<<

International Journal of (1999) 23, Suppl 1, 54±58 ß 1999 Stockton Press All rights reserved 0307±0565/99 $12.00 http://www.stockton-press.co.uk/ijo -4 : A novel signalling pathway involved in body weight regulation

SL Fisher1, KA Yagaloff1 and P Burn1*

1Department of Metabolic Diseases, Hoffmann LaRoche, Nutley, NJ 07110, USA

For many years, genetically obese mouse strains have provided models for human obesity. The Avy=-agouti mouse, one of the oldest obese mouse models, is characterized by maturity-onset obesity and diabetes as a result of ectopic expression of the secreted , agouti protein. Agouti protein is normally expressed in hair follicles to regulate pigmentation through antagonism of the melanocortin-1 receptor, but in-vitro studies have demonstrated that the hormone also has potent antagonist activity for the melanocortin-4 receptor (MC4-R). Subsequent develop- ment of the MC4-R knockout mouse model demonstrated that MC4-R plays a role in weight homeostasis as these mice recapitulated the metabolic defects of the agouti mouse. Further evidence for this hypothesis was obtained from pharmacological studies utilizing with MC4-R activity, that inhibitied food intake (when administered intracerebrally). Additional studies with antagonists have now implicated the MC4-R in the signalling pathway. Finally, evidence that the MC4-R may play a role in human obesity has been obtained from the identi®cation of a dis-functional variant of the receptor in genetically obese subjects.

Keywords: obesity; diabetes; agouti; melanocortin; POMC; leptin; ob

Introduction There has been an explosion in obesity research and with this has come an understanding of the molecular mechanisms that underly the disease. This explosion Obesity is reaching epidemic status in the developed was sparked by the cloning of the ob from the world and is rapidly threatening the near-developed obese mouse model ob=ob5 and the subsequent regions of the globe. The clinically obese comprise demonstration that the protein product is a centrally nearly 30% of the total population in the US alone1 active hormone.6 These initial observations have led and it is estimated over $60 billion are spent annually to an increasingly complex view of the factors and in treatments for this disease,2 which is characterized regulatory processes that govern weight homeostasis by increased storage of energy input as in which the OB protein (or leptin) plays a central role as a consequence of unbalanced energy intake and coordinating a number of metabolic and reproductive energy expenditure. Obese patients suffer from an functions.7±10 Use of mouse genetics as a predictor increased risk of a number of severe and debilitating and model of human disease has provided the frame- diseases including non- dependent diabetes work for the identi®cation of novel drug discovery mellitus (NIDDM), hypertension, hyperlipidemia, targets. Our goal in this report is to describe the sleep apnea, and certain cancers.3 While previous current understanding of the weight regulation pro- generations regarded obesity as a social disease that cesses underlying another genetically obese mouse could be remedied with appropriate diet and exercise model, the agouti viable yellow Avy=-mouse. regimens, it is now clear that human obesity is a multifactorial disease with both environmental and genetic components. Studies in human populations4 suggest that at least 35% (and potentially as high as 50%) of the obese population is genetically predis- Agouti mouse model posed to become overweight. This segment of the population is most likely to bene®t from therapeutic vy intervention and it is hoped that with proper diet and The A =-mouse is the oldest known mouse model of obesity and was initially prized by mouse fanciers for exercise, therapeutic treatment will allow for long- 11 term weight reduction and weight maintenance. its golden yellow coat color (Figure 1). In addition to the change in coat color, these mice display an age- onset (10 ± 20 weeks) metabolic defect characterized by hyperphagia, increased adiposity, hyperglycemia, hyperinsulinemia and increased somatic growth. *Correspondence: Paul Burn, Department of Metabolic Dieases, Unlike other mouse models, the increased adipocyte Hoffmann LaRoche, Nutley, NJ 07110, USA; 12 Stewart Fisher, Biochemistry Dept, Astra Research Center, mass results from adipocyte hypertropy Ðan attri- Boston, Cambridge MA 02139, USA. bute that, along with the age-onset of development, Melanocortin-4 receptor SL Fisher et al 55 puri®ed protein product con®rmed this assumption. The agouti protein was found to be a potent ( < 2 nM) antagonist for the MC1-R,16 but interestingly was also found to be a potent anatagonist of the MC4-R. Agouti does not bind to MC2, MC3 or MC 5 receptors. The activity at the MC4-R was puzzling due to the selective tissue expression patterns of agouti and the MC4-R, but careful analysis of the Avy=-mouse iden- ti®ed a in the promoter region of the agouti gene that results in high-level, ectopic expression of the gene.17 This suggested that the characteristic phenotypes of the Avy=-mouse arise from separable antagonist activities at the MC1-R and MC4-R as shown in Figure 2; the yellow coat color from MC1- Figure 1 Agouti mice (right) display a yellow coat color and age- R antagonism and the observed metabolic defects onset obesity compared to wild-type mice (left). from MC4-R antagonism. This hypothesis was the foundation for the development of an MC4-R knock- closely resembles the characteristics most commonly out mouse model to directly assess the role of MC4-R observed in human obesity. in the regulation of weight homeostasis. The underlying cause of the Avy=-mouse metabolic defect remained an enigma until the early 1990s. The yellow coat color strongly suggested that the pigmen- tation regulatory pathways governed by the melano- Studies of the MC4-R cortin-1 receptor (MC1-R) were affected. Primarily expressed in the skin, the MC1-R is a G-protein coupled receptor which regulates pigmentation in The phenotype of the MC4-R knockout mouse pro- response to the stimulating hormone (a- vided con®rmation of the model depicted in Figure 2. 13 These mice completely recapitulated the metabolic MSH), a potent agonist of MC1-R. Agonism of the vy 18 MC1-R results in eumelanin production (and therefore defect of the A =-mice. The MC4-R knockout mice black pigmentation) whereas antagonism produces have a normal coat color, but develop age-onset phaeomelanin (resulting in bleaching of the skin obesity (12 ± 20 weeks) that results from hyperphagia color). There are ®ve known melanocortin receptors and adipocyte hypertrophy. In addition, the mice based on that ranges from 35 ± become hyperglycemic and hyperinsulinemic in a 60% homology between family members.13 The similar age-dependent manner. Furthermore, these MC2-R, also known as the adrenocorticotropin hor- effects are attenuated approx 2-fold in the hetero- mone (ACTH) receptor, is expressed in the adrenal zygote mice ± a ®nding consistent with the expected 2- cortex and regulates corticosteroid synthesis in fold increase in receptor density relative to the knock- response to ACTH levels. The MC3-R and MC4-R out mice. The mice have normal corticosteriod con- are both expressed in the central nervous system centrations, demonstrating an intact hypothalamic ± (CNS), and little was known about the function of pituitary ± adrenal (HPA) axis, but also show an these receptors when the agouti gene was cloned. The increase in the somatic growth as observed in the MC5-R is expressed in speci®c exocrine glands agouti mice. The extent of hyperglycemia and hyper- throughout the body (for example, Harderian and insulinemia observed in the knockout mice is gender perputial glands) and has been implicated in the dependent, with male offspring exhibiting more control of lipid and production in these severe elevation than females in both cases. The sites.14 Melanocyte stimulating are pro- overall phenotype clearly demostrates the separate duced from pro-opiomelanocortin (POMC), a pro- activities of the agouti protein on the melanocortin hormone of 131 amino acids that is processed into three classes of hormones; the (a, b, and g), ACTH and various (for example, ).13 POMC is produced primarily in the pituitary, but also in the arcuate nucleus of the . The cloning of the agouti gene in 1992 represented a breakthrough for understanding the Avy=-mouse.15 The agouti gene codes for small (131 residues), soluble, secreted protein, that is normally expressed only in the skin, consistent with the premise that this gene plays a role in coat color regulation through Figure 2 Agouti protein antagonism of MC1-R and MC4-R leads MC1-R interactions. Pharmacological studies on the to yellow pigmentation and obesity. Melanocortin-4 receptor SL Fisher et al 56 receptors in vivo (Figure 2) and implicates the MC4-R concentrations of POMC expression were observed in as a regulator of energy balance. fasted mice (low leptin concentrations), but POMC Additional proof of the concept for this model expression was elevated in response to rising leptin (Figure 2) was provided by studies that relied on concentrations after feeding.24,26 Acute measurements two cyclic heptapeptide analogs of aMSH, MT-II of POMC levels in Long ± Evans rats were found to be and SHU-9119, which display potent ( < 1 nM) directly in¯uenced by the administration of leptin; MC4-R agonist and antagonist activities, respec- after leptin administration, the POMC concentrations tively.19 The authors found that injection of MT-II rose by nearly 39%.26 In addition, repeated dosing of into the third ventricle of the brain caused a dose- ob mice with leptin (single dose 5 d) showed elevated dependent inhibition of food intake that was long POMC concentrations, when compared to control lasting ( > 8 h) and reversible when co-administered mice.25,26 Similar changes were not observed in the with the antagonist SHU-9119. Intraperitoneal admin- db=db mouse, demonstrating the requirement for an istration of higher doses of MT-II had similar effects intact ob-signalling pathway (in this case functional on food intake, clearly demonstrating that these mole- ob-R) for POMC modulation. cules, although relatively large (MW > 1300), were Additional evidence for melanocortin effects on capable of crossing the blood ± brain barrier in con- leptin signalling was obtained from studies using the centrations high enough to result in ef®cacy. To sum antagonist, SHU-9119, in Long-Evans rats.27 The up, these studies provided the ®rst demonstration that authors ®rst identi®ed a dose that was suf®cient to MC4-R can inhibit food intake and provide a interact with the receptor, but below the threshold foundation for the development of small molecule required for modulation of food intake when adminis- anti-obesity agents. tered by intracerebral injection into the thrid ventricle (iv3t, dose ˆ 0.5 nmol). Under these conditions, both the control animals and those treated with the antago- nist demonstrate normal feeding behavior after injec- Evidence for MC4-R involvement in tion, while those treated with leptin (3.5 mg) show a leptin signalling signi®cant reduction in food intake after 4 h ( 7 56%). Interestingly, animals subjected to co-administration of the antagonist with leptin exhibit normal feeding There were several lines of evidence suggesting the behavior, clearly showing a pharmacological blockade MC4-R played a role in the control of metabolic of leptin signalling by the antagonist. Similar results pathways regulated by leptin. First, agouti mice were obtained upon assessment of cFos activation in were highly resistant to leptin effects on food intake the paraventricular nucleus (PVN) ± a region known to inhibition.20 Second, functional (long-form) ob-R be activated by leptin and the MC4-R receptor. As receptors were found in the neurons of the arcuate expected from previous work,22 animals treated with nucleus that produce POMC, providing a basis for the leptin exhibited a signi®cant activation of cFos nuclei hypothesis that activation of the leptin signalling when the mice were pre-treated with vehicle (254% pathway in these neurons could result in elevated increase), but this effect was completely abolished POMC production and processing to yield MC4-R when the mice were pretreated with SHU-9119. These agonists.21 In addition, comparative analysis (using studies indicate that the MC4-R is downstream of ob- cFos immunohistochemistry analysis of brain sections R in the leptin pathway; consistent with the working of mice treated with leptin or MC4-R agonist) demon- model shown in Figure 3. This simple model has strated that overlapping activation patterns were support from known mouse models of obesity. At observed in a number of sites in the brain, for each of the steps in the signalling cascade, there is a example, paraventricular nucleus (PVN), dorsomedial hypothalamus (DMH) and the central nucleus of the Amygdala (CeA).22; 23 Furthermore, the long duration of action observed in food intake inhibition upon administration of MC4-R agonists, was consistent with similar results observed with leptin dosing ± in both cases, the effects extend well beyond the mea- sured half-life of the administered agent. These obser- vations, while largely circumstantial, led to detailed mechanistic studies of leptin and MC4-R signalling activation. The ®rst direct evidence that leptin in¯uenced the melanocortin signalling pathway was obtained from in-situ analysis of POMC neurons in the arcuate nucleus of a number of mouse and rat models.24 ± 26 Expression of POMC was found to be proportional to Figure 3 Proposed model for MC4-R in the leptin signaling circulating leptin concentrations in the serum; low pathway. Melanocortin-4 receptor SL Fisher et al 57 known genetic mutation that would inhibit the path- activities at other melanocortin receptors.33 In addi- way and result in the expected obesity phenotype. tion, there is evidence to suggest that there are addi- Whilte this model is appealing and correlates well tional melanocortin receptors that remain to be to the data mentioned above, there is a report that identi®ed.32 As a result, con®rmation of additional challenges its underlying tenets. A genetic cross of the MC4-R speci®c regulatory pathways will await the Ay=a and the ob=ob mice was prepared and the generation of more speci®c compounds and the poten- phenotype of these mice was examined for leptin tial discovery of new melanocortin receptors. sensitivity.28 Unexpectedly, these mice exhibited a near normal response to leptin administration, when measured as a function of body weight reduction, demonstrating that the leptin signalling pathway Evidence for melanocortin remained intact in these animals despite the expected regulation of human obesity antagonism of the MC4-R by the agouti protein. In addition, deletion of the leptin gene in agouti mice restored the senstivity to leptin administration, sug- While the data supporting a role for MC4-R in weight gesting that the development of obesity in the Ay=a regulation in rodent models of weight homeostasis is animals is achieved through a pathway that is inde- quite strong, there is growing evidence that the pendent of leptin signalling. Resolution of the appar- melanocortins also play a similar role in human ent con¯ict between the conclusions of this paper and obesity. A quantitative trait (QTL) associated with obesity in a human population was recently the data supporting the model in Figure 3 awaits 34 further investigation, but it should be noted that the identi®ed near the POMC gene locus, suggesting two studies differ considerably in their design (acute that in POMC production=processing may administration vs chronic antagonism by agouti) and play a role in human obesity. Furthermore, a novel in their measurement of leptin ef®cacy (food intake agouti-related transcript (ART, also known as agouti- inhibition and cFos activation vs body weight reduc- related protein or AGRP) has been identi®ed in both tion). One possibility is that the amount of endogen- rodents and man. This protein is produced in the ous agouti protein present in the hypothalamus is not hypothalamus and has potent antagonist activity for both MC3-R and MC4-R, despite sharing consider- able to inhibit ob signalling through the MC4-R 35,36 receptor. In the end, the differing results from the able sequence homology to agouti protein. This two studies may represent the likely possibility that homology, along with the expression pattern and the MC4-R and the leptin signalling pathways, while pharmacological pro®le, suggests that AGRP plays a clearly connected under some conditions, may not be role in allowing speci®c melanocortin subtype (MC3- completely overlapping under all situations. R and MC4-R) regulation similar to that observed with the agouti protein and MC1-R in pigmentaion control. Consistent with this hypothesis, expression levels of ART have recently been shown to be Other melanocortin effects modulated by leptin in an inverse relationship to POMC.37 The current model therefore suggests that attributed to MC4-R regulation the MC4-R is speci®cally switched between activation and inactivation by the action of two secreted hor- Recent studies have suggested that the MC4-R may be mones, MSH and AGRP. involved in the control of additional physiological processes such as grooming behaviour29,30 and blood 31 pressure regulation. The conclusions from these Conclusion studies were bases on selective administration of various MSH analogs (a, b, and g) and the antagonist (SHU-9119) into regions of the brain known to The rodent data supporting the role of MC4-R regula- express MC4-R. For example, administration of a- tion in weight homeostasis is compelling and the MSH into the dorsal motor nucleus of the vagus nerve recent data supporting a similar function in humans (a site where MC4-R is expressed32) results in lower will no doubt be expanded in the near future. This blood pressure and bradycardia. This effect is blocked strong proof-of-concept, along with the rich history of by administration of SHU-9119. Additional side therapeutic agents derived from G-protein coupled effects have been noted at high doses of both MT-II receptors, augurs well for the development of novel and SHU-9119, which includes barrel-rolling in rats therapeutic anti-obesity agents targeted to the MC4-R. and the development of conditioned-taste aversions.23 These side effects are inconsistent with agonism of the MC4-R. It should be noted that although these com- References pounds were originally reported as speci®c for the 1 Parker S. Obesity: Trends and Treatments. In: Scrip Reports. 19 PJB Publications Surrey 1996, pp 6 ± 15. MC4-R, subsequent pharmacological studies have 2 Jung RT. Obesity as a disease. Br Med Bull 1997; 53: shown that both MT-II and SHU-9119 have potent 307 ± 321. Melanocortin-4 receptor SL Fisher et al 58 3 Pi-Sunyer FX. Medical hazards of obesity. Ann Intern Med 22 Van Dijk G, Thiele TE, Donahey JCK, Camp®eld LA, Smith 1993; 119: 655 ± 660. FJ, Burn P, Bernstein IL, Woods SC, Seeley RJ. Central 4 Bouchard C, Perusse L. . Annu Rev Nutr infusions of leptin and GLP-1-(7-36) amide differentially 1993; 13: 337 ± 354. stimulate c-FLI in the rat brain. Am J Phys Reg Int Comp 5 Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Phys 1996; 40: R1096 ± R1100. Freidman JM. Positional cloning of the mouse obese gene 23 Thiele TE, van Dijk G, Yagaloff KA, Fisher Sl, Schwartz and its human homologue. Nature 1994; 372: 425 ± 431. MW, Burn P, Seeley RJ. Central infusion of melanocortin 6 Camp®eld LA, Smith FJ, Guisez Y, Devos R, Burn P. agonist MT-II in rats ± assessment of c-Fos expression and Recombinant mouse OB protein: evidence for a peripheral taste aversion. Am J Phys Reg Int Comp Phys 1998; 43: signal linking adiposity and central neural networks. Science R248 ± R254. 1995; 269: 546 ± 549. 24 Mizuno Tm, Kleopoulos Sp, Bergen HT, Roberts JL, Preist 7 Camp®eld LA, Smith FJ, Burn P. OB Protein: a hormonal CA, Mobbs CV. Hypothalamic pro-opiomelanocortin mRNA controller of central neural networks mediating behavioral, is reduced by fasting in ob=ob and db=db mice, but stimulated metabolic, and neuroendocrine responses. Endocrin Metab by leptin. Diabetes 1998; 47: 294 ± 297. 1997; 4: 81 ± 102. 25 Thornton JE, Cheung CC, Clifton DK, Steiner RA. Regulation 8 Camp®eld LA, Smith FJ, Burn P. The OB protein (leptin) of hypothalamic mRNA by leptin in pathway ± a link between adipose tissue mass and central ob=ob mice. Endocrinology 1997; 138: 5063 ± 5066. neural networks. Horm Metab Res 1996; 28: 619 ± 632. 26 Schwartz MW, Seeley RJ, Woods SC, Weigle DS, Camp®eld 9 Camp®eld LA, Smith FJ, Burn P. Strategies and potential LA, Burn P, Baskin DG. Leptin increases hypothalamic molecular targets for obesity treatment. Science 1998; 280: proopiomelanocortin mRNA expression in the rostral arcuate 1383 ± 1387. nucleus. Diabetes 1997; 46: 2119 ± 2123. 10 Woods SC, Seeley RJ, Porte D, Schwartz MW. Signals that 27 Seeley RJ, Yagaloff KA, Fisher SL, Burn P, Thiele TE, van regulate food intake and energy homeostasis. Science 1998; Dijk G, Baskin DG, Schwartz MW. Melanocorin receptors in 280: 1378 ± 1382. leptin effects. Nature 1997; 390: 349. 11 Barsh GS. The genetics of pigmentation: from fancy to 28 Boston BA, Blaydon KM, Varnerin J, Cone RD. Independent complex traits. Trends Gene 1996; 12: 299 ± 305. and additive effects of central POMC and leptin pathways on 12 Johnson PR, Hirsch J. Cellularity of adipose depots in six murine obesity. Science 1997; 278: 1641 ± 1644. strains of genetically obese mice. J Lipid Res 1972; 13: 2 ± 11. 29 Gipsen WH, Wiegant VM. Opiate antagonists suppress ACTH 13 Cone RD, Lu DS, Koppula S, Vage DI, Klungland H, Boston B, (1-24) induced excessive grooming in the rat. Neurosci Let Chen WB, Orth DN, Pouton C, Kesterson RA. The melanocor- 1976; 2: 159 ± 164. tin receptors ± agonists, antagonists and the hormonal- 30 Hirsch MD, O'Donohue TL, WIlson R, Sawyer TK, Hruby control of pigmentation. Rec Prog Horm Res 1996; 51: VJ, Hadley ME, Vody WL, Knittel JJ, Crawley JN. Structural 287 ± 318. and conformational modi®cations of aMSH=ACTH(1-10) pro- 14 Chen WB, Kelly MA, Opitzaraya X, Thomas RE, Low MJ, vide melanotropin analogues with high potent behavioral Cone RD. Exocrine gland dysfunction in MC5-R-de®cient activities. Peptides 1984; 5: 1197 ± 1201. mice ± Evidence for coordinated regulation of exocrine 31 Li S-J, Varga K, Archer P, Hruby VJ, Sharma SD, Kesterson gland-function by melanocortin peptides. 1997; 91: RA, Cone RD, Kunos G. Melanocortin antagonists de®ne two 789 ± 798. distinct pathways of cardiovascular control by a- and 16: 15 Bultman SJ, Michaud EJ, Woychik RP. Molecular character- 5182 ± 5188. ization of the mouse agouti locus. Cell 1992; 71: 1195 ± 1204. 32 Adan RAH, Gispen WH. Brain melanocortin receptors: from 16 Lu DS, Willard D, Patel IR, Kadwell S, Overton L, Lost T, cloning to function. Peptides 1997; 18: 1279 ± 1287. Luther M, Chen W, Woychik RP, Wilkinson WO, Cone RD. 33 Schioth HB, Muceniece R, Mutulis F, Prusis P, Lindeberg G, Agouti protein is an antagonist of the melanocyte-stimulating Sharma SD, Hruby VJ, Wikberg JES. Selectivity of cyclic [D- hormone receptor. Nature 1994; 371: 799 ± 802. Nal(7)] and [D-Phe-(7)] substituted MSH analogs for 17 Yen T, Gill AM, Frigeri LG, Barsh GS, Wolff GL. Obesity, the subtypes. Peptides 1997; 18: diabetes and neoplasia in yellow Avy=-mice: ectopic expres- 1009 ± 1013. sion of the agouti gene. FASEB J 1994; 8: 479 ± 488. 34 Comuzzie AG, Hixson JE, Almasy L, Mitchell BD, Mahaney 18 Huszar D, Lynch CA, Fairchildhuntress V, Dunmore JH, Fang MC, Dyer TD, Stern MP, MacCluer JW, Blangero J. A major Q, Berkemeier LR, Gu W, Kesterson RA, Boston BA, Cone quantitative trait locus determining serum leptin lelels and fat RD, Smith FJ, Camp®eld LA, Burn P, Lee F. Targeted mass is located on human 2. Nat Gene 1997; 15: disruption of the melanocorin-4 receptor results in obesity in 273 ± 276. mice. Cell 1997; 88: 131 ± 141. 35 Graham M, Shutter JR, Sarmiento U, Sarosi I, Stark KL. 19 Fan W, Boston BA, Kesterson RA, Hruby VJ, Cone RD. Role Overexpression of Agrt leads to obesity in transgenic mice. of melanocortinergic neurons in feeding and the agouti obesity Nat Gene 1997; 17: 273 ± 274. syndrome. Nature 1997; 385: 165 ± 168. 36 Ollmann MM, Wilson BD, Yang Y-K, Kerns JA, Chen Y, 20 Halaas JL, Boozer C, Blair-West J, Fidahusein N, Denton DA, Gantz I, Barsh GS. Antagonistm of central melanocortin Friedman JM. Physiological response to long-term peripheral receptors in vitro and in vivo by agouti-related protein. Science and central leptin infusion in lean and obese mice. Proc Natl 1997; 278: 135 ± 138. Acad Sci USA 1997; 94: 8878 ± 8883. 37 Shutter JR, Graham M, Kinsey AC, Scully S, Luthy R, Stark 21 Cheung CC, Clifton DK, Steiner RA. Proopiomelanocortin KL. Hypothalamic expression of ART, a novel gene related to neurons are direct targets for leptin in the hypothalamus. agouti, is up-regulated in obese and diabetic mutant mice. Endocrinology 1997; 138: 4489 ± 4492. Genes Dev 1997; 11: 593 ± 602.