Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-m42fx Total loading time: 0 Render date: 2024-07-20T00:25:46.729Z Has data issue: false hasContentIssue false

4 - OBESITY: NEW MECHANISMS AND TRANSLATIONAL PARADIGMS

Published online by Cambridge University Press:  04 April 2011

Gregory Gaich
Affiliation:
Eli Lilly & Co.
David E. Moller
Affiliation:
Eli Lilly & Co.
Bruce H. Littman
Affiliation:
Translational Medicine Associates
Rajesh Krishna
Affiliation:
Merck Research Laboratories
Get access

Summary

Introduction

Medical Need and History of Failure

From 1985 through 2005, the prevalence of obesity doubled in the United States. In 2005, only four states had obesity prevalence rates less than 20%, whereas 17 states had prevalence rates of at least 25%, with several exceeding 30% (Centers for Disease Control and Prevention's [CDC's] Behavioral Risk Factor Surveillance System [BRFSS]). Obesity is a growing health epidemic outside the United States as well. Conditions of overweight and obesity are the sixth most important risk factor contributing to the overall global disease burden. According to the International Obesity Task Force and the World Health Organization (WHO), globally more than 1.7 billion adults are overweight, including more than 312 million who are obese. The disease burden of excess adiposity contributes to increased risk of additional diseases, including hypertension, type 2 diabetes, atherosclerosis, osteoarthritis, and some cancers.

Lifestyle interventions are the first-line treatments for obesity, yet this precipitous increase in the prevalence of obesity has occurred despite many years of lifestyle intervention as the primary mode of therapy. The underlying reason may be that survival is dependent on avoiding a negative energy balance, and both eating and activity behaviors required to maintain a neutral or positive energy balance are regulated by hundreds of genes in scores of biochemical and neural pathways. The active regulation of energy balance of both energy intake and energy expenditure is extremely precise with the average adult maintaining energy balance to within 0.3% over several decades.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2011

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Haslam, DW, & James, WP. (2005). Obesity. Lancet. 366, 1197–1209.CrossRef
Rankinen, T, Zuberi, A, Chagnon, C, Weisnagel, SJ, Argyropoulos, G, Walts, B, et al. (2006). The human obesity gene map: The 2005 update. Obesity. 14, 529–644.CrossRefGoogle ScholarPubMed
Rosenbaum, M, Leibel, RL, & Hirsch, J. (1998). Obesity. N. Engl. J. Med. 338, 555.Google Scholar
Colman, E. (2005). Anorectics on trial: A half century of federal regulation of prescription appetite suppressants. Ann. Intern. Med. 143, 380–385.CrossRefGoogle ScholarPubMed
Department of Health and Human Services, Food and Drug Administration, Final rule declaring dietary supplements containing ephedrine alkaloids adulterated because they present an unreasonable risk, Federal Register, 21 CFR, Part 119, (2004).69, 6787–6854.
Li, Z, Maglione, M, Tu, W, Mojica, W, Arterburn, D, Shugarman, L, et al. (2005). Meta-analysis: Pharmacologic treatment of obesity. Ann. Intern. Med. 142, 532–546.CrossRefGoogle ScholarPubMed
Sims, EAH. (1976). Experimental obesity, dietary-induced thermogenesis, and their clinical implications. Clin. Endocrinol. Metab. 5, 377–395.CrossRefGoogle ScholarPubMed
Phelan, S, Roberts, M, Lang, W, Raynor, H, & Wing, R. (2006). Initial results of the LITE (Living Lean in a Toxic Environment) Study: Successful weight losers work harder than never overweight individuals to maintain a normal body weight. Obesity. 14(Suppl), A32.Google Scholar
Wing, RR, & Phelan, S. (2005). Long-term weight loss maintenance. Am. J. Clin. Nutr. 82(Suppl), 222S–225S.CrossRefGoogle ScholarPubMed
Heinrichs, SC. (2003). Nonexercise muscle tension and behavioral fidgeting are positively correlated with food availability/palatability and body weight in rats. Physiol. Behav. 79, 199–207.CrossRefGoogle ScholarPubMed
Bouchard, C, Tremblay, A, Despres, JP, Nadeau, A, Lupien, PJ, Theriault, G, et al. (1990). The response to long-term overfeeding in identical twins. N. Engl. J. Med. 322, 1477–1482.CrossRefGoogle ScholarPubMed
Levine, JA, Eberhardt, NL, & Jensen, MD. (1999). Role of nonexercise activity thermogenesis in resistance to fat gain in humans. Science. 283, 212–214.CrossRefGoogle ScholarPubMed
Moller, DE. (2001). New drug targets for type 2 diabetes and the metabolic syndrome. Nature. 414, 821–827.CrossRefGoogle ScholarPubMed
Zhang, Y, Proenca, R, Maffei, M, Barone, M, Leopold, L, & Friedman, JM. (1994). Positional cloning of the mouse obese gene and its human homologue. Nature. 372, 425–432.CrossRefGoogle ScholarPubMed
Montague, CT, Farooqi, IS, Whitehead, JP, Soos, MA, Rau, H, Wareham, NJ, et al. (1997). Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 387, 903–908.CrossRefGoogle ScholarPubMed
Morton, G, Cummings, D, Baskin, D, Barsh, G, & Schwartz, MW. (2006). Central nervous system control of food intake and body weight. Nature. 443, 289–295.CrossRefGoogle ScholarPubMed
Jobst, E, Enriori, P, Sinnayah, P, & Cowley, M. (2006). Hypothalamic regulatory pathways and potential obesity treatment targets. Endocrine. 29, 33–48.CrossRefGoogle ScholarPubMed
O'Rahilly, S, & Farooqi, I. (2006). Genetics of obesity. Phil. Trans. R. Soc. B. 361, 1095–1105.CrossRefGoogle ScholarPubMed
Luquet, S, Perez, F, Hnasko, T, & Palmiter, R. (2005). NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science. 310, 683–685.CrossRefGoogle ScholarPubMed
Rivera, G, Bocanegra-García, V, Galiano, S, Cirauqui, N, Ceras, J, Pérez, S, et al. (2008). Melanin-concentrating hormone receptor 1 antagonists: A new perspective for the pharmacologic treatment of obesity. Curr. Med. Chem. 15, 1025–1043.CrossRefGoogle ScholarPubMed
Qu, D, Ludwig, DS, Gammeltoft, S, Piper, M, Pelleymounter, MA, Cullen, MJ, et al. (1996). A role for melanin-concentrating hormone in the central regulation of feeding behaviour. Nature. 380, 243–247.CrossRefGoogle ScholarPubMed
Dryden, S, Wang, Q, Frankish, HM, Pickavance, L, & Williams, G. (1995). The serotonin (5-HT) antagonist methysergide increases neuropeptide Y (NPY) synthesis and secretion in the hypothalamus of the rat. Brain Res. 699, 12–18.CrossRefGoogle ScholarPubMed
Tecott, LH, Sun, LM, Akana, SF, Strack, AM, Lowenstein, DH, Dallman, MF, et al. (1995). Eating disorder and epilepsy in mice lacking 5-HT2c serotonin receptors. Nature. 374, 542–546.CrossRefGoogle ScholarPubMed
Kyrou, I, Valsamakis, G, & Tsigos, C. (2006). The endocannabinoid system as a target for the treatment of visceral obesity and metabolic syndrome. Ann. New York Acad. Sci. 1083, 270–305.CrossRefGoogle ScholarPubMed
Horvath, T. (2006). The unfolding cannabinoid story on energy homeostasis: Central or peripheral site of action. Int. J. Obesity. 30, S30–S32.CrossRefGoogle ScholarPubMed
Lam, TKT, Schwartz, GJ, & Rossetti, L. (2005). Hypothalamic sensing of fatty acids. Nat. Neurosci. 8, 579–584.CrossRefGoogle ScholarPubMed
Kusunoki, J, Kanatani, A, & Moller, DE. (2006). Modulation of fatty acid metabolism as a potential approach to the treatment of obesity and the metabolic syndrome. Endocrine. 29, 91–100.CrossRefGoogle ScholarPubMed
Minokoshi, Y, Alqular, T, Furukawa, N, Kim, Y-B, Lee, A, Xue, B, et al. (2004). AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature. 428, 569–574.CrossRefGoogle ScholarPubMed
Winder, W, & Hardie, D. (1999). AMP-activated protein kinase, a metabolic master switch: Possible roles in type 2 diabetes. Am. J. Physiol. 40, E1–E10.Google Scholar
Liddle, RA. (1997). Cholecystokinin cells. Ann. Rev. Physiol. 59, 221–242.CrossRefGoogle ScholarPubMed
Sherrill, R, Berman, J, Birkemo, L, Croom, D, Dezube, M, Ervin, G, et al. (2001). 1,4-Benzodiazepine peripheral cholecystokinin (CCK-A) receptor agonists. Bioorg. Med. Chem. Lett. 11, 1145–1148.CrossRefGoogle ScholarPubMed
Drucker, D. (2006). The biology of incretin hormones. Cell Metab. 3, 153–165.CrossRefGoogle ScholarPubMed
Stanley, S, Wynne, K, & Bloom, S. (2003). Gastrointestinal satiety signals III. Glucagon-like peptide I, oxyntomodulin, peptide YY and pancreatic polypeptide. Am. J. Physiol. 286, G693–G697.Google Scholar
Wynne, K, Park, A, Small, C, Patterson, M, Ellis, S, Murphy, K, et al. (2005). Subcutaneous oxyntomodulin reduces body weight in overweight and obese subjects. Diabetes. 54, 2390–2395.CrossRefGoogle ScholarPubMed
Renshaw, D, & Batterham, R. (2005). Peptide YY: A potential therapy for obesity. Curr. Drug Targets. 6, 171–179.CrossRefGoogle ScholarPubMed
Wortley, K, del Rincon, J-P, Murray, J, Garcia, K, Iida, K, Thorner, M, et al. (2006). Absence of ghrelin protects against early-onset obesity. J. Clin. Invest. 115, 3573–3578.CrossRefGoogle Scholar
Shearman, L, Wang, S, Helmling, S, Stribling, D, Mazur, P, Ge, L, et al. (2006). Ghrelin neutralization by a ribonucleic acid-SPM ameliorates obesity in diet-induced obese mice. Endocrinol. 147, 1517–1526.CrossRefGoogle ScholarPubMed
Yang, J, Brown, M, Liang, G, Grishin, N, & Goldstein, J. (2008). Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone. Cell. 132, 387–396.CrossRefGoogle ScholarPubMed
Gutierrez, J, Solenberg, P, Perkins, D, Willency, J, Knierman, M, Jin, Z, et al. (2008). Ghrelin octanoylation mediated by an orphan lipid transferase. Proc. Natl. Acad. Sci. U. S. A. 105, 6320–6325.CrossRefGoogle ScholarPubMed
Zhou, G, Myers, R, Li, Y, Chen, Y, Shen, X, Fenyk-Melody, J, et al. (2001). Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Invest. 108, 1167–1174.CrossRefGoogle ScholarPubMed
Chen, H, & Farese, R. (2000). DGAT and triglyceride synthesis: A new target for obesity treatment? Trends Cardiovasc. Med. 10, 188–192.CrossRefGoogle ScholarPubMed
Nawrocki, A, & Scherer, P. (2005). Keynote review: The adipocyte as a drug discovery target. Drug Discovery Today. 10, 1219–1230.CrossRefGoogle ScholarPubMed
Cohen, P, Miyazaki, M, Socci, N, Hagge-Greenberg, A, Liedtke, W, Soukas, A, et al. (2002). Role for stearoyl-CoA desaturase-1 in leptin-mediated weight loss. Science. 297, 240–243.CrossRefGoogle ScholarPubMed
Jiang, G, Li, Z, Liu, F, Ellsworth, K, Dallas-Yang, Q, Wu, M, et al. (2005). Prevention of obesity in mice by anti-sense oligonucleotide inhibitors of stearoyl-CoA desaturase-1. J. Clin. Invest. 115, 1030–1038.CrossRefGoogle Scholar
Lowell, BB, S-Susulic, V, Hamann, A, Lawitts, J, Himms-Hagen, J, Boyer, BB, et al. (1993). Development of obesity in transgenic mice after genetic ablation of brown adipose tissue. Nature. 366, 740–742.CrossRefGoogle ScholarPubMed
Lowell, B, & Spiegelman, B. (2000). Towards a molecular understanding of adaptive thermogenesis. Nature. 404, 652–660.CrossRefGoogle ScholarPubMed
Grujic, D, Susulic, VS, Harper, ME, Himms-Hagen, J, Cunningham, BA, Corkey, BE, et al. (1997). Beta3-adrenergic receptors on white and brown adipocytes mediate beta3-selective agonist-induced effects on energy expenditure, insulin secretion, and food intake. J. Biol. Chem. 272, 17686–17693.CrossRefGoogle Scholar
Walker, BR, & Seckl, JR. (2003). 11 β-hydroxysteroid dehydrogenase type 1 as a novel therapeutic target in metabolic and neurodegenerative disease. Expert Opin. Ther. Targets. 7, 771–783.CrossRefGoogle Scholar
Kolonin, M, Saha, P, Chan, L, Pasqualini, R, & Arap, W. (2004). Reversal of obesity by targeted ablation of adipose tissue. Nat. Med. 10, 625–632.CrossRefGoogle ScholarPubMed
Innes, JA, Watson, ML, Ford, MJ, Munro, JF, Stoddart, ME, & Campbell, DB. (1977). Plasma fenfluramine levels, weight loss, and side effects. Br. Med. J. 2, 1322–1325.CrossRefGoogle ScholarPubMed
Weintraub, M, Hasday, JD, Mushlin, AI, & Lockwood, DH. (1984). A double-blind clinical trial in weight control. Use of fenfluramine and phentermine alone and in combination. Arch. Intern. Med. 144, 1143–1148.CrossRefGoogle ScholarPubMed
Bray, GA, Blackburn, GL, Ferguson, JM, Greenway, FL, Jain, AK, Mendel, CM, et al. (1999). Sibutramine produces dose-related weight loss. Obes. Res. 7, 189–198.CrossRefGoogle ScholarPubMed
Addy, C, Wright, H, Laere, K, Gantz, I, & Erondu, N. (2008). The acyclic CB1R inverse agonist taranabant mediates weight loss by increasing energy expenditure and decreasing caloric intake. Cell Metab. 7, 68–78.CrossRefGoogle ScholarPubMed
Scheen, AJ. (2008). CB1 receptor blockade and its impact on cardiometabolic risk factors: Overview of the RIO programme with rimonabant. J. Neuroendocrinol. 20(Suppl 1), 139–146.CrossRefGoogle ScholarPubMed
Christensen, R, Kristensen, PK, Bartels, EM, Bliddal, H, & Astrup, A. (2007). Efficacy and safety of the weight-loss drug rimonabant: A meta-analysis of randomised trials. Lancet. 370, 1706–1713.CrossRefGoogle ScholarPubMed
Steiner, MA, Marsicano, G, Nestler, EJ, Holsboer, F, Lutz, B, & Wotjak, CT. (2008). Antidepressant-like behavioral effects of impaired cannabinoid receptor type 1 signaling coincide with exaggerated corticosterone secretion in mice. Psychoneuroendocrinol. 33, 54–67.CrossRefGoogle ScholarPubMed
Greenway, F, Fujioka, K, Anderson, J, Raj, YP, Gupta, A, O'Neil, P, et al. (2007). A double-blind, placebo-, bupropion-, naltrexone-controlled study of the efficacy and safety of 3 doses. Obesity. 15, A85.Google Scholar
Fujioka, K, Greenway, F, Cowley, M, Guttadauria, M, Robinson, J, Landbloom, R, et al. (2007). The 24-week experience with a combination sustained release product of zonisamide and bupropion: Evidence of an encouraging benefit:risk profile. Obesity. 15, A85.Google Scholar
Greenway, F, Fujioka, K, Gupta, A, Smith, S, Guttadauria, M, Tollefson, G, et al. (2007). A double-blind, placebo-, bupropion- and naltrexone-controlled study of the efficacy and safety of 3 doses of naltrexone-bupropion SR combination therapy in obesity: Effects on total and visceral adipose tissue and CV risk markers. Obesity. 15, A85.Google Scholar
Gadde, KM, Yonish, GM, Foust, MS, Tam, PY, & Najarian, T. (2006). A 24-week randomized controlled trial of VI-0521, a combination weight loss therapy, in obese adults. Obesity. 14, A17.Google Scholar
Erondu, N, Gantz, I, Musser, B, Suryawanshi, S, Mallick, M, Addy, C, et al. (2006). Neuropeptide Y5 receptor antagonism does not induce clinically meaningful weight loss in overweight and obese adults. Cell Metab. 4, 275–282.CrossRefGoogle Scholar
Batterham, RL, Cohen, MA, Ellis, SM, Le Roux, CW, Withers, DJ, Frost, GS, et al. (2003). Inhibition of food intake in obese subjects by peptide YY3–36. N. Engl. J. Med. 349, 941–948.CrossRefGoogle ScholarPubMed
Batterham, RL, Le Roux, CW, Cohen, MA, Park, AJ, Ellis, SM, Patterson, M, et al. (2003). Pancreatic polypeptide reduces appetite and food intake in humans. J. Clin. Endocrinol. Metab. 88, 3989–3992.CrossRefGoogle ScholarPubMed
Gantz, I, Erondu, N, Mallick, M, Musser, B, Krishna, R, Tanaka, W, et al. (2007). Efficacy and safety of intranasal peptide YY3–36 for weight reduction in obese adults. J. Clin. Endocrinol. Metab. 92, 1754–1757.CrossRefGoogle ScholarPubMed
Riddle, MC, Henry, RR, Poon, TH, Zhang, B, Mac, SM, Holcombe, JH, et al. (2006). Exenatide elicits sustained glycaemic control and progressive reduction of body weight in patients with type 2 diabetes inadequately controlled by sulphonylureas with or without metformin. Diabetes Metab. Res. Rev. 22, 483–491.CrossRefGoogle ScholarPubMed
Significant weight loss sustained in obese people treated with liraglutide for one year. (2008). Novo Nordisk. Available at: http://www.novonordisk.com
Souza, CJ, & Burkey, BF. (2001). Beta 3-adrenoceptor agonists as anti-diabetic and anti-obesity drugs in humans. Curr. Pharm. Des. 7, 1433–1449.CrossRefGoogle ScholarPubMed
Redman, LM, Jonge, L, Fang, X, Gamlin, B, Recker, D, Greenway, FL, et al. (2007). Lack of an effect of a novel beta3-adrenoceptor agonist, TAK-677, on energy metabolism in obese individuals: A double-blind, placebo-controlled randomized study. J. Clin. Endocrinol. Metab. 92, 527–531.CrossRefGoogle ScholarPubMed
Cohade, C, Mourtzikos, KA, & Wahl, RL. (2003). “USA-Fat”: Prevalence is related to ambient outdoor temperature-evaluation with 18F-FDG PET/CT. J. Nucl. Med. 44, 1267–1270.Google ScholarPubMed
Wilson, DM, Varvel, SA, Harloe, JP, Martin, BR, & Lichtman, AH. (2006). SR 141716 (rimonabant) precipitates withdrawal in marijuana-dependent mice. Pharmacol. Biochem. Behav. 85, 105–113.CrossRefGoogle ScholarPubMed
Verebey, K, Volavka, J, Mule, SJ, & Resnick, RB. (1976). Naltrexone: disposition, metabolism, and effects after acute and chronic dosing. Clin. Pharmacol. Ther. 20, 315–328.CrossRefGoogle ScholarPubMed
Fairchild, MD, Jenden, DJ, Mickey, MR, & Yale, C. (1980). EEG effects of hallucinogens and cannabinoids using sleep-waking behavior as baseline. Pharmacol. Biochem. Behav. 12, 99–105.CrossRefGoogle ScholarPubMed
Tschop, M, Smiley, DL, & Heiman, ML. (2000). Ghrelin induces adiposity in rodents. Nature. 407, 908–913.CrossRefGoogle ScholarPubMed
Cervera, A, Wajcberg, E, Sriwijitkamol, A, Fernandez, M, Zuo, P, Triplitt, C, et al. (2008). Mechanism of action of exenatide to reduce postprandial hyperglycemia in type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 294, E846–E852.CrossRefGoogle ScholarPubMed
Vedala, A, Wang, W, Neese, RA, Christiansen, MP, & Hellerstein, MK. (2006). Delayed secretory pathway contributions to VLDL-triglycerides from plasma NEFA, diet, and de novo lipogenesis in humans. J. Lipid Res. 47, 2562–2574.CrossRefGoogle ScholarPubMed
Ahnen, DJ, Guerciolini, R, Hauptman, J, Blotner, S, Woods, CJ, & Wargovich, MJ. (2007). Effect of orlistat on fecal fat, fecal biliary acids, and colonic cell proliferation in obese subjects. Gastroenterol. Hepatol. 5, 1291–1299.Google ScholarPubMed
Weyer, C, Tataranni, PA, Snitker, S, Danforth, EJ, & Ravussin, E. (1998). Increase in insulin action and fat oxidation after treatment with CL 316,243, a highly selective beta3-adrenoceptor agonist in humans. Diabetes Metab. Res. Rev. 47, 1555–1561.Google ScholarPubMed
Yeomans, MR, & Gray, RW. (1997). Effects of naltrexone on food intake and changes in subjective appetite during eating: Evidence for opioid involvement in the appetizer effect. Physiol. Behav. 62, 15–21.CrossRefGoogle ScholarPubMed
Chapelot, D, Marmonier, C, Thomas, F, & Hanotin, C. (2000). Modalities of the food intake-reducing effect of sibutramine in humans. Physiol. Behav. 68, 299–308.CrossRefGoogle ScholarPubMed
Rolls, BJ, Shide, DJ, Thorwart, ML, & Ulbrecht, JS. (1998). Sibutramine reduces food intake in non-dieting women with obesity. Obes. Res. 6, 1–11.CrossRefGoogle ScholarPubMed
Gregersen, NT, Flint, A, Bitz, C, Blundell, JE, Raben, A, & Astrup, A. (2008). Reproducibility and power of ad libitum energy intake assessed by repeated single meals. Am. J. Clin. Nutr. 87, 1277–1281.CrossRefGoogle ScholarPubMed
Atkinson, RL, Berke, LK, Drake, CR, Bibbs, ML, Williams, FL, & Kaiser, DL. (1985). Effects of long-term therapy with naltrexone on body weight in obesity. Clin. Pharmacol. Ther. 38, 419–422.CrossRefGoogle ScholarPubMed
Malcolm, R, O'Neil, PM, Sexauer, JD, Riddle, FE, Currey, HS, & Counts, C. (1985). A controlled trial of naltrexone in obese humans. Int. J. Obes. 9, 347–353.Google ScholarPubMed
Mitchell, JE, Morley, JE, Levine, AS, Hatsukami, D, Gannon, M, & Pfohl, D. (1987). High-dose naltrexone therapy and dietary counseling for obesity. Biol. Psychiatr. 22, 35–42.CrossRefGoogle ScholarPubMed
Goldstein, DJ, Rampey, AH, Enas, GG, Potvin, JH, Fludzinski, , & Levine, LR. (1994). Fluoxetine: A randomized clinical trial in the treatment of obesity. Int. J. Obes. 18, 129–135.Google ScholarPubMed
Wren, JA, Gossellin, J, & Sunderland, SJ. (2007). Dirlotapide: A review of its properties and role in the management of obesity in dogs. J. Vet. Pharmacol. Ther. 30(Suppl 1), 11–16.CrossRefGoogle ScholarPubMed
Lipworth, BJ. (1996). Clinical pharmacology of beta 3-adrenoceptors. Br. J. Clin. Pharmacol. 42, 291–300.CrossRefGoogle ScholarPubMed
Grosch, S, Niederberger, E, Lotsch, J, Skarke, C, & Geisslinger, G. (2001). A rapid screening method for a single nucleotide polymorphism (SNP) in the human MOR gene. Br. J. Clin. Pharmacol. 52, 711–714.CrossRefGoogle ScholarPubMed
Monteleone, P, Tortorella, A, Martiadis, V, Di Filippo, C, Canestrelli, B, & Maj, M. (2008). The cDNA 385C to A missense polymorphism of the endocannabinoid degrading enzyme fatty acid amide hydrolase (FAAH) is associated with overweight/obesity but not with binge eating disorder in overweight/obese women. Psychoneuroendocrinol. 33, 546–550.CrossRefGoogle Scholar
Teixeira, PJ, Going, SB, Sardinha, LB, & Lohman, TG. (2005). A review of psychosocial pre-treatment predictors of weight control. Obes. Rev. 6, 43–65.CrossRefGoogle ScholarPubMed
Astrup, A, Buemann, B, Gluud, C, Bennett, P, Tjur, T, & Christensen, N. (1995). Prognostic markers for diet-induced weight loss in obese women. Int. J. Obes. 19, 275–278.Google ScholarPubMed
Wadden, TA, Foster, GD, Wang, J, Pierson, RN, Yang, MU, Moreland, K, et al. (1992). Clinical correlates of short- and long-term weight loss. Am. J. Clin. Nutr. 56(1 Suppl), 271S–274S.CrossRefGoogle Scholar
Foster, GD, Wadden, TA, Swain, RM, Stunkard, AJ, Platte, P, & Vogt, RA. (1998). The Eating Inventory in obese women: Clinical correlates and relationship to weight loss. Int. J. Obes. 22, 778–785.CrossRefGoogle ScholarPubMed
Weintraub, M, Taves, DR, Hasday, JD, Mushlin, AI, & Lockwood, DH. (1981). Determinants of response to anorexiants. Clin. Pharmacol. Ther. 30, 528–533.CrossRefGoogle ScholarPubMed
Womble, LG, Williamson, DA, Greenway, FL, & Redmann, SM. (2001). Psychological and behavioral predictors of weight loss during drug treatment for obesity. Int. J. Obes. 25, 340–345.CrossRefGoogle ScholarPubMed
Shimazaki, T, Yoshimizu, T, & Chaki, S. (2006). Melanin-concentrating hormone MCH1 receptor antagonists: A potential new approach to the treatment of depression and anxiety disorders. CNS Drugs. 20, 801–811.CrossRefGoogle ScholarPubMed
Farooqi, I, Keogh, J, Yeo, G, Lank, E, Cheetham, T, & O'Rahilly, S. (2003). Clinical spectrum of obesity and mutations in the melanocortin 4 receptor. N. Engl. J. Med. 348, 1085–1095.CrossRefGoogle ScholarPubMed
Loos, R, & Bouchard, C. (2008). FTO: The first gene contributing to common forms of human obesity. Obes. Rev. 9, 246–250.CrossRefGoogle ScholarPubMed
Naggert, JK, Fricker, LD, Varlamov, O, Nishina, PM, Rouille, Y, Steiner, DF, et al. (1995). Hyperproinsulinaemia in obese fat/fat mice associated with a carboxypeptidase E mutation which reduces enzyme activity. Nat. Genet. 10, 135–142.CrossRefGoogle ScholarPubMed
Noben-Trauth, K, Naggert, JK, North, MA, & Nishina, PM. (1996). A candidate gene for the mouse mutation tubby. Nature. 380, 534–538.CrossRefGoogle ScholarPubMed
Ohki-Hamazaki, H, Watase, K, Yamamoto, K, Ogura, H, Yamano, M, Yamada, K, et al. (1997). Mice lacking bombesin receptor subtype-3 develop metabolic defects and obesity. Nature. 390, 165–169.CrossRefGoogle ScholarPubMed
Ladenheim, E, Hamilton, N, Behles, R, Bi, S, Hampton, L, Battey, J, et al. (2008). Factors contributing to obesity in bombesin receptor subtype-3-deficient mice. Endocrinol. 149, 971–978.CrossRefGoogle ScholarPubMed
Erlanson-Albertsson, C, & York, D. (1997). Enterostatin–a peptide regulating fat intake. Obes. Res. 5, 360–372.CrossRefGoogle ScholarPubMed
Doggrell, S. (2005). Neuromedin U–a new target in obesity. Expert Opin. Ther. Targets. 9, 875–877.CrossRefGoogle ScholarPubMed
Charon, C, Dupuy, F, Marie, V, & Bazin, R. (1995). Effect of the b-adrenoceptor agonist BRL-35135 on development of obesity in suckling Zucker (fa/fa) rats. Am. J. Physiol. 268, E1039–E1045.Google Scholar
Greenberg, AS, Egan, JJ, Wek, SA, Garty, NB, Blanchette-Mackie, EJ, & Londos, C. (1991). Perilipin, a major hormonally regulated adipocyte-specific phosphoprotein associated with the periphery of lipid storage droplets. J. Biol. Chem. 266, 11341–11346.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×