary

    sensors in the Arc and nTS and ultimately engage

    regions that initiate and organize behavioural,

    autonomic and endocrine response patterns. Histo-

    chemical studies have revealed that (i) the Arc

    projections diverge widely throughout the brain

    [194197] including, via indirect pathways, amassive cortical innervation [198] (Fig. 4), (ii) the

    NPY and POMC populations project in remarkably

    parallel paths [196] and may converge on the same

    cells as supported by the widespread coexistence of

    Y1 and MC4 receptors [199] (Fig. 3d), and (iii) the

    nTS innervates largely the same nuclei as the

    ascending projections from the Arc [196, 200

    202]. This circuitry indicates that integration

    between the primary metabolic sensors in the Arc

    and nTS is a distributed phenomenon, and it has

    been suggested that this arrangement allows for

    motivational state to be weighed into the networkbefore reconvergence and the final decision for a

    proper metabolic response is made [40]. In addition

    to this scheme, extensive reciprocal projections

    connect the Arc with its target regions [203]. The

    functional implications of this arrangement are not

    clear at present. There may exist a sequential

    arrangement hidden within the network that has

    so far eluded the techniques used to investigate the

    system. Another alternative is that the reciprocity

    may produce a reverberating signal, such as the

    large-scale oscillations described in, e.g. thalamo-

    cortical systems [204]. Possibly, such persistent

    activity may be important in the triggering and

    maintenance of an anabolic response. The hierar-chical organization of the metabolic circuits and its

    relationship to behaviour presents a major future

    scientific challenge. (For further discussion of the

    systems organization of energy balance regulation,

    the reader is referred to recent exhaustive reviews

    [37, 38].)

    Coordinating the metabolic output

    It also remains to explain how the divergence

    convergence organization of the Arc/nTS projec-

    tions interdigitates with the efferent networksunderlying the final metabolic response. As

    already mentioned, motivated behaviours (classic-

    ally divided into ingestive, reproductive and defen-

    sive) involve three distinct outlets: components of

    the autonomic and neuroendocrine systems as

    well as coordinating the overall behaviour of the

    animal [44]. Activity within these three effector

    systems is hypothesized to be organized by a

    collection of cell groups within the medial hypo-

    thalamus area, collectively termed the hypotha-

    lamic visceromotor pattern generator network

    [205], which subsequently recruits elements with-in a behavioural control column, spanning the

    mes- and diencephalic midline [206]. Separate

    groups of control column nuclei produce ingestive,

    reproductive and defensive behaviours, but con-

    nections between these networks allow them to

    interact for purposes of mutual exclusion so that

    only one behaviour is expressed at once [207].

    The paraventricular nucleus (PVH; Fig. 3c) is a

    crucial control column module for ingestive beha-

    viour. The PVH collects metabolic information

    from oropharyngeo- and viscerosensory receptors

    and humoral signals (both directly and via theArc), and is regulated by biological rhythms via

    the suprachiasmatic nucleus and by the overall

    state of the animal as reported from the cerebral

    hemispheres via relays in the septum [206].

    Output from the PVH, in turn, employs all

    three outlets described above: endocrine via neuro-

    endocrine neurones controlling pituitary hormone

    release, autonomic via direct projections to

    2005 Blackwell Publishing Ltd Journal of Internal Medicine 258: 301327

    R E V I EW : B R A I N C O N T RO L O F F O O D I N T A KE 3 1 1

  • 8/12/2019 Food Intake Review 2005

    12/27

    preganglionic neurones in the spinal cord inter-

    mediolateral cell column (sympathetic) and the

    dorsal motor nucleus of the vagus (parasympathe-

    tic), and behavioural (i.e. somatomotor) via several

    pathways innervating the brainstem [137, 208

    210] (Figs 1 and 4). The hierarchical arrangementupstream of these motor nuclei bears some simi-

    larity to the basal ganglia organization for the

    control of conscious movement [206]. The details

    of the underlying anatomy remain mysterious, but

    it is clear that, rather than a simple sequential

    organization where information flows neatly from

    one collection of neurones to another, we are

    faced with an interconnected series of hubs that

    collect, integrate and disseminate information.

    Food intake in lower organisms: models for

    the organization of behaviour

    Intriguingly, an improved understanding of the

    organization of feeding behaviour is now coming

    from studies of lower organisms. Animals such as

    the nematode Caenorhabditis elegans and the sea slug

    Aplysia present several technical advantages: beha-

    viour is easily divided into discrete sequences, the

    nervous system consists of a limited number of

    neurones whose electrical properties and intercon-

    nectivity has been characterized in detail, and, in the

    case ofC. elegans, the genome is highly accessible for

    molecular manipulation. This knowledge makes itpossible to understand how nature organizes phys-

    ical networks to efficiently initiate, organize and

    terminate behaviour.

    Caenorhabditis elegans

    Feeding in C. elegans is polymorphic; depending on

    genetic background animals will feed either alone or

    in aggregates [211213]. Naturally occurring var-

    iations in a single amino acid position of the

    neuropeptide receptor NPR-1 (for neuropeptide

    receptor resemblance-1) translate into either asolitary or a social feeding phenotype [212]. Acti-

    vation of NPR-1, by shifting network properties,

    leads to activation of social feeding behaviour, and

    the amino acid substitution in NPR-1 determines the

    response to stimulation with the neuropeptide

    ligands flp 18 or )21 [214]. Conversely, null

    mutations in the npr-1 locus alter the balance in

    favour of solitary feeding [214]. Thus, a single gene

    is sufficient to redirect behaviour. Intriguingly, the

    predicted transmembrane domains of NPR-1 display

    considerable homology to mammalian NPY recep-

    tors [212], suggesting that similar molecular com-

    ponents underlie related behaviours throughout

    evolution. Recent data demonstrate that within thisnetwork an important upstream regulator of NPR-1-

    mediated feeding is oxygen concentration, showing

    how external cues reset behaviour [215, 216].

    Aplysia

    In Aplysia, a meticulously dissected network has

    been highly informative in characterizing the neural

    mechanisms underlying various behavioural com-

    ponents. Aplysia feeding can be initiated by sensory

    stimulation of the lip and is consolidated by arousal

    caused by the exposure to food. In parallel with thefeeding central pattern generator (CPG) circuit, a

    network controlling arousal is triggered, which then

    feeds back into the CPG [217]. Termination of food

    intake is achieved by switching ingestion to the

    opposite behaviour of egestion [218]. Separate

    motor neurones within the CPG effectuate ingestion

    and egestion. Switching the balance between these

    neurones is elegantly accomplished by recruiting a

    single additional interneurone into the circuitry via

    an electrical synapse [219]. Feeding patterns in this

    social mollusc also differ substantially in the absence

    or presence of other animals; company is animportant incentive for feeding. Pheromones secre-

    ted from other Aplysia stimulate a neurone directly

    driving the appetitive phase of feeding, which also

    excites a control neurone for the consummatory

    phase so that these behaviours are properly organ-

    ized, leading to larger and more frequent eating

    bouts [220]. These studies provide information on

    how organization within a neural system translates

    into behaviour, with potentially broad implications

    considering the surprisingly large overlaps between

    human and mollusc feeding behaviour. Importantly,

    they may shed light on the process of selectionwithin the behavioural repertoire and why we eat in

    apparent violation of homeostatic mechanisms

    when our fat stores are filled to the brim.

    Food reward: role of the nucleus accumbens

    However, to elucidate the neural organization of

    feeding in complex vertebrates, it is necessary to

    2005 Blackwell Publishing Ltd Journal of Internal Medicine258: 301327

    3 12 C . B RO BE RG ER

  • 8/12/2019 Food Intake Review 2005

    13/27

    consider that our choice of food is not simply a

    function of energy supply and demand, but also very

    much linked to reward value. Homeostatic systems

    active within the brain operate at the mercy of

    motivational states. The motivational state is a

    product of, inter alia, limbic influences relayed inpart via the amygdala, and reward factors. Obvi-

    ously, adding reward experience to food is a means

    of, e.g. avoiding consumption of foods whose taste

    indicate the presence of invasive microorganisms,

    but also promoting those whose taste signals

    particular nutritional value. As a result, pairing

    feeding with pleasure may override normal satiety

    mechanisms, resulting in hyperphagia and obesity.

    The concept of reward is intimately linked to that of

    addiction, and it has been suggested that obesity is a

    consequence of an addiction to food [221]. Drugs of

    abuse converge upon the mesolimbocortical systemto produce reward, specifically by enhancing dop-

    amine release in the nucleus accumbens (Acb) of the

    forebrain as a final common pathway [222]. There

    is little doubt that changes in dopaminergic trans-

    mission affects food intake. Indeed, animals unable

    to produce brain dopamine die of starvation unless

    fed by gavage [223], and a common side-effect of

    neuroleptics affecting dopamine signalling is obesity

    [224]. However, novel data suggest that dopamine

    primarily acts to reinforce behaviours at the initial

    encounter with a novel reward, but the release of

    dopamine decreases once the behaviour has beenestablished within the behavioural repertoire [225].

    Thus, whilst dopamine modulates learning and

    locomotion associated with motivational behaviour,

    it appears not to modulate feeding behaviour per se;

    blocking Acb dopamine signalling does not alter

    total food consumption in starved rats, although it

    suppresses ambulation associated with feeding

    [226].

    However, transmitter systems other than the

    dopaminergic system connect Acb to components

    of the metabolic circuitry [227]. Notably, chronic

    access to a preferred flavour (chocolate-fat solution)produces the same transmitter changes in the Acb

    as chronic morphine or ethanol, suggesting a

    common reward mechanism for palatable food

    and conventional drugs of abuse [228]. These

    changes include increased transcription of opioid

    peptides such as enkephalin. In turn, stimulating l-

    opioid receptors in the Acb increases intake of fat-

    enriched foods with high palatability value [229],

    which may be interpreted as positive reinforcement.

    This effect is not seen following inhibition of neural

    transmission in the basolateral amygdala (BLA) or

    the LHA [230]. The connection between the BLA

    and forebrain cortical regions has been implicated

    in gauging food palatability, and an intact BLA isrequired for determining the reward value embed-

    ded in sensory input [231]. In contrast, the central

    amygdala (CeA) serves more like a general gate-

    keeper of feeding as inactivation of this subnucleus

    blocks consumption of all foods [230]. This dichot-

    omy may be explained by the different projections

    of the BLA (innervating higher forebrain regions

    such as the prefrontal cortex) and the CeA (aimed

    towards the postulated hypothalamic and brain-

    stem feeding pattern generators). The connection

    between the Acb and the LHA has also been shown

    to modulate food intake (Fig. 4). Kelley andcolleagues have shown that blocking excitation of

    the GABA-encoded Acb results in hyperphagia, an

    effect contingent upon intact transmission in the

    LHA [232]. Glutamatergic excitation in the Acb is

    predominantly supplied by the cortex, so that this

    cortex-Acb-LHA pathway could offer the prefrontal

    cortex in particular a channel for inhibiting feeding

    behaviour in favour of other behaviours [227].

    These data indicate that in the regulation of

    ingestive behaviour, the Acb is indeed at the

    interface between motivation and action as ori-

    ginally postulated for this structure [233].

    Studies on the food intake experience inhumans and monkeys: relevance forunderstanding obesity

    Defining the cortical involvement in feeding beha-

    viour is likely vital for understanding human

    eating disorders. Crosstalk between the cerebral

    cortex and primary sensors is extensive; impres-

    sively, the hypothalamus provides the largest

    external cortical input with the exception of the

    thalamus [198], and reciprocal connections areplenteous [234]. Investigations of monkeys and

    humans by Rolls and collaborators have revealed

    that the sensory properties of food are processed in

    two steps in the cortex [see 235]. The insular

    cortex, functionally and neuroanatomically impli-

    cated as viscerosensory cortex [236], acts as a

    primary taste cortex where these individual fea-

    tures, i.e. taste, appearance, smell and texture, are

    2005 Blackwell Publishing Ltd Journal of Internal Medicine 258: 301327

    R E V I EW : B R A I N C O N T RO L O F F O O D I N T A KE 3 1 3

  • 8/12/2019 Food Intake Review 2005

    14/27

    represented to determine which food is being

    ingested. Individual neurones are sensitive to

    single stimuli, and do not adapt their firing even

    when the stimulus (e.g. glucose) has been present

    for a long time [237]. The orbitofrontal cortex

    (which receives direct input from the insularcortex), however, acts as a higher-order taste

    cortex, which determines how pleasant a particular

    food is. Orbitofrontal neurones are broadly tuned

    to react to multiple sensory features, so that the

    firing of these cells result from the combined

    inputs from several sensory modalities [238],

    although different cells respond to different quan-

    titative combinations of stimuli. The subjective

    pleasantness rating is proportional to orbitofrontal

    activation and this activation drops accordingly

    when a particular food is eaten to satiety [239]

    (Fig. 6). Thus, computations within the orbitofron-tal cortex confer hedonic qualities upon the

    feeding circuitry, a role whose importance in

    human appetite may be underestimated when

    extrapolating from rodent studies. The orbitofron-

    tal cortex feeds directly into the LHA, which may

    thus constitute an important nexus for linking the

    subjective experience of food with homeostatic

    signals.

    The relationship of the hedonic/pleasure experi-

    ence to human obesity has begun to be addressed in

    neuroimaging studies, further emphasizing the cor-

    tical parcellation of different feeding-related process-

    ing. Whereas during hunger, activation is observed

    predominantly in regions associated with the regu-

    lation of emotions (limbic and paralimbic cortex),

    satiety is followed by activation in the prefrontalcortex, postulated to play a role in the inhibition of

    inappropriate behaviours [240]. The presentation of

    a palatable sweet solution after a day and a half

    of fast resulted in increased signal from the insular

    cortex [241]. Moreover, almost all observed changes

    are accentuated in obese compared with lean

    subjects, i.e. both increases and decreases in activity

    are of greater amplitude [241] (Fig. 7). These

    differences are not exclusively accounted for by the

    hyperglycaemia and hyperinsulinaemia manifest in

    obese subjects. The functional implication of the

    intensified patterns of activation and inactivation inobese subjects is not clear at present, but may be of

    pathophysiological importance. Indeed, these re-

    sponses persist also after weight loss in postobese

    subjects [242] suggesting that they are not a

    consequence of the overweight as such, although

    it is not known if such accentuated responses are

    seen also prior to the development of obesity.

    Elucidating these mechanisms may be significant

    also for understanding the human response to food

    advertisements.

    10

    20

    40

    0 50 80

    +1

    +2

    0

    -1

    -2

    pe

    on

    ct

    banana odour

    mango odour

    30

    mL

    Volume of 20% blackcurrant juice

    food

    satiating

    response to

    Behavioural

    Firing rate

    (spikes s1)Blackcurrant odour

    Spontaneous

    Fig. 6 Neurones in the orbitofron-

    tal cortex decrease their firing in

    response to a particular food as that

    food is consumed to satiety. Extra-

    cellular recording from an odour-

    responsive neurone in the orbito-

    frontal cortex of a male macaque.

    The firing rate of one neurone in

    response to different odours was

    recorded whilst the animal con-

    sumed a blackcurrant juice. As

    satiety increases (lower panel), the

    electrophysiological response toblackcurrant odour diminishes,

    whereas the response to unrelated

    odours [banana, mango, phenyl

    ethanal (pe), citral (ct), onion (on)]

    is unaffected or elevated. Figure

    produced by H.D. Critchley and

    E.T. Rolls and is used with permis-

    sion from the American Physiolo-

    gical Society; J Neurophysiol

    1996;75:16731686.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine258: 301327

    3 14 C . B RO BE RG ER

  • 8/12/2019 Food Intake Review 2005

    15/27

    Development of the feeding circuitry

    A series of recent studies have shed light both on the

    normal ontogenetic development of hypothalamic

    circuits as well as how these processes are influenced

    by the nutritional state of the young animal, with

    important implications for the aetiology of obesity. At

    birth, the hypothalamus is rather sparsely inner-

    vated by Arc NPYergic fibres, and in, e.g. the PVH asubstantial NPY/AGRP innervation is seen first at

    postnatal day (P) 15 [197, 243, 244]. Similarly,

    whilst Arc NPY and AGRP mRNAs are detectable

    from birth, levels peak at P15, and drop to adult

    levels by P30 [244] (Fig. 8). This development

    parallels the maturation of the ability to regulate

    suckling in response to caloric demands [245],

    linking changes within the Arc metabolic sensor

    and adult control of feeding. Notably, failure in the

    development of the Arc is associated with fatal

    anorexia in a genetic model [246, 247]. It should be

    mentioned, that during the early postnatal period,

    and under certain physiological circumstances, tran-

    sient expression of NPY in other hypothalamic nuclei

    can be seen [243, 248]. The role of these transitory

    NPY projections remains to be determined.

    As the Arc provides a main conduit for leptin, it isperhaps not surprising, given these data, that pups

    are unable to respond to leptin by changing their

    food intake [249], nor to ghrelin [250]. Yet, there is

    a pronounced peak in serum leptin prior to weaning

    [251]. Recent data from Bouret et al. [252] suggest

    that a postnatal leptin surge is essential for the

    development of Arc projections. In adult ob/obmice,

    there is a distinct paucity of Arc-derived terminals.

    Fig. 7 Lean and obese subjects show differences in brain activation during different states of hunger. Statistical parametric maps of

    significant brain responses (P 0.005, not corrected for multiple comparisons) to hunger and early satiety in obese (top row) and lean

    (bottom row) subjects, respectively, at 4 mm above (left images), 4 mm below (middle images), and 16 mm below (right images) a

    horizontal plane passing through the anterior and posterior commissures (coordinates from the Montreal Neurological Institute). The right

    hemisphere in each section is on the readers right. The T-value colour-coded areas were regions of the brain in which significant

    changes in blood flow (a marker of neural activity) were detected in response to hunger (from yellow to white, in increasing order of

    T-value), as stimulated by a 36-h fast, or in response to early satiety (from blue to green, in increasing order of T-value), as stimulated by

    consumption of a satiating liquid meal [304]. The figure is intended for visual inspection only of several brain regions, where significantly

    greater responses in obese compared with lean individuals were detected, including the middle temporal gyrus (TEMP) insula (INS),dorsolateral prefrontal cortex (DLPFC), hippocampus (HIPP), temporal pole (T.POLE), orbitofrontal cortex (OFC), and ventrolateral

    prefrontal cortex (VLPFC). Figure generously provided by Dr Angelo DelParigi.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine 258: 301327

    R E V I EW : B R A I N C O N T RO L O F F O O D I N T A KE 3 1 5

  • 8/12/2019 Food Intake Review 2005

    16/27

    However, administration of leptin to ob/ob mice

    during early development, but not in adulthood,

    results in innervation patterns similar to what is

    seen in lean littermates, in parallel with a normal-

    ization of body weight. These data provide a novelmechanism for how nutritional signals in the early

    postnatal stage exerts long-lasting effects on the

    metabolic wiring in the adult. It will be of interest to

    determine if similar mechanisms are at play in the

    development of the brainstem-vagal system. In this

    context, the importance of the prenatal metabolic

    environment should also be remembered. Gesta-

    tional diabetes and obesity is associated with obesity

    in the offspring of both rats [253] and humans

    [254]. An extensive long-term study is currently in

    progress to investigate what changes can be seen in

    central metabolic circuits in nonhuman primates

    following intrauterine exposure to diabetes [255].Conversely, changes in hypothalamic circuitry dur-

    ing senescence may contribute to the loss of appetite

    that often accompanies ageing [17]. Such changes

    are observed in older rats, who display significantly

    lower levels of expression of NPY [256] and POMC,

    as well as POMC-positive neurones [257] and

    diminished dendritic arbours [258] in the arcuate

    nucleus compared with younger animals.

    Fig. 8 Arc NPY/AGRP projections to the PVH develop during the postnatal period in the rat. Figure displays confocal micrographs of

    double-label immunofluorescence for NPY (red) and AGRP (green) in the PVH. Double-labelled fibres are shown in yellow. These images

    demonstrate that at postnatal day 5 (P5) that there are minimal Arc NPY/AGRP projections to the PVH, whilst there is an abundance

    of NPY fibres that originate from other sources. By P10 there is a significant concentration of Arc NPY/AGRP projection in the PVH, but

    they do not reach the adult levels until around P15. Images represent a 10- lm thick collection of optical sections collected at 0.5-lm

    intervals. Images were captured with a 25 oil objective (0.75 NA) and represent an area of 400 400 lm. Figure generously provided

    by Dr Kevin Grove.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine258: 301327

    3 16 C . B RO BE RG ER

  • 8/12/2019 Food Intake Review 2005

    17/27

    Mechanisms of anorexia in infection andcancer

    The central systems mediate not only obesity but also

    anorexia of various aetiologies. Infection-based anor-

    exia is a well-established and clinically highly rele-vant model [259], which has been instrumental in

    uncovering the pathways through which microor-

    ganisms cause reduced food intake. Bacterial compo-

    nents such as LPS from Gram-negative bacteria and

    muramyl dipeptide from Gram-positive bacteria acti-

    vate CD14- and Toll-like receptors on host T-cells to

    induce production of cytokines such as interferon-c;

    these results are corroborated by experiments

    employing genetic removal of strategic components

    along the pathway [260]. Cytokines are produced

    peripherally, but activate vagal afferents [261]. In

    addition, cytokines stimulate prostaglandin produc-tion via the enzyme cyclooxygenase 2 (COX-2) in

    cerebral endothelial and perivascular cells [262]. This

    latter pathway is important for the induction of LPS-

    induced anorexia, and can be blocked with indo-

    methacin and other, more selective, COX-2 inhibitors

    [263]. Further downstream, the prostaglandins acti-

    vate neurones producing serotonin (5-HT; [264]), a

    known anorexigenic transmitter [265, 266], which

    in turn has recently been shown, via 5-HT2Crecep-

    tors, to stimulate melanocortin signalling [267].

    Thus, the signalling cascade set off by pathogenic

    bacteria ultimately results in activation of the centralanorexigenic system. These results may shed light

    also on the anorexia accompanying noninfectious

    inflammatory conditions. In this context it is inter-

    esting to note that, based on structure and signalling

    pathways, leptin itself belongs to the cytokine family

    [268, 269].

    While anorexia is an important component also of

    wasting in cancer patients, nutritional supplements

    only alleviate part of the cachectic syndrome [270],

    which accounts for a fifth of cancer deaths [271].

    Cachexia differs from starvation in that both adipose

    and lean mass is lost, whereas starvation primarilydecreases fat stores [272]. A main explanation for

    this relationship is that many cancer tumours

    secrete proteins, e.g. proteolysis-inducing factor

    [273], which suppress protein synthesis in skeletal

    muscle, via activation of an ubiquitin proteolytic

    pathway [274]. Parenteral nutrition may thus be of

    very limited value to the patient if such catabolic

    mechanisms are not interrupted. However, it turns

    out that a polyunsaturated fatty acid found in fish,

    eicosapentaenoic acid (EPA), suppresses the activity

    of the proteolytic complex whilst simultaneously

    inhibiting tumour growth [275]. This finding has

    promising bedside implications; adding EPA to the

    diet of cancer patients attenuates muscle degrada-tion and stabilizes body weight [276].

    From rodent to human: genetic dissection

    In few fields of medicine is the question of nature

    versus nurture more immediate than in the regula-

    tion of body weight. While it is evident that the

    incidence of obesity has accelerated far more rapidly

    than can be explained by population shifts within the

    genome, it is also true that individual differences

    determine how we react in an energy-dense environ-

    ment. Aptly summed up by Olden and Wilson [277]:genes load the gun, environment pulls the trigger.

    Studies of monozygotic twins show that the heritable

    component of obesity equals that of height and

    surpasses virtually every other major disease studied,

    e.g. breast cancer, schizophrenia, cardiovascular

    disease [278280]; some 40% of obesity can be

    attributed to genetic causes [281]. In a series of

    elegant studies, in particular by ORahilly, Farooqi

    and their colleagues, it has been shown that muta-

    tions in several components of the anorexigenicsignal

    chain, including leptin [282] (Fig. 9), the leptin

    receptor [283], POMC [284] and neuropeptide pro-cessing enzymes [285] result in severe early-onset

    obesity in humans. While these monogenic disorders

    incontrovertibly demonstrate that genetic abnormal-

    ities can cause obesity, it cannot automatically be

    concluded that mutations and sequence variants are

    common causes of the metabolic syndrome.

    However, it is now becoming apparent that

    mutation of a particular key signalling protein, the

    MC4R [286], accounts for as many as 5% of cases

    of severe obesity [140, 287289]. There is a

    remarkably solid relationship between the severity

    of the mutation, as revealed in in vitro assays, andthe size of a test meal consumed by the patient

    [140]. Indeed, the heritable component is almost

    exclusively represented by increased intake of

    energy, with only a small component accounted

    for by changes in resting metabolic rate [140]. (In

    contrast, in the mouse MC4R)/) counterpart,

    deficient energy expenditure is an important factor

    underlying overweight [136].) The sometimes

    2005 Blackwell Publishing Ltd Journal of Internal Medicine 258: 301327

    R E V I EW : B R A I N C O N T RO L O F F O O D I N T A KE 3 1 7

  • 8/12/2019 Food Intake Review 2005

    18/27

    modest phenotypes observed in rodent gene knock-

    out experiments (e.g. NPY gene deletion [125])

    have been interpreted as evidence for a high

    degree of redundancy within the system under-

    lying metabolic regulation. However, genetic stud-

    ies such as these (as well as, notably, observed in

    nematodes [212]), suggest that there are weakpoints distributed throughout the system where

    minute changes in nucleotide sequence can have

    profound effects on the expression of behaviour.

    These vulnerable links along the metabolic signal-

    ling chain offer promising targets for therapeutic

    intervention.

    Therapeutic prospects

    As pointed out in the Introduction, effective treat-

    ments for eating disorders are short in supply and

    urgently needed. While much can be gained simplyby increased efforts to educate patients in the

    benefits of weight loss and exercise [290, 291], it

    is also clear that such therapies in many cases fail in

    the absence of adequate pharmacological buttress.

    The anti-obesity drugs presently used in clinical

    practice have relatively modest effects, and others

    still have been withdrawn due to intolerable cardio-

    vascular adverse effects [22]. These drugs have often

    targeted broadly distributed systems, e.g. serotonin

    pharmacology, and it is perhaps not surprising that

    alterations ensue within multiple body functions.

    Another issue to consider is the timing of adminis-

    tration of feeding-regulatory drugs, which may be

    more crucial than in any other therapeutic applica-tion (T. Bartfai, personal communication). However,

    based largely on the research summarized above,

    several novel compounds are now being tested in

    clinical trials, phases II and III [22, 292]. Many of

    these compounds target neuropeptide systems. The

    selective neuroanatomical distribution of many

    neuropeptides may provide an advantage in

    attempting to minimize side-effects, and whilst the

    field of peptide-based pharmaceuticals has not lived

    up to initial hopes, the clinical experience from

    opioid drugs such as morphine and naloxone [293]

    suggest that interfering with peptide signalling canproduce powerful neuropsychiatric effects in

    humans [294]. In particular, drug development

    initiatives have centred on the melanocortin system

    [295], encouraged by the human genetic

    data reviewed above, and several nonpeptide com-

    pounds are now being tested in a clinical setting

    [22].

    The cannabinoid system also provides an attract-

    ive target, with promising early results with the

    inverse CB1 receptor agonist, rimonabant [296],

    currently in phase III clinical trial. Initial evalua-

    tions suggest that rimonabant decreases bodyweight by ca 510%, with beneficial effects also on

    insulin resistance and dyslipidaemia [292]. Interest-

    ingly, this drug has the added benefit of facilitating

    smoke cessation. However, the full safety profile of

    rimonabant has not yet been released. Conversely,

    the active component of marijuana, D9-tetrahydro-

    cannabinol, is also being evaluated for treatment of

    AIDS-associated anorexia [297].

    Early hopes for a leptin-based obesity regimen

    were quelled by clinical trials showing only very

    limited weight loss following leptin administration to

    obese subjects [298], and there are also indicationsthat such treatment may in itself produce the leptin

    resistance hypothesized to be a part of obesity [299].

    It should be pointed out, however, that in the rare

    cases of genetic leptin-deficiency, treatment with

    recombinant leptin has virtually normalized body

    weight in near-fatally obese children, whilst simul-

    taneously inducing age-appropriate puberty [300]

    (Fig. 9). The same treatment has also been used to

    Fig. 9 Leptin deficiency in humans responds to leptin treatment.

    A 3-year-old boy with congenital leptin deficiency with severe

    obesity (body weight 38 kg; BMI SD 7.2) (left). On the right,

    the same patient, after four years of daily subcutaneous admin-istration of recombinant leptin. Leptin treatment results in a

    dramatic decrease in adiposity (body weight 29 kg; BMI SD

    0.9) and normalization of all metabolic abnormalities including

    hyperinsulinaemia. Figure generously provided by Drs Sadaf

    Farooqi and Stephen ORahilly.

    2005 Blackwell Publishing Ltd Journal of Internal Medicine258: 301327

    3 18 C . B RO BE RG ER

  • 8/12/2019 Food Intake Review 2005

    19/27

    successfully rectify several of the metabolic abnor-

    malities associated with lipodystrophy [301, 302].

    Furthermore, pertaining to a not uncommon diag-

    nosis, leptin treatment has recently been demon-

    strated as a highly promising therapy for functional

    amenorrhoea [303].

    Conclusion

    In summary, significant advances in our under-

    standing of feeding behaviour have been achieved

    using a combination of clinical, behavioural, elec-

    trophysiological, anatomical, genetic and imaging

    techniques. This investigation has defined an under-

    lying circuitry and neuropharmacology, which can

    now be probed for therapeutic targets. Thus, whilst

    many questions remain to be answered, in partic-

    ular regarding the causal mechanisms of obesity andanorexia as well as the central circuitry bridging

    metabolic input and output, there is reason for hope

    in offering effective treatments for these exception-

    ally common and debilitating disorders.

    Conflict of interest statement

    No conflict of interest was declared.

    Acknowledgements

    The author gratefully acknowledges generous finan-cial support from Wenner-Gren Stiftelserna, Vet-

    enskapsradet, Jeanssons Stiftelser, Hagbergs Stiftelse,

    Rut och Arvid Wolffs Stiftelse, Thurings Stiftelse,

    Svenska Lakaresallskapet, Ake Wibergs Stiftelse, Kgl.

    Vetenskapsakademien, Hedlunds Stiftelse, Magnus

    Bergvalls Stiftelse, Lars Hiertas Minne, Axel Linders

    Stiftelse, Langmanska Kulturfonden, Teodor Neran-

    ders Fond and internal funds of Karolinska Institutet.

    References

    1 Galbraith JK.The New Industrial State. Boston, MA: Hough-ton Mifflin, 1967.

    2 World Health Organization.Obesity: Preventing and Managing

    the Global Epidemic. Geneva: World Health Organization,

    2000.

    3 National Task Force on the Prevention and Treatment of

    Obesity. Overweight, obesity, and health risk. Arch Intern

    Med2000; 160: 898904.

    4 World Health Organization. Diet, nutrition and the preven-

    tion of chronic diseases. World Health Organ Tech Rep Ser

    2003; 916: iviii, 1149, backcover.

    5 Maclure KM, Hayes KC, Colditz GA, Stampfer MJ, Speizer FE,

    Willett WC. Weight, diet, and the risk of symptomatic gall-

    stones in middle-aged women. N Engl J Med 1989; 321:

    5639.

    6 Felson DT, Anderson JJ, Naimark A, Walker AM, Meenan

    RF. Obesity and knee osteoarthritis. The Framingham Study.

    Ann Intern Med1988; 109: 1824.

    7 Grodstein F, Goldman MB, Cramer DW. Body mass index

    and ovulatory infertility. Epidemiology 1994; 5: 24750.

    8 Rexrode KM, Hennekens CH, Willett WCet al.A prospective

    study of body mass index, weight change, and risk of stroke

    in women. JAMA 1997; 277: 153945.

    9 Scheinfeld NS. Obesity and dermatology. Clin Dermatol

    2004; 22: 3039.

    10 Gottschlich MM, Mayes T, Khoury JC, Warden GD. Signifi-

    cance of obesity on nutritional, immunologic, hormonal,

    and clinical outcome parameters in burns. J Am Diet Assoc

    1993; 93: 12618.

    11 Calle EE, Thun MJ, Petrelli JM, Rodriguez C, Heath CW Jr.

    Body-mass index and mortality in a prospective cohort of

    U.S. adults. N Engl J Med1999; 341: 1097105.

    12 Puhl R, Brownell KD. Bias, discrimination, and obesity.Obes

    Res 2001; 9: 788805.

    13 Pontiroli AE. Type 2 diabetes mellitus is becoming the most

    common type of diabetes in school children. Acta Diabetol

    2004; 41: 8590.

    14 Hoek HW, van Hoeken D. Review of the prevalence and

    incidence of eating disorders. Int J Eat Disord2003;34:383

    96.

    15 Plata-Salaman CR. Cytokines and feeding.News Physiol Sci

    1998; 13: 298304.

    16 Strasser F, Bruera ED. Update on anorexia and cachexia.

    Hematol Oncol Clin North Am 2002; 16: 589617.

    17 Briefel RR, McDowell MA, Alaimo Ket al. Total energy in-

    take of the US population: the third National Health and

    Nutrition Examination Survey, 19881991. Am J Clin Nutr

    1995; 62: 1072S80S.

    18 Johnson D, Drenick EJ. Therapeutic fasting in morbid obes-

    ity. Ar