Emerging aspects of pharmacotherapy for obesity and metabolic syndrome

Emerging aspects of pharmacotherapy for obesity and metabolic syndrome

Pharmacological Research 50 (2004) 453–469 Emerging aspects of pharmacotherapy for obesity and metabolic syndrome Enzo Nisoli a,b,∗ , Michele O. Carr...

470KB Sizes 2 Downloads 88 Views

Pharmacological Research 50 (2004) 453–469

Emerging aspects of pharmacotherapy for obesity and metabolic syndrome Enzo Nisoli a,b,∗ , Michele O. Carruba a,b a

Department of Preclinical Sciences, Center for Study and Research on Obesity, L. Sacco Hospital, University of Milan, LITA Vialba, via G.B. Grassi 74, 20157 Milan, Italy b Istituto Auxologico Italiano, Milan, Italy Accepted 9 February 2004

Abstract Obesity is a multifactorial, chronic disorder that has reached epidemic proportions in most industrialized countries and is threatening to become a global epidemic. Obese patients are at higher risk from coronary artery disease, hypertension, hyperlipidemia, diabetes mellitus, cancers, cerebrovascular accidents, osteoarthritis, restrictive pulmonary disease, and sleep apnoea. In particular, visceral fat accumulation is usually accompanied by insulin resistance or type 2 diabetes mellitus, hypertension, hypertriglyceridemia, high uremic acid levels, low high density lipoprotein (HDL) cholesterol to define a variously named syndrome or metabolic syndrome. Metabolic syndrome is now considered a major cardiovascular risk factor in a large percentage of population in worldwide. Both obesity and metabolic syndrome are particularly challenging clinical conditions to treat because of their complex pathophysiological basis. Indeed, body weight represents the integration of many biological and environmental components and relationships among fat and glucose tolerance or blood pressure are not completely understood. Efforts to develop innovative anti-obesity drugs, with benefits for metabolic syndrome, have been recently intensified. In general two distinct strategies can be adopted: first, to reduce energy intake; second, to increase energy expenditure. Here we review some among the most promising avenues in these two fields of drug therapy of obesity and, consequently, of metabolic syndrome. © 2004 Elsevier Ltd. All rights reserved. Keywords: Obesity; Anti-obesity drugs; Metabolic fitness; Obesity-related disorders; Metabolic syndrome; Leptin; Thermogenic drugs; Mitochondria

1. Introduction Obesity is a multifactorial, chronic disorder that has become a global epidemic (globesity) [1]. It is not just a concern for adults as the number of overweight/obese children and adolescents has doubled in the past 2–3 decades in the US [2]. Overweight children and adolescents are more likely to become overweight or obese adults [2,3]. Obese patients are at higher risk from coronary artery disease, hypertension, hyperlipidemia, diabetes mellitus, cancers, cerebrovascular accidents, osteoarthritis, restrictive pulmonary disease, and sleep apnoea [4,5]. The risk of morbidity and mortality increases with an increase in body weight beyond a body mass index (BMI) (weight in kg/height in m2 ) of 27 and with an increase in waist circumference (as an index of visceral localization of fat). Obesity reduces life expectancy [1]. In the Nurses’ Health Study, in which over 115,000 women 30–55 ∗ Corresponding author. Tel.: +39-02-50319682; fax: +39-02-50319683. E-mail address: [email protected] (E. Nisoli).

1043-6618/$ – see front matter © 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.phrs.2004.02.004

years of age and without known cardiovascular disease were followed for 16 years, the risk of death was 60–70% higher among subjects with a BMI between 29 and 32 kg/m2 than among subjects with a BMI of 25–27 kg/m2 . These figures translate into 1260 excess lives lost per million women per year as a consequence of an average weight difference of only 13 kg (28 lbs) [6]. In the US, approximately 300,000 deaths a year are currently associated with overweight and obesity [3]. People with the metabolic syndrome are at increased risk for developing diabetes mellitus [7] and cardiovascular disease [8] as well as increased mortality from cardiovascular disease and all causes [9]. The recently released Third Report of the National Cholesterol Education Program Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III; ATP III) draws attention to the importance of the metabolic syndrome and provides a working definition of this syndrome for the first time [10]. In addition, the report has drawn specific attention to the importance of this syndrome as a new target of risk-reduction therapy.

454

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

A recent report suggested that the prevalence of the metabolic syndrome increases with age, affecting more than 40% of those older than 60 years [11]. Based on ageadjusted estimates, about a quarter of the population has the metabolic syndrome, representing about 47 million Americans [11]. As detailed in the ATP III report, participants having three or more of the following criteria were defined as having the metabolic syndrome: abdominal obesity (waist circumference >102 cm in men and >88 cm in women); hypertriglyceridemia (150 mg/dl or 1.69 mmol/l); low high density lipoprotein (HDL) cholesterol (<40 mg/dl (1.04 mmol/l) in men and <50 mg/dl (1.29 mmol/l) in women); high blood pressure (130/85 mmHg); high fasting glucose (110 mg/dl or 6.1 mmol/l). Williamson et al. [12] reported that the association between intentional weight loss and longevity in middle-aged overweight women appears to depend on their health status: in women with obesity-related health conditions (n = 15,069), intentional weight loss of any amount was associated with a 20% reduction in all-cause mortality, primarily due to a 40–50% reduction in mortality from obesity-related cancers; in women with no preexisting illness (n = 28,388), intentional weight loss of greater than or equal to 9.1 kg that occurred within the previous year was associated with about a 25% reduction in all-cause, cardiovascular, and cancer mortality; however, loss of <9.1 kg or loss that occurred over an interval of >1 year was generally associated with small to modest increases in mortality. More recently, to examine the relationships among intention to lose weight, weight loss, and all-cause mortality, a prospective cohort study using a probability sample of the US population was performed [13]. Participants were 6391 overweight and obese persons (BMI ≥ 25 kg/m2 ) who were at least 35 years of age. Intention to lose weight and weight change during the past year were assessed by self-report in 1989. Vital status was followed for 9 years. Hazard rate ratios (HRRs) were adjusted for age, sex, ethnicity, education, smoking, health status, health care utilization, and initial BMI. Compared with persons not trying to lose weight and reporting no weight change, those reporting intentional weight loss had a 24% lower mortality rate (HRR: 0.76 (95% CI, 0.60–0.97)) and those with unintentional weight loss had a 31% higher mortality rate (HRR: 1.31 (CI, 1.01–1.70)). However, mortality rates were lower in persons who reported trying to lose weight than those in not trying to lose weight, independent of actual weight change. Compared with persons not trying to lose weight and reporting no weight change, persons trying to lose weight had the following HRRs: no weight change, 0.80 (CI, 0.65–0.99); gained weight, 0.94 (CI, 0.65–1.37); lost weight, 0.76 (CI, 0.60–0.97). Thus, attempted weight loss was associated with lower all-cause mortality, independent of weight change. Self-reported intentional weight loss was associated with lower mortality rates, and weight loss was associated with higher mortality rates only if it is unintentional (see also Section 2).

2. Management of obesity and metabolic syndrome Obesity is a particularly challenging clinical condition to treat because of its complex pathophysiological basis. Indeed, body weight represents the integration of many biological and environmental components [14,15]. Rather than focusing primarily on body weight, many experts are focusing on the so-called “metabolic fitness”, that tracks the metabolic health of obese individuals. Metabolic fitness is defined as the absence of biochemical risk factors associated with obesity (elevated fasting concentrations of cholesterol, triglycerides, glucose, or insulin; impaired glucose tolerance; or elevated blood pressure). Thus, weight loss should be viewed as a modality to improve health [16]. Modest weight reduction, in the range of 5–10% of initial body weight, has been shown to improve obesity-related morbidity and mortality [1]. In women 40–60 years of age who had never smoked, moderate but intentional weight loss reduced all-cause mortality by 20% and diabetes-associated mortality by 30–40% [12]. Modest weight reduction has also been associated with clinically significant improvements in hypertension [17], lipid abnormalities [18] and glycaemic control [19,20]. Recently, the Finnish Diabetes Program [21] and the Diabetes Prevention Program [22] both reported that, in overweight patients losing approximately 5% of their body weight and increasing their physical activity the risk of developing type 2 diabetes was reduced by 58%. Caloric restriction, physical exercise and behavioral modification constitute the standard model for obesity treatment. Successful weight management implies not only initial weight loss over a short period of time, but also maintenance of reduced weight over a period of years. In most cases, dietary changes, exercise and behavioral modification, either alone or in combination, are generally met with poor long-term outcomes [23]. Pharmacological therapy is an adjunct to the treatment of obesity. Anti-obesity drugs must be used only in the context of a comprehensive management programme that includes the standard model. If physiological intervention does not induce weight loss after 6 months, the use of anti-obesity drugs may be considered for weight management in high-risk patients [24–26]. Thus anti-obesity drugs may have a role in weight reduction in patients whose condition is refractory to nonpharmacological measures and for the maintenance of weight loss. To date, agents for the management of obesity have been limited and unsatisfactory. Amphetamine have profound euphoric actions and carry the potential for abuse [27]. Phentermine has stimulant and sympathomimetic effects through catecholaminergic pathways [27]. Phenylpropanolamine-containing appetite suppressants have been associated with increased risk of hemorrhagic stroke in women [28], which resulted in their withdrawal by the U.S. Food and Drug Administration (FDA). Fenfluramine and dexfenfluramine, which are also centrally acting appetite suppressants, act predominantly by releasing serotonin. These agents were also withdrawn from the market

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

because of their association with pulmonary hypertension and heart valve damage [28,29]. Sibutramine and orlistat, two newer therapies with relatively favorable efficacy and safety profiles, have been approved for weight management in conjunction with lifestyle modifications [161]. These mentioned drugs remain “symptomatic” and they are not “curative” drugs. Thus, exploding research has been finding many new potential drug targets. This review will focus on central and peripheral molecular processes involved in the maintenance of energy balance.

3. Central and peripheral processes that control energy balance Both central and peripheral mechanisms are involved in energy balance. In particular, fat depots are able to storage energy as fatty acids in response to fuel availability, and this information can be transfer to brain centers by means of different molecules. In particular, the production of the hormone leptin by adipose tissue is regulated by energy balance. When energy (i.e. fat) stores are replete, leptin production is high. Conversely, leptin production is inhibited when energy stores are depleted during, for example, prolonged fasting [30,31]. Circulating leptin levels thus reflect the status of body energy reserves, and energy balance throughout the body is regulated via leptin-mediated control of processes that are involved in energy intake and utilization. Thus, the fall in leptin levels when energy stores are inadequate (e.g. during starvation) enhances appetite and decreases energy utilization. By contrast, when energy stores are adequate, high leptin levels decrease the drive to eat and enable utilization of energy by the systems described below. There are multiple leptin receptor (LR) isoforms, which result from alternative mRNA splicing of the transcript of the lepr gene and/or from proteolytic processing of the subsequent protein products [32,33]. The lepr gene contains 17 common exons and several alternatively spliced exons. In mice, the five distinct LR isoforms that have been identified are designated LRa–LRe. In all species, LR isoforms can be divided into three classes: secreted, short and long. The secreted forms are either alternative splice products (e.g. murine LRe, which contains only the first 14 exons of lepr) or proteolytic cleavage products of membrane-bound forms of LR. These secreted forms contain only extracellular domains that bind circulating leptin, perhaps regulating the concentration of free leptin [34]. Short forms (LRa, LRc and LRd in mice) and the long form (LRb in mice) contain exons 1–17 of lepr and therefore have identical extracellular and transmembrane domains as well as the same first 29 intracellular amino acids, but then diverge in sequence because of alternative splicing of 3 exons. LRb is crucial for leptin action. Indeed, the db/db mice described originally lack only LRb because of a mutation that causes mis-splicing of LRb mRNA, but have a phenotype

455

that is indistinguishable from that of db3J /db3J mice (which are deficient in all LR isoforms) and of leptin-deficient ob/ob animals [30–33]. The function of short-form LRs is less clear, although proposed roles include the transport of leptin across the blood–brain barrier [35,36]. Many of the effects of leptin are attributed to effects in the CNS, particularly in the basomedial hypothalamus, the site of highest LRb mRNA expression [37–39]. LRb is present with the highest levels in neurons of the nuclei of the basomedial hypothalamus, including the arcuate (ARC), dorsomedial hypothalamic (DMH) and ventromedial hypothalamic (VMH) nuclei [38,39]. Chemical and physical ablation of these nuclei results in increased feeding and neuroendocrine abnormalities that are similar to the phenotypes of db/db and ob/ob mice. This indicates that these hypothalamic nuclei, which make up the so-called ‘satiety center’, are crucial sites of leptin action [37,40]. Within these basomedial hypothalamic nuclei, LRb mRNA is expressed most highly in the ARC, where it is found in at least two distinct populations of neurons (Fig. 1). One population synthesizes neuropeptide Y (NPY) and agouti-related peptide (AgRP) and the other synthesizes pro-opiomelanocortin (POMC) [37,40]. POMC is processed to produce the powerful anorectic (appetite-suppressing) peptide ␣-melanocyte-stimulating hormone (␣-MSH) in LRb/POMC neurons. LRb stimulates the synthesis of POMC and activates LRb/POMC neurons [40,41]. AgRP inhibits ␣-MSH signaling and NPY is an orexigenic (appetite-stimulating) hormone that also suppresses the central LRb-mediated growth and reproductive axes [42–45]. Leptin acts via LRb to inhibit NPY/AgRP neurons and suppress expression of these neuropeptides. Thus, LRb signaling stimulates the production of anorectic neuropeptides and suppresses levels of orexigenic peptides. Conversely, a decrease or deficiency in leptin activity (e.g. during starvation and in ob/ob and db/db mice) stimulates appetite by suppressing synthesis of anorectic neuropeptides (e.g. POMC) and increasing expression of orexigenic peptides (e.g. NPY and AgRP) [37,40].

4. Interventions to reduce energy intake 4.1. Leptin and leptin receptor The complete congenital absence of the adipocyte-derived hormone leptin leads to a syndrome of intense hyperphagia and morbid obesity in humans [46,47] and rodents [48], which can be reversed by the administration of recombinant leptin [49,50]. So, for children with the rare congenital leptin deficiency, recombinant leptin represents a pronounced and, indeed, life-saving therapy. Unfortunately, severe leptin deficiency is not often found in human obesity, and leptin supplementation in obese individuals seems to have only modest effects on fat mass [51]. Given that they are fat, but

456

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

Fig. 1. Diagrams of rat brain, showing major hypothalamic regions implicated in adiposity signaling and regulation of food intake. The small figure at the top is a longitudinal view of a rat brain, with olfactory bulb at the anterior end on the left and the caudal hindbrain on the right. Cross-sections of the brain at two levels (indicated by vertical lines) are shown at middle (left and right). First-order neurons responding to adiposity signals are located in the arcuate nucleus (ARC) and project anteriorly to the PVN as well as the PFH. Other regions implicated in regulating food intake include the ventromedial nucleus (VMN) and dorsomedial nucleus (DMN). Abbreviations of brain structures: AM, amygdala; CC, corpus callosum; CCX, cerebral cortex; HI, hippocampus; OC, optic chiasm; SE, septum; TH, thalamus; 3V, third ventricle. NPY/AGRP and POMC/CART neurons in the ARC (bottom, right), adjacent to the third ventricle, are first-order neurons in the hypothalamic response to the circulating adiposity signals insulin and leptin. NPY and POMC are expressed in discrete populations of arcuate nucleus neurons. NPY release in the PVN and LHA/PFA regions stimulates eating, whereas release of ␣-MSH (derived from POMC) in the PVN has an anorexic effect. Locations of candidate second-order neurons (bottom, left) involved in the hypothalamic response to insulin and leptin adiposity signaling. Second-order neurons include those that express TRH and CRH in the PVN (which cause anorexia), and neurons that express orexins and MCH in the PFA and LHA (which increase feeding).

generally have high plasma levels of leptin, obese subjects can be considered to be ‘leptin resistant’ [50]. There is some evidence for impaired transport of leptin into the CNS in states of leptin resistance [52]. But resistance is also likely to be occurring at the level of neurons in the CNS that express the leptin receptor, and their downstream targets. The recent observation that subjects who are heterozygous for leptin mutations have modest reductions in plasma leptin and show increased fat mass [53], provides some hope that there might be a subgroup of subjects with relative hypoleptinaemia who might respond to leptin supplementation.

Leptin passes through the blood–brain barrier and enters the brain. The problems of peripheral delivery and CNS penetration for agonists of the leptin receptor could, of course, be solved by using a small-molecule agonist for the leptin receptor. An alternative approach has been to search for ligands that mimic the actions of leptin and might be able to either overcome or bypass leptin resistance. One such molecule is ciliary neurotrophic factor (CNTF) [54] and its analogue axokine. These molecules seem to activate signal-transduction mechanisms within the hypothalamus similar to those that

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

are activated by leptin. In contrast to leptin, these agents cause weight loss in animals with mutated leptin receptors, and also seem to be effective in diet-induced obesity [55]. 4.2. Small molecules insulin mimetics Insulin has been suggested to function as one of the adiposity signals to the brain for modulation of energy balance. Administration of insulin into the brain reduces food intake and body weight [15], and mice with a genetic deletion of neuronal insulin receptors are hyperphagic and obese [56]. However, insulin is also an anabolic factor; when administered systemically, pharmacological levels of insulin are associated with body weight gain in patients [57]. Air et al. [58] investigated the efficacy and feasibility of small molecule insulin mimetic compounds (Cpd1 and Cpd2) [59,60] to regulate key parameters of energy homeostasis. Central intracerebroventricular (i.c.v.) administration of an insulin mimetic resulted in a dose-dependent reduction of food intake and body weight in rats, and altered the expression of hypothalamic genes known to regulate food intake and body weight. In particular, POMC expression, normally suppressed by food deprivation, increased 183% in fasted, Cpd1-treated animals compared to those treated with vehicle [58]. Neuropeptide Y (NPY) expression, normally elevated in fasted animals, was suppressed by Cpd1 to 38% of fasted, vehicle-treated animals [58]. This effect is larger than that seen following insulin administration [61]. These data indicate that Cpd1 reduces food intake and body weight, at least in part, by upregulating the catabolic melanocortin system and downregulating the anabolic NPY system in the hypothalamus. Oral administration of a mimetic in a mouse model of high-fat diet-induced obesity reduced body weight gain, adiposity and insulin resistance. Thus, insulin mimetics have a unique advantage over insulin in the control of body weight and hold potential as a novel anti-obesity treatment. 4.3. Pro-opiomelanocortin and melanocortin receptors As previously mentioned a large body of both genetic and pharmacological research clearly implicates hypothalamic neurons that express pro-opiomelanocortin (POMC) as key elements in the control of appetite and energy balance [62]. A role for melanocortin signaling in the control of energy homeostasis first emerged after the cloning of the MC3- and MC4-receptor genes and the demonstration that they are expressed primarily in the brain [63]. This discovery was followed by evidence that a synthetic agonist of these receptors suppresses food intake, whereas a synthetic antagonist has the opposite effect [42]. The report that mice lacking the MC4 receptor (owing to gene targeting) are hyperphagic and very obese [64] indicates that tonic signaling by MC4 receptors limits food intake and body fat mass. Mice heterozygous for the deleted MC4 allele also become obese, although less so than homozygous knockouts [64]. Lack of a full complement of central MC4 receptors, there-

457

fore, predisposes to hyperphagia and pathological weight gain. This finding has since been extended to humans with MC4-receptor mutations [65–67]. The subsequent cloning of the Agrp gene [68] identified a peptide, AGRP, with homology to agouti that is an antagonist of MC3 and MC4 receptors [69]. The demonstration that hypothalamic AGRP expression, like that of NPY and POMC, is localized to ARC, and that it is upregulated by fasting and by leptin deficiency, indicates that antagonism of CNS melanocortin receptors is important in body-weight regulation (see [40]). Consistent with its role as an anabolic signaling molecule, AGRP causes hyperphagia when administered i.c.v. [70] or expressed transgenically [40], and the increase of food intake following a single i.c.v. injection of AGRP is sustained for up to a week [70]. Thus, the central role of the MC4 receptor in controlling body weight makes it a very attractive target for anti-obesity drug development. Interesting at this point it is the observation that i.c.v. infusion of ␣-MSH at a dose below that needed to reduce food intake, both insulin-stimulated glucose disposal and insulin-induced suppression of hepatic glucose production were increased [71]. Increased neuronal melanocortin receptor signaling is therefore sufficient to increase insulin sensitivity in vivo, although it remains to be determined whether this effect occurs independently of melanocortin-induced decreases of body adiposity. 4.4. Neuropeptide Y and its receptors NPY is one of the most abundant peptides of the hypothalamus and, when administered centrally, has the powerful and sustained effect of increasing food intake [72]. Conversely, reduction in endogenous NPY by means of antisense-oligonucleotide [73] or immunoneutralization techniques [74] leads to a decrease in food intake. NPYergic neurons are present in the ARC, where they co-express Agrp and are negatively regulated by leptin. Paradoxically for the receptors of an orexigenic factor, mice that lack Y1 , Y2 and Y5 receptors all develop mild late-onset obesity [75–77]. Npy1r- and Npy2r-deficient animals [72–77] show a normal response to the orexigenic effects of NPY, whereas the Npy5r knockout mouse [78] has a greatly reduced feeding response to NPY administration. Despite the discrepancy between pharmacological and genetic studies, Y1 - and Y5 -receptor antagonists remain attractive candidates for drug development, and several such compounds are now in development. 4.5. Other neuropeptides/receptors There are an increasing number of neuropeptides for which a central role in the control of food intake and energy balance has been proposed (Table 1). In all cases, there is good evidence that the particular peptide is synthesized in and/or acts on the hypothalamus. Furthermore, all of these peptides have been shown to reduce food intake after acute

458

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

Table 1 Peptides and neurotransmitters affecting feeding behavior Peptide/neurotransmitter

Acute effects on feeding

Chronic effects on feeding

Leptin ␣-MSH CART GLP1 CRF Urocortin Neuromedin U CCK IL-6 CNTF PYY3–36 Oxyntomodulin AGRP NPY MCH Galanin Ghrelin Endocannabinoids Orexins

− − − − − − − − − − − − + + + + + + +

− − − − − ND ND ND ND − ND ND + + + ND + ND +/−

−: Inhibition of food intake; +: stimulation of food intake; ND: experiment has not been done.

administration. However, in contrast to the situation with leptin and POMC, in few of the cases described below are there either compelling genetic experiments or convincing results from long-term administration of the peptides on fat mass. (i) CART and its receptor (yet to be identified). Fasted animals show a decrease in expression of CART (cocaine- and amphetamine-regulated transcript) messenger RNA in the ARC [79]. However, the CART knockout mouse does not become obese on a normal diet [80]. In addition, CART has effects on the control of movement, which could complicate food-intake studies [81]. Initial evidence suggested an anorexigenic role for this peptide and decreased hypothalamic CART expression was found in animal models with an obese phenotype [79]. However, recent data challenged previous observations on the appetite-inhibiting effect of CART, showing that intrahypothalamic injection of the peptide might even increase food intake [82]. (ii) Corticotrophin-releasing factor (CRF) and urocortin, and their receptors. Although the effects of CRF and urocortin on appetite seem compelling [83], it is notable that deletion of neither the CRF1 nor the CRF2 receptor [84,85] has any pronounced effect on body weight. (iii) GLP-1. The glucagon-like peptide 1 (GLP1) is a product of post-translational modification of proglucagon in the pancreas, intestinal cells and the CNS [86]. Intracerebroventricular GLP1 suppresses appetite and, more impressively, i.c.v. administration of a GLP1 antagonist increases food intake [87]. However, animals that are deficient in the GLP1 receptor show no systematic alteration in mass of body fat [88].

(iv) IL-6. Interleukin-6 (IL-6) is expressed both in adipose tissue and centrally in the hypothalamus. IL-6-deficient mice develop mature-onset obesity that is partially reversed by central IL-6 administration [89]. (v) BDNF. Brain-derived neurotrophic factor (BDNF) was studied initially for its role in the development of sensory neurons. Homozygous Bdnf-deficient mice die soon after birth, but heterozygous knockout mice show a tendency towards obesity. However, these mice also have abnormalities in locomotor activity, which would have important implications for any attempt to develop BDNF mimetics [90]. (vi) Other peptides. Finally, other peptide–receptor systems that might be worthwhile targets for anorexic drugs include the prolactin-releasing peptide and its receptor GRP10 (G-protein-coupled receptor 10) [91], as well as neuromedin U and the Neuromedin U receptor 2 (FM4), which is selectively expressed in brain [92], and oxyntomodulin, which is released from the gut postprandially in proportion to energy intake [93]. 4.6. Melanin-concentrating hormone and its receptors The melanin-concentrating hormone (MCH) is an orexigenic neuropeptide that is found in the lateral hypothalamus [94]. The Mch transcript is upregulated in the hypothalamus of leptin-deficient (ob/ob) mice, and fasting further increases this expression in both normal and ob/ob mice [95]. Intracerebroventricular injections of MCH into normal rats lead to a marked increase in food intake [95]. Borosky et al. [96] have recently reported that SNAP-7941, a selective, high-affinity MCH1 receptor (MCH1-R) antagonist, inhibited food intake stimulated by central administration of MCH, reduced consumption of palatable food, and, after chronic administration to rats with diet-induced obesity, resulted in a marked, sustained decrease in body weight. Interestingly, besides these effects on food intake, SNAP-7941 produced effects similar to clinically used antidepressants and anxiolytics in three animal models of depression/anxiety. Given these observations, an MCH1-R antagonist may be useful not only in the management of obesity but also as a treatment for depression and/or anxiety. 4.7. Ghrelin and its receptor Great excitement has been engendered by the discovery that the stomach can secrete a peptide, termed ghrelin [97], which, on administration, whether peripherally or centrally, can increase food intake in rodents [98]. Consistent with a role in initiating feeding, ghrelin levels are increased by fasting and decreased after a meal [99]. The feeding response to ghrelin seems to be dependent on NPY and AGRP, as central administration of neutralizing antibodies or antagonists to these agents abolishes the acute effect of ghrelin on feeding [98]. Agonists that interfere with the action of ghrelin on its receptor and inverse agonists

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

459

that modulate ghrelin receptor activity might be candidates for anti-obesity molecules [162], although impairment of growth-hormone secretion, with resultant adverse effects on metabolism, is a potential risk.

tor down-regulation may limit the efficacy of prolonged PYY3–36 administration.

4.8. PYY3–36

The appetite-stimulating effect of marijuana in humans has been well known for centuries [104]. Several reports have demonstrated that administration of cannabinoids stimulates food intake in animal models [105,106]. Both peripheral and central administration of anandamide, one of the major endocannabinoids, increase food intake in rodents [107–109]. On the basis of the observation that cannabinoid receptor 1 (CB1) and endocannabinoids are present in the brain regions controlling food intake [110], the endogenous cannabinoid system has been proposed as a putative modulator of feeding behavior [111,112]. This concept has been further substantiated by the use of specific CB1 antagonists, which provided evidence for the role of CB1 in mediating the orexigenic effect of exogenous or endogenous cannabinoids [113]. Importantly, the levels of hypothalamic endocannabinoids were shown to be decreased after leptin administration, and the blockade of CB1 was demonstrated to inhibit starvation-induced hyperphagia in mice [114]. However, the phenotype of neurons in the hypothalamic appetite center that are directly affected by cannabinoids had not been elucidated. Cota et al. [115] have recently provided compelling evidence that endocannabinoids act at various regions of the hypothalamus that regulate energy homeostasis, including the lateral hypothalamus, ARC, and paraventricular nucleus, in conjunction with direct effects in fat cells on lipogenesis to promote a positive energy balance. If the receptor, CB1, is blocked (in this case by knockout of the gene), the overall effect will be to decrease appetite and lipogenesis in white fat: CB−/− animals are leaner because their caloric intake is lower when they are young. However, in older animals, increased peripheral energy expenditure appears to be the predominant defense against adiposity in these animals compared with their wild-type littermates [115]. Cota et al. have shown that CB1 is expressed in key hypothalamic peptidergic systems of appetite regulation, including those producing CRH in the paraventricular nucleus, CART in the ARC, and MCH and orexin in the lateral hypothalamus–perifornical region. Therefore, it is likely that by modulating food intake and mechanisms regulating energy expenditure, endocannabinoids or their antagonists will affect the signaling flow between these hypothalamic circuits and other neuronal networks with which these peptidergic systems interact. This central mechanism may dominate cannabinoid effects at younger ages but appears to diminish with age [115]. Whether it is chronological aging per se or other developmental processes, for example, puberty, that affects the function of relevant hypothalamic circuitry and thereby enables this impairment of cannabinoid signaling still needs to be clarified. In addition, it must be determined whether such a shift in endocannabinoid signaling during the aging process also occurs in hu-

The NPY Y2 receptor (Y2R), a putative inhibitory presynaptic receptor, is highly expressed on NPY neurons. As we seen, NPY is the most potent orexigenic hypothalamic petide, and NPY neurons are in the ARC, which is accessible to peripheral hormones. Peptide YY3–36 (PYY3–36 ), a Y2R agonist, is released from the gastrointestinal tract postprandially in proportion to the calorie content of a meal [100]. Batterham et al. [101] have recently shown that peripheral injection of PYY3–36 in rats inhibits food intake and reduces weight gain. PYY3–36 also inhibits food intake in mice but not in Y2r-null mice, which suggests that the anorectic effect requires the Y2R. Peripheral administration of PYY3–36 increases c-Fos immunoreactivity in the ARC and decreases hypothalamic NPY messenger RNA. Intra-arcuate injection of PYY3–36 inhibits food intake. PYY3–36 also inhibits electrical activity of NPY nerve terminals, thus activating adjacent POMC neurons [102]. In humans, infusion of normal postprandial concentrations of PYY3–36 significantly decreases appetite and reduces food intake by 33% over 24 h [101]. These results suggest that a gut–hypothalamic pathway that involves postprandial PYY3–36 acting at the arcuate Y2R has a role in regulating feeding. Thus, the PYY3–36 system may provide a therapeutic target for the treatment of obesity. Given, however, that the majority of obese subjects are resistant to the effects of leptin, the effects of PYY3–36 infusion on appetite and food intake were compared in obese and lean subjects. Batterham et al. [103] have found more recently that a single infusion of PYY3–36 , as compared with an infusion of saline, reduced appetite and food consumption by approximately 30% at an all-you-want-to-eat buffet lunch provided 2 h after the infusion. In the obese subjects, the endogenous postprandial PYY3–36 response was diminished as compared with that in the lean subjects, even though the obese subjects consumed a greater number of calories. The PYY3–36 infusion reduced hunger in both the obese and lean groups and had no effect on subjects’ reports of the palatability of the meals or their feelings of nausea. The study by Batterham et al. [103] also shows that infusion of PYY3–36 decreases fasting concentrations of the orexigenic peptide ghrelin. However, the extent to which suppression of ghrelin secretion contributes to a PYY3–36 -mediated reduction in food intake is unclear. Although single intraperitoneal injections of PYY3–36 decrease food intake for up to 7 days in rats, the results of a single infusion in humans cannot be extrapolated to predict long-term outcomes. The use of PYY3–36 may prevent counterregulatory mechanisms from overriding the stimulation of anorexigenic pathways. However, the development of antibodies or tachyphylaxis through recep-

4.9. Endocannabinoids and CB1 cannabinoid receptor

460

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

mans. Nevertheless, the observation that CB1 is present in adipocytes and that CB1 activation affects lipogenesis provides an alternative and/or complementary mechanism for the unidirectional effect of cannabinoids on energy balance. 4.10. FAS inhibitors An important step toward development of drugs to treat obesity was the serendipitous discovery that fatty acid synthase (FAS) inhibitors, originally developed to treat cancer, including cerulenin, produce profound weight loss and reduction in food intake [116]. Despite these promising results, mechanisms mediating the weight-reducing effects of FAS inhibitors remained to be elucidated. A particularly intriguing finding was that FAS inhibitors are more potent in obese than in lean mice. This observation suggested that FAS inhibitors may enhance the effects of factors such as insulin, glucose and fatty acids, which are elevated in obese individuals and act through hypothalamic mechanisms to reduce body weight. A common nutrition-sensing pathway through which these factors act might involve the stimulation of acetyl CoA carboxylase through a PI(3) kinase-dependent pathway [117], which appears to mediate some effects of leptin [118] and insulin [119]. In turn, increased acetyl CoA

carboxylase activity stimulated malonyl CoA production. Hypothalamic responses to glucose appeared to be mediated through a mechanism that would also lead to enhanced production of malonyl CoA [120]. The accumulation of hypothalamic fatty acids, which produce profound metabolic effects [121], was also enhanced by malonyl CoA because this inhibits the enzyme carnitine palmitoyltransferase-1 (CPT1), which is required for entry of long-chain fatty acids (LCFAs) into mitochondria, where they are oxidized. Although a single injection of C75 could influence NPY and POMC mRNA [122], the chronic effects of FAS inhibitors appeared to be independent of at least these known hypothalamic regulators of body weight, suggesting that as yet undiscovered hypothalamic neuropeptides or systems mediated these chronic effects. Recently, Obici et al. [123] have proposed that the mechanism of action of FAS inhibitors does not simply involve increasing concentrations of malonyl-CoA that subsequently inhibit the flux of LCFAs into the mitochondria. Instead, the critical component may be an intracellular increase in LCFAs themselves that then induces a still ill-defined sensing mechanism in hypothalamic neurons (Fig. 2). This is in agreement with their previous observations that hypothalamic infusions of fatty acids decrease food intake and glucose production. The authors

Fig. 2. Neurons in the hypothalamus (in particular, the arcuate nucleus) are primary targets of a number of key hormones and metabolic cues. (Enlargement) Proposed model for function of CPT1 in hypothalamic regulation of food intake. Anorectic drugs such as fatty acid synthase (FAS) inhibitors increase malonyl-CoA, which is derived from the carboxylation of acetyl-CoA by the enzyme acetyl-CoA carboxylase (ACC). Large amounts of malonyl-CoA inhibit CPT1-dependent oxidation of LCFA-CoA molecules. ICV administration of exogenous fatty acids (LCFAs) directly increases cellular LCFA-CoA. The resulting increase in intracellular LCFA-CoA concentration leads to inhibition of feeding behavior.

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

go on to show that increases in LFCAs not only decrease food intake, but also reduce glucose production by the liver. These results and other recent findings suggest that manipulation or dysregulation of hypothalamic pathways can induce hallmarks of type 2 diabetes: hyperinsulinemia, decreased glucose uptake and increased hepatic glucose production. How might LCFAs regulate cellular events? That remains to be determined, but recent studies have indicated that fatty acids act as membrane ligands of G-protein-coupled receptors [124]. Whatever the molecular mechanism, LCFAs may now be viewed as CNS signaling molecules, and FAS and CPT1 as potential pharmaceutical targets for the treatment of obesity and diabetes. Indeed, the potent orexigenic effects of cannabinoids and the potent anorectic effects of CB1 receptor antagonism may also be partly mediated through modulation of hypothalamic CPT1 activity and of LCFA-CoA. In fact, endocannabinoids stimulate CPT1 activity and fatty acid oxidation in cultured astrocytes independently of malonyl-CoA and through interaction with CB1 receptors [114]. Thus, central inhibition of fatty acid oxidation may represent an innovative approach to the prevention and treatment of obesity and type 2 diabetes mellitus. 4.11. Oleylethanolamide Oleylethanolamide (OEA) is a natural analogue of the endogenous cannabinoid anandamide. Like anandamide, OEA is produced in cells in a stimulus-dependent manner and is rapidly eliminated by enzymatic hydrolysis, suggesting a function in cellular signaling. However, OEA does not activate cannabinoid receptors and its biological functions remained unknown for long time. It was demonstrated that, in rats, food deprivation markedly reduced OEA biosynthesis in the small intestine [125]. Moreover, administration of OEA caused a potent and persistent decrease in food intake and gain in body mass. This anorexic effect was behaviorally selective and was associated with the discrete activation of brain regions (the paraventricular hypothalamic nucleus and the nucleus of the solitary tract) involved in the control of satiety. OEA did not affect food intake when injected into the brain ventricles, and its anorexic actions were prevented when peripheral sensory fibres are removed by treatment with capsaicin. These results suggested that OEA is a lipid mediator involved in the peripheral regulation of feeding. Indeed, in a more recent work the same authors have showed that OEA binds with high affinity to the peroxisomeproliferator-activated receptor-␣ (PPAR-␣), a nuclear receptor that regulates several aspects of lipid metabolism [126]. Administration of OEA produces satiety and reduces body weight gain in wild-type mice, but not in mice deficient in PPAR-␣. In the small intestine of wild-type but not PPAR-␣-null mice, OEA regulates the expression of several PPAR-␣ target genes: it initiates the transcription of proteins involved in lipid metabolism and represses inducible nitric oxide synthase (iNOS), an enzyme that may contribute to

461

feeding stimulation [127]. These results, which show that OEA induces satiety by activating PPAR-␣, identify an unexpected role for this nuclear receptor in regulating behavior, and raise possibilities for the treatment of eating disorders.

5. Interventions to increase energy expenditure Two types of adipose tissue are present in mammals: white adipose tissue (WAT) stores energy as triglycerides, and brown adipose tissue (BAT) burns energy for heat production (adaptive thermogenesis). In situations of energy deficit, such as fasting, lipolysis in WAT controls the supply of energy to the body through the release of fatty acids into the plasma. BAT is specialized in adaptive thermogenesis, the part of energy expenditure induced by cold exposure or diet. Fatty acid oxidation and heat production by brown adipose cells are due to intense metabolic activity because of the presence of a large number of mitochondria, and the expression of uncoupling protein 1 (UCP1) [128]. UCP1 allows the dissipation of the proton electrochemical gradient generated by the mitochondrial respiratory chain. Uncoupling between oxygen consumption and ATP synthesis promotes energy dissipation as heat. In neonatal mammals, hibernators and rodents, cold-induced thermogenesis in BAT contributes to the maintenance of body temperature. Fuel is provided as fatty acids from BAT and WAT lipolysis. Thus, two potential drug strategy interventions for the control of fat mass and obesity could be to increase BAT thermogenesis or to induce conversion from white to brown adipocytes. BAT is present throughout the lifespan in rodents but disappears soon after birth in large mammals. In humans, there are no BAT depots in adults and UCP1 mRNA is expressed at low levels in WAT [129]. BAT is not thought to contribute significantly to thermogenesis [130]. However, UCP1 is expressed in hibernomas and in perirenal WAT of adult patients with phaeochromocytoma and primary aldosteronism, revealing that UCP1 expression can be induced in rare tumors and endocrinological disorders [131]. Appearance of brown adipocytes is therefore possible in certain conditions in adults. The thermogenetic relevance of ␤3 -adrenergic receptors, which are selectively expressed in brown adipocytes, has been widely shown in laboratory animals and ␤3 -adrenergic receptor agonists have been proposed as anti-obesity drugs [132]. Recently, Bachman et al. [133] provide direct evidence that ␤-adrenergic receptors directly mediate the stimulatory activity of sympathetic nervous system (SNS) on diet-induced thermogenesis. These authors reveal that mice lacking all three ␤-adrenergic receptors (␤1 AR, ␤2 AR, and ␤3 AR) cannot increase thermogenesis and become massively obese during overfeeding. In contrast to these “␤-less” mice, wild-type mice are able to resist obesity during overfeeding by activating diet-induced thermogenesis. The ␤-less mice are also intolerant to cold exposure, suggesting that the SNS and ␤-adrenergic receptor signaling

462

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

pathways overlap in their control of heat production in response to both diet and cold. Interestingly, the BAT of ␤-less mice is unresponsive to physiological (cold exposure) and pharmacological (nonselective ␤-agonist) stimulation. The earlier discovery that noradrenaline activates thermogenesis in BAT primarily through ␤3 -adrenergic receptor signaling prompted the realization that the SNS-␤3 -adrenergic receptor-UCP-1 axis regulates thermogenesis in response to both diet and cold. Previous attempts to induce obesity in mice by selectively deleting genes encoding UCP-1, ␤3 -adrenergic receptor or other proteins of the SNS have ended in failure. In particular, the production of transgenic mice lacking ␤3 -adrenergic receptor or UCP-1 failed to induce obesity, although varying degrees of cold sensitivity and small increases in body fat (less than two-fold) were observed [134]. These discrepancies may reflect the influence of genetic background on phenotypic outcomes of transgenic manipulations, and the existence of compensatory mechanisms that enable the transgenic mice to stay lean. The importance of PPAR-␥ and its cofactors in the differentiation of brown and white fat cells has been substanti-

ated by several studies (Fig. 3). PPAR-␥ is associated with white fat cell differentiation and hypertrophy. Somewhat unexpectedly, a critical enhancer in rodent and human UCP1 genes contains a PPAR-␥-responsive element that mediates the stimulation induced by thiazolidinediones, a novel class of anti-diabetic drugs [131]. Because PPAR-␥ is expressed in both WAT and BAT, the basis for the specific activation of UCP1 gene transcription in BAT remained elusive until the identification of the PPAR-␥ coactivator 1␣ (PGC-1␣) [135]. PGC-1␣ is expressed at higher levels in BAT than in WAT and its expression is increased in response to cold exposure and ␤3 -adrenergic stimulation. PGC-1␣ coactivates the PPAR–RXR heterodimer to stimulate the UCP1 promoter. The cofactor also plays a crucial role in mitochondrial biogenesis and stimulates the expression of electron-transport chain genes through induction of nuclear respiratory factors 1 and 2 and cooperation with nuclear respiratory factor 1 [136]. Recently, two different experimental approaches have given new insights for understanding whether it is possible to induce a metabolic shift in white fat cells from lipid

Fig. 3. Molecular processes involved in fat cell differentiation. (Enlargement) Coordination of transcription of mitochondrial and nuclear genes encoding subunits of OXPHOS by different extracellular stimuli, such as hormone. In the nucleus, the hormone–receptor complex can interact with the hormone response elements (HREs) of OXPHOS genes, to directly activate them, and also with the HREs of transcription factor genes (NRF, PGC-1␣), to induce transcription factors which exert a positive effect on the OXPHOS genes. The effect of extracellular stimuli, such as hormones, on mitochondrial OXPHOS can be direct, by interaction of the hormone–receptor complex with mitochondrial HREs, or indirect, via induction of nuclear-encoded mitochondrial transcription factors. NR, nuclear receptor.

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

storage towards fatty acid utilization. Firstly, human subcutaneous white adipocytes have been transduced with an adenovirus expressing human PGC-1␣ [137,138]. Expression of the coactivator induced the expression of UCP1, respiratory chain proteins, fatty acid oxidation enzymes and other brown adipocyte markers. The coordinated regulation of gene expression suggested a potential increase in the capacity of fatty acid oxidation in white adipocytes. On the other hand, nitric oxide (NO) production, which is induced by noradrenergic stiimulation of BAT through ␤3 -adrenergic receptors, has been found to increase PGC-1␣ gene expression and, consequently, mitochondrial biogenesis in brown fat cells [139]. The relevance in vivo of these findings was strengthened by studying the BAT functions in wild-type and eNOS−/− mice before and after cold exposure. At both temperatures, histological analysis indicated that eNOS−/− BAT was functionally inactive, and mitochondrial biogenesis was impaired. When the authors looked at the control of biogenesis in the brain, liver and heart of the knockout mice, they found that deletion of eNOS was enough to reduce the number of mitochondria even in tissues that have a basal level of neuronal, and possibly inducible, NOS expression [139]. In eNOS−/− mice, oxygen consumption rates—an indicator of metabolic rate—were decreased, indicating that BAT-dependent thermogenesis might be impaired. In genetic models of obesity, defective energy expenditure is involved in increased food intake and body-weight gain; 8-week-old eNOS−/− mice showed similar food consumption but weighed more than wild-type mice. So, the increased body weight of eNOS−/− mice could be accounted for by higher feed efficiency (i.e. weight gain/food intake) caused by defective energy expenditure. These results suggest that pharmacological manipulation of both NO system and PGC-1␣ transcriptional machinaries could be of relevance for drug treatment of obesity. Two other recent transgenic experiments open up other potential avenues for modulating the brown-fat phenotype. Transgenic mice that overexpress the winged helix–forkhead transcription factor Foxc2 (forkhead box c2) specifically in adipose tissue have enlarged BAT depots, whereas their WAT pads are reduced in size and, on histological examination, have increased numbers of multilocular (as opposed to unilocular) adipocytes [140]. The expression of several genes that are associated with mitochondrial biogenesis and respiration, including UCP1 and PGC-1␣, was also upregulated in WAT. Of further interest, these mice were more insulin sensitive than their non-transgenic littermates, indicating that pharmacological manipulation could lead to a further antidiabetic action. Also subject to genetic manipulation was the eukaryotictranslation-initiation-factor-4E binding protein 1 (EIF4EBP1), which is highly expressed in WAT and reversibly represses cap-dependent translation by preventing the formation of the EIF4E complex [141]. Homozygous EIF4EBP1-deficient mice (Eif4ebp1−/− ) manifested smaller WAT pads than wild-type littermates. In addition,

463

the WAT depots had six-fold higher UCP1 expression and contained increased numbers of multilocular adipocytes that are more characteristic of a brown adipocyte phenotype. Moreover, although these mice showed an unaltered level of PGC-1␣ mRNA expression, they did manifest a two-fold higher PGC-1␣ protein level.

6. Thermogenic drugs 6.1. β3 -Adrenergic receptor agonists First-generation ␤3 -adrenergic receptor agonists were selected on the basis of their thermogenic and anti-obesity activities in rats and mice. In obese rodents, they produce good weight loss (or prevent weight gain) because they increase metabolic rate. There may be an initial reduction in food intake [142], but no effect on total intake is detectable over a few days or longer [143]. All the weight lost is fat, and when given with food, there even may be an increase in body protein [144]. In addition to their anti-obesity activity, ␤3 -adrenergic receptor agonists elicit marked improvements in insulin sensitivity in obese and insulin-resistant rodents. Such effects are seen at doses below those that elicit significant anti-obesity activity. This suggests that ␤3 -adrenergic receptor agonists (and perhaps other thermogenic drugs) have even greater potential as anti-diabetic than as anti-obesity agents [145,146]. An improvement in the blood lipid profile elicited by the ␤3 -adrenergic receptor agonist GR 265261 at a dose that does not have an anti-obesity effect has been described in monkeys [147]. The thermogenic, anti-obesity and anti-diabetic effects of ␤3 -adrenergic receptor agonists in rodents are achieved without significant cardiovascular effects, lowering of blood potassium or tremor, because they lack potency or efficacy at rodent ␤1 - and ␤2 -adrenoceptors. Unfortunately, clinical experience with the first-generation compounds has been less encouraging. Although some compounds have been shown to be thermogenic, to produce weight loss with conservation of protein, or to improve insulin action, efficacy generally has been associated with ␤1 - or ␤2 -adrenoceptor-mediated side effects. Only the American Home Products (Madison, NJ, USA) compound CL-316243, which has little or no efficacy at rodent ␤1 - and ␤2 -adrenoceptors, has shown any beneficial activity without eliciting side effects. Unfortunately, CL-316243 had only 10% oral bioavailability in humans, and it was given at a dose of 1500 mg. Prodrugs with improved oral bioavailability have been described recently [148]. Despite the failure of the first-generation compounds in humans, their effects on metabolic rate and insulin action suggest that the ␤3 -adrenoceptor is a valid target. It might be argued that the beneficial effects of BRL-26830, BRL-35135, and ZD-2079 were due to stimulation of ␤2 - or even ␤1 -adrenoceptors. Indeed, 60% of the thermogenic effects of BRL-35135 was blocked with the ␤1 /␤2 -adrenoceptor antagonist nadolol. Nevertheless,

464

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

the remaining activity does appear to be mediated by the ␤3 -adrenoceptor [149]. Thus, although they may never be as remarkably effective in humans as in rodents, the main problem with the first-generation compounds appears to be that they are not as selective for human as for rodent ␤3 -adrenergic receptors. The reason for this became transparent when the human ␤3 -adrenergic receptor was cloned, and it was found that the first-generation compounds had low efficacies and potencies at the human receptor compared with their efficacies and potencies in rodent tissues that express ␤3 -adrenergic receptor [150]. Since potencies and efficacies at ␤1 - and ␤2 -adrenergic receptor appear similar between humans and rodents, selectivities for ␤3 -adrenergic receptor are low in humans. A suggestion to ameliorate the experimental methodologies, which limit the screening approaches to thermogenetic molecules active in humans as anti-obesity drugs, seems to be implied in a recent paper by Palma et al. [151]. While injecting heterogenic mRNAs, isolated from brains and other tissues, into Xenopus oocytes to express functional neurotransmitter receptors in the oocyte membrane [152], which was used extensively in the last 20 years to study the molecular structure and function of many receptors and voltage-operated channels [153], a method was developed to incorporate in the oocyte membrane foreign receptors that had already been assembled in their native cells [151]. Essentially, this method consists in injecting the oocytes with cell membranes prepared from foreign tissues. This approach has been used to transplant neurotransmitter receptors expressed from cultured cells to the oocytes. Membrane vesicles prepared from a human embryonic kidney cell line (HEK293) stably expressing the rat glutamate receptor 1 were injected into oocytes, and, within a few hours, the oocyte plasma membrane acquired ␣-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptors, which had the same properties as those expressed in the original HEK cells. Analogously, oocytes injected with membranes prepared from rat pituitary GH(4)C1 cells, stably expressing homomeric human neuronal 7 nicotinic acetylcholine receptors (␣7-AcChoRs), incorporated in their plasma membrane AcChoRs that behaved as those expressed in GH(4)C1 cells. Similar results were obtained with HEK cells stably expressing heteromeric human neuronal ␣4␤2-AcChoRs. All this makes the Xenopus oocyte a powerful tool for detailed investigations of ␤3 -adrenergic receptors expressed in the membrane of cultured cells. Finally, new concepts about drug–receptor interaction may add relevant changes in screening procedures to obtain therapeutically effective molecules. It is important to underline that the intimate relationship between affinity and efficacy in drug–receptor interaction is becoming appreciated as new technologies allow the detection of different receptor behaviors. Indeed, efficacy for G-protein-coupled receptors (GPCRs) should not be confined to G-protein

activation, but should also be expanded to the complete range of behaviors of GPCRs, such as receptor internalization, desensitization, oligomerization, phosphorylation and association with other membrane proteins [154]. The idea that affinity is linked to receptor conformational changes compels different views of how GPCRs could be screened for biologically active molecules. This idea also suggests that optimal drug screening processes should use maximal numbers of methods to detect receptor conformational changes. For example, if only one G-protein response is monitored for a given receptor screen (i.e. cAMP production or lipolytic activity by putative ␤3 -adrenergic receptor agonists), then a ligand–receptor interaction that produces a conformational change in the receptor, with a consequent “effect”, but does not produce a specific change in the monitored receptor–G-protein interaction would not be detected. Thus ideally, an assay in which protein conformational changes can be detected directly would optimal for screening for potential therapeutically useful molecules [154]. 6.2. PPAR-δ agonists A recent paper shows that a ligand-activated transcription factor known as peroxisome-proliferator-activated receptor-␦ (PPAR-␦) stimulates fat burning, which indicates that drugs that activate PPAR-␦ might have potential for treating obesity [155]. Selective expression of an activated form of PPAR-␦ in adipose tissue resulted in mice with a lean phenotype, although their food intake on a standard diet was normal. And when these mice were fed a high-fat diet, weight gain and lipid accumulation were markedly reduced compared with control mice [155]. Further experiments with db/db mice, which are predisposed to obesity, showed that expression of activated PPAR-␦ reverses the obesity phenotype [155]. Moreover, age- and weight-matched db/db mice were treated with vehicle or a PPAR-␦-specific agonist, GW501516 [156], for 7 days. The histology of BAT in vehicle-treated db/db mice resembles WAT, each cell filled with a single massive lipid droplet, pushing the nucleus to the cell periphery. Remarkably, in GW501516 treated db/db mice, brown fat adipocytes show increased mitochondria-rich eosinophilic staining, disappearance of large lipid droplets, and replacement with multiple small droplets [155]. Thus, even a relatively short-term agonist treatment leads to a rapid restoration of brown fat appearance with reduced lipid content and enhanced metabolic activity. In addition, the expression of genes involved in fatty acid metabolism was monitored, and it was found that PPAR-␦ activation specifically induces the expression of genes required for fatty acid oxidation and energy dissipation. The ability to induce adaptive thermogenesis and protection against both dietary and genetic obesity suggests that PPAR-␦ agonists may be promising drug candidates for anti-obesity therapies.

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469

6.3. KB-141 KB-141 is a thyroid hormone receptor (TR) agonist that binds to the human (h)TR␤ with a 14-fold higher affinity than to hTR␣ [157]. Recently, KB-141 was used to examine effects of selective activation of TR␤ in control and TR␣−/− mice, cholesterol-fed rats, and cynomolgus monkeys. The latter more closely resemble humans in terms of lipoprotein metabolism and regulation of body weight [158]. 3,5,3 -Triiodi-l-thyronine (T3 ), the major active form of thyroid hormone, increased the metabolic rate in both wild type and TR␣−/− mice, but the increase for the wild type animals was greater than with the TR␣−/− mice [159]. These data imply that both TR␣ and -␤ regulate the metabolic rate. As compared with T3 , KB-141 reduced plasma cholesterol levels selectively versus increasing heart rate in all three models. Relative to T3 , KB-141 also increased whole body oxygen consumption in both mice and rats more than heart rate. In monkeys, KB-141 decreased body weight after 1 week of treatment by up to 7% and Lp(a) by up to 56% without tachycardia. These studies suggest that selective stimulation of the TR␤ might be exploited as a therapeutically effective means to lower weight, plasma cholesterol, and Lp(a) without eliciting deleterious cardiac effects.

7. Conclusions The understanding that obesity is a chronic multifactorial disease, which is poorly treated with the available therapeutic approaches (diet, exercise, behavior), has stimulated a renewed interest in the use of drugs. Over the past 30 years relatively few drugs have been developed, or approved, for the treatment of obesity. Worldwide government regulations and treatment guidelines have been somewhat instrumental in impeding the development of these drugs. However, other drugs that control chronic diseases, such as hypertension or diabetes mellitus, are not expected to increasingly reduce high blood pressure or high blood glucose levels, but rather to maintain a target level [160]. The current view regarding weight loss is that obese patients should not be expected to reach their “ideal” bodyweight, but instead should be encouraged to initially resist further weight gain and subsequently to lose moderate amounts of weight (5–10%) and to maintain this weight loss. At present, few options exist for the pharmacological treatment of obesity. Dexfenfluramine was the only other centrally acting anti-obesity agent recently approved for use longer than 3 months. However, it, along with fenfluramine, has been voluntarily withdrawn from the market worldwide and is undergoing further evaluation because of potential cardiovascular safety concerns. Sibutramine, like dexfenfluramine, is a centrally acting agent which affects satiety, although the mechanism of action of each agent is different. Both agents appear devoid of abuse potential. New drug development is occuring throughout the pharmaceutical industry and several therapeutic agents are

465

around the corner and still others are on the horizon. The advent of fully sequenced genomes and new genomics and proteomics technologies, which permit analyses of gene transcription profiles and protein interactions in complex cell circuitries, will lead to new target selection for drugs able to treat obesity and will enable obese individuals to be classified according to their likely response to an anti-obesity drug.

References [1] World Health Organization. Obesity: preventing and managing the global epidemic. Report of a WHO Consultation Technical Report Series 894, Geneva, Switzerland; 2000. [2] Deckelbaum RJ, Williams CL. Childhood obesity: the health issue. Obes Res 2001;9(Suppl 4):239S–43S. [3] U.S. Department of Health and Human Services. The Surgeon General’s call to action to prevent and decrease overweight and obesity 2001. U.S. Department of Health and Human Services, Public Health Service, Office of the Surgeon General, Rockville, MD; 2001. [4] Pi-Sunyer FX. Medical hazards of obesity. Ann Int Med 1993;119: 655–60. [5] Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 2003;348:1625–38. [6] Manson JE, Willett WC, Stampfer MJ, et al. Body weight and mortality among women. N Engl J Med 1995;333:677–85. [7] Haffner SM, Valdez RA, Hazuda HP, Mitchell BD, Morales PA, Stern MP. Prospective analysis of the insulin-resistance syndrome (syndrome X). Diabetes 1992;41:715–22. [8] Isomaa B, Almgren P, Tuomi T, et al. Cardiovascular morbidity and mortality associated with the metabolic syndrome. Diab Care 2001;24:683–9. [9] Trevisan M, Liu J, Bahsas FB, Menotti A. Syndrome X and mortality: a population-based study. Am J Epidemiol 1998;148:958–66. [10] National Institutes of Health. Third Report of the National Cholesterol Education Program Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). NIH Publication 01-3670. Bethesda, MD: National Institutes of Health; 2001. [11] Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults. JAMA 2002;287:356–9. [12] Williamson DF, Pamuk E, Thun M, et al. Prospective study of intentional weight loss and mortality in never-smoking US white women aged 40–64 years. Am J Epidemiol 1995;141:1128–41. [13] Gregg EW, Gerzoff RB, Thompson TJ, Williamson DF. Intentional weight loss and death in overweight and obese U.S. adults 35 years of age and older. Ann Intern Med 2003;138:383–9. [14] Hill JO, Peters JC. Environmental contributions to the obesity epidemic. Science 1998;280:1371–4. [15] Woods SC, Seeley RJ, Porte Jr D, Schwartz MW. Signals that regulate food intake and energy homeostasis. Science 1998;280:1378– 83. [16] Blackburn L, Miller D, Chan S. Pharmaceutical treatment of obesity. Nurs Clin North Am 1997;32:831–48. [17] Tuck ML, Sowers J, Dornfeld L, et al. The effect of weight reduction on blood pressure. N Engl J Med 1981;304:930–3. [18] Dattilo AM, Kris-Etherton PM. Effects of weight reduction on blood lipids and lipoproteins: a meta-analysis. Am J Clin Nutr 1992;56: 320–8. [19] UK prospective study on maturity onset diabetes. Effect of diet and sulphonylurea, insulin or biguainide therapy on fasting plasma

466

[20]

[21]

[22]

[23]

[24]

[25] [26]

[27] [28]

[29]

[30] [31] [32] [33]

[34]

[35]

[36] [37] [38] [39]

[40] [41]

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469 glucose and body weight over a year. A multicentre study. Diabetologia 1983;24:404–11. Bosello O, Armellini F, Zamboni M, Fitchet M. The benefits of modest weight loss in type 2 diabetes. Int J Obes Relat Metab Disord 1997;21(Suppl 1):S10–3. Tuomilehto J, Lindström J, Eriksson JG, et al. for the Finnish Diabetes Prevention Study Group. Prevention of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired glucose tolerance. N Engl J Med 2001;344:1343–50. U.S. Department of Health and Human Services. Diet and exercise dramatically delay type 2 diabetes: diabetes medication metformin also effective. National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases. Available at: http://www.niddk.nih.gov/welcome/release/8 9 01.htm. Accessed March 12, 2002. National Task Force on the Prevention and Treatment of Obesity. Long-term pharmacotherapy in the management of obesity. JAMA 1996;276:1907–15. National Heart, Lung, and Blood Institute Obesity Education Initiative. The practical guide identification, evaluation, and treatment of overweight and obesity in adults. U.S. Department of Health and Human Services, Public Health Service, National Institutes of Health, National Heart, Lung, and Blood Institute, Rockville, MD. NIH Publication Number 00-4084; October 2000. Manson JE, Faich GA. Pharmacotherapy for obesity—do the benefits outweigh the risks. N Engl J Med 1996;335:659–60. Royal College of Physicians of London. Clinical management of overweight and obese patients with particular reference to the use of drugs. A report of the Royal College of Physicians of London, London; December 1998. Silverstone T. Appetite suppressants. A review. Drugs 1992;43: 820–36. Kernan WN, Viscoli CM, Brass LM, et al. Phenylpropanolamine and the risk of hemorrhagic stroke. N Engl J Med 2000;343: 1826–32. Gardin JM, Schumacher D, Constantine G, et al. Valvular abnormalities and cardiovascular status following exposure to dexfenfluramine or phentermine/fenfluramine. JAMA 2000;283:1703–9. Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature 1998;395:763–70. Elmquist JK, et al. Unraveling the central nervous system pathways underlying responses to leptin. Nat Neurosci 1998;1:445–9. Tartaglia LA. The leptin receptor. J Biol Chem 1997;272:6093–6. Chua Jr SC, et al. Fine structure of the murine leptin receptor gene: splice site suppression is required to form two alternatively spliced transcripts. Genomics 1997;45:264–70. Ge H, et al. Generation of soluble leptin receptor by ectodomain shedding of membrane-spanning receptors in vitro and in vivo. J Biol Chem 2002;277:45898–903. El Haschimi K, et al. Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. J Clin Invest 2000;105:1827–32. Banks WA, et al. Leptin enters the brain by a saturable system independent of insulin. Peptides 1996;17:305–11. Elmquist JK, et al. From lesions to leptin: hypothalamic control of food intake and body weight. Neuron 1999;22:221–32. Elmquist JK, et al. Distributions of leptin receptor mRNA isoforms in the rat brain. J Comp Neurol 1998;395:535–47. Baskin DG, et al. Leptin receptor long-form splice-variant protein expression in neuron cell bodies of the brain and co-localization with neuropeptide Y mRNA in the arcuate nucleus. J Histochem Cytochem 1999;47:353–62. Schwartz MW, et al. Central nervous system control of food intake. Nature 2000;404:661–71. Cowley MA, et al. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 2001;411:480–4.

[42] Fan W, et al. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 1997;385:165–8. [43] Seeley RJ, et al. Melanocortin receptors in leptin effects. Nature 1997;390:349. [44] Erickson JC, et al. Attenuation of the obesity syndrome of ob/ob mice by the loss of neuropeptide Y. Science 1996;274:1704–7. [45] Bates SH, et al. STAT3 signaling is required for leptin regulation of energy balance but not reproduction. Nature 2003;421:856–9. [46] Montague CT, et al. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 1997;387:903–8. [47] Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD. A leptin missense mutation associated with hypogonadism and morbid obesity. Nature Genet 1998;18:213–5. [48] Zhang Y, et al. Positional cloning of the mouse obese gene and its human homologue. Nature 1994;372:425–32. [49] Farooqi IS, et al. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med 1999;341:879–84. [50] Halaas JL, et al. Weight-reducing effects of the plasma protein encoded by the obese gene. Science 1995;269:543–6. [51] Heymsfield SB, et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA 1999;282:1568–75. [52] Caro JF, et al. Decreased cerebrospinal-fluid/serum leptin ratio in obesity: a possible mechanism for leptin resistance. Lancet 1996;348:159–61. [53] Farooqi IS, et al. Metabolism: partial leptin deficiency and human adiposity. Nature 2001;414:34–5. [54] Sleeman MW, Anderson KD, Lambert PD, Yancopoulos GD, Wiegand SJ. The ciliary neurotrophic factor and its receptor, CNTFR. Pharm Acta Helv 2000;74:265–72. [55] Lambert PD, et al. Ciliary neurotrophic factor activates leptin-like pathways and reduces body fat, without cachexia or rebound weight gain, even in leptin-resistant obesity. Proc Natl Acad Sci USA 2001;98:4652–7. [56] Bruning JC, et al. Role of brain insulin receptor in control of body weight and reproduction. Science 2000;289:2122–5. [57] DeFronzo R. Pharmacological therapy for type 2 diabetes. Ann Intern Med 1999;17:281–303. [58] Air EL, Strowski MZ, Benoit SC, Conarello SL, Salituro GM, Guan XM, et al. Small molecule insulin mimetics reduce food intake and body weight and prevent development of obesity. Nat Med 2002;8(2):179–83. [59] Zhang B, et al. Discovery of a small molecule insulin mimetic with antidiabetic activity in mice. Science 1999;284:974–7. [60] Qureshi SA, et al. Activation of insulin signal transduction pathway and anti-diabetic activity of small molecule insulin receptor activators. J Biol Chem 2000;275:36590–5. [61] Sipols AJ, Baskin DG, Schwartz MW. Effect of intracerebroventricular insulin infusion on diabetic hyperphagia and hypothalamic neuropeptide gene expression. Diabetes 1995;44:147–51. [62] Cone RD. The central melanocortin system and energy homeostasis. Trends Endocrinol Metab 1999;10:211–6. [63] Mountjoy K, Mortrud M, Low M, Simerly R, Cone R. Localization of the melanocortin-4 receptor (MC4-R) in neuroendocrine and autonomic control circuits in the brain. Mol Endocrinol 1994;8:1298– 308. [64] Huszar D, et al. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 1997;88:131–41. [65] Vaisse C, Clement K, Guy-Grand B, Froguel P. A frameshift mutation in human MC4R is associated with a dominant form of obesity. Nature Genet 1998;20:113–4. [66] Branson R, Potoczna N, Kral JG, Lentes KU, Hoehe MR, Horber FF. Binge eating as a major phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 2003;348(12):1096–103. [67] Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T, O’Rahilly S. Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med 2003;348(12):1085–95.

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469 [68] Shutter J, et al. Hypothalamic expression of ART, a novel gene related to agouti, is up-regulated in obese and diabetic mutant mice. Genes Dev 1997;11:593–602. [69] Ollmann M, et al. Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 1997;278:135–8. [70] Hagan MM, Rushing PA, Pritchard LM, Schwartz MW, Strack AM, Van Der Ploeg LH, et al. Long-term orexigenic effects of AgRP-(83–132) involve mechanisms other than melanocortin receptor blockade. Am J Physiol Regul Integr Comp Physiol 2000;279(1):R47–52. [71] Obici S, Feng Z, Tan J, Liu L, Karkanias G, Rossetti L. Central melanocortin receptors regulate insulin action. J Clin Invest 2001;108(7):1079–85. [72] Stanley BG, Leibowitz SF. Neuropeptide Y: stimulation of feeding and drinking by injection into the paraventricular nucleus. Life Sci 1984;35:2635–42. [73] Akabayashi A, Wahlestedt C, Alexander JT, Leibowitz SF. Specific inhibition of endogenous neuropeptide Y synthesis in arcuate nucleus by antisense oligonucleotides suppresses feeding behavior and insulin secretion. Brain Res Mol Brain Res 1994;21:55–61. [74] Shibasaki T, Oda T, Imaki T, Ling N, Demura H. Injection of anti-neuropeptide Y-globulin into the hypothalamic paraventricular nucleus decreases food intake in rats. Brain Res 1993;601:313–6. [75] Kushi A, et al. Obesity and mild hyperinsulinemia found in neuropeptide Y Y1 receptor-deficient mice. Proc Natl Acad Sci USA 1998;95:15659–64. [76] Pedrazzini T, et al. Cardiovascular response, feeding behavior and locomotor activity in mice lacking the NPY Y1 receptor. Nature Med 1998;4:722–6. [77] Naveilhan P, et al. Normal feeding behavior, body weight and leptin response require the neuropeptide Y Y2 receptor. Nature Med 1999;5:1188–93. [78] Marsh DJ, Hollopeter G, Kafer KE, Palmiter RD. Role of the Y5 neuropeptide Y receptor in feeding and obesity. Nature Med 1998;4:718–21. [79] Kristensen P, et al. Hypothalamic CART is a new anorectic peptide regulated by leptin. Nature 1998;393:72–6. [80] Asnicar MA, et al. Absence of cocaine- and amphetamine-regulated transcript results in obesity in mice fed a high caloric diet. Endocrinology 2001;142:4394–400. [81] Aja S, Sahandy S, Ladenheim EE, Schwartz GJ, Moran TH. Intracerebroventricular CART peptide reduces food intake and alters motor behavior at a hindbrain site. Am J Physiol Regul Integr Comp Physiol 2001;281:R1862–7. [82] Abbott CR, et al. Evidence of an orexigenic role for cocaine- and amphetamine-regulated transcript after administration into discrete hypothalamic nuclei. Endocrinology 2001;142:3457–63. [83] Spina M, et al. Appetite-suppressing effects of urocortin, a CRF-related neuropeptide. Science 1996;273:1561–4. [84] Coste SC, et al. Abnormal adaptations to stress and impaired cardiovascular function in mice lacking corticotropin-releasing hormone receptor-2. Nature Genet 2000;24:403–9. [85] Bradbury MJ, McBurnie MI, Denton DA, Lee KF, Vale WW. Modulation of urocortin-induced hypophagia and weight loss by corticotropin-releasing factor receptor 1 deficiency in mice. Endocrinology 2000;141:2715–24. [86] Van Dijk G, Thiele TE. Glucagon-like peptide-1 (7–36) amide: a central regulator of satiety and interoceptive stress. Neuropeptides 1999;33:406–14. [87] Bray GA. Afferent signals regulating food intake. Proc Nutr Soc 2000;59:373–84. [88] Scrocchi LA, et al. Glucose intolerance but normal satiety in mice with a null mutation in the glucagon-like peptide 1 receptor gene. Nature Med 1996;2:1254–8. [89] Wallenius V, et al. Interleukin-6-deficient mice develop matureonset obesity. Nature Med 2002;8:75–9.

467

[90] Kernie SG, Liebl DJ, Parada LF. BDNF regulates eating behavior and locomotor activity in mice. EMBO J 2000;19:1290–300. [91] Langmead CJ, et al. Characterization of the binding of [(125)I]human prolactin releasing peptide (PrRP) to GPR10, a novel G protein coupled receptor. Br J Pharmacol 2000;131:683–8. [92] Howard AD, et al. Identification of receptors for neuromedin U and its role in feeding. Nature 2000;406:70–4. [93] Cohen MA, Ellis SM, Le Roux CW, Batterham RL, Park A, Patterson M, et al. Oxyntomodulin suppresses appetite and reduces food intake in humans. J Clin Endocrinol Metab 2003;88(10):4696–701. [94] Bittencourt JC, et al. The melanin-concentrating hormone system of the rat brain: an immuno- and hybridization histochemical characterization. J Comp Neurol 1992;319:218–45. [95] Qu D, et al. A role for melanin-concentrating hormone in the central regulation of feeding behaviour. Nature 1996;380:243–7. [96] Borosky B, Durkin MM, Ogozalek K, et al. Antidepressant, anxiolytic and anorectic effects of a melanin-concentrating hormone-1 receptor anatgonist. Nature Med 2002;8:825–30. [97] Kojima M, et al. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 1999;402:656–60. [98] Nakazato M, et al. A role for ghrelin in the central regulation of feeding. Nature 2001;409:194–218. [99] Tschop M, Smiley DL, Heiman ML. Ghrelin induces adiposity in rodents. Nature 2000;407:908–13. [100] Pedersen-Bjergaard U, et al. Influence of meal composition on postprandial peripheral plasma concentrations of vasoactive peptides in man. Scand J Clin Lab Invest 1996;56:497–503. [101] Batterham RL, Cowley MA, Small CJ, Herzog H, et al. Gut hormone PYY(3–36) physiologically inhibits food intake. Nature 2002;418:650–4. [102] Cowley MA, et al. Leptin activates the anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 2001;411:480–4. [103] Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, et al. Inhibition of food intake in obese subjects by peptide YY3–36. N Engl J Med 2003;349(10):941–8. [104] Abel EL. Cannabis: effects on hunger and thirst. Behav Biol 1975;15:255–81. [105] Williams CM, Rogers PJ, Kirkham TC. Hyperphagia in pre-fed rats following oral delta9-THC. Physiol Behav 1998;65:343–6. [106] Koch JE. Delta(9)-THC stimulates food intake in Lewis rats: effects on chow. Pharmacol Biochem Behav 2001;68:539–43. [107] Williams CM, Kirkham TC. Anandamide induces overeating: mediation by central cannabinoid (CB1) receptors. Psychopharmacology (Berlin) 1999;143:315–7. [108] Jamshidi N, Taylor DA. Anandamide administration into the ventromedial hypothalamus stimulates appetite in rats. Br J Pharmacol 2001;134:1151–4. [109] Hao S, Avraham Y, Mechoulam R, Berry EM. Low dose anandamide affects food intake, cognitive function, neurotransmitter and corticosterone levels in diet-restricted mice. Eur J Pharmacol 2000;392:147–56. [110] Howlett AC, et al. International Union of Pharmacology. XXVII. Classification of cannabinoid receptors. Pharmacol Rev 2002;54:161–202. [111] Mechoulam R, Fride E. A hunger for cannabinoids. Nature 2001;410:763–5. [112] Cota D, et al. The endogenous cannabinoid system as a modulator of food intake. Int J Obes Relat Metab Disord 2003;27:289–301. [113] Simiand J, Keane M, Keane PE, Soubrie P. SR 141716, a CB1 cannabinoid receptor antagonist, selectively reduces sweet food intake in marmoset. Behav Pharmacol 1998;9:179–81. [114] Di Marzo V, et al. Leptin-regulated endocannabinoids are involved in maintaining food intake. Nature 2001;410:822–5. [115] Cota D, Marsicano G, Tschop M, Grubler Y, Flachskamm C, Schubert M, et al. The endogenous cannabinoid system affects energy

468

[116]

[117]

[118] [119]

[120]

[121] [122]

[123]

[124]

[125]

[126]

[127]

[128]

[129]

[130]

[131]

[132]

[133]

[134]

[135] [136]

[137]

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469 balance via central orexigenic drive and peripheral lipogenesis. J Clin Invest 2003;112(3):423–31. Loftus TM, et al. Reduced food intake and body weight in mice treated with fatty acid synthase inhibitors. Science 2000;288:2379– 81. Moule SK, et al. Multiple signalling pathways involved in the stimulation of fatty acid and glycogen synthesis by insulin in rat epididymal fat cells. Biochem J 1995;311:595–601. Niswender KD, et al. Intracellular signalling. Key enzyme in leptin-induced anorexia. Nature 2001;413:794–5. Shepherd PR, Withers DJ, Siddle K. Phosphoinositide 3-kinase: the key switch mechanism in insulin signalling. Biochem J 1998;333:471–90. Yang XJ, Kow LM, Funabashi T, Mobbs CV. Hypothalamic glucose sensor: similarities to and differences from pancreatic-cell mechanisms. Diabetes 1999;48:1763–72. Obici S, et al. Central administration of oleic acid inhibits glucose production and food intake. Diabetes 2002;51:271–5. Shimokawa T, Kumar MV, Lane MD. Effect of a fatty acid synthase inhibitor on food intake and expression of hypothalamic neuropeptides. Proc Natl Acad Sci USA 2002;99:66–71. Obici S, Feng Z, Arduini A, Conti R, Rossetti L. Inhibition of hypothalamic carnitine palmitoyltransferase-1 decreases food intake and glucose production. Nature Med 2003;9(6):756–61. Briscoe CP, et al. The orphan G protein-coupled receptor GPR40 is activated by medium and long chain fatty acids. J Biol Chem 2003;278:11303–11. Rodriguez de Fonseca F, Navarro M, Gomez R, Escuredo L, Nava F, Fu J, et al. An anorexic lipid mediator regulated by feeding. Nature 2001;414(6860):209–12. Fu J, Gaetani S, Oveisi F, Lo Verme J, Serrano A, Rodriguez De Fonseca F, et al. Oleylethanolamide regulates feeding and body weight through activation of the nuclear receptor PPAR-␣. Nature 2003;425(6953):90–3. Sticker-Krongrad A, Beck B, Burlet C. Nitric oxide mediates hyperphagia of obese Zucker rats: relation to specific changes in the microstructure of feeding behavior. Life Sci 1996;58:PL9–PL15. Ricquier D, Bouillaud F. Mitochondrial uncoupling proteins: from mitochondria to the regulation of energy balance. J Physiol 2000;529:3–10. Oberkofler H, et al. Uncoupling protein gene: quantification of expression levels in adipose tissues of obese and non-obese humans. J Lipid Res 1997;38:2125–33. Astrup A, et al. Contribution of BAT and skeletal muscle to thermogenesis induced by ephedrine in man. Am J Physiol 1985;248: E507–15. Del Mar Gonzalez-Barroso M, et al. The human uncoupling protein-1 gene (UCP1): present status and perspectives in obesity research. Obes Rev 2000;1:61–72. Nisoli E, Tonello C, Carruba MO. ␤3 -Adrenergic receptors: really relevant targets for anti-obesity drugs? Curr Med Chem Central Nerv Syst Agents 2003;3(3):257–73. Bachman ES, Dhillon H, Zhang CY, Cinti S, Bianco AC, Kobilka BK, et al. betaAR signaling required for diet-induced thermogenesis and obesity resistance. Science 2002;297:843–5. Harper ME, Himms-Hagen J. Mitochondrial efficiency: lessons learned from transgenic mice. Biochim Biophys Acta 2001;1504: 159–72. Puigserver P, et al. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 1998;92:829–39. Wu Z, et al. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1␤. Cell 1999;98:115–24. Larrouy D, et al. Cloning and mRNA tissue distribution of human PPAR coactivator-1. Int J Obes Relat Metab Disord 1999;23: 1327–32.

[138] Tiraby C, et al. Acquirement of brown fat cell features by human white adipocytes. J Biol Chem 2003;278:33370–6. [139] Nisoli E, Clementi E, Paolucci C, Cozzi V, Tonello C, Sciorati C, et al. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide. Science 2003;299(5608):896–9. [140] Cederberg A, Gronning LM, Ahren B, Tasken K, Carlsson P, Enerback S. FOXC2 is a winged helix gene that counteracts obesity, hypertriglyceridemia, and diet-induced insulin resistance. Cell 2001;106:563–73. [141] Tsukiyama-Kohara K, Poulin F, Kohara M, DeMaria CT, Cheng A, Wu Z, et al. Adipose tissue reduction in mice lacking the translational inhibitor 4E-BP1. Nat Med 2001;7:1128– 32. [142] Tsujii S, Bray GA. A beta-3 adrenergic agonist (BRL-37,344) decreases food intake. Physiol Behav 1998;63:723–8. [143] Arch JRS, Ainsworth AT, Ellis RDM, Piercy V, Thody VE, Thurlby PL, et al. Treatment of obesity with thermogenic beta-adrenoceptor agonists: studies on BRL 26830A in rodents. Int J Obes 1984;8(Suppl 1):1–11. [144] Choo JJ, Horan MA, Little RA, Rothwell NJ. Effects of the beta 2-adrenoceptor agonist, clenbuterol, on muscle atrophy due to food deprivation in the rat. Metabolism 1990;39:647–50. [145] Cawthorne MA, Sennitt MV, Arch JRS, Smith SA. BRL 35135, a potent and selective atypical beta-adrenoceptor agonist. Am J Clin Nutr 1992;55(Suppl 1):S252–7. [146] Largis EE, Burns MG, Muenkel HA, Dolan JA, Claus TH. Drug Dev Res 1994;32:69. [147] Hansen BC. In: Lockwood D, Heffner TG, editors. Handbook of experimental pharmacology. Heidelberg: Springer-Verlag; 2000. p. 461–89. [148] Sum FW, Gilbert A, Venkatesan AM, Lim K, Wong V, O’Dell M, et al. Prodrugs of CL316243: a selective beta3-adrenergic receptor agonist for treating obesity and diabetes. Bioorg Med Chem Lett 1999;9:1921–6. [149] Wheeldon NM, McDevitt DG, McFarlane LC, Lipworth BJ. Beta-adrenoceptor subtypes mediating the metabolic effects of BRL 35135 in man. Clin Sci 1994;86:331–7. [150] Fisher MH, Amend AM, Bach TJ, Barker JM, Brady EJ, Candelore MR, et al. A selective human beta3 adrenergic receptor agonist increases metabolic rate in rhesus monkeys. J Clin Invest 1998;101:2387–93. [151] Palma E, Trettel F, Fucile S, Renzi M, Miledi R, Eusebi F. Microtransplantation of membranes from cultured cells to Xenopus oocytes: a method to study neurotransmitter receptors embedded in native lipids. Proc Natl Acad Sci USA 2003;100:2896– 900. [152] Barnard EA, Miledi R, Sumikawa K. Translation of exogenous messenger RNA coding for nicotinic acetylcholine receptors produces functional receptors in Xenopus oocytes. Proc R Soc London Ser B Biol Sci 1982;215:241–6. [153] Gundersen CB, Miledi R, Parker I. Messenger RNA from human brain induces drug- and voltage-operated channels in Xenopus oocytes. Nature 1984;308:421–4. [154] Kenakin T. Efficacy at G-protein-coupled receptors. Nat Rev Drug Discov 2002;1:103–10. [155] Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H, et al. Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell 2003;113(2):159–70. [156] Oliver Jr WR, Shenk JL, Snaith MR, Russell CS, Plunket KD, Bodkin NL, et al. A selective peroxisome proliferator-activated receptor delta agonist promotes reverse cholesterol transport. Proc Natl Acad Sci USA 2001;98(9):5306–11. [157] Ye L, Li YL, Mellstrom K, Mellin C, Bladh LG, Koehler K, et al. Thyroid receptor ligands. 1. Agonist ligands selective for the thyroid receptor beta1. J Med Chem 2003;46(9): 1580–8.

E. Nisoli, M.O. Carruba / Pharmacological Research 50 (2004) 453–469 [158] Iossa S, Liverini G, Barletta A. Relationship between the resting metabolic rate and hepatic metabolism in rats: effect of hyperthyroidism and fasting for 24 hours. J Endocrinol 1992;135(1):45–51. [159] Grover GJ, Mellstrom K, Ye L, Malm J, Li YL, Bladh LG, et al. Selective thyroid hormone receptor-beta activation: a strategy for reduction of weight, cholesterol, and lipoprotein (a) with reduced cardiovascular liability. Proc Natl Acad Sci USA 2003;100(17):10067– 72.

469

[160] Pi-Sunyer FX. Guidelines for the approval and use of drugs to treat obesity—a position paper for the study of obesity. Obes Res 1995;3:473–8. [161] Nisoli E, Carruba MO. A benefit-risk assessment of sibutramine in the management of obesity. Drug Saf 2003;26:1027–48. [162] Holst B, Cygankiewicz A, Jensen TH, Ankersen M, Schwartz TW. High constitutive signaling of the ghrelin receptor–identification of a potent inverse agonist. Mol Endocrinol 2003;17:2201–10.