You are on page 1of 11

Peptides 26 (2005) 17711781

Review

The Agouti-related protein and its role in energy homeostasis


Adrian M. Stutz, Christopher D. Morrison, George Argyropoulos
Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
Received 14 July 2004; accepted 1 December 2004
Available online 14 June 2005

Abstract
The melanocortin system plays an important role in the regulation of energy homeostasis. The Agouti-related protein (AGRP) is a natural
antagonist of the action of alpha-melanocyte stimulating hormone (-MSH) at the melanocortin receptors (MCR). AGRP is upregulated by
fasting while intracerebroventricular injections of synthetic AGRP lead to increased appetite and food intake. Transgenic mice overexpressing
AGRP are also hyperphagic and eventually become obese. AGRP is, therefore, a significant regulator of energy balance and a candidate
gene for human fatness. Indeed, humans with common single nucleotide polymorphisms (SNPs) in the promoter or the coding region are
leaner and resistant to late-onset obesity than wild-type individuals. AGRP is also expressed in the periphery. Recent studies show that AGRP
in the adrenal gland is upregulated by fasting as much as it is in the hypothalamus. These data open up the possibility for a wider role by
AGRP not only in food intake but also in the regulation of energy balance through its actions on peripheral tissues. This review summarizes
recent advances in the biochemical and physiological properties of AGRP in an effort to enhance our understanding of the role this powerful
neuropeptide plays in mammalian energy homeostasis.
2005 Elsevier Inc. All rights reserved.
Keywords: Obesity; Adrenal; Hypothalamus; Leptin; Polymorphism

Contents
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
AGRP gene structure and tissue expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Physiological properties of AGRP action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Molecular mechanism of AGRP action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Hormonal and substrate regulation of AGRP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
AGRP action downstream of the ARC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
AGRP action in the periphery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Genetic models of AGRP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Human genetics of AGRP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

1772
1772
1772
1773
1773
1774
1775
1776
1776
1777
1777
1777

Abbreviations: AGRP, agouti-related protein; -MSH, alpha-melanocyte stimulating hormone; MC3R & MC4R, melanocortin receptor 3 and 4; i.c.v.,
intracerebroventricular; POMC, proopiomelanocortin; PVN, paraventricular nucleus; LHA, lateral hypothalamic area; ARC, arcuate nucleus; NPY, neuropeptide
Y; SNP, single nucleotide polymorphism; aa, amino acid; RNAi, RNA interference; SHU9119, Ac-Nle-c[Asp-His-Dnal(2 )-Arg-Trp-Lys]-Nh2
Corresponding author. Tel.: +1 225 763 2530; fax: +1 225 763 3030.
E-mail address: argyrog@pbrc.edu (G. Argyropoulos).
0196-9781/$ see front matter 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.peptides.2004.12.024

1772

A.M. Stutz et al. / Peptides 26 (2005) 17711781

1. Introduction
In 1992, Bultman et al. [16] discovered that the ectopic
expression of the Agouti protein was the basis for the monogenic mouse obesity model known as the Ay mouse. Ay is a
dominant mutation that leads to both yellow coat color and
morbid obesity [16,81]. Agouti is normally expressed within
the hair follicle, and promotes yellow hair color by antagonizing the action of alpha-melanocyte stimulating hormone
(-MSH) on the melanocortin receptor 1 (MC1R) [6,88].
The overexpression of Agouti within the hair follicle, thus,
explains the dominant yellow coat color, and it was also
hypothesized that the ectopic expression of Agouti within
regions of the brain controlling body weight homeostasis was
likewise responsible for the obesity and hyperphagia in this
animal model [69].
These potent anabolic effects of Agouti led to the
subsequent hypothesis that an endogenous antagonist of
melanocortin receptors might be produced locally within
the hypothalamus, and that this antagonist might participate in the hypothalamic regulation of energy balance. This
hypothesis was confirmed when Shutter et al. [110] identified a hypothalamic protein with high sequence homology to
Agouti, termed Agouti-related transcript and subsequently
Agouti-related protein (AGRP). These initial observations
are the basis for a growing body of evidence suggesting that
AGRP plays a significant role in the hypothalamic regulation of energy homeostasis [30,71,125,138], and this review
describes recent advances in our understanding of AGRP
action within the hypothalamus, as well as the potential for
AGRP-dependant effects in the periphery.

2. AGRP gene structure and tissue expression


The human AGRP is a relatively short gene, spanning
1.1 kb on chromosome 16q22. It consists of four exons and
encodes a 132 amino acid (aa) protein, with the rodent
ortholog encoding a 131 aa protein [13,76,110]. The AGRP
protein contains nine cysteine residues at conserved positions that form disulfide bridges and are important for its
function [110]. Human and murine AGRP expression profiles showed a predominant presence of both orthologs in
the hypothalamus, the subthalamic nucleus, and a shorter
transcript lacking the 5 non-coding exon in the adrenal and
at lower expression levels in the testis, lung, kidney, spinal
cord, and dorsal root ganglia [7,110]. Although, at present,
it is unclear whether these transcripts arise from differential splicing or from two independently regulated promoters,
the 5 untranslated exon demonstrated significant promoter
activity in a periphery-derived cell line only (Chinese hamster ovary) [13]. This finding suggests that this exon may play
a role in the peripheral expression of AGRP, and supports
the existence of two active promoters, each responsible for
either brain- or periphery-specific (the adrenal cortex, testis,
and lung) expression [13]. AGRP has also been sequenced or

identified in the pig [45], the sheep [1], the fish Fugu (Takifugu rubripes) [63], the zebrafish (Danio rerio) [113], the
goldfish [17], the Japanese quail [10,93] and in ring doves
(Streptopelia risoria) [115,116]. In many of these species
AGRP levels are upregulated by fasting [1,43], underscoring
a conserved role for AGRP in energy homeostasis.

3. Physiological properties of AGRP action


Perhaps the best-documented aspect of AGRP action is its
profound stimulation of food intake. Transgenic mice overexpressing AGRP exhibit severe obesity, have increased body
length, hyperinsulinemia, late-onset hyperglycemia, pancreatic islet hyperplasia, and reduced corticosterone levels [38].
These effects of AGRP are proposed to be mediated predominantly by the brain, as both murine and human AGRP
orthologs stimulate hyperphagia when administered intracerebroventricularly (i.c.v.) [42,102,103,112]. AGRP is one
of the most potent orexigenic peptides known, and is unique
in that it displays long-lasting effects on food intake, with animals displaying hyperphagia even 7 days following a single
i.c.v. injection [42]. While the mechanisms underlying this
long-lasting effect are unclear, it appears to be independent
of a persistent blockade of melanocortin receptors [42,62].
In addition to alterations in feeding behavior, recent data
indicate that AGRP also acts to suppress energy expenditure,
and that this effect contributes significantly to its overall effect
on energy homeostasis. Central administration of AGRP
decreases uncoupling protein 1 (UCP1) expression and temperature within brown adipose tissue (BAT) [111,135], likely
via an inhibition of sympathetic outflow [135]. Interestingly,
administration of N-terminal parts of AGRP increased body
and fat weight in the absence of hyperphagia [37]. Furthermore, reduction of AGRP levels by 50% using RNA interference resulted in increased energy expenditure and loss of
body weight without changes in food intake [73]. Together,
these data indicate that alterations in energy expenditure play
a significant role in the regulation of energy homeostasis by
AGRP.
AGRP also influences physiological systems beyond feeding and energy expenditure. In particular, recent evidence
suggests that AGRP influences neuroendocrine function by
regulating the hypothalamic pituitary axis. The i.c.v. injection of human AGRP into ovariectomized rhesus monkeys
resulted in elevated cortisol, adrenocorticotropin hormone
(ACTH), and prolactin (PRL) release [132]. In addition, i.c.v.
injected AGRP enhanced the ability of IL-1 to increase
ACTH, likely by affecting the function of -MSH at hypothalamic melanocortin receptors. This observation supports a
modulatory role of AGRP in the neuroendocrine responses to
inflammation, which itself may possibly promote obesity and
type 2 diabetes [22]. Evidence also suggests that AGRP has an
inhibitory influence over the hypothalamicpituitarythyroid
axis, with AGRP administration suppressing hypothalamic thyroid stimulating hormone (TSH)-releasing hormone

A.M. Stutz et al. / Peptides 26 (2005) 17711781

(TRH) expression and decreasing circulating levels of thyroid


hormones [31].

4. Molecular mechanism of AGRP action


Available evidence suggests that AGRP is a melanocortin
receptor antagonist, blocking the effect of -MSH and other
agonists to stimulate these receptors [32,83,134]. However,
several studies [47,83] indicate that AGRP may go beyond a
simple antagonist and, additionally, act as an inverse agonist
at the MC4R. Inverse agonist activity would indicate effects
of AGRP on feeding behavior and energy homeostasis that
occur independent of -MSH or other melanocortin receptor
ligands.
In addition to -MSH, other Proopiomelanocortin
(POMC) derived ligands such as -MSH, desacetyl -MSH,
ACTH, and -lipotrophic hormone regulate melanocortin
receptors. Importantly, the orthosterical, antagonistic function of AGRP (aa83132) was the same for all POMC
peptides, suggesting that AGRP (aa83132) inhibits Gscoupled accumulation of intracellular cAMP at the MC4R
in a more general way [94]. Evidence also supports a role
for the cell surface heparan sulfate proteoglycan syndecan-3
in the facilitation of AGRP action at the melanocortin receptor, again influencing the balance of agonist to antagonist
[99,100].
Most studies focusing on in vivo AGRP function have used
carboxyl-terminal AGRP peptides that mimic the effect of the
full-length protein. This approach appears legitimate, as synthetic peptides of the amino-terminus, hAGRP(aa2551), and
the mid-portion, hAGRP(aa5482), of the human AGRP protein were devoid of antagonistic action for -MSH [96,97],
whereas a synthetic variant containing the 46 C-terminal
residues, hAGRP(aa87132), was active and equipotent to
the mature mouse homolog [46,47,119,133]. Moreover, the
hAGRP(aa87132) peptide was able to bind effectively
the melanocortin receptors MC3R, MC4R, and MC5R,
thus, inhibiting binding of -MSH [32,83,134]. In depth
NMR structure analysis of this AGRP domain (aa87132)
revealed an inhibitor cysteine-knot structure [51], which
makes contact with the melanocortin receptor 3 and 4 with
two loops present in this structure [77]. Recently, elongation of the minimum core decapeptide Yc[CRFFNAFC]Y
of AGRP, which acts as an antagonist at the MC4R but
not the MC3R, by two amino acids at either the C- or
the N-terminus conferred antagonistic function also on the
MC3R, resembling the profile of full-length AGRP [53]. All
these reports underscore the importance of the C-terminus
of AGRP in the antagonistic function at the melanocortin
receptors.
Despite this clear importance of the C-terminus, a recent
study suggested that N-terminal parts of AGRP, which
are unable to bind to melanocortin receptors, have effects
on energy expenditure that are, therefore, independent of
melanocortin receptors [37]. A single i.c.v. injection of AGRP

1773

(aa83132) increased cumulative food intake, whereas Nterminal parts AGRP (aa2551) and AGRP (aa5482) did not
affect the amount of food consumption. However, injection of
any of the three portions of AGRP resulted in decreased oxygen consumption and colonic temperature, both readouts for
energy expenditure, and the two N-terminal parts of AGRP
increased body weight and epididymal/mesenteric fat weight,
despite the absence of hyperphagia and cross-reactivity with
MC4R [37]. Additional experiments are necessary to confirm and elucidate this interesting observation. One possible
explanation for this melanocortin independent effect is a
potential interaction with co-receptors. One such example
could be syndecan-3 [99,100], which can only bind to Nterminal AGRP. A repeat of the study, e.g. in syndecan-3
knockout mice or by altering the availability of membranebound syndecan-3, together with further mutagenesis and in
vitro binding experiments would be helpful to understand the
implications of this finding.

5. Hormonal and substrate regulation of AGRP


AGRP is produced predominately by a subpopulation of
neurons within the hypothalamic arcuate nucleus (ARC).
Considering that the ARC mediates the effects of an array
of peripheral metabolic signals, this anatomical localization
suggests that AGRP also plays a role in energy homeostasis.
Indeed, AGRP is predominately localized to a population of
neurons within the medial aspect of the ARC that co-express
neuropeptide Y (NPY). NPY has long been associated with
hypothalamic regulation of feeding behavior, and by coexpressing AGRP and NPY; these so called NPY/AGRP
neurons are considered key mediators in the response to
nutrient depletion and subsequent regulation of energy
balance.
Multiple hormonal signals influence AGRP expression.
Long acting satiety signals such as leptin or insulin act to
decrease AGRP, while maintaining physiological levels of
these hormones blocks fasting induced increases of AGRP
[12,105,128]. Loss of leptin or insulin receptors within the
brain leads to markedly elevated AGRP expression, and
AGRP is also upregulated in leptin-deficient (ob/ob) obese
mice, irrespective of fasting [89,110]. Together these observations suggest that these hormones reduce appetite, in
part, by inhibiting AGRP (and NPY) expression [15,110].
Additionally, recent work indicates that these hormones can
acutely alter membrane potential and reduce neuronal firing from neurons containing NPY, suggesting that leptin and
insulin rapidly inhibit the activity of NPY/AGRP neurons
[124]. Although progress has been made in identifying factors downstream of leptin signaling on AGRP [12,105,128],
it is not clear through which mechanism leptin regulates
AGRP. Potential signaling pathways include the AMP-kinase
pathway [78,79], the JAK-STAT pathway [9,35,44,48], and
the PI3K pathway [84,85]. It remains to be seen which
pathway, if not a combination of two or more, is respon-

1774

A.M. Stutz et al. / Peptides 26 (2005) 17711781

sible in mediating the leptin effect on AGRP expression


and, therefore, reducing food intake and increasing energy
expenditure.
The gut-derived protein ghrelin has also been implicated
in the regulation of AGRP neurons. Unlike leptin and insulin,
ghrelin principally acts to stimulate feeding and body weight
gain [64,121] and activates NPY/AGRP neurons. Ghrelin
is an endogenous ligand for the growth hormone secretagogue receptor (GHS-R) and has been shown to up-regulate
expression of AGRP [5456,82,131]. In addition, administration of ghrelin acutely induces c-Fos (a marker of neuronal
activation) within neurons containing NPY (and presumably
AGRP). Genetic evidence also supports a critical role for
the NPY/AGRP neurons in mediating ghrelin action, since
AGRP/NPY double knockout (k.o.) mice are resistant to
ghrelin-dependant increase in food intake [19]. However,
mice lacking only AGRP or only NPY do not display this
phenotype, whereas AGRP expression was normal in ghrelin
k.o. mice [130].
In addition to leptin, insulin, and ghrelin, other endocrine
signals also influence AGRP levels within the hypothalamus.
Glucocorticoids have been implicated in the regulation of
energy homeostasis and removal of glucocorticoid signaling (for instance, by adrenalectomy) ameliorates obesity in
a number of physiological and genetic models. Adrenalectomy decreases sensitivity to both AGRP [26] and NPY
[136] while increasing the sensitivity to -MSH [26] and
leptin [26,72]. In a different study, adrenalectomy blocked
fasting induced increases in AGRP [70]. Exogenous administration of glucocorticoids, on the other hand, increases food
intake, body weight, as well as AGRP and NPY expression [52]. Another study, supportive of an essential role of
glucocorticoids on AGRP expression, demonstrated that corticosterone secretion temporally coincided with the rising
phase of diurnal AGRP expression [70]. Depletion of corticosterone by adrenalectomy abolished this AGRP diurnal
rhythm, which was restored by exogenous corticosterone
replacement, highlighting its requirement to maintain the normal diurnal AGRP expression cycle [70]. Together, these
observations suggest that glucocorticoids have significant
effects on energy homeostasis potentially mediated by action
on hypothalamic NPY/AGRP neurons. Other hormones that
could potentially regulate AGRP expression include PYY
[30], cholecystokinin (CCK) [80], gastrin [122], glucagonlike peptide 1 [120], and vasopressin [104] that are transmitted as afferent signals by the vagus nerve to the brain [11].
More elaborate studies though are required to address the role
that these hormones play on the regulation of AGRP.
Fatty acids (FAs) may also influence the expression of
AGRP. Food intake and appetite in humans were significantly
affected by intestinal infusions of intralipid and emulsions
of oils enriched with stearic, oleic, and linoleic acids [34],
while i.c.v. administration of oleic acid in animals mimics
the anorexigenic effects of leptin and downregulates NPY
expression [87]. Furthermore, feeding mice a highly saturated
fat diet, but not polyunsaturated FAs (PUFAs), downregu-

lated AGRP expression [127]. Fat-specific satiety has been


reported in humans for fat high in linoleic acid [57], while
the degree of saturation of FAs can influence post-ingestive
satiety with the PUFAs exerting a stronger control than the
monounsaturated fatty acids (MUFAs) [66]. The exact mechanism by which FAs may influence AGRP expression is not
known at this point, but we hypothesize that it could happen through the activation of critical transcription factors
[68,109]. Indeed, FAs are involved in the activation of PPARs
[23,61] and PUFAs, in particular, are linked to the activation
of SREBP [91,92] and the suppression of other transcription
factors [86]. FAs may also be involved in the activation of
leptin [98], thus, employing multiple pathways for regulating AGRP expression.
Finally, changes in glucose concentration are known
to influence AGRP expression potentially linking appetite
to metabolic need. Central administration of 2-Deoxy-dglucose (2DG), a nonmetabolizable glucose analog that
inhibits glucose utilization, increased AGRP gene expression and elicits glucoprivic food intake [33,107]. Recently,
the effect of high glucose levels to decrease AGRP expression could be correlated with decreased phosphorylation of
AMP-kinase [67]. Further studies are needed to confirm these
findings and establish a role for glucose in AGRP regulation
in vivo.
In addition to being upregulated in response to fasting,
AGRP is also increased in other physiological situations
whereby increased food intake is desirable or necessary. For
example, during pregnancy AGRP levels, but not POMC,
MC4R or NPY, were elevated in Wistar rats, suggesting that
AGRP could play a role in pregnancy-associated hyperphagia [101]. Similarly, AGRP is upregulated in lactating sheep
[114] while ring doves express elevated AGRP levels during
the post hatching stages when parents eat more food to feed
their young [115]. Some diseases that result in insufficient
food intake correlate with reduced levels of AGRP, such as a
mouse model of PraderWilli syndrome, in which neonates
display failure-to-thrive [36]. In a rat experimental model
of anorexia nervosa, central infusion of AGRP prevented
self-starvation by counteracting physical hyperactivity and
dominantly stimulating food intake [58]. AGRP treatment in
tumor-bearing animals lead to a maintenance of lean body
mass and circadian activity patterns during tumor growth,
without negatively affecting tumor size [75]. AGRP, therefore, could modulate disease-related changes in food intake
and contribute to maintenance to overall energy balance in
response to hormonal and substrate stimuli.

6. AGRP action downstream of the ARC


The neuronal sites and signaling mechanisms mediating AGRP action within regions downstream of the ARC
are currently not well characterized. Neurons within the
paraventricular nucleus (expressing TRH, corticotrophinreleasing hormone (CRH), opioidergic) and lateral hypotha-

A.M. Stutz et al. / Peptides 26 (2005) 17711781

lamus (expressing melanin-concentrating hormone (MCH)


and orexin) are strong candidates as mediators of downstream
AGRP signaling. Within the PVN, AGRP has been shown to
specifically inhibit TRH neurons [31], in opposition to both
-MSH and leptin, whereas a synthetic melanocortin receptor antagonist (SHU9119) that mimics AGRP action was able
to block leptin induced c-Fos (a marker of neuronal activation) within paraventricular nucleus (PVN) neurons [106].
This model of opposing agonist and antagonist activity is
well documented in the PVN, and is likely replicated in
other areas receiving input from AGRP and POMC neurons.
For instance, AGRP neurons also strongly project to MC4R
expressing neurons within the lateral hypothalamic area
(LHA) [29] and where AGRP increases c-Fos. Interestingly,
both -MSH and leptin are believed to inhibit neurons within
the LHA, indicating that although the agonist/antagonist balance exists in this region, the specific effects on neuronal
activity differ [30].
AGRP neurons also project to a variety of areas outside the
hypothalamus, and MC4 receptors are also expressed in many
of these areas. Central administration of AGRP induces c-Fos
within nucleus accumbens shell, lateral septum, NTS and
central amygdala [40], and the AGRP dependant activation
of these regions was more robust 24 h after AGRP injection as compared to 2 h, suggesting that these regions may
mediate the long-lasting effects of AGRP [40]. AGRP was
also shown to specifically increase c-Fos in orexin neurons
within the LHA even 24 h after i.c.v. injection, suggesting
that an activation of LHA orexin neurons could also mediate
the long-lasting hyperphagia induced by AGRP [137].
Recent work also suggests that AGRP signals in part
via an opiodergic pathway, as combined inhibition of and the -opioid receptors signaling blocked AGRP-induced
feeding [14,41]. Interestingly, one report indicates that mice
injected with AGRP favor calorically dense chow to a palatable sucrose solution, indicating that the main function of
AGRP on food behavior may be to satisfy energy needs rather
than enhance palatability or hedonic value of food [90,129].

7. AGRP action in the periphery


The site of the most abundant hAGRP, human cocaine- and
amphetamine-regulated transcript (hCART), and hPOMC
expression is the adrenal gland [39]. Within the adrenal cortex, the short isoform of the rat AGRP is upregulated during
fasting [8], paralleling the 18-fold increase that occurs within
the hypothalamus [43]. Moreover, plasma levels of AGRP
are increased in obese men [60] and fasted rats [108]. Nevertheless, the function of AGRP in the periphery remains
unresolved. The active isoform of human AGRP (aa83132)
crosses the bloodbrain barrier [59], raising the possibility for
peripherally expressed AGRP to directly access melanocortin
receptors within the brain.
Although only a few studies have focused on peripheral
AGRP action, these studies highlight the potential for cir-

1775

culating or locally-produced AGRP to influence peripheral


tissues. The obvious question is what is the role of AGRP
in the periphery? In all probability, AGRP in the periphery
does the same as it does in the hypothalamus, i.e. binds to the
melanocortin or perhaps other G protein-coupled receptors.
Both AGRP and Agouti have direct effects on adipocytes,
influencing the expression of fatty acid synthase and leptin
[20,21,28], and blocking -MSH dependant effects on leptin gene expression [49]. In addition to effects in adipocytes,
AGRP may also have a paracrine role in adrenal function,
with adrenal-derived AGRP being regulated by glucocorticoids and blocking the induction of corticosterone secretion
by -MSH [24,25]. Furthermore, experiments in the chicken
have revealed that AGRP binds to MC3R in the adrenal
[117,118]. The situation is less clear if a negative feedback
loop exists in which AGRP itself acts as a negative regulator of leptin action, as was first suggested by Ebihara et al.
[28]. In other experiments, however, central administration of
AGRP in rats for 3- and 7-day periods resulted in significant
increases of plasma leptin levels, even when AGRP induced
hyperphagia was prevented [65]. In a recent study, AGRP
was found to be robustly expressed in epididymal fat and to
be upregulated in this tissue by feeding rather than by fasting
[18]. Leptin was also upregulated by feeding in the epididymal fat but unexpectedly also in the adrenal gland. This was
observed in both the obesity-susceptible C57BL/6J (C57)
and the obesity-resistant CAST/Ei mouse strain. Surprisingly, AGRP expression was higher in the fasted state in the
hypothalamus and the adrenal gland in the leaner and obesityresistant CAST/Ei strain instead of in the obesity-susceptible
C57 strain. This result was recapitulated in another obesitysusceptible mouse strain, tub/tub that also had reduced AGRP
expression in the hypothalamus [4]. These two reports present
the fresh idea that gene-specific obesity (tub/tub mutation)
or generalized fatness (C57 mouse) does not correlate positively with AGRP levels. It is also possible for a lean mouse
(CAST/Ei) to express higher than the C57 AGRP levels in the
hypothalamus without becoming obese, provided that energy
is expended through a hyperactive lifestyle. Rather unexpectedly, in vitro leptin treatment of a mouse hypothalamusderived cell line upregulated endogenous AGRP and this
result was recapitulated in adrenal-derived cells, after an
acute 6-h exposure. However, a 60-h exposure to leptin
resulted in downregulation of AGRP [18]. Treatment of
hypothalamic and adrenal cells with AGRP not only upregulated its endogenous expression but also upregulated endogenous leptin in the adrenal cells [18]. These data reveal a
novel feedback loop and reciprocal transcriptional regulation between AGRP and leptin in the hypothalamus and the
periphery. An autocrine role for AGRP in the rat adrenal cortex modulating the actions of POMC has also been proposed
[8]. Therefore, peripheral expression of AGRP, and particularly in the adrenal gland, could play a significant role in
energy homeostasis. This is even more relevant to human
physiology since AGRP (unlike its murine ortholog) is not
expressed in visceral nor subcutaneous fat [18].

1776

A.M. Stutz et al. / Peptides 26 (2005) 17711781

Table 1
Phenotypes of AGRP mouse models
Model

Phenotype

Reference

AGRP transgenic
i.c.v. injection of AGRP (aa87132)
AGRP k.o.
AGRP/NPY double k.o.

Food intake , body weight , body length , corticosterone levels


Food intake , body weight , energy expenditure
Normal weight gain and feeding behavior, slight difference in MCH
Normal weight gain and feeding behavior, unresponsive to ghrelin
administration
50% Inhibition of AGRP expression, metabolic rate , body weight ,
normal food intake

[38]
[37,42,62,102,103,112,116,132,137]
[19,95]
[19,95]

AGRP RNAi injection into


hypothalamus

[73]

, Increase; , decrease.

8. Genetic models of AGRP


In an effort to elucidate the functional properties of AGRP,
mouse models were created to overexpress or be AGRPdeficient (Table 1). Specifically, transgenic mice overexpressing AGRP are hyperphagic, exhibit severe obesity, show
increased body length, hyperinsulinemia, late-onset hyperglycemia, pancreatic islet hyperplasia, and reduced corticosterone levels [38]. These observations suggest that the elevation of AGRP observed following fasting or in response to
hormonal signals, promotes increased feeding behavior. Surprisingly, AGRP knockout mice (AGRP/ ) exhibit normal
feeding behavior without changes in body weight and cumulative food intake, in comparison to their wild-type littermates
[95]. AGRP/ mice are fertile and exhibit no gross histological or pathological abnormalities, and also have comparable
serum levels of glucose, leptin, triglycerides, and free fatty
acids to wild-type mice. Furthermore, AGRP/ mice do not
show a higher preference for high fat or high sucrose food
over their wild-type littermates. AGRP/ , NPY/ and double AGRP/NPY k.o. mice show no feeding or body weight
deficits and maintain a normal response to starvation [95]. In
AGRP/ mice, expression levels of MC3R, MC4R or NPY
were comparable to levels found in wild-type mice. These
data suggest that the imprinted loss of AGRP could result
in the development of compensatory mechanisms. Indeed,
a marginal increase in the orexigenic MCH expression was
observed in the AGRP/ mice [95]. Another contributing
factor to the absence of a robust phenotype of these AGRP/
mice, could be the genetic background of the mice. Passage
of the mutation to another mouse strain could address this
issue.
In another study, however, RNA interference (RNAi) in the
ARC was used to decrease AGRP expression rather than completely delete the gene, which may be a more physiologically

relevant approach. Indeed, RNAi lead to a 50% inhibition of


AGRP message within the hypothalamus, which was associated with increased metabolic rate and reduced body weight
but without changing food intake [73]. This is consistent with
conclusions from pair-fed animals treated with AGRP, which
resulted in increased BAT UCP1 and increased fat mass and
leptin levels, potentially regulating overall energy expenditure [111]. Therefore, an approach to moderately decrease
AGRP expression levels may provide a means to regulate
body weight and increase energy expenditure.

9. Human genetics of AGRP


In addition to animal models that clearly implicate AGRP
in the regulation of energy balance, studies in humans show
promising associations of AGRP single nucleotide polymorphisms (SNPs) with resistance to obesity and perhaps type 2
diabetes mellitus (T2DM) in Black Africans.
AGRP plasma levels were elevated in obese men [60] and,
consistently with data from animal models, during fasting
[108]. The former finding was recently replicated in another
cohort while a 6-day fast increased AGRP plasma levels in
lean individuals but not in obese ones [50].
Mutation screening has revealed several functional SNPs
in AGRP that are associated with various phenotypes of leanness (Table 2). Two SNPs in the 5 -UTR have been reported
in Black but not White individuals. SNP 3019G > A was in
complete linkage disequilibrium (LD) with another promoter
SNP, 38C > T [5]. Despite LD, the 3019G > A SNP exhibited functional dimorphism whereby the two linked alleles
had opposite effects on promoter activity but the net effect
was likely determined by the 38C > T SNP because of its
position in a region with 1000-fold higher promoter activity [5]. The T allele of the 38C > T SNP was associated

Table 2
Described SNPs in the human AGRP gene
SNP

Function

Phenotype

Reference

3019G > A
38
G423A
C690T
Ala67Thr

Alters promoter activity


Alters promoter activity
Silent
Silent
Alters secondary structure (algorithm prediction)

In complete LD with 38C > T


Associated with obesity, T2D
None reported
None reported
Associated with anorexia nervosa, resistance to late-onset obesity

[5]
[2,76]
[123]
[123]
[3,13,27,74,126]

A.M. Stutz et al. / Peptides 26 (2005) 17711781

with leanness in Sierra Leoneans and reduced fatness in the


Blacks of the HERITAGE Family Study [2]. In addition, all
the diabetics (T2DM) recorded in the Sierra Leonean cohort
were CC homozygous (for the 38C > T SNP) suggesting that
the T allele may not only predispose to obesity-resistance in
Blacks on both sides of the Atlantic, but may also provide
a protective mechanism against the development of T2DM
in the Black Africans. Two silent mutations in the coding
regions were not associated with any obese phenotypes [123].
Another SNP in the coding region that changed the alanine
at position 67 to a threonine was not associated with obesity
[13,27] but was associated with anorexia nervosa [126]. Interestingly, this SNP was found in Whites only, in contrast to
the two promoter SNPs (3019G > A and 38C > T) that were
found in Blacks only. Subsequent studies have shown that heterozygotes for the Ala67Thr SNP were resistant to late-onset
obesity [3] and this finding was later replicated in another
cohort for the rare homozygous individuals for the threonine
allele who, again, had reduced body fatness [74]. These data
suggest that AGRP SNPs could affect metabolic phenotypes
in humans, with individuals carrying the less frequent alleles
(either the Black- or White-specific ones) potentially protected from developing obesity in obesigenic environments.

10. Summary
AGRP is a unique neuropeptide representing a rare
endogenous receptor antagonist or inverse agonist. By acting on melanocortin receptors in brain regions controlling
food intake and energy expenditure, AGRP represents a critical component in a complex regulatory system that exists to
maintain energy balance. Its expression in peripheral tissues
and in particular the adrenal gland suggests a wider role for
AGRP in energy homeostasis, potentially involving feedback
regulatory interactions with leptin. Although genetic AGRPdeficiency does not induce a dramatic phenotype in mice,
a large body of evidence suggests that an increase in AGRP
contributes to an increase in appetite and a decrease in energy
expenditure. From the human studies we learn that common
AGRP SNPs predispose individuals to obesity-resistance and
may provide a protective mechanism against fatness under
obesigenic conditions. AGRP is an attractive target gene for
modulating pathological food intake and obesity but its exact
role in energy expenditure and overall energy homeostasis
requires further investigation. Future studies to elucidate the
peripheral actions of AGRP in the context of its binding to
melanocortin or perhaps other G-protein-coupled receptors
should be intensified. A better understanding of both the
peripheral and hypothalamic interactions between AGRP and
leptin would help to identify the best step to modulate this
complex feedback network. Furthermore, the initial observations that the N-terminal part of AGRP may influence energy
homeostasis independent of food intake and melanocortin
signaling should be investigated in more detail. This could
lead to the design of selective drugs that affect certain inter-

1777

actions of AGRP without necessarily affecting signaling of


POMC-derived peptides on melanocortin receptors.

Acknowledgement
This work was supported by the National Institute of Diabetes and Digestive and Kidney Diseases (grant DK62156 to
GA).

References
[1] Adam CL, Archer ZA, Findlay PA, Thomas L, Marie M. Hypothalamic gene expression in sheep for cocaine- and amphetamineregulated transcript, pro-opiomelanocortin, neuropeptide y, agoutirelated peptide and leptin receptor and responses to negative energy
balance. Neuroendocrinology 2002;75(4):2506.
[2] Argyropoulos G, Rankinen T, Bai F, Rice T, Province M, Leon A,
et al. The agouti related protein and body fatness in humans. Int J
Obes 2003;27:27680.
[3] Argyropoulos G, Rankinen T, Neufeld DR, Rice T, Province MA,
Leon AS, et al. A polymorphism in the human agouti-related protein is associated with late-onset obesity. J Clin Endocrinol Metab
2002;87(9):4198202.
[4] Backberg M, Madjid N, Ogren SO, Meister B. Down-regulated
expression of agouti-related protein (AGRP) mRNA in the hypothalamic arcuate nucleus of hyperphagic and obese tub/tub mice. Brain
Res Mol Brain Res 2004;125(12):12939.
[5] Bai F, Rankinen T, Charbonneau C, Belsham DD, Rao DC,
Bouchard C, et al. Functional dimorphism of two hAgRP promoter SNPs in linkage disequilibrium. J Med Genet 2004;41(5):
3503.
[6] Barsh GS, Ollmann MM, Wilson BD, Miller KA, Gunn TM.
Molecular pharmacology of Agouti protein in vitro and in vivo.
Ann N Y Acad Sci 1999;885:14352.
[7] Beltramo M, Campanella M, Tarozzo G, Fredduzzi S, Corradini L,
Forlani A, et al. Gene expression profiling of melanocortin system
in neuropathic rats supports a role in nociception. Brain Res Mol
Brain Res 2003;118(12):1118.
[8] Bicknell AB, Lomthaisong K, Gladwell RT, Lowry PJ. Agouti
related protein in the rat adrenal cortex: implications for
novel autocrine mechanisms modulating the actions of proopiomelanocortin peptides. J Neuroendocrinol 2000;12(10):977
82.
[9] Bjorbaek C, Kahn BB. Leptin signaling in the central nervous
system and the periphery. Recent Prog Horm Res 2004;59:30531.
[10] Boswell T, Li Q, Takeuchi S. Neurons expressing neuropeptide
Y mRNA in the infundibular hypothalamus of Japanese quail are
activated by fasting and co-express agouti-related protein mRNA.
Brain Res Mol Brain Res 2002;100(12):3142.
[11] Bray GA. Afferent signals regulating food intake. Proc Nutr Soc
2000;59(3):37384.
[12] Broberger C, Johansen J, Johansson C, Schalling M, Hokfelt T. The
neuropeptide Y/agouti gene-related protein (AGRP) brain circuitry
in normal, anorectic, and monosodium glutamate-treated mice. Proc
Natl Acad Sci USA 1998;95(25):150438.
[13] Brown AM, Mayfield DK, Volaufova J, Argyropoulos G. The gene
structure and minimal promoter of the human agouti related protein.
Gene 2001;277(12):2318.
[14] Brugman S, Clegg DJ, Woods SC, Seeley RJ. Combined blockade of both micro- and kappa-opioid receptors prevents the
acute orexigenic action of Agouti-related protein. Endocrinology
2002;143(11):426570.

1778

A.M. Stutz et al. / Peptides 26 (2005) 17711781

[15] Bruning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban


PC, et al. Role of brain insulin receptor in control of body weight
and reproduction. Science 2000;289(5487):21225.
[16] Bultman SJ, Michaud EJ, Woychik RP. Molecular characterization
of the mouse agouti locus. Cell 1992;71(7):1195204.
[17] Cerda-Reverter JM, Peter RE. Endogenous melanocortin antagonist in fish: structure, brain mapping, and regulation by fasting of the goldfish agouti-related protein gene. Endocrinology
2003;144(10):455261.
[18] Charbonneau C, Bai F, Richards BS, Argyropoulos G. Central and
peripheral interactions between the agouti-related protein and leptin. Biochem Biophys Res Commun 2004;319(2):51824.
[19] Chen HY, Trumbauer ME, Chen AS, Weingarth DT, Adams JR,
Frazier EG, et al. Orexigenic action of peripheral ghrelin is mediated by neuropeptide Y (NPY) and agouti-related protein (AgRP).
Endocrinology 2004.
[20] Claycombe KJ, Wang Y, Jones BH, Kim S, Wilkison WO, Zemel
MB, et al. Transcriptional regulation of the adipocyte fatty acid
synthase gene by agouti: interaction with insulin. Physiol Genomics
2000;3(3):15762.
[21] Claycombe KJ, Xue BZ, Mynatt RL, Zemel MB, MoustaidMoussa N. Regulation of leptin by agouti. Physiol Genomics
2000;2(3):1015.
[22] Dandona P, Aljada A, Bandyopadhyay A. Inflammation: the link
between insulin resistance, obesity and diabetes. Trends Immunol
2004;25(1):47.
[23] Delerive P, Fruchart JC, Staels B. Peroxisome proliferator-activated
receptors in inflammation control. J Endocrinol 2001;169(3):4539.
[24] Dhillo WS, Small CJ, Gardiner JV, Bewick GA, Whitworth EJ,
Jethwa PH, et al. Agouti-related protein has an inhibitory paracrine
role in the rat adrenal gland. Biochem Biophys Res Commun
2003;301(1):1027.
[25] Doghman M, Delagrange P, Blondet A, Berthelon MC, Durand
P, Naville D, et al. Agouti-related protein antagonizes glucocorticoid production induced through mc4-r activation in bovine adrenal
cells: a possible autocrine control. Endocrinology 2003.
[26] Drazen DL, Wortman MD, Schwartz MW, Clegg DJ, van
Dijk G, Woods SC, et al. Adrenalectomy alters the sensitivity of the central nervous system melanocortin system. Diabetes
2003;52(12):292834.
[27] Dubern B, Clement K, Pelloux V, Froguel P, Girardet JP, GuyGrand B, et al. Mutational analysis of melanocortin-4 receptor,
agouti-related protein, and alpha-melanocyte-stimulating hormone
genes in severely obese children. J Pediatr 2001;139(2):2049.
[28] Ebihara K, Ogawa Y, Katsuura G, Numata Y, Masuzaki H, Satoh
N, et al. Involvement of agouti-related protein, an endogenous
antagonist of hypothalamic melanocortin receptor, in leptin action.
Diabetes 1999;48(10):202833.
[29] Elias CF, Saper CB, Maratos-Flier E, Tritos NA, Lee C, Kelly
J, et al. Chemically defined projections linking the mediobasal
hypothalamus and the lateral hypothalamic area. J Comp Neurol
1998;402(4):44259.
[30] Ellacott KL, Cone RD. The central melanocortin system and the
integration of short- and long-term regulators of energy homeostasis. Recent Prog Horm Res 2004;59:395408.
[31] Fekete C, Sarkar S, Rand WM, Harney JW, Emerson CH,
Bianco AC, et al. Agouti-related protein (AGRP) has a central inhibitory action on the hypothalamic-pituitary-thyroid (HPT)
axis; comparisons between the effect of AGRP and neuropeptide Y on energy homeostasis and the HPT axis. Endocrinology
2002;143(10):384653.
[32] Fong TM, Mao C, MacNeil T, Kalyani R, Smith T, Weinberg
D, et al. ART (protein product of agouti-related transcript) as an
antagonist of MC-3 and MC-4 receptors. Biochem Biophys Res
Commun 1997;237(3):62931.
[33] Fraley GS, Ritter S. Immunolesion of norepinephrine and
epinephrine afferents to medial hypothalamus alters basal and 2-

[34]

[35]

[36]

[37]

[38]

[39]
[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

[48]
[49]

[50]

deoxy-d-glucose-induced neuropeptide Y and agouti gene-related


protein messenger ribonucleic acid expression in the arcuate
nucleus. Endocrinology 2003;144(1):7583.
French SJ, Conlon CA, Mutuma ST, Arnold M, Read NW, Meijer
G, et al. The effects of intestinal infusion of long-chain fatty acids
on food intake in humans. Gastroenterology 2000;119(4):9438.
Gao Q, Wolfgang MJ, Neschen S, Morino K, Horvath TL, Shulman GI, et al. Disruption of neural signal transducer and activator
of transcription 3 causes obesity, diabetes, infertility, and thermal
dysregulation. Proc Natl Acad Sci USA 2004;101(13):46616.
Ge Y, Ohta T, Driscoll DJ, Nicholls RD, Kalra SP. Anorexigenic melanocortin signaling in the hypothalamus is augmented
in association with failure-to-thrive in a transgenic mouse model
for PraderWilli syndrome. Brain Res 2002;957(1):425.
Goto K, Inui A, Takimoto Y, Yuzuriha H, Asakawa A, Kawamura Y, et al. Acute intracerebroventricular administration of either
carboxyl-terminal or amino-terminal fragments of agouti-related
peptide produces a long-term decrease in energy expenditure in
rats. Int J Mol Med 2003;12(3):37983.
Graham M, Shutter JR, Sarmiento U, Sarosi I, Stark KL. Overexpression of Agrt leads to obesity in transgenic mice. Nat Genet
1997;17(3):2734.
Graunke DM, Argyropoulos G. Deciphering transcriptional regulation relevant to eating behavior. Curr Genom 2003;4:62333.
Hagan MM, Benoit SC, Rushing PA, Pritchard LM, Woods SC,
Seeley RJ. Immediate and prolonged patterns of Agouti-related
peptide-(83132)-induced c-Fos activation in hypothalamic and
extrahypothalamic sites. Endocrinology 2001;142(3):10506.
Hagan MM, Rushing PA, Benoit SC, Woods SC, Seeley RJ. Opioid
receptor involvement in the effect of AgRP- (83132) on food
intake and food selection. Am J Physiol Regul Integr Comp Physiol
2001;280(3):814R821.
Hagan MM, Rushing PA, Pritchard LM, Schwartz MW, Strack
AM, Van Der Ploeg LH, et al. Long-term orexigenic effects
of AgRP-(83132) involve mechanisms other than melanocortin
receptor blockade. Am J Physiol Regul Integr Comp Physiol
2000;279(1):R4752.
Hahn TM, Breininger JF, Baskin DG, Schwartz MW. Co-expression
of Agrp and NPY in fasting-activated hypothalamic neurons. Nat
Neurosci 1998;1(4):2712.
Hakansson-Ovesjo ML, Collin M, Meister B. Down-regulated
STAT3 messenger ribonucleic acid and STAT3 protein in the
hypothalamic arcuate nucleus of the obese leptin-deficient (ob/ob)
mouse. Endocrinology 2000;141(11):394655.
Halverson MJ, Donelan KJ, Granholm NH, Cheesbrough TM,
Westby CA, Marshall DM. Rapid communication: sequencing
of the porcine agouti-related protein (AGRP) gene. J Anim Sci
2002;80(5):13889.
Haskell-Luevano C, Cone RD, Monck EK, Wan YP. Structure activity studies of the melanocortin-4 receptor by in vitro mutagenesis:
identification of agouti-related protein (agrp), melanocortin agonist
and synthetic peptide antagonist interaction determinants. Biochemistry 2001;40(20):616479.
Haskell-Luevano C, Monck EK. Agouti-related protein functions as
an inverse agonist at a constitutively active brain melanocortin-4
receptor. Regul Pept 2001;99(1):17.
Hegyi K, Fulop K, Kovacs K, Toth S, Falus A. Leptin-induced
signal transduction pathways. Cell Biol Int 2004;28(3):15969.
Hoggard N, Hunter L, Duncan JS, Rayner DV. Regulation of
adipose tissue leptin secretion by alpha-melanocyte-stimulating hormone and agouti-related protein: further evidence of an interaction
between leptin and the melanocortin signalling system. J Mol
Endocrinol 2004;32(1):14553.
Hoggard N, Johnstone AM, Faber P, Gibney ER, Elia M, Lobley G,
et al. Plasma concentrations of alpha-MSH, AgRP and leptin in lean
and obese men and their relationship to differing states of energy
balance perturbation. Clin Endocrinol (Oxf) 2004;61(1):319.

A.M. Stutz et al. / Peptides 26 (2005) 17711781


[51] Jackson PJ, McNulty JC, Yang YK, Thompson DA, Chai B,
Gantz I, et al. Design, pharmacology, and NMR structure of a
minimized cystine knot with agouti-related protein activity. Biochemistry 2002;41(24):756572.
[52] Jeanrenaud B, Rohner-Jeanrenaud F. CNS-periphery relationships
and body weight homeostasis: influence of the glucocorticoid status. Int J Obes Relat Metab Disord 2000;24(Suppl. 2):S746.
[53] Joseph CG, Bauzo RM, Xiang Z, Shaw AM, Millard WJ,
Haskell-Luevano C. Elongation studies of the human agoutirelated protein (AGRP) core decapeptide (Yc[CRFFNAFC]Y)
results in antagonism at the mouse melanocortin-3 receptor. Peptides 2003;24(2):26370.
[54] Kamegai J, Tamura H, Ishii S, Sugihara H, Wakabayashi I. Thyroid
hormones regulate pituitary growth hormone secretagogue receptor
gene expression. J Neuroendocrinol 2001;13(3):2758.
[55] 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):4797800.
[56] 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):243843.
[57] Kamphuis MM, Westerterp-Plantenga MS, Saris WH. Fat-specific
satiety in humans for fat high in linoleic acid versus fat high in
oleic acid. Eur J Clin Nutr 2001;55(6):499508.
[58] Kas MJ, van Dijk G, Scheurink AJ, Adan RA. Agouti-related protein prevents self-starvation. Mol Psychiatry 2003;8(2):23540.
[59] Kastin AJ, Akerstrom V, Hackler L. Agouti-related protein(83-132)
aggregates and crosses the bloodbrain barrier slowly. Metabolism
2000;49(11):14448.
[60] Katsuki A, Sumida Y, Gabazza EC, Murashima S, Tanaka T, Furuta
M, et al. Plasma levels of agouti-related protein are increased in
obese men. J Clin Endocrinol Metab 2001;86(5):19214.
[61] Kim JB, Spiegelman BM. ADD1/SREBP1 promotes adipocyte differentiation and gene expression linked to fatty acid metabolism.
Genes Dev 1996;10(9):1096107.
[62] Kim MS, Rossi M, Abbott CR, AlAhmed SH, Smith DM,
Bloom SR. Sustained orexigenic effect of Agouti related protein
may be not mediated by the melanocortin 4 receptor. Peptides
2002;23(6):106976.
[63] Klovins J, Haitina T, Fridmanis D, Kilianova Z, Kapa I, Fredriksson R, et al. The melanocortin system in Fugu: determination of
POMC/AGRP/MCR gene repertoire and synteny. As well as pharmacology and anatomical distribution of the MCRs. Mol Biol Evol
2004;21(3):56379.
[64] 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):65660.
[65] Korner J, Wissig S, Kim A, Conwell IM, Wardlaw SL.
Effects of agouti-related protein on metabolism and hypothalamic neuropeptide gene expression. J Neuroendocrinol 2003;15(12):
111621.
[66] Lawton CL, Delargy HJ, Brockman J, Smith FC, Blundell JE. The
degree of saturation of fatty acids influences post-ingestive satiety.
Br J Nutr 2000;83(5):47382.
[67] Lee K, Li B, Xi X, Suh Y, Martin RJ. The role of neuronal energy
status in the regulation of AMP-activated protein kinase, orexigenic neuropeptides expression and feeding behavior. Endocrinology 2004.
[68] Lefevre P, Tripon E, Plumelet C, Douaire M, Diot C. Effects
of polyunsaturated fatty acids and clofibrate on chicken stearoylcoA desaturase 1 gene expression. Biochem Biophys Res Commun
2001;280(1):2531.
[69] Lu D, Willard D, Patel IR, Kadwell S, Overton L, Kost T, et
al. Agouti protein is an antagonist of the melanocyte-stimulatinghormone receptor. Nature 1994;371(6500):799802.

1779

[70] Lu XY, Shieh KR, Kabbaj M, Barsh GS, Akil H, Watson SJ.
Diurnal rhythm of agouti-related protein and its relation to corticosterone and food intake. PG - 3905-3915. Endocrinology
2002;143(10).
[71] MacNeil DJ, Howard AD, Guan X, Fong TM, Nargund RP,
Bednarek MA, et al. The role of melanocortins in body weight
regulation: opportunities for the treatment of obesity. Eur J Pharmacol 2002;440(23):14157.
[72] Madiehe AM, Lin L, White C, Braymer HD, Bray GA, York DA.
Constitutive activation of STAT-3 and downregulation of SOCS-3
expression induced by adrenalectomy. Am J Physiol Regul Integr
Comp Physiol 2001;281(6):R204858.
[73] Makimura H, Mizuno TM, Mastaitis JW, Agami R, Mobbs CV.
Reducing hypothalamic AGRP by RNA interference increases
metabolic rate and decreases body weight without influencing food
intake. BMC Neurosci 2002;3(1):18.
[74] Marks DL, Boucher N, Lanouette CM, Perusse L, Brookhart G,
Comuzzie AG, et al. Ala67Thr polymorphism in the Agouti-related
peptide gene is associated with inherited leanness in humans. Am
J Med Genet 2004;126A(3):26771.
[75] Marks DL, Cone RD. Central melanocortins and the regulation of
weight during acute and chronic disease. Recent Prog Horm Res
2001;56:35975.
[76] Mayfield DK, Brown AM, Page GP, Garvey WT, Shriver MD,
Argyropoulos G. A role for the agouti related protein promoter
in obesity and type 2 diabetes. Biochem Biophys Res Commun
2001;287(2):56873.
[77] Millhauser GL, McNulty JC, Jackson PJ, Thompson DA, Barsh
GS, Gantz I. Loops and links: structural insights into the remarkable function of the agouti-related protein. Ann N Y Acad Sci
2003;994:2735.
[78] Minokoshi Y, Alquier T, Furukawa N, Kim YB, Lee A, Xue B, et
al. AMP-kinase regulates food intake by responding to hormonal
and nutrient signals in the hypothalamus. Nature 2004.
[79] Minokoshi Y, Kahn BB. Role of AMP-activated protein kinase in
leptin-induced fatty acid oxidation in muscle. Biochem Soc Trans
2003;31(Pt 1):196201.
[80] Moran TH, Schwartz GJ. Neurobiology of cholecystokinin. Crit
Rev Neurobiol 1994;9(1):128.
[81] Morgan HD, Sutherland HG, Martin DI, Whitelaw E. Epigenetic inheritance at the agouti locus in the mouse. Nat Genet
1999;23(3):3148.
[82] Nakazato M, Murakami N, Date Y, Kojima M, Matsuo H, Kangawa
K, et al. A role for ghrelin in the central regulation of feeding.
Nature 2001;409(6817):1948.
[83] Nijenhuis WA, Oosterom J, Adan RA. AgRP(83-132) acts as
an inverse agonist on the human-melanocortin-4 receptor. Mol
Endocrinol 2001;15(1):16471.
[84] Niswender KD, Gallis B, Blevins JE, Corson MA, Schwartz MW,
Baskin DG. Immunocytochemical detection of phosphatidylinositol
3-kinase activation by insulin and leptin. J Histochem Cytochem
2003;51(3):27583.
[85] Niswender KD, Morrison CD, Clegg DJ, Olson R, Baskin DG,
Myers Jr MG, et al. Insulin activation of phosphatidylinositol 3kinase in the hypothalamic arcuate nucleus: a key mediator of
insulin-induced anorexia. Diabetes 2003;52(2):22731.
[86] Ntambi JM, Bene H. Polyunsaturated fatty acid regulation of
gene expression. J Mol Neurosci 2001;16(23):2738, discussion
279284.
[87] Obici S, Feng Z, Morgan K, Stein D, Karkanias G, Rossetti L.
Central administration of oleic acid inhibits glucose production
and food intake. Diabetes 2002;51(2):2715.
[88] Ollmann MM, Lamoreux ML, Wilson BD, Barsh GS. Interaction
of Agouti protein with the melanocortin 1 receptor in vitro and in
vivo. Genes Dev 1998;12(3):31630.
[89] Ollmann MM, Wilson BD, Yang YK, Kerns JA, Chen Y, Gantz
I, et al. Antagonism of central melanocortin receptors in vitro

1780

[90]

[91]

[92]

[93]

[94]

[95]

[96]

[97]

[98]
[99]

[100]

[101]

[102]

[103]

[104]

[105]

[106]

A.M. Stutz et al. / Peptides 26 (2005) 17711781


and in vivo by agouti-related protein. Science 1997;278(5335):
1358.
Olszewski PK, Wickwire K, Wirth MM, Levine AS, Giraudo SQ.
Agouti-related protein: appetite or reward? Ann N Y Acad Sci
2003;994:18791.
Osborne TF. Creating a SCAP-less liver keeps SREBPs pinned
in the ER membrane and prevents increased lipid synthesis
in response to low cholesterol and high insulin. Genes Dev
2001;15(15):18738.
Osborne TF. Sterol regulatory element-binding proteins (SREBPs):
key regulators of nutritional homeostasis and insulin action. J Biol
Chem 2000;275(42):3237982.
Phillips-Singh D, Li Q, Takeuchi S, Ohkubo T, Sharp PJ, Boswell
T. Fasting differentially regulates expression of agouti-related peptide, pro-opiomelanocortin, prepro-orexin, and vasoactive intestinal
polypeptide mRNAs in the hypothalamus of Japanese quail. Cell
Tissue Res 2003;313(2):21725.
Pritchard LE, Armstrong D, Davies N, Oliver RL, Schmitz CA,
Brennand JC, et al. Agouti-related protein (83132) is a competitive
antagonist at the human melanocortin-4 receptor: no evidence for
differential interactions with pro-opiomelanocortin-derived ligands.
J Endocrinol 2004;180(1):18391.
Qian S, Chen H, Weingarth D, Trumbauer ME, Novi DE, Guan X,
et al. Neither agouti-related protein nor neuropeptide Y is critically
required for the regulation of energy homeostasis in mice. Mol Cell
Biol 2002;22(14):502735.
Quillan JM, Jayawickreme CK, Lerner MR. Combinatorial
diffusion assay used to identify topically active melanocytestimulating hormone receptor antagonists. Proc Natl Acad Sci USA
1995;92(7):28948.
Quillan JM, Sadee W, Wei ET, Jimenez C, Ji L, Chang JK. A
synthetic human Agouti-related protein-(83132)-NH2 fragment is
a potent inhibitor of melanocortin receptor function. FEBS Lett
1998;428(12):5962.
Rayner DV, Trayhurn P. Regulation of leptin production: sympathetic nervous system interactions. J Mol Med 2001;79(1):820.
Reizes O, Benoit SC, Strader AD, Clegg DJ, Akunuru S, Seeley RJ. Syndecan-3 modulates food intake by interacting with the
melanocortin/AgRP pathway. Ann N Y Acad Sci 2003;994:66
73.
Reizes O, Lincecum J, Wang Z, Goldberger O, Huang L, Kaksonen M, et al. Transgenic expression of syndecan-1 uncovers
a physiological control of feeding behavior by syndecan-3. Cell
2001;106(1):10516.
Rocha M, Bing C, Williams G, Puerta M. Pregnancyinduced hyperphagia is associated with increased gene expression of hypothalamic agouti-related peptide in rats. Regul Pept
2003;114(23):15965.
Rosenfeld RD, Zeni L, Welcher AA, Narhi LO, Hale C, Marasco
J, et al. Biochemical, biophysical, and pharmacological characterization of bacterially expressed human agouti-related protein.
Biochemistry 1998;37(46):1604152.
Rossi M, Kim MS, Morgan DG, Small CJ, Edwards CM, Sunter
D, et al. A C-terminal fragment of Agouti-related protein increases
feeding and antagonizes the effect of alpha-melanocyte stimulating
hormone in vivo. Endocrinology 1998;139(10):442831.
Saravia FE, Grillo CA, Ferrini M, Roig P, Lima AE, de Kloet
ER, et al. Changes of hypothalamic and plasma vasopressin in rats
with deoxycorticosterone-acetate induced salt appetite. J Steroid
Biochem Mol Biol 1999;70(13):4757.
Schwartz MW, Baskin DG, Bukowski TR, Kuijper JL, Foster D,
Lasser G, et al. Specificity of leptin action on elevated blood glucose levels and hypothalamic neuropeptide Y gene expression in
ob/ob mice. Diabetes 1996;45(4):5315.
Seeley RJ, Yagaloff KA, Fisher SL, Burn P, Thiele TE, van
Dijk G, et al. Melanocortin receptors in leptin effects. Nature
1997;390(6658):349.

[107] Sergeyev V, Broberger C, Gorbatyuk O, Hokfelt T. Effect of


2-mercaptoacetate and 2-deoxy-d-glucose administration on the
expression of NPY, AGRP, POMC, MCH and hypocretin/orexin
in the rat hypothalamus. Neuroreport 2000;11(1):11721.
[108] Shen CP, Wu KK, Shearman LP, Camacho R, Tota MR, Fong TM,
et al. Plasma agouti-related protein level: a possible correlation
with fasted and fed states in humans and rats. J Neuroendocrinol
2002;14(8):60710.
[109] Shimokawa T, Kumar MV, Lane MD. Effect of a fatty acid synthase
inhibitor on food intake and expression of hypothalamic neuropeptides. Proc Natl Acad Sci USA 2002;99(1):6671.
[110] Shutter JR, Graham M, Kinsey AC, Scully S, Luthy R, Stark KL.
Hypothalamic expression of ART, a novel gene related to agouti,
is up-regulated in obese and diabetic mutant mice. Genes Dev
1997;11(5):593602.
[111] Small CJ, Kim MS, Stanley SA, Mitchell JR, Murphy K, Morgan DG, et al. Effects of chronic central nervous system administration of agouti-related protein in pair-fed animals. Diabetes
2001;50(2):24854.
[112] Small CJ, Liu YL, Stanley SA, Connoley IP, Kennedy A, Stock
MJ, et al. CNS administration of Agouti-related protein (Agrp)
reduces energy expenditure. Int J Obes Relat Metab Disord
2003;27(4):5303.
[113] Song Y, Golling G, Thacker TL, Cone RD. Agouti-related protein (AGRP) is conserved and regulated by metabolic state in the
zebrafish, Danio rerio. Endocrine 2003;22(3):25765.
[114] Sorensen A, Adam CL, Findlay PA, Marie M, Thomas L, Travers
MT, et al. Leptin secretion and hypothalamic neuropeptide and
receptor gene expression in sheep. Am J Physiol Regul Integr
Comp Physiol 2002;282(4):1227R1235.
[115] Strader AD, Buntin JD. Changes in agouti-related peptide during
the ring dove breeding cycle in relation to prolactin and parental
hyperphagia. J Neuroendocrinol 2003;15(11):104653.
[116] Strader AD, Schioth HB, Buntin JD. The role of the melanocortin
system and the melanocortin-4 receptor in ring dove (Streptopelia
risoria) feeding behavior. Brain Res 2003;960(12):11221.
[117] Takeuchi S, Takahashi S. A possible involvement of melanocortin 3
receptor in the regulation of adrenal gland function in the chicken.
Biochim Biophys Acta 1999;1448(3):5128.
[118] Takeuchi S, Teshigawara K, Takahashi S. Widespread expression of
Agouti-related protein (AGRP) in the chicken: a possible involvement of AGRP in regulating peripheral melanocortin systems in
the chicken. Biochim Biophys Acta 2000;1496(23):2619.
[119] Thirumoorthy R, Holder JR, Bauzo RM, Richards NG,
Edison AS, Haskell-Luevano C. Novel agouti-related-proteinbased melanocortin-1 receptor antagonist. J Med Chem
2001;44(24):411424.
[120] Tritos NA, Vicent D, Gillette J, Ludwig DS, Flier ES, Maratos-Flier
E. Functional interactions between melanin-concentrating hormone,
neuropeptide Y, and anorectic neuropeptides in the rat hypothalamus. Diabetes 1998;47(11):168792.
[121] Tschop M, Smiley DL, Heiman ML. Ghrelin induces adiposity in
rodents. Nature 2000;407(6806):90813.
[122] Uhe AM, Szmukler GI, Collier GR, Hansky J, ODea K, Young
GP. Potential regulators of feeding behavior in anorexia nervosa.
Am J Clin Nutr 1992;55(1):2832.
[123] Vaisse C, Clement K, Durand E, Hercberg S, Guy-Grand B, Froguel
P. Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity. J Clin Invest 2000;106(2):253
62.
[124] van den Top M, Lee K, Whyment AD, Blanks AM, Spanswick D.
Orexigen-sensitive NPY/AgRP pacemaker neurons in the hypothalamic arcuate nucleus. Nat Neurosci 2004;7(5):4934.
[125] Vergoni AV, Bertolini A. Role of melanocortins in the central control of feeding. Eur J Pharmacol 2000;405(13):2532.
[126] Vink T, Hinney A, van Elburg AA, van Goozen SH, Sandkuijl
LA, Sinke RJ, et al. Association between an agouti-related pro-

A.M. Stutz et al. / Peptides 26 (2005) 17711781

[127]

[128]

[129]

[130]

[131]

[132]

tein gene polymorphism and anorexia nervosa. Mol Psychiatry


2001;6(3):3258.
Wang H, Storlien LH, Huang XF. Effects of dietary fat types
on body fatness, leptin, and ARC leptin receptor, NPY, and
AgRP mRNA expression. Am J Physiol Endocrinol Metab
2002;282(6):E13529.
Wilson BD, Ollmann MM, Barsh GS. The role of agoutirelated protein in regulating body weight. Mol Med Today
1999;5(6):2506.
Wirth MM, Olszewski PK, Levine AS, Giraudo SQ. Effect of
Agouti-related protein on development of conditioned taste aversion
and oxytocin neuronal activation. Neuroreport 2002;13(10):13558.
Wortley KE, Anderson K, Garcia K, Murray J, Malinova L, Liu
R, et al. Deletion of ghrelin reveals no effect on food intake, but
a primary role in energy balance. Obes Res 2004;12(1):170.
Wren AM, Small CJ, Abbott CR, Dhillo WS, Seal LJ, Cohen MA,
et al. Ghrelin causes hyperphagia and obesity in rats. Diabetes
2001;50(11):25407.
Xiao E, Xia-Zhang L, Vulliemoz NR, Ferin M, Wardlaw
SL. Agouti-related protein stimulates the hypothalamic-pituitaryadrenal (HPA) axis and enhances the HPA response to interleukin-1
in the primate. Endocrinology 2003;144(5):173641.

1781

[133] Yang YK, Dickinson CJ, Zeng Q, Li JY, Thompson DA, Gantz I.
Contribution of melanocortin receptor exoloops to Agouti-related
protein binding. J Biol Chem 1999;274(20):141006.
[134] Yang YK, Thompson DA, Dickinson CJ, Wilken J, Barsh GS,
Kent SB, et al. Characterization of Agouti-related protein binding to melanocortin receptors. Mol Endocrinol 1999;13(1):148
55.
[135] Yasuda T, Masaki T, Kakuma T, Yoshimatsu H. Hypothalamic melanocortin system regulates sympathetic nerve activity in
brown adipose tissue. Exp Biol Med (Maywood) 2004;229(3):
2359.
[136] Zakrzewska KE, Sainsbury A, Cusin I, Rouru J, Jeanrenaud B,
Rohner-Jeanrenaud F. Selective dependence of intracerebroventricular neuropeptide Y-elicited effects on central glucocorticoids.
Endocrinology 1999;140(7):31837.
[137] Zheng H, Corkern MM, Crousillac SM, Patterson LM, Phifer CB,
Berthoud HR. Neurochemical phenotype of hypothalamic neurons
showing Fos expression 23 h after intracranial AgRP. Am J Physiol
Regul Integr Comp Physiol 2002;282(6):1773R1781.
[138] Zimanyi IA, Pelleymounter MA. The role of melanocortin peptides
and receptors in regulation of energy balance. Curr Pharm Des
2003;9(8):62741.

You might also like