Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Oct 1.
Published in final edited form as: J Steroid Biochem Mol Biol. 2009 Dec 24;122(1-3):65–73. doi: 10.1016/j.jsbmb.2009.12.005

Central Effects of Estradiol in the Regulation of Adiposity

LM Brown 1, DJ Clegg 2,*
PMCID: PMC2889220  NIHMSID: NIHMS167075  PMID: 20035866

Abstract

In recent years, obesity and its associated health disorders and costs have increased. Accumulation of adipose tissue, or fat, in the intra-abdominal adipose depot is associated with an increased risk of developing cardiovascular problems, type-2 diabetes mellitus, certain cancers, and other disorders like the metabolic syndrome. Males and females differ in terms of how and where their body fat is stored, in their hormonal secretions, and in their neural responses to signals regulating weight and body fat distribution. Men and post-menopausal women accumulate more fat in their intra-abdominal depots than pre-menopausal women, resulting in a greater risk of developing complications associated with obesity. The goal of this review is to discuss the current literature on sexual dimorphisms in body weight regulation, adipose tissue accrual and deposition.

Keywords: sex differences, obesity, fat deposition, food intake, neuropeptides, estradiol

Introduction

Gonadal hormones potently control food intake and body weight. In female animals, the activational effects of estradiol acutely and chronically influence body weight homeostasis [13]. In rats and mice, estrogen exerts a tonic inhibitory effect on meal size and daily food intake throughout the ovarian cycle and a cyclic inhibitory effect during the peri-ovulatory phase [1, 35]. Removal of estrogen leads to changes in meal size and duration [6, 7], hyperphagia, and obesity. Estrogen has similar effects in humans where it modulates peri-ovulatory decreases in daily food intake [8]. Additionally, reductions in estrogen are associated with changes in body weight and fat distribution in humans, which parallel the findings in animals [8]. Estrogen has the ability to control energy balance, food intake, and body fat distribution and this may be mediated through its interaction with orexigenic and anorexigenic hormones. This review aims to explore these interactions and discuss the link between estrogen and obesity.

1.0 Estrogen regulates adiposity

The accumulation of fat in a central distribution (intraabdominal) has emerged as a risk factor for the metabolic syndrome [9, 10] which includes a higher risk of diabetes, hypertriglyceridemia, hypertension, and cardiovascular disease [11]. Estrogen promotes the accumulation of subcutaneous fat [12], and the loss of estrogen with menopause is associated with an increase in central fat [10, 13]. The sexual dimorphism in adipose tissue distribution may partially explain the greater risk for the metabolic syndrome in men compared with premenopausal women.

1.1 Estrogen regulates adipose tissue distribution

Visceral fat varies inversely with estrogen levels [14]. When estrogen levels become sufficiently low visceral fat accumulation occurs in females, possibly due to direct effects of estrogen, especially since progesterone and androgen receptors (PR and AR) as well as estrogen receptor (ER) are expressed in adipose tissues [15]. Subcutaneous adipose tissue has higher concentrations of ER and PR; however, visceral adipose tissue has higher concentrations of AR [16]. Additionally, subcutaneous adipose tissue has few androgen receptors, and estrogen down-regulates AR expression in subcutaneous fat [17]. Adipose tissue-specific AR knockout mice have increased intra-adipose estradiol levels, which leads to increased subcutaneous obesity and hyperleptinemia [18].

Ovariectomized (OVX) rats gain fat, specifically visceral fat with no change of subcutaneous fat [19]. Peripheral or central administration of estradiol to OVX rats restores central leptin sensitivity and changes their body fat distribution to mirror that of intact females; additionally, altering the sex hormone milieu in males with estradiol administration increases sensitivity to central leptin and increases subcutaneous fat deposition [19]. An important implication from these findings is that estrogen regulates body fat distribution, interacts with the integrated adiposity message conveyed to the brain by leptin, and enhances leptin’s action in sympathetic activation to the visceral fat, which facilitates fat mobilization in the visceral depot and fat deposition in the subcutaneous depot.

1.1 Estrogen regulates adiposity through estrogen receptors

Estrogen regulates body adiposity and fat distribution through its receptors, ER alpha (ERα) and beta (ERβ) [20, 21]. However, only ERα has been reported to have a major influence on energy homeostasis [22]. ERα is necessary for estradiol’s genomic actions on body weight regulation [23] while ERβ functions more as a modulator of estrogen actions [24]. Rapid, non-genomic actions of estradiol also have been described and some of them appear to involve ERα [25].

Heine et al. [22] reported that male and female mice with total body deletion of ERα, ERα-knock-out (αERKO) mice, have increased adiposity in both male and female mice, suggesting an important role for this estrogen receptor in the regulation of body weight and adiposity. Recently, site-specific knockdown of ERα expression in the VMH, a brain region critical for body weight regulation, demonstrates the role of VMH ERα activity in the regulation of body weight homeostasis [23]. Knockdown of VMH ERα results in obesity due to an anabolic process, with changes in energy expenditure primarily mediating the weight gain [23]. These data are consistent to previous finding in the αERKO mice where it has been demonstrated that the obesity is primarily due to changes in energy expenditure rather than changes in food intake [22, 26] and those mice have increased visceral adiposity (unpublished data). These data suggest that estrogen signaling with critical hypothalamic nuclei is responsible for the regulation of body weight via modulating energy expenditure.

Since ERá is expressed in hypothalamic areas that regulate energy homeostasis [2733], the absence of ERá expression is consistent with changes in body weight. Furthermore, ERá polymorphisms identified in humans have been associated with increased levels of visceral fat.

A ventral medial nucleus (VMN) specific ERá knockdown in both female mice and rats resulted in phenotypes characteristic of a metabolic syndrome [23]. Microinjections of low doses of estradiol directly into the brain were shown to inhibit food intake [19, 34, 35]. Taken together, these observations suggest that the binding of estradiol to ERá in the hypothalamus, or elsewhere in the brain, may represent a mechanism by which estradiol regulates food intake, body weight, and possibly body fat distribution.

1.2 Estrogen regulates adiposity by decreasing inflammation

Obesity is a state of chronic inflammation, and inflammatory signaling pathways in obesity are linked to insulin resistance [3638]. Sex differences where females are protected have been reported in diet-induced obesity, insulin resistance and inflammatory response to a high-fat (HF) diet [3941]. This may be explained in part by the anti-inflammatory properties of estrogen [42]. Recent studies have shown that estradiol may play a role in reducing the inflammatory response in adipose, cardiovascular, and neural systems [43], in addition to being neuroprotective both in vivo and in vitro [44, 45].

ERá (and in some cases ERâ) is expressed in immune and cytokine-producing cells including macrophages and microglia, and in vitro studies have shown estradiol-activated ERá decreases the number of pro-inflammatory cytokines [44, 45]. The anti-inflammatory properties of estradiol can be partially explained by the ability of ERs to act as transcriptional repressors by inhibiting the activity of nuclear factor kappa B (NFêB) through protein-protein interactions between agonist-bound ERs and activated NFêB subunits [42, 46, 47]. Estradiol’s inhibitory effect on NFêB function is not fully understood and may be target selective [4749].

Symptoms of a metabolic syndrome increase when animals are maintained on a high-fat (HF) diet or when females have low ovarian hormone levels. Free fatty acids (FFAs), particularly saturated fatty acids, increase inflammation by activating toll-like receptor 4 (TLR4) [50]. Muscle and liver expression of tumor necrosis factor-alpha (TNFá), interleukin-6 (IL-6), and NFêB also increase with HF diets [50]. Estradiol has been shown to be neuroprotective and to increase the expression of growth factors and proteins involved in apoptosis [51, 52].

Estradiol signaling pathways are active in monocytes and macrophages, and ERs are expressed by these cells [45, 53]. Therefore, the protective effects of estradiol in neurodegenerative diseases can be mediated by inhibiting the inflammatory response, and consequently, hormone withdrawal can increase inflammation [53].

Both in a model of brain inflammation and in primary cultures of microglial cells, estradiol inhibited the synthesis of inflammatory mediators induced by lipopolysaccharide (LPS) [44, 45, 53, 54]. Moreover, hormone loss in OVX mice resulted in increased microglial activation [54], while estradiol replacement decreased microglia activation [53]. These data provide strong evidence that chronic inflammation in the brain can be regulated by estradiol.

2.0 Estrogen interacts with orexigenic neuropeptides

Estrogen has been proposed to act directly and indirectly to decrease orexigenic peptides and decrease food intake. In this section, we will review the literature and describing the interactions of estrogen and neuropeptides that increase food intake.

2.1Neuropeptide Y

Neuropeptide Y (NPY) is an important central regulator of energy homeostasis in a many hypothalamic neurons [55, 56]. NPY is a potent orexigen, which increases feeding behavior in fed and fasted animals [57]. Estrogen acts via the estrogen receptors (ERs) in the hypothalamus to reduce feeding [58], and may mediate its anorectic effects by decreasing NPY expression or release [59]. In addition, Sar et al. reported colocalization of estradiol and NPY immunoreactivity in ARC neurons, suggesting that estradiol may modulate NPY neurosecretion in the hypothalamus [60]. However, it remains unclear whether estrogen directly affects NPY neurons and in which area of the brain this effect occurs [61].

Ovariectomized (OVX) rats experience a rapid weight gain, which can be reversed by estradiol replacement [6, 59]. administered either peripherally or centrally [19, 62]. Estradiol deficiency results in increased NPY concentrations in the paraventricular nucleus (PVN) of the hypothalamus [63] and elevated NPY mRNA expression in the arcuate nucleus (ARC) of the hypothalamus [64, 65]. Bonavera et al. reported that estradiol treatment decreased hypothalamic NPY levels in the PVN of OVX rats [59, 63].

NPY neurons in the hypothalamus not only affect feeding, but they also influence reproduction. Therefore, estradiol can modulate both of these neuroendrocrine systems by regulating NPY gene expression. NPY neurons are activated by signals indicating reduced energy availability, such as decreased levels of circulating glucose, leptin or insulin, which increase NPY release in the PVN to stimulate feeding [66]. This may be due to estradiol stimulating NPY [67], NPY Y1 receptor [68] expression and NPY release [59]. In an ex vivo hypothalamic neuronal cell line, N-38, estradiol affected the expression of NPY in a biphasic manner, which corresponded to changes in the ERá:ERâ ratio. When the ERá:ERâ ratio was high, NPY transcription was repressed; conversely, when the ratio was low, NPY transcription was stimulated. These results provided evidence that the ratio of ERs expression in the hypothalamus may differentially regulate NPY in vivo [61].

2.2 Ghrelin

Ghrelin is produced in the stomach and acts on growth hormone secretagogue receptors (GHSRs) to increase food intake. While mainly synthesized by the stomach, ghrelin is also found the hypothalamus, pituitary gland, hippocampus, brain cortex, adrenal gland, intestine, and pancreas [6971]. Exogenous ghrelin is less potent in intact female rats than in male rats or OVX females [72]. Central intra-third ventricular (i3vt) or peripheral ghrelin administration reliably increased feeding in intact male rats and OVX females [7379]; however, the threshold for a significant increase in feeding using either administration route was significantly greater in intact females than in males or OVX females [72]. When OVX rats were treated with estradiol, moderate intra-peritoneal (i.p.) or i3vt doses of ghrelin no longer stimulated eating. Together, these data demonstrate that estradiol reduces the orexigenic potency of ghrelin. Lastly, estradiol reduced the eating-stimulatory effect of i3vt ghrelin in male rats, indicating that the estrogenic effect exists in both sexes, which is a point of potential therapeutic relevance [72]. Careful attention to sex differences and gonadal hormone status should be included in the development of any ghrelin-based clinical control for eating behaviors.

The eating-stimulatory effect of ghrelin varies across different phases of the ovarian cycle in intact rats [72]. Administration of i3vt ghrelin had no reliable overall effect when cycle day was not taken into account. However, when the cycle day was considered, i3vt ghrelin increased eating during diestrus 1 and diestrus 2, but not during proestrus or estrus. In addition, in estradiol-treated OVX rats, ghrelin increased food intake on the days that modeled diestrus in intact rats, but not on the days that modeled proestrus or estrus. This indicates that there are cyclic variation in eating in rats and mice, and spontaneous food intake is maximal during diestrus and minimal during estrus. Therefore, the analogous peri-ovulatory decreases in eating in women may be due to changes in estrogenic tone that affect ghrelin's eating-stimulatory action [72]. To assess the importance of ghrelin signaling in OVX-induced hyperphagia and obesity, Ghsr−/− mice lacking GHSR were OVX [72]. The OVX mice, which were similar to wild-type mice in body weight and food intake pre-surgery, showed no increase in food intake or body weight gain after surgery. This indicates that estradiol tonically inhibits endogenous ghrelin signaling in mice and that release from this inhibition is necessary for OVX mice to increase food intake and body weight. This mechanism may account for other sex differences in eating and weight regulation previously reported in Ghsr−/− mice. For example, female Ghsr−/− mice accumulated less body weight and adiposity when given a high-fat diet [80]. Also, the magnitude of the differences in adiposity observed between Ghsr−/− and wild-type mice were greater in females than in males [72]. Ghrelin signaling appears to be a necessary component of the estrogenic control of eating and weight regulation [72].

The site of the GHSR-mediated effects on eating remain unclear [78, 8183], so estradiol may influence ghrelin and its receptors either centrally or peripherally [72]. In male brains, GHSR have been implicated in ghrelin's eating-stimulatory effect in the ARC, PVN, ventral tegmental area, and dorsal vagal complex [76, 8388]. Since administration of exogenous ghrelin can reach all of these ER-containing sites, each area could mediate the observed sex differences [72]. In contrast, Currie et al. failed to observe any sex difference following direct ghrelin microinjections to the ARC or PVN [89]. However, since ovarian cycling was not monitored, it is possible that the female rats were acyclic, which may have caused an artificial increase in ghrelin-induced eating. It is also possible that estrogenic actions on nucleus tractus solitaries (NTS) neurons contribute to the observed effects. Exogenous estradiol was shown to act on ERá-expressing neurons in the NTS to inhibit eating [90], and ghrelin microinjection into the NTS stimulated eating in male rats [87].

2.3 Melanin-Concentrating Hormone (MCH)

Since its discovery in hypothalamic neurons [91, 92], MCH has been recognized as an important regulator of energy homeostasis [93]. Central administration of MCH promotes feeding [94, 95], while genetic ablation of the Mch gene produces a lean phenotype [96, 97]. In addition, Mch is upregulated by fasting [94]. MCH neurons in the lateral hypothalamic area (LHA) receive inputs from NPY/AgRP and POMC neurons in the ARC [98103]. Therefore, MCH neurons are in a position to integrate the feeding response because they have projections from the ARC and to brain structures like the nucleus accumbens [104].

Messina et al. investigated the effect of centrally injected MCH on feeding in estradiol- and vehicle-treated OVX rats and male rats. MCH increased the meal size in all three groups. Additionally, intake increased more in rats in diestrus than rats in estrus, although MCH increased food intake in both groups. Overall, estradiol decreased the orexigenic effect of MCH, leading the authors to hypothesize that the decrease in food intake during estrus was mediated by a decrease in MCH signaling [7]. In a second study, Santollo et al. examined whether the behavioral effects of MCH were sexually dimorphic. They observed a greater increase in food intake, meal size, and water intake following MCH treatment in male rats than in estradiol-treated OVX rats [6]. Additionally, they observed that higher MCH doses were necessary to increase food intake in estradiol-treated OVX rats, suggesting that estradiol reduced MCH sensitivity in female rats [6]. Given that circulating levels of estradiol are lower in males than in females, this sex difference may contribute to the increased sensitivity to MCH in male rats.

There are several ways that estradiol could decrease MCH signaling. By acting on nuclear ERs in the lateral hypothalamus (LH) and zona incerta (ZI) [105], estradiol can alter gene transcription. ERs can modulate gene expression locally by decreasing MCH synthesis. In support of this hypothesis, physiological doses of estradiol decreased pre-pro MCH mRNA expression in the ZI of OVX rats [106] and the LH of obese male rats [107]. In addition, chronic estradiol treatment in male rats blocked increases in LH MCH mRNA expression induced by fasting [107]. In contrast, pharmacological doses of estradiol in male mice increased MCH mRNA within hypothalamic tissue punches [108]. These discrepancies emphasize the need for additional research in intact, cycling rats to determine the role of endogenous estradiol in regulating MCH mRNA expression. It is also possible that estradiol, acting at nuclear ERs in brain regions that express MCH-1 receptors [28, 109], can decrease MCH signaling by decreasing the number or binding affinity of MCH-1 receptors. A recent study demonstrated that LH neurons containing MCH-1 receptors and ERs are not coexpressed, but are in close proximity to one another [110].

Several studies evaluated whether MCH affects meal size or number [6, 7, 111]. The size of the first meal increased following MCH treatment in male and estradiol- or vehicle-treated OVX rats. In male and vehicle-treated OVX rats, MCH increased the average meal size throughout the period of increased food intake. There was no change in meal size in the estradiol-treated OVX rats, and there were no differences in meal number in any group, indicating that MCH increases food intake by increasing meal size in both males and females [6].

3.0 Estrogen interacts with anorexigenic neuropeptides

Estrogen has been reported to have an inhibitory effect on body weight gain in animal models [4, 19, 62]. ERá null mice are obese, insulin resistant, and have decreased energy expenditure [22, 112]. This model indicates that ERá is critical for the estrogenic control of feeding behaviors and body weight [112]. Estrogen decreases food intake through its direct effects and through its interactions with other compounds that reduce food intake. In this section we will review the literature on estrogen’s interactions with anorexigenic hormones.

3.1 Insulin

After Kennedy (1953) hypothesized that fat stores produced a hormone that functioned as a negative feedback control for adiposity[113], one early suggestion was that the hormone was insulin [114, 115]. Plasma insulin levels directly correlate with body weight and adiposity [116118]. Obese animals and humans have higher basal insulin levels and secrete more insulin in response to a meal than lean individuals [116, 119]. Insulin increases during meals and other periods of positive energy balance and decreases during fasting and other periods of negative energy balance.

Insulin receptors are distributed in discrete brain areas, including the hypothalamus [120122]. Hypothalamic insulin receptors are thought to mediate food intake and body weight regulation via mechanisms similar to leptin [102, 123126].

Hormones provide important regulatory signals to the brain. Manipulation of gonadal steroid levels can influence leptin and insulin sensitivities and body fat distribution [19]. This implies that the relative amount of androgen and estradiol is a key determinant of the brain’s sensitivity to the catabolic actions of insulin. When there is proportionally less estrogen, this favors insulin sensitivity. Thus, estradiol directly determines body fat distribution through its actions in the brain.

3.2 Leptin

First described in 1994 [127], leptin has proven to be a key metabolic protein with actions throughout the body. Analogous to insulin, plasma leptin levels are directly correlated with adiposity. Circulating leptin is transported into the brain, where leptin receptors exist in many areas, including the ARC [128, 129]. Leptin has many actions within the brain, including reducing food intake and increasing energy expenditure.

Male and female rats differentially respond to centrally administered leptin [62, 130]. In females, estradiol alters the sensitivity to centrally administered leptin and changes the body fat distribution [19]. Peripheral or central administration of estradiol to OVX females restores their central leptin sensitivity and changes their body fat distribution to be more similar to intact females. Additionally, administering estradiol to males increases the sensitivity to central leptin and increases subcutaneous fat deposition [19]. These findings indicate that gonadal steroids mediate body fat distribution and interact with the integrated adiposity message conveyed to the brain by leptin, which results in differential sensitivities of this signal in males and females.

Leptin provides a powerful catabolic signal to the brain, resulting in inhibition of food intake [99, 102, 123, 131136]. Leptin levels are higher in females, even before puberty, compared with males, and these levels are independent of differences in body composition [137139]. After puberty, estrogen and testosterone further modulate leptin synthesis and secretion via sex steroid receptor-dependent transcriptional mechanisms [140]. Leptin is secreted from adipose tissue in direct proportion to fat content, and it penetrates the blood-brain barrier to interact with leptin receptors in the hypothalamus and brainstem [99, 123, 131, 134, 141143]. Although several splice variants of the leptin receptor are known, OB-Rb is the critical variant for regulating energy balance [144].

ERs are also expressed in the brain, including the hypothalamic regions that regulate food intake and body weight [21, 27, 28, 3032, 105, 145]. OB-Rb expression has been colocalized with ERá in the ARC [146], and estrogen has been reported to regulate the expression of OB-Rb mRNA [147], possibly via an estrogen-responsive element on the leptin receptor gene [148]. The extensive hypothalamic colocalization of these two receptors suggests there is a closely coupled interaction for regulating the behavioral and neuroendrocrine mechanisms. Studies indicate that when estrogen levels are low, central leptin sensitivity is reduced, as in OVX females [63] and intact males. Conversely, when estrogen levels are relatively high, as in intact females, and in both estradiol-treated OVX females and males, leptin sensitivity is high [62, 149].

OB-Rb leptin receptors colocalize with several neuropeptides believed to be involved in controlling food intake and reproduction. Leptin has the ability to activate or inhibit hypothalamic neurons [150152]. Thus, leptin is ideally situated to link metabolic status and brain function. Diano et al. reported colocalization of leptin and estrogen receptors [146]. Animals with higher estrogen levels have higher plasma leptin and higher hypothalamic OB-Rb expression levels [19]. There is an inverse relationship between plasma leptin and hypothalamic OB-Rb mRNA, which is not correlated with estrogen status [19]. Additionally, hypothalamic OB-Rb mRNA expression was greater in OVX animals than in intact females, consistent with previous findings [153]. These results were in contrast with one study that observed no difference in hypothalamic OB-Rb mRNA expression between sham and OVX rats [63] and a second that reported decreased OB-Rb expression in OVX rats in comparison to sham-operated females [154]. Kimura et al. detected no change in plasma leptin levels in OVX females, despite observing that the OVX rats weighed significantly more than the sham-operated females [154]. In contrast, in other studies, a significant increase in plasma leptin was observed following the weight gain associated with OVX [19, 63]. These varying results indicate that the assay time may be critical for understanding the physiological roles of estrogen and leptin in regulating body weight. Additionally, since only OB-Rb mRNA expression has been measured, it is unknown how estrogen may impact the OB-Rb protein and signaling.

The differences in leptin sensitivity caused by the presence or absence of estrogen must occur downstream of OB-Rb transcription and translation (Clegg 2006). Additionally, females have greater c-Fos and pSTAT3 immunoreactivity in the ARC than males following i3vt leptin (unpublished data), which suggests that despite having fewer hypothalamic OB-Rbs, females have greater leptin signaling in this brain region. Therefore, there may be an estrogen threshold required to enhance the central sensitivity to leptin. In addition, estradiol treatment in intact males increased their sensitivity to centrally administered leptin.

3.3 Serotonin

Estradiol decreases food intake by selectively influencing the neural controls of meal size, which likely require the serotonergic system [155]. Estradiol heightens the anorexia induced by increased serotonergic neurotransmission [156] and decreases the hyperphagia induced by decreased serotonergic neurotransmission [157, 158]. Estradiol was reported to increase the expression of the serotonin transporter (5-HTT) in the dorsal and median raphe nuclei, allowing estradiol to regulate the serotonin system [159]. Other labs reported that acute injection of estradiol increased 5-HTT mRNA levels in the dorsal raphe nuclei (DRV) of OVX rats [160, 161]. The DRV has reciprocal connections with the hypothalamus and parabrachial nucleus [162, 163] putting the DRV in a position to directly regulate part of the neural network controlling food intake [159]. The estradiol/serotonin interaction may also have a clinical relevance, since human 5-HTT promoter polymorphisms are associated with anorexia nervosa, an eating disorder that primarily affects women [164, 165].

3.4 Cholecystokinin (CCK)

Cholecystokinin (CCK) is synthesized and released from cells of the upper intestine and acts on abdominal CCK-A receptors. It plays a variety of roles in the digestive process, including slowing gastric emptying and intestinal motility (see review, Moran, 2009) [166]. CCK exerts its satiety action primarily by activating subdiaphragmatic vagal afferent neurons [1, 167169]. Administration of CCK antagonists increases food intake by increasing meal size [170]. Several experiments highlighted the interactions between estrogen and CCK. CCK-A antagonists decreased food intake to a greater extent when intact female or estradiol-treated OVX rats were in estrus, and this effect was lessened in diestrus rats [171174].

Because CCK satiation depends on vagal afferents [175177], up-regulation of CCK receptors in the terminals of vagal afferent fibers may account for the increased CCK sensitivity. To investigate this hypothesis, in vitro quantitative autoradiography was used to measure the effects of estradiol on the binding characteristics of CCK receptors in the nucleus of the solitary tract (NTS), a brain area that receives terminal projections of abdominal vagal afferent fibers [177], and in two interconnected areas, the area postrema (AP) and the VMN. Estradiol treatment in OVX animals did not alter the number or affinity of CCK receptors [178], suggesting that the up-regulation of CCK receptors does not mediate the increased sensitivity to CCK following estradiol treatment. Estrogen increased the potency of CCK by increasing the sensitivity of vagal CCK-A receptors, but did not increase CCK secretion or the number of CCK-A receptors [178181].

CCK’s effects on meal size has been characterized in male rats by examining the pattern of c-Fos expression after consumption [182187] or injection of CCK [188192]. Estradiol treatment in OVX rats increased the number of feeding- and CCK-induced c-Fos-positive cells within the NTS, PVN, and the central nucleus of the amygdala [183, 193]. These data suggest that exogenous estradiol may decrease meal size by selectively increasing neuronal activity in multiple brain areas that control meal size. It is currently unknown whether a similar mechanism underlies the decrease in meal size or increase in CCK satiation, which occurs during estrus in cycling rats.

Although there is solid evidence that estradiol increases the potency by which CCK exerts its direct, inhibitory control over meal size, such an interaction does not completely account for the decrease in food intake during estrus in gonadally intact rats or following estradiol treatment in OVX rats. For example, blocking the release of CCK during a meal attenuated, but did not block, the phasic inhibitory decrease in meal size, observed during estrus in gonadally intact rats [174]. Additionally, endogenous CCK does not appear to play a role in the tonic inhibitory action of estradiol [172, 174]. Thus, estradiol must modulate the potency of other stimuli that directly generate negative feedback during a meal.

4.0 Summary

This review focused on the literature describing estrogen’s interactions with orexigenic and anorexigenic neuropeptides and how these interactions affect food intake and adiposity. In addition, estrogen’s steroid structure imparts anti-inflammatory effects that may explain how intact females on HF diets decrease inflammation. Understanding the neurophysiology of estrogen may lead to possible interventions for postmenopausal women, who are at an increased risk of metabolic syndrome.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Geary N. Estradiol, CCK and satiation. Peptides. 2001;22(8):1251–1263. doi: 10.1016/s0196-9781(01)00449-1. [DOI] [PubMed] [Google Scholar]
  • 2.Hrupka BJ, Smith GP, Geary N. Hypothalamic implants of dilute estradiol fail to reduce feeding in ovariectomized rats. Physiol Behav. 2002;77(2–3):233–241. doi: 10.1016/s0031-9384(02)00857-0. [DOI] [PubMed] [Google Scholar]
  • 3.Geary N. The estrogenic inhibition of eating. In: Stricker EM, Woods SC, editors. Handbook of Behavioral Neuroscience. New York: Springer; 2004. pp. 304–343. [Google Scholar]
  • 4.Drewett RF. Sexual behaviour and sexual motivation in the female rat. Nature. 1973;242(5398):476–477. doi: 10.1038/242476a0. [DOI] [PubMed] [Google Scholar]
  • 5.Asarian L, Geary N. Modulation of appetite by gonadal steroid hormones. Philos Trans R Soc Lond B Biol Sci. 2006;361(1471):1251–1263. doi: 10.1098/rstb.2006.1860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Santollo J, Eckel LA. Estradiol decreases the orexigenic effect of neuropeptide Y, but not agouti-related protein, in ovariectomized rats. Behav Brain Res. 2008;191(2):173–177. doi: 10.1016/j.bbr.2008.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Messina MM, Boersma G, Overton JM, Eckel LA. Estradiol decreases the orexigenic effect of melanin-concentrating hormone in ovariectomized rats. Physiol Behav. 2006;88(4–5):523–528. doi: 10.1016/j.physbeh.2006.05.002. [DOI] [PubMed] [Google Scholar]
  • 8.Geary N, Lovejoy J. Sex differences in energy metabolism, obesity and eating behavior. In: Jill KJB, Becker B, Geary Nori, Hampson Elizabeth, Herman JamesP, Young Elizabeth, editors. Sex Differences in the Brain: From Genes to Behavior. New York: Oxford University Press; 2007. [Google Scholar]
  • 9.Kannel WB, Cupples LA, Ramaswami R, Stokes J, Kreger BE, Higgins M. Regional obesity and risk of cardiovascular disease; the Framingham study. J Clin Eipidem. 1991;44:183–190. doi: 10.1016/0895-4356(91)90265-b. [DOI] [PubMed] [Google Scholar]
  • 10.Lee CG, Carr MC, Murdoch SJ, Mitchell E, Woods NF, Wener MH, Chandler WL, Boyko EJ, Brunzell JD. Adipokines, inflammation, and visceral adiposity across the menopausal transition: a prospective study. The Journal of clinical endocrinology and metabolism. 2009;94(4):1104–1110. doi: 10.1210/jc.2008-0701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Despres JP. Abdominal obesity as important component of insulin-resistance syndrome. Nutrition. 1993;9(5):452–459. [PubMed] [Google Scholar]
  • 12.Krotkiewski M, Bjorntorp P, Sjostrom L, Smith U. Impact of obesity on metabolism in men and women. Importance of regional adipose tissue distribution. The Journal of clinical investigation. 1983;72(3):1150–1162. doi: 10.1172/JCI111040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Poehlman ET, Toth MJ, Gardner AW. Changes in energy balance and body composition at menopause: a controlled longitudinal study. Ann Intern Med. 1995;123(9):673–675. doi: 10.7326/0003-4819-123-9-199511010-00005. [DOI] [PubMed] [Google Scholar]
  • 14.Bouchard C, Despres JP, Mauriege P. Genetic and nongenetic determinants of regional fat distribution. Endocrine reviews. 1993;14(1):72–93. doi: 10.1210/edrv-14-1-72. [DOI] [PubMed] [Google Scholar]
  • 15.Crandall DL, Busler DE, Novak TJ, Weber RV, Kral JG. Identification of estrogen receptor beta RNA in human breast and abdominal subcutaneous adipose tissue. Biochem Biophys Res Commun. 1998;248(3):523–526. doi: 10.1006/bbrc.1998.8997. [DOI] [PubMed] [Google Scholar]
  • 16.Lu SF, McKenna SE, Cologer-Clifford A, Nau EA, Simon NG. Androgen receptor in mouse brain: sex differences and similarities in autoregulation. Endocrinology. 1998;139(4):1594–1601. doi: 10.1210/endo.139.4.5863. [DOI] [PubMed] [Google Scholar]
  • 17.Bjorntorp P. Hormonal control of regional fat distribution. Human Reproduction. 1997;12 Suppl 1:21–25. doi: 10.1093/humrep/12.suppl_1.21. [DOI] [PubMed] [Google Scholar]
  • 18.Yu IC, Lin HY, Liu NC, Wang RS, Sparks JD, Yeh S, Chang C. Hyperleptinemia without obesity in male mice lacking androgen receptor in adipose tissue. Endocrinology. 2008;149(5):2361–2368. doi: 10.1210/en.2007-0516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Clegg DJ, Brown LM, Woods SC, Benoit SC. Gonadal hormones determine sensitivity to central leptin and insulin. Diabetes. 2006;55(4):978–987. doi: 10.2337/diabetes.55.04.06.db05-1339. [DOI] [PubMed] [Google Scholar]
  • 20.Matelski H, Greene R, Huberman M, Lokich J, Zipoli T. Randomized trial of estrogen vs. tamoxifen therapy for advanced breast cancer. Am J Clin Oncol. 1985;8(2):128–133. doi: 10.1097/00000421-198504000-00004. [DOI] [PubMed] [Google Scholar]
  • 21.Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA. Cloning of a novel receptor expressed in rat prostate and ovary. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(12):5925–5930. doi: 10.1073/pnas.93.12.5925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(23):12729–12734. doi: 10.1073/pnas.97.23.12729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Musatov S, Chen W, Pfaff DW, Mobbs CV, Yang XJ, Clegg DJ, Kaplitt MG, Ogawa S. Silencing of estrogen receptor alpha in the ventromedial nucleus of hypothalamus leads to metabolic syndrome. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(7):2501–2506. doi: 10.1073/pnas.0610787104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Schomberg DW, Couse JF, Mukherjee A, Lubahn DB, Sar M, Mayo KE, Korach KS. Targeted disruption of the estrogen receptor-alpha gene in female mice: characterization of ovarian responses and phenotype in the adult. Endocrinology. 1999;140(6):2733–2744. doi: 10.1210/endo.140.6.6823. [DOI] [PubMed] [Google Scholar]
  • 25.McEwen BS, Alves SE. Estrogen actions in the central nervous system. Endocrine reviews. 1999;20(3):279–307. doi: 10.1210/edrv.20.3.0365. [DOI] [PubMed] [Google Scholar]
  • 26.Ohlsson C, Hellberg N, Parini P, Vidal O, Bohlooly M, Rudling M, Lindberg MK, Warner M, Angelin B, Gustafsson JA. Obesity and disturbed lipoprotein profile in estrogen receptor-alpha- deficient male mice. Biochem Biophys Res Commun. 2000;278(3):640–645. doi: 10.1006/bbrc.2000.3827. [DOI] [PubMed] [Google Scholar]
  • 27.Merchenthaler I, Lane MV, Numan S, Dellovade TL. Distribution of estrogen receptor alpha and beta in the mouse central nervous system: in vivo autoradiographic and immunocytochemical analyses. The Journal of comparative neurology. 2004;473(2):270–291. doi: 10.1002/cne.20128. [DOI] [PubMed] [Google Scholar]
  • 28.Osterlund M, Kuiper GG, Gustafsson JA, Hurd YL. Differential distribution and regulation of estrogen receptor-alpha and -beta mRNA within the female rat brain. Brain Res Mol Brain Res. 1998;54(1):175–180. doi: 10.1016/s0169-328x(97)00351-3. [DOI] [PubMed] [Google Scholar]
  • 29.Shima N, Yamaguchi Y, Yuri K. Distribution of estrogen receptor beta mRNA-containing cells in ovariectomized and estrogen-treated female rat brain. Anat Sci Int. 2003;78(2):85–97. doi: 10.1046/j.0022-7722.2003.00042.x. [DOI] [PubMed] [Google Scholar]
  • 30.Simerly RB, Chang C, Muramatsu M, Swanson LW. Distribution of androgen and estrogen receptor mRNA-containing cells in the rat brain: an in situ hybridization study. The Journal of comparative neurology. 1990;294(1):76–95. doi: 10.1002/cne.902940107. [DOI] [PubMed] [Google Scholar]
  • 31.Simonian SX, Herbison AE. Differential expression of estrogen receptor alpha and beta immunoreactivity by oxytocin neurons of rat paraventricular nucleus. Journal of neuroendocrinology. 1997;9(11):803–806. doi: 10.1046/j.1365-2826.1997.00659.x. [DOI] [PubMed] [Google Scholar]
  • 32.Voisin DL, Simonian SX, Herbison AE. Identification of estrogen receptor-containing neurons projecting to the rat supraoptic nucleus. Neuroscience. 1997;78(1):215–228. doi: 10.1016/s0306-4522(96)00551-9. [DOI] [PubMed] [Google Scholar]
  • 33.Wilkinson HA, Dahllund J, Liu H, Yudkovitz J, Cai SJ, Nilsson S, Schaeffer JM, Mitra SW. Identification and characterization of a functionally distinct form of human estrogen receptor beta. Endocrinology. 2002;143(4):1558–1561. doi: 10.1210/endo.143.4.8829. [DOI] [PubMed] [Google Scholar]
  • 34.Butera PC, Beikirch RJ. Central implants of diluted estradiol: independent effects on ingestive and reproductive behaviors of ovariectomized rats. Brain research. 1989;491(2):266–273. doi: 10.1016/0006-8993(89)90062-0. [DOI] [PubMed] [Google Scholar]
  • 35.Palmer K, Gray JM. Central vs. peripheral effects of estrogen on food intake and lipoprotein lipase activity in ovariectomized rats. Physiol Behav. 1986;37(1):187–189. doi: 10.1016/0031-9384(86)90404-x. [DOI] [PubMed] [Google Scholar]
  • 36.Kahn BB, Flier JS. Obesity and insulin resistance. The Journal of clinical investigation. 2000;106(4):473–481. doi: 10.1172/JCI10842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wellen KE, Hotamisligil GS. Obesity-induced inflammatory changes in adipose tissue. The Journal of clinical investigation. 2003;112(12):1785–1788. doi: 10.1172/JCI20514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. The Journal of clinical investigation. 2005;115(5):1111–1119. doi: 10.1172/JCI25102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Dhar MS, Sommardahl CS, Kirkland T, Nelson S, Donnell R, Johnson DK, Castellani LW. Mice heterozygous for Atp10c, a putative amphipath, represent a novel model of obesity and type 2 diabetes. J Nutr. 2004;134(4):799–805. doi: 10.1093/jn/134.4.799. [DOI] [PubMed] [Google Scholar]
  • 40.Gallou-Kabani C, Vige A, Gross MS, Boileau C, Rabes JP, Fruchart-Najib J, Jais JP, Junien C. Resistance to high-fat diet in the female progeny of obese mice fed a control diet during the periconceptual, gestation, and lactation periods. Am J Physiol Endocrinol Metab. 2007;292(4):E1095–E1100. doi: 10.1152/ajpendo.00390.2006. [DOI] [PubMed] [Google Scholar]
  • 41.Payette C, Blackburn P, Lamarche B, Tremblay A, Bergeron J, Lemieux I, Despres JP, Couillard C. Sex differences in postprandial plasma tumor necrosis factor-alpha, interleukin-6, and C-reactive protein concentrations. Metabolism. 2009 doi: 10.1016/j.metabol.2009.05.011. [DOI] [PubMed] [Google Scholar]
  • 42.Ghisletti S, Meda C, Maggi A, Vegeto E. 17beta-estradiol inhibits inflammatory gene expression by controlling NF-kappaB intracellular localization. Molecular and cellular biology. 2005;25(8):2957–2968. doi: 10.1128/MCB.25.8.2957-2968.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Turgeon JL, Carr MC, Maki PM, Mendelsohn ME, Wise PM. Complex actions of sex steroids in adipose tissue, the cardiovascular system, and brain: Insights from basic science and clinical studies. Endocrine reviews. 2006;27(6):575–605. doi: 10.1210/er.2005-0020. [DOI] [PubMed] [Google Scholar]
  • 44.Vegeto E, Belcredito S, Etteri S, Ghisletti S, Brusadelli A, Meda C, Krust A, Dupont S, Ciana P, Chambon P, Maggi A. Estrogen receptor-alpha mediates the brain antiinflammatory activity of estradiol. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(16):9614–9619. doi: 10.1073/pnas.1531957100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Vegeto E, Bonincontro C, Pollio G, Sala A, Viappiani S, Nardi F, Brusadelli A, Viviani B, Ciana P, Maggi A. Estrogen prevents the lipopolysaccharide-induced inflammatory response in microglia. J Neurosci. 2001;21(6):1809–1818. doi: 10.1523/JNEUROSCI.21-06-01809.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Stein B, Yang MX. Repression of the interleukin-6 promoter by estrogen receptor is mediated by NF-kappa B and C/EBP beta. Molecular and cellular biology. 1995;15(9):4971–4979. doi: 10.1128/mcb.15.9.4971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kalaitzidis D, Gilmore TD. Transcription factor cross-talk: the estrogen receptor and NF-kappaB. Trends in endocrinology and metabolism: TEM. 2005;16(2):46–52. doi: 10.1016/j.tem.2005.01.004. [DOI] [PubMed] [Google Scholar]
  • 48.Chadwick CC, Chippari C, Matelan E, Borges-Marcucci L, Eckert AM, Keith JC, Jr, Albert LM, Leathurby Y, Harris HA, Bhat RA, Ashwell M, Trybulski E, Winneker RC, Adelman SJ, Steffan RJ, Harnish DC. Identification of pathway-selective estrogen receptor ligands that inhibit NF-kappaB transcriptional activity. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(7):2543–2548. doi: 10.1073/pnas.0405841102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Harris HA, Albert LM, Leathurby Y, Malamas MS, Mewshaw RE, Miller CP, Kharode YP, Marzolf J, Komm BS, Winneker RC, Frail DE, Henderson RA, Zhu Y, Keith JC., Jr Evaluation of an estrogen receptor-beta agonist in animal models of human disease. Endocrinology. 2003;144(10):4241–4249. doi: 10.1210/en.2003-0550. [DOI] [PubMed] [Google Scholar]
  • 50.Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. The Journal of clinical investigation. 2006;116(11):3015–3025. doi: 10.1172/JCI28898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wise PM. Estrogens and neuroprotection. Trends in endocrinology and metabolism: TEM. 2002;13(6):229–230. doi: 10.1016/s1043-2760(02)00611-2. [DOI] [PubMed] [Google Scholar]
  • 52.Maggi A, Ciana P, Belcredito S, Vegeto E. Estrogens in the nervous system: mechanisms and nonreproductive functions. Annual review of physiology. 2004;66:291–313. doi: 10.1146/annurev.physiol.66.032802.154945. [DOI] [PubMed] [Google Scholar]
  • 53.Pozzi S, Benedusi V, Maggi A, Vegeto E. Estrogen action in neuroprotection and brain inflammation. Annals of the New York Academy of Sciences. 2006;1089:302–323. doi: 10.1196/annals.1386.035. [DOI] [PubMed] [Google Scholar]
  • 54.Vegeto E, Belcredito S, Ghisletti S, Meda C, Etteri S, Maggi A. The endogenous estrogen status regulates microglia reactivity in animal models of neuroinflammation. Endocrinology. 2006;147(5):2263–2272. doi: 10.1210/en.2005-1330. [DOI] [PubMed] [Google Scholar]
  • 55.Clark JT, Kalra PS, Crowley WR, Kalra SP. Neuropeptide Y and human pancreatic polypeptide stimulate feeding behavior in rats. Endocrinology. 1984;115(1):427–429. doi: 10.1210/endo-115-1-427. [DOI] [PubMed] [Google Scholar]
  • 56.Stanley BG, Kyrkouli SE, Lampert S, Leibowitz SF. Neuropeptide Y chronically injected into the hypothalamus: A powerful neurochemical inducer of hyperphagia and obesity. Peptides. 1986;7:1189–1192. doi: 10.1016/0196-9781(86)90149-x. [DOI] [PubMed] [Google Scholar]
  • 57.Kalra SP, Dube MG, Pu S, Xu B, Horvath TL, Kalra PS. Interacting appetite-regulating pathways in the hypothalamic regulation of body weight. Endocrine reviews. 1999;20(1):68–100. doi: 10.1210/edrv.20.1.0357. [DOI] [PubMed] [Google Scholar]
  • 58.Wade GN, Gray JM, Bartness TJ. Gonadal influences on adiposity. Int J Obes. 1985;9 Suppl 1:83–92. [PubMed] [Google Scholar]
  • 59.Bonavera JJ, Dube MG, Kalra PS, Kalra SP. Anorectic effects of estrogen may be mediated by decreased neuropeptide-Y release in the hypothalamic paraventricular nucleus. Endocrinology. 1994;134(6):2367–2370. doi: 10.1210/endo.134.6.8194462. [DOI] [PubMed] [Google Scholar]
  • 60.Sar M, Sahu A, Crowley WR, Kalra SP. Localization of neuropeptide-Y immunoreactivity in estradiol-concentrating cells in the hypothalamus. Endocrinology. 1990;127(6):2752–2756. doi: 10.1210/endo-127-6-2752. [DOI] [PubMed] [Google Scholar]
  • 61.Titolo D, Cai F, Belsham DD. Coordinate regulation of neuropeptide Y and agouti-related peptide gene expression by estrogen depends on the ratio of estrogen receptor (ER) alpha to ERbeta in clonal hypothalamic neurons. Molecular endocrinology (Baltimore, Md. 2006;20(9):2080–2092. doi: 10.1210/me.2006-0027. [DOI] [PubMed] [Google Scholar]
  • 62.Clegg DJ, Benoit SC, Fisher ME, Barrera JG, Seeley RJ, Woods SC. Sex hormones determine body fat distribution and sensitivity to adiposity signals. Appetite. 2003;40:324. [Google Scholar]
  • 63.Ainslie DA, Morris MJ, Wittert G, Turnbull H, Proietto J, Thorburn AW. Estrogen deficiency causes central leptin insensitivity and increased hypothalamic neuropeptide Y. Int J Obes Relat Metab Disord. 2001;25(11):1680–1688. doi: 10.1038/sj.ijo.0801806. [DOI] [PubMed] [Google Scholar]
  • 64.Akabayashi A, Watanabe Y, Wahlestedt C, McEwen BS, Paez X, Leibowitz SF. Hypothalamic neuropeptide Y, its gene expression and receptor activity: relation to circulating corticosterone in adrenalectomized rats. Brain research. 1994;665(2):201–212. doi: 10.1016/0006-8993(94)91339-0. [DOI] [PubMed] [Google Scholar]
  • 65.Pelleymounter MA, Baker MB, McCaleb M. Does estradiol mediate leptin's effects on adiposity and body weight? Am J Physiol. 1999;276(5 Pt 1):E955–E963. doi: 10.1152/ajpendo.1999.276.5.E955. [DOI] [PubMed] [Google Scholar]
  • 66.Acosta-Martinez M, Horton T, Levine JE. Estrogen receptors in neuropeptide Y neurons: at the crossroads of feeding and reproduction. Trends in endocrinology and metabolism: TEM. 2007;18(2):48–50. doi: 10.1016/j.tem.2006.12.001. [DOI] [PubMed] [Google Scholar]
  • 67.Bauer-Dantoin AC, Urban JH, Levine JE. Neuropeptide Y gene expression in the arcuate nucleus is increased during preovulatory luteinizing hormone surges. Endocrinology. 1992;131(6):2953–2958. doi: 10.1210/endo.131.6.1446633. [DOI] [PubMed] [Google Scholar]
  • 68.Hill JW, Urban JH, Xu M, Levine JE. Estrogen Induces Neuropeptide Y (NPY) Y1 receptor gene expression and responsiveness to NPY in gonadotrope-enriched pituitary cell cultures. Endocrinology. 2004;145(5):2283–2290. doi: 10.1210/en.2003-1368. [DOI] [PubMed] [Google Scholar]
  • 69.Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402(6762):656–660. doi: 10.1038/45230. [DOI] [PubMed] [Google Scholar]
  • 70.Howard AD, Feighner SD, Cully DF, Arena JP, Liberator PA, Rosenblum CI, Hamelin M, Hreniuk DL, Palyha OC, Anderson J, Paress PS, Diaz C, Chou M, Liu KK, McKee KK, Pong SS, Chaung LY, Elbrecht A, Dashkevicz M, Heavens R, Rigby M, Sirinathsinghji DJ, Dean DC, Melillo DG, Patchett AA, Nargund R, Griffin PR, DeMartino JA, Gupta SK, Schaeffer JM, Smith RG, Van der Ploeg LH. A receptor in pituitary and hypothalamus that functions in growth hormone release. Science (New York, N.Y. 1996;273(5277):974–977. doi: 10.1126/science.273.5277.974. [DOI] [PubMed] [Google Scholar]
  • 71.Kojima M, Kangawa K. Ghrelin: structure and function. Physiol Rev. 2005;85(2):495–522. doi: 10.1152/physrev.00012.2004. [DOI] [PubMed] [Google Scholar]
  • 72.Clegg DJ, Brown LM, Zigman JM, Kemp CJ, Strader AD, Benoit SC, Woods SC, Mangiaracina M, Geary N. Estradiol-dependent decrease in the orexigenic potency of ghrelin in female rats. Diabetes. 2007;56(4):1051–1058. doi: 10.2337/db06-0015. [DOI] [PubMed] [Google Scholar]
  • 73.Nakazato M, Murakami N, Date Y, Kojima M, Matsuo H, Kangawa K, Matsukura S. A role for ghrelin in the central regulation of feeding. Nature. 2001;409(6817):194–198. doi: 10.1038/35051587. [DOI] [PubMed] [Google Scholar]
  • 74.Tschop M, Smiley DL, Heiman ML. Ghrelin induces adiposity in rodents. Nature. 2000;407(6806):908–913. doi: 10.1038/35038090. [DOI] [PubMed] [Google Scholar]
  • 75.Wren AM, Small CJ, Abbott CR, Dhillo WS, Seal LJ, Cohen MA, Batterham RL, Taheri S, Stanley SA, Ghatei MA, Bloom SR. Ghrelin causes hyperphagia and obesity in rats. Diabetes. 2001;50(11):2540–2547. doi: 10.2337/diabetes.50.11.2540. [DOI] [PubMed] [Google Scholar]
  • 76.Horvath TL, Diano S, Sotonyi P, Heiman M, Tschop M. Minireview: ghrelin and the regulation of energy balance--a hypothalamic perspective. Endocrinology. 2001;142(10):4163–4169. doi: 10.1210/endo.142.10.8490. [DOI] [PubMed] [Google Scholar]
  • 77.Wren AM, Seal LJ, Cohen MA, Brynes AE, Frost GS, Murphy KG, Dhillo WS, Ghatei MA, Bloom SR. Ghrelin enhances appetite and increases food intake in humans. The Journal of clinical endocrinology and metabolism. 2001;86(12):5992. doi: 10.1210/jcem.86.12.8111. [DOI] [PubMed] [Google Scholar]
  • 78.Arnold M, Mura A, Langhans W, Geary N. Gut vagal afferents are not necessary for the eating-stimulatory effect of intraperitoneally injected ghrelin in the rat. J Neurosci. 2006;26(43):11052–11060. doi: 10.1523/JNEUROSCI.2606-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Davidson TL, Kanoski SE, Tracy AL, Walls EK, Clegg D, Benoit SC. The interoceptive cue properties of ghrelin generalize to cues produced by food deprivation. Peptides. 2005;26(9):1602–1610. doi: 10.1016/j.peptides.2005.02.014. [DOI] [PubMed] [Google Scholar]
  • 80.Zigman JM, Nakano Y, Coppari R, Balthasar N, Marcus JN, Lee CE, Jones JE, Deysher AE, Waxman AR, White RD, Williams TD, Lachey JL, Seeley RJ, Lowell BB, Elmquist JK. Mice lacking ghrelin receptors resist the development of diet-induced obesity. The Journal of clinical investigation. 2005;115(12):3564–3572. doi: 10.1172/JCI26002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kamegai J, Tamura H, Shimizu T, Ishii S, Sugihara H, Wakabayashi I. Central effect of ghrelin, an endogenous growth hormone secretagogue, on hypothalamic peptide gene expression. Endocrinology. 2000;141(12):4797–4800. doi: 10.1210/endo.141.12.7920. [DOI] [PubMed] [Google Scholar]
  • 82.Kamegai J, Tamura H, Shimizu T, Ishii S, Sugihara H, Wakabayashi I. Chronic central infusion of ghrelin increases hypothalamic neuropeptide Y and Agouti-related protein mRNA levels and body weight in rats. Diabetes. 2001;50(11):2438–2443. doi: 10.2337/diabetes.50.11.2438. [DOI] [PubMed] [Google Scholar]
  • 83.Shintani M, Ogawa Y, Ebihara K, Aizawa-Abe M, Miyanaga F, Takaya K, Hayashi T, Inoue G, Hosoda K, Kojima M, Kangawa K, Nakao K. Ghrelin, an endogenous growth hormone secretagogue, is a novel orexigenic peptide that antagonizes leptin action through the activation of hypothalamic neuropeptide Y/Y1 receptor pathway. Diabetes. 2001;50(2):227–232. doi: 10.2337/diabetes.50.2.227. [DOI] [PubMed] [Google Scholar]
  • 84.Horvath TL, Diano S, Tschop M. Ghrelin in hypothalamic regulation of energy balance. Curr Top Med Chem. 2003;3(8):921–927. doi: 10.2174/1568026033452230. [DOI] [PubMed] [Google Scholar]
  • 85.Naleid AM, Grace MK, Cummings DE, Levine AS. Ghrelin induces feeding in the mesolimbic reward pathway between the ventral tegmental area and the nucleus accumbens. Peptides. 2005;26(11):2274–2279. doi: 10.1016/j.peptides.2005.04.025. [DOI] [PubMed] [Google Scholar]
  • 86.Willesen MG, Kristensen P, Romer J. Co-localization of growth hormone secretagogue receptor and NPY mRNA in the arcuate nucleus of the rat. Neuroendocrinology. 1999;70(5):306–316. doi: 10.1159/000054491. [DOI] [PubMed] [Google Scholar]
  • 87.Faulconbridge LF, Cummings DE, Kaplan JM, Grill HJ. Hyperphagic effects of brainstem ghrelin administration. Diabetes. 2003;52(9):2260–2265. doi: 10.2337/diabetes.52.9.2260. [DOI] [PubMed] [Google Scholar]
  • 88.Cowley MA, Smith RG, Diano S, Tschop M, Pronchuk N, Grove KL, Strasburger CJ, Bidlingmaier M, Esterman M, Heiman ML, Garcia-Segura LM, Nillni EA, Mendez P, Low MJ, Sotonyi P, Friedman JM, Liu H, Pinto S, Colmers WF, Cone RD, Horvath TL. The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron. 2003;37(4):649–661. doi: 10.1016/s0896-6273(03)00063-1. [DOI] [PubMed] [Google Scholar]
  • 89.Currie PJ, Mirza A, Fuld R, Park D, Vasselli JR. Ghrelin is an orexigenic and metabolic signaling peptide in the arcuate and paraventricular nuclei. Am J Physiol Regul Integr Comp Physiol. 2005;289(2):R353–R358. doi: 10.1152/ajpregu.00756.2004. [DOI] [PubMed] [Google Scholar]
  • 90.Thammacharoen S, Lutz TA, Geary N, Asarian L. Hindbrain administration of estradiol inhibits feeding and activates estrogen receptor-alpha-expressing cells in the nucleus tractus solitarius of ovariectomized rats. Endocrinology. 2008;149(4):1609–1617. doi: 10.1210/en.2007-0340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Bittencourt JC, Presse F, Arias C, Peto C, Vaughan J, Nahon JL, Vale W, Sawchenko PE. The melanin-concentrating hormone system of the rat brain: an immuno- and hybridization histochemical characterization. The Journal of comparative neurology. 1992;319(2):218–245. doi: 10.1002/cne.903190204. [DOI] [PubMed] [Google Scholar]
  • 92.Vaughan JM, Fischer WH, Hoeger C, Rivier J, Vale W. Characterization of melanin-concentrating hormone from rat hypothalamus. Endocrinology. 1989;125(3):1660–1665. doi: 10.1210/endo-125-3-1660. [DOI] [PubMed] [Google Scholar]
  • 93.Pissios P, Bradley RL, Maratos-Flier E. Expanding the scales: The multiple roles of MCH in regulating energy balance and other biological functions. Endocrine reviews. 2006;27(6):606–620. doi: 10.1210/er.2006-0021. [DOI] [PubMed] [Google Scholar]
  • 94.Qu D, Ludwig DS, Gammeltoft S, Piper M, Pelleymounter MA, Cullen MJ, Mathes WF, Przypek R, Kanarek R, Maratos-Flier E. A role for melanin-concentrating hormone in the central regulation of feeding behaviour. Nature. 1996;380(6571):243–247. doi: 10.1038/380243a0. [DOI] [PubMed] [Google Scholar]
  • 95.Rossi M, Choi SJ, O'Shea D, Miyoshi T, Ghatei MA, Bloom SR. Melanin-concentrating hormone acutely stimulates feeding, but chronic administration has no effect on body weight. Endocrinology. 1997;138(1):351–355. doi: 10.1210/endo.138.1.4887. [DOI] [PubMed] [Google Scholar]
  • 96.Shimada M, Tritos N, Lowell B, Flier J, Maratos-Flier E. Mice lacking melanin-concentrating hormone are hypophagic and lean. Nature. 1998 Dec 17;396:670–674. doi: 10.1038/25341. [DOI] [PubMed] [Google Scholar]
  • 97.Alon T, Friedman JM. Late-onset leanness in mice with targeted ablation of melanin concentrating hormone neurons. J Neurosci. 2006;26(2):389–397. doi: 10.1523/JNEUROSCI.1203-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Broberger C, De Lecea L, Sutcliffe JG, Hokfelt T. Hypocretin/orexin- and melanin-concentrating hormone-expressing cells form distinct populations in the rodent lateral hypothalamus: relationship to the neuropeptide Y and agouti gene-related protein systems. Journal of Comparative Neurology. 1998;402:460–474. [PubMed] [Google Scholar]
  • 99.Elias CF, Aschkenasi C, Lee C, Kelly J, Ahima RS, Bjorbaek C, Flier JS, Saper CB, Elmquist JK. Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area. Neuron. 1999;23(4):775–786. doi: 10.1016/s0896-6273(01)80035-0. [DOI] [PubMed] [Google Scholar]
  • 100.Elias CF, Saper CB, Maratos-Flier E, Tritos NA, Lee C, Kelly J, Tatro JB, Hoffman GE, Ollmann MM, Barsh GS, Sakurai T, Yanagisawa M, Elmquist JK. Chemically defined projections linking the mediobasal hypothalamus and the lateral hypothalamic area. The Journal of comparative neurology. 1998;402(4):442–459. [PubMed] [Google Scholar]
  • 101.Flier JS. Obesity wars: molecular progress confronts an expanding epidemic. Cell. 2004;116:337–350. doi: 10.1016/s0092-8674(03)01081-x. [DOI] [PubMed] [Google Scholar]
  • 102.Schwartz MW, Woods SC, Porte DJ, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000;404:661–671. doi: 10.1038/35007534. [DOI] [PubMed] [Google Scholar]
  • 103.Zigman JM, Elmquist JK. Minireview: From anorexia to obesity--the yin and yang of body weight control. Endocrinology. 2003;144(9):3749–3756. doi: 10.1210/en.2003-0241. [DOI] [PubMed] [Google Scholar]
  • 104.Georgescu D, Sears RM, Hommel JD, Barrot M, Bolanos CA, Marsh DJ, Bednarek MA, Bibb JA, Maratos-Flier E, Nestler EJ, DiLeone RJ. The hypothalamic neuropeptide melanin-concentrating hormone acts in the nucleus accumbens to modulate feeding behavior and forced-swim performance. J Neurosci. 2005;25(11):2933–2940. doi: 10.1523/JNEUROSCI.1714-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Shughrue PJ, Lane MV, Merchenthaler I. Comparative distribution of estrogen receptor-alpha and -beta mRNA in the rat central nervous system. The Journal of comparative neurology. 1997;388(4):507–525. doi: 10.1002/(sici)1096-9861(19971201)388:4<507::aid-cne1>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
  • 106.Murray JF, Baker BI, Levy A, Wilson CA. The influence of gonadal steroids on pre-pro melanin-concentrating hormone mRNA in female rats. Journal of neuroendocrinology. 2000;12(1):53–59. doi: 10.1046/j.1365-2826.2000.00425.x. [DOI] [PubMed] [Google Scholar]
  • 107.Morton GJ, Mystkowski P, Matsumoto AM, Schwartz MW. Increased hypothalamic melanin concentrating hormone gene expression during energy restriction involves a melanocortin-independent, estrogen-sensitive mechanism. Peptides. 2004;25(4):667–674. doi: 10.1016/j.peptides.2004.02.007. [DOI] [PubMed] [Google Scholar]
  • 108.Tritos NA, Segal-Lieberman G, Vezeridis PS, Maratos-Flier E. Estradiol-induced anorexia is independent of leptin and melanin-concentrating hormone. Obes Res. 2004;12(4):716–724. doi: 10.1038/oby.2004.84. [DOI] [PubMed] [Google Scholar]
  • 109.Hervieu GJ, Cluderay JE, Harrison D, Meakin J, Maycox P, Nasir S, Leslie RA. The distribution of the mRNA and protein products of the melanin-concentrating hormone (MCH) receptor gene, slc-1, in the central nervous system of the rat. Eur J Neurosci. 2000;12(4):1194–1216. doi: 10.1046/j.1460-9568.2000.00008.x. [DOI] [PubMed] [Google Scholar]
  • 110.Muschamp JW, Hull EM. Melanin concentrating hormone and estrogen receptor-alpha are coexstensive but not coexpressed in cells of male rat hypothalamus. Neurosci Lett. 2007;427(3):123–126. doi: 10.1016/j.neulet.2007.09.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Smith GP. The controls of eating: a shift from nutritional homeostasis to behavioral neuroscience. Nutrition. 2000;16:814–820. doi: 10.1016/s0899-9007(00)00457-3. [DOI] [PubMed] [Google Scholar]
  • 112.Geary N, Asarian L, Korach KS, Pfaff DW, Ogawa S. Deficits in E2-dependent control of feeding, weight gain, and cholecystokinin satiation in ER-alpha null mice. Endocrinology. 2001;142(11):4751–4757. doi: 10.1210/endo.142.11.8504. [DOI] [PubMed] [Google Scholar]
  • 113.Kennedy GC. The role of depot fat in the hypothalamic control of food intake in the rat. Proc R Soc Lond (Biol) 1953;140:579–592. doi: 10.1098/rspb.1953.0009. [DOI] [PubMed] [Google Scholar]
  • 114.Baskin DG, Figlewicz DP, Woods SC, Porte D, Jr, Dorsa DM. Insulin in the brain. Ann Rev Physiol. 1987;49:335–347. doi: 10.1146/annurev.ph.49.030187.002003. [DOI] [PubMed] [Google Scholar]
  • 115.Woods SC. Insulin and the brain: A mutual dependency. Progress in Psychobiology and Physiological Psychology. 1996;16:53–81. [Google Scholar]
  • 116.Bagdade JD, Bierman EL, Porte D., Jr The significance of basal insulin levels in the evaluation of the insulin response to glucose in diabetic and nondiabetic subjects. The Journal of clinical investigation. 1967;46:1549–1557. doi: 10.1172/JCI105646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Polonsky KS, Given BD, Hirsch L, Shapiro ET, Tillil H, Beebe C, Galloway JA, Frank BH, Karrison T, Van-Cauter E. Quantitative study of insulin secretion and clearance in normal and obese subjects. The Journal of clinical investigation. 1988;81:435–441. doi: 10.1172/JCI113338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Polonsky KS, Given E, Carter V. Twenty-four-hour profiles and pulsatile patterns of insulin secretion in normal and obese subjects. Journal of Clinical Investigation. 1988;81:442–448. doi: 10.1172/JCI113339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Woods SC, Decke E, Vasselli JR. Metabolic hormones and regulation of body weight. Psychol Rev. 1974;81(1):26–43. doi: 10.1037/h0035927. [DOI] [PubMed] [Google Scholar]
  • 120.Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Allen AM, Mendelsohn FA. Localization and characterization of insulin receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry. Endocrinology. 1987;121(4):1562–1570. doi: 10.1210/endo-121-4-1562. [DOI] [PubMed] [Google Scholar]
  • 121.Folli F, Saad MJ, Kahn CR. Insulin receptor/IRS-1/PI 3-kinase signaling system in corticosteroid-induced insulin resistance. Acta Diabetol. 1996;33(3):185–192. doi: 10.1007/BF02048541. [DOI] [PubMed] [Google Scholar]
  • 122.Hill JM, Lesniak MA, Pert CB, Roth J. Autoradiographic localization of insulin receptors in rat brain: prominence in olfactory and limbic areas. Neuroscience. 1986;17:1127–1138. doi: 10.1016/0306-4522(86)90082-5. [DOI] [PubMed] [Google Scholar]
  • 123.Schwartz MW, Porte D., Jr Diabetes, obesity, and the brain. Science (New York, N.Y. 2005;307(5708):375–379. doi: 10.1126/science.1104344. [DOI] [PubMed] [Google Scholar]
  • 124.Niswender KD, Morrison CD, Clegg DJ, Olson R, Baskin DG, Myers MG, Jr, Seeley RJ, Schwartz MW. Insulin activation of phosphatidylinositol 3-kinase in the hypothalamic arcuate nucleus: a key mediator of insulin-induced anorexia. Diabetes. 2003;52(2):227–231. doi: 10.2337/diabetes.52.2.227. [DOI] [PubMed] [Google Scholar]
  • 125.Niswender KD, Schwartz MW. Insulin and leptin revisited: adiposity signals with overlapping physiological and intracellular signaling capabilities. Frontiers in Neuroendocrinology. 2003;24:1–10. doi: 10.1016/s0091-3022(02)00105-x. [DOI] [PubMed] [Google Scholar]
  • 126.Benoit SC, Air EL, Coolen LM, Strauss R, Jackman A, Clegg DJ, Seeley RJ, Woods SC. The catabolic action of insulin in the brain is mediated by melanocortins. Journal of Neuroscience. 2002;22(20):9048–9052. doi: 10.1523/JNEUROSCI.22-20-09048.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372(6505):425–432. doi: 10.1038/372425a0. [DOI] [PubMed] [Google Scholar]
  • 128.Banks WA, Kastin AJ, Huang Wea. Leptin enters the brain by a saturable system independent of insulin. Peptides. 1996;17:305–311. doi: 10.1016/0196-9781(96)00025-3. [DOI] [PubMed] [Google Scholar]
  • 129.Schwartz MW, Seeley RJ, Campfield LA, Burn P, Baskin DG. Identification of hypothalmic targets of leptin action. Journal of Clinical Investigation. 1996;98:1101–1106. doi: 10.1172/JCI118891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Clegg DJ, Riedy CA, Smith KA, Benoit SC, Woods SC. Differential sensitivity to central leptin and insulin in male and female rats. Diabetes. 2003;52(3):682–687. doi: 10.2337/diabetes.52.3.682. [DOI] [PubMed] [Google Scholar]
  • 131.Ahima RS, Kelly J, Elmquist JK, Flier JS. Distinct physiologic and neuronal responses to decreased leptin and mild hyperleptinemia. Endocrinology. 1999;140(11):4923–4931. doi: 10.1210/endo.140.11.7105. [DOI] [PubMed] [Google Scholar]
  • 132.Seeley RJ, Woods SC. Monitoring of stored and av ailable fuel by the CNS: implications for obesity. Nat Rev Neurosci. 2003;4(11):901–909. doi: 10.1038/nrn1245. [DOI] [PubMed] [Google Scholar]
  • 133.Elmquist JK, Elias CF, Saper CB. From lesions to leptin: Hypothalamic control of food intake and body weight. Neuron. 1999;22:221–232. doi: 10.1016/s0896-6273(00)81084-3. [DOI] [PubMed] [Google Scholar]
  • 134.Morton GJ, Niswender KD, Rhodes CJ, Myers MG, Jr, Blevins JE, Baskin DG, Schwartz MW. Arcuate nucleus-specific leptin receptor gene therapy attenuates the obesity phenotype of Koletsky (fa(k)/fa(k)) rats. Endocrinology. 2003;144(5):2016–2024. doi: 10.1210/en.2002-0115. [DOI] [PubMed] [Google Scholar]
  • 135.Woods SC, Schwartz MW, Baskin DG, Seeley RJ. Food intake and the regulation of body weight. Annual Review of Psychology. 2000;51:255–277. doi: 10.1146/annurev.psych.51.1.255. [DOI] [PubMed] [Google Scholar]
  • 136.Balthasar N, Coppari R, McMinn J, Liu SM, Lee CE, Tang V, Kenny CD, McGovern RA, Chua SC, Jr, Elmquist JK, Lowell BB. Leptin receptor signaling in POMC neurons is required for normal body weight homeostasis. Neuron. 2004;42(6):983–991. doi: 10.1016/j.neuron.2004.06.004. [DOI] [PubMed] [Google Scholar]
  • 137.Demerath EW, Towne B, Wisemandle W, Blangero J, Chumlea WC, Siervogel RM. Serum leptin concentration, body composition, and gonadal hormones during puberty. Int J Obes Relat Metab Disord. 1999;23(7):678–685. doi: 10.1038/sj.ijo.0800902. [DOI] [PubMed] [Google Scholar]
  • 138.Shimizu H, Shimomura Y, Nakanishi Y, Futawatari T, Ohtani K, Sato N, Mori M. Estrogen increases in vivo leptin production in rats and human subjects. J Endocrinol. 1997;154(2):285–292. doi: 10.1677/joe.0.1540285. [DOI] [PubMed] [Google Scholar]
  • 139.Wu-Peng S, Rosenbaum M, Nicolson M, Chua SC, Leibel RL. Effects of exogenous gonadal steroids on leptin homeostasis in rats. Obes Res. 1999;7(6):586–592. doi: 10.1002/j.1550-8528.1999.tb00718.x. [DOI] [PubMed] [Google Scholar]
  • 140.Machinal F, Dieudonne MN, Leneveu MC, Pecquery R, Giudicelli Y. In vivo and in vitro ob gene expression and leptin secretion in rat adipocytes: evidence for a regional specific regulation by sex steroid hormones. Endocrinology. 1999;140(4):1567–1574. doi: 10.1210/endo.140.4.6617. [DOI] [PubMed] [Google Scholar]
  • 141.Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Flier-Maratos E, Flier JS. Role of leptin in the neuroendocrine response to fasting. Nature. 1996;382:250–252. doi: 10.1038/382250a0. [DOI] [PubMed] [Google Scholar]
  • 142.Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R, Richards GJ, Campfield LA, Clark FT, Deeds J, Muir C, Sanker S, Moriarty A, Moore KJ, Smutko JS, Mays GG, Woolf EA, Monroe CA, Tepper RI. Identification and expression cloning of a leptin receptor, OB-R. Cell. 1995;83:1263–1271. doi: 10.1016/0092-8674(95)90151-5. [DOI] [PubMed] [Google Scholar]
  • 143.Seeley RJ, van Dijk G, Campfield LA, Smith FJ, Burn P, Nelligan JA, Bell SM, Baskin DG, Woods SC, Schwartz MW. Intraventricular leptin reduces food intake and body weight of lean rats but not obese Zucker rats. Horm Metab Res. 1996;28(12):664–668. doi: 10.1055/s-2007-979874. [DOI] [PubMed] [Google Scholar]
  • 144.Chen H, Charlat O, Tartaglia LA, Woolf EA, Weng X, Ellis SJ, Lakey ND, Culpepper J, Moore KJ, Breitbart RE, Duyk GM, Tepper RI, Morgenstern JP. Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell. 1996;84(3):491–495. doi: 10.1016/s0092-8674(00)81294-5. [DOI] [PubMed] [Google Scholar]
  • 145.Mitra SW, Hoskin E, Yudkovitz J, Pear L, Wilkinson HA, Hayashi S, Pfaff DW, Ogawa S, Rohrer SP, Schaeffer JM, McEwen BS, Alves SE. Immunolocalization of estrogen receptor beta in the mouse brain: comparison with estrogen receptor alpha. Endocrinology. 2003;144(5):2055–2067. doi: 10.1210/en.2002-221069. [DOI] [PubMed] [Google Scholar]
  • 146.Diano S, Kalra SP, Horvath TL. Leptin receptor immunoreactivity is associated with the Golgi apparatus of hypothalamic neurons and glial cells. Journal of neuroendocrinology. 1998;10(9):647–650. doi: 10.1046/j.1365-2826.1998.00261.x. [DOI] [PubMed] [Google Scholar]
  • 147.Bennett PA, Lindell K, Wilson C, Carlsson LM, Carlsson B, Robinson IC. Cyclical variations in the abundance of leptin receptors, but not in circulating leptin, correlate with NPY expression during the oestrous cycle. Neuroendocrinology. 1999;69(6):417–423. doi: 10.1159/000054444. [DOI] [PubMed] [Google Scholar]
  • 148.Lindell K, Bennett PA, Itoh Y, Robinson IC, Carlsson LM, Carlsson B. Leptin receptor 5'untranslated regions in the rat: relative abundance, genomic organization and relation to putative response elements. Mol Cell Endocrinol. 2001;172(1–2):37–45. doi: 10.1016/s0303-7207(00)00382-8. [DOI] [PubMed] [Google Scholar]
  • 149.Clegg DJ, Benoit SC, Barrera JG, Woods SC. Estrogen Mediates Body Fat Distribution and Brain Sensitivity to Adiposity Signals. Diabetes. 2003;52 supplement 1 [Google Scholar]
  • 150.Elmquist JK, Ahima RS, Elias CF, Flier JS, Saper CB. Leptin activates distinct projections from the dorsomedial and ventromedial hypothalamic nuclei. Proceedings of the National Academy of Sciences USA. 1998;95:741–746. doi: 10.1073/pnas.95.2.741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Elmquist JK, Bjorbaek C, Ahima RS, Flier JS, Saper CB. Distributions of leptin receptor mRNA isoforms in the rat brain. The Journal of comparative neurology. 1998;395(4):535–547. [PubMed] [Google Scholar]
  • 152.Elmquist JK, Maratos-Flier E, Saper CB, Flier JS. Unraveling the central nervous system pathways underlying responses to leptin. Nature Neuroscience. 1998;1:445–450. doi: 10.1038/2164. [DOI] [PubMed] [Google Scholar]
  • 153.Bennett PA, Lindell K, Karlsson C, Robinson IC, Carlsson LM, Carlsson B. Differential expression and regulation of leptin receptor isoforms in the rat brain: effects of fasting and oestrogen. Neuroendocrinology. 1998;67(1):29–36. doi: 10.1159/000054295. [DOI] [PubMed] [Google Scholar]
  • 154.Kimura M, Irahara M, Yasui T, Saito S, Tezuka M, Yamano S, Kamada M, Aono T. The obesity in bilateral ovariectomized rats is related to a decrease in the expression of leptin receptors in the brain. Biochem Biophys Res Commun. 2002;290(4):1349–1353. doi: 10.1006/bbrc.2002.6355. [DOI] [PubMed] [Google Scholar]
  • 155.Simansky K. Serotonin and the structure of satiation. In: Smith G, editor. Satiation: From Gut to Brain. New York: Oxford University Press; 1998. pp. 217–262. [Google Scholar]
  • 156.Rivera HM, Eckel LA. The anorectic effect of fenfluramine is increased by estradiol treatment in ovariectomized rats. Physiol Behav. 2005;86(3):331–337. doi: 10.1016/j.physbeh.2005.08.004. [DOI] [PubMed] [Google Scholar]
  • 157.Salamanca S, Uphouse L. Estradiol modulation of the hyperphagia induced by the 5-HT1A agonist, 8-OH-DPAT. Pharmacol Biochem Behav. 1992;43(3):953–955. doi: 10.1016/0091-3057(92)90431-e. [DOI] [PubMed] [Google Scholar]
  • 158.Uphouse L, Salamanca S, Caldarola-Pastuszka M. Gender and estrous cycle differences in the response to the 5-HT1A agonist 8-OH-DPAT. Pharmacol Biochem Behav. 1991;40(4):901–906. doi: 10.1016/0091-3057(91)90104-a. [DOI] [PubMed] [Google Scholar]
  • 159.Rivera HM, Oberbeck DR, Kwon B, Houpt TA, Eckel LA. Estradiol increases Pet-1 and serotonin transporter mRNA in the midbrain raphe nuclei of ovariectomized rats. Brain research. 2009;1259:51–58. doi: 10.1016/j.brainres.2008.12.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.McQueen JK, Wilson H, Fink G. Estradiol-17 beta increases serotonin transporter (SERT) mRNA levels and the density of SERT-binding sites in female rat brain. Brain Res Mol Brain Res. 1997;45(1):13–23. doi: 10.1016/s0169-328x(96)00233-1. [DOI] [PubMed] [Google Scholar]
  • 161.Sumner BE, Grant KE, Rosie R, Hegele-Hartung C, Fritzemeier KH, Fink G. Effects of tamoxifen on serotonin transporter and 5-hydroxytryptamine(2A) receptor binding sites and mRNA levels in the brain of ovariectomized rats with or without acute estradiol replacement. Brain Res Mol Brain Res. 1999;73(1–2):119–128. doi: 10.1016/s0169-328x(99)00243-0. [DOI] [PubMed] [Google Scholar]
  • 162.Petrov T, Krukoff TL, Jhamandas JH. The hypothalamic paraventricular and lateral parabrachial nuclei receive collaterals from raphe nucleus neurons: a combined double retrograde and immunocytochemical study. The Journal of comparative neurology. 1992;318(1):18–26. doi: 10.1002/cne.903180103. [DOI] [PubMed] [Google Scholar]
  • 163.Peyron C, Petit JM, Rampon C, Jouvet M, Luppi PH. Forebrain afferents to the rat dorsal raphe nucleus demonstrated by retrograde and anterograde tracing methods. Neuroscience. 1998;82(2):443–468. doi: 10.1016/s0306-4522(97)00268-6. [DOI] [PubMed] [Google Scholar]
  • 164.Fumeron F, Betoulle D, Aubert R, Herbeth B, Siest G, Rigaud D. Association of a functional 5-HT transporter gene polymorphism with anorexia nervosa and food intake. Mol Psychiatry. 2001;6(1):9–10. doi: 10.1038/sj.mp.4000824. [DOI] [PubMed] [Google Scholar]
  • 165.Heils A, Teufel A, Petri S, Stober G, Riederer P, Bengel D, Lesch KP. Allelic variation of human serotonin transporter gene expression. Journal of neurochemistry. 1996;66(6):2621–2624. doi: 10.1046/j.1471-4159.1996.66062621.x. [DOI] [PubMed] [Google Scholar]
  • 166.Moran TH. Gut peptides in the control of food intake. Int J Obes (Lond) 2009;33 Suppl 1:S7–S10. doi: 10.1038/ijo.2009.9. [DOI] [PubMed] [Google Scholar]
  • 167.Cox JE. Cholecystokinin satiety involves CCKA receptors perfused by the superior pancreaticoduodenal artery. Am J Physiol. 1998;274(5 Pt 2):R1390–R1396. doi: 10.1152/ajpregu.1998.274.5.R1390. [DOI] [PubMed] [Google Scholar]
  • 168.Smith G, Gibbs J. The Satiating Effects of Cholecystokinin and Bombesin-Like Peptides. In: Smith G, editor. Satiation from Gut to Brain. New York: Oxford University Press; 1998. pp. 97–125. [Google Scholar]
  • 169.Beglinger C, Degen L, Matzinger D, D'Amato M, Drewe J. Loxiglumide, a CCK-A receptor antagonist, stimulates calorie intake and hunger feelings in humans. American Journal of Physiology. 2001;280:R1149–R1154. doi: 10.1152/ajpregu.2001.280.4.R1149. [DOI] [PubMed] [Google Scholar]
  • 170.Moran TH, Ameglio PJ, Peyton HJ, Schwartz GJ, McHugh PR. Blockade of type A, but not type B, CCK receptors postpones satiety in rhesus monkeys. American Journal of Physiology. 1993;265:R620–R624. doi: 10.1152/ajpregu.1993.265.3.R620. [DOI] [PubMed] [Google Scholar]
  • 171.Huang YS, Doi R, Chowdhury P, Pasley JN, Nishikawa M, Huang TJ, Rayford PL. Effect of cholecystokinin on food intake at different stages of the estrous cycle in female rats. J Assoc Acad Minor Phys. 1993;4(2):56–58. [PubMed] [Google Scholar]
  • 172.Asarian L, Geary N. Cyclic estradiol treatment phasically potentiates endogenous cholecystokinin's satiating action in ovariectomized rats. Peptides. 1999;20(4):445–450. doi: 10.1016/s0196-9781(99)00024-8. [DOI] [PubMed] [Google Scholar]
  • 173.Asarian L, Geary N. Estradiol enhances cholecystokinin-dependent lipid-induced satiation and activates estrogen receptor-alpha-expressing cells in the nucleus tractus solitarius of ovariectomized rats. Endocrinology. 2007;148(12):5656–5666. doi: 10.1210/en.2007-0341. [DOI] [PubMed] [Google Scholar]
  • 174.Eckel LA, Geary N. Endogenous cholecystokinin's satiating action increases during estrus in female rats. Peptides. 1999;20(4):451–456. doi: 10.1016/s0196-9781(99)00025-x. [DOI] [PubMed] [Google Scholar]
  • 175.Moran TH, Ameglio PJ, Schwartz GJ, McHugh PR. Blockade of type A, not type B, CCK receptors attenuates satiety actions of exogenous and endogenous CCK. Am J Physiol. 1992;262(1 Pt 2):R46–R50. doi: 10.1152/ajpregu.1992.262.1.R46. [DOI] [PubMed] [Google Scholar]
  • 176.Reidelberger RD. Abdominal vagal mediation of the satiety effects of exogenous and endogenous cholecystokinin in rats. Am J Physiol. 1992;263(6 Pt 2):R1354–R1358. doi: 10.1152/ajpregu.1992.263.6.R1354. [DOI] [PubMed] [Google Scholar]
  • 177.Moran TH, Norgren R, Crosby RJ, McHugh PR. Central and peripheral vagal transport of cholecystokinin binding sites occurs in afferent fibers. Brain research. 1990;526(1):95–102. doi: 10.1016/0006-8993(90)90253-8. [DOI] [PubMed] [Google Scholar]
  • 178.Geary N, Smith GP, Corp ES. The increased satiating potency of CCK-8 by estradiol is not mediated by upregulation of NTS CCK receptors. Brain research. 1996;719(1–2):179–186. doi: 10.1016/0006-8993(96)00099-6. [DOI] [PubMed] [Google Scholar]
  • 179.Linden A, Uvnas-Moberg K, Forsberg G, Bednar I, Sodersten P. Involvement of Cholecystokinin in Food Intake: III. Oestradiol Potentiates the Inhibitory Effect of Cholecystokinin Octapeptide on Food Intake in Ovariectomized Rats. Journal of neuroendocrinology. 1990;2(6):797–801. doi: 10.1111/j.1365-2826.1990.tb00643.x. [DOI] [PubMed] [Google Scholar]
  • 180.Butera PC, Bradway DM, Cataldo NJ. Modulation of the satiety effect of cholecystokinin by estradiol. Physiol Behav. 1993;53(6):1235–1238. doi: 10.1016/0031-9384(93)90387-u. [DOI] [PubMed] [Google Scholar]
  • 181.Geary N, Asarian L. Cyclic estradiol treatment normalizes body weight and test meal size in ovariectomized rats. Physiol Behav. 1999;67(1):141–147. doi: 10.1016/s0031-9384(99)00060-8. [DOI] [PubMed] [Google Scholar]
  • 182.DiNardo LA, Travers JB. Distribution of fos-like immunoreactivity in the medullary reticular formation of the rat after gustatory elicited ingestion and rejection behaviors. J Neurosci. 1997;17(10):3826–3839. doi: 10.1523/JNEUROSCI.17-10-03826.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Eckel LA, Geary N. Estradiol treatment increases feeding-induced c-Fos expression in the brains of ovariectomized rats. Am J Physiol Regul Integr Comp Physiol. 2001;281(3):R738–R746. doi: 10.1152/ajpregu.2001.281.3.R738. [DOI] [PubMed] [Google Scholar]
  • 184.Rinaman L, Baker EA, Hoffman GE, Stricker EM, Verbalis JG. Medullary c-Fos activation in rats after ingestion of a satiating meal. American Journal of Physiology. 1998;275:R262–R268. doi: 10.1152/ajpregu.1998.275.1.R262. [DOI] [PubMed] [Google Scholar]
  • 185.Park TH, Carr KD. Neuroanatomical patterns of fos-like immunoreactivity induced by a palatable meal and meal-paired environment in saline- and naltrexone-treated rats. Brain research. 1998;805(1–2):169–180. doi: 10.1016/s0006-8993(98)00719-7. [DOI] [PubMed] [Google Scholar]
  • 186.Emond MH, Weingarten HP. Fos-like immunoreactivity in vagal and hypoglossal nuclei in different feeding states: a quantitative study. Physiol Behav. 1995;58(3):459–465. doi: 10.1016/0031-9384(95)00069-u. [DOI] [PubMed] [Google Scholar]
  • 187.Fraser KA, Davison JS. Meal-induced c-fos expression in brain stem is not dependent on cholecystokinin release. Am J Physiol. 1993;265(1 Pt 2):R235–R239. doi: 10.1152/ajpregu.1993.265.1.R235. [DOI] [PubMed] [Google Scholar]
  • 188.Rinaman L, Hoffman GE, Dohanics J, Le WW, Stricker EM, Verbalis JG. Cholecystokinin activates catecholaminergic neurons in the caudal medulla that innervate the paraventricular nucleus of the hypothalamus in rats. Journal of Comparative Neurology. 1995;360:246–256. doi: 10.1002/cne.903600204. [DOI] [PubMed] [Google Scholar]
  • 189.Li BH, Rowland NE. Cholecystokinin- and dexfenfluramine-induced anorexia compared using devazepide and c-fos expression in the rat brain. Regulatory Peptides. 1994;50:223–233. doi: 10.1016/0167-0115(94)90003-5. [DOI] [PubMed] [Google Scholar]
  • 190.Rinaman L, Verbalis JG, Stricker EM, Hoffman GE. Distribution and neurochemical phenotypes of caudal medullary neurons activated to express cFos following peripheral administration of cholecystokinin. The Journal of comparative neurology. 1993;338(4):475–490. doi: 10.1002/cne.903380402. [DOI] [PubMed] [Google Scholar]
  • 191.Olson BR, Hoffman GE, Sved AF, Stricker EM, Verbalis JG. Cholecystokinin induces c-fos expression in hypothalamic oxytocinergic neurons projecting to the dorsal vagal complex. Brain research. 1992;569(2):238–248. doi: 10.1016/0006-8993(92)90635-m. [DOI] [PubMed] [Google Scholar]
  • 192.Chen DY, Deutsch JA, Gonzalez MF, Gu Y. The induction and suppression of c-fos expression in the rat brain by cholecystokinin and its antagonist L364,718. Neurosci Lett. 1993;149(1):91–94. doi: 10.1016/0304-3940(93)90355-o. [DOI] [PubMed] [Google Scholar]
  • 193.Eckel LA, Houpt TA, Geary N. Estradiol treatment increases CCK-induced c-Fos expression in the brains of ovariectomized rats. Am J Physiol Regul Integr Comp Physiol. 2002;283(6):R1378–R1385. doi: 10.1152/ajpregu.00300.2002. [DOI] [PubMed] [Google Scholar]

RESOURCES