Hostname: page-component-8448b6f56d-tj2md Total loading time: 0 Render date: 2024-04-20T00:04:05.722Z Has data issue: false hasContentIssue false

Peripheral signalling involved in energy homeostasis control

Published online by Cambridge University Press:  23 November 2012

Andoni Lancha
Affiliation:
Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
Gema Frühbeck
Affiliation:
Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
Javier Gómez-Ambrosi*
Affiliation:
Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
*
*Corresponding author: Dr Javier Gómez-Ambrosi, fax +34 948 425652, email jagomez@unav.es
Rights & Permissions [Opens in a new window]

Abstract

The alarming prevalence of obesity has led to a better understanding of the molecular mechanisms controlling energy homeostasis. Regulation of energy intake and expenditure is more complex than previously thought, being influenced by signals from many peripheral tissues. In this sense, a wide variety of peripheral signals derived from different organs contributes to the regulation of body weight and energy expenditure. Besides the well-known role of insulin and adipokines, such as leptin and adiponectin, in the regulation of energy homeostasis, signals from other tissues not previously thought to play a role in body weight regulation have emerged in recent years. The role of fibroblast growth factor 21 (FGF21), insulin-like growth factor 1 (IGF-I), and sex hormone-binding globulin (SHBG) produced by the liver in the regulation of body weight and insulin sensitivity has been recently described. Moreover, molecules expressed by skeletal muscle such as myostatin have also been involved in adipose tissue regulation. Better known is the involvement of ghrelin, cholecystokinin, glucagon-like peptide 1 (GLP-1) and PYY3–36, produced by the gut, in energy homeostasis. Even the kidney, through the production of renin, appears to regulate body weight, with mice lacking this hormone exhibiting resistance to diet-induced obesity. In addition, the skeleton has recently emerged as an endocrine organ, with effects on body weight control and glucose homeostasis through the actions of bone-derived factors such as osteocalcin and osteopontin. The comprehension of these signals will help in a better understanding of the aetiopathology of obesity, contributing to the potential development of new therapeutic targets aimed at tackling excess body fat accumulation.

Type
Review Article
Copyright
Copyright © The Authors 2012

Introduction

The obesity epidemic poses one of the most serious public health challenges. The prevalence of obesity has increased three-fold over the past three to four decades(Reference Swinburn, Sacks and Hall1). More than half of all adults in Europe and the USA are overweight. Of these, one-third is already obese and figures are increasing(Reference Finucane, Stevens and Cowan2). Obesity is a serious medical problem because it increases the risk of type 2 diabetes mellitus (T2DM), CVD, fatty liver, sleep-breathing disorders, and certain forms of cancer, among others(Reference Kahn, Hull and Utzschneider3Reference Fabbrini, Sullivan and Klein8). Moreover, obesity adversely affects the quality of life and shortens life expectancy(Reference Pischon, Boeing and Hoffmann9, Reference Berrington de Gonzalez, Hartge and Cerhan10).

The alarming prevalence of obesity has led to a greater understanding of the molecular mechanisms that regulate energy homeostasis and body weight control(Reference O'Rahilly11). Energy balance regulation is an extremely complex process that integrates multiple interacting homeostatic and behavioural pathways. In recent years, awareness has been raised regarding the increasing number of neuropeptides involved in the hypothalamic integration of peripheral signals derived mainly from the pancreas and adipose tissue(Reference Woods12). The existing evidence collected over recent years through targeted expression or knockout of specific genes involved in the pathways controlling energy intake, energy expenditure, adiposity or fat distribution has contributed to disentangling the mechanisms controlling energy homeostasis(Reference Frühbeck and Gómez-Ambrosi13). Thus, regulation of energy intake and expenditure is more complex than previously thought, being influenced by signals from many other peripheral tissues(Reference Takahashi, Li and Hua14Reference Somogyi, Gyorffy and Scalise18). In this sense, a wide variety of peripheral signals derived from different organs contributes to the regulation of body weight and energy expenditure. In addition to the well-known role of insulin (from the pancreas) and adipokines, such as leptin and adiponectin (from adipose tissue), in the regulation of energy homeostasis, signals from other tissues not previously thought to be involved in body weight regulation have emerged in recent years. The role of emerging proteins produced by the liver such as fibroblast growth factor (FGF) 21 in the regulation of body weight and insulin sensitivity has been described recently. Moreover, molecules expressed by skeletal muscle such as myostatin have also been involved in adipose tissue regulation. Better known is the involvement of ghrelin, cholecystokinin (CCK), glucagon-like peptide (GLP)-1 and peptide YY (PYY)3–36, produced by the gut, in energy homeostasis(Reference Field, Chaudhri and Bloom19). Even the kidney, through the production of renin, appears to regulate body weight, because mice lacking this hormone exhibit resistance to diet-induced obesity(Reference Takahashi, Li and Hua14). In addition, the skeleton has recently emerged as an endocrine organ, with effects on body weight control and glucose homeostasis through the actions of bone-derived factors such as osteocalcin and osteopontin(Reference Gómez-Ambrosi, Rodríguez and Catalán15). The cross-talk between adipose tissue and the skeleton constitutes a homeostatic feedback system, with adipokines and molecules secreted by osteoblasts and osteoclasts representing the links of an active bone–adipose tissue axis. The present review summarises the current state of knowledge of the peripherals signals involved in the regulation of energy intake and expenditure.

Signals from adipose tissue

The concept that circulating signals secreted in proportion to body fat stores regulate energy intake and expenditure in a coordinated manner to regulate body weight was proposed more than 50 years ago(Reference Kennedy20). According to this model, changes in energy balance sufficient to alter body fat stores were signalled via one or more circulating factors acting in the hypothalamus for triggering compensatory changes in order to match energy intake to energy expenditure(Reference Frühbeck and Gómez Ambrosi21). This was formulated as the ‘lipostatic theory’, presuming that as adipose tissue mass expands, a factor that operates as a sensing hormone or ‘lipostat’ in a negative feedback control from adipose tissue to hypothalamic receptors informs the brain about the size of the body fat stores, thereby allowing feeding behaviour and energy expenditure to be coupled to the nutritional state of the organism. Current knowledge has fostered the idea of a far more complex system than initially thought from the formulation of the lipostatic theory, involving the integration of a multitude of factors(Reference Frühbeck and Gómez Ambrosi21). This pleiotropic nature relies on the ability of adipocytes to secrete a large number of hormones, growth factors, enzymes, cytokines and matrix proteins, collectively termed as adipokines, which warrant an appropriate feedback response to changes in adipose tissue mass(Reference Frühbeck and Salvador22Reference Catalán, Gómez-Ambrosi and Rodríguez25).

Leptin

Leptin is an adipokine mainly produced by adipocytes in proportion to fat stores(Reference Zhang, Proenca and Maffei26). It was originally thought to be only involved in energy intake inhibition and body weight regulation acting at its hypothalamic receptors. This inhibition of appetite takes place mainly through the inhibition of neuropeptide Y (NPY)- and Agouti-related peptide (AgRP)-expressing neurons and the stimulation of pro-opiomelanocortin (POMC)-expressing neurons in the hypothalamic arcuate nucleus(Reference Morton, Cummings and Baskin27). However, a significant number of leptin receptor-expressing neurons lie outside the hypothalamic arcuate nucleus, suggesting that other brain regions known to modulate energy balance are involved in leptin's anorectic effect(Reference Myers, Munzberg and Leinninger28). Although food intake regulation is a major role of leptin, its receptor is expressed in almost all tissues(Reference Tartaglia, Dembski and Weng29Reference Frühbeck, Gómez-Ambrosi and Martínez31), underlining a high functional pleiotropism involving energy homeostasis, glucose metabolism, reproduction, angiogenesis, immunity, gastrointestinal function, wound healing, bone remodelling and cardiovascular function(Reference Frühbeck, Jebb and Prentice32Reference Gautron and Elmquist38). Plasma leptin concentrations are increased in obese patients, being strongly correlated with BMI and the percentage of body fat, as well as with leptin mRNA expression in adipose tissue(Reference Frühbeck, Gómez-Ambrosi and Muruzábal23, Reference Gómez-Ambrosi and Frühbeck39, Reference Frühbeck40). The failure of high leptin concentrations to suppress feeding and mediate weight loss in common obesity defines what has been termed leptin resistance(Reference Myers, Cowley and Münzberg36).

Leptin-deficient ob/ob mice and leptin receptor-deficient db/db mutants show early-onset obesity, diabetes and reduced energy expenditure(Reference Lindström41). Leptin administration induces a dramatic loss of adipose mass in rodents(Reference Pelleymounter, Cullen and Baker42, Reference Halaas, Gajiwala and Maffei43). This effect is not only mediated by a reduction in energy intake, but also by a direct effect on adipose tissue(Reference Halaas, Gajiwala and Maffei43). In this sense, leptin inhibits lipogenesis and stimulates lipolysis in adipocytes through a direct effect(Reference Siegrist-Kaiser, Pauli and Juge-Aubry44, Reference Frühbeck, Aguado and Gómez-Ambrosi45) or a centrally mediated action(Reference Gallardo, Bonzón-Kulichenko and Fernández-Agulloó46) without the release of NEFA, which are intracellularly oxidised(Reference Wang, Lee and Unger47). Furthermore, leptin replacement in leptin-deficient mice increases energy expenditure(Reference Pelleymounter, Cullen and Baker42, Reference Hwa, Fawzi and Graziano48). A few leptin-deficient patients who also exhibit severe early-onset obesity and hormonal alterations have been identified(Reference Montague, Farooqi and Whitehead49, Reference Strobel, Issad and Camoin50). Surprisingly, BMR and total energy expenditure were similar to those of age-, sex- and weight-matched controls(Reference Farooqi, Jebb and Langmack51). Leptin replacement in humans reduces body weight mainly at the expense of the fat compartment and reverses metabolic and hormonal alterations(Reference Farooqi, Jebb and Langmack51Reference Paz-Filho, Wong and Licinio54). Interestingly, the weight loss-associated decrease in energy expenditure that takes places after prolonged negative energy balance was less pronounced in leptin-treated patients than in obese leptin-replete controls following a weight-loss programme(Reference Farooqi, Jebb and Langmack51, Reference Farooqi, Matarese and Lord52, Reference Rosenbaum, Goldsmith and Bloomfield55, Reference Galgani, Greenway and Caglayan56). Therefore, leptin prevents the reduction in metabolic rate that is associated with weight loss(Reference Farooqi, Matarese and Lord52, Reference Rosenbaum, Goldsmith and Bloomfield55, Reference Galgani, Greenway and Caglayan56).

Adiponectin

The adipokine adiponectin is highly expressed in adipose tissue and its circulating concentrations are considerably high, accounting for approximately 0·01 % of total serum protein(Reference Kadowaki and Yamauchi57Reference Kadowaki, Yamauchi and Kubota59). Adiponectin exerts a wide variety of physiological roles through the binding of at least three different receptors called AdipoR1, AdipoR2 and T-cadherin(Reference Yamauchi, Kamon and Ito60, Reference Hug, Wang and Ahmad61). Serum concentrations of adiponectin are decreased in obese subjects(Reference Arita, Kihara and Ouchi62, Reference Weyer, Funahashi and Tanaka63) and increase with weight loss(Reference Yang, Lee and Funahashi64). Hypoadiponectinaemia is associated with insulin resistance and patients with T2DM are reported to have decreased concentrations of adiponectin(Reference Weyer, Funahashi and Tanaka63, Reference Hotta, Funahashi and Arita65, Reference Spranger, Kroke and Mohlig66). Patients with CVD also exhibit lower adiponectin concentrations(Reference Ouchi, Kihara and Arita67). In addition, administration of adiponectin induces glucose-lowering effects and improves insulin sensitivity in rodent models of obesity-associated diabetes(Reference Berg, Combs and Du68, Reference Combs, Wagner and Berger69). Moreover, adiponectin also exhibits anti-atherosclerotic properties(Reference Matsuzawa, Funahashi and Kihara70). Adiponectin-deficient mice as well as mice lacking adiponectin receptors confirm the protective effects of this adipokine in the development of insulin resistance and atherosclerosis(Reference Maeda, Shimomura and Kishida71Reference Yamauchi, Nio and Maki73).

Although the insulin-sensitising effects of adiponectin are clear, the effects of this adipokine in energy intake and energy expenditure regulation are still a matter of controversy(Reference Dridi and Taouis74). A lack of effect of intracerebroventricularly administered adiponectin on energy intake has been described. At the same time, weight-reducing actions through an increase in energy expenditure in normal mice have been shown(Reference Qi, Takahashi and Hileman75). These observations are confirmed in the long term after a 4-week treatment(Reference Park, Kim and Kwon76). However, other authors have reported that adiponectin increases energy intake through the stimulation of AMP-activated protein kinase (AMPK) in the hypothalamic arcuate nucleus via AdipoR1 with a concomitant decrease in oxygen consumption(Reference Kubota, Yano and Kubota77). Adiponectin- or AdipoR1-deficient mice show no changes in energy intake under a normal diet(Reference Maeda, Shimomura and Kishida71, Reference Yamauchi, Nio and Maki73, Reference Frühbeck, Alonso and Marzo78) but, when challenged with a high-fat diet, adiponectin knock-out mice show a reduced energy intake accompanied by an increase in oxygen consumption. The absence of lower food consumption in mice lacking adiponectin under a normal diet can be explained by a compensatory increase in the orexigenic pathways in order to avoid excessive weight loss. This reduction in food intake could be detrimental given the increase in energy expenditure observed after adiponectin administration(Reference Kubota, Yano and Kubota77).

Resistin

Resistin is another adipokine which was initially proposed as a link between increased adiposity and T2DM(Reference Steppan, Bailey and Bhat79). Resistin has been reported to be highly expressed in adipose tissue in mice. Under physiological circumstances, resistin appears to oppose insulin action in adipocytes and to impair glucose tolerance and insulin sensitivity in mice. Moreover, insulin-stimulated glucose uptake by adipocytes is enhanced by resistin neutralisation and is reduced by resistin treatment(Reference Steppan, Bailey and Bhat79, Reference McTernan, Kusminski and Kumar80). Different genetic and dietary models of rodent obesity exhibit increased serum concentrations of resistin(Reference Steppan, Bailey and Bhat79, Reference Gómez-Ambrosi and Frühbeck81). Moreover, transgenic overexpression of resistin leads to insulin resistance in mice(Reference Rangwala, Rich and Rhoades82) and rats(Reference Satoh, Nguyen and Miles83). In this sense, ob/ob mice simultaneously lacking resistin exhibit an improved glucose tolerance and insulin sensitivity(Reference Qi, Nie and Lee84).

The exact role of resistin in human physiology and whether or not resistin is involved in the development of insulin resistance still need to be completely elucidated(Reference McTernan, Kusminski and Kumar80, Reference Kusminski, McTernan and Kumar85, Reference Schwartz and Lazar86). Several groups have described increased concentrations of resistin in obesity(Reference Azuma, Katsukawa and Oguchi87, Reference Degawa-Yamauchi, Bovenkerk and Juliar88), while others report no existing differences(Reference Silha, Krsek and Skrha89Reference Heilbronn, Rood and Janderova91). However, cross-sectional and prospective epidemiological investigations reveal that human resistin is not significantly involved in the development of insulin resistance(Reference Fehmann and Heyn92Reference Heidemann, Sun and van Dam94) or the metabolic syndrome(Reference Utzschneider, Carr and Tong95). Rather than a role in the development of insulin resistance, considerable evidence links resistin to inflammation(Reference Gómez-Ambrosi and Frühbeck81, Reference Kusminski, McTernan and Kumar85, Reference Gómez-Ambrosi and Frühbeck96). In this respect, it has been reported that resistin markedly up-regulates pro-inflammatory cytokines(Reference Bokarewa, Nagaev and Dahlberg97, Reference Silswal, Singh and Aruna98) and that inflammation up-regulates resistin in human macrophages(Reference Kaser, Kaser and Sandhofer99, Reference Lehrke, Reilly and Millington100). Furthermore, it has been confirmed that resistin is related to markers of inflammation, being a predictive factor of atherosclerosis(Reference Reilly, Lehrke and Wolfe101), and that it also influences pro-inflammatory cytokine release from human adipocytes(Reference Kusminski, da Silva and Creely102). However, the precise physiological role of resistin in the pathogenesis and perpetuation of inflammation and CVD in humans remains unclear(Reference Gómez-Ambrosi and Frühbeck81).

Central administration of resistin induces a short-term decrease in food intake in rats(Reference Tovar, Nogueiras and Tung103Reference Cifani, Durocher and Pathak105), mainly associated with a suppression of the normal fasting-induced increase in the orexigenic neuropeptides NPY and AgRP, and a suppression of the decrease in cocaine and amphetamine-regulated transcript (CART)(Reference Vázquez, González and Varela106). However, other authors have described an up-regulation of hypothalamic NPY after intracerebroventricular administration of resistin in mice without measuring the potential effect on food intake(Reference Singhal, Lazar and Ahima107). Furthermore, leptin-deficient ob/ob mice lacking resistin exhibit higher body weight and body adiposity than ob/ob mice with normal resistin levels(Reference Qi, Nie and Lee84). This observation has been attributed to a lower metabolic rate, uncovering a crucial role of resistin in the regulation of thermogenesis. In addition to its inhibitory effect on adipogenesis(Reference Kim, Lee and Moon108), the anorexigenic and thermogenic effects of resistin underscore the relevance of this molecule as an interesting regulator of energy homeostasis. It still remains to be elucidated whether resistin acts on the brain or peripheral tissues to control metabolic rate. The complex actions of resistin on energy expenditure and the expression of resistin in several hypothalamic nuclei suggest that resistin may act in an autocrine/paracrine fashion for regulating body weight, deserving further research(Reference Nogueiras, Novelle and Vazquez109).

Acylation-stimulating protein

Acylation-stimulating protein (ASP) is an adipokine predominantly produced by fully differentiated adipocytes(Reference Maslowska, Sniderman and Germinario110). ASP results from the cleavage of the complement C3 via factor B and adipsin (factor D) interaction producing C3a, which is rapidly desarginated to give ASP (also known as C3adesArg)(Reference Cianflone, Xia and Chen111). ASP increases after a fat-containing meal and stimulates TAG synthesis and storage(Reference Frühbeck, Gómez-Ambrosi and Muruzábal23, Reference Yasruel, Cianflone and Sniderman112). ASP also stimulates translocation of GLUT1 to the cell surface(Reference Germinario, Sniderman and Manuel113). Differences between adipose tissue depots have been observed, with greater ASP binding and action in subcutaneous compared with omental fat, in females compared with males, and in obese compared with non-obese subjects, suggesting that ASP could be a female lipogenic factor(Reference Saleh, Al-Wardy and Farhan114). Circulating concentrations of ASP have been reported to be increased in obese and T2DM subjects(Reference Cianflone, Xia and Chen111).

Mice lacking complement C3 are deficient in ASP. These mice exhibit a striking phenotype, being leaner in spite of having a significantly increased food intake, which is counterbalanced by increased energy expenditure(Reference Xia, Stanhope and Digitale115, Reference Xia, Sniderman and Cianflone116). Similarly, mice lacking the ASP receptor C5L2 showed a phenotype similar to ASP-deficient mice, with delayed postprandial TAG clearance, increased dietary food intake, and increased muscle fatty acid oxidation(Reference Paglialunga, Schrauwen and Roy117). Furthermore, exogenously administered ASP increases energy storage while ASP antibody neutralisation increases whole body energy utilisation in mice(Reference Paglialunga, Fisette and Munkonda118). Recently, it has been suggested that injections of ASP in the third ventricle inhibit food intake in rats through an increase in POMC expression(Reference Roy, Roy and Gauvreau119). Therefore, ASP is pointed out as a potent anabolic hormone that may also be a mediator of energy expenditure. The potential contribution of ASP as a regulator of food intake and energy expenditure in humans remains to be elucidated.

TNF-α

TNF-α is a cytokine involved in the metabolic disturbances of chronic inflammation, playing a major role in pathophysiological processes such as insulin resistance and anorexia(Reference Frühbeck, Gómez-Ambrosi and Muruzábal23, Reference Rodríguez, Catalán and Gómez-Ambrosi120). In addition, TNF-α is a potent negative regulator of adipocyte differentiation(Reference Cawthorn, Heyd and Hegyi121). Adipose tissue is both a source of and a target for TNF-α(Reference Cawthorn and Sethi122). It has been suggested that TNF-α is a candidate mediator of insulin resistance in obesity, as it is overexpressed in the adipose tissue of obese rodents and humans(Reference Hotamisligil123). This cytokine blocks the action of insulin in adipose tissue and skeletal muscle in vitro and in vivo. In this sense, TNF-α-deficient mice exhibit protection from the development of obesity-induced insulin resistance(Reference Uysal, Wiesbrock and Marino124).

The anorexigenic and cachexic effects of TNF-α are well known(Reference Tisdale125, Reference de Kloet, Pacheco-Lopez and Langhans126). This cytokine modulates the expression of neurotransmitters involved in the control of energy homeostasis, favouring anorexia and energy expenditure(Reference Amaral, Barbuio and Milanski127). Brown adipose tissue (BAT) is a particular form of adipose tissue, functionally and morphologically distinct from white adipose tissue, specialised in dissipating energy in the form of heat(Reference Frühbeck, Sesma and Burrell128, Reference Frühbeck, Becerril and Sáinz129). BAT has been suggested to play a role in human energy homeostasis(Reference Frühbeck, Becerril and Sáinz129, Reference Cypess, Lehman and Williams130). Several controversial studies suggest that TNF-α may modulate the thermogenic capacity of BAT(Reference Cawthorn and Sethi122). Administration of TNF-α increases BAT thermogenic activity(Reference Masaki, Yoshimatsu and Chiba131). This is consistent with the catabolic role of TNF-α. In contrast, other studies suggest that TNF-α decreases the activity of BAT(Reference Valladares, Roncero and Benito132). Accordingly, the lack of TNF-α receptor 1 improves the thermoadaptive capacity of obese animals(Reference Romanatto, Roman and Arruda133). However, information regarding the role of TNF-α in energy expenditure in humans, beyond its adipose tissue-mobilising effect, is scarce. Disentangling the exact role of TNF-α action in insulin resistance and energy expenditure in humans may provide the basis for the development of novel strategies for treating obesity and the metabolic syndrome.

IL-6

IL-6 is an inflammatory cytokine with pleiotropic effects on a variety of tissues, including stimulation of acute-phase protein synthesis and regulation of glucose and lipid metabolism(Reference Frühbeck and Salvador22, Reference Frühbeck, Gómez-Ambrosi and Muruzábal23). Adipose tissue produces IL-6, with circulating levels of IL-6 being proportional to adipose mass and the magnitude of insulin resistance(Reference Bastard, Maachi and Van Nhieu134, Reference Tilg and Moschen135). Although increased concentrations of IL-6 have been detected in obese subjects, mice lacking IL-6 develop mature-onset obesity, with the obese phenotype being only partly ameliorated by IL-6 replacement(Reference Wallenius, Wallenius and Ahren136). Interestingly, acute IL-6 treatment has been reported to produce an increase in insulin-stimulated glucose disposal in humans in vivo and to induce fatty acid oxidation, glucose transport, and GLUT4 translocation to the plasma membrane in vitro (Reference Carey, Steinberg and Macaulay137). In this sense, IL-6 is considered an autocrine/paracrine regulator of adipocyte function. Its involvement in the development of insulin resistance is still not completely understood(Reference Tilg and Moschen135).

Similarly to TNF-α, IL-6 has been shown to trigger catabolic effects(Reference Tisdale125). In this sense, IL-6 exerts anti-obesity effects centrally by increasing energy expenditure(Reference Wallenius, Wallenius and Sunter138), with mice lacking IL-6 exhibiting adult-onset obesity(Reference Wallenius, Wallenius and Ahren136, Reference Chida, Osaka and Hashimoto139) through decreased expression of corticotrophin-releasing hormone (CRH) and oxytocin in the hypothalamus. Due to the fact that CRH is known to stimulate energy expenditure and oxytocin has anorexigenic effects, a reduction in both neuropeptides may be contributing to the obese phenotype of mice lacking IL-6(Reference Benrick, Schele and Pinnock140). In humans, exogenous administration of IL-6 increases energy expenditure and activates the hypothalamic–pituitary–adrenal axis, thereby suggesting that CRH may be mediating this effect, as is observed in mice(Reference Stouthard, Romijn and Van der Poll141, Reference Tsigos, Papanicolaou and Defensor142). Moreover, a polymorphism in the IL-6 gene has been shown to influence energy expenditure(Reference Kubaszek, Pihlajamaki and Punnonen143). It seems clear that IL-6 regulates energy expenditure; however, its exact involvement in the development of obesity in humans and its potential therapeutic utility remain to be fully elucidated. It should be noted that the treatment of obesity with IL-6 is not currently under way due to potential major side effects and the lack of knowledge regarding the relative contribution of different target organs to IL-6-induced thermogenesis(Reference Hoene and Weigert144).

Visfatin

Visfatin, previously identified as nicotinamide phosphoribosyl transferase and colony-enhancing factor of pre-B cells, was originally identified as a modulator of B cell differentiation expressed in lymphocytes, bone marrow, skeletal muscle and liver(Reference Ouchi, Parker and Lugus145). It was named visfatin because it is highly secreted by visceral fat of both mice and humans and its expression levels in serum increase during the development of obesity(Reference Ouchi, Parker and Lugus145, Reference Fukuhara, Matsuda and Nishizawa146). Visfatin was first reported to have insulin-like activity(Reference Fukuhara, Matsuda and Nishizawa146). However, these findings are currently controversial, with the authors having been forced to retract some of their original conclusions(Reference Fukuhara, Matsuda and Nishizawa147). Surprisingly, plasma visfatin correlates with measures of obesity but not with visceral fat mass or variables of insulin sensitivity in humans. Furthermore, visfatin mRNA expression does not differ between visceral and subcutaneous adipose tissue(Reference Berndt, Klöting and Kralisch148, Reference Catalán, Gómez-Ambrosi and Rodríguez149). Nicotinamide phosphoribosyl transferase is thought to play an important role in insulin secretion by pancreatic β-cells(Reference Revollo, Korner and Mills150) and appears to also be involved in the regulation of the inflammatory response(Reference Moschen, Kaser and Enrich151).

There is little information regarding the effect of visfatin on energy homeostasis. Central administration of visfatin to Sprague–Dawley rats decreased food intake and locomotor activity, and also increased body temperature(Reference Park, Jin and Park152). These effects resemble those produced by other pro-inflammatory cytokines and they take place via the melanocortin pathway(Reference Park, Jin and Park152). Undoubtedly, more studies are needed in order to fully understand the real implications of visfatin in glucose metabolism and energy homeostasis(Reference Chen, Chung and Chang93, Reference Arner153).

Visceral adipose tissue-derived serpin

Visceral adipose tissue-derived serpin (vaspin) is a member of the serine protease inhibitor family. Vaspin is highly expressed in adipocytes of visceral adipose tissue at the same time that obesity and insulin levels peak in Otsuka Long-Evans Tokushima fatty (OLETF) diabetic obese rats. Administration of vaspin to obese insulin-resistant mice improves glucose tolerance and insulin sensitivity. These findings indicate that vaspin exerts an insulin-sensitising effect in states of obesity(Reference Hida, Wada and Eguchi154). Human vaspin mRNA expression in adipose tissue is not detectable in lean normoglycaemic individuals, but is induced by increased fat mass and decreased insulin sensitivity, which could represent a compensatory mechanism associated with obesity and T2DM(Reference Klöting, Berndt and Kralisch155). However, no differences in the levels of vaspin between individuals with normal glucose tolerance and T2DM have been detected(Reference Youn, Klöting and Kratzsch156). The potential involvement of vaspin in glucose homeostasis certainly requires further investigation(Reference Zvonic, Lefevre and Kilroy157).

Vaspin mRNA in adipose tissue decreases after fasting and its levels are partially recovered after leptin treatment(Reference González, Caminos and Vázquez158). Circulating vaspin concentrations follow a meal-related circadian variation in humans, similar to that seen for ghrelin, suggesting a role for vaspin in the regulation of food intake. Serum vaspin levels exhibit a preprandial rise followed by a rapid decline after meals(Reference Jeong, Youn and Kim159). Both peripheral and central vaspin administration decrease food intake in mice(Reference Klöting, Kovacs and Kern160). Furthermore, intrahypothalamic vaspin administration reduces food intake in rats, decreasing NPY and increasing POMC mRNA expression(Reference Brunetti, Di Nisio and Recinella161). Therefore, vaspin exhibits anorexigenic and glucose-lowering effects, suggesting its potential use as a therapeutic tool for the treatment of obesity and related diseases. However, the effect of vaspin on energy expenditure needs to be addressed.

Chemerin

Chemerin, also known as retinoic acid receptor responder 2, is a secreted chemoattractant protein with an important role in adaptive and innate immunity(Reference Catalán, Gómez-Ambrosi and Rodríguez25, Reference Ernst and Sinal162). Chemerin has been recently described as an adipokine associated with obesity and the metabolic syndrome(Reference Bozaoglu, Bolton and McMillan163, Reference Goralski, McCarthy and Hanniman164). Furthermore, chemerin contributes to inflammation by stimulating macrophage adhesion to extracellular matrix proteins and by promoting chemotaxis(Reference Hart and Greaves165). Increased mRNA expression of chemerin has been found in mouse and human adipocytes, while the knockdown of chemerin indicates a major role for this adipokine in regulating adipogenesis and metabolic homeostasis in the adipocyte(Reference Goralski, McCarthy and Hanniman164). Increased circulating levels of chemerin have been found in morbidly obese patients, with a significant decrease after bariatric surgery(Reference Sell, Divoux and Poitou166, Reference Catalán, Gómez-Ambrosi and Rodríguez167).

Central administration of chemerin does not modify food intake 24 h after treatment in rats(Reference Brunetti, Di Nisio and Recinella161). However, chemerin treatment increases both AgRP and POMC mRNA expression in the hypothalamus. This could lead to a null effect, considering that AgRP is orexigenic and POMC is anorexigenic, but it suggests a role of chemerin in the regulation of neuropeptides involved in food intake regulation(Reference Brunetti, Di Nisio and Recinella161). The changes in chemerin concentrations after weight loss may merely reflect the reduction in body adiposity, but also a putative role in body weight homeostasis(Reference Sell, Divoux and Poitou166, Reference Catalán, Gómez-Ambrosi and Rodríguez167).

Omentin

Omentin, also named intelectin or intestinal lactoferrin receptor, is another recently described visceral fat depot-specific adipokine(Reference Catalán, Gómez-Ambrosi and Rodríguez25, Reference Schaffler, Neumeier and Herfarth168). It is a secreted protein likely to act as both an endocrine factor to modulate systemic metabolism and an autocrine/paracrine factor to regulate adipocyte biology locally(Reference Schaffler, Neumeier and Herfarth168, Reference Yang, Lee and Hu169). Obesity negatively regulates omentin expression and its release into the circulation, with reduced plasma omentin levels having been observed in obese subjects(Reference de Souza Batista, Yang and Lee170). Omentin increases insulin action by enhancing insulin-mediated glucose transport in isolated adipocytes(Reference Schaffler, Neumeier and Herfarth168, Reference Yang, Lee and Hu169). Omentin has also been related to inflammation, exerting an anti-inflammatory action and displaying beneficial effects on the metabolic syndrome(Reference Tan, Adya and Randeva171).

Little is known regarding the role of omentin in energy homeostasis. Central administration of omentin produces a slight but non-significant increase in food intake in rats(Reference Brunetti, Di Nisio and Recinella161). In humans, circulating omentin concentrations change oppositely to what takes place in energy balance, thereby rising after prolonged negative energy balance, as is the case after dietary-induced weight loss(Reference Moreno-Navarrete, Catalán and Ortega172). Further studies are necessary in order to gain more insight regarding the involvement of omentin in the regulation of appetite and energy expenditure.

Angiontensin II

Murine models of obesity show increased local formation of angiotensin (Ang) II due to elevated secretion of its precursor, angiotensinogen, from adipocytes(Reference Frühbeck, Gómez-Ambrosi and Muruzábal23, Reference Yvan-Charvet and Quignard-Boulange173, Reference Kalupahana and Moustaid-Moussa174), with deficiency or overexpression of angiotensinogen affecting body weight regulation(Reference Frühbeck and Gómez-Ambrosi13). Given the close relationship between Ang II and insulin resistance and the fact that the renin–Ang system is inappropriately activated in obesity(Reference Katovich and Pachori175, Reference Luther and Brown176), the participation of the adipose tissue–renin–Ang system in the development of insulin resistance and the metabolic syndrome is conceivable in humans, but has to be evaluated in more detail(Reference Yvan-Charvet and Quignard-Boulange173, Reference Engeli, Schling and Gorzelniak177).

Mice lacking angiotensinogen or any of its two major receptor subtypes, type 1 (AT1R) and type 2, show protection against the development of obesity without notable changes in food intake(Reference Massiera, Seydoux and Geloen178Reference Yvan-Charvet, Even and Bloch-Faure180). By contrast, it is clear that these knockout mice exhibit higher energy expenditure, particularly when a high-fat diet is consumed(Reference Kouyama, Suganami and Nishida179, Reference Yvan-Charvet, Even and Bloch-Faure180). Furthermore, mice deficient in Ang-converting enzyme (ACE), which is responsible for the conversion of Ang I to the bioactive peptide Ang II, also have increased energy expenditure, with reduced fat mass and improved glucose clearance(Reference Jayasooriya, Mathai and Walker181). Paradoxically, administration of Ang II to rats by means of subcutaneous osmotic minipumps produces a maintained reduction of food intake with a transient decrease in oxygen consumption(Reference Brink, Wellen and Delafontaine182, Reference Cassis, Helton and English183). Furthermore, ACE inhibition with captopril reduces food intake and protects against the development of diet-induced obesity and glucose intolerance in rats(Reference de Kloet, Krause and Kim184). However, Ang II may exert different effects on metabolism, depending on the tissues of action. In this sense, accumulating evidence suggests that increased Ang II activity locally within the brain promotes negative energy balance(Reference Porter and Potratz185, Reference de Kloet, Krause and Scott186). Taken together, these studies suggest that reduced systemic Ang II signalling protects against diet-induced adipose tissue enlargement by increasing energy expenditure in rodents(Reference Yvan-Charvet and Quignard-Boulange173). In humans, although ACE inhibitors and AT1R blockers are widely used as antihypertensive agents and are beginning to be used for promoting insulin sensitivity, there is minimal evidence that these agents significantly affect energy homeostasis(Reference de Kloet, Krause and Woods187).

Sex steroids

Adipose tissue expresses different sex steroid-metabolising enzymes that promote the conversion of oestrogens from androgenic precursors, which are produced by the gonads or adrenal glands, thereby regulating the synthesis and bioavailability of endogenous sex steroids, oestrogens and androgens, through different mechanisms(Reference Tchernof and Despres188Reference Mauvais-Jarvis191). In this sense, in men and postmenopausal women, adipose tissue is the main source of oestrogen synthesis, and circulating levels of oestrogens are directly related to BMI. Sex steroid hormones play a critical role in adipose tissue metabolism, distribution and accretion(Reference Tchernof and Despres188, Reference Mayes and Watson189). In women, menopause-induced oestrogen deficiency and increased androgenicity are associated with increased visceral obesity and with the subsequent cardiometabolic alterations(Reference Tchernof and Despres188). Moreover, hormone replacement therapy with oestradiol treatment for 1 year decreased intra-abdominal fat(Reference Mattiasson, Rendell and Tornquist192). Ageing in men is associated with a progressive deficit in androgen production and reduced concentrations of testosterone have been related to increased visceral obesity and the metabolic syndrome(Reference Tsai, Boyko and Leonetti193, Reference Kupelian, Page and Araujo194). Therefore, treating middle-aged obese men with testosterone reduces abdominal fat(Reference Marin, Holmang and Jonsson195).

In females, oestrogens regulate energy homeostasis via oestrogen receptor (ER)α and ERβ, by reducing food intake(Reference Somogyi, Gyorffy and Scalise18, Reference Brown, Gent and Davis190, Reference Eckel196) and adiposity(Reference Heine, Taylor and Iwamoto197), enhancing energy expenditure(Reference Musatov, Chen and Pfaff198, Reference Ropero, Alonso-Magdalena and Quesada199), and improving insulin sensitivity(Reference Ropero, Alonso-Magdalena and Quesada199). In males, testosterone is converted to oestrogen and controls energy homeostasis via ER and androgen receptors, which share related functions for increasing energy expenditure, reducing fat accumulation(Reference Fan, Yanase and Nomura200) and ameliorating glucose homeostasis(Reference Mauvais-Jarvis191). It has been recently reported that distinct hypothalamic neurons mediate oestrogenic effects on food intake and energy homeostasis. Food intake is regulated by ERα in POMC neurons while energy expenditure and fat accumulation is controlled by ERα in steroidogenic factor-1 neurons(Reference Xu, Nedungadi and Zhu201).

Signals from the pancreas

The major physiological function of the endocrine pancreas is the maintenance of glucose homeostasis. The pancreas senses the concentration of glucose in blood and, through the release of insulin and glucagon, regulates glucose uptake and utilisation by peripheral tissues. However, insulin and glucagon, as well as pancreatic polypeptide (PP) and amylin, also exert a regulatory effect on energy homeostasis(Reference Plum, Belgardt and Brüning202).

Insulin

Insulin was the first hormone to be involved in the control of body weight by the central nervous system(Reference Plum, Belgardt and Brüning202Reference Brüning, Gautam and Burks204). To date, insulin and leptin are the only hormones that fulfil the criteria to be considered an adiposity signal. Both hormones circulate at concentrations proportional to the amount of body fat and enter the central nervous system where leptin and insulin receptors are expressed by neurons involved in energy homeostasis; administration of either molecule into the brain reduces food intake, whereas its deficiency does the opposite(Reference Morton, Cummings and Baskin27, Reference Schwartz, Woods and Porte205). The breeding of mice with brain-specific insulin receptor deficiency, which translates into an increased food intake and diet-sensitive obesity, demonstrated a critical role for brain insulin signalling in the central regulation of energy disposal and fuel metabolism(Reference Brüning, Gautam and Burks204). The central effect of insulin on the reduction of food intake is mainly mediated through an inhibition of NPY/AgRP neurons and the stimulation of POMC neurons(Reference Morton, Cummings and Baskin27, Reference Plum, Belgardt and Brüning202). Besides these basic homeostatic circuits, food palatability and reward are thought to be major factors involved in the regulation of food intake elicited by insulin and leptin(Reference Könner, Klöckener and Brüning206). Although it was initially considered that the effect of insulin was dose dependent, with low doses stimulating thermogenesis and high doses decreasing it(Reference Rothwell and Stock207), further studies have shown the thermogenic effect of insulin(Reference Menéndez and Atrens208, Reference Dulloo and Girardier209).

Glucagon

Glucagon is secreted by the α-cells of the pancreatic islets. It has catabolic properties, functioning as a counter-regulatory hormone opposing the actions of insulin. Glucagon maintains blood glucose concentrations during fasting by promoting glycogenolysis and gluconeogenesis as well as by inhibiting glycogenesis and glycolysis in the liver, thereby preventing hypoglycaemia(Reference Jiang and Zhang210, Reference Gómez-Ambrosi, Catalan and Frühbeck211). Obese patients exhibit increased glucagon levels(Reference Kalkhoff, Gossain and Matute212, Reference Starke, Erhardt and Berger213). Inappropriately elevated concentrations of insulin and glucagon, together with insulin resistance, contribute to the obesity-associated impaired glucose homeostasis(Reference Koeslag, Saunders and Terblanche214).

Glucagon exerts many extrahepatic actions. It increases lipolysis in adipose tissue and reduces food intake, acting as a satiety factor in the brain(Reference Gómez-Ambrosi, Catalan and Frühbeck211). In humans, pharmacological doses of glucagon decrease the amount of food that is eaten(Reference Penick and Hinkle215). Although this effect was initially attributed to an indirect action of glucagon via the increase in portal glucose levels(Reference Geary, Langhans and Scharrer216), it has been clearly demonstrated that glucagon has central actions in the brain to reduce food intake(Reference Habegger, Heppner and Geary217). Administration of glucagon has been shown to produce weight loss in humans and rats(Reference Schulman, Carleton and Whitney218, Reference Billington, Briggs and Link219). This can be explained by the fact that glucagon causes an increase in energy expenditure(Reference Davidson, Salter and Best220) via activation of BAT(Reference Billington, Briggs and Link219). The effect of glucagon on human BAT remains to be fully clarified. However, this effect of glucagon promoting energy expenditure contrasts with the phenotype observed in glucagon receptor knockout mice that exhibit lower adiposity, despite having normal growth rates, body weight, food intake and resting energy expenditure(Reference Gelling, Du and Dichmann221). Similar striking observations are reported for mice lacking synaptotagmin-7, a Ca sensor for insulin and glucagon granule exocytosis, which show normal insulin concentrations but severely reduced glucagon levels. This mouse model exhibits a lean phenotype with increased lipolysis and energy expenditure(Reference Lou, Gustavsson and Wang222). A potential counter-regulatory increase in GLP-1 in mice lacking glucagon signalling could explain these discrepancies(Reference Vuguin and Charron223).

Pancreatic polypeptide

PP is a thirty-six-amino acid peptide belonging to the family of peptides including NPY and PYY, which is secreted postprandially under vagal control by pancreatic islet PP cells(Reference Field, Chaudhri and Bloom19, Reference Schwartz, Holst and Fahrenkrug224). Circulating levels of PP are apparently normal in obese patients(Reference Jorde and Burhol225, Reference Pieramico, Malfertheiner and Nelson226) but the rapid increase in response to a meal observed in healthy subjects is significantly impaired in obese individuals(Reference Lassmann, Vague and Vialettes227, Reference Holst, Schwartz and Lovgreen228).

Peripheral administration of PP acutely reduces food intake and gastric emptying and increases energy expenditure in mice, whereas repeated administration during 2 weeks leads to reductions in body weight gain(Reference Asakawa, Inui and Yuzuriha229). Similar effects are observed in transgenic mice overexpressing PP(Reference Ueno, Inui and Iwamoto230). The anorectic effect is mediated through Y4 receptors and is associated with reduced expression of NPY and orexin mRNA in the hypothalamus, being dependent on intact vagal signalling(Reference Asakawa, Inui and Yuzuriha229). In humans, intravenous infusion of PP leads to delayed gastric emptying and reduced cumulative 24 h food intake(Reference Batterham, Le Roux and Cohen231, Reference Schmidt, Näslund and Grybäck232). To our knowledge, there are no data reporting the effect of PP on energy expenditure in humans.

Amylin

Amylin, a peptide co-secreted with insulin postprandially by pancreatic β-cells and, therefore, also named islet amyloid polypeptide, was firstly isolated from diabetic human pancreas(Reference Cooper, Willis and Clark233). Amylin inhibits gastric emptying as well as gastric acid and glucagon secretion(Reference Field, Chaudhri and Bloom19, Reference Cummings and Overduin234, Reference Lutz235). Increased circulating concentrations of amylin have been reported in obese rats and humans, suggesting a role for amylin in the pathophysiology of obesity(Reference Boyle and Lutz236).

Central or peripheral administration of amylin decreases meal size and food intake by promoting meal-ending satiation(Reference Lutz235). The anorectic effects of amylin, in contrast to other gut peptides, take place primarily in the area postrema, showing synergy with PYY, CCK and leptin(Reference Trevaskis, Parkes and Roth237). Mechanistic studies in rodents suggest that amylin reduces body weight in a fat-specific manner, preserving lean mass(Reference Trevaskis, Parkes and Roth237). The synthetic amylin analogue pramlintide is prescribed for the treatment of diabetes but also causes mild progressive weight loss in humans(Reference Aronne, Fujioka and Aroda238). Furthermore, pramlintide also exhibits synergic effects with leptin after 20 weeks of treatment in overweight and obese volunteers(Reference Roth, Roland and Cole239). In addition to its role eliciting satiety, amylin appears to influence energy balance by increasing energy expenditure(Reference Roth, Hughes and Kendall240Reference Osaka, Tsukamoto and Koyama242). Amylin administration increases lipid utilisation, as indicated by a lower respiratory quotient, reducing adiposity(Reference Rushing, Hagan and Seeley243). Finally, it has been suggested that amylin could prevent the compensatory decrease in energy expenditure that typically takes place during or after weight loss(Reference Lutz235).

Signals from the gut

The gastrointestinal tract is the largest endocrine organ in the body, representing an important source of regulatory hormones(Reference Murphy, Dhillo and Bloom244). The gut uses neural and endocrine pathways to coordinately regulate food intake and energy expenditure in the hypothalamus(Reference Badman and Flier245). Ghrelin is considered the only circulating orexigenic hormone, which seems to act as a meal initiator(Reference Kojima and Kangawa246). Satiety signals derived from the gut include GLP-1, PYY, CCK and oxyntomodulin, among others(Reference Cummings and Overduin234, Reference Murphy, Dhillo and Bloom244, Reference Badman and Flier245, Reference Murphy and Bloom247, Reference Wren and Bloom248). Recently, the existence of another peptide, prouroguanylin, produced by the intestine and with satiating effects has been reported(Reference Valentino, Lin and Snook16).

Ghrelin

Ghrelin is a twenty-eight-amino acid peptide secreted by oxyntic cells in the stomach fundus. Ghrelin was first characterised as a natural ligand of the growth hormone secretagogue receptor(Reference Kojima, Hosoda and Date249). In subsequent studies ghrelin was shown to participate in the complex entero-hypothalamic control of food intake signalling(Reference Nakazato, Murakami and Date250). Central or peripheral administration of ghrelin increases food intake and adiposity in rodents(Reference Tschöp, Smiley and Heiman251, Reference Asakawa, Inui and Kaga252) and humans(Reference Wren, Seal and Cohen253, Reference Rodríguez, Gómez-Ambrosi and Catalán254). In humans, plasma ghrelin concentrations have been shown to rise shortly before and fall quickly after every meal, suggesting a role in meal initiation(Reference Cummings, Purnell and Frayo255). Ghrelin concentrations are decreased in human obesity(Reference Tschöp, Weyer and Tataranni256), which has been explained as a physiological adaptation to the positive energy balance associated with obesity, and increase in response to diet-induced weight loss(Reference Cummings, Weigle and Frayo257). However, many studies have shown that ghrelin concentrations do not increase after surgically induced weight loss following procedures that compromise the functionality of the fundus(Reference Frühbeck, Diez Caballero and Gil258, Reference Frühbeck, Diez-Caballero and Gil259), while other studies have reported an increase in ghrelin levels following bariatric surgery(Reference Holdstock, Engström and Öhrvall260).

Chronic central or peripheral administration of ghrelin increases cumulative food intake and decreases energy expenditure, resulting in body weight gain(Reference Nakazato, Murakami and Date250, Reference Tschöp, Smiley and Heiman251, Reference Wren, Seal and Cohen253, Reference Wren, Small and Abbott261). The orexigenic effect of ghrelin is mediated by the activation of NPY/AgRP, since ghrelin does not stimulate feeding in NPY and AgRP double knockout mice(Reference Chen, Trumbauer and Chen262), and it is also mediated by the inhibition of hypothalamic fatty acid biosynthesis(Reference López, Lage and Saha263). In this sense, the absence of ghrelin or its receptors protects against the development of early-onset obesity(Reference Wortley, Del Rincon and Murray264, Reference Zigman, Nakano and Coppari265) and mice lacking simultaneously ghrelin and its receptor exhibit an increased energy expenditure(Reference Pfluger, Kirchner and Günnel266). Therefore, ghrelin signalling inhibition has been suggested as a potential therapeutic tool in obesity treatment, whereas a direct therapeutic application of ghrelin can be contemplated for the treatment of cachexia and anorexia(Reference Wren and Bloom248). In this sense, intravenous administration of ghrelin results in weight gain in patients with cardiac cachexia and chronic obstructive pulmonary disease(Reference Nagaya, Moriya and Yasumura267, Reference Nagaya, Itoh and Murakami268).

Glucose-dependent insulinotropic polypeptide

Insulin secretion is higher in response to orally administered than to intravenous glucose administration. This is known as the incretin effect(Reference Drucker269). Originally named gastric inhibitory polypeptide, glucose-dependent insulinotropic polypeptide (GIP) was the first incretin identified(Reference Brown270). The major stimulus for GIP secretion is nutrient intake. GIP is mainly secreted by K cells in the duodenum and jejunum(Reference Drucker269, Reference Song and Wolfe271). Circulating concentrations of GIP are low in the fasting state and rise within minutes following food intake. Moreover, GIP levels increase after a high-fat diet, with postprandial GIP secretion being significantly higher in obese subjects than in age-matched lean individuals(Reference Song and Wolfe271, Reference Roust, Stesin and Go272). In addition to its role in regulating insulin secretion, GIP stimulates β-cell replication and mass expansion at the same time as it stimulates glucagon secretion(Reference Drucker273, Reference Wideman and Kieffer274). In addition, GIP inhibits gastric acid secretion and gastric emptying, although only at supraphysiological doses(Reference Fehmann, Goke and Goke275).

The GIP receptor (GIPR) is present in adipose tissue, regulating adipocyte growth, and there is a large body of biochemical and animal data suggesting that GIP signalling promotes fat accumulation(Reference Song and Wolfe271, Reference Wasada, McCorkle and Harris276Reference Althage, Ford and Wang278). Chemical or genetic ablation of GIP signalling or targeted reduction of GIP-secreting cells does not modify food intake(Reference Althage, Ford and Wang278Reference Hansotia, Maida and Flock281). However, the absence of GIP signalling produces a significant increase in energy expenditure, protecting from high-fat diet-induced obesity and insulin resistance(Reference Althage, Ford and Wang278, Reference Miyawaki, Yamada and Ban279, Reference Hansotia, Maida and Flock281). Moreover, peripheral administration of synthetic human GIP reduces energy expenditure in healthy subjects but not in patients with T2DM(Reference Daousi, Wilding and Aditya282). Furthermore, emerging evidence suggests that the rapid resolution of diabetes in morbidly obese patients undergoing bypass surgery is mediated, at least in part, by surgical removal of GIP-secreting cells in the upper small intestine(Reference Irwin and Flatt283). Although inhibiting GIP/GIPR signalling may be beneficial as a treatment for obesity(Reference Song and Wolfe271), the mechanisms involved in the regulation of food intake and energy expenditure elicited by GIP in humans remain to be fully understood.

Glucagon-like peptide 1

Through action of prohormone convertases, proglucagon is processed to glicentin, oxyntomodulin, intervening peptides 1 and 2, GLP-1 and GLP-2. To date, only GIP and GLP-1 are considered to be incretin hormones in humans, being responsible for as much as 50 % of postprandial insulin secretion(Reference Wideman and Kieffer274, Reference Baggio and Drucker284). GLP-1 is mainly produced by L-cells in the ileum after meals; in addition to its role as an insulinotropic hormone it participates actively in regulating gastric motility, islet β-cell neogenesis, neuronal plasticity and the suppression of plasma glucagon concentrations(Reference Kieffer and Habener285, Reference Drucker286). Although a clear role for GLP-1 in the aetiology of T2DM has not been proved, a common view states that GLP-1 secretion in patients with T2DM is deficient(Reference Nauck, Vardarli and Deacon287). Similarly, some controversies exist regarding the involvement of GLP-1 in obesity pathophysiology. It has been suggested that obesity is associated with reduced secretion of GLP-1(Reference Ranganath, Beety and Morgan288Reference Torekov, Madsbad and Holst290), which is restored to normal levels after weight loss(Reference Verdich, Toubro and Buemann291), particularly following malabsorptive bariatric surgery(Reference Morínigo, Moize and Musri292).

GLP-1 has an important role in food intake regulation, promoting satiety(Reference Turton, O'Shea and Gunn293Reference Gutzwiller, Goke and Drewe295) even in obese men(Reference Naslund, Barkeling and King296, Reference Flint, Raben and Ersboll297), acting as a short-term satiation signal, limiting the amount of food eaten and prolonging time between meals(Reference Williams, Baskin and Schwartz298). Another important action of this incretin in relation to energy homeostasis is the inhibition of gastric emptying following GLP-1 administration, with the vagus nerve playing an important role(Reference Drucker286, Reference Flint, Raben and Ersboll297). Intravenous administration of GLP-1 increases postprandial energy expenditure via the lower brainstem and the sympathoadrenal system in rats(Reference Osaka, Endo and Yamakawa299). Exogenous administration of GLP-1 to humans reduces postprandial thermogenesis, which can be explained by a reduction in meal size(Reference Flint, Raben and Ersboll297). However, higher fasting plasma concentrations of GLP-1 are associated with higher resting energy expenditure and fat oxidation rates in humans(Reference Pannacciulli, Bunt and Koska300). Data regarding energy expenditure from mice deficient in GLP-1 signalling are conflicting, with some studies finding that loss of function protects against the development of diet-induced obesity by increasing energy expenditure(Reference Hansotia, Maida and Flock281, Reference Knauf, Cani and Ait-Belgnaoui301, Reference Ayala, Bracy and James302), while others show that loss of GLP-1 signalling increases fat accumulation(Reference Nogueiras, Perez-Tilve and Veyrat-Durebex303, Reference Barrera, Jones and Herman304). These differences may be related to species-specific differences and effects on locomotor activity(Reference Hayes, De Jonghe and Kanoski305).

GLP-1 signalling is a potential target for the treatment of both T2DM and obesity. In this sense, liraglutide, a GLP-1 analogue with a prolonged half-life initially developed for the treatment of T2DM, has shown additional beneficial features for body weight control(Reference Vilsboll, Zdravkovic and Le-Thi306, Reference Astrup, Rossner and Van Gaal307). In this context, activation of the GLP-1 receptor is currently proposed as the most effective drug for treating the metabolic syndrome(Reference Day, Ottaway and Patterson308).

GLP-2 has also been involved in the regulation of food intake(Reference Tang Christensen, Larsen and Thulesen309). However, deletion of GLP-2 receptor signalling in ob/ob mice impairs the normal islet adaptive response needed for maintaining glucose homeostasis but has no effect on body weight or food intake(Reference Bahrami, Longuet and Baggio310). GLP-2 has also been associated with gut hypertrophy and intestinal crypt cell proliferation after gastric bypass(Reference le Roux, Borg and Wallis311) but has no effect on energy expenditure(Reference Osaka, Endo and Yamakawa299).

Further evidence of the important involvement of incretins in energy homeostasis arises from studies involving dipeptidyl peptidase 4 (DPP-4). DPP-4 is a member of the prolyl oligopeptidase family of peptidases and is the key enzyme responsible for cleaving and inactivating GIP and GLP-1(Reference Drucker312). Mice lacking DPP-4 exhibit improved glucose tolerance and insulin sensitivity as well as resistance to diet-induced obesity, which can be explained by reduced food intake and increased energy expenditure(Reference Conarello, Li and Ronan313).

Peptide YY

PYY is a thirty-six-amino acid gut hormone so called after the tyrosine residues at each terminus of the peptide that belongs to the NPY family(Reference Tatemoto and Mutt314, Reference Karra and Batterham315). It is secreted mainly from specialised enteroendocrine cells, called L-cells, of the distal gut, with the highest production being in the ileum and colon. Two main endogenous forms of PYY exist, PYY1–36 and PYY3–36. PYY3–36 is the dominant circulating form of the peptide both in the fasted and fed states, accounting for 60 % of postprandial circulating PYY(Reference Grandt, Schimiczek and Beglinger316, Reference Kirchner, Tong and Tschöp317). Circulating concentrations of PYY rise within 15 min after nutrient ingestion. PYY1–36 has specificity for Y1 and Y5 receptors, increasing food intake. However, PYY3–36 binds preferentially to Y2 receptors, thereby stimulating anorectic pathways(Reference Zac-Varghese, De Silva and Bloom318). Lower levels of PYY3–36 have been reported in obese individuals, suggesting that this gut hormone has a role in the pathophysiology of obesity(Reference Batterham, Cohen and Ellis319).

Peripheral injection of PYY3–36 has been shown to reduce food intake and to induce a negative energy balance in mice and rats(Reference Batterham, Cowley and Small320), monkeys(Reference Moran, Smedh and Kinzig321) and humans(Reference Batterham, Cowley and Small320), even in obese patients(Reference Batterham, Cohen and Ellis319). This occurs through modulation of different cortical and hypothalamic brain areas(Reference Batterham, Ffytche and Rosenthal322). However, these anorexigenic effects of PYY3–36 have not been confirmed by others(Reference Tschöp, Castañeda and Joost323). Furthermore, despite the effects of PYY3–36 on food intake inhibition, mice lacking Pyy do not exhibit a clear phenotype, showing normal feeding behaviour, growth and energy expenditure(Reference Schonhoff, Baggio and Ratineau324, Reference Wortley, Garcia and Okamoto325), or even obesity(Reference Boey, Lin and Karl326, Reference Batterham, Heffron and Kapoor327). Intravenous administration of PYY3–36 increases lipolysis and energy expenditure in humans(Reference Sloth, Holst and Flint328), with total PYY being significantly correlated with postprandial energy expenditure(Reference Doucet, Laviolette and Imbeault329). However, this association has not been unequivocally found(Reference Guo, Ma and Enriori330). The extent of PYY3–36 involvement in the regulation of energy homeostasis and the underlying mechanisms mediating the effects of PYY3–36 on energy expenditure in humans are still not fully understood.

Cholecystokinin

CCK is secreted mainly by I-cells in the proximal small intestine in response to lipids and proteins in the meal(Reference Field, Chaudhri and Bloom19, Reference Cummings and Overduin234, Reference Raybould331). The predominant circulating forms of CCK in rodents include CCK octapeptide (CCK-8) and CCK-22, whereas larger molecular forms are also present in human plasma(Reference Liddle, Goldfine and Rosen332). CCK is involved in modulating intestinal motility, stimulating pancreatic enzyme secretion, enhancing gallbladder contraction and regulating meal size but not meal frequency(Reference Lo, Samuelson and Chambers333). A total of two CCK receptors have been cloned so far: CCK1R and CCK2R. Selective CCK1R antagonists block the anorectic effect of CCK, whereas selective antagonism of CCK2R has no effect on food intake(Reference Cummings and Overduin234, Reference Crawley and Corwin334Reference Lo, King and Samuelson336).

The satiating effect of CCK was first described more than three decades ago(Reference Gibbs, Young and Smith337), with vagotomy suppressing the anorectic effects of peripheral CCK(Reference Smith, Jerome and Cushin338). In humans, intravenous infusion of CCK induces a dose-dependent suppression of food intake(Reference Lieverse, Jansen and Masclee339). Administration of CCK before the start of a meal does not delay the onset of eating, but rather reduces the amount of food consumed once eating begins(Reference Woods12). However, long-term CCK1R stimulation failed to produce significant weight loss in obese patients due to the rapid development of tolerance(Reference Crawley and Beinfeld340, Reference Jordan, Greenway and Leiter341), thereby questioning the potential of CCK as an anti-obesity target(Reference Cummings and Overduin234).

Rats deficient in CCK1R show increased meal size and obesity(Reference Moran, Katz and Plata-Salaman342). However, mice lacking CCK or CCK1R exhibit a normal food intake and body weight, apparently indicating that CCK is not essential for the long-term maintenance of body weight(Reference Lo, Samuelson and Chambers333, Reference Kopin, Mathes and McBride335). Interestingly, CCK knockout mice fed on a high-fat diet develop protection against obesity despite having a normal food intake, probably through decreased lipid absorption and increased energy expenditure(Reference Lo, King and Samuelson336).

Oxyntomodulin

Oxyntomodulin is another cleavage product of proglucagon secreted by intestinal L-cells after meals in proportion to the energy content of foods(Reference Bataille, Tatemoto and Gespach343, Reference Le Quellec, Kervran and Blache344). It was named oxyntomodulin after its inhibitory action on the oxyntic glands of the stomach(Reference Dubrasquet, Bataille and Gespach345). Oxyntomodulin inhibits gastric acid secretion and pancreatic enzyme secretion(Reference Schjoldager, Mortensen and Myhre346). Although no oxyntomodulin receptor has been identified yet, it appears that the actions of oxyntomodulin are mediated via the GLP-1 receptor(Reference Schepp, Dehne and Riedel347), since the anorectic effect of oxyntomodulin is abolished in GLP-1 receptor-deficient mice(Reference Baggio, Huang and Brown348). However, GLP-1 and oxyntomodulin appear to activate different hypothalamic pathways(Reference Parkinson, Chaudhri and Kuo349) and, therefore, a separate unidentified oxyntomodulin receptor may exist(Reference Karra and Batterham315).

Central or peripherally administered oxyntomodulin inhibits food intake in fasted and non-fasted rats(Reference Dakin, Gunn and Small350, Reference Dakin, Small and Batterham351). However, the anorectic effect in mice is only observed after intracerebroventricular administration(Reference Baggio, Huang and Brown348). Intravenous administration of oxyntomodulin suppresses appetite and reduces food intake in humans(Reference Cohen, Ellis and Le Roux352). Furthermore, subcutaneous injections of oxyntomodulin resulted in weight loss and a change in the levels of adipokines consistent with a loss of body fat over a 4-week period in overweight and obese subjects(Reference Wynne, Park and Small353). Central administration of oxyntomodulin increases energy expenditure and causes a disproportionate reduction in body weight compared with pair-fed rats(Reference Dakin, Small and Batterham351, Reference Dakin, Small and Park354). In humans, 4 d subcutaneous self-administration of pre-prandial oxyntomodulin three times per d promotes a negative energy balance, increasing energy expenditure while reducing energy intake(Reference Wynne and Bloom355). However, the acute thermogenic effect of oxyntomodulin observed in rats and humans has not been reproduced in mice(Reference Baggio, Huang and Brown348). Further studies are needed in order to investigate whether the effect of oxyntomodulin on energy expenditure in humans is maintained in the long term, but data presented above support the role of oxyntomodulin as a potential anti-obesity tool.

Uroguanylin

Guanylin and uroguanylin have been well-known key paracrine players in intestinal ion and water balance for over 20 years, acting as endogenous ligands of guanylyl cyclase (GUCY) 2C and increasing cyclic guanosine monophosphate (cGMP) production(Reference Potter356). They are secreted by intestinal epithelial cells as prohormones, requiring proteolytic enzymic conversion into active hormones in the target tissue(Reference Seeley and Tschöp357). Physiological functions for these molecules include the modulation of epithelial cell balance in the intestinal epithelium and the regulation of Na balance through actions on the kidney(Reference Carrithers, Ott and Hill358). Recently, Valentino et al. (Reference Valentino, Lin and Snook16) revealed a new endocrine role for uroguanylin in energy homeostasis. The uroguanylin precursor, prouroguanylin, is secreted into the circulation after meals in both mice and humans; it can then be cleaved to uroguanylin in the hypothalamus to activate GUCY2C for decreasing food intake(Reference Valentino, Lin and Snook16). Deletion of GUCY2C in mice disrupts appetite regulation specifically by impairing satiation, producing hyperphagia associated with obesity and glucose intolerance(Reference Valentino, Lin and Snook16). No changes in cold-induced thermogenesis assessed by core body temperature were observed(Reference Valentino, Lin and Snook16). However, the role of uroguanylin in the modulation of energy expenditure needs to be addressed in both rodents and humans. Furthermore, it has been suggested that uroguanylin could exert a direct effect on adipose tissue, regulating lipolysis(Reference Frühbeck359), given the fact that cGMP is a second messenger known to be involved in the lipolytic effect of natriuretic peptides, which are closely related to uroguanylin(Reference Lafontan, Moro and Berlan360). The uroguanylin–GUCY2C endocrine axis may offer a novel therapeutic target for regulating food intake and a weapon against obesity(Reference Valentino, Lin and Snook16, Reference Frühbeck359).

Fibroblast growth factor 19

The family of fibroblast growth factors (FGF) regulates a plethora of processes including organ development, the maintaining of bile acid homeostasis and the activation of hepatic protein and glycogen synthesis(Reference Beenken and Mohammadi361, Reference Kir, Beddow and Samuel362). FGF19 is expressed in the distal small intestine, with the concentration of circulating FGF19 increasing in response to feeding. Transgenic mice expressing human FGF19 exhibit an increased metabolic rate and decreased adiposity despite having increased food intake with an increase in fatty acid oxidation(Reference Tomlinson, Fu and John363, Reference Fu, John and Adams364). However, in addition to its metabolic actions FGF19 also has proliferative effects, with transgenic mice developing hepatocellular carcinoma within 1 year, thereby rendering FGF19, a priori, unsuitable as a candidate for combating obesity(Reference Wu, Ge and Lemon365).

Signals from the liver

The liver plays an important role in energy homeostasis(Reference Hirota and Fukamizu366). Due to its anatomical position the liver has rapid access to incoming nutrients from intestinal absorption. In addition to its role in regulating glucose and fatty acid metabolism, the liver produces several proteins involved in peripheral control of energy homeostasis.

Insulin-like growth factor system

Members of the insulin-like growth factor (IGF) system are functionally related to insulin. The IGF regulatory system consists of IGF (IGF-I and IGF-II), type I and type II IGF receptors, and IGF-binding proteins (IGFBP-1–6)(Reference Juul367, Reference Kawai and Rosen368). IGF are ubiquitously expressed, although the main source of circulating IGF-I is the liver. They exert actions in almost all tissues and are among the major regulators of growth(Reference Kawai and Rosen368, Reference Baker, Liu and Robertson369). While insulin is a short-term regulator of glucose homeostasis, IGF have been suggested to exert long-term regulation of glucose homeostasis(Reference Sandhu, Heald and Gibson370Reference Clemmons372). Insulin and IGF-I show cross-reactivity at the receptor level. After ligand binding-induced autophosphorylation, insulin receptor and IGF-I receptor catalyse the phosphorylation of cellular proteins such as members of the insulin receptor substrate family(Reference Saltiel and Kahn373). Other functions in which IGF play a critical role are the regulation of growth, neuroprotection, tumorigenesis and longevity(Reference Clemmons372, Reference Holzenberger, Dupont and Ducos374, Reference Sanchez-Alavez, Osborn and Tabarean375). Adipose tissue levels of IGF-I have been shown to be higher in both rodent and human obesity(Reference Frystyk, Vestbo and Skjaerbaek376), although the IGF-I-induced signalling cascade is impaired in obese mice(Reference Le Marchand-Brustel, Heydrick and Jullien377).

IGF-I treatment by osmotic minipumps at adult age reduces hyperphagia, obesity, hyperinsulinaemia, hyperleptinaemia and hypertension in rats programmed to develop the metabolic syndrome by fetal programming(Reference Vickers, Ikenasio and Breier378). However, IGF-I administration does not exhibit anorectic effects in sheep(Reference Foster, Ames and Emery379). Singularly, another study reported that central injection of IGF-II, but not IGF-I, reduces short-term food intake in rats(Reference Lauterio, Marson and Daughaday380). It has been recently reported that IGF-I may play an important role in thermogenesis(Reference Sanchez-Alavez, Osborn and Tabarean375). Administration of IGF-I to the preoptic area, a hypothalamic region involved in the control of thermoregulation, produces hyperthermia involving activation of BAT in mice(Reference Sanchez-Alavez, Osborn and Tabarean375). This thermogenic effect was accompanied by a switch from glycolysis to fatty acid oxidation and appears to be dependent of the insulin receptor, since it is absent in mice lacking the neuronal insulin receptor. These findings suggest a more important role of the IGF system in energy expenditure than previously thought(Reference Sanchez-Alavez, Osborn and Tabarean375).

Although IGFBP are generally thought to inhibit the action of IGF through high-affinity binding which prevents interaction with IGF receptors, IGFBP can potentially either inhibit or enhance IGF actions(Reference Kawai and Rosen368, Reference Firth and Baxter381). Overexpression of IGFBP2 by adenovirus prevents weight gain and hyperglycaemia in diet-induced obese mice(Reference Wheatcroft, Kearney and Shah382). Moreover, it reverses diabetes at the same time as reducing food intake and inhibits body weight gain in insulin-resistant ob/ob mice by unexplored mechanisms(Reference Hedbacker, Birsoy and Wysocki383).

Fibroblast growth factor 21

FGF21 is a pleiotropic hormone-like protein that has emerged as a major regulator of energy homeostasis(Reference Kharitonenkov and Larsen384). Production of FGF21 takes place mainly in the liver(Reference Tyynismaa, Raivio and Hakkarainen385) and is regulated by PPARα(Reference Badman, Pissios and Kennedy386). FGF21 has been shown to be a major regulator of hepatic lipid metabolism in ketotic states, being up-regulated during fasting(Reference Badman, Pissios and Kennedy386, Reference Galman, Lundasen and Kharitonenkov387). FGF21 transgenic mice are resistant to diet-induced obesity, with FGF21 administration reducing serum glucose and TAG levels in obese and diabetic ob/ob and db/db mice(Reference Kharitonenkov, Shiyanova and Koester388). In humans, FGF21 correlates with BMI and may be a novel biomarker for fatty liver(Reference Dushay, Chui and Gopalakrishnan389). Since the expected beneficial effects of endogenous FGF21 for improving glucose tolerance and reducing TAG levels are absent in obese mice and men, obesity has been proposed to be a FGF21-resistant state(Reference Fisher, Chui and Antonellis390).

Intraperitoneal injections or central administration of FGF21 increases energy expenditure, improves insulin sensitivity and reverses hepatic steatosis in diet-induced obese mice(Reference Xu, Lloyd and Hale391) and rats(Reference Sarruf, Thaler and Morton392). The thermogenic effect may be related to the activation of BAT, since it has been reported that this adipose depot may be a source of FGF in response to cold, exhibiting an autocrine role in the stimulation of thermogenesis(Reference Hondares, Iglesias and Giralt393). Although the wide interindividual variation in serum FGF21 observed in humans raises some doubts regarding its therapeutic relevance(Reference Kharitonenkov and Larsen384), the reported metabolic effects of FGF21 highlight the need for more research in order to assess the use of FGF21 for treating metabolic diseases.

Sex hormone-binding globulin

Sex hormone-binding globulin (SHBG) transports androgens and oestrogens in blood and regulates their access to target tissues(Reference Hammond394). SHBG is mainly produced by hepatocytes and its secretion fluctuates, being primarily influenced by metabolic and hormonal factors(Reference Hammond394). Obesity results in reduced hepatic synthesis and blood concentrations of SHBG, with blood levels of SHBG correlating negatively with energy expenditure in postmenopausal women(Reference Svendsen, Hassager and Christiansen395) and with fat accumulation in men(Reference Abate, Haffner and Garg396). Moreover, low circulating concentrations of SHBG are a strong predictor of the risk of T2DM in men and women(Reference Ding, Song and Manson397) as well as of the metabolic syndrome in non-obese men(Reference Kupelian, Page and Araujo194). This finding has been related to the suppressive effect of insulin on SHBG. Furthermore, the combined effect of increased levels of sex hormones previously mentioned, together with the reduced concentrations of SHBG, leads to an increase in the bioavailable androgens and oestrogens which may promote cellular proliferation and inhibit apoptosis in target cells, thereby being involved in the increased risk of cancer associated with obesity(Reference Calle and Kaaks398).

Signals from skeletal muscle

In humans, skeletal muscle represents 40 % of the total body mass and accounts for approximately 20–30 % of the total resting oxygen uptake(Reference Zurlo, Larson and Bogardus399). A large part of the adaptive thermogenic response is determined by skeletal muscle via the process of mitochondrial uncoupling(Reference van den Berg, van Marken Lichtenbelt and Willems van Dijk400). Furthermore, skeletal muscle secretes myostatin, which has been shown to play a role in energy homeostasis(Reference Choi, Yablonka-Reuveni and Kaiyala17). Exercise is well known to exert beneficial effects on energy balance control. Recently, irisin has been identified as an exercise-induced hormone secreted by skeletal muscle that promotes brown adipocyte recruitment in white fat, thereby increasing energy expenditure(Reference Boström, Wu and Jedrychowski401).

Myostatin

Myostatin is a secreted member of the transforming growth factor-β (TGF-β) family that acts as a negative regulator of skeletal muscle growth by signalling through activin receptors(Reference Lee402). During adulthood, the myostatin protein is produced by skeletal muscle, circulates in the blood, and limits muscle mass(Reference Lee402). Myostatin is expressed almost exclusively in skeletal muscle, although detectable levels of myostatin mRNA are also present in adipose tissue. Myostatin overexpression in mice induces a dramatic loss of muscle and adipose tissue mass with normal food intake(Reference Zimmers, Davies and Koniaris403). The loss of fat is concordant with the capacity of myostatin to block adipogenesis(Reference Rebbapragada, Benchabane and Wrana404). As can be expected, mice lacking myostatin exhibit increased muscle mass but, surprisingly, show reduced adiposity(Reference McPherron and Lee405). This finding can be explained by an increased fatty acid oxidation in peripheral tissues through the stimulation of enzymes involved in lipolysis and in mitochondrial fatty acid oxidation(Reference Zhang, McFarlane and Lokireddy406). The decreased fat mass of myostatin-null mice can be further explained by a concomitant stimulation of thermogenesis through the activation of BAT(Reference Zhang, McFarlane and Lokireddy406). Inhibition of myostatin signalling either in skeletal muscle or adipose tissue evidenced that body fat loss is an indirect result of metabolic changes in skeletal muscle(Reference Guo, Jou and Chanturiya407), apparently mediated by increased energy expenditure and leptin sensitivity(Reference Choi, Yablonka-Reuveni and Kaiyala17). Interestingly, skeletal muscle myostatin protein levels and plasma concentrations were higher in extremely obese women, with the former being correlated with the severity of insulin resistance(Reference Hittel, Berggren and Shearer408). Leptin replacement increases muscle mass of ob/ob mice, and this effect is associated with a leptin-induced reduction in the skeletal muscle expression of myostatin(Reference Sáinz, Rodríguez and Catalán409). However, leptin administration to hypoleptinaemic women did not decrease serum levels of myostatin, suggesting that leptin is not probably involved in the regulation of the myostatin axis in humans, although expression of myostatin in skeletal muscle was not measured(Reference Brinkoetter, Magkos and Vamvini410). Recently, the association of myostatin gene polymorphisms with obesity in humans has been reported, although the pathophysiological mechanisms remain to be elucidated(Reference Pan, Ping and Zhu411).

Signals from the kidney

Under normal circumstances the kidney is not directly involved in energy homeostasis. However, it exerts a notable role in the peripheral control of energy metabolism, secreting primary molecules that participate in the renin–Ang system(Reference Kalupahana and Moustaid-Moussa174).

Renin

As mentioned previously, an overactive renin–Ang system has been involved in the development of obesity-associated co-morbidities as well as in energy homeostasis(Reference Kalupahana and Moustaid-Moussa174). Renin catalyses the rate-limiting step of Ang II production(Reference Kalupahana and Moustaid-Moussa174). Mice lacking renin exhibit lower blood pressure and undetectable plasma levels of renin, Ang I and Ang II(Reference Takahashi, Lopez and Cowhig412). Unexpectedly, these mice are resistant to diet-induced obesity via an increased metabolic rate and partly through a gastrointestinal loss of dietary fat, but not from increased locomotor activity or reduced food intake(Reference Takahashi, Li and Hua14). Some, but not all, of the observed alterations were reversed after Ang II administration. Furthermore, it has been reported that transgenic rodents overexpressing renin eat significantly more after 24 h than controls(Reference Szczepanska-Sadowska, Paczwa and Dobruch413) and develop obesity(Reference Uehara, Tsuchida and Kanno414) by mechanisms not related to Ang II(Reference Gratze, Boschmann and Dechend415). These findings suggest that renin inhibitors may be a therapeutic tool against obesity, insulin resistance and their cardiometabolic co-morbidities.

Signals from the heart

The discovery of atrial natriuretic peptide (ANP) showed that the heart is not only a mechanical organ pumping blood through the blood vessels, but also an endocrine organ involved in the regulation of the cardiovascular–renal system and energy metabolism(Reference Kishimoto, Tokudome and Nakao416).

Atrial natriuretic peptides

ANP and brain natriuretic peptide (BNP) are synthesised in the heart and they are considered to exert their predominant effects in lowering blood pressure, controlling blood volume and reducing heart overgrowth in pathological conditions(Reference Beleigoli, Diniz and Ribeiro417). Another related peptide, C-type natriuretic peptide (CNP) is expressed mainly in the central nervous system but also in the vascular endothelial cells and chondrocytes(Reference Pandey418). ANP and BNP preferentially bind to GUCY-A, promoting the production of cGMP and the activation of protein kinase G(Reference Kuhn419). CNP is the physiological ligand for GUCY-B.

All members of the system (natriuretic peptides and their receptors) are expressed in adipose tissue, while their expression levels are altered in obesity(Reference Beleigoli, Diniz and Ribeiro417, Reference Sarzani, Dessi-Fulgheri and Paci420Reference Moro and Smith423). ANP and BNP, but not CNP, have been reported to induce potent lipolytic effects in human adipocytes similar to those exerted by the β-adrenoceptor agonist isoproterenol(Reference Lafontan, Moro and Berlan360, Reference Birkenfeld, Boschmann and Moro424). Moreover, ANP inhibits human visceral adipocyte growth in culture at physiological concentrations(Reference Sarzani, Marcucci and Salvi425). ANP availability is decreased in obesity, with BMI being inversely correlated to circulating ANP and BNP concentrations(Reference Wang, Larson and Levy426).

Besides their physiological role as lipid-mobilising agents, natriuretic peptides are also involved in the regulation of food intake and energy expenditure. CNP suppresses oxygen consumption in BAT in mice by attenuating the sympathetic nervous system activity, possibly under the control of the hypothalamus(Reference Inuzuka, Tamura and Yamada427). Furthermore, it has been shown that natriuretic peptides can promote muscle mitochondrial biogenesis and fat oxidation, preventing the development of obesity and insulin resistance in mice(Reference Miyashita, Itoh and Tsujimoto428). In this sense, intravenously administered ANP induces postprandial lipid oxidation in humans and increases energy expenditure(Reference Birkenfeld, Budziarek and Boschmann429). These findings suggest that natriuretic peptides may represent a promising therapeutic tool for combating obesity and T2DM.

Signals from the thyroid gland

The thyroid gland, through the production of thyroid hormones, is a major determinant of overall energy expenditure and BMR(Reference Kim430).

Thyroid hormones

The thyroid gland produces the parental form of thyroid hormone, thyroxine (T4), and lower amounts of the major active form of thyroid hormone, triiodothyronine (T3)(Reference Baxter and Webb431). T3 is produced by deiodination of T4 in target cells by specific deiodinases. Secretion of T4 by the thyroid gland is stimulated by thyroid-stimulating hormone secreted by the pituitary gland(Reference Yen432). Thyroid hormones bind to thyroid hormone receptor (THR)α and THRβ, which are members of the nuclear hormone receptor family(Reference Zhang and Lazar433). Thyroid hormones affect numerous cellular processes that are relevant for energy homeostasis(Reference Kim430). Thyroid-stimulating hormone is usually moderately increased in obesity, which is a consequence rather than a cause of obesity(Reference Reinehr434). Alterations in thyroid hormones affect body weight. Hypothyroidism is frequently associated with a modest weight gain, decreased metabolic rate and cold intolerance, whereas hyperthyroidism is related to weight loss despite increased appetite and elevated metabolic rate(Reference Reinehr434, Reference Silva435).

Hyperthyroidism in humans and rodents causes increased food intake but reduced body weight compared with euthyroid controls due to increased energy expenditure. The increase in oxygen consumption and body temperature is accompanied by enhanced fatty acid oxidation(Reference Baxter and Webb431). Evidence of the critical role of thyroid hormones on energy homeostasis arises from genetic mouse models lacking THR(Reference Alkemade436, Reference Barros and Gustafsson437). Mice lacking THRα(Reference Park, Zhao and Glidewell-Kenney438) or THRβ(Reference Foryst-Ludwig, Clemenz and Hohmann439) exhibit reduced thermogenesis as well as other metabolic alterations. Although the involvement of thyroid hormones in energy homeostasis is critical, the physiological mechanisms explaining this effect remain elusive. The thermogenic effect of thyroid hormones has been related to accelerated ATP turnover and reduced efficiency of ATP synthesis as well as to changes in the efficiency of metabolic processes downstream from the mitochondria ‘futile and substrate cycles’(Reference Kim430, Reference Silva435). Peripheral administration of T3 increases food intake but also energy expenditure(Reference Dillo440). An increase in hypothalamic AMPK may be mediating the orexigenic effect of T3(Reference Ishii, Kamegai and Tamura441). Similarly, it has been recently described that besides the critical role of T3 stimulating thermogenesis in skeletal muscle(Reference Kim430), thyroid hormone-induced modulation of AMPK activity and lipid metabolism in the hypothalamus and subsequent thermogenic activation of BAT is a major regulator of whole-body energy homeostasis(Reference López, Varela and Vázquez442). Although the information available makes the thyroid system an interesting field for the development of therapeutic drugs in the fight against obesity, available data regarding effectiveness of thyroid hormone therapy for treating obesity are inconclusive(Reference Kaptein, Beale and Chan443).

Signals from bone

Both body fat mass and fat-free mass correlate directly with bone mineral density. Obesity has been proposed to exert a protective role in the development of osteoporosis(Reference Zhao, Liu and Liu444). On the contrary, low BMI is a risk factor for low bone quality and osteoporosis which is largely independent of age and sex(Reference Galusca, Zouch and Germain445). There is a putative ‘endocrine’ interplay between adipose tissue and bone, with adipokines and molecules secreted by osteoblasts and osteoclasts (osteokines) being the links of a bone–adipose tissue axis(Reference Gómez-Ambrosi, Rodríguez and Catalán15). In this sense, recent findings suggest that osteokines may exert an endocrine regulation on glucose homeostasis and body weight(Reference Gómez-Ambrosi, Rodríguez and Catalán15, Reference Clemens and Karsenty446).

Osteopontin

Osteopontin, also known as secreted phosphoprotein-1, bone sialoprotein-1 and early T lymphocyte activation (Eta-1) is a phosphoprotein expressed by a wide variety of cell types, such as osteoclasts, macrophages, hepatocytes and vascular smooth muscle cells, among others(Reference Scatena, Liaw and Giachelli447). Osteopontin exerts important actions on bone turnover, serving as attachment for osteoclasts activating resorption(Reference Scatena, Liaw and Giachelli447, Reference Reinholt, Hultenby and Oldberg448). In addition to bone remodelling, osteopontin is also involved in several pathophysiological processes including inflammation, immunity, neoplastic transformation, progression of metastases, wound healing and cardiovascular function(Reference Scatena, Liaw and Giachelli447). Osteopontin has been shown to be also produced by adipocytes and to play an important role in obesity and obesity-associated insulin resistance(Reference Gómez-Ambrosi and Frühbeck39, Reference Gómez-Ambrosi, Catalán and Ramírez449Reference Chapman, Miles and Ofrecio453). Expression of osteopontin is dramatically increased in adipose tissue from obese individuals(Reference Gómez-Ambrosi, Catalán and Ramírez449, Reference Pietiläinen, Naukkarinen and Rissanen454, Reference Hurtado Del Pozo, Calvo and Vesperinas-Garcia455), suggesting the important role that this protein has in the molecular alterations that take place during adipose tissue enlargement. Moreover, osteopontin has been suggested to play a pivotal role linking obesity to insulin resistance development by promoting inflammation and the accumulation of macrophages in adipose tissue(Reference Nomiyama, Perez-Tilve and Ogawa450, Reference Chapman, Miles and Ofrecio453, Reference Kiefer, Zeyda and Gollinger456). Higher levels of expression of osteopontin have been shown to be related to macrophage accumulation in adipose tissue and to liver steatosis in morbidly obese subjects(Reference Bertola, Deveaux and Bonnafous457), which promote fibrosis progression in non-alcoholic steatohepatitis(Reference Syn, Choi and Liaskou458). Furthermore, it has been recently shown that osteopontin expression is increased in omental adipose tissue of colon cancer patients, suggesting a potential role of osteopontin linking increased inflammation in visceral adiposity with neoplastic processes(Reference Catalán, Gómez-Ambrosi and Rodríguez459).

Osteocalcin

Osteocalcin is a non-collagenous protein marker of osteoblastic activity thought to play a role in mineralisation and Ca homeostasis(Reference Calvo, Eyre and Gundberg460). Osteocalcin is secreted mainly by osteoblasts and its levels decrease in malnutrition, starvation and anorexia nervosa. Osteocalcin has been traditionally considered as a biological marker of bone formation(Reference Calvo, Eyre and Gundberg460) but now has also been shown to play a role in the regulation of metabolism and in the development of CVD(Reference Kanazawa, Yamaguchi and Yamamoto461Reference Ducy463).

The regulation of bone remodelling by leptin led to the hypothesis that bone exerts a role in the feedback control of energy homeostasis(Reference Lee, Sowa and Hinoi464). In this sense, mice lacking the osteoblast-secreted molecule osteocalcin exhibit an increased adiposity and insulin resistance(Reference Lee, Sowa and Hinoi464, Reference Martin465). Osteocalcin is able to improve glucose tolerance in vivo through the stimulation of the expression of insulin and β-cell proliferation and the induction of the expression of adiponectin and genes involved in energy expenditure in adipocytes(Reference Lee, Sowa and Hinoi464, Reference Ferron, Hinoi and Karsenty466). In this sense, administration of osteocalcin improves glucose metabolism and prevents the development of T2DM in mice(Reference Ferron, Hinoi and Karsenty466, Reference Ferron, McKee and Levine467).

Serum osteocalcin levels are positively associated with insulin sensitivity and secretion in non-diabetic subjects as well as in patients with type 2 diabetes(Reference Fernández-Real and Ricart462, Reference Kanazawa, Yamaguchi and Tada468). Interestingly, osteocalcin concentrations are reduced in obese individuals and increase after weight loss in parallel to the reduction in visceral fat mass(Reference Fernández-Real, Izquierdo and Ortega469). Finally, a recent work suggests that further molecules secreted by bone, yet to be identified, affect energy metabolism(Reference Yoshikawa, Kode and Xu470).

Conclusions

In summary, adipose tissue mass and energy homeostasis are regulated by a wide array of molecules derived not only by adipose tissue and the pancreas but also by the gut, liver, skeletal muscle, kidney, heart, thyroid gland and bone. This implies that the control of energy homeostasis is more complex than previously described and that the hypothalamus integrates hundreds of signals from many different peripheral organs. Moreover, many of these signals are able to stimulate thermogenesis in organs such as BAT and skeletal muscle (Fig. 1). The comprehension of these signals will help to better understand the aetiopathology of obesity and will contribute to the development of new therapeutic targets aimed at tackling excess body fat accumulation. More exact and precise knowledge regarding the complex interplay between the diverse and numerous peripheral signals as well as the pathophysiological alterations that take place in the different organs will lead to a better understanding of energy homeostasis and the causes and pathogenesis of obesity.

Fig. 1 Peripheral factors exerting a direct effect on energy homeostasis grouped by source organ or system. Although due to their multiple production organs some of the elements might be included in more than one organ or system, they have been included only under one organ or system for simplicity reasons. These molecules play an important role in energy homeostasis mainly, but not uniquely, through direct actions on the brain regulation of food intake and on the thermogenic activity of brown adipose tissue (BAT). In some cases, the effect on BAT activation is mediated via the hypothalamus. Ang II, angiotensin II; ANP, atrial natriuretic peptide; ASP, acylation-stimulating protein; BNP, brain natriuretic peptide; CCK, cholecystokinin; CNP, c-type natriuretic peptide; FGF19, fibroblast growth factor-19; FGF21, fibroblast growth factor-21; GIP, glucose-dependent insulinotropic polypeptide; GLP-1, glucagon-like peptide-1; IGF, insulin-like growth factor; PP, pancreatic polypeptide; PYY3–36, peptide YY (peptide tyrosine-tyrosine); SHBG, sex hormone-binding globulin; T3, triiodothyronine; T4, thyroxine. * Although CNP is mainly expressed in the central nervous system, it is also expressed in vascular cells (A colour version of this figure can be found online at http://www.journals.cambridge.org/nrr).

Acknowledgements

The present review was supported by grants from the Instituto de Salud Carlos III (ISCIII; no. FIS PI081146, PS09/02330, PI09/91029 and PI11/02681) and the Departments of Health (3/2006 and 31/2009) and Education (res228/2008) of the Gobierno de Navarra. CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn) is an initiative of the ISCIII, Spain. The authors gratefully acknowledge Laura Stokes for the diligent English editing of the manuscript.

All authors have contributed to the writing and revision of the text and have approved the final manuscript.

There are no conflicts of interest to declare.

References

1Swinburn, BA, Sacks, G, Hall, KD, et al. (2011) The global obesity pandemic: shaped by global drivers and local environments. Lancet 378, 804814.CrossRefGoogle ScholarPubMed
2Finucane, MM, Stevens, GA, Cowan, MJ, et al. (2011) National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet 377, 557567.CrossRefGoogle ScholarPubMed
3Kahn, SE, Hull, RL & Utzschneider, KM (2006) Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444, 840846.CrossRefGoogle ScholarPubMed
4Van Gaal, LF, Mertens, IL & De Block, CE (2006) Mechanisms linking obesity with cardiovascular disease. Nature 444, 875880.CrossRefGoogle ScholarPubMed
5Donath, MY & Shoelson, SE (2011) Type 2 diabetes as an inflammatory disease. Nat Rev Immunol 11, 98107.CrossRefGoogle ScholarPubMed
6Gregor, MF & Hotamisligil, GS (2011) Inflammatory mechanisms in obesity. Annu Rev Immunol 29, 415445.CrossRefGoogle ScholarPubMed
7Calle, EE (2007) Obesity and cancer. BMJ 335, 11071108.CrossRefGoogle ScholarPubMed
8Fabbrini, E, Sullivan, S & Klein, S (2010) Obesity and nonalcoholic fatty liver disease: biochemical, metabolic, and clinical implications. Hepatology 51, 679689.CrossRefGoogle ScholarPubMed
9Pischon, T, Boeing, H, Hoffmann, K, et al. (2008) General and abdominal adiposity and risk of death in Europe. N Engl J Med 359, 21052120.CrossRefGoogle ScholarPubMed
10Berrington de Gonzalez, A, Hartge, P, Cerhan, JR, et al. (2010) Body-mass index and mortality among 1.46 million white adults. N Engl J Med 363, 22112219.CrossRefGoogle ScholarPubMed
11O'Rahilly, S (2009) Human genetics illuminates the paths to metabolic disease. Nature 462, 307314.CrossRefGoogle ScholarPubMed
12Woods, SC (2009) The control of food intake: behavioral versus molecular perspectives. Cell Metab 9, 489498.CrossRefGoogle ScholarPubMed
13Frühbeck, G & Gómez-Ambrosi, J (2003) Control of body weight: a physiologic and transgenic perspective. Diabetologia 46, 143172.CrossRefGoogle ScholarPubMed
14Takahashi, N, Li, F, Hua, K, et al. (2007) Increased energy expenditure, dietary fat wasting, and resistance to diet-induced obesity in mice lacking renin. Cell Metab 6, 506512.CrossRefGoogle ScholarPubMed
15Gómez-Ambrosi, J, Rodríguez, A, Catalán, V, et al. (2008) The bone–adipose axis in obesity and weight loss. Obes Surg 18, 11341143.CrossRefGoogle ScholarPubMed
16Valentino, MA, Lin, JE, Snook, AE, et al. (2011) A uroguanylin–GUCY2C endocrine axis regulates feeding in mice. J Clin Invest 121, 35783588.CrossRefGoogle ScholarPubMed
17Choi, SJ, Yablonka-Reuveni, Z, Kaiyala, KJ, et al. (2011) Increased energy expenditure and leptin sensitivity account for low fat mass in myostatin-deficient mice. Am J Physiol Endocrinol Metab 300, E1031E1037.CrossRefGoogle ScholarPubMed
18Somogyi, V, Gyorffy, A, Scalise, TJ, et al. (2011) Endocrine factors in the hypothalamic regulation of food intake in females: a review of the physiological roles and interactions of ghrelin, leptin, thyroid hormones, oestrogen and insulin. Nutr Res Rev 24, 132154.CrossRefGoogle ScholarPubMed
19Field, BC, Chaudhri, OB & Bloom, SR (2010) Bowels control brain: gut hormones and obesity. Nat Rev Endocrinol 6, 444453.CrossRefGoogle ScholarPubMed
20Kennedy, GC (1953) The role of depot fat in the hypothalamic control of food intake in the rat. Proc Royal Soc London 140B, 579592.Google Scholar
21Frühbeck, G & Gómez Ambrosi, J (2001) Rationale for the existence of additional adipostatic hormones. FASEB J 15, 19962006.CrossRefGoogle ScholarPubMed
22Frühbeck, G & Salvador, J (2004) Role of adipocytokines in metabolism and disease. Nutr Res 24, 803826.CrossRefGoogle Scholar
23Frühbeck, G, Gómez-Ambrosi, J, Muruzábal, FJ, et al. (2001) The adipocyte: a model for integration of endocrine and metabolic signaling in energy metabolism regulation. Am J Physiol Endocrinol Metab 280, E827E847.CrossRefGoogle Scholar
24Frühbeck, G & Gómez-Ambrosi, J (2005) Adipose tissue. In Encyclopedia of Human Nutrition, 2nd ed., pp. 114 [, editors]. Oxford, UK: Elsevier Ltd.Google Scholar
25Catalán, V, Gómez-Ambrosi, J, Rodríguez, A, et al. (2009) Adipokines in the treatment of diabetes mellitus and obesity. Expert Opin Pharmacother 10, 239254.CrossRefGoogle ScholarPubMed
26Zhang, Y, Proenca, R, Maffei, M, et al. (1994) Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425432.CrossRefGoogle ScholarPubMed
27Morton, GJ, Cummings, DE, Baskin, DG, et al. (2006) Central nervous system control of food intake and body weight. Nature 443, 289295.CrossRefGoogle ScholarPubMed
28Myers, MG Jr, Munzberg, H, Leinninger, GM, et al. (2009) The geometry of leptin action in the brain: more complicated than a simple ARC. Cell Metab 9, 117123.CrossRefGoogle ScholarPubMed
29Tartaglia, LA, Dembski, M, Weng, X, et al. (1995) Identification and expression cloning of a leptin receptor, OB-R. Cell 83, 12631271.CrossRefGoogle ScholarPubMed
30Lee, GH, Proenca, R, Montez, JM, et al. (1996) Abnormal splicing of the leptin receptor in diabetic mice. Nature 379, 632635.CrossRefGoogle ScholarPubMed
31Frühbeck, G, Gómez-Ambrosi, J & Martínez, JA (1999) Pre- and postprandial expression of the leptin receptor splice variants OB-Ra and OB-Rb in murine peripheral tissues. Physiol Res 48, 189195.Google ScholarPubMed
32Frühbeck, G, Jebb, SA & Prentice, AM (1998) Leptin: physiology and pathophysiology. Clin Physiol 18, 399419.CrossRefGoogle ScholarPubMed
33Frühbeck, G & Salvador, J (2000) Relations between leptin and the regulation of glucose metabolism. Diabetologia 43, 312.Google ScholarPubMed
34Muruzábal, FJ, Frühbeck, G, Gómez-Ambrosi, J, et al. (2002) Immunocytochemical detection of leptin in non-mammalian vertebrate stomach. Gen Comp Endocrinol 128, 149152.CrossRefGoogle ScholarPubMed
35Archanco, M, Muruzábal, FJ, Llopiz, D, et al. (2003) Leptin expression in the rat ovary depends on estrous cycle. J Histochem Cytochem 51, 12691277.CrossRefGoogle ScholarPubMed
36Myers, MG, Cowley, MA & Münzberg, H (2008) Mechanisms of leptin action and leptin resistance. Annu Rev Physiol 70, 537556.CrossRefGoogle ScholarPubMed
37Morton, GJ & Schwartz, MW (2011) Leptin and the central nervous system control of glucose metabolism. Physiol Rev 91, 389411.CrossRefGoogle ScholarPubMed
38Gautron, L & Elmquist, JK (2011) Sixteen years and counting: an update on leptin in energy balance. J Clin Invest 121, 20872093.CrossRefGoogle ScholarPubMed
39Gómez-Ambrosi, J & Frühbeck, G (2007) Unlocking the molecular basis of obesity. Future Lipidol 2, 577581.CrossRefGoogle Scholar
40Frühbeck, G (2006) Intracellular signalling pathways activated by leptin. Biochem J 393, 720.CrossRefGoogle ScholarPubMed
41Lindström, P (2007) The physiology of obese–hyperglycemic mice [ob/ob mice]. Sci World J 7, 666685.CrossRefGoogle ScholarPubMed
42Pelleymounter, MA, Cullen, MJ, Baker, MB, et al. (1995) Effects of the obese gene product on body weight regulation in ob/ob mice. Science 269, 540543.CrossRefGoogle ScholarPubMed
43Halaas, JL, Gajiwala, KS, Maffei, M, et al. (1995) Weight-reducing effects of the plasma protein encoded by the obese gene. Science 269, 543546.CrossRefGoogle ScholarPubMed
44Siegrist-Kaiser, CA, Pauli, V, Juge-Aubry, CE, et al. (1997) Direct effects of leptin on brown and white adipose tissue. J Clin Invest 100, 28582864.CrossRefGoogle ScholarPubMed
45Frühbeck, G, Aguado, M, Gómez-Ambrosi, J, et al. (1998) Lipolytic effect of in vivo leptin administration on adipocytes of lean and ob/ob mice, but not db/db mice. Biochem Biophys Res Commun 250, 99102.CrossRefGoogle Scholar
46Gallardo, N, Bonzón-Kulichenko, E, Fernández-Agulloó, T, et al. (2007) Tissue-specific effects of central leptin on the expression of genes involved in lipid metabolism in liver and white adipose tissue. Endocrinology 148, 56045610.CrossRefGoogle ScholarPubMed
47Wang, MY, Lee, Y & Unger, RH (1999) Novel form of lipolysis induced by leptin. J Biol Chem 274, 1754117544.CrossRefGoogle ScholarPubMed
48Hwa, JJ, Fawzi, AB, Graziano, MP, et al. (1997) Leptin increases energy expenditure and selectively promotes fat metabolism in ob/ob mice. Am J Physiol 272, R1204R1209.Google ScholarPubMed
49Montague, CT, Farooqi, IS, Whitehead, JP, et al. (1997) Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387, 903908.CrossRefGoogle ScholarPubMed
50Strobel, A, Issad, T, Camoin, L, et al. (1998) A leptin missense mutation associated with hypogonadism and morbid obesity. Nat Genet 18, 213215.CrossRefGoogle ScholarPubMed
51Farooqi, IS, Jebb, SA, Langmack, G, et al. (1999) Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med 341, 879884.CrossRefGoogle Scholar
52Farooqi, IS, Matarese, G, Lord, GM, et al. (2002) Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest 110, 10931103.CrossRefGoogle ScholarPubMed
53Licinio, J, Caglayan, S, Ozata, M, et al. (2004) Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc Natl Acad Sci U S A 101, 45314536.CrossRefGoogle ScholarPubMed
54Paz-Filho, G, Wong, ML & Licinio, J (2011) Ten years of leptin replacement therapy. Obes Rev 12, e315e323.CrossRefGoogle ScholarPubMed
55Rosenbaum, M, Goldsmith, R, Bloomfield, D, et al. (2005) Low-dose leptin reverses skeletal muscle, autonomic, and neuroendocrine adaptations to maintenance of reduced weight. J Clin Invest 115, 35793586.CrossRefGoogle ScholarPubMed
56Galgani, JE, Greenway, FL, Caglayan, S, et al. (2010) Leptin replacement prevents weight loss-induced metabolic adaptation in congenital leptin-deficient patients. J Clin Endocrinol Metab 95, 851855.CrossRefGoogle ScholarPubMed
57Kadowaki, T & Yamauchi, T (2005) Adiponectin and adiponectin receptors. Endocr Rev 26, 439451.CrossRefGoogle ScholarPubMed
58Trujillo, ME & Scherer, PE (2005) Adiponectin – journey from an adipocyte secretory protein to biomarker of the metabolic syndrome. J Intern Med 257, 167175.CrossRefGoogle ScholarPubMed
59Kadowaki, T, Yamauchi, T, Kubota, N, et al. (2006) Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest 116, 17841792.CrossRefGoogle ScholarPubMed
60Yamauchi, T, Kamon, J, Ito, Y, et al. (2003) Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 423, 762769.CrossRefGoogle ScholarPubMed
61Hug, C, Wang, J, Ahmad, NS, et al. (2004) T-cadherin is a receptor for hexameric and high-molecular-weight forms of Acrp30/adiponectin. Proc Natl Acad Sci U S A 101, 1030810313.CrossRefGoogle ScholarPubMed
62Arita, Y, Kihara, S, Ouchi, N, et al. (1999) Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun 257, 7983.CrossRefGoogle ScholarPubMed
63Weyer, C, Funahashi, T, Tanaka, S, et al. (2001) Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab 86, 19301935.CrossRefGoogle ScholarPubMed
64Yang, WS, Lee, WJ, Funahashi, T, et al. (2001) Weight reduction increases plasma levels of an adipose-derived anti-inflammatory protein, adiponectin. J Clin Endocrinol Metab 86, 38153819.CrossRefGoogle ScholarPubMed
65Hotta, K, Funahashi, T, Arita, Y, et al. (2000) Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol 20, 15951599.CrossRefGoogle ScholarPubMed
66Spranger, J, Kroke, A, Mohlig, M, et al. (2003) Adiponectin and protection against type 2 diabetes mellitus. Lancet 361, 226228.CrossRefGoogle ScholarPubMed
67Ouchi, N, Kihara, S, Arita, Y, et al. (1999) Novel modulator for endothelial adhesion molecules: adipocyte-derived plasma protein adiponectin. Circulation 100, 24732476.CrossRefGoogle ScholarPubMed
68Berg, AH, Combs, TP, Du, X, et al. (2001) The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat Med 7, 947953.CrossRefGoogle ScholarPubMed
69Combs, TP, Wagner, JA, Berger, J, et al. (2002) Induction of adipocyte complement-related protein of 30 kilodaltons by PPARγ agonists: a potential mechanism of insulin sensitization. Endocrinology 143, 9981007.CrossRefGoogle ScholarPubMed
70Matsuzawa, Y, Funahashi, T, Kihara, S, et al. (2004) Adiponectin and metabolic syndrome. Arterioscler Thromb Vasc Biol 24, 2933.CrossRefGoogle ScholarPubMed
71Maeda, N, Shimomura, I, Kishida, K, et al. (2002) Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nat Med 8, 731737.CrossRefGoogle ScholarPubMed
72Kubota, N, Terauchi, Y, Yamauchi, T, et al. (2002) Disruption of adiponectin causes insulin resistance and neointimal formation. J Biol Chem 277, 2586325866.CrossRefGoogle ScholarPubMed
73Yamauchi, T, Nio, Y, Maki, T, et al. (2007) Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat Med 13, 332339.CrossRefGoogle ScholarPubMed
74Dridi, S & Taouis, M (2009) Adiponectin and energy homeostasis: consensus and controversy. J Nutr Biochem 20, 831839.CrossRefGoogle ScholarPubMed
75Qi, Y, Takahashi, N, Hileman, SM, et al. (2004) Adiponectin acts in the brain to decrease body weight. Nat Med 10, 524529.CrossRefGoogle ScholarPubMed
76Park, S, Kim, DS, Kwon, DY, et al. (2011) Long-term central infusion of adiponectin improves energy and glucose homeostasis by decreasing fat storage and suppressing hepatic gluconeogenesis without changing food intake. J Neuroendocrinol 23, 687698.CrossRefGoogle ScholarPubMed
77Kubota, N, Yano, W, Kubota, T, et al. (2007) Adiponectin stimulates AMP-activated protein kinase in the hypothalamus and increases food intake. Cell Metab 6, 5568.CrossRefGoogle ScholarPubMed
78Frühbeck, G, Alonso, R, Marzo, F, et al. (1995) A modified method for the indirect quantitative analysis of phytate in foodstuffs. Anal Biochem 225, 206212.CrossRefGoogle ScholarPubMed
79Steppan, CM, Bailey, ST, Bhat, S, et al. (2001) The hormone resistin links obesity to diabetes. Nature 409, 307312.CrossRefGoogle ScholarPubMed
80McTernan, PG, Kusminski, CM & Kumar, S (2006) Resistin. Curr Opin Lipidol 17, 170175.CrossRefGoogle ScholarPubMed
81Gómez-Ambrosi, J & Frühbeck, G (2005) Evidence for the involvement of resistin in inflammation and cardiovascular disease. Curr Diabetes Rev 1, 227234.CrossRefGoogle ScholarPubMed
82Rangwala, SM, Rich, AS, Rhoades, B, et al. (2004) Abnormal glucose homeostasis due to chronic hyperresistinemia. Diabetes 53, 19371941.CrossRefGoogle ScholarPubMed
83Satoh, H, Nguyen, MT, Miles, PD, et al. (2004) Adenovirus-mediated chronic ‘hyper-resistinemia’ leads to in vivo insulin resistance in normal rats. J Clin Invest 114, 224231.CrossRefGoogle ScholarPubMed
84Qi, Y, Nie, Z, Lee, Y-S, et al. (2006) Loss of resistin improves glucose homeostasis in leptin deficiency. Diabetes 55, 30833090.CrossRefGoogle ScholarPubMed
85Kusminski, CM, McTernan, PG & Kumar, S (2005) Role of resistin in obesity, insulin resistance and type II diabetes. Clin Sci 109, 243256.CrossRefGoogle ScholarPubMed
86Schwartz, DR & Lazar, MA (2011) Human resistin: found in translation from mouse to man. Trends Endocrinol Metab 22, 259265.Google ScholarPubMed
87Azuma, K, Katsukawa, F, Oguchi, S, et al. (2003) Correlation between serum resistin level and adiposity in obese individuals. Obes Res 11, 9971001.CrossRefGoogle ScholarPubMed
88Degawa-Yamauchi, M, Bovenkerk, JE, Juliar, BE, et al. (2003) Serum resistin (FIZZ3) protein is increased in obese humans. J Clin Endocrinol Metab 88, 54525455.CrossRefGoogle ScholarPubMed
89Silha, JV, Krsek, M, Skrha, JV, et al. (2003) Plasma resistin, adiponectin and leptin levels in lean and obese subjects: correlations with insulin resistance. Eur J Endocrinol 149, 331335.CrossRefGoogle ScholarPubMed
90Lee, JH, Chan, JL, Yiannakouris, N, et al. (2003) Circulating resistin levels are not associated with obesity or insulin resistance in humans and are not regulated by fasting or leptin administration: cross-sectional and interventional studies in normal, insulin-resistant, and diabetic subjects. J Clin Endocrinol Metab 88, 48484856.CrossRefGoogle ScholarPubMed
91Heilbronn, LK, Rood, J, Janderova, L, et al. (2004) Relationship between serum resistin concentrations and insulin resistance in nonobese, obese, and obese diabetic subjects. J Clin Endocrinol Metab 89, 18441848.CrossRefGoogle ScholarPubMed
92Fehmann, HC & Heyn, J (2002) Plasma resistin levels in patients with type 1 and type 2 diabetes mellitus and in healthy controls. Horm Metab Res 34, 671673.CrossRefGoogle ScholarPubMed
93Chen, MP, Chung, FM, Chang, DM, et al. (2006) Elevated plasma level of visfatin/pre-B cell colony-enhancing factor in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab 91, 295299.CrossRefGoogle ScholarPubMed
94Heidemann, C, Sun, Q, van Dam, RM, et al. (2008) Total and high-molecular-weight adiponectin and resistin in relation to the risk for type 2 diabetes in women. Ann Intern Med 149, 307316.CrossRefGoogle Scholar
95Utzschneider, KM, Carr, DB, Tong, J, et al. (2005) Resistin is not associated with insulin sensitivity or the metabolic syndrome in humans. Diabetologia 48, 23302333.CrossRefGoogle ScholarPubMed
96Gómez-Ambrosi, J & Frühbeck, G (2001) Do resistin and resistin-like molecules also link obesity to inflammatory diseases? Ann Intern Med 135, 306307.CrossRefGoogle ScholarPubMed
97Bokarewa, M, Nagaev, I, Dahlberg, L, et al. (2005) Resistin, an adipokine with potent proinflammatory properties. J Immunol 174, 57895795.CrossRefGoogle ScholarPubMed
98Silswal, N, Singh, AK, Aruna, B, et al. (2005) Human resistin stimulates the pro-inflammatory cytokines TNF-α and IL-12 in macrophages by NF-κB-dependent pathway. Biochem Biophys Res Commun 334, 10921101.CrossRefGoogle ScholarPubMed
99Kaser, S, Kaser, A, Sandhofer, A, et al. (2003) Resistin messenger-RNA expression is increased by proinflammatory cytokines in vitro. Biochem Biophys Res Commun 309, 286290.CrossRefGoogle ScholarPubMed
100Lehrke, M, Reilly, MP, Millington, SC, et al. (2004) An inflammatory cascade leading to hyperresistinemia in humans. PLoS Med 1, e45.CrossRefGoogle ScholarPubMed
101Reilly, MP, Lehrke, M, Wolfe, ML, et al. (2005) Resistin is an inflammatory marker of atherosclerosis in humans. Circulation 111, 932939.CrossRefGoogle ScholarPubMed
102Kusminski, CM, da Silva, NF, Creely, SJ, et al. (2007) The in vitro effects of resistin on the innate immune signaling pathway in isolated human subcutaneous adipocytes. J Clin Endocrinol Metab 92, 270276.CrossRefGoogle ScholarPubMed
103Tovar, S, Nogueiras, R, Tung, LY, et al. (2005) Central administration of resistin promotes short-term satiety in rats. Eur J Endocrinol 153, R1R5.CrossRefGoogle ScholarPubMed
104Park, S, Hong, SM, Sung, SR, et al. (2008) Long-term effects of central leptin and resistin on body weight, insulin resistance, and β-cell function and mass by the modulation of hypothalamic leptin and insulin signaling. Endocrinology 149, 445454.CrossRefGoogle ScholarPubMed
105Cifani, C, Durocher, Y, Pathak, A, et al. (2009) Possible common central pathway for resistin and insulin in regulating food intake. Acta Physiol 196, 395400.CrossRefGoogle ScholarPubMed
106Vázquez, MJ, González, CR, Varela, L, et al. (2008) Central resistin regulates hypothalamic and peripheral lipid metabolism in a nutritional-dependent fashion. Endocrinology 149, 45344543.CrossRefGoogle Scholar
107Singhal, NS, Lazar, MA & Ahima, RS (2007) Central resistin induces hepatic insulin resistance via neuropeptide Y. J Neurosci 27, 1292412932.CrossRefGoogle ScholarPubMed
108Kim, KH, Lee, K, Moon, YS, et al. (2001) A cysteine-rich adipose tissue-specific secretory factor inhibits adipocyte differentiation. J Biol Chem 276, 1125211256.CrossRefGoogle ScholarPubMed
109Nogueiras, R, Novelle, MG, Vazquez, MJ, et al. (2010) Resistin: regulation of food intake, glucose homeostasis and lipid metabolism. Endocr Dev 17, 175184.CrossRefGoogle ScholarPubMed
110Maslowska, M, Sniderman, AD, Germinario, R, et al. (1997) ASP stimulates glucose transport in cultured human adipocytes. Int J Obes Relat Metab Disord 21, 261266.CrossRefGoogle ScholarPubMed
111Cianflone, K, Xia, Z & Chen, LY (2003) Critical review of acylation-stimulating protein physiology in humans and rodents. Biochim Biophys Acta 1609, 127143.CrossRefGoogle ScholarPubMed
112Yasruel, Z, Cianflone, K, Sniderman, AD, et al. (1991) Effect of acylation stimulating protein on the triacylglycerol synthetic pathway of human adipose tissue. Lipids 26, 495499.CrossRefGoogle ScholarPubMed
113Germinario, R, Sniderman, AD, Manuel, S, et al. (1993) Coordinate regulation of triacylglycerol synthesis and glucose transport by acylation-stimulating protein. Metabolism 42, 574580.CrossRefGoogle ScholarPubMed
114Saleh, J, Al-Wardy, N, Farhan, H, et al. (2011) Acylation stimulating protein: a female lipogenic factor? Obes Rev 12, 440448.CrossRefGoogle ScholarPubMed
115Xia, Z, Stanhope, KL, Digitale, E, et al. (2004) Acylation-stimulating protein (ASP)/complement C3adesArg deficiency results in increased energy expenditure in mice. J Biol Chem 279, 40514057.CrossRefGoogle ScholarPubMed
116Xia, Z, Sniderman, AD & Cianflone, K (2002) Acylation-stimulating protein (ASP) deficiency induces obesity resistance and increased energy expenditure in ob/ob mice. J Biol Chem 277, 4587445879.CrossRefGoogle ScholarPubMed
117Paglialunga, S, Schrauwen, P, Roy, C, et al. (2007) Reduced adipose tissue triglyceride synthesis and increased muscle fatty acid oxidation in C5L2 knockout mice. J Endocrinol 194, 293304.CrossRefGoogle ScholarPubMed
118Paglialunga, S, Fisette, A, Munkonda, M, et al. (2010) The effects of acylation stimulating protein supplementation VS antibody neutralization on energy expenditure in wildtype mice. BMC Physiol 10, 4.CrossRefGoogle ScholarPubMed
119Roy, C, Roy, MC, Gauvreau, D, et al. (2011) Acute injection of ASP in the third ventricle inhibits food intake and locomotor activity in rats. Am J Physiol Endocrinol Metab 301, E232E241.CrossRefGoogle ScholarPubMed
120Rodríguez, A, Catalán, V, Gómez-Ambrosi, J, et al. (2007) Visceral and subcutaneous adiposity: are both potential therapeutic targets for tackling the metabolic syndrome? Curr Pharm Des 13, 21692175.CrossRefGoogle ScholarPubMed
121Cawthorn, WP, Heyd, F, Hegyi, K, et al. (2007) Tumour necrosis factor-α inhibits adipogenesis via a β-catenin/TCF4(TCF7L2)-dependent pathway. Cell Death Differ 14, 13611373.CrossRefGoogle Scholar
122Cawthorn, WP & Sethi, JK (2008) TNF-α and adipocyte biology. FEBS Lett 582, 117131.CrossRefGoogle ScholarPubMed
123Hotamisligil, GS (2006) Inflammation and metabolic disorders. Nature 444, 860867.CrossRefGoogle ScholarPubMed
124Uysal, KT, Wiesbrock, SM, Marino, MW, et al. (1997) Protection from obesity-induced insulin resistance in mice lacking TNF-α function. Nature 389, 610614.CrossRefGoogle ScholarPubMed
125Tisdale, MJ (2009) Mechanisms of cancer cachexia. Physiol Rev 89, 381410.CrossRefGoogle ScholarPubMed
126de Kloet, AD, Pacheco-Lopez, G, Langhans, W, et al. (2011) The effect of TNFα on food intake and central insulin sensitivity in rats. Physiol Behav 103, 1720.CrossRefGoogle ScholarPubMed
127Amaral, ME, Barbuio, R, Milanski, M, et al. (2006) Tumor necrosis factor-α activates signal transduction in hypothalamus and modulates the expression of pro-inflammatory proteins and orexigenic/anorexigenic neurotransmitters. J Neurochem 98, 203212.CrossRefGoogle ScholarPubMed
128Frühbeck, G, Sesma, P & Burrell, MA (2009) PRDM16: the interconvertible adipo-myocyte switch. Trends Cell Biol 19, 141146.CrossRefGoogle ScholarPubMed
129Frühbeck, G, Becerril, S, Sáinz, N, et al. (2009) BAT: a new target for human obesity? Trends Pharmacol Sci 30, 387396.CrossRefGoogle Scholar
130Cypess, AM, Lehman, S, Williams, G, et al. (2009) Identification and importance of brown adipose tissue in adult humans. N Engl J Med 360, 15091517.CrossRefGoogle ScholarPubMed
131Masaki, T, Yoshimatsu, H, Chiba, S, et al. (1999) Tumor necrosis factor-α regulates in vivo expression of the rat UCP family differentially. Biochim Biophys Acta 1436, 585592.CrossRefGoogle ScholarPubMed
132Valladares, A, Roncero, C, Benito, M, et al. (2001) TNF-α inhibits UCP-1 expression in brown adipocytes via ERKs. Opposite effect of p38MAPK. FEBS Lett 493, 611.CrossRefGoogle ScholarPubMed
133Romanatto, T, Roman, EA, Arruda, AP, et al. (2009) Deletion of tumor necrosis factor-α receptor 1 (TNFR1) protects against diet-induced obesity by means of increased thermogenesis. J Biol Chem 284, 3621336222.CrossRefGoogle ScholarPubMed
134Bastard, JP, Maachi, M, Van Nhieu, JT, et al. (2002) Adipose tissue IL-6 content correlates with resistance to insulin activation of glucose uptake both in vivo and in vitro. J Clin Endocrinol Metab 87, 20842089.CrossRefGoogle ScholarPubMed
135Tilg, H & Moschen, AR (2008) Inflammatory mechanisms in the regulation of insulin resistance. Mol Med 14, 222231.CrossRefGoogle ScholarPubMed
136Wallenius, V, Wallenius, K, Ahren, B, et al. (2002) Interleukin-6-deficient mice develop mature-onset obesity. Nat Med 8, 7579.CrossRefGoogle ScholarPubMed
137Carey, AL, Steinberg, GR, Macaulay, SL, et al. (2006) Interleukin-6 increases insulin-stimulated glucose disposal in humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated protein kinase. Diabetes 55, 26882697.CrossRefGoogle ScholarPubMed
138Wallenius, K, Wallenius, V, Sunter, D, et al. (2002) Intracerebroventricular interleukin-6 treatment decreases body fat in rats. Biochem Biophys Res Commun 293, 560565.CrossRefGoogle ScholarPubMed
139Chida, D, Osaka, T, Hashimoto, O, et al. (2006) Combined interleukin-6 and interleukin-1 deficiency causes obesity in young mice. Diabetes 55, 971977.CrossRefGoogle ScholarPubMed
140Benrick, A, Schele, E, Pinnock, SB, et al. (2009) Interleukin-6 gene knockout influences energy balance regulating peptides in the hypothalamic paraventricular and supraoptic nuclei. J Neuroendocrinol 21, 620628.CrossRefGoogle ScholarPubMed
141Stouthard, JM, Romijn, JA, Van der Poll, T, et al. (1995) Endocrinologic and metabolic effects of interleukin-6 in humans. Am J Physiol 268, E813E819.Google ScholarPubMed
142Tsigos, C, Papanicolaou, DA, Defensor, R, et al. (1997) Dose effects of recombinant human interleukin-6 on pituitary hormone secretion and energy expenditure. Neuroendocrinology 66, 5462.CrossRefGoogle ScholarPubMed
143Kubaszek, A, Pihlajamaki, J, Punnonen, K, et al. (2003) The C-174G promoter polymorphism of the IL-6 gene affects energy expenditure and insulin sensitivity. Diabetes 52, 558561.CrossRefGoogle ScholarPubMed
144Hoene, M & Weigert, C (2008) The role of interleukin-6 in insulin resistance, body fat distribution and energy balance. Obes Rev 9, 2029.CrossRefGoogle ScholarPubMed
145Ouchi, N, Parker, JL, Lugus, JJ, et al. (2011) Adipokines in inflammation and metabolic disease. Nat Rev Immunol 11, 8597.CrossRefGoogle ScholarPubMed
146Fukuhara, A, Matsuda, M, Nishizawa, M, et al. (2005) Visfatin: a protein secreted by visceral fat that mimics the effects of insulin. Science 307, 426430.CrossRefGoogle ScholarPubMed
147Fukuhara, A, Matsuda, M, Nishizawa, M, et al. (2007) Visfatin: a protein secreted by visceral fat that mimics the effects of insulin: retraction. Science 318, 565.CrossRefGoogle Scholar
148Berndt, J, Klöting, N, Kralisch, S, et al. (2005) Plasma visfatin concentrations and fat depot-specific mRNA expression in humans. Diabetes 54, 29112916.CrossRefGoogle ScholarPubMed
149Catalán, V, Gómez-Ambrosi, J, Rodríguez, A, et al. (2011) Association of increased visfatin/PBEF/NAMPT circulating concentrations and gene expression levels in peripheral blood cells with lipid metabolism and fatty liver in human morbid obesity. Nutr Metab Cardiovasc Dis 21, 245253.Google ScholarPubMed
150Revollo, JR, Korner, A, Mills, KF, et al. (2007) Nampt/PBEF/visfatin regulates insulin secretion in β cells as a systemic NAD biosynthetic enzyme. Cell Metab 6, 363375.CrossRefGoogle ScholarPubMed
151Moschen, AR, Kaser, A, Enrich, B, et al. (2007) Visfatin, an adipocytokine with proinflammatory and immunomodulating properties. J Immunol 178, 17481758.CrossRefGoogle ScholarPubMed
152Park, BS, Jin, SH, Park, JJ, et al. (2011) Visfatin induces sickness responses in the brain. PLoS ONE 6, e15981.CrossRefGoogle ScholarPubMed
153Arner, P (2006) Editorial: visfatin – a true or false trail to type 2 diabetes mellitus. J Clin Endocrinol Metab 91, 2830.CrossRefGoogle ScholarPubMed
154Hida, K, Wada, J, Eguchi, J, et al. (2005) Visceral adipose tissue-derived serine protease inhibitor: a unique insulin-sensitizing adipocytokine in obesity. Proc Natl Acad Sci U S A 102, 1061010615.CrossRefGoogle ScholarPubMed
155Klöting, N, Berndt, J, Kralisch, S, et al. (2006) Vaspin gene expression in human adipose tissue: association with obesity and type 2 diabetes. Biochem Biophys Res Commun 339, 430436.CrossRefGoogle ScholarPubMed
156Youn, BS, Klöting, N, Kratzsch, J, et al. (2008) Serum vaspin concentrations in human obesity and type 2 diabetes. Diabetes 57, 372377.CrossRefGoogle ScholarPubMed
157Zvonic, S, Lefevre, M, Kilroy, G, et al. (2007) Secretome of primary cultures of human adipose-derived stem cells: modulation of serpins by adipogenesis. Mol Cell Proteomics 6, 1828.CrossRefGoogle ScholarPubMed
158González, CR, Caminos, JE, Vázquez, MJ, et al. (2009) Regulation of visceral adipose tissue-derived serine protease inhibitor by nutritional status, metformin, gender and pituitary factors in rat white adipose tissue. J Physiol 587, 37413750.CrossRefGoogle ScholarPubMed
159Jeong, E, Youn, BS, Kim, DW, et al. (2010) Circadian rhythm of serum vaspin in healthy male volunteers: relation to meals. J Clin Endocrinol Metab 95, 18691875.CrossRefGoogle ScholarPubMed
160Klöting, N, Kovacs, P, Kern, M, et al. (2011) Central vaspin administration acutely reduces food intake and has sustained blood glucose-lowering effects. Diabetologia 54, 18191823.CrossRefGoogle ScholarPubMed
161Brunetti, L, Di Nisio, C, Recinella, L, et al. (2011) Effects of vaspin, chemerin and omentin-1 on feeding behavior and hypothalamic peptide gene expression in the rat. Peptides 32, 18661871.CrossRefGoogle ScholarPubMed
162Ernst, MC & Sinal, CJ (2010) Chemerin: at the crossroads of inflammation and obesity. Trends Endocrinol Metab 21, 660667.CrossRefGoogle Scholar
163Bozaoglu, K, Bolton, K, McMillan, J, et al. (2007) Chemerin is a novel adipokine associated with obesity and metabolic syndrome. Endocrinology 148, 46874694.CrossRefGoogle ScholarPubMed
164Goralski, KB, McCarthy, TC, Hanniman, EA, et al. (2007) Chemerin: a novel adipokine that regulates adipogenesis and adipocyte metabolism. J Biol Chem 282, 2817528188.CrossRefGoogle ScholarPubMed
165Hart, R & Greaves, DR (2010) Chemerin contributes to inflammation by promoting macrophage adhesion to VCAM-1 and fibronectin through clustering of VLA-4 and VLA-5. J Immunol 185, 37283739.CrossRefGoogle ScholarPubMed
166Sell, H, Divoux, A, Poitou, C, et al. (2010) Chemerin correlates with markers for fatty liver in morbidly obese patients and strongly decreases after weight loss induced by bariatric surgery. J Clin Endocrinol Metab 95, 28922896.CrossRefGoogle ScholarPubMed
167Catalán, V, Gómez-Ambrosi, J, Rodríguez, A, et al. (2011) Increased levels of chemerin and its receptor, chemokine-like receptor-1, in obesity are related to inflammation: tumor necrosis factor-α stimulates mRNA levels of chemerin in visceral adipocytes from obese patients. Surg Obes Relat Dis (epublication ahead of print version 10 November 2011).Google ScholarPubMed
168Schaffler, A, Neumeier, M, Herfarth, H, et al. (2005) Genomic structure of human omentin, a new adipocytokine expressed in omental adipose tissue. Biochim Biophys Acta 1732, 96102.CrossRefGoogle ScholarPubMed
169Yang, RZ, Lee, MJ, Hu, H, et al. (2006) Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. Am J Physiol Endocrinol Metab 290, E1253E1261.CrossRefGoogle ScholarPubMed
170de Souza Batista, CM, Yang, RZ, Lee, MJ, et al. (2007) Omentin plasma levels and gene expression are decreased in obesity. Diabetes 56, 16551661.CrossRefGoogle ScholarPubMed
171Tan, BK, Adya, R & Randeva, HS (2010) Omentin: a novel link between inflammation, diabesity, and cardiovascular disease. Trends Cardiovasc Med 20, 143148.CrossRefGoogle ScholarPubMed
172Moreno-Navarrete, JM, Catalán, V, Ortega, F, et al. (2010) Circulating omentin concentration increases after weight loss. Nutr Metab 7, 27.CrossRefGoogle ScholarPubMed
173Yvan-Charvet, L & Quignard-Boulange, A (2011) Role of adipose tissue renin–angiotensin system in metabolic and inflammatory diseases associated with obesity. Kidney Int 79, 162168.CrossRefGoogle ScholarPubMed
174Kalupahana, NS & Moustaid-Moussa, N (2012) The renin–angiotensin system: a link between obesity, inflammation and insulin resistance. Obes Rev 13, 136149.CrossRefGoogle ScholarPubMed
175Katovich, MJ & Pachori, A (2000) Effects of inhibition of the renin–angiotensin system on the cardiovascular actions of insulin. Diabetes Obes Metab 2, 314.CrossRefGoogle ScholarPubMed
176Luther, JM & Brown, NJ (2011) The renin–angiotensin–aldosterone system and glucose homeostasis. Trends Pharmacol Sci 32, 734739.CrossRefGoogle ScholarPubMed
177Engeli, S, Schling, P, Gorzelniak, K, et al. (2003) The adipose-tissue renin–angiotensin–aldosterone system: role in the metabolic syndrome? Int J Biochem Cell Biol 35, 807825.CrossRefGoogle ScholarPubMed
178Massiera, F, Seydoux, J, Geloen, A, et al. (2001) Angiotensinogen-deficient mice exhibit impairment of diet-induced weight gain with alteration in adipose tissue development and increased locomotor activity. Endocrinology 142, 52205225.CrossRefGoogle ScholarPubMed
179Kouyama, R, Suganami, T, Nishida, J, et al. (2005) Attenuation of diet-induced weight gain and adiposity through increased energy expenditure in mice lacking angiotensin II type 1a receptor. Endocrinology 146, 34813489.CrossRefGoogle ScholarPubMed
180Yvan-Charvet, L, Even, P, Bloch-Faure, M, et al. (2005) Deletion of the angiotensin type 2 receptor (AT2R) reduces adipose cell size and protects from diet-induced obesity and insulin resistance. Diabetes 54, 991999.CrossRefGoogle ScholarPubMed
181Jayasooriya, AP, Mathai, ML, Walker, LL, et al. (2008) Mice lacking angiotensin-converting enzyme have increased energy expenditure, with reduced fat mass and improved glucose clearance. Proc Natl Acad Sci U S A 105, 65316536.CrossRefGoogle ScholarPubMed
182Brink, M, Wellen, J & Delafontaine, P (1996) Angiotensin II causes weight loss and decreases circulating insulin-like growth factor I in rats through a pressor-independent mechanism. J Clin Invest 97, 25092516.CrossRefGoogle ScholarPubMed
183Cassis, L, Helton, M, English, V, et al. (2002) Angiotensin II regulates oxygen consumption. Am J Physiol Regul Integr Comp Physiol 282, R445R453.CrossRefGoogle ScholarPubMed
184de Kloet, AD, Krause, EG, Kim, DH, et al. (2009) The effect of angiotensin-converting enzyme inhibition using captopril on energy balance and glucose homeostasis. Endocrinology 150, 41144123.CrossRefGoogle ScholarPubMed
185Porter, JP & Potratz, KR (2004) Effect of intracerebroventricular angiotensin II on body weight and food intake in adult rats. Am J Physiol Regul Integr Comp Physiol 287, R422R428.CrossRefGoogle ScholarPubMed
186de Kloet, AD, Krause, EG, Scott, KA, et al. (2011) Central angiotensin II has catabolic action at white and brown adipose tissue. Am J Physiol Endocrinol Metab 301, E1081E1091.CrossRefGoogle ScholarPubMed
187de Kloet, AD, Krause, EG & Woods, SC (2010) The renin angiotensin system and the metabolic syndrome. Physiol Behav 100, 525534.CrossRefGoogle ScholarPubMed
188Tchernof, A & Despres, JP (2000) Sex steroid hormones, sex hormone-binding globulin, and obesity in men and women. Horm Metab Res 32, 526536.CrossRefGoogle ScholarPubMed
189Mayes, JS & Watson, GH (2004) Direct effects of sex steroid hormones on adipose tissues and obesity. Obes Rev 5, 197216.CrossRefGoogle ScholarPubMed
190Brown, LM, Gent, L, Davis, K, et al. (2010) Metabolic impact of sex hormones on obesity. Brain Res 1350, 7785.CrossRefGoogle ScholarPubMed
191Mauvais-Jarvis, F (2011) Estrogen and androgen receptors: regulators of fuel homeostasis and emerging targets for diabetes and obesity. Trends Endocrinol Metab 22, 2433.CrossRefGoogle ScholarPubMed
192Mattiasson, I, Rendell, M, Tornquist, C, et al. (2002) Effects of estrogen replacement therapy on abdominal fat compartments as related to glucose and lipid metabolism in early postmenopausal women. Horm Metab Res 34, 583588.CrossRefGoogle ScholarPubMed
193Tsai, EC, Boyko, EJ, Leonetti, DL, et al. (2000) Low serum testosterone level as a predictor of increased visceral fat in Japanese-American men. Int J Obes Relat Metab Disord 24, 485491.CrossRefGoogle ScholarPubMed
194Kupelian, V, Page, ST, Araujo, AB, et al. (2006) Low sex hormone-binding globulin, total testosterone, and symptomatic androgen deficiency are associated with development of the metabolic syndrome in nonobese men. J Clin Endocrinol Metab 91, 843850.CrossRefGoogle ScholarPubMed
195Marin, P, Holmang, S, Jonsson, L, et al. (1992) The effects of testosterone treatment on body composition and metabolism in middle-aged obese men. Int J Obes Relat Metab Disord 16, 991997.Google ScholarPubMed
196Eckel, LA (2011) The ovarian hormone estradiol plays a crucial role in the control of food intake in females. Physiol Behav 104, 517524.CrossRefGoogle Scholar
197Heine, PA, Taylor, JA, Iwamoto, GA, et al. (2000) Increased adipose tissue in male and female estrogen receptor-α knockout mice. Proc Natl Acad Sci U S A 97, 1272912734.CrossRefGoogle ScholarPubMed
198Musatov, S, Chen, W, Pfaff, DW, et al. (2007) Silencing of estrogen receptor α in the ventromedial nucleus of hypothalamus leads to metabolic syndrome. Proc Natl Acad Sci U S A 104, 25012506.CrossRefGoogle ScholarPubMed
199Ropero, AB, Alonso-Magdalena, P, Quesada, I, et al. (2008) The role of estrogen receptors in the control of energy and glucose homeostasis. Steroids 73, 874879.CrossRefGoogle ScholarPubMed
200Fan, W, Yanase, T, Nomura, M, et al. (2005) Androgen receptor null male mice develop late-onset obesity caused by decreased energy expenditure and lipolytic activity but show normal insulin sensitivity with high adiponectin secretion. Diabetes 54, 10001008.CrossRefGoogle ScholarPubMed
201Xu, Y, Nedungadi, TP, Zhu, L, et al. (2011) Distinct hypothalamic neurons mediate estrogenic effects on energy homeostasis and reproduction. Cell Metab 14, 453465.CrossRefGoogle ScholarPubMed
202Plum, L, Belgardt, BF & Brüning, JC (2006) Central insulin action in energy and glucose homeostasis. J Clin Invest 116, 17611766.CrossRefGoogle ScholarPubMed
203Woods, SC, Lotter, EC, McKay, LD, et al. (1979) Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature 282, 503505.CrossRefGoogle ScholarPubMed
204Brüning, JC, Gautam, D, Burks, DJ, et al. (2000) Role of brain insulin receptor in control of body weight and reproduction. Science 289, 21222125.CrossRefGoogle ScholarPubMed
205Schwartz, MW, Woods, SC, Porte, D Jr, et al. (2000) Central nervous system control of food intake. Nature 404, 661671.CrossRefGoogle ScholarPubMed
206Könner, AC, Klöckener, T & Brüning, JC (2009) Control of energy homeostasis by insulin and leptin: targeting the arcuate nucleus and beyond. Physiol Behav 97, 632638.CrossRefGoogle ScholarPubMed
207Rothwell, NJ & Stock, MJ (1988) Insulin and thermogenesis. Int J Obes 12, 98103.Google ScholarPubMed
208Menéndez, JA & Atrens, DM (1989) Insulin increases energy expenditure and respiratory quotient in the rat. Pharmacol Biochem Behav 34, 765768.CrossRefGoogle ScholarPubMed
209Dulloo, AG & Girardier, L (1989) Energy expenditure and diet-induced thermogenesis in presence and absence of hyperphagia induced by insulin. Am J Physiol 257, R717R725.Google ScholarPubMed
210Jiang, G & Zhang, BB (2003) Glucagon and regulation of glucose metabolism. Am J Physiol Endocrinol Metab 284, E671E678.CrossRefGoogle ScholarPubMed
211Gómez-Ambrosi, J, Catalan, V & Frühbeck, G (2009) The adipo–hepato–insular axis in glucose homeostasis. In Peptides in Energy Balance and Obesity, pp. 163193 [, editor]. Wallingford: CABI Publishing.CrossRefGoogle Scholar
212Kalkhoff, RK, Gossain, VV & Matute, ML (1973) Plasma glucagon in obesity. Response to arginine, glucose and protein administration. N Engl J Med 289, 465467.CrossRefGoogle ScholarPubMed
213Starke, AA, Erhardt, G, Berger, M, et al. (1984) Elevated pancreatic glucagon in obesity. Diabetes 33, 277280.CrossRefGoogle ScholarPubMed
214Koeslag, JH, Saunders, PT & Terblanche, E (2003) A reappraisal of the blood glucose homeostat which comprehensively explains the type 2 diabetes mellitus–syndrome X complex. J Physiol 549, 333346.CrossRefGoogle ScholarPubMed
215Penick, SB & Hinkle, LE Jr (1961) Depression of food intake induced in healthy subjects by glucagon. N Engl J Med 264, 893897.CrossRefGoogle ScholarPubMed
216Geary, N, Langhans, W & Scharrer, E (1981) Metabolic concomitants of glucagon-induced suppression of feeding in the rat. Am J Physiol 241, R330R335.Google ScholarPubMed
217Habegger, KM, Heppner, KM, Geary, N, et al. (2010) The metabolic actions of glucagon revisited. Nat Rev Endocrinol 6, 689697.CrossRefGoogle ScholarPubMed
218Schulman, JL, Carleton, JL, Whitney, G, et al. (1957) Effect of glucagon on food intake and body weight in man. J Appl Physiol 11, 419421.CrossRefGoogle ScholarPubMed
219Billington, CJ, Briggs, JE, Link, JG, et al. (1991) Glucagon in physiological concentrations stimulates brown fat thermogenesis in vivo. Am J Physiol 261, R501R507.Google ScholarPubMed
220Davidson, IW, Salter, JM & Best, CH (1957) Calorigenic action of glucagon. Nature 180, 1124.CrossRefGoogle ScholarPubMed
221Gelling, RW, Du, XQ, Dichmann, DS, et al. (2003) Lower blood glucose, hyperglucagonemia, and pancreatic α cell hyperplasia in glucagon receptor knockout mice. Proc Natl Acad Sci U S A 100, 14381443.CrossRefGoogle ScholarPubMed
222Lou, PH, Gustavsson, N, Wang, Y, et al. (2011) Increased lipolysis and energy expenditure in a mouse model with severely impaired glucagon secretion. PLoS ONE 6, e26671.CrossRefGoogle Scholar
223Vuguin, PM & Charron, MJ (2011) Novel insight into glucagon receptor action: lessons from knockout and transgenic mouse models. Diabetes Obes Metab 13, Suppl. 1, 144150.CrossRefGoogle ScholarPubMed
224Schwartz, TW, Holst, JJ, Fahrenkrug, J, et al. (1978) Vagal, cholinergic regulation of pancreatic polypeptide secretion. J Clin Invest 61, 781789.CrossRefGoogle ScholarPubMed
225Jorde, R & Burhol, PG (1984) Fasting and postprandial plasma pancreatic polypeptide (PP) levels in obesity. Int J Obes 8, 393397.Google ScholarPubMed
226Pieramico, O, Malfertheiner, P, Nelson, DK, et al. (1990) Interdigestive cycling and post-prandial release of pancreatic polypeptide in severe obesity. Int J Obes 14, 10051011.Google ScholarPubMed
227Lassmann, V, Vague, P, Vialettes, B, et al. (1980) Low plasma levels of pancreatic polypeptide in obesity. Diabetes 29, 428430.CrossRefGoogle ScholarPubMed
228Holst, JJ, Schwartz, TW, Lovgreen, NA, et al. (1983) Diurnal profile of pancreatic polypeptide, pancreatic glucagon, gut glucagon and insulin in human morbid obesity. Int J Obes 7, 529538.Google ScholarPubMed
229Asakawa, A, Inui, A, Yuzuriha, H, et al. (2003) Characterization of the effects of pancreatic polypeptide in the regulation of energy balance. Gastroenterology 124, 13251336.CrossRefGoogle ScholarPubMed
230Ueno, N, Inui, A, Iwamoto, M, et al. (1999) Decreased food intake and body weight in pancreatic polypeptide-overexpressing mice. Gastroenterology 117, 14271432.CrossRefGoogle ScholarPubMed
231Batterham, RL, Le Roux, CW, Cohen, MA, et al. (2003) Pancreatic polypeptide reduces appetite and food intake in humans. J Clin Endocrinol Metab 88, 39893992.CrossRefGoogle ScholarPubMed
232Schmidt, PT, Näslund, E, Grybäck, P, et al. (2005) A role for pancreatic polypeptide in the regulation of gastric emptying and short-term metabolic control. J Clin Endocrinol Metab 90, 52415246.CrossRefGoogle ScholarPubMed
233Cooper, GJ, Willis, AC, Clark, A, et al. (1987) Purification and characterization of a peptide from amyloid-rich pancreases of type 2 diabetic patients. Proc Natl Acad Sci U S A 84, 86288632.CrossRefGoogle ScholarPubMed
234Cummings, DE & Overduin, J (2007) Gastrointestinal regulation of food intake. J Clin Invest 117, 1323.CrossRefGoogle ScholarPubMed
235Lutz, TA (2010) The role of amylin in the control of energy homeostasis. Am J Physiol Regul Integr Comp Physiol 298, R1475R1484.CrossRefGoogle ScholarPubMed
236Boyle, CN & Lutz, TA (2011) Amylinergic control of food intake in lean and obese rodents. Physiol Behav 105, 129137.CrossRefGoogle ScholarPubMed
237Trevaskis, JL, Parkes, DG & Roth, JD (2010) Insights into amylin–leptin synergy. Trends Endocrinol Metab 21, 473479.CrossRefGoogle ScholarPubMed
238Aronne, L, Fujioka, K, Aroda, V, et al. (2007) Progressive reduction in body weight after treatment with the amylin analog pramlintide in obese subjects: a phase 2, randomized, placebo-controlled, dose-escalation study. J Clin Endocrinol Metab 92, 29772983.CrossRefGoogle ScholarPubMed
239Roth, JD, Roland, BL, Cole, RL, et al. (2008) Leptin responsiveness restored by amylin agonism in diet-induced obesity: evidence from nonclinical and clinical studies. Proc Natl Acad Sci U S A 105, 72577562.CrossRefGoogle ScholarPubMed
240Roth, JD, Hughes, H, Kendall, E, et al. (2006) Antiobesity effects of the β-cell hormone amylin in diet-induced obese rats: effects on food intake, body weight, composition, energy expenditure, and gene expression. Endocrinology 147, 58555864.CrossRefGoogle ScholarPubMed
241Mack, C, Wilson, J, Athanacio, J, et al. (2007) Pharmacological actions of the peptide hormone amylin in the long-term regulation of food intake, food preference, and body weight. Am J Physiol Regul Integr Comp Physiol 293, R1855R1863.CrossRefGoogle ScholarPubMed
242Osaka, T, Tsukamoto, A, Koyama, Y, et al. (2008) Central and peripheral administration of amylin induces energy expenditure in anesthetized rats. Peptides 29, 10281035.CrossRefGoogle ScholarPubMed
243Rushing, PA, Hagan, MM, Seeley, RJ, et al. (2000) Amylin: a novel action in the brain to reduce body weight. Endocrinology 141, 850853.CrossRefGoogle ScholarPubMed
244Murphy, KG, Dhillo, WS & Bloom, SR (2006) Gut peptides in the regulation of food intake and energy homeostasis. Endocr Rev 27, 719727.CrossRefGoogle ScholarPubMed
245Badman, MK & Flier, JS (2005) The gut and energy balance: visceral allies in the obesity wars. Science 307, 19091914.CrossRefGoogle ScholarPubMed
246Kojima, M & Kangawa, K (2005) Ghrelin: structure and function. Physiol Rev 85, 495522.CrossRefGoogle ScholarPubMed
247Murphy, KG & Bloom, SR (2006) Gut hormones and the regulation of energy homeostasis. Nature 444, 854859.CrossRefGoogle ScholarPubMed
248Wren, AM & Bloom, SR (2007) Gut hormones and appetite control. Gastroenterology 132, 21162130.CrossRefGoogle ScholarPubMed
249Kojima, M, Hosoda, H, Date, Y, et al. (1999) Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402, 656660.CrossRefGoogle ScholarPubMed
250Nakazato, M, Murakami, N, Date, Y, et al. (2001) A role for ghrelin in the central regulation of feeding. Nature 409, 194198.CrossRefGoogle ScholarPubMed
251Tschöp, M, Smiley, DL & Heiman, ML (2000) Ghrelin induces adiposity in rodents. Nature 407, 908913.CrossRefGoogle ScholarPubMed
252Asakawa, A, Inui, A, Kaga, T, et al. (2001) Ghrelin is an appetite-stimulatory signal from stomach with structural resemblance to motilin. Gastroenterology 120, 337345.CrossRefGoogle ScholarPubMed
253Wren, AM, Seal, LJ, Cohen, MA, et al. (2001) Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab 86, 5992.CrossRefGoogle ScholarPubMed
254Rodríguez, A, Gómez-Ambrosi, J, Catalán, V, et al. (2009) Acylated and desacyl ghrelin stimulate lipid accumulation in human visceral adipocytes. Int J Obes 33, 541552.CrossRefGoogle ScholarPubMed
255Cummings, DE, Purnell, JQ, Frayo, RS, et al. (2001) A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 50, 17141749.CrossRefGoogle ScholarPubMed
256Tschöp, M, Weyer, C, Tataranni, PA, et al. (2001) Circulating ghrelin levels are decreased in human obesity. Diabetes 50, 707709.CrossRefGoogle ScholarPubMed
257Cummings, DE, Weigle, DS, Frayo, RS, et al. (2002) Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med 346, 16231630.CrossRefGoogle ScholarPubMed
258Frühbeck, G, Diez Caballero, A & Gil, MJ (2004) Fundus functionality and ghrelin concentrations after bariatric surgery. N Engl J Med 350, 308309.CrossRefGoogle ScholarPubMed
259Frühbeck, G, Diez-Caballero, A, Gil, MJ, et al. (2004) The decrease in plasma ghrelin concentrations following bariatric surgery depends on the functional integrity of the fundus. Obes Surg 14, 606612.CrossRefGoogle ScholarPubMed
260Holdstock, C, Engström, BE, Öhrvall, M, et al. (2003) Ghrelin and adipose tissue regulatory peptides: effect of gastric bypass surgery in obese humans. J Clin Endocrinol Metab 88, 31773183.CrossRefGoogle ScholarPubMed
261Wren, AM, Small, CJ, Abbott, CR, et al. (2001) Ghrelin causes hyperphagia and obesity in rats. Diabetes 50, 25402547.CrossRefGoogle ScholarPubMed
262Chen, HY, Trumbauer, ME, Chen, AS, et al. (2004) Orexigenic action of peripheral ghrelin is mediated by neuropeptide Y and Agouti-related protein. Endocrinology 145, 26072612.CrossRefGoogle ScholarPubMed
263López, M, Lage, R, Saha, AK, et al. (2008) Hypothalamic fatty acid metabolism mediates the orexigenic action of ghrelin. Cell Metab 7, 389399.CrossRefGoogle ScholarPubMed
264Wortley, KE, Del Rincon, JP, Murray, JD, et al. (2005) Absence of ghrelin protects against early-onset obesity. J Clin Invest 115, 35733578.CrossRefGoogle ScholarPubMed
265Zigman, JM, Nakano, Y, Coppari, R, et al. (2005) Mice lacking ghrelin receptors resist the development of diet-induced obesity. J Clin Invest 115, 35643572.CrossRefGoogle ScholarPubMed
266Pfluger, PT, Kirchner, H, Günnel, S, et al. (2008) Simultaneous deletion of ghrelin and its receptor increases motor activity and energy expenditure. Am J Physiol Gastrointest Liver Physiol 294, G610G618.CrossRefGoogle ScholarPubMed
267Nagaya, N, Moriya, J, Yasumura, Y, et al. (2004) Effects of ghrelin administration on left ventricular function, exercise capacity, and muscle wasting in patients with chronic heart failure. Circulation 110, 36743679.CrossRefGoogle ScholarPubMed
268Nagaya, N, Itoh, T, Murakami, S, et al. (2005) Treatment of cachexia with ghrelin in patients with COPD. Chest 128, 11871193.CrossRefGoogle ScholarPubMed
269Drucker, DJ (2006) The biology of incretin hormones. Cell Metab 3, 153165.CrossRefGoogle ScholarPubMed
270Brown, JC (1971) A gastric inhibitory polypeptide. I. The amino acid composition and the tryptic peptides. Can J Biochem 49, 255261.CrossRefGoogle ScholarPubMed
271Song, DH & Wolfe, MM (2007) Glucose-dependent insulinotropic polypeptide and its role in obesity. Curr Opin Endocrinol Diabetes Obes 14, 4651.CrossRefGoogle ScholarPubMed
272Roust, LR, Stesin, M, Go, VL, et al. (1988) Role of gastric inhibitory polypeptide in postprandial hyperinsulinemia of obesity. Am J Physiol 254, E767E774.Google ScholarPubMed
273Drucker, DJ (2007) The role of gut hormones in glucose homeostasis. J Clin Invest 117, 2432.CrossRefGoogle ScholarPubMed
274Wideman, RD & Kieffer, TJ (2009) Mining incretin hormone pathways for novel therapies. Trends Endocrinol Metab 20, 280286.CrossRefGoogle ScholarPubMed
275Fehmann, HC, Goke, R & Goke, B (1995) Cell and molecular biology of the incretin hormones glucagon-like peptide-I and glucose-dependent insulin releasing polypeptide. Endocr Rev 16, 390410.CrossRefGoogle ScholarPubMed
276Wasada, T, McCorkle, K, Harris, V, et al. (1981) Effect of gastric inhibitory polypeptide on plasma levels of chylomicron triglycerides in dogs. J Clin Invest 68, 11061107.CrossRefGoogle ScholarPubMed
277Song, DH, Getty-Kaushik, L, Tseng, E, et al. (2007) Glucose-dependent insulinotropic polypeptide enhances adipocyte development and glucose uptake in part through Akt activation. Gastroenterology 133, 17961805.CrossRefGoogle ScholarPubMed
278Althage, MC, Ford, EL, Wang, S, et al. (2008) Targeted ablation of glucose-dependent insulinotropic polypeptide-producing cells in transgenic mice reduces obesity and insulin resistance induced by a high fat diet. J Biol Chem 283, 1836518376.CrossRefGoogle ScholarPubMed
279Miyawaki, K, Yamada, Y, Ban, N, et al. (2002) Inhibition of gastric inhibitory polypeptide signaling prevents obesity. Nat Med 8, 738742.CrossRefGoogle ScholarPubMed
280Gault, VA, Irwin, N, Green, BD, et al. (2005) Chemical ablation of gastric inhibitory polypeptide receptor action by daily (Pro3)GIP administration improves glucose tolerance and ameliorates insulin resistance and abnormalities of islet structure in obesity-related diabetes. Diabetes 54, 24362446.CrossRefGoogle ScholarPubMed
281Hansotia, T, Maida, A, Flock, G, et al. (2007) Extrapancreatic incretin receptors modulate glucose homeostasis, body weight, and energy expenditure. J Clin Invest 117, 143152.CrossRefGoogle ScholarPubMed
282Daousi, C, Wilding, JP, Aditya, S, et al. (2009) Effects of peripheral administration of synthetic human glucose-dependent insulinotropic peptide (GIP) on energy expenditure and subjective appetite sensations in healthy normal weight subjects and obese patients with type 2 diabetes. Clin Endocrinol 71, 195201.CrossRefGoogle ScholarPubMed
283Irwin, N & Flatt, PR (2009) Evidence for beneficial effects of compromised gastric inhibitory polypeptide action in obesity-related diabetes and possible therapeutic implications. Diabetologia 52, 17241731.CrossRefGoogle ScholarPubMed
284Baggio, LL & Drucker, DJ (2007) Biology of incretins: GLP-1 and GIP. Gastroenterology 132, 21312157.CrossRefGoogle ScholarPubMed
285Kieffer, TJ & Habener, JF (1999) The glucagon-like peptides. Endocr Rev 20, 876913.CrossRefGoogle ScholarPubMed
286Drucker, DJ (2005) Biologic actions and therapeutic potential of the proglucagon-derived peptides. Nat Clin Pract Endocr Metab 1, 2231.CrossRefGoogle ScholarPubMed
287Nauck, MA, Vardarli, I, Deacon, CF, et al. (2011) Secretion of glucagon-like peptide-1 (GLP-1) in type 2 diabetes: what is up, what is down? Diabetologia 54, 1018.CrossRefGoogle ScholarPubMed
288Ranganath, LR, Beety, JM, Morgan, LM, et al. (1996) Attenuated GLP-1 secretion in obesity: cause or consequence? Gut 38, 916919.CrossRefGoogle ScholarPubMed
289Naslund, E, Gryback, P, Backman, L, et al. (1998) Distal small bowel hormones: correlation with fasting antroduodenal motility and gastric emptying. Dig Dis Sci 43, 945952.CrossRefGoogle ScholarPubMed
290Torekov, SS, Madsbad, S & Holst, JJ (2011) Obesity – an indication for GLP-1 treatment? Obesity pathophysiology and GLP-1 treatment potential. Obes Rev 12, 593601.CrossRefGoogle ScholarPubMed
291Verdich, C, Toubro, S, Buemann, B, et al. (2001) The role of postprandial releases of insulin and incretin hormones in meal-induced satiety – effect of obesity and weight reduction. Int J Obes Relat Metab Disord 25, 12061214.CrossRefGoogle ScholarPubMed
292Morínigo, R, Moize, V, Musri, M, et al. (2006) Glucagon-like peptide-1, peptide YY, hunger, and satiety after gastric bypass surgery in morbidly obese subjects. J Clin Endocrinol Metab 91, 17351740.CrossRefGoogle ScholarPubMed
293Turton, MD, O'Shea, D, Gunn, I, et al. (1996) A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379, 6972.CrossRefGoogle ScholarPubMed
294Flint, A, Raben, A, Astrup, A, et al. (1998) Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J Clin Invest 101, 515520.CrossRefGoogle ScholarPubMed
295Gutzwiller, JP, Goke, B, Drewe, J, et al. (1999) Glucagon-like peptide-1: a potent regulator of food intake in humans. Gut 44, 8186.CrossRefGoogle ScholarPubMed
296Naslund, E, Barkeling, B, King, N, et al. (1999) Energy intake and appetite are suppressed by glucagon-like peptide-1 (GLP-1) in obese men. Int J Obes Relat Metab Disord 23, 304311.CrossRefGoogle ScholarPubMed
297Flint, A, Raben, A, Ersboll, AK, et al. (2001) The effect of physiological levels of glucagon-like peptide-1 on appetite, gastric emptying, energy and substrate metabolism in obesity. Int J Obes Relat Metab Disord 25, 781792.CrossRefGoogle ScholarPubMed
298Williams, DL, Baskin, DG & Schwartz, MW (2009) Evidence that intestinal glucagon-like peptide-1 plays a physiological role in satiety. Endocrinology 150, 16801687.CrossRefGoogle Scholar
299Osaka, T, Endo, M, Yamakawa, M, et al. (2005) Energy expenditure by intravenous administration of glucagon-like peptide-1 mediated by the lower brainstem and sympathoadrenal system. Peptides 26, 16231631.CrossRefGoogle ScholarPubMed
300Pannacciulli, N, Bunt, JC, Koska, J, et al. (2006) Higher fasting plasma concentrations of glucagon-like peptide 1 are associated with higher resting energy expenditure and fat oxidation rates in humans. Am J Clin Nutr 84, 556560.CrossRefGoogle ScholarPubMed
301Knauf, C, Cani, PD, Ait-Belgnaoui, A, et al. (2008) Brain glucagon-like peptide 1 signaling controls the onset of high-fat diet-induced insulin resistance and reduces energy expenditure. Endocrinology 149, 47684777.CrossRefGoogle ScholarPubMed
302Ayala, JE, Bracy, DP, James, FD, et al. (2010) Glucagon-like peptide-1 receptor knockout mice are protected from high-fat diet-induced insulin resistance. Endocrinology 151, 46784687.CrossRefGoogle ScholarPubMed
303Nogueiras, R, Perez-Tilve, D, Veyrat-Durebex, C, et al. (2009) Direct control of peripheral lipid deposition by CNS GLP-1 receptor signaling is mediated by the sympathetic nervous system and blunted in diet-induced obesity. J Neurosci 29, 59165925.CrossRefGoogle ScholarPubMed
304Barrera, JG, Jones, KR, Herman, JP, et al. (2011) Hyperphagia and increased fat accumulation in two models of chronic CNS glucagon-like peptide-1 loss of function. J Neurosci 31, 39043913.CrossRefGoogle ScholarPubMed
305Hayes, MR, De Jonghe, BC & Kanoski, SE (2010) Role of the glucagon-like-peptide-1 receptor in the control of energy balance. Physiol Behav 100, 503510.CrossRefGoogle ScholarPubMed
306Vilsboll, T, Zdravkovic, M, Le-Thi, T, et al. (2007) Liraglutide, a long-acting human glucagon-like peptide-1 analog, given as monotherapy significantly improves glycemic control and lowers body weight without risk of hypoglycemia in patients with type 2 diabetes. Diabetes Care 30, 16081610.CrossRefGoogle ScholarPubMed
307Astrup, A, Rossner, S, Van Gaal, L, et al. (2009) Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet 374, 16061616.CrossRefGoogle ScholarPubMed
308Day, JW, Ottaway, N, Patterson, JT, et al. (2009) A new glucagon and GLP-1 co-agonist eliminates obesity in rodents. Nat Chem Biol 5, 749757.CrossRefGoogle ScholarPubMed
309Tang Christensen, M, Larsen, PJ, Thulesen, J, et al. (2000) The proglucagon-derived peptide, glucagon-like peptide-2, is a neurotransmitter involved in the regulation of food intake. Nat Med 6, 802807.CrossRefGoogle ScholarPubMed
310Bahrami, J, Longuet, C, Baggio, LL, et al. (2010) Glucagon-like peptide-2 receptor modulates islet adaptation to metabolic stress in the ob/ob mouse. Gastroenterology 139, 857868.CrossRefGoogle ScholarPubMed
311le Roux, CW, Borg, C, Wallis, K, et al. (2010) Gut hypertrophy after gastric bypass is associated with increased glucagon-like peptide 2 and intestinal crypt cell proliferation. Ann Surg 252, 5056.CrossRefGoogle ScholarPubMed
312Drucker, DJ (2007) Dipeptidyl peptidase-4 inhibition and the treatment of type 2 diabetes: preclinical biology and mechanisms of action. Diabetes Care 30, 13351343.CrossRefGoogle ScholarPubMed
313Conarello, SL, Li, Z, Ronan, J, et al. (2003) Mice lacking dipeptidyl peptidase IV are protected against obesity and insulin resistance. Proc Natl Acad Sci U S A 100, 68256830.CrossRefGoogle ScholarPubMed
314Tatemoto, K & Mutt, V (1980) Isolation of two novel candidate hormones using a chemical method for finding naturally occurring polypeptides. Nature 285, 417418.CrossRefGoogle ScholarPubMed
315Karra, E & Batterham, RL (2010) The role of gut hormones in the regulation of body weight and energy homeostasis. Mol Cell Endocrinol 316, 120128.CrossRefGoogle ScholarPubMed
316Grandt, D, Schimiczek, M, Beglinger, C, et al. (1994) Two molecular forms of peptide YY (PYY) are abundant in human blood: characterization of a radioimmunoassay recognizing PYY 1–36 and PYY 3–36. Regul Pept 51, 151159.CrossRefGoogle ScholarPubMed
317Kirchner, H, Tong, J, Tschöp, MH, et al. (2010) Ghrelin and PYY in the regulation of energy balance and metabolism: lessons from mouse mutants. Am J Physiol Endocrinol Metab 298, E909E919.CrossRefGoogle ScholarPubMed
318Zac-Varghese, S, De Silva, A & Bloom, SR (2011) Translational studies on PYY as a novel target in obesity. Curr Opin Pharmacol 11, 582585.CrossRefGoogle ScholarPubMed
319Batterham, RL, Cohen, MA, Ellis, SM, et al. (2003) Inhibition of food intake in obese subjects by peptide YY3–36. N Engl J Med 349, 941948.CrossRefGoogle ScholarPubMed
320Batterham, RL, Cowley, MA, Small, CJ, et al. (2002) Gut hormone PYY3–36 physiologically inhibits food intake. Nature 418, 650654.CrossRefGoogle Scholar
321Moran, TH, Smedh, U, Kinzig, KP, et al. (2005) Peptide YY(3–36) inhibits gastric emptying and produces acute reductions in food intake in rhesus monkeys. Am J Physiol Regul Integr Comp Physiol 288, R384R388.CrossRefGoogle ScholarPubMed
322Batterham, RL, Ffytche, DH, Rosenthal, JM, et al. (2007) PYY modulation of cortical and hypothalamic brain areas predicts feeding behaviour in humans. Nature 450, 106109.CrossRefGoogle ScholarPubMed
323Tschöp, M, Castañeda, TR, Joost, HG, et al. (2004) Physiology: does gut hormone PYY3–36 decrease food intake in rodents? Nature 430, 1 p following 165; discussion 2 p following 165.CrossRefGoogle ScholarPubMed
324Schonhoff, S, Baggio, L, Ratineau, C, et al. (2005) Energy homeostasis and gastrointestinal endocrine differentiation do not require the anorectic hormone peptide YY. Mol Cell Biol 25, 41894199.CrossRefGoogle Scholar
325Wortley, KE, Garcia, K, Okamoto, H, et al. (2007) Peptide YY regulates bone turnover in rodents. Gastroenterology 133, 15341543.CrossRefGoogle ScholarPubMed
326Boey, D, Lin, S, Karl, T, et al. (2006) Peptide YY ablation in mice leads to the development of hyperinsulinaemia and obesity. Diabetologia 49, 13601370.CrossRefGoogle Scholar
327Batterham, RL, Heffron, H, Kapoor, S, et al. (2006) Critical role for peptide YY in protein-mediated satiation and body-weight regulation. Cell Metab 4, 223233.CrossRefGoogle ScholarPubMed
328Sloth, B, Holst, JJ, Flint, A, et al. (2007) Effects of PYY1–36 and PYY3–36 on appetite, energy intake, energy expenditure, glucose and fat metabolism in obese and lean subjects. Am J Physiol Endocrinol Metab 292, E1062E1068.CrossRefGoogle ScholarPubMed
329Doucet, E, Laviolette, M, Imbeault, P, et al. (2008) Total peptide YY is a correlate of postprandial energy expenditure but not of appetite or energy intake in healthy women. Metabolism 57, 14581464.CrossRefGoogle ScholarPubMed
330Guo, Y, Ma, L, Enriori, PJ, et al. (2006) Physiological evidence for the involvement of peptide YY in the regulation of energy homeostasis in humans. Obesity 14, 15621570.CrossRefGoogle ScholarPubMed
331Raybould, HE (2007) Mechanisms of CCK signaling from gut to brain. Curr Opin Pharmacol 7, 570574.CrossRefGoogle ScholarPubMed
332Liddle, RA, Goldfine, ID, Rosen, MS, et al. (1985) Cholecystokinin bioactivity in human plasma. Molecular forms, responses to feeding, and relationship to gallbladder contraction. J Clin Invest 75, 11441152.CrossRefGoogle ScholarPubMed
333Lo, CM, Samuelson, LC, Chambers, JB, et al. (2008) Characterization of mice lacking the gene for cholecystokinin. Am J Physiol Regul Integr Comp Physiol 294, R803R810.CrossRefGoogle ScholarPubMed
334Crawley, JN & Corwin, RL (1994) Biological actions of cholecystokinin. Peptides 15, 731755.CrossRefGoogle ScholarPubMed
335Kopin, AS, Mathes, WF, McBride, EW, et al. (1999) The cholecystokinin-A receptor mediates inhibition of food intake yet is not essential for the maintenance of body weight. J Clin Invest 103, 383391.CrossRefGoogle Scholar
336Lo, CM, King, A, Samuelson, LC, et al. (2010) Cholecystokinin knockout mice are resistant to high-fat diet-induced obesity. Gastroenterology 138, 19972005.CrossRefGoogle ScholarPubMed
337Gibbs, J, Young, RC & Smith, GP (1973) Cholycystokinin elicits satiety in rats with open gastric fistulas. Nature 245, 323325.CrossRefGoogle Scholar
338Smith, GP, Jerome, C, Cushin, BJ, et al. (1981) Abdominal vagotomy blocks the satiety effect of cholecystokinin in the rat. Science 213, 10361037.CrossRefGoogle ScholarPubMed
339Lieverse, RJ, Jansen, JB, Masclee, AA, et al. (1995) Satiety effects of a physiological dose of cholecystokinin in humans. Gut 36, 176179.CrossRefGoogle ScholarPubMed
340Crawley, JN & Beinfeld, MC (1983) Rapid development of tolerance to the behavioural actions of cholecystokinin. Nature 302, 703706.CrossRefGoogle Scholar
341Jordan, J, Greenway, FL, Leiter, LA, et al. (2008) Stimulation of cholecystokinin-A receptors with GI181771X does not cause weight loss in overweight or obese patients. Clin Pharmacol Ther 83, 281287.CrossRefGoogle ScholarPubMed
342Moran, TH, Katz, LF, Plata-Salaman, CR, et al. (1998) Disordered food intake and obesity in rats lacking cholecystokinin A receptors. Am J Physiol 274, R618R625.Google ScholarPubMed
343Bataille, D, Tatemoto, K, Gespach, C, et al. (1982) Isolation of glucagon-37 (bioactive enteroglucagon/oxyntomodulin) from porcine jejuno-ileum. Characterization of the peptide. FEBS Lett 146, 7986.CrossRefGoogle ScholarPubMed
344Le Quellec, A, Kervran, A, Blache, P, et al. (1992) Oxyntomodulin-like immunoreactivity: diurnal profile of a new potential enterogastrone. J Clin Endocrinol Metab 74, 14051409.Google ScholarPubMed
345Dubrasquet, M, Bataille, D & Gespach, C (1982) Oxyntomodulin (glucagon-37 or bioactive enteroglucagon): a potent inhibitor of pentagastrin-stimulated acid secretion in rats. Biosci Rep 2, 391395.CrossRefGoogle ScholarPubMed
346Schjoldager, B, Mortensen, PE, Myhre, J, et al. (1989) Oxyntomodulin from distal gut. Role in regulation of gastric and pancreatic functions. Dig Dis Sci 34, 14111419.CrossRefGoogle ScholarPubMed
347Schepp, W, Dehne, K, Riedel, T, et al. (1996) Oxyntomodulin: a cAMP-dependent stimulus of rat parietal cell function via the receptor for glucagon-like peptide-1 (7–36)NH2. Digestion 57, 398405.CrossRefGoogle ScholarPubMed
348Baggio, LL, Huang, Q, Brown, TJ, et al. (2004) Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology 127, 546558.CrossRefGoogle ScholarPubMed
349Parkinson, JR, Chaudhri, OB, Kuo, YT, et al. (2009) Differential patterns of neuronal activation in the brainstem and hypothalamus following peripheral injection of GLP-1, oxyntomodulin and lithium chloride in mice detected by manganese-enhanced magnetic resonance imaging (MEMRI). Neuroimage 44, 10221031.CrossRefGoogle ScholarPubMed
350Dakin, CL, Gunn, I, Small, CJ, et al. (2001) Oxyntomodulin inhibits food intake in the rat. Endocrinology 142, 42444250.CrossRefGoogle ScholarPubMed
351Dakin, CL, Small, CJ, Batterham, RL, et al. (2004) Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocrinology 145, 26872695.CrossRefGoogle ScholarPubMed
352Cohen, MA, Ellis, SM, Le Roux, CW, et al. (2003) Oxyntomodulin suppresses appetite and reduces food intake in humans. J Clin Endocrinol Metab 88, 46964701.CrossRefGoogle ScholarPubMed
353Wynne, K, Park, AJ, Small, CJ, et al. (2005) Subcutaneous oxyntomodulin reduces body weight in overweight and obese subjects: a double-blind, randomized, controlled trial. Diabetes 54, 23902395.CrossRefGoogle ScholarPubMed
354Dakin, CL, Small, CJ, Park, AJ, et al. (2002) Repeated ICV administration of oxyntomodulin causes a greater reduction in body weight gain than in pair-fed rats. Am J Physiol Endocrinol Metab 283, E1173E1177.CrossRefGoogle ScholarPubMed
355Wynne, K & Bloom, SR (2006) The role of oxyntomodulin and peptide tyrosine-tyrosine (PYY) in appetite control. Nat Clin Pract Endocrinol Metab 2, 612620.CrossRefGoogle ScholarPubMed
356Potter, LR (2011) Guanylyl cyclase structure, function and regulation. Cell Signal 23, 19211926.CrossRefGoogle ScholarPubMed
357Seeley, RJ & Tschöp, MH (2011) Uroguanylin: how the gut got another satiety hormone. J Clin Invest 121, 33843386.CrossRefGoogle ScholarPubMed
358Carrithers, SL, Ott, CE, Hill, MJ, et al. (2004) Guanylin and uroguanylin induce natriuresis in mice lacking guanylyl cyclase-C receptor. Kidney Int 65, 4053.CrossRefGoogle ScholarPubMed
359Frühbeck, G (2012) Gastrointestinal hormones: uroguanylin – a new gut-derived weapon against obesity? Nat Rev Endocrinol 8, 56.CrossRefGoogle Scholar
360Lafontan, M, Moro, C, Berlan, M, et al. (2008) Control of lipolysis by natriuretic peptides and cyclic GMP. Trends Endocrinol Metab 19, 130137.CrossRefGoogle ScholarPubMed
361Beenken, A & Mohammadi, M (2009) The FGF family: biology, pathophysiology and therapy. Nat Rev Drug Discov 8, 235253.CrossRefGoogle ScholarPubMed
362Kir, S, Beddow, SA, Samuel, VT, et al. (2011) FGF19 as a postprandial, insulin-independent activator of hepatic protein and glycogen synthesis. Science 331, 16211624.CrossRefGoogle ScholarPubMed
363Tomlinson, E, Fu, L, John, L, et al. (2002) Transgenic mice expressing human fibroblast growth factor-19 display increased metabolic rate and decreased adiposity. Endocrinology 143, 17411747.CrossRefGoogle ScholarPubMed
364Fu, L, John, LM, Adams, SH, et al. (2004) Fibroblast growth factor 19 increases metabolic rate and reverses dietary and leptin-deficient diabetes. Endocrinology 145, 25942603.CrossRefGoogle Scholar
365Wu, X, Ge, H, Lemon, B, et al. (2010) Separating mitogenic and metabolic activities of fibroblast growth factor 19 (FGF19). Proc Natl Acad Sci U S A 107, 1415814163.CrossRefGoogle Scholar
366Hirota, K & Fukamizu, A (2010) Transcriptional regulation of energy metabolism in the liver. J Recept Signal Transduct Res 30, 403409.CrossRefGoogle ScholarPubMed
367Juul, A (2003) Serum levels of insulin-like growth factor I and its binding proteins in health and disease. Growth Horm IGF Res 13, 113170.CrossRefGoogle ScholarPubMed
368Kawai, M & Rosen, CJ (2010) The IGF-I regulatory system and its impact on skeletal and energy homeostasis. J Cell Biochem 111, 1419.CrossRefGoogle ScholarPubMed
369Baker, J, Liu, JP, Robertson, EJ, et al. (1993) Role of insulin-like growth factors in embryonic and postnatal growth. Cell 75, 7382.CrossRefGoogle ScholarPubMed
370Sandhu, MS, Heald, AH, Gibson, JM, et al. (2002) Circulating concentrations of insulin-like growth factor-I and development of glucose intolerance: a prospective observational study. Lancet 359, 17401745.CrossRefGoogle ScholarPubMed
371Murphy, LJ (2003) The role of the insulin-like growth factors and their binding proteins in glucose homeostasis. Experimental Diab Res 4, 213224.CrossRefGoogle ScholarPubMed
372Clemmons, DR (2006) Involvement of insulin-like growth factor-I in the control of glucose homeostasis. Curr Opin Pharmacol 6, 620625.CrossRefGoogle ScholarPubMed
373Saltiel, AR & Kahn, CR (2001) Insulin signalling and the regulation of glucose and lipid metabolism. Nature 414, 799806.CrossRefGoogle ScholarPubMed
374Holzenberger, M, Dupont, J, Ducos, B, et al. (2003) IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421, 182187.CrossRefGoogle ScholarPubMed
375Sanchez-Alavez, M, Osborn, O, Tabarean, IV, et al. (2011) Insulin-like growth factor 1-mediated hyperthermia involves anterior hypothalamic insulin receptors. J Biol Chem 286, 1498314990.CrossRefGoogle ScholarPubMed
376Frystyk, J, Vestbo, E, Skjaerbaek, C, et al. (1995) Free insulin-like growth factors in human obesity. Metabolism 44, 3744.CrossRefGoogle ScholarPubMed
377Le Marchand-Brustel, Y, Heydrick, SJ, Jullien, D, et al. (1995) Effected of insulin and insulin-like growth factor-I on glucose transport and its transporters in soleus muscle of lean and obese mice. Metabolism 44, 1823.CrossRefGoogle ScholarPubMed
378Vickers, MH, Ikenasio, BA & Breier, BH (2001) IGF-I treatment reduces hyperphagia, obesity, and hypertension in metabolic disorders induced by fetal programming. Endocrinology 142, 39643973.CrossRefGoogle ScholarPubMed
379Foster, LA, Ames, NK & Emery, RS (1991) Food intake and serum insulin responses to intraventricular infusions of insulin and IGF-I. Physiol Behav 50, 745749.CrossRefGoogle ScholarPubMed
380Lauterio, TJ, Marson, L, Daughaday, WH, et al. (1987) Evidence for the role of insulin-like growth factor II (IGF-II) in the control of food intake. Physiol Behav 40, 755758.CrossRefGoogle ScholarPubMed
381Firth, SM & Baxter, RC (2002) Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev 23, 824854.CrossRefGoogle ScholarPubMed
382Wheatcroft, SB, Kearney, MT, Shah, AM, et al. (2007) IGF-binding protein-2 protects against the development of obesity and insulin resistance. Diabetes 56, 285294.CrossRefGoogle ScholarPubMed
383Hedbacker, K, Birsoy, K, Wysocki, RW, et al. (2010) Antidiabetic effects of IGFBP2, a leptin-regulated gene. Cell Metab 11, 1122.CrossRefGoogle ScholarPubMed
384Kharitonenkov, A & Larsen, P (2011) FGF21 reloaded: challenges of a rapidly growing field. Trends Endocrinol Metab 22, 8186.CrossRefGoogle ScholarPubMed
385Tyynismaa, H, Raivio, T, Hakkarainen, A, et al. (2011) Liver fat but not other adiposity measures influence circulating FGF21 levels in healthy young adult twins. J Clin Endocrinol Metab 96, E351E355.CrossRefGoogle Scholar
386Badman, MK, Pissios, P, Kennedy, AR, et al. (2007) Hepatic fibroblast growth factor 21 is regulated by PPARα and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab 5, 426437.CrossRefGoogle ScholarPubMed
387Galman, C, Lundasen, T, Kharitonenkov, A, et al. (2008) The circulating metabolic regulator FGF21 is induced by prolonged fasting and PPARα activation in man. Cell Metab 8, 169174.CrossRefGoogle ScholarPubMed
388Kharitonenkov, A, Shiyanova, TL, Koester, A, et al. (2005) FGF-21 as a novel metabolic regulator. J Clin Invest 115, 16271635.CrossRefGoogle ScholarPubMed
389Dushay, J, Chui, PC, Gopalakrishnan, GS, et al. (2010) Increased fibroblast growth factor 21 in obesity and nonalcoholic fatty liver disease. Gastroenterology 139, 456463.CrossRefGoogle ScholarPubMed
390Fisher, FM, Chui, PC, Antonellis, PJ, et al. (2010) Obesity is a fibroblast growth factor 21 (FGF21)-resistant state. Diabetes 59, 27812789.CrossRefGoogle ScholarPubMed
391Xu, J, Lloyd, DJ, Hale, C, et al. (2009) Fibroblast growth factor 21 reverses hepatic steatosis, increases energy expenditure, and improves insulin sensitivity in diet-induced obese mice. Diabetes 58, 250259.CrossRefGoogle ScholarPubMed
392Sarruf, DA, Thaler, JP, Morton, GJ, et al. (2010) Fibroblast growth factor 21 action in the brain increases energy expenditure and insulin sensitivity in obese rats. Diabetes 59, 18171824.CrossRefGoogle ScholarPubMed
393Hondares, E, Iglesias, R, Giralt, A, et al. (2011) Thermogenic activation induces FGF21 expression and release in brown adipose tissue. J Biol Chem 286, 1298312990.CrossRefGoogle ScholarPubMed
394Hammond, GL (2011) Diverse roles for sex hormone-binding globulin in reproduction. Biol Reprod 85, 431441.CrossRefGoogle ScholarPubMed
395Svendsen, OL, Hassager, C & Christiansen, C (1993) Impact of regional and total body composition and hormones on resting energy expenditure in overweight postmenopausal women. Metabolism 42, 15881591.CrossRefGoogle ScholarPubMed
396Abate, N, Haffner, SM, Garg, A, et al. (2002) Sex steroid hormones, upper body obesity, and insulin resistance. J Clin Endocrinol Metab 87, 45224527.CrossRefGoogle ScholarPubMed
397Ding, EL, Song, Y, Manson, JE, et al. (2009) Sex hormone-binding globulin and risk of type 2 diabetes in women and men. N Engl J Med 361, 11521163.CrossRefGoogle ScholarPubMed
398Calle, EE & Kaaks, R (2004) Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer 4, 579591.CrossRefGoogle ScholarPubMed
399Zurlo, F, Larson, K, Bogardus, C, et al. (1990) Skeletal muscle metabolism is a major determinant of resting energy expenditure. J Clin Invest 86, 14231427.CrossRefGoogle Scholar
400van den Berg, SA, van Marken Lichtenbelt, W, Willems van Dijk, K, et al. (2011) Skeletal muscle mitochondrial uncoupling, adaptive thermogenesis and energy expenditure. Curr Opin Clin Nutr Metab Care 14, 243249.CrossRefGoogle ScholarPubMed
401Boström, P, Wu, J, Jedrychowski, MP, et al. (2012) A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481, 463468.CrossRefGoogle ScholarPubMed
402Lee, SJ (2004) Regulation of muscle mass by myostatin. Annu Rev Cell Dev Biol 20, 6186.CrossRefGoogle ScholarPubMed
403Zimmers, TA, Davies, MV, Koniaris, LG, et al. (2002) Induction of cachexia in mice by systemically administered myostatin. Science 296, 14861488.CrossRefGoogle ScholarPubMed
404Rebbapragada, A, Benchabane, H, Wrana, JL, et al. (2003) Myostatin signals through a transforming growth factor β-like signaling pathway to block adipogenesis. Mol Cell Biol 23, 72307242.CrossRefGoogle ScholarPubMed
405McPherron, AC & Lee, SJ (2002) Suppression of body fat accumulation in myostatin-deficient mice. J Clin Invest 109, 595601.CrossRefGoogle ScholarPubMed
406Zhang, C, McFarlane, C, Lokireddy, S, et al. (2012) Inhibition of myostatin protects against diet-induced obesity by enhancing fatty acid oxidation and promoting a brown adipose phenotype in mice. Diabetologia 55, 183193.CrossRefGoogle Scholar
407Guo, T, Jou, W, Chanturiya, T, et al. (2009) Myostatin inhibition in muscle, but not adipose tissue, decreases fat mass and improves insulin sensitivity. PLoS ONE 4, e4937.CrossRefGoogle Scholar
408Hittel, DS, Berggren, JR, Shearer, J, et al. (2009) Increased secretion and expression of myostatin in skeletal muscle from extremely obese women. Diabetes 58, 3038.CrossRefGoogle ScholarPubMed
409Sáinz, N, Rodríguez, A, Catalán, V, et al. (2009) Leptin administration favors muscle mass accretion by decreasing FoxO3a and increasing PGC-1α in ob/ob mice. PLoS ONE 4, e6808.CrossRefGoogle ScholarPubMed
410Brinkoetter, M, Magkos, F, Vamvini, M, et al. (2011) Leptin treatment reduces body fat but does not affect lean body mass or the myostatin–follistatin–activin axis in lean hypoleptinemic women. Am J Physiol Endocrinol Metab 301, E99E104.CrossRefGoogle ScholarPubMed
411Pan, H, Ping, XC, Zhu, HJ, et al. (2012) Association of myostatin gene polymorphisms with obesity in Chinese north Han human subjects. Gene 494, 237241.CrossRefGoogle ScholarPubMed
412Takahashi, N, Lopez, ML, Cowhig, JE Jr, et al. (2005) Ren1c homozygous null mice are hypotensive and polyuric, but heterozygotes are indistinguishable from wild-type. J Am Soc Nephrol 16, 125132.CrossRefGoogle ScholarPubMed
413Szczepanska-Sadowska, E, Paczwa, P & Dobruch, J (2003) Enhanced food and water intake in renin transgenic rats. J Physiol Pharmacol 54, 8188.Google ScholarPubMed
414Uehara, S, Tsuchida, M, Kanno, T, et al. (2003) Late-onset obesity in mice transgenic for the human renin gene. Int J Mol Med 11, 723727.Google ScholarPubMed
415Gratze, P, Boschmann, M, Dechend, R, et al. (2009) Energy metabolism in human renin-gene transgenic rats: does renin contribute to obesity? Hypertension 53, 516523.CrossRefGoogle ScholarPubMed
416Kishimoto, I, Tokudome, T, Nakao, K, et al. (2011) Natriuretic peptide system: an overview of studies using genetically engineered animal models. FEBS J 278, 18301841.CrossRefGoogle ScholarPubMed
417Beleigoli, AM, Diniz, MF & Ribeiro, AL (2009) Natriuretic peptides: linking heart and adipose tissue in obesity and related conditions – a systematic review. Obes Rev 10, 617626.CrossRefGoogle ScholarPubMed
418Pandey, KN (2011) The functional genomics of guanylyl cyclase/natriuretic peptide receptor-A: perspectives and paradigms. FEBS J 278, 17921807.CrossRefGoogle ScholarPubMed
419Kuhn, M (2003) Structure, regulation, and function of mammalian membrane guanylyl cyclase receptors, with a focus on guanylyl cyclase-A. Circ Res 93, 700709.CrossRefGoogle ScholarPubMed
420Sarzani, R, Dessi-Fulgheri, P, Paci, VM, et al. (1996) Expression of natriuretic peptide receptors in human adipose and other tissues. J Endocrinol Invest 19, 581585.CrossRefGoogle ScholarPubMed
421Engeli, S & Sharma, AM (2001) The renin–angiotensin system and natriuretic peptides in obesity-associated hypertension. J Mol Med 70, 2129.CrossRefGoogle Scholar
422Gómez-Ambrosi, J, Catalán, V, Diez-Caballero, A, et al. (2004) Gene expression profile of omental adipose tissue in human obesity. FASEB J 18, 215217.CrossRefGoogle ScholarPubMed
423Moro, C & Smith, SR (2009) Natriuretic peptides: new players in energy homeostasis. Diabetes 58, 27262728.CrossRefGoogle ScholarPubMed
424Birkenfeld, AL, Boschmann, M, Moro, C, et al. (2005) Lipid mobilization with physiological atrial natriuretic peptide concentrations in humans. J Clin Endocrinol Metab 90, 36223628.CrossRefGoogle ScholarPubMed
425Sarzani, R, Marcucci, P, Salvi, F, et al. (2008) Angiotensin II stimulates and atrial natriuretic peptide inhibits human visceral adipocyte growth. Int J Obes 32, 259267.CrossRefGoogle ScholarPubMed
426Wang, TJ, Larson, MG, Levy, D, et al. (2004) Impact of obesity on plasma natriuretic peptide levels. Circulation 109, 594600.CrossRefGoogle ScholarPubMed
427Inuzuka, M, Tamura, N, Yamada, N, et al. (2010) C-type natriuretic peptide as a new regulator of food intake and energy expenditure. Endocrinology 151, 36333642.CrossRefGoogle ScholarPubMed
428Miyashita, K, Itoh, H, Tsujimoto, H, et al. (2009) Natriuretic peptides/cGMP/cGMP-dependent protein kinase cascades promote muscle mitochondrial biogenesis and prevent obesity. Diabetes 58, 28802892.CrossRefGoogle ScholarPubMed
429Birkenfeld, AL, Budziarek, P, Boschmann, M, et al. (2008) Atrial natriuretic peptide induces postprandial lipid oxidation in humans. Diabetes 57, 31993204.CrossRefGoogle ScholarPubMed
430Kim, B (2008) Thyroid hormone as a determinant of energy expenditure and the basal metabolic rate. Thyroid 18, 141144.CrossRefGoogle ScholarPubMed
431Baxter, JD & Webb, P (2009) Thyroid hormone mimetics: potential applications in atherosclerosis, obesity and type 2 diabetes. Nat Rev Drug Discov 8, 308320.CrossRefGoogle ScholarPubMed
432Yen, PM (2001) Physiological and molecular basis of thyroid hormone action. Physiol Rev 81, 10971142.CrossRefGoogle ScholarPubMed
433Zhang, J & Lazar, MA (2000) The mechanism of action of thyroid hormones. Annu Rev Physiol 62, 439466.CrossRefGoogle ScholarPubMed
434Reinehr, T (2010) Obesity and thyroid function. Mol Cell Endocrinol 316, 165171.CrossRefGoogle ScholarPubMed
435Silva, JE (2003) The thermogenic effect of thyroid hormone and its clinical implications. Ann Intern Med 139, 205213.CrossRefGoogle ScholarPubMed
436Alkemade, A (2010) Central and peripheral effects of thyroid hormone signalling in the control of energy metabolism. J Neuroendocrinol 22, 5663.CrossRefGoogle ScholarPubMed
437Barros, RP & Gustafsson, JA (2011) Estrogen receptors and the metabolic network. Cell Metab 14, 289299.CrossRefGoogle ScholarPubMed
438Park, CJ, Zhao, Z, Glidewell-Kenney, C, et al. (2011) Genetic rescue of nonclassical ERα signaling normalizes energy balance in obese Erα-null mutant mice. J Clin Invest 121, 604612.CrossRefGoogle ScholarPubMed
439Foryst-Ludwig, A, Clemenz, M, Hohmann, S, et al. (2008) Metabolic actions of estrogen receptor beta (ERβ) are mediated by a negative cross-talk with PPARγ. PLoS Genet 4, e1000108.CrossRefGoogle ScholarPubMed
440Dillo, WS (2007) Appetite regulation: an overview. Thyroid 17, 433445.CrossRefGoogle Scholar
441Ishii, S, Kamegai, J, Tamura, H, et al. (2008) Triiodothyronine (T3) stimulates food intake via enhanced hypothalamic AMP-activated kinase activity. Regul Pept 151, 164169.CrossRefGoogle ScholarPubMed
442López, M, Varela, L, Vázquez, MJ, et al. (2010) Hypothalamic AMPK and fatty acid metabolism mediate thyroid regulation of energy balance. Nat Med 16, 10011008.CrossRefGoogle ScholarPubMed
443Kaptein, EM, Beale, E & Chan, LS (2009) Thyroid hormone therapy for obesity and nonthyroidal illnesses: a systematic review. J Clin Endocrinol Metab 94, 36633675.CrossRefGoogle ScholarPubMed
444Zhao, LJ, Liu, YJ, Liu, PY, et al. (2007) Relationship of obesity with osteoporosis. J Clin Endocrinol Metab 92, 16401646.CrossRefGoogle ScholarPubMed
445Galusca, B, Zouch, M, Germain, N, et al. (2008) Constitutional thinness: unusual human phenotype of low bone quality. J Clin Endocrinol Metab 93, 110117.CrossRefGoogle ScholarPubMed
446Clemens, TL & Karsenty, G (2011) The osteoblast: an insulin target cell controlling glucose homeostasis. J Bone Miner Res 26, 677680.CrossRefGoogle ScholarPubMed
447Scatena, M, Liaw, L & Giachelli, CM (2007) Osteopontin. A multifunctional molecule regulating chronic inflammation and vascular disease. Arterioscler Thromb Vasc Biol 27, 23022309.CrossRefGoogle ScholarPubMed
448Reinholt, FP, Hultenby, K, Oldberg, A, et al. (1990) Osteopontin – a possible anchor of osteoclasts to bone. Proc Natl Acad Sci U S A 87, 44734475.CrossRefGoogle ScholarPubMed
449Gómez-Ambrosi, J, Catalán, V, Ramírez, B, et al. (2007) Plasma osteopontin levels and expression in adipose tissue are increased in obesity. J Clin Endocrinol Metab 92, 37193727.CrossRefGoogle ScholarPubMed
450Nomiyama, T, Perez-Tilve, D, Ogawa, D, et al. (2007) Osteopontin mediates obesity-induced adipose tissue macrophage infiltration and insulin resistance in mice. J Clin Invest 117, 28772888.CrossRefGoogle ScholarPubMed
451Kiefer, FW, Zeyda, M, Todoric, J, et al. (2008) Osteopontin expression in human and murine obesity: extensive local upregulation in adipose tissue but minimal systemic alterations. Endocrinology 149, 13501357.CrossRefGoogle ScholarPubMed
452Catalán, V, Gómez-Ambrosi, J, Rodríguez, A, et al. (2009) Increased adipose tissue expression of lipocalin-2 in obesity is related to inflammation and matrix metalloproteinase-2 and -9 activity in humans. J Mol Med 87, 803813.CrossRefGoogle ScholarPubMed
453Chapman, J, Miles, PD, Ofrecio, JM, et al. (2010) Osteopontin is required for the early onset of high fat diet-induced insulin resistance in mice. PLoS ONE 5, e13959.CrossRefGoogle ScholarPubMed
454Pietiläinen, KH, Naukkarinen, J, Rissanen, A, et al. (2008) Global transcript profiles of fat in monozygotic twins discordant for BMI: pathways behind acquired obesity. PLoS Med 5, e51.CrossRefGoogle ScholarPubMed
455Hurtado Del Pozo, C, Calvo, RM, Vesperinas-Garcia, G, et al. (2010) IPO8 and FBXL10: new reference genes for gene expression studies in human adipose tissue. Obesity 18, 897903.CrossRefGoogle ScholarPubMed
456Kiefer, FW, Zeyda, M, Gollinger, K, et al. (2010) Neutralization of osteopontin inhibits obesity-induced inflammation and insulin resistance. Diabetes 59, 935946.CrossRefGoogle ScholarPubMed
457Bertola, A, Deveaux, V, Bonnafous, S, et al. (2009) Elevated expression of osteopontin may be related to adipose tissue macrophage accumulation and liver steatosis in morbid obesity. Diabetes 58, 125133.CrossRefGoogle ScholarPubMed
458Syn, WK, Choi, SS, Liaskou, E, et al. (2011) Osteopontin is induced by hedgehog pathway activation and promotes fibrosis progression in nonalcoholic steatohepatitis. Hepatology 53, 106115.CrossRefGoogle ScholarPubMed
459Catalán, V, Gómez-Ambrosi, J, Rodríguez, A, et al. (2011) Up-regulation of the novel proinflammatory adipokines lipocalin-2, chitinase-3 like-1 and osteopontin as well as angiogenic-related factors in visceral adipose tissue of patients with colon cancer. J Nutr Biochem 22, 634641.CrossRefGoogle ScholarPubMed
460Calvo, MS, Eyre, DR & Gundberg, CM (1996) Molecular basis and clinical application of biological markers of bone turnover. Endocr Rev 17, 333368.Google ScholarPubMed
461Kanazawa, I, Yamaguchi, T, Yamamoto, M, et al. (2009) Serum osteocalcin level is associated with glucose metabolism and atherosclerosis parameters in type 2 diabetes mellitus. J Clin Endocrinol Metab 94, 4549.CrossRefGoogle ScholarPubMed
462Fernández-Real, JM & Ricart, W (2011) Osteocalcin: a new link between bone and energy metabolism. Some evolutionary clues. Curr Opin Clin Nutr Metab Care 14, 360366.CrossRefGoogle ScholarPubMed
463Ducy, P (2011) The role of osteocalcin in the endocrine cross-talk between bone remodelling and energy metabolism. Diabetologia 54, 12911297.CrossRefGoogle ScholarPubMed
464Lee, NK, Sowa, H, Hinoi, E, et al. (2007) Endocrine regulation of energy metabolism by the skeleton. Cell 130, 456469.CrossRefGoogle ScholarPubMed
465Martin, TJ (2007) A skeleton key to metabolism. Nat Med 13, 10211023.CrossRefGoogle ScholarPubMed
466Ferron, M, Hinoi, E, Karsenty, G, et al. (2008) Osteocalcin differentially regulates β cell and adipocyte gene expression and affects the development of metabolic diseases in wild-type mice. Proc Natl Acad Sci U S A 105, 52665270.CrossRefGoogle ScholarPubMed
467Ferron, M, McKee, MD, Levine, RL, et al. (2012) Intermittent injections of osteocalcin improve glucose metabolism and prevent type 2 diabetes in mice. Bone 50, 568575.CrossRefGoogle ScholarPubMed
468Kanazawa, I, Yamaguchi, T, Tada, Y, et al. (2011) Serum osteocalcin level is positively associated with insulin sensitivity and secretion in patients with type 2 diabetes. Bone 48, 720725.CrossRefGoogle ScholarPubMed
469Fernández-Real, JM, Izquierdo, M, Ortega, F, et al. (2009) The relationship of serum osteocalcin concentration to insulin secretion, sensitivity, and disposal with hypocaloric diet and resistance training. J Clin Endocrinol Metab 94, 237245.CrossRefGoogle ScholarPubMed
470Yoshikawa, Y, Kode, A, Xu, L, et al. (2011) Genetic evidence points to an osteocalcin-independent influence of osteoblasts on energy metabolism. J Bone Miner Res 26, 20122025.CrossRefGoogle Scholar
Figure 0

Fig. 1 Peripheral factors exerting a direct effect on energy homeostasis grouped by source organ or system. Although due to their multiple production organs some of the elements might be included in more than one organ or system, they have been included only under one organ or system for simplicity reasons. These molecules play an important role in energy homeostasis mainly, but not uniquely, through direct actions on the brain regulation of food intake and on the thermogenic activity of brown adipose tissue (BAT). In some cases, the effect on BAT activation is mediated via the hypothalamus. Ang II, angiotensin II; ANP, atrial natriuretic peptide; ASP, acylation-stimulating protein; BNP, brain natriuretic peptide; CCK, cholecystokinin; CNP, c-type natriuretic peptide; FGF19, fibroblast growth factor-19; FGF21, fibroblast growth factor-21; GIP, glucose-dependent insulinotropic polypeptide; GLP-1, glucagon-like peptide-1; IGF, insulin-like growth factor; PP, pancreatic polypeptide; PYY3–36, peptide YY (peptide tyrosine-tyrosine); SHBG, sex hormone-binding globulin; T3, triiodothyronine; T4, thyroxine. * Although CNP is mainly expressed in the central nervous system, it is also expressed in vascular cells (A colour version of this figure can be found online at http://www.journals.cambridge.org/nrr).