201
|
Ellacott KL, Cone RD. The role of the central melanocortin system in the regulation of food intake and energy homeostasis: lessons from mouse models. Philos Trans R Soc Lond B Biol Sci 2007; 361:1265-74. [PMID: 16815803 PMCID: PMC1642695 DOI: 10.1098/rstb.2006.1861] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A little more than a decade ago, the molecular basis of the lipostat was largely unknown. At that time, many laboratories were at work attempting to clone the genes encoding the obesity, diabetes, fatty, tubby and agouti loci, with the hope that identification of these obesity genes would help shed light on the process of energy homeostasis, appetite and energy expenditure. Characterization of obesity and diabetes elucidated the nature of the adipostatic hormone leptin and its receptor, respectively, while cloning of the agouti gene eventually led to the identification and characterization of one of the key neural systems upon which leptin acts to regulate intake and expenditure. In this review, we describe the neural circuitry known as the central melanocortin system and discuss the current understanding of its role in feeding and other processes involved in energy homeostasis.
Collapse
|
202
|
Abstract
The rising tide of obesity is one of the most pressing health issues of our time, yet existing medicines to combat the problem are disappointingly limited in number and effectiveness. Fortunately, a recent burgeoning of mechanistic insights into the neuroendocrine regulation of body weight provides an expanding list of molecular targets for novel, rationally designed antiobesity pharmaceuticals. In this review, we articulate a set of conceptual principles that we feel could help prioritize among these molecules in the development of obesity therapeutics, based on an understanding of energy homeostasis. We focus primarily on central targets, highlighting selected strategies to stimulate endogenous catabolic signals or inhibit anabolic signals. Examples of the former approach include methods to enhance central leptin signaling through intranasal leptin delivery, use of superpotent leptin-receptor agonists, and mechanisms to increase leptin sensitivity by manipulating SOCS-3, PTP-1B, ciliary neurotrophic factor, or simply by first losing weight with traditional interventions. Techniques to augment signaling by neurochemical mediators of leptin action that lie downstream of at least some levels of obesity-associated leptin resistance include activation of melanocortin receptors or 5-HT2C and 5-HT1B receptors. We also describe strategies to inhibit anabolic molecules, such as neuropeptide Y, melanin-concentrating hormone, ghrelin, and endocannabinoids. Modulation of gastrointestinal satiation and hunger signals is discussed as well. As scientists continue to provide fundamental insights into the mechanisms governing body weight, the future looks bright for development of new and better antiobesity medications to be used with diet and exercise to facilitate substantial weight loss.
Collapse
Affiliation(s)
- Karen E Foster-Schubert
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Veterans Administration Puget Sound Health Care System, Seattle, Washington 98108, USA
| | | |
Collapse
|
203
|
Abstract
The melanocortin system refers to a set of hormonal, neuropeptidergic, and paracrine signaling pathways that are defined by components that include the five G protein-coupled melanocortin receptors; peptide agonists derived from the proopiomelanocortin preprohormone precursor; and the endogenous antagonists, agouti and agouti-related protein. This signaling system regulates a remarkably diverse array of physiological functions including pigmentation, adrenocortical steroidogenesis, energy homeostasis, natriuresis, erectile responses, energy homeostasis, and exocrine gland secretion. There are many complex and unique aspects of melanocortin signaling, such as the existence of endogenous antagonists, the agouti proteins, that act at three of the five melanocortin receptors. However, there is an aspect of melanocortin signaling that has facilitated highly reductionist approaches aimed at understanding the physiological functions of each receptor and peptide: in contrast to many peptides, the melanocortin agonists and antagonists are expressed in a limited number of very discrete locations. Similarly, the melanocortin receptors are also expressed in a limited number of discrete locations where they tend to be involved in rather circumscribed physiological functions. This review examines my laboratory's participation in the cloning of the melanocortin receptors and characterization of their physiological roles.
Collapse
Affiliation(s)
- Roger D Cone
- Center for the Study of Weight Regulation and Associated Disorders, and Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098, USA.
| |
Collapse
|
204
|
Vaughan CH, Haskell-Luevano C, Andreasen A, Rowland NE. Effects of oral preload, CCK or bombesin administration on short term food intake of melanocortin 4-receptor knockout (MC4RKO) mice. Peptides 2006; 27:3226-33. [PMID: 16963158 DOI: 10.1016/j.peptides.2006.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Revised: 07/30/2006] [Accepted: 08/03/2006] [Indexed: 11/25/2022]
Abstract
We investigated whether either heterozygous (HET) or homozygous (knockout, KO) disruption of the melanocortin type 4 receptor (MC4R) gene alters post ingestive responsiveness of mice. Specifically, we tested the hypothesis that hyperphagia in MC4RKO mice might be due to a deficit in processes that sustain intermeal intervals (satiety) and/or processes that terminate ongoing episodes of eating (satiation). To test satiety, mice drank an oral preload and then we monitored intake of a subsequent liquid diet test meal. To test satiation, we examined the effect of exogenous administration of cholecystokinin (CCK) and bombesin (BN) on the size of a liquid diet meal. Experiment 1 was comprised of two studies. In the first, we determined that the intake of all three genotypes following fasts of either 6, 12, or 24h were comparable, and so chose 12h deprivation for the subsequent studies. In the second, 12h fasted mice were allowed to consume a fixed preload, approximately 50% of their expected mean intake and, following delays of either 30 or 60 min, were allowed to consume to satiation. Compared with no preload, the preload significantly reduced meal size comparably in all three genotypes. The reduction in intake was greater when the test meal was presented 30 compared with 60 min after the preload, again with no genotype differences in this decay of satiety. In experiment 2, we administered either CCK or BN and examined suppression of meal size after a 12h fast. Mice were tested repeatedly with CCK-8 (2, 6, or 18 microg/kg ip) or BN (2, 4 or 8 microg/kg ip) with vehicle injection days intervening. The 30 min intakes of HET and KO mice were suppressed more than those of WT following either CCK or BN. These experiments suggest that diminished responsiveness to nutrients or gut satiety hormones is not responsible for hyperphagia in MC4RKO mice.
Collapse
Affiliation(s)
- C H Vaughan
- Department of Psychology, University of Florida, Gainesville, FL 32611, USA
| | | | | | | |
Collapse
|
205
|
Van der Ploeg LHT, Kanatani A, MacNeil D, Ming Fong T, Strack A, Nargund R, Guan XM. Design and synthesis of (ant)-agonists that alter appetite and adiposity. PROGRESS IN BRAIN RESEARCH 2006; 153:107-18. [PMID: 16876570 DOI: 10.1016/s0079-6123(06)53005-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Over the past decade, hypothalamic circuits have been described that impact energy homeostasis in rodents and humans. Our drug development efforts for the treatment of obesity and the metabolic syndrome have largely focused on selected genetic and/or pharmacologically validated pathways. The translation of these pathways into therapeutics for the treatment of obesity will find its first clinical successes over the coming decade. Initial efforts have focused on gaining a better understanding of the relevance of rodent pharmacological and genetic observations for the development of therapeutics for the treatment of human obesity. We pursue pathways defined by the expression of the ghrelin receptor, melanin-concentrating hormone receptors, melanocortin receptors, cannabinoid receptors and neuropeptide Y1 and Y5 receptors. In this review, we will discuss drug development efforts for the treatment of obesity, focused on selective melanocortin 4 receptor agonists and neuropeptide Y1 and Y5 receptor antagonists. These drug development efforts required an in-depth understanding of cell-based observations which drive the development of compound structure-activity relationships. These include understanding of receptor function in selected cell-based backgrounds and early evaluation and validation of ex vivo observations in appropriate in vivo models. In order to develop selective and safe anti-obesity drugs, diverse approaches are needed to increase the likelihood of clinical success, including: (i) developing a detailed understanding of the predictive value of rodent pathways for treatment of human disease; (ii) knowledge of the exact location of targeted receptor subtypes for the clinical indication under study in order to derive a suitable compound profile; (iii) predictive measures of in vivo and/or ex vivo receptor occupancy required to bring about a desired physiological effect; (iv) predictive parameters that outline that the drug-derived effects are safe and mechanism-based; and (v) the refinement of selected compound classes, aimed at their clinical use.
Collapse
|
206
|
Abstract
In recent years, scientific interest in melanocortins (MC) has progressively increased due to their wide range of effects and expression on various tissues. Primarily discovered as mediators of skin pigmentation, recent research has shown their important roles in various body functions, such as energy homeostasis, sexual function, and inflammation. The anti-inflammatory and immunomodulatory properties discovered so far have led to the hypothesis that alpha-melanocyte-stimulating hormone (MSH) and its cognate receptors might present potential anti-inflammatory treatment options.
Collapse
Affiliation(s)
- Christian Maaser
- Department of Medicine B, University of Muenster, Albert-Schweitzer Strasse 33, 48129 Muenster, Germany.
| | | | | |
Collapse
|
207
|
Adan RAH, Tiesjema B, Hillebrand JJG, la Fleur SE, Kas MJH, de Krom M. The MC4 receptor and control of appetite. Br J Pharmacol 2006; 149:815-27. [PMID: 17043670 PMCID: PMC2014686 DOI: 10.1038/sj.bjp.0706929] [Citation(s) in RCA: 178] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mutations in the human melanocortin (MC)4 receptor have been associated with obesity, which underscores the relevance of this receptor as a drug target to treat obesity. Infusion of MC4R agonists decreases food intake, whereas inhibition of MC receptor activity by infusion of an MC receptor antagonist or with the inverse agonist AgRP results in increased food intake. This review addresses the role of the MC system in different aspects of feeding behaviour. MC4R activity affects meal size and meal choice, but not meal frequency, and the type of diet affects the efficacy of MC4R agonists to reduce food intake. The central sites involved in the different aspects of feeding behaviour that are affected by MC4R signalling are being unravelled. The paraventricular nucleus plays an important role in food intake per se, whereas MC signalling in the lateral hypothalamus is associated with the response to a high fat diet. MC4R signalling in the brainstem has been shown to affect meal size. Further genetic, behavioural and brain-region specific studies need to clarify how the MC4R agonists affect feeding behaviour in order to determine which obese individuals would benefit most from treatment with these drugs. Application of MCR agonists in humans has already revealed side effects, such as penile erections, which may complicate introduction of these drugs in the treatment of obesity.
Collapse
MESH Headings
- Agouti-Related Protein
- Animals
- Anti-Obesity Agents/pharmacology
- Appetite Depressants/pharmacology
- Appetite Regulation/drug effects
- Brain/metabolism
- Diet
- Energy Intake
- Feeding Behavior
- Food Preferences
- Humans
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Ligands
- Melanocortins/metabolism
- Mice
- Mice, Transgenic
- Mutation
- Nutritional Physiological Phenomena
- Obesity/genetics
- Obesity/metabolism
- Obesity/physiopathology
- Pro-Opiomelanocortin/genetics
- Pro-Opiomelanocortin/metabolism
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 4/drug effects
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- Signal Transduction/drug effects
- Time Factors
Collapse
Affiliation(s)
- R A H Adan
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Centre Utrecht, Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
208
|
Dembele K, Yao XH, Chen L, Nyomba BLG. Intrauterine ethanol exposure results in hypothalamic oxidative stress and neuroendocrine alterations in adult rat offspring. Am J Physiol Regul Integr Comp Physiol 2006; 291:R796-802. [PMID: 16614051 DOI: 10.1152/ajpregu.00633.2005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Prenatal ethanol (EtOH) exposure is associated with low birth weight, followed by increased appetite, catch-up growth, insulin resistance, and impaired glucose tolerance in the rat offspring. Because EtOH can induce oxidative stress, which is a putative mechanism of insulin resistance, and because of the central role of the hypothalamus in the regulation of energy homeostasis and insulin action, we investigated whether prenatal EtOH exposure causes oxidative damage to the hypothalamus, which may alter its function. Female rats were given EtOH by gavage throughout pregnancy. At birth, their offspring were smaller than those of non-EtOH rats. Markers of oxidative stress and expression of neuropeptide Y and proopiomelanocortin (POMC) were determined in hypothalami of postnatal day 7 (PD7) and 3-mo-old (adult) rat offspring. In both PD7 and adult rats, prenatal EtOH exposure was associated with decreased levels of glutathione and increased expression of MnSOD. The concentrations of lipid peroxides and protein carbonyls were normal in PD7 EtOH-exposed offspring, but were increased in adult EtOH-exposed offspring. Both PD7 and adult EtOH-exposed offspring had normal neuropeptide Y and POMC mRNA levels, but the adult offspring had reduced POMC protein concentration. Thus only adult offspring preexposed to EtOH had increased hypothalamic tissue damage and decreased levels of POMC, which could impair melanocortin signaling. We conclude that prenatal EtOH exposure causes hypothalamic oxidative stress, which persists into adult life and alters melanocortin action during adulthood. These neuroendocrine alterations may explain weight gain and insulin resistance in rats exposed to EtOH early in life.
Collapse
Affiliation(s)
- Korami Dembele
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada R3E3P4
| | | | | | | |
Collapse
|
209
|
Dube MG, Kalra SP, Kalra PS. The hypothalamic paraventricular nucleus is not essential for orexigenic NPY or anorexigenic melanocortin action. Peptides 2006; 27:2239-48. [PMID: 16759748 DOI: 10.1016/j.peptides.2006.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2006] [Revised: 04/11/2006] [Accepted: 04/11/2006] [Indexed: 10/24/2022]
Abstract
Bilateral electrolytic lesions of the paraventricular nucleus of the hypothalamus (PVN) produce hyperphagia with excess weight gain. The orexigenic neuropeptide Y (NPY) system and the anorexigenic melanocortin system act in the PVN to regulate food intake, and participate in mediating the anorexic effects of leptin. We hypothesized that changes in the responsiveness of these systems may contribute to the hyperphagia observed in PVN-lesioned rats. Adult female Sprague-Dawley rats received either sham or electrolytic lesions in the PVN immediately followed by implantation of a guide cannula into the third cerebroventricle. Twenty-five days following surgery groups of sham and hyperphagic PVN-lesioned rats were injected intracerebroventricularly (i.c.v.) with either 118 pmole or 470 pmole of NPY and food intake was measured for 3 h. Food intake in response to NPY was nearly three-fold higher in PVN-lesioned rats as compared to sham rats. However, the response to 5 microg leptin i.c.v. was not different in lesioned versus sham rats. The effect of the melanocortin agonist MTII on food intake was tested in additional rats beginning either 7-14 days or 30-40 days following surgery. Doses of 0.1 nmole or 1.0 nmole of MTII were injected immediately before lights-off and food intake was measured at 2 h, 24 h and 48 h post-injection. Suppression of food intake in PVN-lesioned rats was not different from that in sham-lesioned rats. These data suggest that hyper-responsiveness to NPY may account in part for the hyperphagia observed in PVN-lesioned rats. Furthermore, based on the similarities of responses of PVN-lesioned and sham control rats to the anorexigenic agents MTII and leptin and the hypersensitivity of lesioned rats to NPY, we conclude that the PVN is not essential for NPY stimulation of food intake or for melanocortin suppression of food intake and that NPY and melanocortin receptors outside of the PVN are sufficient to produce these effects.
Collapse
Affiliation(s)
- Michael G Dube
- Department of Physiology and Functional Genomics, McKnight Brain Institute, University of Florida, College of Medicine, P.O. Box 100274, Gainesville, FL 32610-0274, USA
| | | | | |
Collapse
|
210
|
Ryabinin AE, Weitemier AZ. The urocortin 1 neurocircuit: Ethanol-sensitivity and potential involvement in alcohol consumption. ACTA ACUST UNITED AC 2006; 52:368-80. [PMID: 16766036 DOI: 10.1016/j.brainresrev.2006.04.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Revised: 04/26/2006] [Accepted: 04/29/2006] [Indexed: 11/27/2022]
Abstract
One of the hallmarks of alcoholism is continued excessive consumption of alcohol-containing beverages despite the negative consequences of such behavior. The neurocircuitry regulating alcohol consumption is not well understood. Recent studies have shown that the neuropeptide urocortin 1 (Ucn1), a member of the corticotropin-releasing factor (CRF) family of peptides, could be an important player in the regulation of alcohol consumption. This evidence is accumulated along three directions of research: (1) Ucn 1-containing neurons are extremely sensitive to alcohol; (2) the Ucn1 neurocircuit may contribute to the genetic predisposition to high alcohol intake in mice and rats; (3) manipulation of the Ucn1 system alters alcohol consumption and sensitivity. This paper reviews the current knowledge of the Ucn1 neurocircuit and the evidence for its involvement in alcohol-related behaviors, and proposes a mechanism for its involvement in the regulation of alcohol consumption.
Collapse
Affiliation(s)
- Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health and Science University, L470, 3181 SW Sam Jackson Park Road, Portland, 97239, USA.
| | | |
Collapse
|
211
|
Hen G, Yosefi S, Simchaev V, Shinder D, Hruby VJ, Friedman-Einat M. The melanocortin circuit in obese and lean strains of chicks. J Endocrinol 2006; 190:527-35. [PMID: 16899585 PMCID: PMC2730167 DOI: 10.1677/joe.1.06783] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Agonists of membranal melanocortin 3 and 4 receptors (MC3/4Rs) are known to take part in the complex control mechanism of energy balance. In this study, we compared the physiological response to an exogenous MC3/4R agonist and the hypothalamic expression of proopic melanocortin (POMC) gene, encoding few MC3/4R ligands, between broiler and layer chicken strains. These strains, representing the two most prominent commercial strains of chickens grown for meat (broilers) and egg production (layers), differ in their food intake, fat accumulation, and reproductive performance and, therefore, form a good model of obese and lean phenotypes, respectively. A single i.v. injection of the synthetic peptide melanotan-II (MT-II; 1 mg/kg body weight) into the wing vein of feed-restricted birds led to attenuation of food intake upon exposure to feeding ad libitum in both broiler and layer chickens. A study of the POMC mRNA encoding the two prominent natural MC3/4R agonists, alpha-MSH and ACTH, also revealed a general similarity between the strains. Under feeding conditions ad libitum, POMC mRNA levels were highly similar in chicks of both strains and this level was significantly reduced upon feed restriction. However, POMC mRNA down-regulation upon feed restriction was more pronounced in layers than in broilers. These results suggest: (i) a role for MC3/4R agonists in the control of appetite; (ii) that the physiological differences between broilers and layers are not related to unresponsiveness of broiler chickens to the satiety signal of MC3/4R ligands. Therefore, these findings suggest that artificial activation of this circuit in broiler chicks could help to accommodate with their agricultural shortcomings of overeating, fattening, and impaired reproduction.
Collapse
Affiliation(s)
- Gideon Hen
- Institute of Animal Science, Agricultural Research Organization, Volcani Center, PO Box 6, Bet Dagan 50250, Israel
| | | | | | | | | | | |
Collapse
|
212
|
Nahon JL. The melanocortins and melanin-concentrating hormone in the central regulation of feeding behavior and energy homeostasis. C R Biol 2006; 329:623-38; discussion 653-5. [PMID: 16860280 DOI: 10.1016/j.crvi.2006.03.021] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2006] [Accepted: 03/08/2006] [Indexed: 11/16/2022]
Abstract
A number of different neuropeptides exert powerful concerted controls on feeding behavior and energy balance, most of them being produced in hypothalamic neuronal networks under stimulation by anabolic and catabolic peripheral hormones such as ghrelin and leptin, respectively. These peptide-expressing neurons interconnect extensively to integrate the multiple opposing signals that mediate changes in energy expenditure. In the present review I have summarized our current knowledge about two key peptidic systems involved in regulating appetite and energy homeostasis, the melanocortin system (alpha-MSH, agouti and Agouti-related peptides, MC receptors and mahogany protein) and the melanin-concentrating hormone system (proMCH-derived peptides and MCH receptors) that contribute to satiety and feeding-initiation, respectively, with concurrent effects on energy expenditure. I have focused particularly on recent data concerning transgenic mice and the ongoing development of MC/MCH receptor antagonists/agonists that may represent promising drugs to treat human eating disorders on both sides of the energy balance (anorexia, obesity).
Collapse
Affiliation(s)
- Jean-Louis Nahon
- Institut de pharmacologie moléculaire et cellulaire, UMR 6097, Centre national de la recherche scientifique (CNRS), 660, route des Lucioles, Sophia-Antipolis, 06560 Valbonne, France.
| |
Collapse
|
213
|
Abstract
Obesity is a major public health concern and environmental factors are involved in its development. The hypothalamus is a primary site for the integration of signals for the regulation of energy homeostasis. Dysregulation of these pathways can lead to weight loss or gain. Some drugs in development can have favourable effects on body weight, acting on some of these pathways and leading to responses resulting in weight loss. Strategies for the management of weight reduction include exercise, diet, behavioural therapy, drug therapy and surgery. Investigational antiobesity medications can modulate energy homeostasis by stimulating catabolic or inhibiting anabolic pathways. Investigational drugs stimulating catabolic pathways consist of leptin, agonists of melanocortin receptor-4, 5-HT and dopamine; bupropion, growth hormone fragments, cholecystokinin subtype 1 receptor agonist, peptide YY3-36, oxyntomodulin, ciliary neurotrophic factor analogue, beta3-adrenergic receptor agonists, adiponectin derivatives and glucagon-like peptide-1. On the other hand, investigational drugs inhibiting anabolic pathways consist of the ghrelin receptor, neuropeptide Y receptor and melanin-concentrating hormone-1 antagonists; somatostatin analogues, peroxisome proliferator-activated receptor-gamma and -beta/delta antagonists, gastric emptying retardation agents, pancreatic lipase inhibitors, topiramate and cannabinoid-1 receptor antagonists. These differing approaches are reviewed and commented on in this article.
Collapse
MESH Headings
- Animals
- Anti-Obesity Agents/pharmacology
- Anti-Obesity Agents/therapeutic use
- Body Weight
- Drugs, Investigational/pharmacology
- Drugs, Investigational/therapeutic use
- Energy Metabolism
- Humans
- Hypothalamus/drug effects
- Hypothalamus/metabolism
- Leptin/genetics
- Leptin/pharmacology
- Leptin/therapeutic use
- Obesity/drug therapy
- Obesity/metabolism
- Peroxisome Proliferator-Activated Receptors/drug effects
- Peroxisome Proliferator-Activated Receptors/metabolism
- Randomized Controlled Trials as Topic
- Receptor, Melanocortin, Type 4/agonists
- Receptor, Melanocortin, Type 4/metabolism
- Receptor, Serotonin, 5-HT1B/metabolism
- Receptor, Serotonin, 5-HT2C/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Ghrelin
- Receptors, Neuropeptide Y/antagonists & inhibitors
- Receptors, Neuropeptide Y/metabolism
- Recombinant Proteins/pharmacology
- Recombinant Proteins/therapeutic use
- Serotonin 5-HT1 Receptor Agonists
- Serotonin 5-HT2 Receptor Agonists
- Serotonin Receptor Agonists/pharmacology
- Serotonin Receptor Agonists/therapeutic use
Collapse
Affiliation(s)
- Marcio C Mancini
- Sao Paulo University, Obesity & Metabolic Syndrome Group of the Endocrinology & Metabology Service, Faculty of Medicine, Hospital das Clínicas, Sao Paulo, Brazil.
| | | |
Collapse
|
214
|
Aja S, Bi S, Knipp SB, McFadden JM, Ronnett GV, Kuhajda FP, Moran TH. Intracerebroventricular C75 decreases meal frequency and reduces AgRP gene expression in rats. Am J Physiol Regul Integr Comp Physiol 2006; 291:R148-54. [PMID: 16484442 DOI: 10.1152/ajpregu.00041.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
3-Carboxy-4-alkyl-2-methylenebutyrolactone (C75), an inhibitor of fatty acid synthase and stimulator of carnitine palmitoyltransferase-1, reduces food intake and body weight in rodents when given systemically or centrally. Intracellular molecular mechanisms involving changes in cellular energy status are proposed to initiate the feeding and body weight reductions. However, effectors that lie downstream of these initial steps are not yet fully identified. Present experiments characterize the time courses of hypophagia and weight loss after single injections of C75 into the lateral cerebroventicle in rats and go on to identify specific meal pattern changes and coinciding alterations in gene expression for feeding-related hypothalamic neuropeptides. C75 reduced chow intake and body weight dose dependently. Although the principal effects occurred on the first day, weight losses relative to vehicle control were maintained over multiple days. C75 did not affect generalized locomotor activity. C75 began to reduce feeding after a 6-h delay. The hypophagia was due primarily to decreased meal number during 6–12 h without a significant effect on meal size, suggesting that central C75 reduced the drive to initiate meals. C75 prevented the anticipated hypophagia-induced increases in mRNA for AgRP in the arcuate nucleus at 22 h and at 6 h when C75 begins to suppress feeding. Overall, the data suggest that gene expression changes leading to altered melanocortin signaling are important for the hypophagic response to intracerebroventricular C75.
Collapse
Affiliation(s)
- Susan Aja
- Department of Psychiatry and Behavioural Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | |
Collapse
|
215
|
Abstract
Although the presence of hyperleptinemia with leptin resistance and obesity has long been recognized, a causal role of elevated leptin in these biological states remains unclear. This article summarizes some recent work from our laboratory supporting the concept that leptin, in and of itself, promotes leptin resistance and such resistance compounds the metabolic impact of diet-induced obesity. Results from multiple studies demonstrate that (1) chronically elevated central leptin decreases hypothalamic leptin receptor expression and protein levels and impairs leptin signaling; (2) leptin resistance and obesity are associated with reduced leptin receptors and diminished maximal leptin signaling capacity; and (3) leptin resistance confers increased susceptibility to diet-induced obesity. In essence, the augmented leptin accompanying obesity contributes to leptin resistance, and this leptin resistance promotes further obesity, leading to a vicious cycle of escalating metabolic devastation.
Collapse
Affiliation(s)
- Yi Zhang
- Research Service, Department of Veterans Affairs Medical Center, Gainesville, FL 32608, USA
| | | |
Collapse
|
216
|
Tallam LS, da Silva AA, Hall JE. Melanocortin-4 receptor mediates chronic cardiovascular and metabolic actions of leptin. Hypertension 2006; 48:58-64. [PMID: 16754792 DOI: 10.1161/01.hyp.0000227966.36744.d9] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study tested whether the melanocortin 4-receptor (MC4R) is essential for the chronic cardiovascular and metabolic actions of leptin. Twenty- to 22-week-old male wild-type (WT) C57BL/6J and obese MC4R (-/-) mice (N=5 to 6 per group) were implanted with radiotelemetric transmitters and catheters for measuring mean arterial pressure (MAP) and heart rate 24 hours per day and intravenous infusions. After a 3-day stable control period, leptin was infused (2 microg/kg per minute IV) for 7 days in WT, obese ad libitum-fed MC4R (-/-), and nonobese pair-fed MC4R (-/-) mice. WT mice receiving vehicle for 7 days served as controls. MC4 (-/-) mice were 30% heavier and had 4- and 11-fold increases in plasma insulin and leptin levels, respectively, compared with WT mice. Despite obesity, MAP and heart rate tended to be lower in MC4R (-/-) mice compared with WT mice. Chronic leptin infusion in the different groups increased plasma leptin levels to 45 to 65 ng/mL. Seven-day leptin infusion in WT mice increased MAP by 12+/-3 mm Hg despite a 35% reduction in food intake and an 8% reduction in body weight. Leptin did not alter plasma glucose but reduced plasma insulin in WT mice (5.9+/-1.0 versus 3.0+/-0.5 microU/mL). These cardiovascular and metabolic actions of leptin were abolished in obese and nonobese MC4R (-/-) mice. These data suggest that MC4R deficiency, and not obesity-induced leptin resistance, abolished the cardiovascular and metabolic actions of leptin in obese MC4R (-/-) mice. Thus, a functional MC4R is essential for the chronic cardiovascular and metabolic actions of leptin.
Collapse
Affiliation(s)
- Lakshmi S Tallam
- Department of Physiology, School of Medicine, University of Mississippi Medical Center, Jackson, Miss 39216, USA
| | | | | |
Collapse
|
217
|
Sutton GM, Trevaskis JL, Hulver MW, McMillan RP, Markward NJ, Babin MJ, Meyer EA, Butler AA. Diet-genotype interactions in the development of the obese, insulin-resistant phenotype of C57BL/6J mice lacking melanocortin-3 or -4 receptors. Endocrinology 2006; 147:2183-96. [PMID: 16469808 PMCID: PMC2735103 DOI: 10.1210/en.2005-1209] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Loss of brain melanocortin receptors (Mc3rKO and Mc4rKO) causes increased adiposity and exacerbates diet-induced obesity (DIO). Little is known about how Mc3r or Mc4r genotype, diet, and obesity affect insulin sensitivity. Insulin resistance, assessed by insulin and glucose tolerance tests, Ser(307) phosphorylation of insulin receptor substrate 1, and activation of protein kinase B, was examined in control and DIO wild-type (WT), Mc3rKO and Mc4rKO C57BL/6J mice. Mc4rKO mice were hyperphagic and had increased metabolic efficiency (weight gain per kilojoule consumed) relative to WT; both parameters increased further on high-fat diet. Obesity of Mc3rKO was more dependent on fat intake, involving increased metabolic efficiency. Fat mass of DIO Mc3rKO and Mc4rKO was similar, although Mc4rKO gained weight more rapidly. Mc4rKO develop hepatic insulin resistance and severe hepatic steatosis with obesity, independent of diet. DIO caused further deterioration of insulin action in Mc4rKO of either sex and, in male Mc3rKO, compared with controls, associated with increased fasting insulin, severe glucose intolerance, and reduced insulin signaling in muscle and adipose tissue. DIO female Mc3rKO exhibited very modest perturbations in glucose metabolism and insulin sensitivity. Consistent with previous data suggesting impaired fat oxidation, both Mc3rKO and Mc4rKO had reduced muscle oxidative metabolism, a risk factor for weight gain and insulin resistance. Energy expenditure was, however, increased in Mc4rKO compared with Mc3rKO and controls, perhaps due to hyperphagia and metabolic costs associated with rapid growth. In summary, DIO affects insulin sensitivity more severely in Mc4rKO compared with Mc3rKO, perhaps due to a more positive energy balance.
Collapse
MESH Headings
- Adipose Tissue/metabolism
- Animal Feed
- Animals
- Calorimetry
- Cell Proliferation
- Diet
- Energy Metabolism
- Fatty Acids/metabolism
- Female
- Genotype
- Glucose/metabolism
- Insulin/metabolism
- Insulin Resistance
- Liver/metabolism
- Liver/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Obese
- Muscle, Skeletal/metabolism
- Obesity
- Oxygen/metabolism
- Phenotype
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- RNA/metabolism
- Receptor, Insulin/metabolism
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/physiology
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/physiology
- Regression Analysis
- Risk Factors
- Sensitivity and Specificity
- Serine/chemistry
- Sex Factors
- Signal Transduction
- Time Factors
Collapse
Affiliation(s)
- Gregory M Sutton
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, 70808, USA
| | | | | | | | | | | | | | | |
Collapse
|
218
|
Salomon G, Niv-Spector L, Gussakovsky EE, Gertler A. Large-scale preparation of biologically active mouse and rat leptins and their L39A/D40A/F41A muteins which act as potent antagonists. Protein Expr Purif 2006; 47:128-36. [PMID: 16289983 DOI: 10.1016/j.pep.2005.09.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2005] [Revised: 09/23/2005] [Accepted: 09/24/2005] [Indexed: 11/25/2022]
Abstract
Expression plasmids encoding mouse and rat leptins and their L39A/D40A/F41A muteins were prepared. The proteins were expressed in Escherichia coli, refolded and purified to homogeneity, yielding electrophoretically pure, over 98% monomeric protein. Circular dichroism (CD) analysis revealed that the mutations hardly affect the leptins' secondary structure, and they were similar to previously reported CD spectra for human leptin. Both mouse and rat leptins were biologically active in promoting proliferation in BAF/3 cells stably transfected with the long form of human leptin receptor. The mutations did not change the binding properties to BAF/3 cells as compared, respectively, to non-mutated mouse, rat or human leptins, or their ability to form 1:1 complexes with the leptin-binding domain of chicken leptin receptor. In contrast, their biological activity, tested in a BAF/3 proliferation assay, was abolished and both became potent antagonists. As the LDF (amino acids 39-41) sequence is preserved in all known leptins, the present results substantiate the hypothesis that this sequence plays a pivotal role in leptins' site III and that interaction of leptin with its receptors resembles the corresponding interactions of interleukin-6 and granulocyte colony-stimulating factor their receptors.
Collapse
Affiliation(s)
- Gili Salomon
- Faculty of Agricultural, Food and Environmental Quality Sciences, The Hebrew University, Rehovot 76100, Israel
| | | | | | | |
Collapse
|
219
|
Jokubka R, Maak S, Kerziene S, Swalve HH. Association of a melanocortin 4 receptor (MC4R) polymorphism with performance traits in Lithuanian White pigs. J Anim Breed Genet 2006; 123:17-22. [PMID: 16420261 DOI: 10.1111/j.1439-0388.2006.00559.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The melanocortin 4 receptor is expressed in virtually all brain regions of mammals and plays an important role in energy homeostasis. Polymorphisms in this gene may thus be related to growth and obesity. In pigs, a non-synonymous polymorphic site was described (Asp298Asn) and demonstrated to affect cAMP production and to alter adenylyl cyclase signalling. Association studies revealed significant linkage of this mutation with production trait in pigs. In this study, 207 Lithuanian White pigs were genotyped at the MC4R locus and analysed on relationships between genotype and breeding values for several performance traits. The observed allele and genotype frequencies did not deviate significantly from Hardy-Weinberg equilibrium (wildtype allele 0.59; mutant allele 0.41) and are comparable with those described in other Large White populations. The mutant Asn298 allele of the MC4R gene was significantly associated with increased test daily gain, higher lean meat percentage and lower backfat thickness. There was a trend towards an improved feed conversion ratio (p = 0.065) in animals with the mutant allele whereas no significant effect was found on lifetime daily gain. These results indicate that the MC4R polymorphism should be integrated in selection programmes in the Lithuanian White to improve carcass composition.
Collapse
Affiliation(s)
- R Jokubka
- Department of Animal Breeding and Genetics, Lithuanian Veterinary Academy, Kaunas, Lithuania.
| | | | | | | |
Collapse
|
220
|
Abstract
Obesity is rapidly becoming an epidemic in developed countries. Currently available anti-obesity therapeutics are only modestly effective and are accompanied by significant adverse effects. In recent years, substantial advances have been made in the basic understanding of brain control of feeding behaviour and metabolism. As a result, several compounds have progressed to Phase III development, with additional compounds at various stages of Phase II development. Most of the late-stage development candidates are CNS agents, which reflects the consensus that the brain exerts a dominant control on feeding behaviour and peripheral metabolism through the autonomic nervous system. Homeostatic mechanisms encompassing hypothalamic/brainstem pathways have long been recognised in obesity research. In addition, non-homeostatic mechanisms encompassing the reward circuit and volitional control need to be targeted to control feeding behaviour and physical activity, especially in humans. While recognising the importance of CNS control, certain peripherally acting agents can affect mitochondrial metabolism, lipolysis, nutrient absorption or the vagal feedback pathway, such that these peripherally acting agents can potentially be combined with CNS agents to achieve maximal efficacy. It is expected that newer generations of anti-obesity therapeutics will be superior to existing agents and will facilitate lifestyle modification.
Collapse
Affiliation(s)
- Tung M Fong
- Department of Metabolic Disorders, Merck Research Laboratories, R80M-213, PO Box 2000, Rahway, NJ 07065, USA.
| |
Collapse
|
221
|
Elmquist JK, Coppari R, Balthasar N, Ichinose M, Lowell BB. Identifying hypothalamic pathways controlling food intake, body weight, and glucose homeostasis. J Comp Neurol 2006; 493:63-71. [PMID: 16254991 DOI: 10.1002/cne.20786] [Citation(s) in RCA: 319] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The past decade has greatly increased our understanding and appreciation of the ability of the central nervous system (CNS) to regulate food intake and body weight. This was spearheaded by the discovery of key molecules regulating body weight homeostasis. It is now also apparent that the CNS, especially the hypothalamus, plays a primary role in directly regulating glucose homeostasis, independently of effects on body weight. These discoveries are important given the increasing incidences of obesity and type II diabetes in Western societies. In this article, we will highlight recent data from genetically modified mice. These data and other models have helped to dissect the CNS pathways regulating body weight and glucose homeostasis. Finally, although these studies have been illustrative, they also underscore our relative lack of knowledge and highlight the need for more definitive approaches to unravel the functional significance of these pathways.
Collapse
Affiliation(s)
- Joel K Elmquist
- Department of Medicine and Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | |
Collapse
|
222
|
Hillebrand JJG, Kas MJH, Adan RAH. To eat or not to eat; regulation by the melanocortin system. Physiol Behav 2006; 89:97-102. [PMID: 16574172 DOI: 10.1016/j.physbeh.2006.01.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Accepted: 01/10/2006] [Indexed: 10/24/2022]
Abstract
The central melanocortin (MC) system is one of the best-characterized neuropeptidergic systems involved in the regulation of energy balance. This short review describes the role of the central MC system in feeding behavior. Pharmacological, anatomical and genetic studies show that activation of the MC system reduces meal size, whereas de-activation of the MC system increases meal size. Several brain regions, including distinct hypothalamic nuclei and the hindbrain, are involved in this process. Further dissection of MC pathways in feeding behavior is the subject of recent and probably future studies. As the MC system is involved in animal models of obesity and (possibly) anorexia, it appears that this is a target system for development of drugs for the treatment of disturbed human eating behavior.
Collapse
Affiliation(s)
- Jacquelien J G Hillebrand
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | |
Collapse
|
223
|
Zhang Y, Kilroy GE, Henagan TM, Prpic-Uhing V, Richards WG, Bannon AW, Mynatt RL, Gettys TW. Targeted deletion of melanocortin receptor subtypes 3 and 4, but not CART, alters nutrient partitioning and compromises behavioral and metabolic responses to leptin. FASEB J 2006; 19:1482-91. [PMID: 16126916 DOI: 10.1096/fj.05-3851com] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mouse lines with targeted disruption of the cocaine amphetamine-related transcript (CART), melanocortin receptor 3 (MCR3), or melanocortin receptor 4 (MCR4) were used to assess the role of each component in mediating the anorectic and metabolic effects of leptin, and in regulating the partitioning of nutrient energy between fat and protein deposition. Leptin was administered over a 3 day period using either intraperitoneal or intracerebroventricular routes of injection. The absence of MCR4 blocked leptin's ability to increase UCP1 mRNA in both brown and white adipose tissue, but not its ability to reduce food consumption. In contrast, deletion of MCR3 compromised leptin's ability to reduce food consumption, but not its ability to reduce fat deposition or increase UCP1 expression in adipose tissue. Leptin-dependent effects on food consumption and adipocyte gene expression were unaffected by the absence of CART. Repeated measures of body composition over time indicate that the absence of either MCR3 or MCR4, but not CART, increased lipid deposition and produced comparable degrees of adiposity in both lines. Moreover, modest increases in fat content of the diet (4 to 11%) accentuated fat deposition and produced a rapid and comparable 10-12% increase in % body fat in both genotypes. The results indicate that nutrient partitioning, as well as the anorectic and metabolic responses to leptin, are dependent on integrated but separable inputs from the melanocortin 3 and 4 receptor subtypes.
Collapse
MESH Headings
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Animals
- Body Composition/drug effects
- Carrier Proteins/genetics
- Eating/drug effects
- Energy Metabolism/drug effects
- Gene Expression Regulation/drug effects
- Ion Channels
- Leptin/pharmacology
- Male
- Membrane Proteins/genetics
- Mice
- Mitochondrial Proteins
- Nerve Tissue Proteins/physiology
- RNA, Messenger/analysis
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/physiology
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/physiology
- Receptors, Leptin
- Uncoupling Protein 1
Collapse
Affiliation(s)
- Yubin Zhang
- Division of Experimental Obesity, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | | | | | | | | | | | | | | |
Collapse
|
224
|
|
225
|
Zhang Y, Scarpace PJ. Circumventing central leptin resistance: lessons from central leptin and POMC gene delivery. Peptides 2006; 27:350-64. [PMID: 16274846 DOI: 10.1016/j.peptides.2005.01.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2004] [Accepted: 01/10/2005] [Indexed: 10/25/2022]
Abstract
We identified that leptin resistance in aged-obese rats has both peripheral and central components. The central resistance is characterized by diminished hypothalamic leptin receptors and impaired leptin signal transduction. We developed a new model of leptin-induced leptin resistance in which application of the central leptin gene delivery produces unabated hypothalamic leptin over-expression. The chronic central elevation of leptin precipitates leptin resistance in young animals devoid of obesity and exacerbates it in mature or aged animals with obesity. Despite leptin resistance, our aged obese, DIO, and leptin-induced leptin resistant rats were fully responsive to central pharmacological melanocortin activation. We propose that the central leptin resistance resides between leptin receptor and melanocortin receptor activation. Our central POMC gene therapy overcame leptin resistance, producing weight and fat loss and improved insulin sensitivity in obese Zucker and aged rats. This success highlights the central melanocortin system as a useful drug target for combating obesity.
Collapse
Affiliation(s)
- Yi Zhang
- Geriatric Research, Education and Clinical Center, Department of Veterans Affairs Medical Center, Gainesville, FL 32608-1197, USA
| | | |
Collapse
|
226
|
Abstract
The melanocortins, a family of peptides produced from the post-translational processing of pro-opiomelanocortin (POMC), regulate ingestive behavior and energy expenditure. Loss of function mutations of genes encoding POMC, or of either of two melanocortin receptors expressed in the central nervous system (MC3R, MC4R), are associated with obesity. The analyses of MC4R knockout mice indicate that activation of this receptor is involved in the regulation of appetite, the adaptive metabolic response to excess caloric consumption, and negative energy balance associated with cachexia induced by cytokines. In contrast, MC3R knockout mice exhibit a normal, or even exaggerated, response to signals that induce a state of negative energy balance. However, loss of the MC3R also results in an increase in adiposity. This article discusses the regulation of energy balance by the melanocortins. Published and newly presented data from studies analyzing of energy balance of MC3R and MC4R knockout mice indicate that increased adiposity observed in both models involves an imbalance in fat intake and oxidation.
Collapse
|
227
|
Perez-Tilve D, Nogueiras R, Mallo F, Benoit SC, Tschoep M. Gut hormones ghrelin, PYY, and GLP-1 in the regulation of energy balance [corrected] and metabolism. Endocrine 2006; 29:61-71. [PMID: 16622293 DOI: 10.1385/endo:29:1:61] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 11/30/1999] [Accepted: 11/15/2005] [Indexed: 02/07/2023]
Abstract
The first hormone discovered in the gastrointestinal tract was secretin, isolated from duodenal mucosa. Some years later, two additional gastrointestinal hormones, gastrin and cholecystokinin (CCK), were discovered, but it was not until the 1970s that gastrointestinal endocrinology studies became more prevalent, resulting in the discovery of many more hormones. Here, we examine the role of gut hormones in energy balance regulation and their possible use as pharmaceutical targets for obesity.
Collapse
Affiliation(s)
- Diego Perez-Tilve
- Department of Psychiatry, University of Cincinnati Genome Research Institute, Cincinnati, OH 45237, USA
| | | | | | | | | |
Collapse
|
228
|
Abstract
Sequencing of the human genome has yielded thousands of potential drug targets. The difficulty now is in determining which targets have real therapeutic value and should be the focus of a drug discovery effort. The available evidence suggests that knockout technology can be used prospectively to identify targets that are amenable to drug development for the treatment of a variety of diseases. This review compares the knockout phenotypes of 21 potential obesity targets with the effects of therapeutics designed for those targets on rodents and, when data were available, on humans. The phenotypes of obesity target knockouts model the effects seen when therapeutics designed for those obesity targets are delivered to rodents; of the 21 obesity targets reviewed, 16 showed a correspondence between knockout phenotype and drug effect in mice and/or rats. This suggests that, at least in terms of evaluating obesity targets, it is rare for compensatory developmental changes caused by the gene knockout to prevent detection of the relevant phenotype. In the majority of cases, the knockout phenotypes also modelled the effects seen when the relevant therapeutics were delivered to humans. Thus, it seems rational to use mouse knockout technology prospectively to identify genes that regulate body fat in vivo, and then to develop anti-obesity therapeutics by targeting the human protein products of these genes. Ultimately, the value of using this approach to identify novel targets for human anti-obesity therapies will be judged by future studies examining the anti-obesity effect, in humans, of the therapeutics that result from this approach.
Collapse
Affiliation(s)
- D R Powell
- Department of Endocrinology, Pharmaceutical Biology, Lexicon Genetics Incorporated, 8800 Technology Forest Place, The Woodlands, TX 77381-1160, USA.
| |
Collapse
|
229
|
MacKenzie RG. Obesity-associated mutations in the human melanocortin-4 receptor gene. Peptides 2006; 27:395-403. [PMID: 16274851 DOI: 10.1016/j.peptides.2005.03.064] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2004] [Accepted: 03/07/2005] [Indexed: 10/25/2022]
Abstract
Mutations in the human melanocortin-4 receptor (MC4R) gene have been associated with severe obesity. Many of the mutations result in partial or complete loss-of-function based on the nature of the mutation or the function of mutated receptors when tested in heterologous expression systems. This review discusses the role of MC4R in the central regulation of body weight, the pathogenic mechanisms of the mutations, and the validity of MC4R as an anti-obesity drug target.
Collapse
Affiliation(s)
- Robert G MacKenzie
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, 3228 Scott Hall, 540 E. Canfield, Detroit, MI 48201, USA.
| |
Collapse
|
230
|
Yoshimatsu H. The neuronal histamine H(1) and pro-opiomelanocortin-melanocortin 4 receptors: independent regulation of food intake and energy expenditure. Peptides 2006; 27:326-32. [PMID: 16343692 DOI: 10.1016/j.peptides.2005.02.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2004] [Accepted: 02/21/2005] [Indexed: 11/29/2022]
Abstract
Hypothalamic neuronal histamine and its H(1) receptor (H(1)-R) form part of the leptin signaling pathway in the brain, and regulate body weight and adiposity by affecting food intake and energy expenditure. The pro-opiomelanocortin (POMC)-melanocortin 4 receptor (MC4-R) is also important for leptin signaling. We investigated whether and how these two neuronal pathways interact in regulating energy metabolism. From studies of agouti yellow (A(y)/a) obese mice, a model of a defect in POMC-MC4-R signaling, we concluded that the histamine H(1)-R signaling pathway is independent of the POMC-MC4-R complex in regulating food intake, energy metabolism, and adiposity.
Collapse
Affiliation(s)
- Hironobu Yoshimatsu
- Department of Internal Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama, Oita 879-5593, Japan.
| |
Collapse
|
231
|
Ellacott KLJ, Halatchev IG, Cone RD. Interactions between gut peptides and the central melanocortin system in the regulation of energy homeostasis. Peptides 2006; 27:340-9. [PMID: 16309792 DOI: 10.1016/j.peptides.2005.02.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Accepted: 02/21/2005] [Indexed: 12/20/2022]
Abstract
Genetic and pharmacological studies have shown that the central melanocortin system plays a critical role in the regulation of energy homeostasis. Animals and humans with defects in the central melanocortin system display a characteristic melanocortin obesity phenotype typified by increased adiposity, hyperphagia, metabolic defects and increased linear growth. In addition to interacting with long-term regulators of energy homeostasis such as leptin, more recent data suggest that the central melanocortin system also responds to gut-released peptides involved in mediating satiety. In this review, we discuss the interactions between these systems, with particular emphasis on cholecystokinin (CCK), ghrelin and PYY(3-36).
Collapse
Affiliation(s)
- Kate L J Ellacott
- Vollum Institute and The Center for Weight Regulation and Associated Disorders, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239-3098, USA
| | | | | |
Collapse
|
232
|
Jobst EE, Enriori PJ, Sinnayah P, Cowley MA. Hypothalamic regulatory pathways and potential obesity treatment targets. Endocrine 2006; 29:33-48. [PMID: 16622291 DOI: 10.1385/endo:29:1:33] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 11/30/1999] [Accepted: 11/08/2005] [Indexed: 12/25/2022]
Abstract
With an ever-growing population of obese people as well as comorbidities associated with obesity, finding effective weight loss strategies is more imperative than ever. One of the challenges in curbing the obesity crisis is designing successful strategies for long-term weight loss and weight-loss maintenance. Currently, weight-loss strategies include promotion of therapeutic lifestyle changes (diet and exercise), pharmacological therapy, and bariatric surgery. This review focuses on several pharmacological targets that activate central nervous system pathways that normally limit food intake and body weight. Though it is likely that no single therapy will prove effective for everyone, this review considers several recent pre-clinical targets, and several compounds that have been in human clinical trials.
Collapse
Affiliation(s)
- Erin E Jobst
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| | | | | | | |
Collapse
|
233
|
Smart JL, Tolle V, Low MJ. Glucocorticoids exacerbate obesity and insulin resistance in neuron-specific proopiomelanocortin-deficient mice. J Clin Invest 2006; 116:495-505. [PMID: 16440060 PMCID: PMC1350998 DOI: 10.1172/jci25243] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2005] [Accepted: 11/29/2005] [Indexed: 12/12/2022] Open
Abstract
Null mutations of the proopiomelanocortin gene (Pomc) cause obesity in humans and rodents, but the contributions of central versus pituitary POMC deficiency are not fully established. To elucidate these roles, we introduced a POMC transgene (Tg) that selectively restored peripheral melanocortin and corticosterone secretion in Pomc mice. Rather than improving energy balance, the genetic replacement of pituitary POMC in PomcTg mice aggravated their metabolic syndrome with increased caloric intake and feed efficiency, reduced oxygen consumption, increased subcutaneous, visceral, and hepatic fat, and severe insulin resistance. Pair-feeding of PomcTg mice to the daily intake of lean controls normalized their rate of weight gain but did not abolish obesity, indicating that hyperphagia is a major but not sole determinant of the phenotype. Replacement of corticosterone in the drinking water of Pomc mice recapitulated the hyperphagia, excess weight gain and fat accumulation, and hyperleptinemia characteristic of genetically rescued PomcTg mice. These data demonstrate that CNS POMC peptides play a critical role in energy homeostasis that is not substituted by peripheral POMC. Restoration of pituitary POMC expression to create a de facto neuronal POMC deficiency exacerbated the development of obesity, largely via glucocorticoid modulation of appetite, metabolism, and energy partitioning.
Collapse
Affiliation(s)
- James L Smart
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | | | | |
Collapse
|
234
|
Costa JL, Hochgeschwender U, Brennan M. The Role of Melanocyte-Stimulating Hormone in Insulin Resistance and Type 2 Diabetes Mellitus. ACTA ACUST UNITED AC 2006; 5:7-13. [PMID: 16396514 DOI: 10.2165/00024677-200605010-00002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
In humans, mice, and other mammals, the melanocortin system consists of four peptide hormones with a core amino acid sequence of histidine-phenylalanine-arginine-tryptophan and five melanocortin receptors. Both the melanocortin hormones and their receptors are produced in diverse tissues throughout the body. The ligand of primary interest for treatment of insulin resistance is alpha-melanocyte-stimulating hormone (alpha-MSH), which is derived, as are all melanocortins, from tissue-specific post-translational proteolytic processing of the pro-opiomelanocortin (POMC) precursor protein. Recent results have shown that alpha-MSH is the complement of leptin in the endocrine circuit, regulating bodyweight, food intake, and metabolic rate. alpha-MSH can decrease bodyweight, weight gain, and food intake in mice with diet-induced and genetic obesity. As obesity is a major risk factor for type 2 diabetes mellitus, it was reasonable to investigate the endocrine agents involved in obesity for their involvement in diabetes. alpha-MSH analogs have also been shown to affect blood glucose levels in some mouse models of obesity. For instance, the POMC null mouse is extremely sensitive to insulin in an insulin tolerance test, while being otherwise euglycemic. The results from rodent studies with alpha-MSH suggest reciprocal effects: alpha-MSH appears to increase sensitivity to insulin when present in the CNS, while alpha-MSH in the periphery is necessary for insulin resistance. Should these trends be validated in humans, alpha-MSH-based therapeutics specifically active in the CNS or peripheral circulation may be promising for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Jessica Lynn Costa
- Eleanor Roosevelt Institute at the University of Denver, Denver, Colorado, USA
| | | | | |
Collapse
|
235
|
Balthasar N, Dalgaard LT, Lee CE, Yu J, Funahashi H, Williams T, Ferreira M, Tang V, McGovern RA, Kenny CD, Christiansen LM, Edelstein E, Choi B, Boss O, Aschkenasi C, Zhang CY, Mountjoy K, Kishi T, Elmquist JK, Lowell BB. Divergence of melanocortin pathways in the control of food intake and energy expenditure. Cell 2005; 123:493-505. [PMID: 16269339 DOI: 10.1016/j.cell.2005.08.035] [Citation(s) in RCA: 824] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Revised: 07/19/2005] [Accepted: 08/18/2005] [Indexed: 12/16/2022]
Abstract
Activation of melanocortin-4-receptors (MC4Rs) reduces body fat stores by decreasing food intake and increasing energy expenditure. MC4Rs are expressed in multiple CNS sites, any number of which could mediate these effects. To identify the functionally relevant sites of MC4R expression, we generated a loxP-modified, null Mc4r allele (loxTB Mc4r) that can be reactivated by Cre-recombinase. Mice homozygous for the loxTB Mc4r allele do not express MC4Rs and are markedly obese. Restoration of MC4R expression in the paraventricular hypothalamus (PVH) and a subpopulation of amygdala neurons, using Sim1-Cre transgenic mice, prevented 60% of the obesity. Of note, increased food intake, typical of Mc4r null mice, was completely rescued while reduced energy expenditure was unaffected. These findings demonstrate that MC4Rs in the PVH and/or the amygdala control food intake but that MC4Rs elsewhere control energy expenditure. Disassociation of food intake and energy expenditure reveals unexpected divergence in melanocortin pathways controlling energy balance.
Collapse
Affiliation(s)
- Nina Balthasar
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, 99 Brookline Avenue, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Thompson MD, Burnham WM, Cole DEC. The G protein-coupled receptors: pharmacogenetics and disease. Crit Rev Clin Lab Sci 2005; 42:311-92. [PMID: 16281738 DOI: 10.1080/10408360591001895] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Genetic variation in G-protein coupled receptors (GPCRs) is associated with a wide spectrum of disease phenotypes and predispositions that are of special significance because they are the targets of therapeutic agents. Each variant provides an opportunity to understand receptor function that complements a plethora of available in vitro data elucidating the pharmacology of the GPCRs. For example, discrete portions of the proximal tail of the dopamine D1 receptor have been discovered, in vitro, that may be involved in desensitization, recycling and trafficking. Similar in vitro strategies have been used to elucidate naturally occurring GPCR mutations. Inactive, over-active or constitutively active receptors have been identified by changes in ligand binding, G-protein coupling, receptor desensitization and receptor recycling. Selected examples reviewed include those disorders resulting from mutations in rhodopsin, thyrotropin, luteinizing hormone, vasopressin and angiotensin receptors. By comparison, the recurrent pharmacogenetic variants are more likely to result in an altered predisposition to complex disease in the population. These common variants may affect receptor sequence without intrinsic phenotype change or spontaneous induction of disease and yet result in significant alteration in drug efficacy. These pharmacogenetic phenomena will be reviewed with respect to a limited sampling of GPCR systems including the orexin/hypocretin system, the beta2 adrenergic receptors, the cysteinyl leukotriene receptors and the calcium-sensing receptor. These developments will be discussed with respect to strategies for drug discovery that take into account the potential for the development of drugs targeted at mutated and wild-type proteins.
Collapse
Affiliation(s)
- Miles D Thompson
- Department of Laboratory Medicine and Pathobiology, Banting Institute, University of Toronto, ON, Canada.
| | | | | |
Collapse
|
237
|
Thorpe AJ, Cleary JP, Levine AS, Kotz CM. Centrally administered orexin A increases motivation for sweet pellets in rats. Psychopharmacology (Berl) 2005; 182:75-83. [PMID: 16075284 DOI: 10.1007/s00213-005-0040-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Accepted: 04/24/2005] [Indexed: 11/24/2022]
Abstract
RATIONALE Centrally administered orexin A induces both feeding and locomotion in rats. Thus, the feeding response following orexin A administration may be secondary to general increases in activity rather than a specific motivation to eat. OBJECTIVE The aim of the study is to determine whether orexin A increases the motivation to eat. METHODS The effect of orexin A (0, 31.25, 62.5, 125, 250, and 500 pmol) on breakpoint was determined in male Sprague-Dawley rats with rostro-lateral hypothalamic cannulae under a progressive ratio of five schedule (PR5). The effect of orexin A (0, 31.25, 125, and 500 pmol) on pressing rate under a fixed ratio (20) schedule was obtained to analyze the time course of orexin-A-induced pressing. The effect of 24-h food deprivation on breakpoint under PR5 and the effect of orexin A (125 pmol) on free feeding (sweet pellets) and on open-field locomotor activity (0, 100, 500, and 1,000 pmol) were also tested. RESULTS Orexin A significantly augmented free feeding of sweet pellets, open-field locomotor activity, rate of pressing (FR20 schedule), and breakpoint (PR5 schedule), although compared to 24-h deprivation, the effect of orexin A on breakpoint was mild. However, there was a differential dose response relationship and time course of stimulation between orexin A's effects on locomotion and lever pressing. CONCLUSION These data indicate that infusion of orexin A enhances free feeding by enhancing and possibly prolonging motivation to eat.
Collapse
Affiliation(s)
- A J Thorpe
- Department of Neuroscience, University of Minnesota, 1334 Eckles Avenue, Saint Paul, MN 55108, USA
| | | | | | | |
Collapse
|
238
|
Trevaskis JL, Butler AA. Double leptin and melanocortin-4 receptor gene mutations have an additive effect on fat mass and are associated with reduced effects of leptin on weight loss and food intake. Endocrinology 2005; 146:4257-65. [PMID: 15994342 DOI: 10.1210/en.2005-0492] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Melanocortin-4 receptors (MC4Rs) are involved in the regulation of food intake, sympathetic nervous activity, and adrenal and thyroid function by leptin. The role of MC4Rs in regulating energy balance by leptin was investigated using double heterozygote or homozygous leptin (Lep(ob)) and Mc4r gene mutant mice. Double heterozygous or homozygous mutants were generated by crossing MC4R knockout (Mc4r-/-) mice, backcrossed onto C57BL/6J, with B6.V-Lep(ob) mice. Energy expenditure was measured using indirect calorimetry. The effect of leptin on food intake, weight loss, insulin, and corticosterone was compared for Lep(ob)/Lep(ob)Mc4r-/- mice and Lep(ob)/Lep(ob) mice. Double heterozygous and homozygous mutants exhibited an additive effect on fat mass. The 2-fold increase in body weight associated with severe obesity of Lep(ob)/Lep(ob) mice was associated with a significantly higher 24 h total and resting energy expenditure. The effect of obesity on energy expenditure was attenuated by 50% in Lep(ob)/Lep(ob) Mc4r+/- and Lep(ob)/Lep(ob) Mc4r-/- mice. Loss of MC4Rs did not affect basal food intake of Lep(ob)/Lep(ob) mice but was associated with partial leptin resistance in terms of food intake and weight loss. Leptin suppression of insulin and corticosterone in Lep(ob)/Lep(ob) mice were not significantly affected by Mc4r genotype. These results suggest a complex interaction between the Lep and Mc4r genes in energy homeostasis and suggest that MC4Rs retain significant anti-obesity function in the obese leptin-deficient state. Increased adiposity with double mutations may involve a reduction in energy expenditure. MC4Rs might have a modest role in the regulation of energy balance by exogenously administered leptin, primarily effecting food intake.
Collapse
Affiliation(s)
- James L Trevaskis
- Neuropeptides Laboratory, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, Louisiana 70808, USA
| | | |
Collapse
|
239
|
Daniels D, Suzuki A, Shapiro E, Luo L, Yee DK, Fluharty SJ. Rattus norvegicus melanocortin 3 receptor: a corrected sequence. Peptides 2005; 26:1835-41. [PMID: 15985309 DOI: 10.1016/j.peptides.2004.11.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2004] [Accepted: 11/22/2004] [Indexed: 11/25/2022]
Abstract
Examination of the Rattus norvegicus genome reveals differences in the melanocortin 3 receptor (MC3R) compared with the published sequence (accession X70667). To clarify these differences, we used RT-PCR to clone MC3R from Sprague Dawley rats. These efforts revealed a sequence for the rat MC3R consistent with that predicted by the rat genome, but different from the published receptor by three amino acids, all of which were located in the predicted second transmembrane domain (TM2). Analysis of these residues revealed that TM2 of the rat MC3R is more homologous with other species than previously considered. The presently described sequence maps onto chromosome 3 of the rat genome, which shows highly conserved synteny with the mouse chromosome 2 and the human chromosome 20. Transient expression revealed high affinity binding of [125I]-NDP-MSH and a concentration-dependent cAMP response to the synthetic agonist MTII. These data both clarify the sequence of the MC3R and demonstrate the great utility of genomic information recently made available.
Collapse
Affiliation(s)
- Derek Daniels
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, 220E, Philadelphia, PA 19104, USA.
| | | | | | | | | | | |
Collapse
|
240
|
Irani BG, Haskell-Luevano C. Feeding effects of melanocortin ligands--a historical perspective. Peptides 2005; 26:1788-99. [PMID: 16046247 DOI: 10.1016/j.peptides.2004.11.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Accepted: 11/18/2004] [Indexed: 11/25/2022]
Abstract
The process of energy homeostasis is a highly regulated process involving interacting signals between a variety of anorexigenic and orexigenic peptides, proteins and signaling molecules. The melanocortin system is an important component of this complex regulatory network. Involvement of the melanocortin pathway in the control of food intake and body weight regulation has been studied extensively in the past two decades. Previous studies that involve central administration of melanocortin molecules and examination of molecules that effect food intake in melanocortin knockout (KO) mice (MC3R, MC4R, POMC, AGRP and NPY) have been examined. In this review, we have summarized feeding studies that have resulted in the recognition of the melanocortin system as a major contributor to the complex neuroendocrine system regulating energy homeostasis.
Collapse
Affiliation(s)
- Boman G Irani
- Department of Medicinal Chemistry, University of Florida, P.O. Box 100485, Gainesville, FL-32610, USA
| | | |
Collapse
|
241
|
Fan W, Voss-Andreae A, Cao WH, Morrison SF. Regulation of thermogenesis by the central melanocortin system. Peptides 2005; 26:1800-13. [PMID: 15979759 DOI: 10.1016/j.peptides.2004.11.033] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2004] [Accepted: 11/05/2004] [Indexed: 12/21/2022]
Abstract
Adaptive thermogenesis represents one of the important homeostatic mechanisms by which the body maintains appropriate levels of stored energy and its core temperature. Dysregulation of adaptive thermogenesis promotes obesity. The central melanocortin system, in particular the melanocortin 4 receptor (MC4R) signaling pathway, influences the regulation of every aspect of energy balance, including thermogenesis, and plays a critical role in energy homeostasis in both rodent and man. This review will outline our current understanding of adaptive thermogenesis, focusing on the role of the central melanocortin pathway in the regulation of thermogenesis.
Collapse
Affiliation(s)
- Wei Fan
- Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L474, Portland, OR 97239-3098, USA.
| | | | | | | |
Collapse
|
242
|
Schiöth HB, Haitina T, Ling MK, Ringholm A, Fredriksson R, Cerdá-Reverter JM, Klovins J. Evolutionary conservation of the structural, pharmacological, and genomic characteristics of the melanocortin receptor subtypes. Peptides 2005; 26:1886-900. [PMID: 15985310 DOI: 10.1016/j.peptides.2004.11.034] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2004] [Accepted: 11/21/2004] [Indexed: 11/21/2022]
Abstract
We have cloned melanocortin receptors (MCRs) from several species of fish. The MC4R and MC5R subtypes arose early in vertebrate evolution and their primary structure is remarkably conserved. Expression and pharmacological characterization of the MCRs in fish has revealed that they bind and respond to melanocortin peptides with high potency. Detailed characterization of the binding properties of the different subtypes suggests that MCRs in early vertebrates had preference for adrenocorticotropic hormone (ACTH) peptides, while the high sensitivity for the shorter proopiomelanocortin (POMC) products, such as the alpha-, beta-, and gamma-melanocyte-stimulating hormone (MSH), has appeared later, perhaps as the MCR subtypes gained more specialized functions. The MCR repertoire shows in general high similarities in their primary structures, while they are however not similar in terms of functional roles. The MCRs serve therefore as an interesting model family to understand the molecular mechanisms of how functions of the genes can diverge during evolution. In this review, we provide an overview of our recent studies on the cloning, expression, pharmacology, 3D modeling, and genomic studies of the MCRs in non-mammalian species.
Collapse
Affiliation(s)
- Helgi B Schiöth
- Department of Neuroscience, Uppsala University, Biomedical Centre, Box 593, SE75124 Uppsala, Sweden.
| | | | | | | | | | | | | |
Collapse
|
243
|
Della-Fera MA, Baile CA. Roles for melanocortins and leptin in adipose tissue apoptosis and fat deposition. Peptides 2005; 26:1782-7. [PMID: 16002187 DOI: 10.1016/j.peptides.2004.12.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2004] [Accepted: 12/12/2004] [Indexed: 10/25/2022]
Abstract
Leptin has a wide range of effects on physiological functions related to the regulation of body energy balance. Many of leptin's effects are mediated through neuropeptide-containing neurons and neuropeptide receptors in the hypothalamus. The melanocortin system includes both agonist (alpha-melanocyte stimulating hormone, alphaMSH) and antagonist peptides (agouti related peptide, AGRP). Increased melanocortin receptor stimulation following leptin administration plays an important role in leptin-induced hypophagia and increased sympathetic nervous system activity and is partly responsible for leptin-induced weight loss. However, melanocortins do not appear to mediate some of the more striking centrally-mediated effects of leptin on adipose tissue, including adipose tissue apoptosis, that lead to the extensive depletion of fat.
Collapse
Affiliation(s)
- Mary Anne Della-Fera
- Department of Animal and Dairy Science, University of Georgia, 444 Animal Science Complex, Athens, GA 30602-2771, USA
| | | |
Collapse
|
244
|
Chen Y, Snieder H, Wang X, Kaviya B, McCaffrey C, Spector TD, Carter ND, O'Dell SD. Proopiomelanocortin gene variants are associated with serum leptin and body fat in a normal female population. Eur J Hum Genet 2005; 13:772-80. [PMID: 15812563 DOI: 10.1038/sj.ejhg.5201407] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A major quantitative trait locus (QTL) determining leptin levels has been linked to the proopiomelanocortin (POMC) region on chromosome 2. Most studies, based on under 350 lean or obese subjects, have shown no association between POMC SNP 8246 C/T and serum leptin, but significant associations have been reported with RsaI 8246 C/T SNP haplotypes. We have investigated association of four POMC SNPs with body composition and serum leptin in 2758 normal Caucasian female subjects (mean age 47.4+/-12.5 years), from the St Thomas' UK Adult Twin Registry (Twins UK): RsaI and 51 G/C in the 5'UTR and 8246 C/T and 7965 C/T in the 3'UTR. Under the recessive model, the 8246 T allele (freq. 0.18) was significantly associated with higher mean BMI (P=0.032) and total fat (P=0.046, both after age adjustment). Significant associations were maintained in sib-TDT with waist (P=0.049), total fat (P=0.037) and emerged with serum leptin (P=0.016). Initial significant associations between RsaI (-) allele (freq. 0.30) and higher waist (P=0.04) or % central fat (P=0.02) were not maintained in sib-TDT. No significant associations were found between body composition or serum leptin and RsaI/8246 C/T haplotype and none with 51 G/C (freq. 0.01) or 7965 C/T (freq. 0.004). There was minimal pairwise LD between the four loci, apart from RsaI and 8246 C/T (D'=-0.78 (P<0.0001)). Associations of BMI, weight and total fat with SNPs in regions flanking the POMC gene in this powerful study suggest that regulation of POMC expression may be influential in determining body weight.
Collapse
Affiliation(s)
- Yuanneng Chen
- Department of Clinical Developmental Sciences, St George's Hospital Medical School, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
245
|
Tallam LS, Stec DE, Willis MA, da Silva AA, Hall JE. Melanocortin-4 receptor-deficient mice are not hypertensive or salt-sensitive despite obesity, hyperinsulinemia, and hyperleptinemia. Hypertension 2005; 46:326-32. [PMID: 16027245 DOI: 10.1161/01.hyp.0000175474.99326.bf] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The purpose of this study was to test whether the melanocortin-4 receptor (MC4R) is critical in the development of hypertension associated with obesity and its metabolic disorders. MC4R-deficient homozygous (-/-) and heterozygous (+/-) and wild-type (WT) C57BL/6J mice 17 to 19 weeks old (n=5 to 7 per group) were implanted with telemetry devices for monitoring 24-hour mean arterial pressure (MAP) and heart rate (HR). After 3-day stable control measurements on normal-salt diet (NSD; 0.4% NaCl), mice received a high-salt diet (HSD; 4% NaCl) for 7 days, followed by 3-day recovery on NSD. MC4R (-/-) mice were severely obese compared with MC4R (+/-) and WT mice (body weight 48+/-1.5 versus 31+/-0.6 and 30+/-0.5 g respectively). On NSD, MAP was similar in all groups of mice (MC4R (-/-) 110+/-3 mm Hg; MC4R (+/-) 109+/-2 mm Hg; WT 114+/-2 mm Hg), and HR in MC4R (-/-) was lower than in WT (604+/-5 versus 645+/-9 bpm; P<0.05) but not different from MC4R (+/-) (625+/-13 bpm) mice. HSD did not significantly alter MAP or HR in any of the groups. Epididymal and retroperitoneal fat weights and plasma leptin levels were several-fold greater in MC4R (-/-) compared with MC4R (+/-) and WT mice. Plasma insulin and glucose levels were also significantly greater in MC4R (-/-) than in MC4R (+/-) and WT mice. These data suggest that despite obesity, visceral adiposity, hyperleptinemia, and hyperinsulinemia, MC4R (-/-) mice are neither hypertensive nor salt sensitive, indicating that a functional MC4R may be necessary for the development of hypertension associated with obesity and its metabolic abnormalities.
Collapse
Affiliation(s)
- Lakshmi S Tallam
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216-4505, USA.
| | | | | | | | | |
Collapse
|
246
|
Tao YX. Molecular mechanisms of the neural melanocortin receptor dysfunction in severe early onset obesity. Mol Cell Endocrinol 2005; 239:1-14. [PMID: 15975705 DOI: 10.1016/j.mce.2005.04.012] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2005] [Revised: 04/10/2005] [Accepted: 04/20/2005] [Indexed: 11/23/2022]
Abstract
The neural melanocortin receptors, melanocortin-3 and -4 receptors (MC3R and MC4R), have been shown to regulate different aspects of energy homeostasis in rodents. Human genetic studies showed that mutations in the MC4R gene are the most common monogenic form of obesity. Functional analyses of the mutant receptors revealed multiple defects. A classification scheme is presented for cataloguing the ever-increasing array of MC4R mutations. Functional analysis of the only inactivating MC3R mutation is also summarized. Insights from the analyses of the naturally occurring mutations in the MC3R and MC4R on the structure and function of these receptors are highlighted.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849, USA.
| |
Collapse
|
247
|
Scott V, Kimura N, Stark JA, Luckman SM. Intravenous peptide YY3-36 and Y2 receptor antagonism in the rat: effects on feeding behaviour. J Neuroendocrinol 2005; 17:452-7. [PMID: 15946163 DOI: 10.1111/j.1365-2826.2005.01330.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Systemic injection of peptide YY3-36 reduces food intake in rodents and humans, although some groups have reported a lack of response. PYY3-36 is thought to act via the Y2 receptor to presynaptically inhibit the release of neuropeptide Y and GABA from hypothalamic arcuate neurones. Due to the controversy surrounding its action in rodents, we tested the peptide intravenously on feeding behaviour in rats and attempted to block its actions with the Y2 receptor antagonist BIIE0246. PYY3-36 significantly decreased food intake during the first hour in male Sprague-Dawley rats fasted overnight and then re-fed. BIIE0246 had no effect alone on re-feeding, but completely blocked the action of PYY3-36. In a second experiment of similar design, the behavioural satiety sequence (BSS) was studied. Normal rats eat, drink, explore and groom before entering rest. PYY3-36 significantly reduced food eaten maintaining the normal BSS, although shifting it to the left as expected for a natural satiety factor. The latency to rest occurred earlier for animals given PYY3-36 alone and PYY3-36 tended to increase the total time in rest compared with controls. These behavioural effects of PYY3-36 were blocked by BIIE0246, and BIIE0246 alone did not have an effect on the BSS. These results support the role of PYY3-36 as a natural satiety factor acting through Y2 receptors.
Collapse
Affiliation(s)
- V Scott
- Faculty of Life Science, University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
248
|
de Rijke CE, Jackson PJ, Garner KM, van Rozen RJ, Douglas NR, Kas MJH, Millhauser GL, Adan RAH. Functional analysis of the Ala67Thr polymorphism in agouti related protein associated with anorexia nervosa and leanness. Biochem Pharmacol 2005; 70:308-16. [PMID: 15927146 DOI: 10.1016/j.bcp.2005.04.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2005] [Revised: 04/21/2005] [Accepted: 04/22/2005] [Indexed: 11/19/2022]
Abstract
AgRP is a neuropeptide that stimulates food intake through inhibition of central melanocortin receptors (MCRs). In humans, the non-conservative amino acid substitution Alanine (Ala) 67 Threonine (Thr) has been associated with Anorexia Nervosa and with leanness. In the present study, the cellular distribution, processing and in vitro and in vivo activities of Ala67 and Thr67 AgRP were investigated. Western blots of media and lysates of BHK cells stably transfected with Ala67 or Thr67 expression constructs showed identical AgRP bands. Both Ala67 and Thr67 AgRP colocalised with the Golgi apparatus, but not with the ER or lysosomes when expressed in Att20 D16V cells. Also, no differences were observed between the potencies of bacterially expressed Ala67 and Thr67 AgRP to stimulate MC4R in a reporter gene assay or inhibit food intake in rats. Taken together, no evidence was found for a functional defect of Thr67 AgRP related to MC4R interactions.
Collapse
Affiliation(s)
- Corine E de Rijke
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
249
|
|
250
|
Myung CS, Kim BT, Choi SH, Song GY, Lee SY, Jahng JW. Role of neuropeptide Y and proopiomelanocortin in fluoxetine-induced anorexia. Arch Pharm Res 2005; 28:716-21. [PMID: 16042082 DOI: 10.1007/bf02969363] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Fluoxetine is an anorexic agent known to reduce food intake and weight gain. However, the molecular mechanism by which fluoxetine induces anorexia has not been well-established. We examined mRNA expression levels of neuropeptide Y (NPY) and proopiomelanocortin (POMC) in the brain regions of rats using RT-PCR and in situ hybridization techniques after 2 weeks of administering fluoxetine daily. Fluoxetine persistently suppressed food intake and weight gain during the experimental period. The pair-fed group confirmed that the reduction in body weight in the fluoxetine treated rats resulted primarily from decreased food intake. RT-PCR analyses showed that mRNA expression levels of both NPY and POMC were markedly reduced by fluoxetine treatment in all parts of the brain examined, including the hypothalamus. POMC mRNA in situ signals were significantly decreased, NPY levels tended to increase in the arcuate nucleus (ARC) of fluoxetine treated rats (compared to the vehicle controls). In the pair-fed group, NPY mRNA levels did not change, but the POMC levels decreased (compared with the vehicle controls). These results reveal that the chronic administration of fluoxetine decreases expression levels in both NPY and POMC in the brain, and suggests that fluoxetine-induced anorexia may not be mediated by changes in the ARC expression of either NPY or POMC. It is possible that a fluoxetine raised level of 5-HT play an inhibitory role in the orectic action caused by a reduced expression of ARC POMC (alpha-MSH).
Collapse
Affiliation(s)
- Chang-Seon Myung
- Laboratory in Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea.
| | | | | | | | | | | |
Collapse
|