Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Blackwell Publishing LtdOxford, UKOBRObesity Reviews1467-7881 2007 Queens Printer and Controller of HMSO; published with permission; Journal

compilation 2007 The International Association for the Study of Obesity? 200785561Review ArticleAnimal models of obesity J. Speakman et al.

obesity reviews

Animal models of obesity


J. Speakman, C. Hambly, S. Mitchell and E. Krl

Aberdeen Centre for Energy Regulation and Obesity, School of Biological Sciences, University of Aberdeen, Aberdeen, UK

Keywords: Obesity, genetics, transgenic, animal models.

Accepted 19 September 2006

Address for correspondence: J Speakman, Aberdeen Centre for Energy Regulation and Obesity, School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK. E-mail: j.speakman@abdn.ac.uk

OnlineOpen: This article is available free online at www.blackwell-synergy.com

obesity reviews (2007) 8 (Suppl. 1), 5561

Background
Obesity stems from a prolonged imbalance between the levels of energy intake and expenditure, with the resultant surplus being stored as body lipids. Our understanding of the regulation of food intake and the physiological basis of differences in energy expenditure is owed, in large part, to studies made on animals. Moreover, animal models have been a cornerstone of studies of environmental effects, such as epigenetics, responses to high-fat and low-calorie diets and the identication and development of pharmaceuticals for obesity treatment. This review provides some examples of the animal work that has been performed, and focuses on the variation in approaches that have been taken and their potential, rather than aiming to be a comprehensive summary.

The physiological and genetic basis of obesity Spontaneous single-gene loss-of-function mutations
The genetically obese ob/ob mouse is a classic case of a spontaneous single-gene loss-of-function mutation that generates massive obesity. Characterizing the genetic basis of this mutation revealed that the defect is a single base pair deletion, which results in a premature stop codon in a gene expressed almost exclusively in adipocytes. The gene product was called leptin (1). The gene is recessive, but

mice that are heterozygotic (i.e. with one copy only, rather than two) for the defect produce reduced amounts of leptin and are moderately overweight. There are now at least 10 known single-gene loss-of-function defects that cause massive obesity and have been completely genetically characterized. In all the cases where such defects have been discovered, they have initially resulted from spontaneous mutational events in large breeding establishments. Discovery of the defect in the rst place has been down to sharp-eyed observers noticing an abnormal obese phenotype (a phenotype describes the observable physical and biochemical characteristics of an individual) and then selectively breeding to expose the responsible gene in homozygotes (which have two copies of the gene). In some cases, the gap between discovery of the original phenotype and its subsequent genetic and physiological characterization has been very long, for example, in the case of leptin, about 50 years. These delays were largely down to the fact that, prior to the 1980s, the tools were not available to clone genes and sequence them. The development of highthroughput sequencing capability, and the completion of the mouse and rat genomes in the early part of the new millennium, means that such delays between the discovery of a new mutant and its characterization are likely to get progressively shorter. Nevertheless, there is an inherent randomness in the discovery of single-gene defects, which depends primarily on unusual animals being identied in routine colony checks. This means that genes with only minor effects on

This paper was commissioned by the Foresight programme of the Ofce of Science and Innovation, Department of Trade and Industry 2007 Queens Printer and Controller of HMSO; published with permission Journal compilation 2007 The International Association for the Study of Obesity. obesity reviews 8 (Suppl. 1), 5561

55

56

Animal models of obesity

J. Speakman et al.

obesity reviews

the heterozygote are unlikely to be discovered. This ability to only detect genetic defects that have major loss-offunction effects in heterozygotes means that, almost by denition, these defects will only affect a minor proportion of the population. Consequently, while the discovery of the ob/ob mouse and the leptin gene was a major leap forwards, genetic screens of the human population have revealed trivially small numbers of obese individuals that have loss-of-function mutations in this gene (2). The same is true for all the other genes that have been discovered as spontaneous single-gene defects and characterized genetically and functionally (3). The real progress that study of these genes allows is to further our understanding of how the energy regulation system works. In fact, many of the genes that appear important in single-gene mutation events seem to be involved in a common pathway that includes leptin and insulin as signalling molecules (4). Our knowledge of this pathway has been crucially informed by characterization studies of spontaneous single-gene loss-of-function defects.

Articially generated single-gene loss-of-function mutations


The arbitrary nature of relying on spontaneous mutational events resulting in major loss-of-function mutations of critical genes has led to attempts to accelerate the process by increasing the mutation rate articially. This is performed by treating animals with mutagenic chemicals, or exposing them to radiation (57). Several countries have programmes of research that include such articial mutagenesis studies (e.g. Gailus-Durner et al. (8)). The animals generated from these experiments may inform not only our understanding of energy regulation but many other aspects of animal function. The major problem with this approach is the cost of phenotyping. As the mouse genome consists of around 30 00040 000 genes and that a given mutation may only produce a loss-of-function effect in say 35% of cases, to discover the effect of loss of function in a given gene might require phenotyping over half a million animals. Just measuring body weight of animals on a single occasion in these sorts of numbers would be prohibitively expensive. Moreover, as with spontaneous genetic mutations, the effects still need to be present and of considerable effect in heterozygotes. Little progress has therefore been made from these studies in the context of understanding energy regulation. Transgenic models An enormous number of transgenic models with obese or lean phenotypes have been created since the characterization of the rst obesity genes (9,10). The 2005 update of the human obesity gene map cited 248 genes that, when

mutated or expressed as transgenes in the mouse, result in phenotypes that affect body weight and adiposity (3). With traditional transgenic technologies, there was little control over where or how many copies of genes were introduced into the genome. However, current sophisticated genetargeting strategies permit investigators to manipulate the genome in ways that essentially allow the introduction of virtually any desired change. Furthermore, advanced techniques allow alterations to the genome that act only at specic times, or that are expressed only in specic tissues or cell types (11). Over-expression of target genes was the rst widely used technique. The full-length coding sequence of the gene is cloned downstream of a promoter, which may provide global, or tissue-specic expression, resulting in transgenic offspring over-expressing the target gene. Although relatively straightforward and inexpensive, the level of gene and protein expression does not always generate a physiological effect. More predictable and reproducible than overexpression models are global-knockout models. Here, the phenotype is created through total ablation of the target gene in all tissues. These knockout models have often resulted in unpredicted effects of the target genes, which in some cases allow an unexpected insight into the action of the target. An example of an unexpected insight developed from a transgenic knockout is the axl mouse, originally developed to determine whether the tyrosine kinase receptor, axl, played a role in leukaemia, was found by to be associated with non-insulin-dependent diabetes mellitus. In addition to global and tissue-specic knockouts, transgenic work has also involved knock-in models, that is, replacing the endogenous gene with a mutated form. Knock-in mice have the ability to address more specic roles and can be used to determine the effects of subtle changes in protein structure or function. For example, the s/s mice created by Bates et al. were developed to investigate leptin signalling by introducing a knock-in mutation that disrupted the dominant intracellular signalling pathway through which leptin was believed to have its major action (the STAT3 pathway) (12). An obvious problem with conventional transgenic knockouts is the potential that manipulating the gene may result in early embryonic death. A less obvious problem is that a genetic manipulation that acts over the whole life may be compensated for during the period of development. So, what is normally a key gene in the process of energy regulation may appear to have little importance when it is knocked out because its action has been taken over by compensatory mechanisms. A case in point is the neuropeptide Y (NPY) gene. When introduced directly into the brain, this neuropeptide is one of the most potent stimulators of feeding behaviour. When NPY was knocked out, however, the resultant mouse had no obvious abnormal phenotype (1315).

This paper was commissioned by the Foresight programme of the Ofce of Science and Innovation, Department of Trade and Industry 2007 Queens Printer and Controller of HMSO; published with permission Journal compilation 2007 The International Association for the Study of Obesity. obesity reviews 8 (Suppl. 1), 5561

obesity reviews

Animal models of obesity

J. Speakman et al.

57

The Cre/loxP system is a tool for tissue-specic and timespecic knockout of target genes, which permits investigation of such genes. This system involves two separate transgenic lines, one expressing Cre recombinase (Cre) and the other in which Cre recombinase recognition (loxP) sites are strategically positioned at either side of the target gene. When Cre is expressed in mice harbouring a loxPcontaining target gene, the desired gene is excised. Depending on the tissue specicity and timing of recombinase expression, these modications can be restricted to certain cell types or developmental stages (16). An example of the Cre/lox system is its use to create mice with disruptions of the insulin receptor in fat tissue (the FIRKO mouse) (17). The FIRKO mouse has a low-fat mass, loss of the normal relationship between plasma leptin and body weight, and appears protected against age-related and hypothalamic lesion-induced obesity. Polygenic models It is clear that in most circumstances, obesity, and its related energetic precursors, are polygenic traits, resulting from the combined actions of many genes. In the light of this, several research programmes have turned to the study of the polygenic basis of obesity, and many of these have employed animal models. Although an animals phenotype is often easily measured, identifying the genes underlying this trait can be a laborious process. Compared with human studies, however, using animal models is often faster as enhanced techniques and some destructive methods (i.e. full body dissection) allow more accurate description of the phenotype. In addition, the power to detect quantitative trait loci (QTLs) using model animal species is improved because of the larger family size and formalized pedigree structure. Although several animal models have been used, such as rats (18), chickens (19) and pigs (20), the mouse is the most common genetic model species and recent advances in determining the mouse genetic map has driven their increased use. Currently, the molecular marker map for mouse consists of over 6500 PCR-based microsatellite markers (21). There are also a large number of different mouse populations, which offers a wide range of alternative genetic variances, and their short breeding cycle combined with a large litter size facilitates rapid proliferation of generations, which is necessary for inbreeding and selected line studies. In the search to locate regions of the genome responsible for polygenic obese phenotypes, QTL mapping is a valuable tool that screens an organisms genome for statistical associations between phenotypic and marker information. To date, over 200 obesity QTLs have been located in the mouse, although many of these are at the same chromosomal region and may therefore reect the same gene(s) (22). A key advantage of this approach is that the technique requires no prior knowledge concerning the biological

nature of the trait under examination (23). Obesity-related traits that have been divergently selected over many generations include body mass (2426), body composition (27,28), food intake (27), heat loss (29,30) and spontaneous activity (31). A detailed list of those where QTL mapping has occurred is available in Brockmann and Bevova (32).

Exotic models
In addition to more traditional rodent models of obesity, there have been several studies that have used more exotic animal models. These models generally involve wild animals that undergo natural patterns of variation in their fat mass, for example, seals (33,34) and bats (35,36). In practice, the usefulness of such models is probably constrained in two ways. First, the tools to explore the genetic basis of such effects are a considerable way from being developed. Second, these animals pose signicant challenges when it comes to establishing laboratory-based colonies that are amenable to investigation. There are two areas, however, where such exotic models are probably of far more utility and will probably provide signicant insights into aspects of the regulation of body fat storage. The rst include studies of non-human primates. The second involves studies of non-standard small rodents that undergo cycles of seasonally induced fat storage.

Non-human primates
Separation of the primate and rodent lineages is a relatively ancient event (6585 million years ago (37)). In contrast, the separation of the Hominoidea (humans and the other great apes) and the Cercopithecoidea (the Old World monkeys) occurred relatively recently (about 25 million years ago (38)). So, Old World monkeys (such as macaques, rhesus monkey and baboons) may provide a genetically more appropriate model for studying human obesity (39,40). It has been demonstrated that 1015% of captive macaque and rhesus monkeys develop age-related obesity when maintained on a relatively low-fat (10% of energy) ad libitum diet (41). Interestingly, the reduced locomotor activity arising from caging appears not to be a key factor contributing to obesity in monkeys.

Seasonal models of obesity


Many small mammals exhibit annual cycles of reproduction that are accompanied by variations in body mass and adiposity (4247). Many species (e.g. hamsters Phodopus sp. and Mesocricetus sp., voles Microtus sp. and lemmings Dicrostonyx sp.) rely on environmental cues such as increasing or decreasing day lengths to trigger these events (45,48,49). An advantage of this system is that the

This paper was commissioned by the Foresight programme of the Ofce of Science and Innovation, Department of Trade and Industry 2007 Queens Printer and Controller of HMSO; published with permission Journal compilation 2007 The International Association for the Study of Obesity. obesity reviews 8 (Suppl. 1), 5561

58

Animal models of obesity

J. Speakman et al.

obesity reviews

responses to photoperiod can be mimicked in the laboratory by acutely transferring animals between long-day (LD) and short-day (SD) photoperiods. This makes hamsters, voles and lemmings attractive study species for investigating the mechanisms underlying the regulation of body mass (45,50). Although these robust rhythms of adiposity occur in a variety of mammalian species, they have been studied most extensively in the Siberian or Djungarian hamster (Phodopus sungorus), Syrian or golden hamster (Mesocricetus auratus) and the collared lemming (Dicrostonyx groenlandicus). However, seasonal changes in adiposity linked to photoperiod have also been demonstrated in several other species (5155), which may also provide useful models. One species that has recently received considerable attention as a potential model of seasonal obesity is the eld vole (Microtus agrestis), which is a non-hibernating arvicoline rodent. Cold-acclimated adult male voles transferred from SD (8-h light) to LD (16-h light) undergo a substantial increase in adiposity (56), which depends on photoperiodtriggered leptin resistance (57).

Animal studies of environmental effects Exposure to high-fat and palatable diets


One hypothesis for the rapidity of the obesity epidemic is the possibility that our regulatory systems have become overwhelmed by high-fathigh-density palatable foods, which have become increasingly available in the past 20 30 years. In rodents, there have been many studies that have attempted to characterize the responses of animals exposed to high-fat diets. Two types of response have been observed some animals show profound increases in their body fatness (for example, the C57BL/6 mouse (58)) and have been termed diet-induced obese (DIO) rodents. Many studies have used this DIO paradigm to examine the regulation of food intake under conditions of high-fat intake as a model of human obesity. On the other hand, some rodent strains, termed dietary-resistant (DR) strains, and many wild rodents seem resistant to weight gain when given high-fat diets. In the outbred Sprague-Dawley rat, many studies have been performed examining the responses to a high-fat high-palatability diet (59,60). These outbred animals show a diversity of responses that include some animals showing DIO and others showing DR responses (59,61,62). Levin and colleagues have derived inbred lines from those most resistant and most susceptible to developing obesity (63) and have directed considerable effort to identifying those aspects of physiology (particularly in the brain) (64) and genetics (via QTL mapping) that may underlie the difference between these lines. This may give us insights into why some individuals develop obesity

and others do not when exposed to the same obesogenic environment. A major difference between these experimental designs and the situation in humans is that a large aspect of human susceptibility to obesity may not depend on the ability to resist weight gain when force-fed a high-fat diet; rather, it may hinge on individual differences in the propensity to choose high-fat foods in the rst place. Relatively few studies have explored this aspect of choice behaviour in animal models (65,66). Two different types of study have been made. These focus on the role of brain neuropeptides in macronutrient choice, while others studies have focused on peripheral aspects of the taste and olfaction system. This work has highlighted the fact that expression levels of certain neuropeptides may be linked to dietary intake (4,6771). In the eld of studies of taste preference, inbred strains show very large differences in their preference for sweet-tasting water, suggesting there may be signicant differences in individuals in their propensity to select different foods based on polymorphic variation in their taste receptors. Ultimately, this may be a much more rewarding avenue for investigation compared with the models where animals are force-fed high-fat diets without choice, as humans rarely face this latter scenario.

Epigenetic effects
A popular recent hypothesis concerning obesity is that our susceptibility may be programmed during the period of our lives that we spend in utero (72,73). By characterizing the health of adults in the Netherlands whose mothers had endured an enforced period of famine during the Second World War, compared with children born immediately prior to and following the famine, strong experimental support for the hypothesis has been gathered in humans (7476). (and see contribution by Barker). The in utero effects are also often called epigenetic effects, because they can be difcult to separate from genetic effects. This is one area where an obesity-related phenomenon has been rst discovered in humans rather than in animal models, but in spite of this, animal models provide a very valuable tool for the study of the mechanisms by which such epigenetic effects arise. Animals provide two very clear advantages. First, it would be ethically unacceptable to impose deliberate restriction on human foetuses. Moreover, the timescale of the effects would be impossible to study. Small mammals, however, which live a median of 2 3 years, provide an opportunity to overcome these problems. Many intervention studies have been made in rats and mice that involve changes in the intrauterine number of foetuses (77) or manipulations of foetal nutrition by ligation of the placenta (reviewed in Holemans et al. (78)). We can expect considerable progress during the next decade in this area.

This paper was commissioned by the Foresight programme of the Ofce of Science and Innovation, Department of Trade and Industry 2007 Queens Printer and Controller of HMSO; published with permission Journal compilation 2007 The International Association for the Study of Obesity. obesity reviews 8 (Suppl. 1), 5561

obesity reviews

Animal models of obesity

J. Speakman et al.

59

Responses to low-calorie and other weight-loss diets


Caloric restriction is the most frequently prescribed treatment for obesity. However, its success is limited. This is primarily because, to be effective, the restricted intake has to be maintained indenitely, which is often impossible to achieve. Moreover, during caloric restriction, there is a compensatory reaction to the restriction by adjustment of other components of the energy budget. These compensations oppose the energy decit and make it harder to sustain weigh loss. In humans, this lack of continued weight loss may be a primary factor leading to non-compliance with the regime. Relying on humans to self-report their food intake is fraught with errors and frequent underreporting, which places in question the accuracy of many dietary intervention studies (79). Animals are ideal to investigate factors associated with caloric restriction because their energy intake can be easily monitored under strict environmental conditions, and each aspect of the energy budget can be more easily quantied.

potential obesity pharmaceuticals, rodents will remain a crucial model. An example of a drug target that was identied in animals, leading to the development of a class of potential obesity therapeutics, which ultimately generated a useable drug that has just nished clinical trials, was the development of the cannabinoid receptor type 1 antagonist rimonabant.

Conclusion
The use of animal models to study phenomena that underlie obesity (genetic, physiological, epigenetic and environmental) as well as investigations of potential treatments in animals has provided an enormous amount of information that has had both direct and indirect impacts on our understanding of the condition. As is often the case, however, as knowledge expands in an area, it generally serves to document the extent of our ignorance. This is certainly the case in the study of energy regulation and the resultant modulations of energy storage. We are not yet at a stage where replacing animals with computer-based models of energy regulation, or focusing on cell-culture-based work could realistically overtake the use of live animals in experimental investigations of energy balance and obesity. Consequently, it is very unlikely that signicant reductions in the numbers of animals involved in obesity research could be envisaged in the near future. The use of animal models has therefore been and will continue to be the cornerstone of our understanding of the underlying physiological and genetic basis of energy regulation, taste and smell perception and food choice behaviour. Moreover, animal models will also continue to provide a vital test bed for putative pharmaceuticals and novel dietary intervention techniques that may impact on the epidemic.

Development of pharmaceuticals for the treatment of obesity


Powell highlighted the important role that knockout mice may play in the future prospective identication of putative pharmaceutical targets for drug development (80). He reviewed the phenotypes of 21 different types of knockout mice where the gene knocked out was a potential therapeutic target for obesity. These were compared with the phenotypes of mice treated with therapeutics designed for the same targets. Of the 21 obesity gene targets considered, 16 showed a close correspondence between the knockout phenotype and drug effect in mice and/or rats. This suggests that, as far as the evaluation of drug targets for obesity is concerned, compensatory developmental changes that are precipitated by the whole-life knockout do not normally prevent detection of the relevant phenotype. Importantly, it was also found that, where data were available, the knockout phenotypes mimicked not only the effects of therapeutics in rodents, but also the effects when relevant therapeutics targeting the same genes were delivered to humans. Transgenic mouse technology may therefore be a valuable tool to prospectively identify genes that regulate body fat in vivo, and then to develop anti-obesity therapeutics by targeting the human protein products of these genes. An important message from the review by Powell (80) is that not only does the transgenic mouse represent a valuable tool for therapeutic development, but also that therapeutics designed to interfere with levels of fat storage in rodents also generally have the same effects on fat storage in humans. As an identication and testing ground for

Conict of Interest Statement


No conict of interest was declared.

References
1. Zhang Y, Proenca R, Maffei M et al. Positional cloning of the mouse obese gene and its human homologue. Nature 1994; 372: 425432. 2. Farooqi IS, ORahilly S. Monogenic obesity in humans. Annu Rev Med 2005; 56: 443458. 3. Rankinen T, Zuberi A, Chagnon YC et al. The human obesity gene map: the 2005 update. Obesity 2006; 14: 529644. 4. Schwartz MW, Woods SC, Porte D Jr et al. Central nervous system control of food intake. Nature 2000; 404: 661671. 5. Russell LB, Russell WL. Frequency and nature of specic-locus mutations induced in female mice by radiations and chemicals: a review. Mutat Res 1992; 296: 107127. 6. Dhar M, Webb LS, Smith L et al. A novel ATPase on mouse chromosome 7 is a candidate gene for increased body fat. Physiol Genomics 2000; 4: 93100.

This paper was commissioned by the Foresight programme of the Ofce of Science and Innovation, Department of Trade and Industry 2007 Queens Printer and Controller of HMSO; published with permission Journal compilation 2007 The International Association for the Study of Obesity. obesity reviews 8 (Suppl. 1), 5561

60

Animal models of obesity

J. Speakman et al.

obesity reviews

7. Dhar MS, Sommardahl CS, Kirkland T et al. Mice heterozygous for Atp10c, a putative amphipath, represent a novel model of obesity and type 2 diabetes. J Nutr 2004; 134: 799805. 8. Gailus-Durner V, Fuchs H, Becker L et al. Introducing the german mouse clinic: open access platform for standardized phenotyping. Nat Methods 2005; 2: 403404. 9. Inui A. Transgenic approach to the study of body weight regulation. Pharmacol Rev 2000; 52: 3561. 10. Salton SR, Hahm S, Mizuno TM. Of mice and MEN: what transgenic models tell us about hypothalamic control of energy balance. Neuron 2000; 25: 265268. 11. Davey RA, Maclean HE. Current and future approaches using genetically modied mice in endocrine research. Am J Physiol Endocrinol Metab 2006. 12. Bates SH, Stearns WH, Dundon TA et al. STAT3 signalling is required for leptin regulation of energy balance but not reproduction. Nature 2003; 421: 856859. 13. Erickson JC, Clegg KE, Palmiter RD. Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y. Nature 1996a; 381: 415421. 14. Erickson JC, Hollopeter G, Palmiter RD. Attenuation of the obesity syndrome of Ob/Ob mice by the loss of neuropeptide Y. Science 1996b; 274: 17041707. 15. Palmiter RD, Erickson JC, Hollopeter G et al. Life without Neuropeptide Y. Recent Prog Horm Res 1998; 53: 163199. 16. Kuhn R, Torres RM. Cre/loxP recombination system and gene targeting. Methods Mol Biol 2002; 180: 175204. 17. Bluher M, Michael MD, Peroni OD et al. Adipose tissue selective insulin receptor knockout protects against obesity and obesity-related glucose intolerance. Dev Cell 2002; 3: 2538. 18. Watanabe TK, Okuno S, Oga K et al. Genetic dissection of OLETF, a rat model for non-insulin-dependent diabetes mellitus: quantitative trait locus analysis of (OLETF x BN) x OLETF. Genomics 1999; 58: 233239. 19. Jennen DG, Vereijken AL, Bovenhuis H et al. Conrmation of quantitative trait loci affecting fatness in chickens. Genet Sel Evol 2005; 37: 215228. 20. Andersson L, Haley CS, Ellegren H et al. Genetic mapping of quantitative trait loci for growth and fatness in pigs. Science 1994; 263: 17711774. 21. Dietrich WF, Miller J, Steen R et al. A comprehensive genetic map of the mouse genome. Nature 1996; 380: 149152. 22. Snyder EE, Walts B, Perusse L et al. The human obesity gene map: the 2003 update. Obes Res 2004; 12: 369439. 23. Fisler JS, Warden CH. Mapping of mouse obesity genes: a generic approach to a complex trait. J Nutr 1997; 127: 1909S 1916S. 24. White JM, Legates JE, Eisen EJ. Maternal effects among lines of mice selected for body weight. Genetics 1968; 60: 395 408. 25. Hastings IM, Bootland LH, Hill WG. The role of growth hormone in lines of mice divergently selected on body weight. Genet Res 1993; 61: 101106. 26. Bunger L, Hill WG. Inbred lines derived from long-term divergent selection on fat content and body weight. Mamm Genome 1999; 10: 645648. 27. Sharp G, Hill WG, Robertson A. Effects of selection on growth, body composition and food intake in mice. Genet Res 1984; 43: 7592. 28. Hastings IM, Hill WG. A Note on the effect of different selection criteria on carcass composition in mice. Anim Prod 1989; 48: 229233. 29. Nielsen MK, Freking BA, Jones LD et al. Divergent selection for heat loss in mice. II. Correlated responses in feed intake, body

mass, body composition and number born through fteen generations. J Anim Sci 1997; 75: 14691476. 30. Nielsen MK, Jones LD, Freking BA, Deshazer JA. Divergent Selection for heat loss in mice. I. Selection applied and direct response through fteen generations. J Anim Sci 1997; 75: 1461 1468. 31. Swallow JG, Koteja P, Carter PA, Garland T Jr. Food consumption and body composition in mice selected for high wheelrunning activity. J Comp Physiol B 2001; 171: 651659. 32. Brockmann GA, Bevova MR. Using mouse models to dissect the genetics of obesity. Trends Genet 2002; 18: 367376. 33. Ortiz RM, Noren DP, Litz B, Ortiz CL. A new perspective on adiposity in a naturally obese mammal. Am J Physiol Endocrinol Metab 2001; 281: E1347E1351. 34. Arnould JP, Morris MJ, Rawlins DR, Boyd IL. Variation in plasma leptin levels in response to fasting in antarctic fur seals (Arctocephalus gazella). J Comp Physiol B 2002; 172: 2734. 35. Widmaier EP, Gornstein ER, Hennessey JL et al. High plasma cholesterol, but low triglycerides and plaque-free arteries, in mexican free-tailed bats. Am J Physiol 1996; 271: R1101R1106. 36. Kronfeld-Schor N, Richardson C, Silvia BA et al. Dissociation of leptin secretion and adiposity during prehibernatory fattening in little brown bats. Am J Physiol Regul Integr Comp Physiol 2000; 279: R1277R1281. 37. Eizirik E, Murphy WJ, OBrien SJ. Molecular dating and biogeography of the early placental mammal radiation. J Hered 2001; 92: 212219. 38. Page SL, Goodman M. Catarrhine phylogeny: noncoding DNA evidence for a diphyletic origin of the mangabeys and for a humanchimpanzee clade. Mol Phylogenet Evol 2001; 18: 1425. 39. West DB, York B. Dietary fat, genetic predisposition, and obesity: lessons from animal models. Am J Clin Nutr 1998; 67: 505S512S. 40. Wagner JE, Kavanagh K, Ward GM et al. Old world nonhuman primate models of type 2 diabetes mellitus. Inst Lab Anim Resour J 2006; 47: 259271. 41. Kemnitz JW. Obesity in macaques: spontaneous and induced. Adv Vet Sci Comp Med 1984; 28: 81114. 42. Heldmaier G, Steinlechner S. Seasonal control of energy requirements for thermoregulation in the djungarian hamster (Phodopus sungorus), living in natural photoperiod. J Comp Physiol 1981; 142: 429437. 43. Stebbins LL. Overwintering activity of Peromyscus maniculatus, Clethrionomys gapperi, C. rutilus, Eutamias amoenus and Microtus pennsylvanicus. In: Merritt J (ed.). Winter Ecology of Small Mammals. Special Publication of Carnegie Museum Natural History, Vol. 10. Xxxx: Pittsburgh, 1984, pp. 201213. 44. Bartness TJ, Wade GN. Photoperiodic control of seasonal body weight cycles in hamsters. Neurosci Biobehav Rev 1985; 9: 599612. 45. Bartness TJ, Demas GE, Song CK. Seasonal changes in adiposity: the roles of photoperiod, melatonin and other hormones, and sympathetic nervous system. Exp Biol Med 2002; 227: 363 376. 46. Klingenspor M, Dickopp A, Heldmaier G, Klaus S. Short photoperiod reduces leptin gene expression in white and brown adipose tissue of djungarian hamsters. FEBS Lett 1996; 399: 290 294. 47. Drazen DL, Jasnow AM, Nelson RJ, Demas GE. Exposure to short days, but not short-term melatonin, enhances humoral immunity of male syrian hamsters (Mesocricetus auratus). J Pineal Res 2002; 33: 118124.

This paper was commissioned by the Foresight programme of the Ofce of Science and Innovation, Department of Trade and Industry 2007 Queens Printer and Controller of HMSO; published with permission Journal compilation 2007 The International Association for the Study of Obesity. obesity reviews 8 (Suppl. 1), 5561

obesity reviews

Animal models of obesity

J. Speakman et al.

61

48. Dark J, Zucker I, Wade GN. Photoperiodic regulation of body mass, food intake, and reproduction in the meadow vole (Microtus pennsylvanicus). Am J Physiol 1983; 245: R334R338. 49. Mrosovsky N. Cyclical obesity in hibernators: the search for the adjustable regulator. In: Hirsch J, Van Itallie TB (eds). Recent Advances in Obesity Research: IV. John Libbey & Co Ltd: London, 1983, pp. 4556. 50. Mercer JG, Speakman JR. Hypothalamic neuropeptide mechanisms for regulating energy balance: from rodent models to human obesity. Neurosci Biobehav Rev 2001; 25: 101116. 51. Canguilhem B, Vaultier J-P, Pevet P et al. Photoperiodic regulation of body mass, food intake, hibernation, and reproduction in intact and castrated european hamsters, Cricetus cricetus. J Comp Physiol A 1988; 163: 549557. 52. Dark J, Zucker I. Gonadal and photoperiodic control of seasonal body weight changes in male vole. Am J Physiol 1984; 247: R84R88. 53. Petterborg LJ. Effect of photoperiod on body weight in the vole, Microtus montanus. Can J Zool 1978; 56: 431435. 54. Wang D, Wang Z. Seasonal variations in thermogenesis and energy requirements of plateau pikas Ochotona curzoniae and root voles Microtus oeconomus. Acta Theriol 1996; 41: 225236. 55. Peacock WL, Krl E, Moar KM et al. Photoperiodic effects on body mass, energy balance and hypothalamic gene expression in the bank vole. J Exp Biol 2004; 207: 165177. 56. Krl E, Redman P, Thomson PJ et al. Effect of photoperiod on body mass, food intake and body composition in the eld vole, Microtus agrestis. J Exp Biol 2005; 208: 571584. 57. Krl E, Duncan JS, Redman P et al. Photoperiod regulates leptin sensitivity in eld voles, Microtus agrestis. J Comp Physiol B 2006; 176: 153163. 58. Surwit RS, Kuhn CM, Cochrane C et al. Diet-induced type II diabetes in C57BL/6J mice. Diabetes 1988; 37: 11631167. 59. Archer ZA, Rayner DV, Rozman J et al. Normal distribution of body weight gain in male sprague-dawley rats fed a high-energy diet. Obes Res 2003; 11: 13761383. 60. Levin BE, Dunn-Meynell AA. Defense of body weight depends on dietary composition and palatability in rats with diet-induced obesity. Am J Physiol Regul Integr Comp Physiol 2002; 282: R46 R54. 61. Levin BE, Keesey RE. Defense of differing body weight set points in diet-induced obese and resistant rats. Am J Physiol 1998; 274: R412R419. 62. Mercer JG, Archer ZA. Diet-induced obesity in the spraguedawley rat: dietary manipulations and their effect on hypothalamic neuropeptide energy balance systems. Biochem Soc Trans 2005; 33: 10681072. 63. Levin BE, Dunn-Meynell AA, Balkan B, Keesey RE. Selective breeding for diet-induced obesity and resistance in SpragueDawley rats. Am J Physiol Regul Integr Comp Physiol 1997; 273: R725R730.

64. Ricci MR, Levin BE. Ontogeny of diet-induced obesity in selectively bred sprague-dawley rats. Am J Physiol Regul Integr Comp Physiol 2003; 285: R610R618. 65. Smith BK, Kelly LA, Pina R et al. Preferential fat intake increases adiposity but not body weight in sprague-dawley rats. Appetite 1998; 31: 127139. 66. Smith BK, Andrews PK, West DB. Macronutrient diet selection in thirteen mouse strains. Am J Physiol Regul Integr Comp Physiol 2000; 278: R797R805. 67. Crawley JN. The role of galanin in feeding behavior. Neuropeptides 1999; 33: 369375. 68. Odorizzi M, Fernette B, Angel E et al. Galanin receptor antagonists decrease fat preference in brattleboro rat. Neuropharmacology 2002; 42: 134141. 69. Smith BK, York DA, Bray GA. Chronic cerebroventricular galanin does not induce sustained hyperphagia or obesity. Peptides 1994; 15: 12671272. 70. Smith BK, Berthoud HR, York DA, Bray GA. Differential effects of baseline macronutrient preferences on macronutrient selection after galanin, npy, and an overnight fast. Peptides 1997; 18: 207211. 71. Jhanwar-Uniyal M, Beck B, Jhanwar YS et al. Neuropeptide Y projection from arcuate nucleus to parvocellular division of paraventricular nucleus: specic relation to the ingestion of carbohydrate. Brain Res 1993; 631: 97106. 72. Barker DJB (ed.). Fetal and Infant Origins of Adult Disease. BMJ Publishing Group: London, 1992. 73. Barker DJP. Mothers, Babies, and Disease in Later Life. BMJ Publishing Group: London, 1994. 74. Ravelli ACJ, van der Meulen JHP, Michels RPJ et al. Glucose tolerance in adults after prenatal exposure to famine. Lancet 1998; 351: 173177. 75. Roseboom TJ, van der Meulen JH, Ravelli AC et al. Effects of prenatal exposure to the dutch famine on adult disease in later life: an overview. Mol Cell Endocrinol 2001; 185: 9398. 76. Painter RC, Roseboom TJ, Bossuyt PM et al. Adult mortality at age 57 after prenatal exposure to the dutch famine. Eur J Epidemiol 2005; 20: 673676. 77. Nagasawa H, Yanai R. Quantitative participation of placental mammotropic hormones in mammary development during pregnancy of mice. Endocrinol Jpn 1971; 18: 507510. 78. Holemans K, Aerts L, Van Assche FA. Fetal growth restriction and consequences for the offspring in animal models. J Soc Gynecol Invest 2003; 10: 392399. 79. Poppitt SD, Swann D, Black AE, Prentice AM. Assessment of selective under-reporting of food intake by both obese and nonobese women in a metabolic facility. Int J Obes Relat Metab Disord 1998; 4: 303311. 80. Powell DR. Obesity drugs and their targets: correlation of mouse knockout phenotypes with drug effects in vivo. Obes Rev 2006; 7: 89108.

This paper was commissioned by the Foresight programme of the Ofce of Science and Innovation, Department of Trade and Industry 2007 Queens Printer and Controller of HMSO; published with permission Journal compilation 2007 The International Association for the Study of Obesity. obesity reviews 8 (Suppl. 1), 5561

You might also like